1
|
Albrecht FB, Schick AK, Klatt A, Schmidt FF, Nellinger S, Kluger PJ. Exploring Morphological and Molecular Properties of Different Adipose Cell Models: Monolayer, Spheroids, Gellan Gum-Based Hydrogels, and Explants. Macromol Biosci 2024:e2400320. [PMID: 39450850 DOI: 10.1002/mabi.202400320] [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: 09/18/2024] [Revised: 10/04/2024] [Indexed: 10/26/2024]
Abstract
White adipose tissue (WAT) plays a crucial role in energy homeostasis and secretes numerous adipokines with far-reaching effects. WAT is linked to diseases such as diabetes, cardiovascular disease, and cancer. There is a high demand for suitable in vitro models to study diseases and tissue metabolism. Most of these models are covered by 2D-monolayer cultures. This study aims to evaluate the performance of different WAT models to better derive potential applications. The stability of adipocyte characteristics in spheroids and two 3D gellan gum hydrogels with ex situ lobules and 2D-monolayer culture is analyzed. First, the differentiation to achieve adipocyte-like characteristics is determined. Second, to evaluate the maintenance of differentiated ASC-based models, an adipocyte-based model, and explants over 3 weeks, viability, intracellular lipid content, perilipin A expression, adipokine, and gene expression are analyzed. Several advantages are supported using each of the models. Including, but not limited to, the strong differentiation in 2D-monolayers, the self-assembling within spheroids, the long-term stability of the stem cell-containing hydrogels, and the mature phenotype within adipocyte-containing hydrogels and the lobules. This study highlights the advantages of 3D models due to their more in vivo-like behavior and provides an overview of the different adipose cell models.
Collapse
Affiliation(s)
- Franziska B Albrecht
- Reutlingen Research Institute, Reutlingen University, Alteburgstraße 150, 72762, Reutlingen, Germany
- Faculty of Natural Science, University of Hohenheim, Schloss Hohenheim 1, 70599, Stuttgart, Germany
| | - Ann-Kathrin Schick
- Faculty of Science, Energy and Building Services, Esslingen University, Kanalstraße 33, 73728, Esslingen, Germany
| | - Annemarie Klatt
- Reutlingen Research Institute, Reutlingen University, Alteburgstraße 150, 72762, Reutlingen, Germany
| | - Freia F Schmidt
- Reutlingen Research Institute, Reutlingen University, Alteburgstraße 150, 72762, Reutlingen, Germany
| | - Svenja Nellinger
- Reutlingen Research Institute, Reutlingen University, Alteburgstraße 150, 72762, Reutlingen, Germany
| | - Petra J Kluger
- School of Life Sciences, Reutlingen University, Alteburgstraße 150, 72762, Reutlingen, Germany
| |
Collapse
|
2
|
Krumins E, Lentz JC, Sutcliffe B, Sohaib A, Jacob PL, Brugnoli B, Cuzzucoli Crucitti V, Cavanagh R, Owen R, Moloney C, Ruiz-Cantu L, Francolini I, Howdle SM, Shusteff M, Rose FRAJ, Wildman RD, He Y, Taresco V. Glycerol-based sustainably sourced resin for volumetric printing. GREEN CHEMISTRY : AN INTERNATIONAL JOURNAL AND GREEN CHEMISTRY RESOURCE : GC 2024; 26:1345-1355. [PMID: 38323306 PMCID: PMC10840650 DOI: 10.1039/d3gc03607c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2023] [Accepted: 01/02/2024] [Indexed: 02/08/2024]
Abstract
Volumetric Additive Manufacturing (VAM) represents a revolutionary advancement in the field of Additive Manufacturing, as it allows for the creation of objects in a single, cohesive process, rather than in a layer-by-layer approach. This innovative technique offers unparalleled design freedom and significantly reduces printing times. A current limitation of VAM is the availability of suitable resins with the required photoreactive chemistry and from sustainable sources. To support the application of this technology, we have developed a sustainable resin based on polyglycerol, a bioderived (e.g., vegetable origin), colourless, and easily functionisable oligomer produced from glycerol. To transform polyglycerol-6 into an acrylate photo-printable resin we adopted a simple, one-step, and scalable synthesis route. Polyglycerol-6-acrylate fulfils all the necessary criteria for volumetric printing (transparency, photo-reactivity, viscosity) and was successfully used to print a variety of models with intricate geometries and good resolution. The waste resin was found to be reusable with minimal performance issues, improving resin utilisation and minimising waste material. Furthermore, by incorporating dopants such as poly(glycerol) adipate acrylate (PGA-A) and 10,12-pentacosadyinoic acid (PCDA), we demonstrated the ability to print objects with a diverse range of functionalities, including temperature sensing probes and a polyester excipient, highlighting the potential applications of these new resins.
Collapse
Affiliation(s)
- Eduards Krumins
- School of Chemistry, University of Nottingham Nottingham NG7 2RD UK
| | - Joachim C Lentz
- School of Chemistry, University of Nottingham Nottingham NG7 2RD UK
| | - Ben Sutcliffe
- School of Pharmacy, Nottingham Biodiscovery Institute, University of Nottingham NG7 2RD Nottingham UK
| | - Ali Sohaib
- Faculty of Engineering, University of Nottingham Nottingham NG7 2RD UK
| | - Philippa L Jacob
- School of Chemistry, University of Nottingham Nottingham NG7 2RD UK
| | - Benedetta Brugnoli
- Department of Chemistry, Sapienza University of Rome Piazzale A. Moro 5 00185 Rome Italy
| | | | - Robert Cavanagh
- School of Pharmacy, Nottingham Biodiscovery Institute, University of Nottingham NG7 2RD Nottingham UK
- School of Medicine, University of Nottingham Biodiscovery Institute, University of Nottingham NG7 2RD UK
| | - Robert Owen
- School of Pharmacy, Nottingham Biodiscovery Institute, University of Nottingham NG7 2RD Nottingham UK
| | - Cara Moloney
- School of Medicine, University of Nottingham Biodiscovery Institute, University of Nottingham NG7 2RD UK
| | - Laura Ruiz-Cantu
- Faculty of Engineering, University of Nottingham Nottingham NG7 2RD UK
| | - Iolanda Francolini
- Department of Chemistry, Sapienza University of Rome Piazzale A. Moro 5 00185 Rome Italy
| | - Steven M Howdle
- School of Chemistry, University of Nottingham Nottingham NG7 2RD UK
| | - Maxim Shusteff
- Lawrence Livermore National Laboratory Livermore CA 94550 USA
| | - Felicity R A J Rose
- School of Pharmacy, Nottingham Biodiscovery Institute, University of Nottingham NG7 2RD Nottingham UK
| | - Ricky D Wildman
- Faculty of Engineering, University of Nottingham Nottingham NG7 2RD UK
| | - Yinfeng He
- Faculty of Engineering, University of Nottingham Nottingham NG7 2RD UK
- Nottingham Ningbo China Beacons of Excellence Research and Innovation Institute University of Nottingham Ningbo China Ningbo 315100 China
| | - Vincenzo Taresco
- School of Chemistry, University of Nottingham Nottingham NG7 2RD UK
| |
Collapse
|
3
|
Ogino S, Yamada A, Nakano T, Lee S, Yamanaka H, Tsuge I, Sowa Y, Sakamoto M, Kyoko F, Kambe Y, Kato Y, Arata J, Yamauchi K, Yamaoka T, Morimoto N. Long term observation of de novo adipogenesis using novel bioabsorbable implants with larger size in a porcine model. Regen Ther 2023; 24:324-331. [PMID: 37649673 PMCID: PMC10463193 DOI: 10.1016/j.reth.2023.08.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 07/27/2023] [Accepted: 08/13/2023] [Indexed: 09/01/2023] Open
Abstract
Introduction The regeneration of adipose tissue in patients after breast cancer surgery would be desirable without the use of growth factors or cells to avoid potential recurrence and metastasis. We reported that prolate spheroidal-shaped poly-L-lactic acid (PLLA) mesh implants of approximately 18-mm polar diameter and 7.5-mm greatest equatorial diameter containing collagen sponge (CS) would be replaced by regenerated adipose tissue after implantation, thereby suggesting an innovative method for breast reconstruction. Our study aimed to evaluate the adipose tissue regeneration ability of implant aggregates in a porcine model. Methods We prepared implant aggregates consisting of thirty PLLA mesh implants containing CS packed in a woven poly (glycolic acid) bag. The implant aggregates were inserted under the mammary glands in the porcine abdomen for a year. Single and double groups were classified by inserting either one or two implant aggregates on each side of the abdomen, respectively. Results In both groups, the volume of the implant aggregates decreased over time, and the formation of adipose tissue peaked between 6 and 9 months. Histologically, the formation of adipose tissue was confirmed in the area that was in contact with native adipose tissue. Conclusions Our implant aggregates could induce the autologous adipose tissue after long term implantation in vivo, without the use of any growth factor or cell treatment, presenting a potential novel method of breast reconstruction.
Collapse
Affiliation(s)
- Shuichi Ogino
- Department of Plastic and Reconstructive Surgery, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga 520-2192, Japan
| | - Atsushi Yamada
- Department of Research and Development for Innovative Medical Devices and Systems, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga 520-2192, Japan
| | - Takashi Nakano
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin, Kawahara-cho, Sakyou-ku, Kyoto 606-8507, Japan
| | - Sunghee Lee
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin, Kawahara-cho, Sakyou-ku, Kyoto 606-8507, Japan
| | - Hiroki Yamanaka
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin, Kawahara-cho, Sakyou-ku, Kyoto 606-8507, Japan
| | - Itaru Tsuge
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin, Kawahara-cho, Sakyou-ku, Kyoto 606-8507, Japan
| | - Yoshihiro Sowa
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin, Kawahara-cho, Sakyou-ku, Kyoto 606-8507, Japan
| | - Michiharu Sakamoto
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin, Kawahara-cho, Sakyou-ku, Kyoto 606-8507, Japan
| | - Fukazawa Kyoko
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe Shin-machi, Suita, Osaka 564-8565, Japan
| | - Yusuke Kambe
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe Shin-machi, Suita, Osaka 564-8565, Japan
| | - Yuki Kato
- Gunze QOL Research Center Laboratory, 1 Zeze, Aono-cho, Ayabe, Kyoto 623-0051, Japan
| | - Jun Arata
- Department of Plastic and Reconstructive Surgery, Shiga University of Medical Science, Seta Tsukinowa-cho, Otsu, Shiga 520-2192, Japan
| | - Koji Yamauchi
- Gunze QOL Research Center Laboratory, 1 Zeze, Aono-cho, Ayabe, Kyoto 623-0051, Japan
| | - Tetsuji Yamaoka
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, 6-1 Kishibe Shin-machi, Suita, Osaka 564-8565, Japan
| | - Naoki Morimoto
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, 54 Shogoin, Kawahara-cho, Sakyou-ku, Kyoto 606-8507, Japan
| |
Collapse
|
4
|
Ky A, McCoy AJ, Flesher CG, Friend NE, Li J, Akinleye K, Patsalis C, Lumeng CN, Putnam AJ, O’Rourke RW. Matrix density regulates adipocyte phenotype. Adipocyte 2023; 12:2268261. [PMID: 37815174 PMCID: PMC10566443 DOI: 10.1080/21623945.2023.2268261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/22/2023] [Accepted: 09/29/2023] [Indexed: 10/11/2023] Open
Abstract
Alterations of the extracellular matrix contribute to adipose tissue dysfunction in metabolic disease. We studied the role of matrix density in regulating human adipocyte phenotype in a tunable hydrogel culture system. Lipid accumulation was maximal in intermediate hydrogel density of 5 weight %, relative to 3% and 10%. Adipogenesis and lipid and oxidative metabolic gene pathways were enriched in adipocytes in 5% relative to 3% hydrogels, while fibrotic gene pathways were enriched in 3% hydrogels. These data demonstrate that the intermediate density matrix promotes a more adipogenic, less fibrotic adipocyte phenotype geared towards increased lipid and aerobic metabolism. These observations contribute to a growing literature describing the role of matrix density in regulating adipose tissue function.
Collapse
Affiliation(s)
- Alexander Ky
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Atticus J. McCoy
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Carmen G. Flesher
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Graduate Program, University of Pennsylvania, Philadelphia, PA, USA
| | - Nicole E. Friend
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Jie Li
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Kore Akinleye
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
| | - Christopher Patsalis
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI, USA
- Department of Bioinformatics, University of Michigan, Ann Arbor, MI, USA
| | - Carey N. Lumeng
- Department of Pediatrics and Communicable Diseases, University of Michigan, Ann Arbor, MI, USA
- Graduate Program in Immunology, University of Michigan, Ann Arbor, MI, USA
- Graduate Program in Cellular and Molecular Biology, University of Michigan, Ann Arbor, MI, USA
| | - Andrew J. Putnam
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Robert W. O’Rourke
- Department of Surgery, University of Michigan, Ann Arbor, MI, USA
- Department of Surgery, Veterans Affairs Ann Arbor Healthcare System, Ann Arbor, MI, USA
| |
Collapse
|
5
|
Karanfil AS, Louis F, Sowa Y, Matsusaki M. ECM proteins and cationic polymers coating promote dedifferentiation of patient-derived mature adipocytes to stem cells. Biomater Sci 2023; 11:7623-7638. [PMID: 37830400 DOI: 10.1039/d3bm00934c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/14/2023]
Abstract
Reprogramming of mature adipocytes is an attractive research area due to the plasticity of these cells. Mature adipocytes can be reprogrammed in vitro, transforming them into dedifferentiated fat cells (DFATs), which are considered a new type of stem cell, and thereby have a high potential for use in tissue engineering and regenerative medicine. However, there are still no reports or findings on in vitro controlling the dedifferentiation. Although ceiling culture performed in related studies is a relatively simple method, its yield is low and does not allow manipulation of mature adipocytes to increase or decrease the dedifferentiation. In this study, to understand the role of physicochemical surface effects on the dedifferentiation of patient-derived mature adipocytes, the surfaces of cell culture flasks were coated with extracellular matrix, basement membrane proteins, and cationic/anionic polymers. Extracellular matrix such as fibronectin and collagen type I, and basement membrane proteins such as collagen type IV and laminin strongly promoted dedifferentiation of mature adipocytes, with laminin showing the highest effect with a DFAT ratio of 2.98 (±0.84). Interestingly, cationic polymers also showed a high dedifferentiation effect, but anionic polymers did not, and poly(diallyl dimethylammonium chloride) showed the highest DFAT ratio of 2.27 (±2.8) among the cationic polymers. Protein assay results revealed that serum proteins were strongly adsorbed on the surfaces of the cationic polymer coating, including inducing high mature adipocyte adhesion. This study demonstrates for the first time the possibility of regulating the transformation of mature adipocytes to DFAT stem cells by controlling the physicochemical properties of the surface of conventional cell culture flasks.
Collapse
Affiliation(s)
- Aslı Sena Karanfil
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Japan.
| | - Fiona Louis
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Japan
| | - Yoshihiro Sowa
- Department of Plastic and Reconstructive Surgery, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Japan
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, Japan
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Japan.
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Japan
| |
Collapse
|
6
|
Footner E, Firipis K, Liu E, Baker C, Foley P, Kapsa RMI, Pirogova E, O'Connell C, Quigley A. Layer-by-Layer Analysis of In Vitro Skin Models. ACS Biomater Sci Eng 2023; 9:5933-5952. [PMID: 37791888 DOI: 10.1021/acsbiomaterials.3c00283] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
In vitro human skin models are evolving into versatile platforms for the study of skin biology and disorders. These models have many potential applications in the fields of drug testing and safety assessment, as well as cosmetic and new treatment development. The development of in vitro skin models that accurately mimic native human skin can reduce reliance on animal models and also allow for more precise, clinically relevant testing. Recent advances in biofabrication techniques and biomaterials have led to the creation of increasingly complex, multilayered skin models that incorporate important functional components of skin, such as the skin barrier, mechanical properties, pigmentation, vasculature, hair follicles, glands, and subcutaneous layer. This improved ability to recapitulate the functional aspects of native skin enhances the ability to model the behavior and response of native human skin, as the complex interplay of cell-to-cell and cell-to-material interactions are incorporated. In this review, we summarize the recent developments in in vitro skin models, with a focus on their applications, limitations, and future directions.
