1
|
Sun Q, Zhang Y, Hu B, Feng Q, Xia Y, Yu L, Zhang C, Liu W, Liu Z, Yao H, Lang Y. Development of a dual-responsive injectable GelMA/F127DA hydrogel for enhanced cartilage regeneration in osteoarthritis: Harnessing MMP-triggered and mechanical stress-induced release of therapeutic agents. Int J Biol Macromol 2025; 304:140823. [PMID: 39924046 DOI: 10.1016/j.ijbiomac.2025.140823] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 02/05/2025] [Accepted: 02/07/2025] [Indexed: 02/11/2025]
Abstract
Osteoarthritis (OA) presents a significant challenge in clinical settings due to the limited self-renewal capability of cartilage tissue. To address this, engineered biomaterials employing biomimetic strategies have been developed to modulate and enhance cell-microenvironment interactions, facilitating cartilage regeneration. Nonetheless, excessive mechanical stress on joint structures can induce inflammatory responses, thereby impeding the process of cartilage repair. In this study, we focus on the OA microenvironment, characterized by the overexpression of matrix metalloproteinases (MMPs), and the mechanical stimuli due to joint movement. We engineered a dual-responsive injectable hydrogel: a blend of MMP-responsive, thermo-sensitive GelMA and mechanically robust, reverse thermo-sensitive F127DA. This hydrogel was designed to deliver TGF-β and KGN in a controlled manner via simple temperature modulation. The hydrophilic properties of GelMA and the hydrophobic nature of F127DA allow for efficient intra-articular delivery of diverse drug types, optimizing their therapeutic effects. Photocrosslinking the hydrogel in situ effectively seals cartilage defects and prevents further degradation. The overexpressed MMP in the OA environment triggers the release of TGF-β, recruiting bone marrow-derived stem cells (BMSCs), while mechanical pressure from joint movements releases KGN, promoting chondrogenic differentiation and mitigating inflammation. In summary, our injectable hydrogel, responsive to both the OA microenvironment and mechanical stress, shows promise in enhancing cartilage regeneration in OA. This approach holds significant potential for advancing the field of OA cartilage tissue engineering.
Collapse
Affiliation(s)
- Qi Sun
- Department of Orthopedics, Hangzhou Fuyang Hospital of Orthopedics of Traditional Chinese Medicine, Hangzhou 311499, China
| | - Yuanbin Zhang
- Department of Orthopedics, Hangzhou Fuyang Hospital of Orthopedics of Traditional Chinese Medicine, Hangzhou 311499, China
| | - Baisong Hu
- Department of Orthopedics, Hangzhou Fuyang Hospital of Orthopedics of Traditional Chinese Medicine, Hangzhou 311499, China
| | - Qi Feng
- Department of Orthopedics, Hangzhou Fuyang Hospital of Orthopedics of Traditional Chinese Medicine, Hangzhou 311499, China
| | - Yuanyuan Xia
- Biomedical and Health Technology Innovation Platform, National University of Singapore (Suzhou) Research Institute, Suzhou 215123, China
| | - Lili Yu
- Biomedical and Health Technology Innovation Platform, National University of Singapore (Suzhou) Research Institute, Suzhou 215123, China
| | - Chunye Zhang
- Biomedical and Health Technology Innovation Platform, National University of Singapore (Suzhou) Research Institute, Suzhou 215123, China
| | - Wenjun Liu
- Department of Research and Development, Zhejiang Shangyue Biotechnology Research Center, Hangzhou 310000, China
| | - Zhao Liu
- Biomedical and Health Technology Innovation Platform, National University of Singapore (Suzhou) Research Institute, Suzhou 215123, China.
| | - Hai Yao
- Biomedical and Health Technology Innovation Platform, National University of Singapore (Suzhou) Research Institute, Suzhou 215123, China.
| | - Yong Lang
- Department of Orthopedics, Hangzhou Fuyang Hospital of Orthopedics of Traditional Chinese Medicine, Hangzhou 311499, China.
| |
Collapse
|
2
|
Heltmann-Meyer S, Detsch R, Hazur J, Kling L, Pechmann S, Kolan RR, Osterloh J, Boccaccini AR, Christiansen S, Geppert CI, Arkudas A, Horch RE, Steiner D. Biofunctionalization of ADA-GEL Hydrogels Based on the Degree of Cross-Linking and Polymer Concentration Improves Angiogenesis. Adv Healthc Mater 2025; 14:e2500730. [PMID: 40095294 DOI: 10.1002/adhm.202500730] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2025] [Revised: 02/26/2025] [Indexed: 03/19/2025]
Abstract
The creation of bioartificial tissues is a promising option for the reconstruction of large-volume defects. The vascularization of tissue engineering constructs, as well as the material properties of the carrier matrix, are important factors for successful clinical application. In this regard, hydrogels are promising biomaterials, providing an extracellular matrix-like milieu that enables the possibility of cell transplantation and de novo tissue formation. Furthermore, biofunctionalization allows for a certain fine-tuning of angiogenic properties. This study aims to investigate vascularization and tissue formation of highly cross-linked alginate dialdehyde (ADA) and gelatin (GEL). This highly cross-linked network is created using a dural cross-linking mechanism combining ionic (Ca2+ ions) and enzymatic (human transglutaminase (hTG)) cross-linking, resulting in reduced swelling and moderate degradation rates. Vascularization of the ADA-GEL-hTG constructs is induced surgically using arteriovenous (AV) loops. Biocompatibility, tissue formation, and vascularization are analyzed by histology and X-ray microscopy. After only 2 weeks, vascularization of the ADA-GEL-hTG constructs is already present. After 4 weeks, both de novo tissue formation and vascularization of the ADA-GEL-hTG matrix increase. In conclusion, ADA-GEL-hTG-based hydrogels are shown to be promising scaffold materials for tissue engineering applications.
Collapse
Affiliation(s)
- Stefanie Heltmann-Meyer
- Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Rainer Detsch
- Institute of Biomaterials, University of Erlangen-Nürnberg, 91058, Erlangen, Germany
| | - Jonas Hazur
- Institute of Biomaterials, University of Erlangen-Nürnberg, 91058, Erlangen, Germany
| | - Lasse Kling
- Institute for Nanotechnology and Correlative Microscopy gGmbH (INAM gGmbH), 91301, Forchheim, Germany
| | - Sabrina Pechmann
- Department for Correlative Microscopy and Materials Data, Fraunhofer Institute for Ceramic Technologies and Systems (IKTS), 91301, Forchheim, Germany
| | - Rajkumar Reddy Kolan
- Institute for Nanotechnology and Correlative Microscopy gGmbH (INAM gGmbH), 91301, Forchheim, Germany
| | - Justus Osterloh
- Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Department of Plastic and Hand Surgery, University of Freiburg Medical Center, 79106, Freiburg, Germany
| | - Aldo R Boccaccini
- Institute of Biomaterials, University of Erlangen-Nürnberg, 91058, Erlangen, Germany
| | - Silke Christiansen
- Department for Correlative Microscopy and Materials Data, Fraunhofer Institute for Ceramic Technologies and Systems (IKTS), 91301, Forchheim, Germany
- Fachbereich Physik, Freie Universität Berlin (FU Berlin), 14195, Berlin, Germany
| | - Carol I Geppert
- Institute of Pathology, University Hospital of Erlangen, Friedrich-Alexander-Universität, Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), University Hospital Erlangen, FAU Erlangen-Nuremberg, 91054, Erlangen, Germany
| | - Andreas Arkudas
- Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Raymund E Horch
- Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Dominik Steiner
- Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Department of Hand, Plastic, Reconstructive, and Burn Surgery, BG Trauma Clinic, University of Tübingen, 72076, Tübingen, Germany
| |
Collapse
|
3
|
Alkazemi H, Mitchell GM, Lokmic-Tomkins Z, Heath DE, O'Connor AJ. Hierarchically vascularized and suturable tissue constructs created through angiogenesis from tissue-engineered vascular grafts. Acta Biomater 2024; 189:168-178. [PMID: 39368723 DOI: 10.1016/j.actbio.2024.09.052] [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: 06/13/2024] [Revised: 09/01/2024] [Accepted: 09/26/2024] [Indexed: 10/07/2024]
Abstract
A major roadblock in implementing engineered tissues clinically lies in their limited vascularization. After implantation, such tissues do not integrate with the host's circulation as quickly as needed, commonly resulting in loss of viability and functionality. This study presents a solution to the vascularization problem that could enable the survival and function of large, transplantable, and vascularized engineered tissues. The technique allows vascularization of a cell laden hydrogel through angiogenesis from a suturable tissue-engineered vascular graft (TEVG) constructed from electrospun polycaprolactone with macropores. The graft is surrounded by a layer of cell-laden gelatin-methacryloyl hydrogel. The constructs are suturable and possess mechanical properties like native vessels. Angiogenesis occurs through the pores in the graft, resulting in a hydrogel containing an extensive vascular network that is connected to an implantable TEVG. The size of the engineered tissue and the degree of vascularization can be increased by adding multiple TEVGs into a single construct. The engineered tissue has the potential to be immediately perfused by the patient's blood upon surgical anastomosis to host vessels, enabling survival of implanted cells. These findings provide a meaningful step to address the longstanding problem of fabricating suturable pre-vascularized tissues which could survive upon implantation in vivo. STATEMENT OF SIGNIFICANCE: Creating vascularized engineered tissues that can be transplanted and rapidly perfused by the host blood supply is a major challenge which has limited the clinical impact of tissue engineering. In this study we demonstrate a technique to fabricate vascularized tissue constructs via angiogenesis from a suturable tissue-engineered vascular graft. The macroporous graft is surrounded with hydrogel, allowing endothelial cells to migrate from the lumen and vascularize the hydrogel layer with capillary-like structures connected to the macrovessel. The graft has comparable mechanical properties to native blood vessels and larger constructs can be fabricated by incorporating multiple grafts. These constructs could potentially be connected surgically to the circulation at an implantation site to support their immediate perfusion and survival.
Collapse
Affiliation(s)
- Hazem Alkazemi
- Department of Biomedical Engineering, Graeme Clark Institute, University of Melbourne, Victoria 3010, Australia
| | - Geraldine M Mitchell
- O'Brien Institute Department of Vincent's Institute of Medical Research, Victoria 3065, Australia; Faculty of Health Sciences, Australian Catholic University, Victoria 3065, Australia; Department of Surgery at St Vincent's Hospital Melbourne, University of Melbourne, Fitzroy, Victoria 3065, Australia
| | | | - Daniel E Heath
- Department of Biomedical Engineering, Graeme Clark Institute, University of Melbourne, Victoria 3010, Australia
| | - Andrea J O'Connor
- Department of Biomedical Engineering, Graeme Clark Institute, University of Melbourne, Victoria 3010, Australia; Aikenhead Centre for Medical Discovery (ACMD), Fitzroy, Victoria 3065, Australia.
| |
Collapse
|
4
|
Tanadchangsaeng N, Pasanaphong K, Tawonsawatruk T, Rattanapinyopituk K, Tangketsarawan B, Rawiwet V, Kongchanagul A, Srikaew N, Yoyruerop T, Panupinthu N, Sangpayap R, Panaksri A, Boonyagul S, Hemstapat R. 3D bioprinting of fish skin-based gelatin methacryloyl (GelMA) bio-ink for use as a potential skin substitute. Sci Rep 2024; 14:23240. [PMID: 39369014 PMCID: PMC11455937 DOI: 10.1038/s41598-024-73774-1] [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: 07/19/2024] [Accepted: 09/20/2024] [Indexed: 10/07/2024] Open
Abstract
Gelatin methacryloyl (GelMA), typically derived from mammalian sources, has recently emerged as an ideal bio-ink for three-dimensional (3D) bioprinting. Herein, we developed a fish skin-based GelMA bio-ink for the fabrication of a 3D GelMA skin substitute with a 3D bioprinter. Several concentrations of methacrylic acid anhydride were used to fabricate GelMA, in which their physical-mechanical properties were assessed. This fish skin-based GelMA bio-ink was loaded with human adipose tissue-derived mesenchymal stromal cells (ASCs) and human platelet lysate (HPL) and then printed to obtain 3D ASCs + HPL-loaded GelMA scaffolds. Cell viability test and a preliminary investigation of its effectiveness in promoting wound closure were evaluated in a critical-sized full thickness skin defect in a rat model. The cell viability results showed that the number of ASCs increased significantly within the 3D GelMA hydrogel scaffold, indicating its biocompatibility property. In vivo results demonstrated that ASCs + HPL-loaded GelMA scaffolds could delay wound contraction, markedly enhanced collagen deposition, and promoted the formation of new blood vessels, especially at the wound edge, compared to the untreated group. Therefore, this newly fish skin-based GelMA bio-ink developed in this study has the potential to be utilized for the printing of 3D GelMA skin substitutes.
Collapse
Affiliation(s)
| | | | - Tulyapruek Tawonsawatruk
- Department of Orthopaedics, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Kasem Rattanapinyopituk
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | | | - Visut Rawiwet
- Central Animal Facility, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Alita Kongchanagul
- Center for Vaccine Development, Institute of Molecular Biosciences, Mahidol University, Bangkok, Thailand
| | - Narongrit Srikaew
- Research Center, Faculty of Medicine Ramathibodi Hospital, Mahidol University, Bangkok, Thailand
| | - Thanaporn Yoyruerop
- Mahidol University-Frontier Research Facility (MU-FRF), Mahidol University, Nakhon Pathom, Thailand
| | - Nattapon Panupinthu
- Department of Physiology, Faculty of Science, Mahidol University, Bangkok, Thailand
- Center of Calcium and Bone Research (COCAB), Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Ratirat Sangpayap
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand
| | - Anuchan Panaksri
- College of Biomedical Engineering, Rangsit University, Pathum Thani, Thailand
| | - Sani Boonyagul
- College of Biomedical Engineering, Rangsit University, Pathum Thani, Thailand
| | - Ruedee Hemstapat
- Department of Pharmacology, Faculty of Science, Mahidol University, Bangkok, Thailand.
| |
Collapse
|
5
|
Schlauch D, Ebbecke JP, Meyer J, Fleischhammer TM, Pirmahboub H, Kloke L, Kara S, Lavrentieva A, Pepelanova I. Development of a Human Recombinant Collagen for Vat Polymerization-Based Bioprinting. Biotechnol J 2024; 19:e202400393. [PMID: 39380502 DOI: 10.1002/biot.202400393] [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: 06/20/2024] [Revised: 08/13/2024] [Accepted: 08/30/2024] [Indexed: 10/10/2024]
Abstract
In light-based 3D-bioprinting, gelatin methacrylate (GelMA) is one of the most widely used materials, as it supports cell attachment, and shows good biocompatibility and degradability in vivo. However, as an animal-derived material, it also causes safety concerns when used in medical applications. Gelatin is a partial hydrolysate of collagen, containing high amounts of hydroxyproline. This causes the material to form a thermally induced gel at ambient temperatures, a behavior also observed in GelMA. This temperature-dependent gelation requires precise temperature control during the bioprinting process to prevent the gelation of the material. To avoid safety concerns associated with animal-derived materials and reduce potential issues caused by thermal gelation, a recombinant human alpha-1 collagen I fragment was expressed in Komagataella phaffii without hydroxylation. The resulting protein was successfully modified with methacryloyl groups and underwent rapid photopolymerization upon ultraviolet light exposure. The developed material exhibited slightly slower polymerization and lower storage modulus compared to GelMA, while it showed higher stretchability. However, unlike the latter, the material did not undergo physical gelation at ambient temperatures, but only when cooled down to below 10°C, a characteristic that has not been described for comparable materials so far. This gelation was not caused by the formation of triple-helical structures, as shown by the absence of the characteristic peak at 220 nm in CD spectra. Moreover, the developed recombinant material facilitated cell adherence with high cell viability after crosslinking via light to a 3D structure. Furthermore, desired geometries could be easily printed on a stereolithographic bioprinter.
Collapse
Affiliation(s)
- Domenic Schlauch
- Cellbricks GmbH, Berlin, Germany
- Leibniz University Hannover, Hannover, Germany
| | - Jan Peter Ebbecke
- Cellbricks GmbH, Berlin, Germany
- Leibniz University Hannover, Hannover, Germany
| | | | | | | | | | - Selin Kara
- Leibniz University Hannover, Hannover, Germany
| | | | | |
Collapse
|
6
|
Zhang J, Suttapreyasri S, Leethanakul C, Samruajbenjakun B. Fabrication of vascularized tissue-engineered bone models using triaxial bioprinting. J Biomed Mater Res A 2024; 112:1093-1106. [PMID: 38411369 DOI: 10.1002/jbm.a.37694] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2023] [Revised: 01/01/2024] [Accepted: 02/14/2024] [Indexed: 02/28/2024]
Abstract
Bone tissue is a highly vascularized tissue. When constructing tissue-engineered bone models, both the osteogenic and angiogenic capabilities of the construct should be carefully considered. However, fabricating a vascularized tissue-engineered bone to promote vascular formation and bone generation, while simultaneously establishing nutrition channels to facilitate nutrient exchange within the constructs, remains a significant challenge. Triaxial bioprinting, which not only allows the independent encapsulation of different cell types while simultaneously forming nutrient channels, could potentially emerge as a strategy for fabricating vascularized tissue-engineered bone. Moreover, bioinks should also be applied in combination to promote both osteogenesis and angiogenesis. In this study, employing triaxial bioprinting, we used a blend bioink of gelatin methacryloyl (GelMA), sodium alginate (Alg), and different concentrations of nano beta-tricalcium phosphate (nano β-TCP) encapsulated MC3T3-E1 preosteoblasts as the outer layer, a mixed bioink of GelMA and Alg loaded with human umbilical vein endothelial cells (HUVEC) as the middle layer, and gelatin as a sacrificial material to form nutrient channels in the inner layer to fabricate vascularized bone constructs simulating the microenvironment for bone and vascular tissues. The results showed that the addition of nano β-TCP could adjust the mechanical, swelling, and degradation properties of the constructs. Biological assessments revealed the cell viability of constructs containing different concentrations of nano β-TCP was higher than 90% on day 7, The cell-laden constructs containing 3% (w/v) nano β-TCP exhibited better osteogenic (higher Alkaline phosphatase activity and larger Osteocalcin positive area) and angiogenic (the gradual increased CD31 positive area) potential. Therefore, using triaxial bioprinting technology and employing GelMA, Alg, and nano β-TCP as bioink components could fabricate vascularized bone tissue constructs, offering a novel strategy for vascularized bone tissue engineering.
Collapse
Affiliation(s)
- Junbiao Zhang
- Orthodontic Section, Department of Preventive Dentistry, Faculty of Dentistry, Prince of Songkla University, Songkhla, Thailand
- Guiyang Hospital of Stomatology, Guiyang, People's Republic of China
| | - Srisurang Suttapreyasri
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Prince of Songkla University, Hat Yai, Thailand
| | - Chidchanok Leethanakul
- Orthodontic Section, Department of Preventive Dentistry, Faculty of Dentistry, Prince of Songkla University, Songkhla, Thailand
| | - Bancha Samruajbenjakun
- Orthodontic Section, Department of Preventive Dentistry, Faculty of Dentistry, Prince of Songkla University, Songkhla, Thailand
| |
Collapse
|
7
|
Imere A, Foster NC, Hajiali H, Okur KE, Wright AL, Barroso IA, Haj AJE. Enhanced chondrogenic potential in GelMA-based 3D cartilage model via Wnt3a surface immobilization. Sci Rep 2024; 14:15022. [PMID: 38951570 PMCID: PMC11217376 DOI: 10.1038/s41598-024-65970-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2024] [Accepted: 06/25/2024] [Indexed: 07/03/2024] Open
Abstract
Cartilage tissue engineering aims to develop functional substitutes for treating cartilage defects and osteoarthritis. Traditional two-dimensional (2D) cell culture systems lack the complexity of native cartilage, leading to the development of 3D regenerative cartilage models. In this study, we developed a 3D model using Gelatin Methacryloyl (GelMA)-based hydrogels seeded with Y201 cells, a bone marrow mesenchymal stem cell line. The model investigated chondrogenic differentiation potential in response to Wnt3a stimulation within the GelMA scaffold and validated using known chondrogenic agonists. Y201 cells demonstrated suitability for the model, with increased proteoglycan content and upregulated chondrogenic marker expression under chondrogenic conditions. Wnt3a enhanced cell proliferation, indicating activation of the Wnt/β-catenin pathway, which plays a role in cartilage development. GelMA hydrogels provided an optimal scaffold, supporting cell viability and proliferation. The 3D model exhibited consistent responses to chondrogenic agonists, with TGF-β3 enhancing cartilage-specific extracellular matrix (ECM) production and chondrogenic differentiation. The combination of Wnt3a and TGF-β3 showed synergistic effects, promoting chondrogenic differentiation and ECM production. This study presents a 3D regenerative cartilage model with potential for investigating cartilage biology, disease mechanisms, and drug screening. The model provides insights into complex cartilage regeneration mechanisms and offers a platform for developing therapeutic approaches for cartilage repair and osteoarthritis treatment.