Collapse
Affiliation(s)
- Elizabeth Footner
- Electrical and Biomedical Engineering, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia
- Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
| | - Kate Firipis
- Electrical and Biomedical Engineering, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia
- Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
| | - Emily Liu
- Electrical and Biomedical Engineering, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia
- Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
| | - Chris Baker
- Department of Dermatology, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
- Skin Health Institute, Carlton, VIC 3053, Australia
- Department of Medicine, University of Melbourne, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
| | - Peter Foley
- Department of Dermatology, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
- Skin Health Institute, Carlton, VIC 3053, Australia
- Department of Medicine, University of Melbourne, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
| | - Robert M I Kapsa
- Electrical and Biomedical Engineering, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia
- Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
- Department of Medicine, University of Melbourne, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
- Centre for Clinical Neurosciences and Neurological Research, St. Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
| | - Elena Pirogova
- Electrical and Biomedical Engineering, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia
- Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
| | - Cathal O'Connell
- Electrical and Biomedical Engineering, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia
- Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
| | - Anita Quigley
- Electrical and Biomedical Engineering, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia
- Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
- Department of Medicine, University of Melbourne, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
- Centre for Clinical Neurosciences and Neurological Research, St. Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia
| |
Collapse
|
7
|
Vasella M, Arnke K, Dranseikiene D, Guzzi E, Melega F, Reid G, Klein HJ, Schweizer R, Tibbitt MW, Kim BS. Methacrylated Gelatin as a Scaffold for Mechanically Isolated Stromal Vascular Fraction for Cutaneous Wound Repair. Int J Mol Sci 2023; 24:13944. [PMID: 37762247 PMCID: PMC10530931 DOI: 10.3390/ijms241813944] [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: 08/03/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Mechanically processed stromal vascular fraction (mSVF) is a highly interesting cell source for regenerative purposes, including wound healing, and a practical alternative to enzymatically isolated SVF. In the clinical context, SVF benefits from scaffolds that facilitate viability and other cellular properties. In the present work, the feasibility of methacrylated gelatin (GelMA), a stiffness-tunable, light-inducible hydrogel with high biocompatibility is investigated as a scaffold for SVF in an in vitro setting. Lipoaspirates from elective surgical procedures were collected and processed to mSVF and mixed with GelMA precursor solutions. Non-encapsulated mSVF served as a control. Viability was measured over 21 days. Secreted basic fibroblast growth factor (bFGF) levels were measured on days 1, 7 and 21 by ELISA. IHC was performed to detect VEGF-A, perilipin-2, and CD73 expression on days 7 and 21. The impact of GelMA-mSVF on human dermal fibroblasts was measured in a co-culture assay by the same viability assay. The viability of cultured GelMA-mSVF was significantly higher after 21 days (p < 0.01) when compared to mSVF alone. Also, GelMA-mSVF secreted stable levels of bFGF over 21 days. While VEGF-A was primarily expressed on day 21, perilipin-2 and CD73-positive cells were observed on days 7 and 21. Finally, GelMA-mSVF significantly improved fibroblast viability as compared with GelMA alone (p < 0.01). GelMA may be a promising scaffold for mSVF as it maintains cell viability and proliferation with the release of growth factors while facilitating adipogenic differentiation, stromal cell marker expression and fibroblast proliferation.
Collapse
Affiliation(s)
- Mauro Vasella
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (M.V.); (G.R.)
| | - Kevin Arnke
- Center for Preclinical Development, University Hospital Zurich, 8091 Zurich, Switzerland;
| | - Dalia Dranseikiene
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, 8092 Zurich, Switzerland; (D.D.); (E.G.); (M.W.T.)
| | - Elia Guzzi
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, 8092 Zurich, Switzerland; (D.D.); (E.G.); (M.W.T.)
| | - Francesca Melega
- Institute of Pathology and Molecular Pathology, University Hospital Zurich, 8091 Zurich, Switzerland;
| | - Gregory Reid
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (M.V.); (G.R.)
| | - Holger Jan Klein
- Department of Plastic Surgery and Hand Surgery, Cantonal Hospital Aarau, 5001 Aarau, Switzerland;
| | - Riccardo Schweizer
- Department of Plastic, Reconstructive and Aesthetic Surgery, Regional Hospital Lugano, 6900 Lugano, Switzerland;
| | - Mark W. Tibbitt
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, 8092 Zurich, Switzerland; (D.D.); (E.G.); (M.W.T.)
| | - Bong-Sung Kim
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (M.V.); (G.R.)
| |
Collapse
|
8
|
Major G, Longoni A, Simcock J, Magon NJ, Harte J, Bathish B, Kemp R, Woodfield T, Lim KS. Clinical Applicability of Visible Light-Mediated Cross-linking for Structural Soft Tissue Reconstruction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2300538. [PMID: 37424046 PMCID: PMC10502829 DOI: 10.1002/advs.202300538] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 05/28/2023] [Indexed: 07/11/2023]
Abstract
Visible light-mediated cross-linking has utility for enhancing the structural capacity and shape fidelity of laboratory-based polymers. With increased light penetration and cross-linking speed, there is opportunity to extend future applications into clinical spheres. This study evaluated the utility of a ruthenium/sodium persulfate photocross-linking system for increasing structural control in heterogeneous living tissues as an example, focusing on unmodified patient-derived lipoaspirate for soft tissue reconstruction. Freshly-isolated tissue is photocross-linked, then the molar abundance of dityrosine bonds is measured using liquid chromatography tandem mass spectrometry and the resulting structural integrity assessed. The cell function and tissue survival of photocross-linked grafts is evaluated ex vivo and in vivo, with tissue integration and vascularization assessed using histology and microcomputed tomography. The photocross-linking strategy is tailorable, allowing progressive increases in the structural fidelity of lipoaspirate, as measured by a stepwise reduction in fiber diameter, increased graft porosity and reduced variation in graft resorption. There is an increase in dityrosine bond formation with increasing photoinitiator concentration, and tissue homeostasis is achieved ex vivo, with vascular cell infiltration and vessel formation in vivo. These data demonstrate the capability and applicability of photocrosslinking strategies for improving structural control in clinically-relevant settings, potentially achieving more desirable patient outcomes using minimal manipulation in surgical procedures.
Collapse
Affiliation(s)
- Gretel Major
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurch8011New Zealand
| | - Alessia Longoni
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurch8011New Zealand
| | - Jeremy Simcock
- Department of SurgeryUniversity of OtagoChristchurch8011New Zealand
| | - Nicholas J Magon
- Centre for Free Radical ResearchDepartment of Pathology and Biomedical ScienceUniversity of OtagoChristchurch8011New Zealand
| | - Jessica Harte
- Jacqui Wood Cancer CentreDivision of Cellular MedicineNinewells Hospital and Medical SchoolUniversity of DundeeDundeeScotlandDD2 1GZUK
| | - Boushra Bathish
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurch8011New Zealand
- Jacqui Wood Cancer CentreDivision of Cellular MedicineNinewells Hospital and Medical SchoolUniversity of DundeeDundeeScotlandDD2 1GZUK
| | - Roslyn Kemp
- Department of Microbiology and ImmunologyUniversity of OtagoDunedin9016New Zealand
| | - Tim Woodfield
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurch8011New Zealand
| | - Khoon S Lim
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurch8011New Zealand
- Light‐Activated Biomaterials GroupSchool of Medical SciencesUniversity of SydneySydney2006Australia
| |
Collapse
|
9
|
Asim S, Tabish TA, Liaqat U, Ozbolat IT, Rizwan M. Advances in Gelatin Bioinks to Optimize Bioprinted Cell Functions. Adv Healthc Mater 2023; 12:e2203148. [PMID: 36802199 PMCID: PMC10330013 DOI: 10.1002/adhm.202203148] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2022] [Revised: 01/31/2023] [Indexed: 02/21/2023]
Abstract
Gelatin is a widely utilized bioprinting biomaterial due to its cell-adhesive and enzymatically cleavable properties, which improve cell adhesion and growth. Gelatin is often covalently cross-linked to stabilize bioprinted structures, yet the covalently cross-linked matrix is unable to recapitulate the dynamic microenvironment of the natural extracellular matrix (ECM), thereby limiting the functions of bioprinted cells. To some extent, a double network bioink can provide a more ECM-mimetic, bioprinted niche for cell growth. More recently, gelatin matrices are being designed using reversible cross-linking methods that can emulate the dynamic mechanical properties of the ECM. This review analyzes the progress in developing gelatin bioink formulations for 3D cell culture, and critically analyzes the bioprinting and cross-linking techniques, with a focus on strategies to optimize the functions of bioprinted cells. This review discusses new cross-linking chemistries that recapitulate the viscoelastic, stress-relaxing microenvironment of the ECM, and enable advanced cell functions, yet are less explored in engineering the gelatin bioink. Finally, this work presents the perspective on the areas of future research and argues that the next generation of gelatin bioinks should be designed by considering cell-matrix interactions, and bioprinted constructs should be validated against currently established 3D cell culture standards to achieve improved therapeutic outcomes.
Collapse
Affiliation(s)
- Saad Asim
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI, 49931 USA
| | - Tanveer A. Tabish
- Cardiovascular Division, Radcliff Department of Medicine, University of Oxford, Oxford, United Kingdom
| | - Usman Liaqat
- Department of Materials Engineering, School of Chemical and Materials Engineering (SCME), National University of Sciences & Technology (NUST), Pakistan
| | - Ibrahim T. Ozbolat
- Engineering Science and Mechanics, Penn State, University Park, PA 16802, USA
- Department of Biomedical Engineering, Penn State, University Park, PA 16802, USA
- Department of Neurosurgery, Penn State, Hershey, PA 16802, USA
- Department of Medical Oncology, Cukurova University, Adana 01330, Turkey
| | - Muhammad Rizwan
- Department of Biomedical Engineering, Michigan Technological University, Houghton, MI, 49931 USA
- Health Research Institute, Michigan Technological University, Houghton, MI, 49931 USA
| |
Collapse
|
10
|
Mekhileri NV, Major G, Lim K, Mutreja I, Chitcholtan K, Phillips E, Hooper G, Woodfield T. Biofabrication of Modular Spheroids as Tumor-Scale Microenvironments for Drug Screening. Adv Healthc Mater 2023; 12:e2201581. [PMID: 36495232 PMCID: PMC11468982 DOI: 10.1002/adhm.202201581] [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: 06/30/2022] [Revised: 11/13/2022] [Indexed: 12/14/2022]
Abstract
To streamline the drug discovery pipeline, there is a pressing need for preclinical models which replicate the complexity and scale of native tumors. While there have been advancements in the formation of microscale tumor units, these models are cell-line dependent, time-consuming and have not improved clinical trial success rates. In this study, two methods for generating 3D tumor microenvironments are compared, rapidly fabricated hydrogel microspheres and traditional cell-dense spheroids. These modules are then bioassembled into 3D printed thermoplastic scaffolds, using an automated biofabrication process, to form tumor-scale models. Modules are formed with SKOV3 and HFF cells as monocultures and cocultures, and the fabrication efficiency, cell architecture, and drug response profiles are characterized, both as single modules and as multimodular constructs. Cell-encapsulated Gel-MA microspheres are fabricated with high-reproducibility and dimensions necessary for automated tumor-scale bioassembly regardless of cell type, however, only cocultured spheroids form compact modules suitable for bioassembly. Chemosensitivity assays demonstrate the reduced potency of doxorubicin in coculture bioassembled constructs and a ≈five-fold increase in drug resistance of cocultured cells in 3D modules compared with 2D monolayers. This bioassembly system is efficient and tailorable so that a variety of relevant-sized tumor constructs could be developed to study tumorigenesis and modernize drug discovery.
Collapse
Affiliation(s)
- Naveen Vijayan Mekhileri
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurchCanterbury8011New Zealand
| | - Gretel Major
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurchCanterbury8011New Zealand
| | - Khoon Lim
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurchCanterbury8011New Zealand
| | - Isha Mutreja
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurchCanterbury8011New Zealand
| | - Kenny Chitcholtan
- Department of Obstetrics and GynaecologyGynaecological Cancer Research GroupUniversity of OtagoChristchurchCanterbury8011New Zealand
| | - Elisabeth Phillips
- Mackenzie Cancer Research GroupDepartment of Pathology and Biomedical ScienceUniversity of OtagoChristchurchCanterbury8011New Zealand
| | - Gary Hooper
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurchCanterbury8011New Zealand
| | - Tim Woodfield
- Department of Orthopaedic Surgery and Musculoskeletal MedicineCentre for Bioengineering & NanomedicineUniversity of OtagoChristchurchCanterbury8011New Zealand
| |
Collapse
|
11
|
Karanfil AS, Louis F, Matsusaki M. Biofabrication of vascularized adipose tissues and their biomedical applications. MATERIALS HORIZONS 2023; 10:1539-1558. [PMID: 36789675 DOI: 10.1039/d2mh01391f] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/10/2023]
Abstract
Recent advances in adipose tissue engineering and cell biology have led to the development of innovative therapeutic strategies in regenerative medicine for adipose tissue reconstruction. To date, the many in vitro and in vivo models developed for vascularized adipose tissue engineering cover a wide range of research areas, including studies with cells of various origins and types, polymeric scaffolds of natural and synthetic derivation, models presented using decellularized tissues, and scaffold-free approaches. In this review, studies on adipose tissue types with different functions, characteristics and body locations have been summarized with 3D in vitro fabrication approaches. The reason for the particular focus on vascularized adipose tissue models is that current liposuction and fat transplantation methods are unsuitable for adipose tissue reconstruction as the lack of blood vessels results in inadequate nutrient and oxygen delivery, leading to necrosis in situ. In the first part of this paper, current studies and applications of white and brown adipose tissues are presented according to the polymeric materials used, focusing on the studies which could show vasculature in vitro and after in vivo implantation, and then the research on adipose tissue fabrication and applications are explained.
Collapse
Affiliation(s)
- Aslı Sena Karanfil
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Japan.
| | - Fiona Louis
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Japan
| | - Michiya Matsusaki
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Japan.
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Japan
| |
Collapse
|
12
|
Hofmann E, Fink J, Pignet AL, Schwarz A, Schellnegger M, Nischwitz SP, Holzer-Geissler JCJ, Kamolz LP, Kotzbeck P. Human In Vitro Skin Models for Wound Healing and Wound Healing Disorders. Biomedicines 2023; 11:biomedicines11041056. [PMID: 37189674 DOI: 10.3390/biomedicines11041056] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 03/23/2023] [Accepted: 03/24/2023] [Indexed: 05/17/2023] Open
Abstract
Skin wound healing is essential to health and survival. Consequently, high amounts of research effort have been put into investigating the cellular and molecular components involved in the wound healing process. The use of animal experiments has contributed greatly to the knowledge of wound healing, skin diseases, and the exploration of treatment options. However, in addition to ethical concerns, anatomical and physiological inter-species differences often influence the translatability of animal-based studies. Human in vitro skin models, which include essential cellular and structural components for wound healing analyses, would improve the translatability of results and reduce animal experiments during the preclinical evaluation of novel therapy approaches. In this review, we summarize in vitro approaches, which are used to study wound healing as well as wound healing-pathologies such as chronic wounds, keloids, and hypertrophic scars in a human setting.
Collapse
Affiliation(s)
- Elisabeth Hofmann
- COREMED-Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Julia Fink
- COREMED-Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Anna-Lisa Pignet
- COREMED-Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Anna Schwarz
- COREMED-Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Marlies Schellnegger
- COREMED-Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Sebastian P Nischwitz
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Judith C J Holzer-Geissler
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Lars-Peter Kamolz
- COREMED-Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Petra Kotzbeck
- COREMED-Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft mbH, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| |
Collapse
|
13
|
Hofmann E, Schwarz A, Fink J, Kamolz LP, Kotzbeck P. Modelling the Complexity of Human Skin In Vitro. Biomedicines 2023; 11:biomedicines11030794. [PMID: 36979772 PMCID: PMC10045055 DOI: 10.3390/biomedicines11030794] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2023] [Revised: 02/17/2023] [Accepted: 02/19/2023] [Indexed: 03/08/2023] Open
Abstract
The skin serves as an important barrier protecting the body from physical, chemical and pathogenic hazards as well as regulating the bi-directional transport of water, ions and nutrients. In order to improve the knowledge on skin structure and function as well as on skin diseases, animal experiments are often employed, but anatomical as well as physiological interspecies differences may result in poor translatability of animal-based data to the clinical situation. In vitro models, such as human reconstructed epidermis or full skin equivalents, are valuable alternatives to animal experiments. Enormous advances have been achieved in establishing skin models of increasing complexity in the past. In this review, human skin structures are described as well as the fast evolving technologies developed to reconstruct the complexity of human skin structures in vitro.