Collapse
Affiliation(s)
- Angela Imere
- Healthcare Technologies Institute, Institute of Translational Medicine, National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK
| | - Nicola C Foster
- Healthcare Technologies Institute, Institute of Translational Medicine, National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK
| | - Hadi Hajiali
- Healthcare Technologies Institute, Institute of Translational Medicine, National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK.
| | - Kerime Ebrar Okur
- Healthcare Technologies Institute, Institute of Translational Medicine, National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK
| | - Abigail L Wright
- Healthcare Technologies Institute, Institute of Translational Medicine, National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK
| | - Ines A Barroso
- Healthcare Technologies Institute, Institute of Translational Medicine, National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK
| | - Alicia J El Haj
- Healthcare Technologies Institute, Institute of Translational Medicine, National Institute for Health and Care Research (NIHR) Birmingham Biomedical Research Centre, School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK.
| |
Collapse
|
8
|
Pramanik S, Alhomrani M, Alamri AS, Alsanie WF, Nainwal P, Kimothi V, Deepak A, Sargsyan AS. Unveiling the versatility of gelatin methacryloyl hydrogels: a comprehensive journey into biomedical applications. Biomed Mater 2024; 19:042008. [PMID: 38768611 DOI: 10.1088/1748-605x/ad4df7] [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: 02/20/2024] [Accepted: 05/20/2024] [Indexed: 05/22/2024]
Abstract
Gelatin methacryloyl (GelMA) hydrogels have gained significant recognition as versatile biomaterials in the biomedical domain. GelMA hydrogels emulate vital characteristics of the innate extracellular matrix by integrating cell-adhering and matrix metalloproteinase-responsive peptide motifs. These features enable cellular proliferation and spreading within GelMA-based hydrogel scaffolds. Moreover, GelMA displays flexibility in processing, as it experiences crosslinking when exposed to light irradiation, supporting the development of hydrogels with adjustable mechanical characteristics. The drug delivery landscape has been reshaped by GelMA hydrogels, offering a favorable platform for the controlled and sustained release of therapeutic actives. The tunable physicochemical characteristics of GelMA enable precise modulation of the kinetics of drug release, ensuring optimal therapeutic effectiveness. In tissue engineering, GelMA hydrogels perform an essential role in the design of the scaffold, providing a biomimetic environment conducive to cell adhesion, proliferation, and differentiation. Incorporating GelMA in three-dimensional printing further improves its applicability in drug delivery and developing complicated tissue constructs with spatial precision. Wound healing applications showcase GelMA hydrogels as bioactive dressings, fostering a conducive microenvironment for tissue regeneration. The inherent biocompatibility and tunable mechanical characteristics of GelMA provide its efficiency in the closure of wounds and tissue repair. GelMA hydrogels stand at the forefront of biomedical innovation, offering a versatile platform for addressing diverse challenges in drug delivery, tissue engineering, and wound healing. This review provides a comprehensive overview, fostering an in-depth understanding of GelMA hydrogel's potential impact on progressing biomedical sciences.
Collapse
Affiliation(s)
- Sheersha Pramanik
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai 600036, Tamil Nadu, India
| | - Majid Alhomrani
- Department of Clinical Laboratory Sciences, The faculty of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
- Centre of Biomedical Sciences Research (CBSR), Deanship of Scientific Research, Taif University, Taif, Saudi Arabia
| | - Abdulhakeem S Alamri
- Department of Clinical Laboratory Sciences, The faculty of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
- Centre of Biomedical Sciences Research (CBSR), Deanship of Scientific Research, Taif University, Taif, Saudi Arabia
| | - Walaa F Alsanie
- Department of Clinical Laboratory Sciences, The faculty of Applied Medical Sciences, Taif University, Taif, Saudi Arabia
- Centre of Biomedical Sciences Research (CBSR), Deanship of Scientific Research, Taif University, Taif, Saudi Arabia
| | - Pankaj Nainwal
- School of Pharmacy, Graphic Era Hill University, Dehradun 248001, India
| | - Vishwadeepak Kimothi
- Himalayan Institute of Pharmacy and Research, Rajawala, Dehradun, Uttrakhand, India
| | - A Deepak
- Saveetha Institute of Medical and Technical Sciences, Saveetha School of Engineering, Chennai, Tamil Nadu 600128, India
| | - Armen S Sargsyan
- Scientific and Production Center 'Armbiotechnology' NAS RA, 14 Gyurjyan Str., Yerevan 0056, Armenia
| |
Collapse
|
9
|
Simińska-Stanny J, Nicolas L, Chafai A, Jafari H, Hajiabbas M, Dodi G, Gardikiotis I, Delporte C, Nie L, Podstawczyk D, Shavandi A. Advanced PEG-tyramine biomaterial ink for precision engineering of perfusable and flexible small-diameter vascular constructs via coaxial printing. Bioact Mater 2024; 36:168-184. [PMID: 38463551 PMCID: PMC10924180 DOI: 10.1016/j.bioactmat.2024.02.019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 02/09/2024] [Accepted: 02/16/2024] [Indexed: 03/12/2024] Open
Abstract
Vascularization is crucial for providing nutrients and oxygen to cells while removing waste. Despite advances in 3D-bioprinting, the fabrication of structures with void spaces and channels remains challenging. This study presents a novel approach to create robust yet flexible and permeable small (600-1300 μm) artificial vessels in a single processing step using 3D coaxial extrusion printing of a biomaterial ink, based on tyramine-modified polyethylene glycol (PEG-Tyr). We combined the gelatin biocompatibility/activity, robustness of PEG-Tyr and alginate with the shear-thinning properties of methylcellulose (MC) in a new biomaterial ink for the fabrication of bioinspired vessels. Chemical characterization using NMR and FTIR spectroscopy confirmed the successful modification of PEG with Tyr and rheological characterization indicated that the addition of PEG-Tyr decreased the viscosity of the ink. Enzyme-mediated crosslinking of PEG-Tyr allowed the formation of covalent crosslinks within the hydrogel chains, ensuring its stability. PEG-Tyr units improved the mechanical properties of the material, resulting in stretchable and elastic constructs without compromising cell viability and adhesion. The printed vessel structures displayed uniform wall thickness, shape retention, improved elasticity, permeability, and colonization by endothelial-derived - EA.hy926 cells. The chorioallantoic membrane (CAM) and in vivo assays demonstrated the hydrogel's ability to support neoangiogenesis. The hydrogel material with PEG-Tyr modification holds promise for vascular tissue engineering applications, providing a flexible, biocompatible, and functional platform for the fabrication of vascular structures.
Collapse
Affiliation(s)
- Julia Simińska-Stanny
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
| | - Lise Nicolas
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
- European School of Materials Science and Engineering, University of Lorraine, Nancy, France
| | - Adam Chafai
- Université Libre de Bruxelles (ULB), Micro-milli Platform, Avenue F.D. Roosevelt, 50 - CP 165/67, 1050, Brussels, Belgium
| | - Hafez Jafari
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
| | - Maryam Hajiabbas
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
- Université Libre de Bruxelles (ULB), Faculté de Médecine, Campus Erasme - CP 611, Laboratory of Pathophysiological and Nutritional Biochemistry, Route de Lennik, 808, 1070, Bruxelles, Belgium
| | - Gianina Dodi
- Faculty of Medical Bioengineering, Grigore T. Popa, University of Medicine and Pharmacy of Iasi, Romania
| | - Ioannis Gardikiotis
- Advanced Research and Development Center for Experimental Medicine, Grigore T. Popa, University of Medicine and Pharmacy of Iasi, Romania
| | - Christine Delporte
- Université Libre de Bruxelles (ULB), Faculté de Médecine, Campus Erasme - CP 611, Laboratory of Pathophysiological and Nutritional Biochemistry, Route de Lennik, 808, 1070, Bruxelles, Belgium
| | - Lei Nie
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
- College of Life Science, Xinyang Normal University, Xinyang, China
| | - Daria Podstawczyk
- Department of Process Engineering and Technology of Polymer and Carbon Materials, Faculty of Chemistry, Wroclaw University of Science and Technology, Norwida 4/6, 50-373, Wroclaw, Poland
| | - Amin Shavandi
- Université Libre de Bruxelles (ULB), École polytechnique de Bruxelles, 3BIO-BioMatter, Avenue F.D. Roosevelt, 50 - CP 165/61, 1050, Brussels, Belgium
| |
Collapse
|
10
|
Schmid R, Schmidt SK, Schrüfer S, Schubert DW, Heltmann-Meyer S, Schicht M, Paulsen F, Horch RE, Bosserhoff AK, Kengelbach-Weigand A, Arkudas A. A vascularized in vivo melanoma model suitable for metastasis research of different tumor stages using fundamentally different bioinks. Mater Today Bio 2024; 26:101071. [PMID: 38736612 PMCID: PMC11081803 DOI: 10.1016/j.mtbio.2024.101071] [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: 02/12/2024] [Revised: 04/21/2024] [Accepted: 04/25/2024] [Indexed: 05/14/2024] Open
Abstract
Although 2D cancer models have been the standard for drug development, they don't resemble in vivo properties adequately. 3D models can potentially overcome this. Bioprinting is a promising technique for more refined models to investigate central processes in tumor development such as proliferation, dormancy or metastasis. We aimed to analyze bioinks, which could mimic these different tumor stages in a cast vascularized arteriovenous loop melanoma model in vivo. It has the advantage to be a closed system with a defined microenvironment, supplied only with one vessel-ideal for metastasis research. Tested bioinks showed significant differences in composition, printability, stiffness and microscopic pore structure, which led to different tumor stages (Matrigel and Alg/HA/Gel for progression, Cellink Bioink for dormancy) and resulted in different primary tumor growth (Matrigel significantly higher than Cellink Bioink). Light-sheet fluorescence microscopy revealed differences in vascularization and hemorrhages with no additional vessels found in Cellink Bioink. Histologically, typical human melanoma with different stages was demonstrated. HMB-45-positive tumors in progression inks were infiltrated by macrophages (CD163), highly proliferative (Ki67) and metastatic (MITF/BRN2, ATX, MMP3). Stainings of lymph nodes revealed metastases even without significant primary tumor growth in Cellink Bioink. This model can be used to study tumor pathology and metastasis of different tumor stages and therapies.
Collapse
Affiliation(s)
- Rafael Schmid
- Laboratory for Tissue-Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Krankenhausstraße 12, 91054, Erlangen, Germany
| | - Sonja K. Schmidt
- Institute of Biochemistry, Friedrich-Alexander University Erlangen-Nürnberg, Fahrstraße 17, 91054, Erlangen, Germany
| | - Stefan Schrüfer
- Institute of Polymer Materials, Friedrich-Alexander University Erlangen-Nürnberg, Martensstraße 7, 91058, Erlangen, Germany
| | - Dirk W. Schubert
- Institute of Polymer Materials, Friedrich-Alexander University Erlangen-Nürnberg, Martensstraße 7, 91058, Erlangen, Germany
| | - Stefanie Heltmann-Meyer
- Laboratory for Tissue-Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Krankenhausstraße 12, 91054, Erlangen, Germany
| | - Martin Schicht
- Department of Functional and Clinical Anatomy, Friedrich-Alexander University Erlangen-Nürnberg, Universitätsstraße 19, 91054, Erlangen, Germany
| | - Friedrich Paulsen
- Department of Functional and Clinical Anatomy, Friedrich-Alexander University Erlangen-Nürnberg, Universitätsstraße 19, 91054, Erlangen, Germany
| | - Raymund E. Horch
- Laboratory for Tissue-Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Krankenhausstraße 12, 91054, Erlangen, Germany
| | - Anja K. Bosserhoff
- Institute of Biochemistry, Friedrich-Alexander University Erlangen-Nürnberg, Fahrstraße 17, 91054, Erlangen, Germany
| | - Annika Kengelbach-Weigand
- Laboratory for Tissue-Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Krankenhausstraße 12, 91054, Erlangen, Germany
| | - Andreas Arkudas
- Laboratory for Tissue-Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Krankenhausstraße 12, 91054, Erlangen, Germany
| |
Collapse
|
11
|
Cirves E, Vargas A, Wheeler EE, Leach JK, Simon SI, Gonzalez‐Fernandez T. Neutrophil Granulopoiesis Optimized Through Ex Vivo Expansion of Hematopoietic Progenitors in Engineered 3D Gelatin Methacrylate Hydrogels. Adv Healthc Mater 2024; 13:e2301966. [PMID: 38345178 PMCID: PMC11144100 DOI: 10.1002/adhm.202301966] [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: 06/21/2023] [Revised: 02/07/2024] [Indexed: 02/27/2024]
Abstract
Neutrophils are the first line of defense of the innate immune system. In response to methicillin-resistant Staphylococcus aureus infection in the skin, hematopoietic stem, and progenitor cells (HSPCs) traffic to wounds and undergo extramedullary granulopoiesis, producing neutrophils necessary to resolve the infection. This prompted the engineering of a gelatin methacrylate (GelMA) hydrogel that encapsulates HSPCs within a matrix amenable to subcutaneous delivery. The authors study the influence of hydrogel mechanical properties to produce an artificial niche for granulocyte-monocyte progenitors (GMPs) to efficiently expand into functional neutrophils that can populate infected tissue. Lin-cKIT+ HSPCs, harvested from fluorescent neutrophil reporter mice, are encapsulated in GelMA hydrogels of varying polymer concentration and UV-crosslinked to produce HSPC-laden gels of specific stiffness and mesh sizes. Softer 5% GelMA gels yield the most viable progenitors and effective cell-matrix interactions. Compared to suspension culture, 5% GelMA results in a twofold expansion of mature neutrophils that retain antimicrobial functions including degranulation, phagocytosis, and ROS production. When implanted dermally in C57BL/6J mice, luciferase-expressing neutrophils expanded in GelMA hydrogels are visualized at the site of implantation for over 5 days. They demonstrate the potential of GelMA hydrogels for delivering HSPCs directly to the site of skin infection to promote local granulopoiesis.
Collapse
Affiliation(s)
- Evan Cirves
- Department of Biomedical EngineeringUniversity of California at Davis451 East Health Sciences Drive, 2303 GBSFDavisCA95616USA
| | - Alex Vargas
- Department of Biomedical EngineeringUniversity of California at Davis451 East Health Sciences Drive, 2303 GBSFDavisCA95616USA
| | - Erika E. Wheeler
- Department of Biomedical EngineeringUniversity of California at Davis451 East Health Sciences Drive, 2303 GBSFDavisCA95616USA
- Department of Orthopaedic SurgeryUC Davis Health4860 Y Street, Suite 3800SacramentoCA95817USA
| | - Jonathan Kent Leach
- Department of Orthopaedic SurgeryUC Davis Health4860 Y Street, Suite 3800SacramentoCA95817USA
| | - Scott I. Simon
- Department of Biomedical Engineering and DermatologyUniversity of California at DavisDavisCA95616USA
| | - Tomas Gonzalez‐Fernandez
- Department of BioengineeringLehigh University124 E Morton Street, Health Science and Technology BuildingBethlehemPA18015USA
| |
Collapse
|
12
|
Xu Q, Bai Y, Li S, Hou W, Hao Y, Yang R, Li X, Zhang X. Enhancing osteogenesis and angiogenesis functions for Ti-24Nb-4Zr-8Sn scaffolds with methacrylated gelatin and deferoxamine. Front Bioeng Biotechnol 2024; 12:1372636. [PMID: 38707506 PMCID: PMC11066197 DOI: 10.3389/fbioe.2024.1372636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Accepted: 04/08/2024] [Indexed: 05/07/2024] Open
Abstract
Repair of large bone defects remains challenge for orthopedic clinical treatment. Porous titanium alloys have been widely fabricated by the additive manufacturing, which possess the elastic modulus close to that of human cortical bone, good osteoconductivity and osteointegration. However, insufficient bone regeneration and vascularization inside the porous titanium scaffolds severely limit their capability for repair of large-size bone defects. Therefore, it is crucially important to improve the osteogenic function and vascularization of the titanium scaffolds. Herein, methacrylated gelatin (GelMA) were incorporated with the porous Ti-24Nb-4Zr-8Sn (Ti2448) scaffolds prepared by the electron beam melting (EBM) method (Ti2448-GelMA). Besides, the deferoxamine (DFO) as an angiogenic agent was doped into the Ti2448-GelMA scaffold (Ti2448-GelMA/DFO), in order to promote vascularization. The results indicate that GelMA can fully infiltrate into the pores of Ti2448 scaffolds with porous cross-linked network (average pore size: 120.2 ± 25.1 μm). Ti2448-GelMA scaffolds facilitated the differentiation of MC3T3-E1 cells by promoting the ALP expression and mineralization, with the amount of calcium contents ∼2.5 times at day 14, compared with the Ti2448 scaffolds. Impressively, the number of vascular meshes for the Ti2448-GelMA/DFO group (∼7.2/mm2) was significantly higher than the control group (∼5.3/mm2) after cultivation for 9 h, demonstrating the excellent angiogenesis ability. The Ti2448-GelMA/DFO scaffolds also exhibited sustained release of DFO, with a cumulative release of 82.3% after 28 days. Therefore, Ti2448-GelMA/DFO scaffolds likely provide a new strategy to improve the osteogenesis and angiogenesis for repair of large bone defects.
Collapse
Affiliation(s)
- Qian Xu
- Department of Materials Physics and Chemistry, School of Materials Science and Engineering, Key Laboratory for Anisotropy and Texture of Materials, Ministry of Education, Northeastern University, Shenyang, Liaoning, China
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, Liaoning, China
| | - Yun Bai
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, Liaoning, China
- School of Materials Science and Engineering, University of Science and Technology of China, Hefei, Anhui, China
| | - Shujun Li
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, Liaoning, China
- School of Materials Science and Engineering, University of Science and Technology of China, Hefei, Anhui, China
| | - Wentao Hou
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, Liaoning, China
- School of Materials Science and Engineering, University of Science and Technology of China, Hefei, Anhui, China
| | - Yulin Hao
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, Liaoning, China
- School of Materials Science and Engineering, University of Science and Technology of China, Hefei, Anhui, China
| | - Rui Yang
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, Liaoning, China
- School of Materials Science and Engineering, University of Science and Technology of China, Hefei, Anhui, China
| | - Xiaowu Li
- Department of Materials Physics and Chemistry, School of Materials Science and Engineering, Key Laboratory for Anisotropy and Texture of Materials, Ministry of Education, Northeastern University, Shenyang, Liaoning, China
| | - Xing Zhang
- Institute of Metal Research, Chinese Academy of Sciences, Shenyang, Liaoning, China
- School of Materials Science and Engineering, University of Science and Technology of China, Hefei, Anhui, China
| |
Collapse
|
13
|
Xu Z, Arkudas A, Munawar MA, Schubert DW, Fey T, Weisbach V, Mengen LM, Horch RE, Cai A. Schwann Cells Do Not Promote Myogenic Differentiation in the EPI Loop Model. Tissue Eng Part A 2024; 30:244-256. [PMID: 38063005 DOI: 10.1089/ten.tea.2023.0215] [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: 01/12/2024] Open
Abstract
In skeletal muscle tissue engineering, innervation and vascularization play an essential role in the establishment of functional skeletal muscle. For adequate three-dimensional assembly, biocompatible aligned nanofibers are beneficial as matrices for cell seeding. The aim of this study was to analyze the impact of Schwann cells (SC) on myoblast (Mb) and adipogenic mesenchymal stromal cell (ADSC) cocultures on poly-ɛ-caprolactone (PCL)-collagen I-nanofibers in vivo. Human Mb/ADSC cocultures, as well as Mb/ADSC/SC cocultures, were seeded onto PCL-collagen I-nanofiber scaffolds and implanted into the innervated arteriovenous loop model (EPI loop model) of immunodeficient rats for 4 weeks. Histological staining and gene expression were used to compare their capacity for vascularization, immunological response, myogenic differentiation, and innervation. After 4 weeks, both Mb/ADSC and Mb/ADSC/SC coculture systems showed similar amounts and distribution of vascularization, as well as immunological activity. Myogenic differentiation could be observed in both groups through histological staining (desmin, myosin heavy chain) and gene expression (MYOD, MYH3, ACTA1) without significant difference between groups. Expression of CHRNB and LAMB2 also implied neuromuscular junction formation. Our study suggests that the addition of SC did not significantly impact myogenesis and innervation in this model. The implanted motor nerve branch may have played a more significant role than the presence of SC.
Collapse
Affiliation(s)
- Zhou Xu
- Laboratory for Tissue Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
- Department of Thyroid and Breast Surgery, Affiliated Hospital of North Sichuan Medical College, Nanchong, China
| | - Andreas Arkudas
- Laboratory for Tissue Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Muhammad Azeem Munawar
- Department of Materials Science and Engineering, Institute of Polymer Materials, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Dirk W Schubert
- Department of Materials Science and Engineering, Institute of Polymer Materials, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Tobias Fey
- Department of Materials Science and Engineering, Institute of Glass and Ceramics, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Volker Weisbach
- Department of Transfusion Medicine, University Hospital of Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Lilly M Mengen
- Laboratory for Tissue Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Raymund E Horch
- Laboratory for Tissue Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Aijia Cai
- Laboratory for Tissue Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-University Erlangen-Nürnberg (FAU), Erlangen, Germany
| |
Collapse
|
14
|
Feng B, Dong T, Song X, Zheng X, Jin C, Cheng Z, Liu Y, Zhang W, Wang X, Tao Y, Wu H. Personalized Porous Gelatin Methacryloyl Sustained-Release Nicotinamide Protects Against Noise-Induced Hearing Loss. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2305682. [PMID: 38225752 DOI: 10.1002/advs.202305682] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 11/11/2023] [Indexed: 01/17/2024]
Abstract
There are no Food and Drug Administration-approved drugs for treating noise-induced hearing loss (NIHL), reflecting the absence of clear specific therapeutic targets and effective delivery strategies. Noise trauma is demonstrated results in nicotinamide adenine dinucleotide (NAD+) downregulation and mitochondrial dysfunction in cochlear hair cells (HCs) and spiral ganglion neurons (SGNs) in mice, and NAD+ boosted by nicotinamide (NAM) supplementation maintains cochlear mitochondrial homeostasis and prevents neuroexcitatory toxic injury in vitro and ex vivo, also significantly ameliorated NIHL in vivo. To tackle the limited drug delivery efficiency due to sophisticated anatomical barriers and unique clearance pathway in ear, personalized NAM-encapsulated porous gelatin methacryloyl (PGMA@NAM) are developed based on anatomy topography of murine temporal bone by micro-computed tomography and reconstruction of round window (RW) niche, realizing hydrogel in situ implantation completely, NAM sustained-release and long-term auditory preservation in mice. This study strongly supports personalized PGMA@NAM as NIHL protection drug with effective inner ear delivery, providing new inspiration for drug-based treatment of NIHL.