Collapse
Affiliation(s)
- Elisabeth Hofmann
- COREMED—Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Anna Schwarz
- COREMED—Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Julia Fink
- COREMED—Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Lars-Peter Kamolz
- COREMED—Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
| | - Petra Kotzbeck
- COREMED—Centre of Regenerative and Precision Medicine, JOANNEUM RESEARCH Forschungsgesellschaft, 8010 Graz, Austria
- Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, 8036 Graz, Austria
- Correspondence:
| |
Collapse
|
14
|
Zanon M, Montalvillo-Jiménez L, Bosch P, Cue-López R, Martínez-Campos E, Sangermano M, Chiappone A. Photocurable Thiol-yne Alginate Hydrogels for Regenerative Medicine Purposes. Polymers (Basel) 2022; 14:4709. [PMID: 36365703 PMCID: PMC9654832 DOI: 10.3390/polym14214709] [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: 09/30/2022] [Revised: 10/18/2022] [Accepted: 10/27/2022] [Indexed: 04/03/2024] Open
Abstract
Every year millions of people worldwide undergo surgical interventions, with the occurrence of mild or severe post-treatment consequences meaning that rehabilitation plays a key role in modern medicine. Considering the cases of burns and plastic surgery, the pressing need for new materials that can be used for wound patches or body fillers and are able to sustain tissue regeneration and promote cell adhesion and proliferation is clear. The challenges facing next-generation implant materials also include the need for improved structural properties for cellular organization and morphogenic guidance together with optimal mechanical, rheological, and topographical behavior. Herein, we propose for the first time a sodium alginate hydrogel obtained by a thiol-yne reaction, easily synthesized using carbodiimide chemistry in a two-step reaction. The hydrogels were formed in all cases within a few minutes of light irradiation, showing good self-standing properties under solicitation. The mechanical, rheological, topographical, and swelling properties of the gels were also tested and reported. Lastly, no cytotoxicity was detected among the hydrogels. Soluble extracts in culture media allowed cell proliferation, and no differences between samples were detected in terms of metabolic activity and DNA content. These results suggest the potential use of these cytocompatible hydrogels in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Michael Zanon
- Dipartimento di Scienza Applicata e Tecnologia, Politecnico di Torino, C.so Duca Degli Abruzzi 24, 10129 Turin, Italy
- Departamento de Química Macromolecular Aplicada, Instituto de Ciencia y Tecnología de Polímeros, Consejo Superior de Investigaciones Científicas (CSIC), C/Juan de la Cierva 3, 28006 Madrid, Spain
| | - Laura Montalvillo-Jiménez
- Departamento de Química Macromolecular Aplicada, Instituto de Ciencia y Tecnología de Polímeros, Consejo Superior de Investigaciones Científicas (CSIC), C/Juan de la Cierva 3, 28006 Madrid, Spain
| | - Paula Bosch
- Departamento de Química Macromolecular Aplicada, Instituto de Ciencia y Tecnología de Polímeros, Consejo Superior de Investigaciones Científicas (CSIC), C/Juan de la Cierva 3, 28006 Madrid, Spain
| | - Raquel Cue-López
- Departamento de Química Macromolecular Aplicada, Instituto de Ciencia y Tecnología de Polímeros, Consejo Superior de Investigaciones Científicas (CSIC), C/Juan de la Cierva 3, 28006 Madrid, Spain
- Grupo de Síntesis Orgánica y Bioevaluación, Instituto Pluridisciplinar (UCM), Unidad Asociada al Instituto de Ciencia y Tecnología de Polímeros, Instituto de Química Médica (CSIC), Paseo de Juan XXIII 1, 28040 Madrid, Spain
| | - Enrique Martínez-Campos
- Departamento de Química Macromolecular Aplicada, Instituto de Ciencia y Tecnología de Polímeros, Consejo Superior de Investigaciones Científicas (CSIC), C/Juan de la Cierva 3, 28006 Madrid, Spain
- Grupo de Síntesis Orgánica y Bioevaluación, Instituto Pluridisciplinar (UCM), Unidad Asociada al Instituto de Ciencia y Tecnología de Polímeros, Instituto de Química Médica (CSIC), Paseo de Juan XXIII 1, 28040 Madrid, Spain
| | - Marco Sangermano
- Dipartimento di Scienza Applicata e Tecnologia, Politecnico di Torino, C.so Duca Degli Abruzzi 24, 10129 Turin, Italy
| | - Annalisa Chiappone
- Dipartimento di Scienze Chimiche e Geologiche, Università Degli Studi di Cagliari, Via Università 40, 09124 Cagliari, Italy
| |
Collapse
|
15
|
Grilli F, Pitton M, Altomare L, Farè S. Decellularized fennel and dill leaves as possible 3D channel network in GelMA for the development of an in vitro adipose tissue model. Front Bioeng Biotechnol 2022; 10:984805. [DOI: 10.3389/fbioe.2022.984805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 10/17/2022] [Indexed: 11/13/2022] Open
Abstract
The development of 3D scaffold-based models would represent a great step forward in cancer research, offering the possibility of predicting the potential in vivo response to targeted anticancer or anti-angiogenic therapies. As regards, 3D in vitro models require proper materials, which faithfully recapitulated extracellular matrix (ECM) properties, adequate cell lines, and an efficient vascular network. The aim of this work is to investigate the possible realization of an in vitro 3D scaffold-based model of adipose tissue, by incorporating decellularized 3D plant structures within the scaffold. In particular, in order to obtain an adipose matrix capable of mimicking the composition of the adipose tissue, methacrylated gelatin (GelMA), UV photo-crosslinkable, was selected. Decellularized fennel, wild fennel and, dill leaves have been incorporated into the GelMA hydrogel before crosslinking, to mimic a 3D channel network. All leaves showed a loss of pigmentation after the decellularization with channel dimensions ranging from 100 to 500 µm up to 3 μm, comparable with those of human microcirculation (5–10 µm). The photo-crosslinking process was not affected by the embedded plant structures in GelMA hydrogels. In fact, the weight variation test, performed on hydrogels with or without decellularized leaves showed a weight loss in the first 96 h, followed by a stability plateau up to 5 weeks. No cytotoxic effects were detected comparing the three prepared GelMA/D-leaf structures; moreover, the ability of the samples to stimulate differentiation of 3T3-L1 preadipocytes in mature adipocytes was investigated, and cells were able to grow and proliferate in the structure, colonizing the entire microenvironment and starting to differentiate. The developed GelMA hydrogels mimicked adipose tissue together with the incorporated plant structures seem to be an adequate solution to ensure an efficient vascular system for a 3D in vitro model. The obtained results showed the potentiality of the innovative proposed approach to mimic the tumoral microenvironment in 3D scaffold-based models.
Collapse
|
16
|
Albrecht FB, Schmidt FF, Volz AC, Kluger PJ. Bioprinting of 3D Adipose Tissue Models Using a GelMA-Bioink with Human Mature Adipocytes or Human Adipose-Derived Stem Cells. Gels 2022; 8:gels8100611. [PMID: 36286112 PMCID: PMC9601941 DOI: 10.3390/gels8100611] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/21/2022] [Accepted: 09/21/2022] [Indexed: 12/31/2022] Open
Abstract
Adipose tissue is related to the development and manifestation of multiple diseases, demonstrating the importance of suitable in vitro models for research purposes. In this study, adipose tissue lobuli were explanted, cultured, and used as an adipose tissue control to evaluate in vitro generated adipose tissue models. During culture, lobule exhibited a stable weight, lactate dehydrogenase, and glycerol release over 15 days. For building up in vitro adipose tissue models, we adapted the biomaterial gelatin methacryloyl (GelMA) composition and handling to homogeneously mix and bioprint human primary mature adipocytes (MA) and adipose-derived stem cells (ASCs), respectively. Accelerated cooling of the bioink turned out to be essential for the homogeneous distribution of lipid-filled MAs in the hydrogel. Last, we compared manual and bioprinted GelMA hydrogels with MA or ASCs and the explanted lobules to evaluate the impact of the printing process and rate the models concerning the physiological reference. The viability analyses demonstrated no significant difference between the groups due to additive manufacturing. The staining of intracellular lipids and perilipin A suggest that GelMA is well suited for ASCs and MA. Therefore, we successfully constructed physiological in vitro models by bioprinting MA-containing GelMA bioinks.
Collapse
Affiliation(s)
- Franziska B. Albrecht
- Reutlingen Research Institute, Reutlingen University, 72762 Reutlingen, Germany
- Faculty of Natural Science, University of Hohenheim, 70599 Stuttgart, Germany
| | - Freia F. Schmidt
- Reutlingen Research Institute, Reutlingen University, 72762 Reutlingen, Germany
| | - Ann-Cathrin Volz
- Reutlingen Research Institute, Reutlingen University, 72762 Reutlingen, Germany
| | - Petra J. Kluger
- Faculty of Applied Chemistry, Reutlingen University, 72762 Reutlingen, Germany
- Correspondence: ; Tel.: +49-7121-271-2061
| |
Collapse
|
17
|
Rogal J, Roosz J, Teufel C, Cipriano M, Xu R, Eisler W, Weiss M, Schenke‐Layland K, Loskill P. Autologous Human Immunocompetent White Adipose Tissue-on-Chip. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2104451. [PMID: 35466539 PMCID: PMC9218765 DOI: 10.1002/advs.202104451] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/07/2021] [Revised: 03/03/2022] [Indexed: 05/07/2023]
Abstract
Obesity and associated diseases, such as diabetes, have reached epidemic proportions globally. In this era of "diabesity", white adipose tissue (WAT) has become a target of high interest for therapeutic strategies. To gain insights into mechanisms of adipose (patho-)physiology, researchers traditionally relied on animal models. Leveraging Organ-on-Chip technology, a microphysiological in vitro model of human WAT is introduced: a tailored microfluidic platform featuring vasculature-like perfusion that integrates 3D tissues comprising all major WAT-associated cellular components (mature adipocytes, organotypic endothelial barriers, stromovascular cells including adipose tissue macrophages) in an autologous manner and recapitulates pivotal WAT functions, such as energy storage and mobilization as well as endocrine and immunomodulatory activities. A precisely controllable bottom-up approach enables the generation of a multitude of replicates per donor circumventing inter-donor variability issues and paving the way for personalized medicine. Moreover, it allows to adjust the model's degree of complexity via a flexible mix-and-match approach. This WAT-on-Chip system constitutes the first human-based, autologous, and immunocompetent in vitro adipose tissue model that recapitulates almost full tissue heterogeneity and can become a powerful tool for human-relevant research in the field of metabolism and its associated diseases as well as for compound testing and personalized- and precision medicine applications.
Collapse
Affiliation(s)
- Julia Rogal
- Department for Microphysiological Systems, Institute of Biomedical EngineeringEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGBNobelstr. 12Stuttgart70569Germany
| | - Julia Roosz
- NMI Natural and Medical Sciences Institute at the University of TübingenMarkwiesenstr. 55Reutlingen72770Germany
| | - Claudia Teufel
- Department for Microphysiological Systems, Institute of Biomedical EngineeringEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
| | - Madalena Cipriano
- Department for Microphysiological Systems, Institute of Biomedical EngineeringEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
- 3R‐Center for In vitro Models and Alternatives to Animal TestingEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
| | - Raylin Xu
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGBNobelstr. 12Stuttgart70569Germany
- Harvard Medical School (HMS)25 Shattuck StBostonMA02115USA
| | - Wiebke Eisler
- Clinic for PlasticReconstructiveHand and Burn SurgeryBG Trauma CenterEberhard Karls University TübingenSchnarrenbergstraße 95Tübingen72076Germany
| | - Martin Weiss
- NMI Natural and Medical Sciences Institute at the University of TübingenMarkwiesenstr. 55Reutlingen72770Germany
- Department of Women's HealthEberhard Karls University TübingenCalwerstrasse 7Tübingen72076Germany
| | - Katja Schenke‐Layland
- NMI Natural and Medical Sciences Institute at the University of TübingenMarkwiesenstr. 55Reutlingen72770Germany
- Department of Medicine/CardiologyCardiovascular Research LaboratoriesDavid Geffen School of Medicine at UCLA675 Charles E. Young Drive South, MRL 3645Los AngelesCA90095USA
- Cluster of Excellence iFIT (EXC2180) “Image‐Guided and Functionally Instructed Tumor Therapies”Eberhard Karls University TuebingenRöntgenweg 11Tuebingen72076Germany
- Department for Medical Technologies and Regenerative MedicineInstitute of Biomedical EngineeringEberhard Karls University TübingenSilcherstr. 7/1Tübingen72076Germany
| | - Peter Loskill
- Department for Microphysiological Systems, Institute of Biomedical EngineeringEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
- NMI Natural and Medical Sciences Institute at the University of TübingenMarkwiesenstr. 55Reutlingen72770Germany
- 3R‐Center for In vitro Models and Alternatives to Animal TestingEberhard Karls University TübingenÖsterbergstr. 3Tübingen72074Germany
| |
Collapse
|
18
|
Preliminary report of de novo adipogenesis using novel bioabsorbable implants and image evaluation using a porcine model. J Artif Organs 2022; 25:245-253. [PMID: 35235081 PMCID: PMC9418277 DOI: 10.1007/s10047-022-01313-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2021] [Accepted: 01/17/2022] [Indexed: 11/24/2022]
Abstract
Our bioabsorbable poly-l-lactic acid (PLLA) mesh implants containing collagen sponge are replaced with adipose tissue after implantation, and this is an innovative method for breast reconstruction. In this preliminary study, we investigated the formation of adipose tissue and evaluated the process via multimodal images in a porcine model using an implant aggregate to generate the larger adipose tissue. The implant aggregate consists of PLLA mesh implants containing collagen sponge and a poly-glycolic acid woven bag covering them. We inserted the implant aggregates under the porcine mammary glands. Magnetic resonance imaging (MRI), ultrasonography (USG), and 3-dimensional (3D) surface imaging and histological evaluations were performed to evaluate the formation of adipose tissue over time. The volume of the implant aggregate and the formed adipose tissue inside the implant aggregate could be evaluated over time via MRI. The space within the implant aggregate was not confirmed on USG due to the acoustic shadow of the PLLA threads. The change in volume was not confirmed precisely using 3D surface imaging. Histologically, the newly formed adipose tissue was confirmed on the skin side of the implant aggregate. This implant aggregate has the ability to regenerate adipose tissue, and MRI is an appropriate method for the evaluation of the volume of the implant aggregation and the formation of adipose tissue.
Collapse
|
19
|
Vassallo V, Tsianaka A, Alessio N, Grübel J, Cammarota M, Tovar GEM, Southan A, Schiraldi C. Evaluation of novel biomaterials for cartilage regeneration based on gelatin methacryloyl interpenetrated with extractive chondroitin sulfate or unsulfated biotechnological chondroitin. J Biomed Mater Res A 2022; 110:1210-1223. [PMID: 35088923 PMCID: PMC9306773 DOI: 10.1002/jbm.a.37364] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 01/11/2022] [Accepted: 01/13/2022] [Indexed: 01/10/2023]
Abstract
Gelatin is widely proposed as scaffold for cartilage tissue regeneration due to its high similarities to the extracellular matrix. However, poor mechanical properties and high sensitivity to enzymatic degradation encouraged the scientific community to develop strategies to obtain better performing hydrogels. Gelatin networks, specifically gelatin‐methacryloyl (GM), have been coupled to hyaluronan or chondroitin sulfate (CS). In this study, we evaluated the biophysical properties of an innovative photocross‐linked hydrogel based on GM with the addition of CS or a new unsulfated biotechnological chondroitin (BC). Biophysical, mechanical, and biochemical characterization have been assessed to compare GM hydrogels to the chondroitin containing networks. Moreover, mesenchymal stem cells (MSCs) were seeded on these biomaterials in order to evaluate the differentiation toward the chondrocyte phenotype in 21 days. Rheological characterization showed that both CS and BC increased the stiffness (G' was about 2‐fold), providing a stronger rigid matrix, with respect to GM alone. The biological tests confirmed the onset of MSCs differentiation process starting from 14 days of in vitro culture. In particular, the combination GM + BC resulted to be more effective than GM + CS in the up‐regulation of key genes such as collagen type 2A1 (COLII), SOX‐9, and aggrecan). In addition, the scanning microscope analyses revealed the cellular adhesion on materials and production of extracellular vesicles. Immunofluorescence staining confirmed an increase of COLII in presence of both chondroitins. Finally, the outcomes suggest that BC entangled within cross‐linked GM matrix may represent a promising new biomaterial with potential applications in cartilage regeneration.