Collapse
Affiliation(s)
- Baoyi Feng
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639, Zhizaoju Road, Shanghai, 200011, P. R. China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, No.115, Jinzun Road, Shanghai, 200125, P. R. China
- Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, No.115, Jinzun Road, Shanghai, 200125, P. R. China
| | - Tingting Dong
- Ear Institute, Shanghai Jiao Tong University School of Medicine, No.115, Jinzun Road, Shanghai, 200125, P. R. China
- Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, No.115, Jinzun Road, Shanghai, 200125, P. R. China
- Biobank of Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.115, Jinzun Road, Shanghai, 200125, P. R. China
| | - Xinyu Song
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Xiaofei Zheng
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639, Zhizaoju Road, Shanghai, 200011, P. R. China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, No.115, Jinzun Road, Shanghai, 200125, P. R. China
- Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, No.115, Jinzun Road, Shanghai, 200125, P. R. China
| | - Chenxi Jin
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639, Zhizaoju Road, Shanghai, 200011, P. R. China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, No.115, Jinzun Road, Shanghai, 200125, P. R. China
- Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, No.115, Jinzun Road, Shanghai, 200125, P. R. China
| | - Zhenzhe Cheng
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639, Zhizaoju Road, Shanghai, 200011, P. R. China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, No.115, Jinzun Road, Shanghai, 200125, P. R. China
- Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, No.115, Jinzun Road, Shanghai, 200125, P. R. China
| | - Yiqing Liu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639, Zhizaoju Road, Shanghai, 200011, P. R. China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, No.115, Jinzun Road, Shanghai, 200125, P. R. China
- Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, No.115, Jinzun Road, Shanghai, 200125, P. R. China
| | - Wenjie Zhang
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, P. R. China
| | - Xueling Wang
- Ear Institute, Shanghai Jiao Tong University School of Medicine, No.115, Jinzun Road, Shanghai, 200125, P. R. China
- Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, No.115, Jinzun Road, Shanghai, 200125, P. R. China
- Biobank of Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.115, Jinzun Road, Shanghai, 200125, P. R. China
| | - Yong Tao
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639, Zhizaoju Road, Shanghai, 200011, P. R. China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, No.115, Jinzun Road, Shanghai, 200125, P. R. China
- Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, No.115, Jinzun Road, Shanghai, 200125, P. R. China
| | - Hao Wu
- Department of Otolaryngology-Head and Neck Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, No.639, Zhizaoju Road, Shanghai, 200011, P. R. China
- Ear Institute, Shanghai Jiao Tong University School of Medicine, No.115, Jinzun Road, Shanghai, 200125, P. R. China
- Shanghai Key Laboratory of Translation Medicine on Ear and Nose Disease, No.115, Jinzun Road, Shanghai, 200125, P. R. China
| |
Collapse
|
15
|
Carpentier N, Ye S, Delemarre MD, Van der Meeren L, Skirtach AG, van der Laan LJW, Schneeberger K, Spee B, Dubruel P, Van Vlierberghe S. Gelatin-Based Hybrid Hydrogels as Matrices for Organoid Culture. Biomacromolecules 2024; 25:590-604. [PMID: 38174962 DOI: 10.1021/acs.biomac.2c01496] [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: 01/05/2024]
Abstract
The application of liver organoids is very promising in the field of liver tissue engineering; however, it is still facing some limitations. One of the current major limitations is the matrix in which they are cultured. The mainly undefined and murine-originated tumor matrices derived from Engelbreth-Holm-Swarm (EHS) sarcoma, such as Matrigel, are still the standard culturing matrices for expansion and differentiation of organoids toward hepatocyte-like cells, which will obstruct its future clinical application potential. In this study, we exploited the use of newly developed highly defined hydrogels as potential matrices for the culture of liver organoids and compared them to Matrigel and two hydrogels that were already researched in the field of organoid research [i.e., polyisocyanopeptides, enriched with laminin-entactin complex (PIC-LEC) and gelatin methacryloyl (GelMA)]. The newly developed hydrogels are materials that have a physicochemical resemblance with native liver tissue. Norbornene-modified dextran cross-linked with thiolated gelatin (DexNB-GelSH) has a swelling ratio and macro- and microscale properties that highly mimic liver tissue. Norbornene-modified chondroitin sulfate cross-linked with thiolated gelatin (CSNB-GelSH) contains chondroitin sulfate, which is a glycosaminoglycan (GAG) that is present in the liver ECM. Furthermore, CSNB-GelSH hydrogels with different mechanical properties were evaluated. Bipotent intrahepatic cholangiocyte organoids (ICOs) were applied in this work and encapsulated in these materials. This research revealed that the newly developed materials outperformed Matrigel, PIC-LEC, and GelMA in the differentiation of ICOs toward hepatocyte-like cells. Furthermore, some trends indicate that an interplay of both the chemical composition and the mechanical properties has an influence on the relative expression of certain hepatocyte markers. Both DexNB-GelSH and CSNB-GelSH showed promising results for the expansion and differentiation of intrahepatic cholangiocyte organoids. The stiffest CSNB-GelSH hydrogel even significantly outperformed Matrigel based on ALB, BSEP, and CYP3A4 gene expression, being three important hepatocyte markers.
Collapse
Affiliation(s)
- Nathan Carpentier
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent 9000, Belgium
| | - Shicheng Ye
- Department Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CT, The Netherlands
| | - Maarten D Delemarre
- Department Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CT, The Netherlands
| | - Louis Van der Meeren
- Nano-Biotechnology Laboratory, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent 9000, Belgium
| | - André G Skirtach
- Nano-Biotechnology Laboratory, Department of Biotechnology, Faculty of Bioscience Engineering, Ghent University, Ghent 9000, Belgium
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC-University Medical Center, Rotterdam 3000 CA, The Netherlands
| | - Kerstin Schneeberger
- Department Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CT, The Netherlands
| | - Bart Spee
- Department Clinical Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht 3584 CT, The Netherlands
| | - Peter Dubruel
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent 9000, Belgium
| | - Sandra Van Vlierberghe
- Polymer Chemistry & Biomaterials Group, Centre of Macromolecular Chemistry, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent 9000, Belgium
| |
Collapse
|
16
|
Lee J, Lee H. Sacrificial-Rotating Rod-Based 3D Bioprinting Technique for the Development of an In Vitro Cardiovascular Model. J Funct Biomater 2023; 15:2. [PMID: 38276475 PMCID: PMC10817312 DOI: 10.3390/jfb15010002] [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: 10/26/2023] [Revised: 12/11/2023] [Accepted: 12/16/2023] [Indexed: 01/27/2024] Open
Abstract
Several studies have attempted to develop complex cardiovascular models, but the use of multiple cell types and poor cell alignments after fabrication have limited the practical application of these models. Among various bioprinting methods, extrusion-based bioprinting is the most widely used in the bioengineering field. This method not only has the potential to construct complex 3D biological structures but it also enables the alignment of cells in the printing direction owing to the application of shear stress to the cells during the printing process. Therefore, this study developed an in vitro cardiovascular model using an extrusion-based bioprinting method that utilizes a rotating rod as a printing platform. The rotating rod was made of polyvinyl alcohol (PVA) and used as a sacrificial rod. This rotating platform approach enabled the printing of longer tubular-vascular structures of multiple shapes, including disease models, and the water-soluble properties of PVA facilitated the isolation of the printed vascular models. In addition, this method enabled the printing of the endothelial cells in the bloodstream direction and smooth muscle cells in the circumferential direction to better mimic the anatomy of real blood vessels. Consequently, a cardiovascular model was successfully printed using a gelatin methacryloyl bioink with cells. In conclusion, the proposed fabrication method can facilitate the fabrication of various cardiovascular models that mimic the alignment of real blood vessels.
Collapse
Affiliation(s)
- Jooyoung Lee
- Department of Smart Health Science and Technology, Kangwon National University (KNU), Chuncheon 24341, Republic of Korea;
| | - Hyungseok Lee
- Department of Smart Health Science and Technology, Kangwon National University (KNU), Chuncheon 24341, Republic of Korea;
- Department of Mechanical and Biomedical Engineering, Kangwon National University (KNU), Chuncheon 24341, Republic of Korea
| |
Collapse
|
17
|
Lee DN, Park JY, Seo YW, Jin X, Hong J, Bhattacharyya A, Noh I, Choi SH. Photo-crosslinked gelatin methacryloyl hydrogel strengthened with calcium phosphate-based nanoparticles for early healing of rabbit calvarial defects. J Periodontal Implant Sci 2023; 53:321-335. [PMID: 36919004 PMCID: PMC10627735 DOI: 10.5051/jpis.2203220161] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 10/16/2022] [Accepted: 10/24/2022] [Indexed: 02/05/2023] Open
Abstract
PURPOSE The aim of this study was to investigate the efficacy of photo-crosslinked gelatin methacryloyl (GelMa) hydrogel containing calcium phosphate nanoparticles (CNp) when applying different fabrication methods for bone regeneration. METHODS Four circular defects were created in the calvaria of 10 rabbits. Each defect was randomly allocated to the following study groups: 1) the sham control group, 2) the GelMa group (defect filled with crosslinked GelMa hydrogel), 3) the CNp-GelMa group (GelMa hydrogel crosslinked with nanoparticles), and 4) the CNp+GelMa group (crosslinked GelMa loaded with nanoparticles). At 2, 4, and 8 weeks, samples were harvested, and histological and micro-computed tomography analyses were performed. RESULTS Histomorphometric analysis showed that the CNp-GelMa and CNp+GelMa groups at 2 weeks had significantly greater total augmented areas than the control group (P<0.05). The greatest new bone area was observed in the CNp-GelMa group, but without statistical significance (P>0.05). Crosslinked GelMa hydrogel with nanoparticles exhibited good biocompatibility with a minimal inflammatory reaction. CONCLUSIONS There was no difference in the efficacy of bone regeneration according to the synthesized method of photo-crosslinked GelMa hydrogel with nanoparticles. However, these materials could remain within a bone defect up to 2 weeks and showed good biocompatibility with little inflammatory response. Further improvement in mechanical properties and resistance to enzymatic degradation would be needed for the clinical application.