Collapse
Affiliation(s)
- Valentina Vassallo
- Department of Experimental Medicine, Section of Biotechnology, Medical Histology and Molecular Biology, University of Campania "Luigi Vanvitelli", Naples
| | - Anastasia Tsianaka
- Institute of Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Stuttgart, Germany
| | - Nicola Alessio
- Department of Experimental Medicine, Section of Biotechnology, Medical Histology and Molecular Biology, University of Campania "Luigi Vanvitelli", Naples
| | - Jana Grübel
- Institute of Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Stuttgart, Germany
| | - Marcella Cammarota
- Department of Experimental Medicine, Section of Biotechnology, Medical Histology and Molecular Biology, University of Campania "Luigi Vanvitelli", Naples
| | - Günter E M Tovar
- Institute of Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Stuttgart, Germany.,Fraunhofer Institute of Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
| | - Alexander Southan
- Institute of Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Stuttgart, Germany
| | - Chiara Schiraldi
- Department of Experimental Medicine, Section of Biotechnology, Medical Histology and Molecular Biology, University of Campania "Luigi Vanvitelli", Naples
| |
Collapse
|
20
|
Shahabipour F, Tavafoghi M, Aninwene GE, Bonakdar S, Oskuee RK, Shokrgozar MA, Potyondy T, Alambeigi F, Ahadian S. Coaxial 3D bioprinting of tri-polymer scaffolds to improve the osteogenic and vasculogenic potential of cells in co-culture models. J Biomed Mater Res A 2022; 110:1077-1089. [PMID: 35025130 DOI: 10.1002/jbm.a.37354] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2021] [Revised: 12/20/2021] [Accepted: 12/27/2021] [Indexed: 12/20/2022]
Abstract
The crosstalk between osteoblasts and endothelial cells is critical for bone vascularization and regeneration. Here, we used a coaxial 3D bioprinting method to directly print an osteon-like structure by depositing angiogenic and osteogenic bioinks from the core and shell regions of the coaxial nozzle, respectively. The bioinks were made up of gelatin, gelatin methacryloyl (GelMA), alginate, and hydroxyapatite (HAp) nanoparticles and were loaded with human umbilical vascular endothelial cells (HUVECs) and osteoblasts (MC3T3) in the core and shell regions, respectively. Conventional monoaxial 3D bioprinting was used as a control method, where the hydrogels, HAp nanoparticles, MC3T3 cells, and HUVECs were all mixed in one bioink and printed from the core nozzle. As a result, the bioprinted scaffolds were composed of cell-laden fibers with either a core-shell or homogenous structure, providing a non-contact (indirect) or contact (direct) co-culture of MC3T3 cells and HUVECs, respectively. Both structures supported the 3D culture of HUVECs and osteoblasts over a long period. The scaffolds also supported the expression of osteogenic and angiogenic factors. However, the gene expression was significantly higher for the core-shell structure than the homogeneous structure due to the well-defined distribution of osteoblasts and endothelial cells and the formation of vessel-like structures in the co-culture system. Our results indicated that the coaxial bioprinting technique, with the ability to create a non-contact co-culture of cells, can provide a more efficient bioprinting strategy for printing highly vascularized and bioactive bone structures.
Collapse
Affiliation(s)
- Fahimeh Shahabipour
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran.,National Cell Bank of Iran, Pasteur Institute of Iran, Tehran, Iran.,Harvard-MIT Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, Massachusetts, USA
| | - Maryam Tavafoghi
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, California, USA.,Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, California, USA
| | - George E Aninwene
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, California, USA.,Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, California, USA.,California NanoSystems Institute (CNSI), University of California-Los Angeles, Los Angeles, California, USA
| | - Shahin Bonakdar
- Skin Research Center, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Reza Kazemi Oskuee
- Biomedical Applied Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Tyler Potyondy
- Department of Bioengineering, Henry Samueli School of Engineering and Applied Sciences, University of California-Los Angeles, Los Angeles, California, USA.,Center for Minimally Invasive Therapeutics (C-MIT), University of California-Los Angeles, Los Angeles, California, USA
| | - Farshid Alambeigi
- Walker Department of Mechanical Engineering, University of Texas at Austin, Austin, Texas, USA
| | - Samad Ahadian
- Terasaki Institute for Biomedical Innovation, Los Angeles, California, USA
| |
Collapse
|
21
|
Louis F, Sowa Y, Kitano S, Matsusaki M. High-throughput drug screening models of mature adipose tissues which replicate the physiology of patients' Body Mass Index (BMI). Bioact Mater 2022; 7:227-241. [PMID: 34466729 PMCID: PMC8379425 DOI: 10.1016/j.bioactmat.2021.05.020] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Revised: 04/27/2021] [Accepted: 05/07/2021] [Indexed: 12/27/2022] Open
Abstract
Obesity is a complex and incompletely understood disease, but current drug screening strategies mostly rely on immature in vitro adipose models which cannot recapitulate it properly. To address this issue, we developed a statistically validated high-throughput screening model by seeding human mature adipocytes from patients, encapsulated in physiological collagen microfibers. These drop tissues ensured the maintenance of adipocyte viability and functionality for controlling glucose and fatty acids uptake, as well as glycerol release. As such, patients' BMI and insulin sensitivity displayed a strong inverse correlation: the healthy adipocytes were associated with the highest insulin-induced glucose uptake, while insulin resistance was confirmed in the underweight and severely obese adipocytes. Insulin sensitivity recovery was possible with two type 2 diabetes treatments, rosiglitazone and melatonin. Finally, the addition of blood vasculature to the model seemed to more accurately recapitulate the in vivo physiology, with particular respect to leptin secretion metabolism.
Collapse
Affiliation(s)
- Fiona Louis
- Osaka University, Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, 2-1 Yamadaoka, Suita Osaka, 565-0871, Japan
| | - Yoshihiro Sowa
- Kyoto Prefectural University of Medicine, Department of Plastic and Reconstructive Surgery, Graduate School of Medical Sciences, Kamigyo-ku Kajii-cho, Kawaramachi-Hirokoji, Kyoto, 602-8566, Japan
- Corresponding author. Kyoto, 602-8566, Kamigyo-ku Kajii-cho, Kawaramachi-Hirokoji, Japan.
| | - Shiro Kitano
- Osaka University, Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, 2-1 Yamadaoka, Suita Osaka, 565-0871, Japan
- TOPPAN PRINTING CO., LTD., Technical Research Institute, 4-2-3 Takanodaiminami, Sugito-machi, Saitama, 345-8508, Japan
| | - Michiya Matsusaki
- Osaka University, Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, 2-1 Yamadaoka, Suita Osaka, 565-0871, Japan
- Osaka University, Graduate School of Engineering, Department of Applied Chemistry, 2-1 Yamadaoka, Suita Osaka, 565-0871, Japan
- Corresponding author. Osaka, 565-0871, 2-1 Yamadaoka, Suita, Japan.
| |
Collapse
|
22
|
Ravichandran A, Meinert C, Bas O, Hutmacher DW, Bock N. Engineering a 3D bone marrow adipose composite tissue loading model suitable for studying mechanobiological questions. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 128:112313. [PMID: 34474864 DOI: 10.1016/j.msec.2021.112313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/22/2020] [Revised: 07/06/2021] [Accepted: 07/07/2021] [Indexed: 10/20/2022]
Abstract
Tissue engineering strategies are widely used to model and study the bone marrow microenvironment in healthy and pathological conditions. Yet, while bone function highly depends on mechanical stimulation, the effects of biomechanical stimuli on the bone marrow niche, specifically on bone marrow adipose tissue (BMAT) is poorly understood due to a lack of representative in vitro loading models. Here, we engineered a BMAT analog made of a GelMA (gelatin methacryloyl) hydrogel/medical-grade polycaprolactone (mPCL) scaffold composite to structurally and biologically mimic key aspects of the bone marrow microenvironment, and exploited an innovative bioreactor to study the effects of mechanical loading. Highly reproducible BMAT analogs facilitated the successful adipogenesis of human mesenchymal bone marrow stem cells. Upon long-term intermittent stimulation (1 Hz, 2 h/day, 3 days/week, 3 weeks) in the novel bioreactor, cellular proliferation and lipid accumulation were similar to unloaded controls, yet there was a significant reduction in the secretion of adipokines including leptin and adiponectin, in line with clinical evidence of reduced adipokine expression following exercise/activity. Ultimately, this innovative loading platform combined with reproducibly engineered BMAT analogs provide opportunities to study marrow physiology in greater complexity as it accounts for the dynamic mechanical microenvironment context.
Collapse
Affiliation(s)
- Akhilandeshwari Ravichandran
- Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove 4059, QLD, Australia; Translational Research Institute (TRI), QUT, Woolloongabba 4102, QLD, Australia
| | - Christoph Meinert
- Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove 4059, QLD, Australia; Metro North Hospital and Health Service, Herston 4029, QLD, Australia
| | - Onur Bas
- Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove 4059, QLD, Australia; Australian Research Council (ARC) Training Centre in Additive Biomanufacturing, QUT, Kelvin Grove 4059, QLD, Australia
| | - Dietmar W Hutmacher
- Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove 4059, QLD, Australia; Translational Research Institute (TRI), QUT, Woolloongabba 4102, QLD, Australia; Bone and Joint Disorders Program, School of Chemistry, Physics and Mechanical Engineering, Science and Engineering Faculty (SEF), QUT, Brisbane 4000, QLD, Australia; School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane 4000, QLD, Australia
| | - Nathalie Bock
- Centre in Regenerative Medicine, IHBI, QUT, Kelvin Grove 4059, QLD, Australia; Translational Research Institute (TRI), QUT, Woolloongabba 4102, QLD, Australia; School of Biomedical Sciences, Faculty of Health and Australian Prostate Cancer Research Centre (APCRC-Q), Institute of Health and Biomedical Innovation (IHBI), Queensland University of Technology (QUT), Brisbane 4000, QLD, Australia; ARC Industrial Transformation Training Centre for Multiscale 3D Imaging, Modelling and Manufacturing, QUT, Kelvin Grove 4059, QLD, Australia.
| |
Collapse
|
23
|
Chen J, Wang C, Wen W, Ni J, Jiang J, Ge Z, Wang M, Zhang F. Feasibility studies of intraocular use of gelatin methacryloyl hydrogel to patch retinal tears. POLYM ADVAN TECHNOL 2021. [DOI: 10.1002/pat.5328] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023]
Affiliation(s)
- Juan Chen
- Affiliated Hospital of Hangzhou Normal University Hangzhou China
| | - Chunyi Wang
- Affiliated Hospital of Hangzhou Normal University Hangzhou China
| | - Wen Wen
- Affiliated Hospital of Hangzhou Normal University Hangzhou China
| | - Jie Ni
- Affiliated Hospital of Hangzhou Normal University Hangzhou China
| | - Jingjie Jiang
- Affiliated Hospital of Hangzhou Normal University Hangzhou China
| | - Zhongjun Ge
- The Fourth People's Hospital of Lin'an Hangzhou China
| | - Mingwei Wang
- Affiliated Hospital of Hangzhou Normal University Hangzhou China
| | - Fang Zhang
- Affiliated Hospital of Hangzhou Normal University Hangzhou China
| |
Collapse
|
24
|
Chen H, Wang X, Wang J, Shi X, Li X, Wang J, Li D, Zhu Y, Tan W, Tan Z. In vitroadipogenesis and long-term adipocyte culture in adipose tissue-derived cell banks. Biofabrication 2021; 13. [PMID: 34044385 DOI: 10.1088/1758-5090/ac0610] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2020] [Accepted: 05/27/2021] [Indexed: 11/12/2022]
Abstract
There is a critical need to developin vitroculture systems appropriate for the expansion of adipose tissue, in order to gain new insights into metabolic diseases and to assist in the restoration of tissue defects. Conventional two- or three-dimensional (2D or 3D)in vitromodels of adipocytes require a combination of supplements to induce adipocyte maturation that greatly increases the cost of large-scale industrial production. In the present study, a microporous, perforated bacterial cellulose (BC)-assisted culture system was developed that promoted the adhesion, proliferation, and adipogenic differentiation of preadipocytes. Additionally, the system maintained the cells as mature unilocular adipocytesex vivoin normal cell culture medium in long-term culture. All cells were derived from isolated adipose tissue without the use of expensive enzymes for tissue digestion. In contrast to culture in hard tissue culture plates, preadipocytes in the soft 3D environments formed multidimensional interlaced cell contacts, undergoing significant spontaneous lipid accumulation and could be cultured for up to threemonths in maintenance medium. More importantly, the cultured adipose tissue-derived cell bank created here was able to produce injury repair activators that promoted the proliferation of fibroblasts with little fibrosis and the functional differentiation of myoblasts, displaying the potential for use in adipose reconstruction. Thus, the present study demonstrates the potential of a mechanically flexible BC scaffold to generate volume tunable adipose constructs and provides a low-cost and user-friendly strategy for large-scale industrial production of adipose tissue.
Collapse
Affiliation(s)
- Haoxiang Chen
- College of Biology, Hunan University, Changsha, Hunan 410082, People's Republic of China.,State Key Laboratory for Chemo/Biosensing and Chemometrics, Hunan University, Changsha, Hunan 410082, People's Republic of China
| | - Xiaocheng Wang
- College of Biology, Hunan University, Changsha, Hunan 410082, People's Republic of China
| | - Jian Wang
- College of Biology, Hunan University, Changsha, Hunan 410082, People's Republic of China.,State Key Laboratory for Chemo/Biosensing and Chemometrics, Hunan University, Changsha, Hunan 410082, People's Republic of China
| | - Xuelei Shi
- College of Biology, Hunan University, Changsha, Hunan 410082, People's Republic of China
| | - Xinghuan Li
- College of Biology, Hunan University, Changsha, Hunan 410082, People's Republic of China
| | - Jianlong Wang
- Department of Orthopedics, The Third Xiangya Hospital, Central South University, Changsha, Hunan 410013, People's Republic of China
| | - Dan Li
- College of Biology, Hunan University, Changsha, Hunan 410082, People's Republic of China
| | - Yonghua Zhu
- College of Biology, Hunan University, Changsha, Hunan 410082, People's Republic of China
| | - Weihong Tan
- College of Biology, Hunan University, Changsha, Hunan 410082, People's Republic of China
| | - Zhikai Tan
- College of Biology, Hunan University, Changsha, Hunan 410082, People's Republic of China.,State Key Laboratory for Chemo/Biosensing and Chemometrics, Hunan University, Changsha, Hunan 410082, People's Republic of China
| |
Collapse
|
25
|
Overcoming functional challenges in autologous and engineered fat grafting trends. Trends Biotechnol 2021; 40:77-92. [PMID: 34016480 DOI: 10.1016/j.tibtech.2021.04.006] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Revised: 04/06/2021] [Accepted: 04/06/2021] [Indexed: 12/11/2022]
Abstract
Autologous fat grafting offers significant promise for the repair of soft tissue deformities; however, high resorption rates indicate that engineered solutions are required to improve adipose tissue (AT) survival. Advances in material development and biofabrication have laid the foundation for the generation of functional AT constructs; however, a balance needs to be struck between clinically feasible delivery and improved structural integrity of the grafts. A new approach combining the objectives from both the clinical and research communities will assist in developing morphologically and genetically mature AT constructs, with controlled spatial arrangement and increased potential for neovascularization. In a rapidly progressing field, this review addresses research in both the preclinical and bioengineering domains and assesses their ability to resolve functional challenges.
Collapse
|
26
|
Van Damme L, Van Hoorick J, Blondeel P, Van Vlierberghe S. Toward Adipose Tissue Engineering Using Thiol-Norbornene Photo-Crosslinkable Gelatin Hydrogels. Biomacromolecules 2021; 22:2408-2418. [PMID: 33950675 DOI: 10.1021/acs.biomac.1c00189] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
Nowadays, breast implants, lipofilling, and microsurgical free tissue transfer are the most often applied procedures to repair soft tissue defects resulting from mastectomies/lumpectomies following breast cancer. Due to the drawbacks and limitations associated with these conventional clinical practices, there is a need for alternative reconstructive strategies. The development of biomimetic materials able to promote cell proliferation and adipogenic differentiation has gained increasing attention in the context of adipose reconstructive purposes. Herein, thiol-norbornene crosslinkable gelatin-based materials were developed and benchmarked to the current commonly applied methacryloyl-modified gelatin (GelMA) with different degrees of substitutions focussing on bottom-up tissue engineering. The developed hydrogels resulted in similar gel fractions, swelling, and in vitro biodegradation properties compared to the benchmark materials. Furthermore, the thiol-ene hydrogels exhibited mechanical properties closer to those of native fatty tissue compared to GelMA. The mechanical cues of the equimolar GelNB DS55% + GelSH DS75% composition resulted not only in similar biocompatibility but also, more importantly, in superior differentiation of the encapsulated cells into the adipogenic lineage, as compared to GelMA. It can be concluded that the photo-crosslinkable thiol-ene systems offer a promising strategy toward adipose tissue engineering through cell encapsulation compared to the benchmark GelMA.