Collapse
Affiliation(s)
- Da-Na Lee
- Department of Periodontology, Research Institute for Periodontal Regeneration, Yonsei University College of Dentistry, Seoul, Korea
| | - Jin-Young Park
- Department of Periodontology, Research Institute for Periodontal Regeneration, Yonsei University College of Dentistry, Seoul, Korea
- Medical & Dental Devices Usability Test Center, Yonsei University Dental Hospital, Seoul, Korea
| | - Young-Wook Seo
- Department of Periodontology, Research Institute for Periodontal Regeneration, Yonsei University College of Dentistry, Seoul, Korea
| | - Xiang Jin
- Department of Periodontology, Research Institute for Periodontal Regeneration, Yonsei University College of Dentistry, Seoul, Korea
| | - Jongmin Hong
- Department of Chemical and Biomolecular Engineering, Seoul National University of Science and Technology, Seoul, Korea
| | - Amitava Bhattacharyya
- Department of Chemical and Biomolecular Engineering, Seoul National University of Science and Technology, Seoul, Korea
- Convergence Institute of Biomedical Engineering and Biomaterials, Seoul National University of Science and Technology, Seoul, Korea
| | - Insup Noh
- Department of Chemical and Biomolecular Engineering, Seoul National University of Science and Technology, Seoul, Korea
- Convergence Institute of Biomedical Engineering and Biomaterials, Seoul National University of Science and Technology, Seoul, Korea
| | - Seong-Ho Choi
- Department of Periodontology, Research Institute for Periodontal Regeneration, Yonsei University College of Dentistry, Seoul, Korea
- Medical & Dental Devices Usability Test Center, Yonsei University Dental Hospital, Seoul, Korea.
| |
Collapse
|
18
|
Roldan L, Montoya C, Solanki V, Cai KQ, Yang M, Correa S, Orrego S. A Novel Injectable Piezoelectric Hydrogel for Periodontal Disease Treatment. ACS APPLIED MATERIALS & INTERFACES 2023; 15:43441-43454. [PMID: 37672788 DOI: 10.1021/acsami.3c08336] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/08/2023]
Abstract
Periodontal disease is a multifactorial, bacterially induced inflammatory condition characterized by the progressive destruction of periodontal tissues. The successful nonsurgical treatment of periodontitis requires multifunctional technologies offering antibacterial therapies and promotion of bone regeneration simultaneously. For the first time, in this study, an injectable piezoelectric hydrogel (PiezoGEL) was developed after combining gelatin methacryloyl (GelMA) with biocompatible piezoelectric fillers of barium titanate (BTO) that produce electrical charges when stimulated by biomechanical vibrations (e.g., mastication, movements). We harnessed the benefits of hydrogels (injectable, light curable, conforms to pocket spaces, biocompatible) with the bioactive effects of piezoelectric charges. A thorough biomaterial characterization confirmed piezoelectric fillers' successful integration with the hydrogel, photopolymerizability, injectability for clinical use, and electrical charge generation to enable bioactive effects (antibacterial and bone tissue regeneration). PiezoGEL showed significant reductions in pathogenic biofilm biomass (∼41%), metabolic activity (∼75%), and the number of viable cells (∼2-3 log) compared to hydrogels without BTO fillers in vitro. Molecular analysis related the antibacterial effects to be associated with reduced cell adhesion (downregulation of porP and fimA) and increased oxidative stress (upregulation of oxyR) genes. Moreover, PiezoGEL significantly enhanced bone marrow stem cell (BMSC) viability and osteogenic differentiation by upregulating RUNX2, COL1A1, and ALP. In vivo, PiezoGEL effectively reduced periodontal inflammation and increased bone tissue regeneration compared to control groups in a mice model. Findings from this study suggest PiezoGEL to be a promising and novel therapeutic candidate for the treatment of periodontal disease nonsurgically.
Collapse
Affiliation(s)
- Lina Roldan
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, Pennsylvania 19140, United States
- Bioengineering Research Group (GIB), Universidad EAFIT, Medellín 050037, Colombia
| | - Carolina Montoya
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, Pennsylvania 19140, United States
| | - Varun Solanki
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, Pennsylvania 19140, United States
| | - Kathy Q Cai
- Histopathology Facility, Fox Chase Cancer, Temple University, Philadelphia, Pennsylvania 19140, United States
| | - Maobin Yang
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, Pennsylvania 19140, United States
- Department of Endodontology, Kornberg School of Dentistry, Temple University, Philadelphia, Pennsylvania 19140, United States
| | - Santiago Correa
- Bioengineering Research Group (GIB), Universidad EAFIT, Medellín 050037, Colombia
| | - Santiago Orrego
- Department of Oral Health Sciences, Kornberg School of Dentistry, Temple University, Philadelphia, Pennsylvania 19140, United States
- Bioengineering Department, College of Engineering, Temple University. Philadelphia, Pennsylvania 19122, United States
| |
Collapse
|
19
|
Ramey-Ward A, Dong Y, Yang J, Ogasawara H, Bremer-Sai EC, Brazhkina O, Franck C, Davis M, Salaita K. Optomechanically Actuated Hydrogel Platform for Cell Stimulation with Spatial and Temporal Resolution. ACS Biomater Sci Eng 2023; 9:5361-5375. [PMID: 37604774 PMCID: PMC10498418 DOI: 10.1021/acsbiomaterials.3c00516] [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: 04/18/2023] [Accepted: 08/02/2023] [Indexed: 08/23/2023]
Abstract
Cells exist in the body in mechanically dynamic environments, yet the vast majority of in vitro cell culture is conducted on static materials such as plastic dishes and gels. To address this limitation, we report an approach to transition widely used hydrogels into mechanically active substrates by doping optomechanical actuator (OMA) nanoparticles within the polymer matrix. OMAs are composed of gold nanorods surrounded by a thermoresponsive polymer shell that rapidly collapses upon near-infrared (NIR) illumination. As a proof of concept, we crosslinked OMAs into laminin-gelatin hydrogels, generating up to 5 μm deformations triggered by NIR pulsing. This response was tunable by NIR intensity and OMA density within the gel and is generalizable to other hydrogel materials. Hydrogel mechanical stimulation enhanced myogenesis in C2C12 myoblasts as evidenced by ERK signaling, myocyte fusion, and sarcomeric myosin expression. We also demonstrate rescued differentiation in a chronic inflammation model as a result of mechanical stimulation. This work establishes OMA-actuated biomaterials as a powerful tool for in vitro mechanical manipulation with broad applications in the field of mechanobiology.
Collapse
Affiliation(s)
- Allison
N. Ramey-Ward
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, Georgia 30322, United States
| | - Yixiao Dong
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Jin Yang
- Department
of Mechanical Engineering, University of
Wisconsin − Madison, Madison, Wisconsin 53706, United States
| | - Hiroaki Ogasawara
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| | - Elizabeth C. Bremer-Sai
- Department
of Mechanical Engineering, University of
Wisconsin − Madison, Madison, Wisconsin 53706, United States
| | - Olga Brazhkina
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, Georgia 30322, United States
| | - Christian Franck
- Department
of Mechanical Engineering, University of
Wisconsin − Madison, Madison, Wisconsin 53706, United States
| | - Michael Davis
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, Georgia 30322, United States
| | - Khalid Salaita
- Wallace
H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, Georgia 30322, United States
- Department
of Chemistry, Emory University, Atlanta, Georgia 30322, United States
| |
Collapse
|
20
|
Zhang J, Suttapreyasri S, Leethanakul C, Samruajbenjakun B. Triaxial bioprinting large-size vascularized constructs with nutrient channels. Biomed Mater 2023; 18:055026. [PMID: 37604152 DOI: 10.1088/1748-605x/acf25a] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Accepted: 08/21/2023] [Indexed: 08/23/2023]
Abstract
Bioprinting has demonstrated great advantages in tissue and organ regeneration. However, constructing large-scale tissue and organsin vitrois still a huge challenge due to the lack of some strategies for loading multiple types of cells precisely while maintaining nutrient channels. Here, a new 3D bioprinting strategy was proposed to construct large-scale vascularized tissue. A mixture of gelatin methacrylate (GelMA) and sodium alginate (Alg) was used as a bioink, serving as the outer and middle layers of a single filament in the triaxial printing process, and loaded with human bone marrow mesenchymal stem cells and human umbilical vein endothelial cells, respectively, while a calcium chloride (CaCl2) solution was used as the inner layer. The CaCl2solution crosslinked with the middle layer bioink during the printing process to form and maintain hollow nutrient channels, then a stable large-scale construct was obtained through photopolymerization and ion crosslinking after printing. The feasibility of this strategy was verified by investigating the properties of the bioink and construct, and the biological performance of the vascularized construct. The results showed that a mixture of 5% (w/v) GelMA and 1% (w/v) Alg bioink could be printed at room temperature with good printability and perfusion capacity. Then, the construct with and without channels was fabricated and characterized, and the results revealed that the construct with channels had a similar degradation profile to that without channels, but lower compressive modulus and higher swelling rate. Biological investigation showed that the construct with channels was more favorable for cell survival, proliferation, diffusion, migration, and vascular network formation. In summary, it was demonstrated that constructing large-scale vascularized tissue by triaxial printing that can precisely encapsulate multiple types of cells and form nutrient channels simultaneously was feasible, and this technology could be used to prepare large-scale vascularized constructs.
Collapse
Affiliation(s)
- Junbiao Zhang
- Orthodontic Section, Department of Preventive Dentistry, Faculty of Dentistry, Prince of Songkla University, Hat Yai 90112, Songkhla, Thailand
- Guiyang Hospital of Stomatology, Guiyang 550002, People's Republic of China
| | - Srisurang Suttapreyasri
- Department of Oral and Maxillofacial Surgery, Faculty of Dentistry, Prince of Songkla University, Hat Yai 90112, Thailand
| | - Chidchanok Leethanakul
- Orthodontic Section, Department of Preventive Dentistry, Faculty of Dentistry, Prince of Songkla University, Hat Yai 90112, Songkhla, Thailand
| | - Bancha Samruajbenjakun
- Orthodontic Section, Department of Preventive Dentistry, Faculty of Dentistry, Prince of Songkla University, Hat Yai 90112, Songkhla, Thailand
| |
Collapse
|
21
|
Martyniak K, Kennedy S, Karimzadeh M, Cruz MA, Jeon O, Alsberg E, Kean TJ. Optimizing Bioink Composition for Human Chondrocyte Expression of Lubricin. Bioengineering (Basel) 2023; 10:997. [PMID: 37760099 PMCID: PMC10526043 DOI: 10.3390/bioengineering10090997] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2023] [Revised: 07/31/2023] [Accepted: 08/03/2023] [Indexed: 09/29/2023] Open
Abstract
The surface zone of articular cartilage is the first area impacted by cartilage defects, commonly resulting in osteoarthritis. Chondrocytes in the surface zone of articular cartilage synthesize and secrete lubricin, a proteoglycan that functions as a lubricant protecting the deeper layers from shear stress. Notably, 3D bioprinting is a tissue engineering technique that uses cells encapsulated in biomaterials to fabricate 3D constructs. Gelatin methacrylate (GelMA) is a frequently used biomaterial for 3D bioprinting cartilage. Oxidized methacrylated alginate (OMA) is a chemically modified alginate designed for its tunable degradation rate and mechanical properties. To determine an optimal combination of GelMA and OMA for lubricin expression, we used our novel high-throughput human articular chondrocyte reporter system. Primary human chondrocytes were transduced with PRG4 (lubricin) promoter-driven Gaussia luciferase, allowing for temporal assessment of lubricin expression. A lubricin expression-driven Design of Experiment screen and subsequent validation identified 14% GelMA/2% OMA for further study. Therefore, DoE optimized 14% GelMA/2% OMA, 14% GelMA control, and 16% GelMA (total solid content control) were 3D bioprinted. The combination of lubricin protein expression and shape retention over the 22 days in culture, successfully determined the 14% GelMA/2%OMA to be the optimal formulation for lubricin secretion. This strategy allows for rapid analysis of the role(s) of biomaterial composition, stiffness or other cell manipulations on lubricin expression by chondrocytes, which may improve therapeutic strategies for cartilage regeneration.
Collapse
Affiliation(s)
- Kari Martyniak
- Biionix Cluster, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Sean Kennedy
- Biionix Cluster, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Makan Karimzadeh
- Biionix Cluster, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Maria A. Cruz
- Biionix Cluster, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| | - Oju Jeon
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL 60607, USA; (O.J.); (E.A.)
| | - Eben Alsberg
- Department of Biomedical Engineering, University of Illinois Chicago, Chicago, IL 60607, USA; (O.J.); (E.A.)
| | - Thomas J. Kean
- Biionix Cluster, College of Medicine, University of Central Florida, Orlando, FL 32827, USA
| |
Collapse
|
22
|
Lin S, Maekawa H, Moeinzadeh S, Lui E, Alizadeh HV, Li J, Kim S, Poland M, Gadomski BC, Easley JT, Young J, Gardner M, Mohler D, Maloney WJ, Yang YP. An osteoinductive and biodegradable intramedullary implant accelerates bone healing and mitigates complications of bone transport in male rats. Nat Commun 2023; 14:4455. [PMID: 37488113 PMCID: PMC10366099 DOI: 10.1038/s41467-023-40149-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Accepted: 07/12/2023] [Indexed: 07/26/2023] Open
Abstract
Bone transport is a surgery-driven procedure for the treatment of large bone defects. However, challenging complications include prolonged consolidation, docking site nonunion and pin tract infection. Here, we develop an osteoinductive and biodegradable intramedullary implant by a hybrid tissue engineering construct technique to enable sustained delivery of bone morphogenetic protein-2 as an adjunctive therapy. In a male rat bone transport model, the eluting bone morphogenetic protein-2 from the implants accelerates bone formation and remodeling, leading to early bony fusion as shown by imaging, mechanical testing, histological analysis, and microarray assays. Moreover, no pin tract infection but tight osseointegration are observed. In contrast, conventional treatments show higher proportion of docking site nonunion and pin tract infection. The findings of this study demonstrate that the novel intramedullary implant holds great promise for advancing bone transport techniques by promoting bone regeneration and reducing complications in the treatment of bone defects.
Collapse
Affiliation(s)
- Sien Lin
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Hirotsugu Maekawa
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Seyedsina Moeinzadeh
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Elaine Lui
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA, 94305, USA
- Department of Mechanical Engineering, School of Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Hossein Vahid Alizadeh
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Jiannan Li
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Sungwoo Kim
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Michael Poland
- Orthopaedic Bioengineering Research Laboratory, Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, 80523, USA
| | - Benjamin C Gadomski
- Orthopaedic Bioengineering Research Laboratory, Department of Mechanical Engineering, Colorado State University, Fort Collins, CO, 80523, USA
| | - Jeremiah T Easley
- Preclinical Surgical Research Laboratory, Department of Clinical Sciences, Colorado State University, Fort Collins, CO, 80523, USA
| | - Jeffrey Young
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Michael Gardner
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - David Mohler
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - William J Maloney
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Yunzhi Peter Yang
- Department of Orthopaedic Surgery, School of Medicine, Stanford University, Stanford, CA, 94305, USA.
- Department of Materials Science and Engineering, School of Engineering, Stanford University, Stanford, CA, 94305, USA.
- Department of Bioengineering, School of Medicine, Stanford University, Stanford, CA, 94305, USA.
| |
Collapse
|
23
|
Asadi N, Sadeghzadeh H, Rahmani Del Bakhshayesh A, Nezami Asl A, Dadashpour M, Karimi Hajishoreh N, Kaamyabi S, Akbarzadeh A. Preparation and characterization of propolis reinforced eggshell membrane/ GelMA composite hydrogel for biomedical applications. BMC Biotechnol 2023; 23:21. [PMID: 37434201 DOI: 10.1186/s12896-023-00788-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2023] [Accepted: 06/15/2023] [Indexed: 07/13/2023] Open
Abstract
Gelatin methacrylate-based hydrogels (GelMA) were widely used in tissue engineering and regenerative medicine. However, to manipulate their various chemical and physical properties and create high-efficiency hydrogels, different materials have been used in their structure. Eggshell membrane (ESM) and propolis are two nature-derived materials that could be used to improve the various characteristics of hydrogels, especially structural and biological properties. Hence, the main purpose of this study is the development of a new type of GelMA hydrogel containing ESM and propolis, for use in regenerative medicine. In this regard, in this study, after synthesizing GelMA, the fragmented ESM fibers were added to it and the GM/EMF hydrogel was made using a photoinitiator and visible light irradiation. Finally, GM/EMF/P hydrogels were prepared by incubating GM/EMF hydrogels in the propolis solution for 24 h. After various structural, chemical, and biological characterizations, it was found that the hydrogels obtained in this study offer improved morphological, hydrophilic, thermal, mechanical, and biological properties. The developed GM/EMF/P hydrogel presented more porosity with smaller and interconnected pores compared to the other hydrogels. GM/EMF hydrogels due to possessing EMF showed compressive strength up to 25.95 ± 1.69 KPa, which is more than the compressive strength provided by GM hydrogels (24.550 ± 4.3 KPa). Also, GM/EMF/P hydrogel offered the best compressive strength (44.65 ± 3.48) due to the presence of both EMF and propolis. GM scaffold with a contact angle of about 65.41 ± 2.199 θ showed more hydrophobicity compared to GM/EMF (28.67 ± 1.58 θ), and GM/EMF/P (26.24 ± 0.73 θ) hydrogels. Also, the higher swelling percentage of GM/EMF/P hydrogels (343.197 ± 42.79) indicated the high capacity of this hydrogel to retain more water than other scaffolds. Regarding the biocompatibility of the fabricated structures, MTT assay results showed that GM/EMF/P hydrogel significantly (p-value < 0.05) supported cell viability. Based on the results, it seems that GM/EMF/P hydrogel could be a promising biomaterial candidate for use in various fields of regenerative medicine.
Collapse
Affiliation(s)
- Nahideh Asadi
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Hadi Sadeghzadeh
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Azizeh Rahmani Del Bakhshayesh
- Department of Tissue Engineering, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Mehdi Dadashpour
- Department of Medical Biotechnology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | | | - Sharif Kaamyabi
- Department of Chemistry, Farhangian University, Tehran, Iran
| | - Abolfazl Akbarzadeh
- Department of Medical Nanotechnology, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
24
|
Schulik J, Salehi S, Boccaccini AR, Schrüfer S, Schubert DW, Arkudas A, Kengelbach-Weigand A, Horch RE, Schmid R. Comparison of the Behavior of 3D-Printed Endothelial Cells in Different Bioinks. Bioengineering (Basel) 2023; 10:751. [PMID: 37508778 PMCID: PMC10376299 DOI: 10.3390/bioengineering10070751] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2023] [Revised: 06/19/2023] [Accepted: 06/21/2023] [Indexed: 07/30/2023] Open
Abstract
Biomaterials with characteristics similar to extracellular matrix and with suitable bioprinting properties are essential for vascular tissue engineering. In search for suitable biomaterials, this study investigated the three hydrogels alginate/hyaluronic acid/gelatin (Alg/HA/Gel), pre-crosslinked alginate di-aldehyde with gelatin (ADA-GEL), and gelatin methacryloyl (GelMA) with respect to their mechanical properties and to the survival, migration, and proliferation of human umbilical vein endothelial cells (HUVECs). In addition, the behavior of HUVECs was compared with their behavior in Matrigel. For this purpose, HUVECs were mixed with the inks both as single cells and as cell spheroids and printed using extrusion-based bioprinting. Good printability with shape fidelity was determined for all inks. The rheological measurements demonstrated the gelling consistency of the inks and shear-thinning behavior. Different Young's moduli of the hydrogels were determined. However, all measured values where within the range defined in the literature, leading to migration and sprouting, as well as reconciling migration with adhesion. Cell survival and proliferation in ADA-GEL and GelMA hydrogels were demonstrated for 14 days. In the Alg/HA/Gel bioink, cell death occurred within 7 days for single cells. Sprouting and migration of the HUVEC spheroids were observed in ADA-GEL and GelMA. Similar behavior of the spheroids was seen in Matrigel. In contrast, the spheroids in the Alg/HA/Gel ink died over the time studied. It has been shown that Alg/HA/Gel does not provide a good environment for long-term survival of HUVECs. In conclusion, ADA-GEL and GelMA are promising inks for vascular tissue engineering.
Collapse
Affiliation(s)
- Jana Schulik
- Laboratory for Tissue-Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery University Hospital of Erlangen, Krankenhausstraße 12, 91054 Erlangen, Germany
| | - Sahar Salehi
- Chair of Biomaterials, University of Bayreuth, Prof.-Rüdiger-Bormann-Str. 1, 95447 Bayreuth, Germany
| | - Aldo R Boccaccini
- Institute of Biomaterials, Friedrich-Alexander University Erlangen-Nürnberg, Cauerstrasse 6, 91058 Erlangen, Germany
| | - Stefan Schrüfer
- Institute of Polymer Materials, Friedrich-Alexander University Erlangen-Nürnberg, Martensstraße 7, 91058 Erlangen, Germany
| | - Dirk W Schubert
- Institute of Polymer Materials, Friedrich-Alexander University Erlangen-Nürnberg, Martensstraße 7, 91058 Erlangen, Germany
| | - Andreas Arkudas
- Laboratory for Tissue-Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery University Hospital of Erlangen, Krankenhausstraße 12, 91054 Erlangen, Germany
| | - Annika Kengelbach-Weigand
- Laboratory for Tissue-Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery University Hospital of Erlangen, Krankenhausstraße 12, 91054 Erlangen, Germany
| | - Raymund E Horch
- Laboratory for Tissue-Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery University Hospital of Erlangen, Krankenhausstraße 12, 91054 Erlangen, Germany
| | - Rafael Schmid
- Laboratory for Tissue-Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery University Hospital of Erlangen, Krankenhausstraße 12, 91054 Erlangen, Germany
| |
Collapse
|
25
|
Dubey N, Ribeiro JS, Zhang Z, Xu J, Ferreira JA, Qu L, Mei L, Fenno JC, Schwendeman A, Schwendeman SP, Nör JE, Bottino MC. Gelatin methacryloyl hydrogel as an injectable scaffold with multi-therapeutic effects to promote antimicrobial disinfection and angiogenesis for regenerative endodontics. J Mater Chem B 2023; 11:3823-3835. [PMID: 36946228 PMCID: PMC10160005 DOI: 10.1039/d2tb02788g] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2023]
Abstract
Regenerative endodontics represents a paradigm shift in dental pulp therapy for necrotic young permanent teeth. However, there are still challenges associated with attaining maximum root canal disinfection while supporting angiogenesis and preserving resident stem cells viability and differentiation capacity. Here, we developed a hydrogel system by incorporating antibiotic-eluting fiber-based microparticles in gelatin methacryloyl (GelMA) hydrogel to gather antimicrobial and angiogenic properties while prompting minimum cell toxicity. Minocycline (MINO) or clindamycin (CLIN) was introduced into a polymer solution and electrospun into fibers, which were further cryomilled to attain MINO- or CLIN-eluting fibrous microparticles. To obtain hydrogels with multi-therapeutic effects, MINO- or CLIN-eluting microparticles were suspended in GelMA at distinct concentrations. The engineered hydrogels demonstrated antibiotic-dependent swelling and degradability while inhibiting bacterial growth with minimum toxicity in dental-derived stem cells. Notably, compared to MINO, CLIN hydrogels enhanced the formation of capillary-like networks of endothelial cells in vitro and the presence of widespread vascularization with functioning blood vessels in vivo. Our data shed new light onto the clinical potential of antibiotic-eluting gelatin methacryloyl hydrogel as an injectable scaffold with multi-therapeutic effects to promote antimicrobial disinfection and angiogenesis for regenerative endodontics.