Collapse
Affiliation(s)
- Lana Van Damme
- Polymer Chemistry & Biomaterials Group-Centre of Macromolecular Chemistry (CMaC)-Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281, S4-Bis, 9000 Ghent, Belgium.,Department of Plastic & Reconstructive Surgery, Ghent University Hospital, Corneel Heymanslaan 10, 2K12, 9000 Ghent, Belgium
| | - Jasper Van Hoorick
- Polymer Chemistry & Biomaterials Group-Centre of Macromolecular Chemistry (CMaC)-Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281, S4-Bis, 9000 Ghent, Belgium
| | - Philip Blondeel
- Department of Plastic & Reconstructive Surgery, Ghent University Hospital, Corneel Heymanslaan 10, 2K12, 9000 Ghent, Belgium
| | - Sandra Van Vlierberghe
- Polymer Chemistry & Biomaterials Group-Centre of Macromolecular Chemistry (CMaC)-Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281, S4-Bis, 9000 Ghent, Belgium
| |
Collapse
|
27
|
Ortega MA, Rodríguez-Comas J, Yavas O, Velasco-Mallorquí F, Balaguer-Trias J, Parra V, Novials A, Servitja JM, Quidant R, Ramón-Azcón J. In Situ LSPR Sensing of Secreted Insulin in Organ-on-Chip. BIOSENSORS-BASEL 2021; 11:bios11050138. [PMID: 33924867 PMCID: PMC8144989 DOI: 10.3390/bios11050138] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Revised: 04/17/2021] [Accepted: 04/25/2021] [Indexed: 01/10/2023]
Abstract
Organ-on-a-chip (OOC) devices offer new approaches for metabolic disease modeling and drug discovery by providing biologically relevant models of tissues and organs in vitro with a high degree of control over experimental variables for high-content screening applications. Yet, to fully exploit the potential of these platforms, there is a need to interface them with integrated non-labeled sensing modules, capable of monitoring, in situ, their biochemical response to external stimuli, such as stress or drugs. In order to meet this need, we aim here to develop an integrated technology based on coupling a localized surface plasmon resonance (LSPR) sensing module to an OOC device to monitor the insulin in situ secretion in pancreatic islets, a key physiological event that is usually perturbed in metabolic diseases such as type 2 diabetes (T2D). As a proof of concept, we developed a biomimetic islet-on-a-chip (IOC) device composed of mouse pancreatic islets hosted in a cellulose-based scaffold as a novel approach. The IOC was interfaced with a state-of-the-art on-chip LSPR sensing platform to monitor the in situ insulin secretion. The developed platform offers a powerful tool to enable the in situ response study of microtissues to external stimuli for applications such as a drug-screening platform for human models, bypassing animal testing.
Collapse
Affiliation(s)
- María A. Ortega
- Biosensors for Bioengineering Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri I Reixac, 10-12, 08028 Barcelona, Spain; (M.A.O.); (J.R.-C.); (F.V.-M.); (J.B.-T.); (V.P.)
| | - Júlia Rodríguez-Comas
- Biosensors for Bioengineering Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri I Reixac, 10-12, 08028 Barcelona, Spain; (M.A.O.); (J.R.-C.); (F.V.-M.); (J.B.-T.); (V.P.)
| | - Ozlem Yavas
- Plasmon Nano-Optics Group, ICFO-Institute for Photonics Sciences, The Barcelona Institute of Science and Technology, 08860 Barcelona, Spain; (O.Y.); (R.Q.)
| | - Ferran Velasco-Mallorquí
- Biosensors for Bioengineering Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri I Reixac, 10-12, 08028 Barcelona, Spain; (M.A.O.); (J.R.-C.); (F.V.-M.); (J.B.-T.); (V.P.)
| | - Jordina Balaguer-Trias
- Biosensors for Bioengineering Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri I Reixac, 10-12, 08028 Barcelona, Spain; (M.A.O.); (J.R.-C.); (F.V.-M.); (J.B.-T.); (V.P.)
| | - Victor Parra
- Biosensors for Bioengineering Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri I Reixac, 10-12, 08028 Barcelona, Spain; (M.A.O.); (J.R.-C.); (F.V.-M.); (J.B.-T.); (V.P.)
| | - Anna Novials
- Diabetes and Obesity Research Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (A.N.); (J.M.S.)
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), 28029 Madrid, Spain
| | - Joan M. Servitja
- Diabetes and Obesity Research Laboratory, Institut d’Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), 08036 Barcelona, Spain; (A.N.); (J.M.S.)
- Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas (CIBERDEM), 28029 Madrid, Spain
| | - Romain Quidant
- Plasmon Nano-Optics Group, ICFO-Institute for Photonics Sciences, The Barcelona Institute of Science and Technology, 08860 Barcelona, Spain; (O.Y.); (R.Q.)
- Nanophotonic Systems Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, 8092 Zurich, Switzerland
- ICREA-Institució Catalana de Recerca i Estudis Avançats, 08010 Barcelona, Spain
| | - Javier Ramón-Azcón
- Biosensors for Bioengineering Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Baldiri I Reixac, 10-12, 08028 Barcelona, Spain; (M.A.O.); (J.R.-C.); (F.V.-M.); (J.B.-T.); (V.P.)
- ICREA-Institució Catalana de Recerca i Estudis Avançats, 08010 Barcelona, Spain
- Correspondence:
| |
Collapse
|
28
|
Abdollahi Baghban S, Ebrahimi M, Bagheri-Khoulenjani S, Khorasani M. A highly efficient microwave-assisted synthesis of an LED-curable methacrylated gelatin for bio applications. RSC Adv 2021; 11:14996-15009. [PMID: 35424032 PMCID: PMC8697925 DOI: 10.1039/d1ra01269j] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2021] [Accepted: 04/10/2021] [Indexed: 12/14/2022] Open
Abstract
This study deals with the development of an LED-curable methacrylated gelatin (GelMA) synthesis via microwave (MW) irradiation with a reaction and purification time-, energy-, and methacrylation reagent-saving approach. To investigate the efficiency of MW irradiation in GelMA synthesis, characteristics of the GelMAs prepared by using glycidyl methacrylate (GMA) or methacrylic anhydride (MA) via the MW-assisted (MWA) method were compared comprehensively with those synthesized via the conventional heating method. Moreover, MWA reaction conditions were optimized in terms of methacrylation reagent concentrations (C), reaction time (t), and MW power (P). Characterization and assessment of the GelMAs were conducted with 1H NMR, FT-IR, and Raman spectroscopy along with physical-mechanical, thermal, and hydrophilicity analysis. The results demonstrated that the MWA synthesized GMA-GelMA hydrogels were possessed of increased methacrylation degree (MD), gel fraction (GF), tensile strength (TS), elongation at break (EB), glass transition temperature (T g), and water contact angle (WCA) as well as decreased swelling degree (SD) values in comparison to those of MA-GelMA and GMA-GelMA hydrogels prepared via the MWA and conventional method, respectively. Enhanced properties of the MWA synthesized GMA-hydrogels suggested an effective methacryloyl conjugation leading to a greater amount of covalent crosslinking density justified by the dipolar moment calculations. Optimal GMA C, t, P, and purification time for a highly crosslinked GelMA hydrogel (MD: 96.1%, GF: 98.3%, SD: 10.11%, TS: 6.7 MPa, EB: 175.2%, T g: 75.34 °C, and WCA: 72.22°) were found to be a 5 times molar excess over the primary amine groups of gelatin, 5 min, 500 W, and 24 h, respectively. Thus, the optimized MW conditions offer a promising green method to prepare GelMAs for bio applications.
Collapse
Affiliation(s)
- Sahar Abdollahi Baghban
- Department of Polymer and Color Engineering, Amirkabir University of Technology 350 Hafez Ave. 15875-4413 Tehran Iran
| | - Morteza Ebrahimi
- Department of Polymer and Color Engineering, Amirkabir University of Technology 350 Hafez Ave. 15875-4413 Tehran Iran
| | - Shadab Bagheri-Khoulenjani
- Department of Polymer and Color Engineering, Amirkabir University of Technology 350 Hafez Ave. 15875-4413 Tehran Iran
| | - Manoucher Khorasani
- Department of Polymer and Color Engineering, Amirkabir University of Technology 350 Hafez Ave. 15875-4413 Tehran Iran
| |
Collapse
|
29
|
The Inflammatory Profile of Obesity and the Role on Pulmonary Bacterial and Viral Infections. Int J Mol Sci 2021; 22:ijms22073456. [PMID: 33810619 PMCID: PMC8037155 DOI: 10.3390/ijms22073456] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2021] [Revised: 03/19/2021] [Accepted: 03/23/2021] [Indexed: 12/16/2022] Open
Abstract
Obesity is a globally increasing health problem, entailing diverse comorbidities such as infectious diseases. An obese weight status has marked effects on lung function that can be attributed to mechanical dysfunctions. Moreover, the alterations of adipocyte-derived signal mediators strongly influence the regulation of inflammation, resulting in chronic low-grade inflammation. Our review summarizes the known effects regarding pulmonary bacterial and viral infections. For this, we discuss model systems that allow mechanistic investigation of the interplay between obesity and lung infections. Overall, obesity gives rise to a higher susceptibility to infectious pathogens, but the pathogenetic process is not clearly defined. Whereas, viral infections often show a more severe course in obese patients, the same patients seem to have a survival benefit during bacterial infections. In particular, we summarize the main mechanical impairments in the pulmonary tract caused by obesity. Moreover, we outline the main secretory changes within the expanded adipose tissue mass, resulting in chronic low-grade inflammation. Finally, we connect these altered host factors to the influence of obesity on the development of lung infection by summarizing observations from clinical and experimental data.
Collapse
|
30
|
Louis F, Piantino M, Liu H, Kang DH, Sowa Y, Kitano S, Matsusaki M. Bioprinted Vascularized Mature Adipose Tissue with Collagen Microfibers for Soft Tissue Regeneration. CYBORG AND BIONIC SYSTEMS 2021; 2021:1412542. [PMID: 36285131 PMCID: PMC9494725 DOI: 10.34133/2021/1412542] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2020] [Accepted: 02/06/2021] [Indexed: 12/02/2022] Open
Abstract
The development of soft tissue regeneration has recently gained importance due to safety concerns about artificial breast implants. Current autologous fat graft implantations can result in up to 90% of volume loss in long-term outcomes due to their limited revascularization. Adipose tissue has a highly vascularized structure which enables its proper homeostasis as well as its endocrine function. Mature adipocytes surrounded by a dense vascular network are the specific features required for efficient regeneration of the adipose tissue to perform host anastomosis after its implantation. Recently, bioprinting has been introduced as a promising solution to recreate in vitro this architecture in large-scale tissues. However, the in vitro induction of both the angiogenesis and adipogenesis differentiations from stem cells yields limited maturation states for these two pathways. To overcome these issues, we report a novel method for obtaining a fully vascularized adipose tissue reconstruction using supporting bath bioprinting. For the first time, directly isolated mature adipocytes encapsulated in a bioink containing physiological collagen microfibers (CMF) were bioprinted in a gellan gum supporting bath. These multilayered bioprinted tissues retained high viability even after 7 days of culture. Moreover, the functionality was also confirmed by the maintenance of fatty acid uptake from mature adipocytes. Therefore, this method of constructing fully functional adipose tissue regeneration holds promise for future clinical applications.
Collapse
Affiliation(s)
- Fiona Louis
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Japan
| | - Marie Piantino
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Japan
| | - Hao Liu
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Japan
| | - Dong-Hee Kang
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Japan
| | - Yoshihiro Sowa
- Department of Plastic and Reconstructive Surgery, Graduate School of Medical Sciences, Kyoto Prefectural University of Medicine, Japan
| | - Shiro Kitano
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Japan
- Toppan Printing Co., Ltd., Tokyo, Japan
| | - Michiya Matsusaki
- Joint Research Laboratory (TOPPAN) for Advanced Cell Regulatory Chemistry, Graduate School of Engineering, Osaka University, Japan
- Department of Applied Chemistry, Graduate School of Engineering, Osaka University, Japan
| |
Collapse
|
31
|
Olăreț E, Drăgușin DM, Serafim A, Lungu A, Șelaru A, Dobranici A, Dinescu S, Costache M, Boerașu I, Vasile BȘ, Steinmüller-Nethl D, Iovu H, Stancu IC. Electrospinning Fabrication and Cytocompatibility Investigation of Nanodiamond Particles-Gelatin Fibrous Tubular Scaffolds for Nerve Regeneration. Polymers (Basel) 2021; 13:polym13030407. [PMID: 33514051 PMCID: PMC7865256 DOI: 10.3390/polym13030407] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/24/2021] [Accepted: 01/25/2021] [Indexed: 12/17/2022] Open
Abstract
This paper reports the electrospinning fabrication of flexible nanostructured tubular scaffolds, based on fish gelatin (FG) and nanodiamond nanoparticles (NDs), and their cytocompatibility with murine neural stem cells. The effects of both nanofiller and protein concentration on the scaffold morphology, aqueous affinity, size modification at rehydration, and degradation are assessed. Our findings indicate that nanostructuring with low amounts of NDs may modify the fiber properties, including a certain regional parallel orientation of fiber segments. NE-4C cells form dense clusters that strongly adhere to the surface of FG50-based scaffolds, while also increasing FG concentration and adding NDs favor cellular infiltration into the flexible fibrous FG70_NDs nanocomposite. This research illustrates the potential of nanostructured NDs-FG fibers as scaffolds for nerve repair and regeneration. We also emphasize the importance of further understanding the effect of the nanofiller-protein interphase on the microstructure and properties of electrospun fibers and on cell-interactivity.
Collapse
Affiliation(s)
- Elena Olăreț
- Advanced Polymer Materials Group, University Politehnica of Bucharest, 011061 Bucharest, Romania; (E.O.); (D.-M.D.); (A.S.); (A.L.); (H.I.)
| | - Diana-Maria Drăgușin
- Advanced Polymer Materials Group, University Politehnica of Bucharest, 011061 Bucharest, Romania; (E.O.); (D.-M.D.); (A.S.); (A.L.); (H.I.)
| | - Andrada Serafim
- Advanced Polymer Materials Group, University Politehnica of Bucharest, 011061 Bucharest, Romania; (E.O.); (D.-M.D.); (A.S.); (A.L.); (H.I.)
| | - Adriana Lungu
- Advanced Polymer Materials Group, University Politehnica of Bucharest, 011061 Bucharest, Romania; (E.O.); (D.-M.D.); (A.S.); (A.L.); (H.I.)
| | - Aida Șelaru
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania; (A.Ș.); (A.D.); (S.D.); (M.C.)
- Department of Immunology, National Institute for Research and Development in Biomedical Pathology and Biomedical Sciences “Victor Babes”, 050096 Bucharest, Romania
| | - Alexandra Dobranici
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania; (A.Ș.); (A.D.); (S.D.); (M.C.)
| | - Sorina Dinescu
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania; (A.Ș.); (A.D.); (S.D.); (M.C.)
- The Research Institute of the University of Bucharest, 050663 Bucharest, Romania
| | - Marieta Costache
- Department of Biochemistry and Molecular Biology, Faculty of Biology, University of Bucharest, 050095 Bucharest, Romania; (A.Ș.); (A.D.); (S.D.); (M.C.)
- The Research Institute of the University of Bucharest, 050663 Bucharest, Romania
| | - Iulian Boerașu
- National Research Center for Micro and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 060042 Bucharest, Romania; (I.B.); (B.Ș.V.)
| | - Bogdan Ștefan Vasile
- National Research Center for Micro and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 060042 Bucharest, Romania; (I.B.); (B.Ș.V.)
- National Research Center for Food Safety, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 060042 Bucharest, Romania
| | | | - Horia Iovu
- Advanced Polymer Materials Group, University Politehnica of Bucharest, 011061 Bucharest, Romania; (E.O.); (D.-M.D.); (A.S.); (A.L.); (H.I.)
| | - Izabela-Cristina Stancu
- Advanced Polymer Materials Group, University Politehnica of Bucharest, 011061 Bucharest, Romania; (E.O.); (D.-M.D.); (A.S.); (A.L.); (H.I.)
- Correspondence:
| |
Collapse
|
32
|
McCarthy M, Brown T, Alarcon A, Williams C, Wu X, Abbott RD, Gimble J, Frazier T. Fat-On-A-Chip Models for Research and Discovery in Obesity and Its Metabolic Comorbidities. TISSUE ENGINEERING. PART B, REVIEWS 2020; 26:586-595. [PMID: 32216545 PMCID: PMC8196547 DOI: 10.1089/ten.teb.2019.0261] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/24/2019] [Accepted: 03/12/2020] [Indexed: 12/11/2022]
Abstract
The obesity epidemic and its associated comorbidities present a looming challenge to health care delivery throughout the world. Obesity is characterized as a sterile inflammatory process within adipose tissues leading to dysregulated secretion of bioactive adipokines such as adiponectin and leptin, as well as systemic metabolic dysfunction. The majority of current obesity research has focused primarily on preclinical animal models in vivo and two-dimensional cell culture models in vitro. Neither of these generalized approaches is optimal due to interspecies variability, insufficient accuracy with respect to predicting human outcomes, and failure to recapitulate the three-dimensional (3D) microenvironment. Consequently, there is a growing demand and need for more sophisticated microphysiological systems to reproduce more physiologically accurate human white and brown/beige adipose depots. To address this research need, human and murine cell lines and primary cultures are being combined with bioscaffolds to create functional 3D environments that are suitable for metabolically active adipose organoids in both static and perfusion bioreactor cultures. The development of these technologies will have considerable impact on the future pace of discovery for novel small molecules and biologics designed to prevent and treat metabolic syndrome and obesity in humans. Furthermore, when these adipose tissue models are integrated with other organ systems they will have applicability to obesity-related disorders such as diabetes, nonalcoholic fatty liver disease, and osteoarthritis. Impact statement The current review article summarizes the advances made within the organ-onchip field, as it pertains to adipose tissue models of obesity and obesity-related syndromes, such as diabetes, non-alcoholic fatty liver disease, and osteoarthritis. As humanized 3D adipose-derived constructs become more accessible to the research community, it is anticipated that they will accelerate and enhance the drug discovery pipeline for obesity, diabetes, and metabolic diseases by reducing the preclinical evaluation process and improving predictive accuracy. Such developments, applications, and usages of existing technologies can change the paradigm of personalized medicine and create substantial progress in our approach to modern medicine.
Collapse
Affiliation(s)
| | - Theodore Brown
- Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Andrea Alarcon
- LaCell LLC, New Orleans, Louisiana, USA
- Obatala Sciences, Inc., New Orleans, Louisiana, USA
| | | | - Xiying Wu
- LaCell LLC, New Orleans, Louisiana, USA
- Obatala Sciences, Inc., New Orleans, Louisiana, USA
| | - Rosalyn D. Abbott
- Materials Science and Engineering Department, Carnegie Mellon University, Pittsburgh, Pennsylvania, USA
| | - Jeffrey Gimble
- LaCell LLC, New Orleans, Louisiana, USA
- Obatala Sciences, Inc., New Orleans, Louisiana, USA
| | - Trivia Frazier
- LaCell LLC, New Orleans, Louisiana, USA
- Obatala Sciences, Inc., New Orleans, Louisiana, USA
| |
Collapse
|
33
|
Photocrosslinkable nanocomposite ink for printing strong, biodegradable and bioactive bone graft. Biomaterials 2020; 263:120378. [PMID: 32932140 DOI: 10.1016/j.biomaterials.2020.120378] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 08/25/2020] [Accepted: 09/07/2020] [Indexed: 01/29/2023]
Abstract
3D printing is known as a cost-effective technique that shows huge potential in fabrication of graft substitutes for bone tissue regeneration. However, the tradeoff between 3D printability, mechanical strength and bioactivity of the printed materials (i.e., inks) remains a challenge. In this work, we present a novel photocrosslinkable nanocomposite ink composed of tri-block poly (lactide-co-propylene glycol-co-lactide) dimethacrylate (PmLnDMA, m and n respectively represent the unit length of propylene glycol and lactide) and hydroxyethyl methacrylate (HEMA)-functionalized hydroxyapatite nanoparticles (nHAMA). The reactive HEMA-conjugated nHAMA, is designed to covalently crosslink with the surrounding polymer matrix to further increase the interfacial bonding between them. We find that the nHAMA can rapidly interact with PmLnDMA upon light exposure within 140 s and form an inorganic-organic co-crosslinked nanocomposite network, further enhancing the nanofiller-matrix interfacial compatibility. Notably, our nanocomposites possess significantly improved mechanical performances compared to the polymer, with compressive modulus increasing by nearly 10 times (from ⁓40 to ⁓400 MPa). Moreover, thanks to the low exothermic heat generation (<37 °C) during photocrosslinking, our nanocomposite ink enables facile encapsulation and long-term release of heat-labile biomolecules like bone morphogenic protein-2 (BMP-2). Furthermore, it demonstrates a readily tunable rheological property, wettability, degradation, and printability as a 3D bone scaffold. Together with its superior osteogenic ability both in vitro and in vivo, we envision that our nanocomposite ink holds great promise in 3D printing of bone grafts.
Collapse
|
34
|
Abdul-Al M, Zaernia A, Sefat F. Biomaterials for breast reconstruction: Promises, advances, and challenges. J Tissue Eng Regen Med 2020; 14:1549-1569. [PMID: 32841503 DOI: 10.1002/term.3121] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 07/31/2020] [Accepted: 08/05/2020] [Indexed: 12/23/2022]
Abstract
Breast reconstruction is the opportunity that provides the chance of having breast after undergoing surgical removal of the breast tissue due to cancer-related surgery. However, this varies on the stage of the cancer diagnosis and the procedure undertaken. There are many regenerative medicine methods that provide several initiatives and direct solutions to problems such as the development of "bioactive tissue," which can regenerate adipose tissues with similar normal functions and structures. There have been several studies which have previously explored for the improvement of breast reconstruction including different variations of biomaterials, different fabrication and processing techniques, cells as well as growth factors which enable bioengineers and tissue engineers to reconstruct a suitable breast for patients with breast cancer. Many factors such as shape, proper volume, mechanical properties have been studies but very scattered with not adequate solution for existing patients worldwide. This review article aims to cover recent advances in the biomaterials, which can be used for reconstruction of breasts as well as looking at the various factors that might lead to individuals needing reconstruction and the materials that are available. The focus would be to look at the various biomaterials that are available to use for reconstruction, their properties, and their structural integrity.
Collapse
Affiliation(s)
- Mohamed Abdul-Al
- Department of Biomedical and Electronics Engineering, School of Engineering, University of Bradford, Bradford, UK
| | - Amir Zaernia
- Department of Biomedical and Electronics Engineering, School of Engineering, University of Bradford, Bradford, UK
| | - Farshid Sefat
- Department of Biomedical and Electronics Engineering, School of Engineering, University of Bradford, Bradford, UK.,Interdisciplinary Research Centre in Polymer Science & Technology (Polymer IRC), University of Bradford, Bradford, UK
| |
Collapse
|
35
|
Nellinger S, Schmidt I, Heine S, Volz A, Kluger PJ. Adipose stem cell‐derived extracellular matrix represents a promising biomaterial by inducing spontaneous formation of prevascular‐like structures by mvECs. Biotechnol Bioeng 2020; 117:3160-3172. [DOI: 10.1002/bit.27481] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Revised: 05/24/2020] [Accepted: 06/24/2020] [Indexed: 12/31/2022]
Affiliation(s)
- Svenja Nellinger
- Reutlingen Research Institute Reutlingen University Reutlingen Germany
| | - Isabelle Schmidt
- School of Applied Chemistry Reutlingen University Reutlingen Germany
| | - Simon Heine
- Reutlingen Research Institute Reutlingen University Reutlingen Germany
| | - Ann‐Cathrin Volz
- Reutlingen Research Institute Reutlingen University Reutlingen Germany
| | - Petra J. Kluger
- School of Applied Chemistry Reutlingen University Reutlingen Germany
| |
Collapse
|
36
|
Schmidt FF, Nowakowski S, Kluger PJ. Improvement of a Three-Layered in vitro Skin Model for Topical Application of Irritating Substances. Front Bioeng Biotechnol 2020; 8:388. [PMID: 32457884 PMCID: PMC7225271 DOI: 10.3389/fbioe.2020.00388] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2020] [Accepted: 04/07/2020] [Indexed: 12/23/2022] Open
Abstract
In the field of skin tissue engineering, the development of physiologically relevant in vitro skin models comprising all skin layers, namely epidermis, dermis, and subcutis, is a great challenge. Increasing regulatory requirements and the ban on animal experiments for substance testing demand the development of reliable and in vivo-like test systems, which enable high-throughput screening of substances. However, the reproducibility and applicability of in vitro testing has so far been insufficient due to fibroblast-mediated contraction. To overcome this pitfall, an advanced 3-layered skin model was developed. While the epidermis of standard skin models showed an 80% contraction, the initial epidermal area of our advanced skin models was maintained. The improved barrier function of the advanced models was quantified by an indirect barrier function test and a permeability assay. Histochemical and immunofluorescence staining of the advanced model showed well-defined epidermal layers, a dermal part with distributed human dermal fibroblasts and a subcutis with round-shaped adipocytes. The successful response of these advanced 3-layered models for skin irritation testing demonstrated the suitability as an in vitro model for these clinical tests: only the advanced model classified irritative and non-irritative substances correctly. These results indicate that the advanced set up of the 3-layered in vitro skin model maintains skin barrier function and therefore makes them more suitable for irritation testing.
Collapse
Affiliation(s)
- Freia F Schmidt
- Reutlingen Research Institute, Reutlingen University, Reutlingen, Germany
| | - Sophia Nowakowski
- Reutlingen Research Institute, Reutlingen University, Reutlingen, Germany
| | - Petra J Kluger
- Reutlingen Research Institute, Reutlingen University, Reutlingen, Germany
| |
Collapse
|
37
|
Liu P, Li Q, Yang Q, Zhang S, Lin C, Zhang G, Tang Z. Three-dimensional cell printing of gingival fibroblast/acellular dermal matrix/gelatin-sodium alginate scaffolds and their biocompatibility evaluation in vitro. RSC Adv 2020; 10:15926-15935. [PMID: 35493638 PMCID: PMC9052548 DOI: 10.1039/d0ra02082f] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Accepted: 04/15/2020] [Indexed: 11/21/2022] Open
Abstract
Tissue engineering has emerged as a promising approach for soft tissue regeneration. Three-dimensional (3D) cell printing showed great potential for producing cell-encapsulated scaffolds to repair tissue defects. The advantage of 3D cell printing technology is precise cell loading in scaffolds to achieve tissue regeneration instead of only relying on the cells from surrounding tissue or blood. A new acellular dermal matrix/gelatin-sodium alginate (ADM/A/G) scaffold with living gingival fibroblasts was constructed by 3D cell printing technology for potential oral soft tissue regeneration in this study, and the biological characteristics of the 3D cell printing scaffolds were evaluated. The residue of nucleic acid and growth factors in ADM were detected. Three biomaterials were mixed at an appropriate radio with human gingival fibroblasts (hGFs) to prepare bioinks. Two kinds of layer scaffolds were fabricated by 3D cell printing technology. The mechanical strength and degradability of the scaffolds were determined by measuring their compressive modulus and mass loss. CCK-8 assay and calcein-AM/PI staining were conducted to detect the cell proliferation and viability in 3D cell printing scaffolds. The morphology of the hGFs in the scaffolds were observed using SEM and FITC-phalloidin staining. The expression of COL1A1, PECAM1, and VEGF-A of hGFs in the scaffolds were quantified by qRT-PCR. The gelatin-sodium alginate (A/G) scaffolds were used as control group in all experiments. Compared with the control group, 3D cell printing ADM/A/G scaffolds showed better mechanical strength and longer degradation time. The ADM/A/G scaffolds obviously had a better promotion effect on cell proliferation and viability. Most of the hGFs observed had a fully extended spindle morphology in the ADM/A/G scaffolds but oval morphology in the control group. The expression of COL1A1 was significantly higher than in the control group with time, and the expression of PECAM1 and VEGF-A was slightly higher in ADM/A/G scaffolds on day 14. 3D cell printing gingival fibroblast-ADM/A/G scaffolds showed excellent biological properties, which could be potentially useful in oral soft tissue regeneration.
Collapse
Affiliation(s)
- Peng Liu
- Second Clinical Division, Peking University School and Hospital of Stomatology Beijing 100101 P. R. China
- National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing 100081 P. R. China
| | - Qing Li
- Second Clinical Division, Peking University School and Hospital of Stomatology Beijing 100101 P. R. China
- Center of Digital Dentistry, Peking University School and Hospital of Stomatology Beijing 100081 P. R. China
- National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing 100081 P. R. China
| | - Qiaolin Yang
- Department of Orthodontics, Peking University School and Hospital of Stomatology Beijing 100081 P. R. China
| | - Shihan Zhang
- Second Clinical Division, Peking University School and Hospital of Stomatology Beijing 100101 P. R. China
| | - Chunping Lin
- Second Clinical Division, Peking University School and Hospital of Stomatology Beijing 100101 P. R. China
| | - Guifeng Zhang
- State Key Laboratories of Biochemical Engineering, Institute of Process Engineering Beijing 100190 P. R. China
| | - Zhihui Tang
- Second Clinical Division, Peking University School and Hospital of Stomatology Beijing 100101 P. R. China
- National Engineering Laboratory for Digital and Material Technology of Stomatology Beijing 100081 P. R. China
| |
Collapse
|
38
|
Ogino S, Sakamoto M, Lee S, Yamanaka H, Tsuge I, Arata J, Sakamoto Y, Kambe Y, Yamaoka T, Morimoto N. De novo adipogenesis using a bioabsorbable implant without additional cells or growth factors. J Tissue Eng Regen Med 2020; 14:920-930. [PMID: 32293793 DOI: 10.1002/term.3041] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Revised: 03/26/2020] [Accepted: 03/31/2020] [Indexed: 11/09/2022]
Abstract
Adipose tissue regeneration in breast cancer patients without additional growth factors or adipose-tissue-derived stromal cells is desirable because of the possibility of recurrence and metastasis. We report that a poly-L-lactic acid (PLLA) mesh implant containing a collagen sponge (CS) maintained the internal space in vivo for up to 12 months and substituted for adipose tissue. We developed a PLLA capsule that maintained the internal space longer than that of PLLA mesh and compared adipose tissue formation at 12 and 24 months after implantation between the PLLA mesh with CS implant and the PLLA capsule implant with or without CS in a rabbit model. After 12 months, all implants maintained the internal space, and the adipose tissue that formed in all implant groups was larger than that in the control group. At 24 months, PLLA mesh maintained the internal space just as well as that at 12 months, while the PLLA capsule collapsed and accumulated a large number of macrophages. The formed adipose tissue in the PLLA mesh group was maintained up to 24 months; however, those in two PLLA capsule groups decreased and showed no difference from the control group. In conclusion, the internal space of the PLLA mesh implant with CS was substituted for adipose tissue at 12 months and sustained the formed adipose tissue after 24 months. The PLLA mesh implant containing CS is a desirable bioabsorbable implant that can be replaced by autologous adipose tissue after implantation in vivo without using any growth factors or cells.
Collapse
Affiliation(s)
- Shuichi Ogino
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, Sakyou-ku, Kyoto, Japan
| | - Michiharu Sakamoto
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, Sakyou-ku, Kyoto, Japan
| | - Sunghee Lee
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, Sakyou-ku, Kyoto, Japan
| | - Hiroki Yamanaka
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, Sakyou-ku, Kyoto, Japan
| | - Itaru Tsuge
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, Sakyou-ku, Kyoto, Japan
| | - Jun Arata
- Department of Plastic and Reconstructive Surgery, National Hospital Organization Kyoto Medical Center, Fushimiku, Kyoto, Japan
| | - Yuki Sakamoto
- Gunze QOL Research Center Laboratory, Ayabe, Kyoto, Japan
| | - Yusuke Kambe
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Tetsuji Yamaoka
- Department of Biomedical Engineering, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan
| | - Naoki Morimoto
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, Kyoto University, Sakyou-ku, Kyoto, Japan
| |
Collapse
|
39
|
Leucht A, Volz AC, Rogal J, Borchers K, Kluger PJ. Advanced gelatin-based vascularization bioinks for extrusion-based bioprinting of vascularized bone equivalents. Sci Rep 2020; 10:5330. [PMID: 32210309 PMCID: PMC7093518 DOI: 10.1038/s41598-020-62166-w] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 02/27/2020] [Indexed: 12/19/2022] Open
Abstract
Bone tissue is highly vascularized. The crosstalk of vascular and osteogenic cells is not only responsible for the formation of the strongly divergent tissue types but also for their physiological maintenance and repair. Extrusion-based bioprinting presents a promising fabrication method for bone replacement. It allows for the production of large-volume constructs, which can be tailored to individual tissue defect geometries. In this study, we used the all-gelatin-based toolbox of methacryl-modified gelatin (GM), non-modified gelatin (G) and acetylated GM (GMA) to tailor both the properties of the bioink towards improved printability, and the properties of the crosslinked hydrogel towards enhanced support of vascular network formation by simple blending. The vasculogenic behavior of human dermal microvascular endothelial cells (HDMECs) and human adipose-derived stem cells (ASCs) was evaluated in the different hydrogel formulations for 14 days. Co-culture constructs including a vascular component and an osteogenic component (i.e. a bone bioink based on GM, hydroxyapatite and ASCs) were fabricated via extrusion-based bioprinting. Bioprinted co-culture constructs exhibited functional tissue-specific cells whose interplay positively affected the formation and maintenance of vascular-like structures. The setup further enabled the deposition of bone matrix associated proteins like collagen type I, fibronectin and alkaline phosphatase within the 30-day culture.
Collapse
Affiliation(s)
- A Leucht
- Institute of Interfacial Process Engineering and Plasmatechnology IGVP, University of Stuttgart, Stuttgart, Germany
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
| | - A-C Volz
- Reutlingen Research Institute, Reutlingen University, Reutlingen, Germany
| | - J Rogal
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
- RWTH Aachen University, Aachen, Germany
| | - K Borchers
- Institute of Interfacial Process Engineering and Plasmatechnology IGVP, University of Stuttgart, Stuttgart, Germany
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany
| | - P J Kluger
- Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Stuttgart, Germany.
- Reutlingen Research Institute, Reutlingen University, Reutlingen, Germany.
| |
Collapse
|
40
|
Colle J, Blondeel P, De Bruyne A, Bochar S, Tytgat L, Vercruysse C, Van Vlierberghe S, Dubruel P, Declercq H. Bioprinting predifferentiated adipose-derived mesenchymal stem cell spheroids with methacrylated gelatin ink for adipose tissue engineering. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2020; 31:36. [PMID: 32206922 DOI: 10.1007/s10856-020-06374-w] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Accepted: 03/03/2020] [Indexed: 06/10/2023]
Abstract
The increasing number of mastectomies results in a greater demand for breast reconstruction characterized by simplicity and a low complication profile. Reconstructive surgeons are investigating tissue engineering (TE) strategies to overcome the current surgical drawbacks. 3D bioprinting is the rising technique for the fabrication of large tissue constructs which provides a potential solution for unmet clinical needs in breast reconstruction building on decades of experience in autologous fat grafting, adipose-derived mesenchymal stem cell (ASC) biology and TE. A scaffold was bioprinted using encapsulated ASC spheroids in methacrylated gelatin ink (GelMA). Uniform ASC spheroids with an ideal geometry and diameter for bioprinting were formed, using a high-throughput non-adhesive agarose microwell system. ASC spheroids in adipogenic differentiation medium (ADM) were evaluated through live/dead staining, histology (HE, Oil Red O), TEM and RT-qPCR. Viable spheroids were obtained for up to 14 days post-printing and showed multilocular microvacuoles and successful differentiation toward mature adipocytes shown by gene expression analysis. Moreover, spheroids were able to assemble at random in GelMA, creating a macrotissue. Combining the advantage of microtissues to self-assemble and the controlled organization by bioprinting technologies, these ASC spheroids can be useful as building blocks for the engineering of soft tissue implants.
Collapse
Affiliation(s)
- Julien Colle
- Tissue Engineering Group, Bioprint Facility-Department of Human Structure and Repair-Faculty of Medicine and Health Sciences, Gent University, Corneel Heymanslaan 10 (6B3), 9000, Gent, Belgium.
- Department of Plastic Surgery, University Hospital Gent, Gent, Belgium.
| | - Phillip Blondeel
- Department of Plastic Surgery, University Hospital Gent, Gent, Belgium
| | - Axelle De Bruyne
- Tissue Engineering Group, Bioprint Facility-Department of Human Structure and Repair-Faculty of Medicine and Health Sciences, Gent University, Corneel Heymanslaan 10 (6B3), 9000, Gent, Belgium
| | - Silke Bochar
- Tissue Engineering Group, Bioprint Facility-Department of Human Structure and Repair-Faculty of Medicine and Health Sciences, Gent University, Corneel Heymanslaan 10 (6B3), 9000, Gent, Belgium
| | - Liesbeth Tytgat
- Polymer Chemistry and Biomaterials Group, Department of Organic Chemistry, Faculty of Sciences, Gent University, Krijgslaan 281, 9000, Gent, Belgium
| | - Chris Vercruysse
- Tissue Engineering Group, Bioprint Facility-Department of Human Structure and Repair-Faculty of Medicine and Health Sciences, Gent University, Corneel Heymanslaan 10 (6B3), 9000, Gent, Belgium
| | - Sandra Van Vlierberghe
- Polymer Chemistry and Biomaterials Group, Department of Organic Chemistry, Faculty of Sciences, Gent University, Krijgslaan 281, 9000, Gent, Belgium
| | - Peter Dubruel
- Polymer Chemistry and Biomaterials Group, Department of Organic Chemistry, Faculty of Sciences, Gent University, Krijgslaan 281, 9000, Gent, Belgium
| | - Heidi Declercq
- Tissue Engineering Group, Bioprint Facility-Department of Human Structure and Repair-Faculty of Medicine and Health Sciences, Gent University, Corneel Heymanslaan 10 (6B3), 9000, Gent, Belgium
| |
Collapse
|
41
|
Tytgat L, Kollert MR, Van Damme L, Thienpont H, Ottevaere H, Duda GN, Geissler S, Dubruel P, Van Vlierberghe S, Qazi TH. Evaluation of 3D Printed Gelatin-Based Scaffolds with Varying Pore Size for MSC-Based Adipose Tissue Engineering. Macromol Biosci 2020; 20:e1900364. [PMID: 32077631 DOI: 10.1002/mabi.201900364] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2019] [Revised: 12/19/2019] [Indexed: 12/31/2022]
Abstract
Adipose tissue engineering aims to provide solutions to patients who require tissue reconstruction following mastectomies or other soft tissue trauma. Mesenchymal stromal cells (MSCs) robustly differentiate into the adipogenic lineage and are attractive candidates for adipose tissue engineering. This work investigates whether pore size modulates adipogenic differentiation of MSCs toward identifying optimal scaffold pore size and whether pore size modulates spatial infiltration of adipogenically differentiated cells. To assess this, extrusion-based 3D printing is used to fabricate photo-crosslinkable gelatin-based scaffolds with pore sizes in the range of 200-600 µm. The adipogenic differentiation of MSCs seeded onto these scaffolds is evaluated and robust lipid droplet formation is observed across all scaffold groups as early as after day 6 of culture. Expression of adipogenic genes on scaffolds increases significantly over time, compared to TCP controls. Furthermore, it is found that the spatial distribution of cells is dependent on the scaffold pore size, with larger pores leading to a more uniform spatial distribution of adipogenically differentiated cells. Overall, these data provide first insights into the role of scaffold pore size on MSC-based adipogenic differentiation and contribute toward the rational design of biomaterials for adipose tissue engineering in 3D volumetric spaces.
Collapse
Affiliation(s)
- Liesbeth Tytgat
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry (CMaC), Ghent University, Krijgslaan 281, S4-Bis, 9000, Ghent, Belgium.,Brussels Photonics, Vrije Universiteit Brussel, Pleinlaan 2, 1050, Brussels, Belgium
| | - Matthias R Kollert
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Berlin-Brandenburg Center and School for Regenerative Therapies, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Lana Van Damme
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry (CMaC), Ghent University, Krijgslaan 281, S4-Bis, 9000, Ghent, Belgium
| | - Hugo Thienpont
- Brussels Photonics, Vrije Universiteit Brussel, Pleinlaan 2, 1050, Brussels, Belgium
| | - Heidi Ottevaere
- Brussels Photonics, Vrije Universiteit Brussel, Pleinlaan 2, 1050, Brussels, Belgium
| | - Georg N Duda
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Berlin-Brandenburg Center and School for Regenerative Therapies, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Sven Geissler
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Berlin-Brandenburg Center and School for Regenerative Therapies, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany
| | - Peter Dubruel
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry (CMaC), Ghent University, Krijgslaan 281, S4-Bis, 9000, Ghent, Belgium
| | - Sandra Van Vlierberghe
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry (CMaC), Ghent University, Krijgslaan 281, S4-Bis, 9000, Ghent, Belgium.,Brussels Photonics, Vrije Universiteit Brussel, Pleinlaan 2, 1050, Brussels, Belgium
| | - Taimoor H Qazi
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Berlin-Brandenburg Center and School for Regenerative Therapies, Charité Universitätsmedizin Berlin, Augustenburger Platz 1, 13353, Berlin, Germany.,Department of Bioengineering, University of Pennsylvania, 210 South 33rd Street, Philadelphia, 19104, USA
| |
Collapse
|
42
|
Žigon-Branc S, Markovic M, Van Hoorick J, Van Vlierberghe S, Dubruel P, Zerobin E, Baudis S, Ovsianikov A. Impact of Hydrogel Stiffness on Differentiation of Human Adipose-Derived Stem Cell Microspheroids. Tissue Eng Part A 2019; 25:1369-1380. [PMID: 30632465 PMCID: PMC6784494 DOI: 10.1089/ten.tea.2018.0237] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 01/07/2019] [Indexed: 12/27/2022] Open
Abstract
Hydrogels represent an attractive material platform for realization of three-dimensional (3D) tissue-engineered constructs, as they have tunable mechanical properties, are compatible with different types of cells, and resemble elements found in natural extracellular matrices. So far, numerous hydrogel-cartilage/bone tissue engineering (TE)-related studies were performed by utilizing a single cell encapsulation approach. Although multicellular spheroid cultures exhibit advantageous properties for cartilage or bone TE, the chondrogenic or osteogenic differentiation potential of stem cell microspheroids within hydrogels has not been investigated much. This study explores, for the first time, how stiffness of gelatin-based hydrogels (having a storage modulus of 538, 3584, or 7263 Pa) affects proliferation and differentiation of microspheroids formed from telomerase-immortalized human adipose-derived stem cells (hASC/hTERT). Confocal microscopy indicates that all tested hydrogels supported cell viability during their 3- to 5-week culture period in the control, chondrogenic, or osteogenic medium. Although in the softer hydrogels cells from neighboring microspheroids started outgrowing and interconnecting within a few days, their protrusion was slower or limited in stiffer hydrogels or those cultured in chondrogenic medium, respectively. High expressions of chondrogenic markers (SOX9, ACAN, COL2A1), detected in all tested hydrogels, proved that the chondrogenic differentiation of hASC/hTERT microspheroids was very successful, especially in the two softer hydrogels, where superior cartilage-specific properties were confirmed by Alcian blue staining. These chondrogenically induced samples also expressed COL10A1, a marker of chondrocyte hypertrophy. Interestingly, the hydrogel itself (with no differentiation medium) showed a slight chondrogenic induction. Regardless of the hydrogel stiffness, in the samples stimulated with osteogenic medium, the expression of selected markers RUNX2, BGLAP, ALPL, and COL1A1 was not conclusive. Nevertheless, the von Kossa staining confirmed the presence of calcium deposits in osteogenically stimulated samples in the two softer hydrogels, suggesting that these also favor osteogenesis. This observation was also confirmed by Alizarin red quantification assay, with which higher amounts of calcium were detected in the osteogenically induced hydrogels than in their controls. The presented data indicate that the encapsulation of adipose-derived stem cell microspheroids in gelatin-based hydrogels show promising potential for future applications in cartilage or bone TE. Impact Statement Osteochondral defects represent one of the leading causes of disability in the world. Although numerous tissue engineering (TE) approaches have shown success in cartilage and bone tissue regeneration, achieving native-like characteristics of these tissues remains challenging. This study demonstrates that in the presence of a corresponding differentiation medium, gelatin-based hydrogels support moderate osteogenic and excellent chondrogenic differentiation of photo-encapsulated human adipose-derived stem cell microspheroids, the extent of which depends on hydrogel stiffness. Because photosensitive hydrogels are a convenient material platform for creating stiffness gradients in three dimensions, the presented microspheroid-hydrogel encapsulation strategy holds promise for future strategies of cartilage or bone TE.
Collapse
Affiliation(s)
- Sara Žigon-Branc
- Institute of Materials Science and Technology, Technische Universität Wien (TU Wien), Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Austria
| | - Marica Markovic
- Institute of Materials Science and Technology, Technische Universität Wien (TU Wien), Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Austria
| | - Jasper Van Hoorick
- Department of Organic and Macromolecular Chemistry, Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium
- Brussels Photonics, Department of Applied Physics and Photonics, Vrije Universiteit Brussel and Flanders Make, Elsene, Belgium
| | - Sandra Van Vlierberghe
- Department of Organic and Macromolecular Chemistry, Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium
- Brussels Photonics, Department of Applied Physics and Photonics, Vrije Universiteit Brussel and Flanders Make, Elsene, Belgium
| | - Peter Dubruel
- Department of Organic and Macromolecular Chemistry, Polymer Chemistry and Biomaterials Group, Centre of Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Elise Zerobin
- Austrian Cluster for Tissue Regeneration, Austria
- Division of Macromolecular Chemistry, Institute of Applied Synthetic Chemistry, Technische Universität Wien (TU Wien), Vienna, Austria
| | - Stefan Baudis
- Austrian Cluster for Tissue Regeneration, Austria
- Division of Macromolecular Chemistry, Institute of Applied Synthetic Chemistry, Technische Universität Wien (TU Wien), Vienna, Austria
| | - Aleksandr Ovsianikov
- Institute of Materials Science and Technology, Technische Universität Wien (TU Wien), Vienna, Austria
- Austrian Cluster for Tissue Regeneration, Austria
| |
Collapse
|
43
|
Şelaru A, Drăgușin DM, Olăreț E, Serafim A, Steinmüller-Nethl D, Vasile E, Iovu H, Stancu IC, Costache M, Dinescu S. Fabrication and Biocompatibility Evaluation of Nanodiamonds-Gelatin Electrospun Materials Designed for Prospective Tissue Regeneration Applications. MATERIALS (BASEL, SWITZERLAND) 2019; 12:E2933. [PMID: 31514289 PMCID: PMC6766245 DOI: 10.3390/ma12182933] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/05/2019] [Revised: 09/04/2019] [Accepted: 09/09/2019] [Indexed: 01/12/2023]
Abstract
Due to the reduced ability of most harmed tissues to self-regenerate, new strategies are being developed in order to promote self-repair assisted or not by biomaterials, among these tissue engineering (TE). Human adipose-derived mesenchymal stem cells (hASCs) currently represent a promising tool for tissue reconstruction, due to their low immunogenicity, high differentiation potential to multiple cell types and easy harvesting. Gelatin is a natural biocompatible polymer used for regenerative applications, while nanodiamond particles (NDs) are used as reinforcing nanomaterial that might modulate cell behavior, namely cell adhesion, viability, and proliferation. The development of electrospun microfibers loaded with NDs is expected to allow nanomechanical sensing due to local modifications of both nanostructure and stiffness. Two aqueous suspensions with 0.5 and 1% w/v NDs in gelatin from cold water fish skin (FG) were used to generate electrospun meshes. Advanced morpho- and micro-structural characterization revealed homogeneous microfibers. Nanoindentation tests confirmed the reinforcing effect of NDs. Biocompatibility assays showed an increased viability and proliferation profile of hASCs in contact with FG_NDs, correlated with very low cytotoxic effects of the materials. Moreover, hASCs developed an elongated cytoskeleton, suggesting that NDs addition to FG materials encouraged cell adhesion. This study showed the FG_NDs fibrous scaffolds potential for advanced TE applications.
Collapse
Affiliation(s)
- Aida Şelaru
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania.
| | - Diana-Maria Drăgușin
- Advanced Polymer Materials Group, University Politehnica of Bucharest, 011061 Bucharest, Romania.
| | - Elena Olăreț
- Advanced Polymer Materials Group, University Politehnica of Bucharest, 011061 Bucharest, Romania.
| | - Andrada Serafim
- Advanced Polymer Materials Group, University Politehnica of Bucharest, 011061 Bucharest, Romania.
| | | | - Eugeniu Vasile
- Department of Science and Engineering of Oxide Materials and Nanomaterials, University Politehnica of Bucharest, 011061 Bucharest, Romania.
| | - Horia Iovu
- Advanced Polymer Materials Group, University Politehnica of Bucharest, 011061 Bucharest, Romania.
| | - Izabela-Cristina Stancu
- Advanced Polymer Materials Group, University Politehnica of Bucharest, 011061 Bucharest, Romania.
| | - Marieta Costache
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania.
- Research Institute of University of Bucharest, 050107 Bucharest, Romania.
| | - Sorina Dinescu
- Department of Biochemistry and Molecular Biology, University of Bucharest, 050095 Bucharest, Romania.
- Research Institute of University of Bucharest, 050107 Bucharest, Romania.
| |
Collapse
|
44
|
Volz AC, Omengo B, Gehrke S, Kluger PJ. Comparing the use of differentiated adipose-derived stem cells and mature adipocytes to model adipose tissue in vitro. Differentiation 2019; 110:19-28. [PMID: 31568881 DOI: 10.1016/j.diff.2019.09.002] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2019] [Revised: 08/19/2019] [Accepted: 09/03/2019] [Indexed: 12/14/2022]
Abstract
In vitro models of human adipose tissue may serve as beneficial alternatives to animal models to study basic biological processes, identify new drug targets, and as soft tissue implants. With this approach, we aimed to evaluate adipose-derived stem cells (ASC) and mature adipocytes (MA) comparatively for the application in the in vitro setup of adipose tissue constructs to imitate native adipose tissue physiology. We used human primary MAs and human ASCs, differentiated for 14 days, and encapsulated them in collagen type I hydrogels to build up a three-dimensional (3D) adipose tissue model. The maintenance of the models was analyzed after seven days based on a viability staining. Further, the expression of the adipocyte specific protein perilipin A and the release of leptin and glycerol were evaluated. Gene transcription profiles of models based on dASCs and MAs were analyzed with regard to native adipose tissue. Compared to MAs, dASCs showed an immature differentiation state. Further, gene transcription of MAs suggests a behavior closer to native tissue in terms of angiogenesis, which supports MAs as preferred cell type. In contrast to native adipose tissue, genes of de novo lipogenesis and tissue remodeling were upregulated in the in vitro attempts.
Collapse
Affiliation(s)
- Ann-Cathrin Volz
- Reutlingen Research Institute, Reutlingen University, Alteburgstrasse 150, 72762, Reutlingen, Germany; University of Hohenheim, Schloss Hohenheim 1, 70599, Stuttgart, Germany
| | - Birgit Omengo
- Institute of Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Nobelstrasse 12, 70569, Stuttgart, Germany
| | - Sandra Gehrke
- Research & Development, Research Special Skincare, Beiersdorf AG, Unnastrasse 48, 20253, Hamburg, Germany
| | - Petra Juliane Kluger
- Reutlingen Research Institute, Reutlingen University, Alteburgstrasse 150, 72762, Reutlingen, Germany; Fraunhofer Institute for Interfacial Engineering and Biotechnology IGB, Cell and Tissue Engineering, Nobelstrasse 12, 70569, Stuttgart, Germany.
| |
Collapse
|
45
|
Tytgat L, Van Damme L, Van Hoorick J, Declercq H, Thienpont H, Ottevaere H, Blondeel P, Dubruel P, Van Vlierberghe S. Additive manufacturing of photo-crosslinked gelatin scaffolds for adipose tissue engineering. Acta Biomater 2019; 94:340-350. [PMID: 31136829 DOI: 10.1016/j.actbio.2019.05.062] [Citation(s) in RCA: 64] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2019] [Revised: 05/20/2019] [Accepted: 05/23/2019] [Indexed: 01/22/2023]
Abstract
There exists a clear clinical need for adipose tissue reconstruction strategies to repair soft tissue defects which outperform the currently available approaches. In this respect, additive manufacturing has shown to be a promising alternative for the development of larger constructs able to support adipose tissue engineering. In the present work, a thiol-ene photo-click crosslinkable gelatin hydrogel was developed which allowed extrusion-based additive manufacturing into porous scaffolds. To this end, norbornene-functionalized gelatin (Gel-NB) was combined with thiolated gelatin (Gel-SH). The application of a macromolecular gelatin-based thiolated crosslinker holds several advantages over conventional crosslinkers including cell-interactivity, less chance at phase separation between scaffold material and crosslinker and the formation of a more homogeneous network. Throughout the paper, these photo-click scaffolds were benchmarked to the conventional methacrylamide-modified gelatin (Gel-MA). The results indicated that stable scaffolds could be realized which were further characterized physico-chemically by performing swelling, mechanical and in vitro biodegradability assays. Furthermore, the seeded adipose tissue-derived stem cells (ASCs) remained viable (>90%) up to 14 days and were able to proliferate. In addition, the cells could be differentiated into the adipogenic lineage on the photo-click crosslinked scaffolds, thereby performing better than the cells supported by the frequently reported Gel-MA scaffolds. As a result, the developed photo-click crosslinked scaffolds can be considered a promising candidate towards adipose tissue engineering and a valuable alternative for the omnipresent Gel-MA. STATEMENT OF SIGNIFICANCE: The field of adipose tissue engineering has emerged as a promising strategy to repair soft tissue defects. Herein, Gel-NB/Gel-SH gelatin-based hydrogel scaffolds were produced using extrusion-based additive manufacturing. Using a cell-interactive, thiolated gelatin crosslinker, a homogeneous network was formed and the risk of phase separation between norbornene-modified gelatin and macromolecular crosslinkers was reduced. UV-induced crosslinking of these materials is based on step growth polymerization which requires less free radicals to enable polymerization. Our results demonstrated the potential of the developed scaffolds, due to their favourable physico-chemical characteristics as well as their adipogenic differentiation potential when benchmarked to Gel-MA scaffolds. Hence, the hydrogels could be of great interest towards future development of adipose tissue constructs and tissue engineering in general.
Collapse
Affiliation(s)
- Liesbeth Tytgat
- Brussels Photonics (B-PHOT) - Department of Applied Physics and Photonics, Vrije Universiteit Brussel and Flanders Make, Pleinlaan 2, 1050 Brussels, Belgium; Polymer Chemistry & Biomaterials Group - Centre of Macromolecular Chemistry (CMaC) - Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281, S4-Bis, 9000 Ghent, Belgium
| | - Lana Van Damme
- Polymer Chemistry & Biomaterials Group - Centre of Macromolecular Chemistry (CMaC) - Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281, S4-Bis, 9000 Ghent, Belgium; Department of Plastic & Reconstructive Surgery, Ghent University Hospital, Corneel Heymanslaan 10, 2K12, 9000 Ghent, Belgium
| | - Jasper Van Hoorick
- Brussels Photonics (B-PHOT) - Department of Applied Physics and Photonics, Vrije Universiteit Brussel and Flanders Make, Pleinlaan 2, 1050 Brussels, Belgium; Polymer Chemistry & Biomaterials Group - Centre of Macromolecular Chemistry (CMaC) - Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281, S4-Bis, 9000 Ghent, Belgium
| | - Heidi Declercq
- Tissue Engineering and Biomaterials Group - Department of Human Structure and Repair, Ghent University, Corneel Heymanslaan 10, 6B3, 9000 Ghent, Belgium
| | - Hugo Thienpont
- Brussels Photonics (B-PHOT) - Department of Applied Physics and Photonics, Vrije Universiteit Brussel and Flanders Make, Pleinlaan 2, 1050 Brussels, Belgium
| | - Heidi Ottevaere
- Brussels Photonics (B-PHOT) - Department of Applied Physics and Photonics, Vrije Universiteit Brussel and Flanders Make, Pleinlaan 2, 1050 Brussels, Belgium
| | - Phillip Blondeel
- Department of Plastic & Reconstructive Surgery, Ghent University Hospital, Corneel Heymanslaan 10, 2K12, 9000 Ghent, Belgium
| | - Peter Dubruel
- Polymer Chemistry & Biomaterials Group - Centre of Macromolecular Chemistry (CMaC) - Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281, S4-Bis, 9000 Ghent, Belgium
| | - Sandra Van Vlierberghe
- Brussels Photonics (B-PHOT) - Department of Applied Physics and Photonics, Vrije Universiteit Brussel and Flanders Make, Pleinlaan 2, 1050 Brussels, Belgium; Polymer Chemistry & Biomaterials Group - Centre of Macromolecular Chemistry (CMaC) - Department of Organic and Macromolecular Chemistry, Ghent University, Krijgslaan 281, S4-Bis, 9000 Ghent, Belgium
| |
Collapse
|
46
|
Expanding the Range of Available Isoelectric Points of Highly Methacryloylated Gelatin. MACROMOL CHEM PHYS 2019. [DOI: 10.1002/macp.201900097] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
47
|
Yu H, Zhang X, Song W, Pan T, Wang H, Ning T, Wei Q, Xu HH, Wu B, Ma D. Effects of 3-dimensional Bioprinting Alginate/Gelatin Hydrogel Scaffold Extract on Proliferation and Differentiation of Human Dental Pulp Stem Cells. J Endod 2019; 45:706-715. [DOI: 10.1016/j.joen.2019.03.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2018] [Revised: 03/06/2019] [Accepted: 03/12/2019] [Indexed: 12/28/2022]
|
48
|
Hauptmann N, Lian Q, Ludolph J, Rothe H, Hildebrand G, Liefeith K. Biomimetic Designer Scaffolds Made of D,L-Lactide- ɛ-Caprolactone Polymers by 2-Photon Polymerization. TISSUE ENGINEERING. PART B, REVIEWS 2019; 25:167-186. [PMID: 30632460 PMCID: PMC6589497 DOI: 10.1089/ten.teb.2018.0284] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/28/2018] [Accepted: 01/01/2019] [Indexed: 11/21/2022]
Abstract
IMPACT STATEMENT In tissue engineering (TE), the establishment of cell targeting materials, which mimic the conditions of the physiological extracellular matrix (ECM), seems to be a mission impossible without advanced materials and fabrication techniques. With this in mind we established a toolbox based on (D,L)-lactide-ɛ-caprolactone methacrylate (LCM) copolymers in combination with a nano-micromaskless lithography technique, the two-photon polymerization (2-PP) to mimic the hierarchical structured and complex milieu of the natural ECM. To demonstrate the versatility of this toolbox, we choose two completely different application scenarios in bone and tumor TE to show the high potential of this concept in therapeutic and diagnostic application.
Collapse
Affiliation(s)
- Nicole Hauptmann
- Department of Biomaterials, Institute for Bioprocessing and Analytical Measurement Techniques e.V. (iba), Rosenhof, Heilbad Heiligenstadt, Germany
| | - Qilin Lian
- Department of Biomaterials, Institute for Bioprocessing and Analytical Measurement Techniques e.V. (iba), Rosenhof, Heilbad Heiligenstadt, Germany
| | - Johanna Ludolph
- Department of Biomaterials, Institute for Bioprocessing and Analytical Measurement Techniques e.V. (iba), Rosenhof, Heilbad Heiligenstadt, Germany
| | - Holger Rothe
- Department of Biomaterials, Institute for Bioprocessing and Analytical Measurement Techniques e.V. (iba), Rosenhof, Heilbad Heiligenstadt, Germany
| | - Gerhard Hildebrand
- Department of Biomaterials, Institute for Bioprocessing and Analytical Measurement Techniques e.V. (iba), Rosenhof, Heilbad Heiligenstadt, Germany
| | - Klaus Liefeith
- Department of Biomaterials, Institute for Bioprocessing and Analytical Measurement Techniques e.V. (iba), Rosenhof, Heilbad Heiligenstadt, Germany
| |
Collapse
|
49
|
Contessi Negrini N, Bonnetier M, Giatsidis G, Orgill DP, Farè S, Marelli B. Tissue-mimicking gelatin scaffolds by alginate sacrificial templates for adipose tissue engineering. Acta Biomater 2019; 87:61-75. [PMID: 30654214 DOI: 10.1016/j.actbio.2019.01.018] [Citation(s) in RCA: 61] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2018] [Revised: 01/02/2019] [Accepted: 01/11/2019] [Indexed: 12/15/2022]
Abstract
When adipose tissue (AT) is impaired by trauma or disease, AT engineering could provide a shelf-ready structural and functional restoration as alternative to current clinical treatments, which mainly aim at aesthetic replacement. Yet, the lack of an efficient vascular network within the scaffolds represents a major limitation to their translation application in patients. Here, we propose the use of microstructured crosslinked gelatin hydrogels with an embedded prevascular channel as scaffolding materials for AT engineering. The scaffolds are fabricated using - simultaneously - alginate-based microbeads and 3D printed filaments as sacrificial material encapsulated in gelatin at the point of material fabrication and removed post-crosslinking. This method yields the formation of microstructures that resemble the micro-architecture of physiological human fat tissue and of microvessels that can facilitate vascularization through anastomosis with patients' own blood vessels. The cytocompatible method used to prepare the gelatin scaffolds showed structural stability over time while allowing for cell infiltration and protease-based remodeling/degradation. Scaffolds' mechanical properties were also designed to mimic the one of natural breast adipose tissue, a key parameter for AT regeneration. Scaffold's embedded channel (∅ = 300-400 µm) allowed for cell infiltration and enabled blood flow in vitro when an anastomosis with a rat blood artery was performed using surgical glue. In vitro tests with human mesenchymal stem cells (hMSC) showed colonization of the porous structure of the gelatin hydrogels, differentiation into adipocytes and accumulation of lipid droplets, as shown by Oil Red O staining. STATEMENT OF SIGNIFICANCE: The potential clinical use of scaffolds for adipose tissue (AT) regeneration is currently limited by an unmet simultaneous achievement of adequate structural/morphological properties together with a promoted scaffold vascularization. Sacrificial materials, currently used either to obtain a tissue-mimicking structure or hollow channels to promote scaffold' vascularization, are powerful versatile tools for the fabrication of scaffolds with desired features. However, an integrated approach by means of sacrificial templates aiming at simultaneously achieving an adequate AT-mimicking structure and hollow channels for vascularization is missing. Here, we prove the suitability of crosslinked gelatin scaffolds obtained by using sacrificial alginate microbeads and 3D printed strands to achieve proper features and hollow channels useful for scaffolds vascularization.
Collapse
Affiliation(s)
- Nicola Contessi Negrini
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA, United States; Department of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milan, Italy; INSTM, National Interuniversity Consortium of Materials Science and Technology, Local Unit Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milan, Italy
| | - Mathilde Bonnetier
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA, United States
| | - Giorgio Giatsidis
- Division of Plastic Surgery, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, United States
| | - Dennis P Orgill
- Division of Plastic Surgery, Brigham and Women's Hospital, 75 Francis Street, Boston, MA, United States
| | - Silvia Farè
- Department of Chemistry, Materials and Chemical Engineering "G. Natta", Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milan, Italy; INSTM, National Interuniversity Consortium of Materials Science and Technology, Local Unit Politecnico di Milano, Piazza Leonardo da Vinci 32, 20133 Milan, Italy.
| | - Benedetto Marelli
- Department of Civil and Environmental Engineering, Massachusetts Institute of Technology, 77 Massachusetts Ave, Cambridge, MA, United States.
| |
Collapse
|
50
|
Stier S, Rebers L, Schönhaar V, Hoch E, Borchers K. Advanced formulation of methacryl- and acetyl-modified biomolecules to achieve independent control of swelling and stiffness in printable hydrogels. JOURNAL OF MATERIALS SCIENCE. MATERIALS IN MEDICINE 2019; 30:35. [PMID: 30840139 DOI: 10.1007/s10856-019-6231-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 02/03/2019] [Indexed: 06/09/2023]
Abstract
Biobased hydrogels are considered to mimic native extracellular matrix due to their high water content and are considered as adequate matrices for cell encapsulation. However, the equilibrium degree of swelling (EDS) and stiffness of simple hydrogel formulations are typically confined: Increasing polymer concentration results in increasing stiffness and simultaneously decreasing EDS. The aim of this contribution was to decouple this standard correlation between polymer content, stiffness and EDS as well as the assembly of hydrogels with graded composition of hydrogels by layer-wise printing. We investigated two sets of formulations, which consisted of three different compositions with increasing total biopolymer concentration (10.6%, 11.5%, 13.0%). Within these compositions the amount of gelatin methacryloyl acetyl (GMA) was constant (10%), whereas the proportion of methacrylated hyaluronic acid and chondroitin sulfate increased. In the first set of formulations GMA with one fixed degree of methacryloylation (DM) was used, whereby the storage modulus (G') increased from ~10 to ~25 kPa and the EDS decreased from ~700 to ~600%. In the second set of formulations we gradually lowered the DM of the GMA in parallel to increase of polymer concentration and achieved an increase of both, G' from ~11 to ~18 kPa and EDS from ~690 to ~790%. By dispensing these compositions, we created a glycosaminoglycan-graded hydrogel. We proved the cytocompatibility of the dispensing process, the used photoinitiator lithium phenyl-2,4,6-trimethylbenzoylphosphinate, and layer-wise UVA irradiation. Glycosaminoglycan gradient was proved stable for 28 d,encapsulated chondrocytes were viable and produced new matrix.
Collapse
Affiliation(s)
- Sandra Stier
- Fraunhofer-Institute for Interfacial Engineering and Biotechnology IGB, Nobelstraße 12, 70569, Stuttgart, Germany
| | - Lisa Rebers
- Institute of Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Nobelstraße 12, 70569, Stuttgart, Germany
| | - Veronika Schönhaar
- Fraunhofer-Institute for Interfacial Engineering and Biotechnology IGB, Nobelstraße 12, 70569, Stuttgart, Germany
| | - Eva Hoch
- Institute of Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Nobelstraße 12, 70569, Stuttgart, Germany
| | - Kirsten Borchers
- Fraunhofer-Institute for Interfacial Engineering and Biotechnology IGB, Nobelstraße 12, 70569, Stuttgart, Germany.
- Institute of Interfacial Process Engineering and Plasma Technology IGVP, University of Stuttgart, Nobelstraße 12, 70569, Stuttgart, Germany.
| |
Collapse
|