Collapse
Affiliation(s)
- Nileshkumar Dubey
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, Michigan, USA.
- Faculty of Dentistry, National University of Singapore, Singapore
| | - Juliana S Ribeiro
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, Michigan, USA.
- Department of Dentistry, Federal University of Santa Catarina, Florianópolis, SC, Brazil
| | - Zhaocheng Zhang
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, Michigan, USA.
| | - Jinping Xu
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, Michigan, USA.
| | - Jessica A Ferreira
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, Michigan, USA.
| | - Liu Qu
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, Michigan, USA.
- Department of Endodontics, School and Hospital of Stomatology, China Medical University, Liaoning Provincial Key Laboratory of Oral Diseases, Shenyang, China
| | - Ling Mei
- Department of Pharmaceutical Sciences and the Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - J Christopher Fenno
- Department of Biologic and Materials Sciences & Prosthodontics, University of Michigan School of Dentistry, Ann Arbor, Michigan, USA
| | - Anna Schwendeman
- Department of Pharmaceutical Sciences and the Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Steven P Schwendeman
- Department of Pharmaceutical Sciences and the Biointerfaces Institute, University of Michigan, Ann Arbor, Michigan, USA
| | - Jacques E Nör
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, Michigan, USA.
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Marco C Bottino
- Department of Cariology, Restorative Sciences, and Endodontics, School of Dentistry, University of Michigan, Ann Arbor, Michigan, USA.
- Department of Biomedical Engineering, College of Engineering, University of Michigan, Ann Arbor, Michigan, USA
| |
Collapse
|
26
|
Properties and Printability of the Synthesized Hydrogel Based on GelMA. Int J Mol Sci 2023; 24:ijms24032121. [PMID: 36768446 PMCID: PMC9917366 DOI: 10.3390/ijms24032121] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2022] [Revised: 01/14/2023] [Accepted: 01/18/2023] [Indexed: 01/25/2023] Open
Abstract
Gelatin methacryloyl (GelMA) has recently attracted increasing attention. Unlike other hydrogels, it allows for the adjustment of the mechanical properties using such factors as degree of functionalization, concentration, and photocrosslinking parameters. In this study, GelMA with a high degree of substitution (82.75 ± 7.09%) was synthesized, and its suitability for extrusion printing, cytocompatibility, and biocompatibility was studied. Satisfactory printing quality was demonstrated with the 15% concentration hydrogel. The high degree of functionalization led to a decrease in the ability of human adipose-derived stem cells (ADSCs) to adhere to the GelMA surface. During the first 3 days after sowing, proliferation was observed. Degradation in animals after subcutaneous implantation was slowed down.
Collapse
|
27
|
The effect of culture conditions on the bone regeneration potential of osteoblast-laden 3D bioprinted constructs. Acta Biomater 2023; 156:190-201. [PMID: 36155098 DOI: 10.1016/j.actbio.2022.09.042] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 09/15/2022] [Accepted: 09/16/2022] [Indexed: 01/18/2023]
Abstract
Three Dimensional (3D) bioprinting is one of the most recent additive manufacturing technologies and enables the direct incorporation of cells within a highly porous 3D-bioprinted construct. While the field has mainly focused on developing methods for enhancing printing resolution and shape fidelity, little is understood about the biological impact of bioprinting on cells. To address this shortcoming, this study investigated the in vitro and in vivo response of human osteoblasts subsequent to bioprinting using gelatin methacryloyl (GelMA) as the hydrogel precursor. First, bioprinted and two-dimensional (2D) cultured osteoblasts were compared, demonstrating that the 3D microenvironment from bioprinting enhanced bone-related gene expression. Second, differentiation regimens of 2-week osteogenic pre-induction in 2D before bioprinting and/or 3-week post-printing osteogenic differentiation were assessed for their capacity to increase the bioprinted construct's biofunctionality towards bone regeneration. The combination of pre-and post-induction regimens showed superior osteogenic gene expression and mineralisation in vitro. Moreover, a rat calvarial model using microtomography and histology demonstrated bone regeneration potential for the pre-and post-differentiation procedure. This study shows the positive impact of bioprinting on cells for osteogenic differentiation and the increased in vivo osteogenic potential of bioprinted constructs via a pre-induction method. STATEMENT OF SIGNIFICANCE: 3D bioprinting, one of the most recent technologies for tissue engineering has mostly focussed on developing methods for enhancing printing properties, little is understood on the biological impact of bioprinting and /or subsequent in vitro maturation methods on cells. Therefore, we addressed these fundamental questions by investigating osteoblast gene expression in bioprinted construct and assessed the efficacy of several induction regimen towards osteogenic differentiation in vitro and in vivo. Osteogenic induction of cells prior to seeding in scaffolds used in conventional tissue engineering applications has been demonstrated to increase the osteogenic potential of the resulting construct. However, to the best of our knowledge, pre-induction methods have not been investigated in 3D bioprinting.
Collapse
|
28
|
Sun H, Xu J, Wang Y, Shen S, Xu X, Zhang L, Jiang Q. Bone microenvironment regulative hydrogels with ROS scavenging and prolonged oxygen-generating for enhancing bone repair. Bioact Mater 2023; 24:477-496. [PMID: 36714330 PMCID: PMC9843284 DOI: 10.1016/j.bioactmat.2022.12.021] [Citation(s) in RCA: 64] [Impact Index Per Article: 32.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/21/2022] [Accepted: 12/21/2022] [Indexed: 01/11/2023] Open
Abstract
Large bone defects resulting from fractures and disease are a major clinical challenge, being often unable to heal spontaneously by the body's repair mechanisms. Lines of evidence have shown that hypoxia-induced overproduction of ROS in bone defect region has a major impact on delaying bone regeneration. However, replenishing excess oxygen in a short time cause high oxygen tension that affect the activity of osteoblast precursor cells. Therefore, reasonably restoring the hypoxic condition of bone microenvironment is essential for facilitating bone repair. Herein, we designed ROS scavenging and responsive prolonged oxygen-generating hydrogels (CPP-L/GelMA) as a "bone microenvironment regulative hydrogel" to reverse the hypoxic microenvironment in bone defects region. CPP-L/GelMA hydrogels comprises an antioxidant enzyme catalase (CAT) and ROS-responsive oxygen-releasing nanoparticles (PFC@PLGA/PPS) co-loaded liposome (CCP-L) and GelMA hydrogels. Under hypoxic condition, CPP-L/GelMA can release CAT for degrading hydrogen peroxide to generate oxygen and be triggered by superfluous ROS to continuously release the oxygen for more than 2 weeks. The prolonged oxygen enriched microenvironment generated by CPP-L/GelMA hydrogel significantly enhanced angiogenesis and osteogenesis while inhibited osteoclastogenesis. Finally, CPP-L/GelMA showed excellent bone regeneration effect in a mice skull defect model through the Nrf2-BMAL1-autophagy pathway. Hence, CPP-L/GelMA, as a bone microenvironment regulative hydrogel for bone tissue respiration, can effectively scavenge ROS and provide prolonged oxygen supply according to the demand in bone defect region, possessing of great clinical therapeutic potential.
Collapse
Key Words
- Alizarin red staining, ARS
- Alkaline phosphatase, ALP
- Bone defect
- Bone marrow mesenchymal stem cells, BMSC
- Bovine serum albumin, BSA
- Brain and muscle arnt-like protein 1
- Brain and muscle arnt-like protein 1, BMAL1
- Catalase, CAT
- Fetal liver kinase-1, Flk-1
- Human umbilical vein endothelial cells, HUVEC
- Hypoxic microenvironment
- Liposome, Lip
- Microtubule-associated proteins light chain 3, LC3
- Nuclear factor (erythroid-derived 2)-like 2, NRF2
- Osteocalcin, OCN
- Osteopontin, OPN
- Perfluorocarbon, PFC
- Phosphate-buffered saline, PBS
- Poly (D, L-lactide-co-glycolide), PLGA
- Poly (propylene sulphide), PPS
- Prolonged oxygen generation
- Reactive oxygen species responsiveness
- Reactive oxygen species, ROS
- Receptor activator of nuclear factor-kappa B ligand, RANKL
- Runt-related transcription factor 2, RUNX2
- Short interfering RNA, siRNA
- Soy phosphatidylcholine, SPC
- Type I collagen, Col I
- Western blot, WB
Collapse
Affiliation(s)
- Han Sun
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China,Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China,Co-innovation Center of Neuroregeneration, Nantong University, 9 Seyuan Road, Nantong, 226019, Jiangsu, PR China,Articular Orthopaedics, The Third Affiliated Hospital of Soochow University, 185 Juqian Road, Changzhou, 213003, Jiangsu, PR China
| | - Juan Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China,Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China,Co-innovation Center of Neuroregeneration, Nantong University, 9 Seyuan Road, Nantong, 226019, Jiangsu, PR China
| | - Yangyufan Wang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China,Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China,Co-innovation Center of Neuroregeneration, Nantong University, 9 Seyuan Road, Nantong, 226019, Jiangsu, PR China
| | - Siyu Shen
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China,Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China,Co-innovation Center of Neuroregeneration, Nantong University, 9 Seyuan Road, Nantong, 226019, Jiangsu, PR China
| | - Xingquan Xu
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China,Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China,Co-innovation Center of Neuroregeneration, Nantong University, 9 Seyuan Road, Nantong, 226019, Jiangsu, PR China,Corresponding author. State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China.
| | - Lei Zhang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China,Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China,Co-innovation Center of Neuroregeneration, Nantong University, 9 Seyuan Road, Nantong, 226019, Jiangsu, PR China,Corresponding author. State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China.
| | - Qing Jiang
- State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China,Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, PR China,Co-innovation Center of Neuroregeneration, Nantong University, 9 Seyuan Road, Nantong, 226019, Jiangsu, PR China,Corresponding author. State Key Laboratory of Pharmaceutical Biotechnology, Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, 321 Zhongshan Road, Nanjing, 210008, Jiangsu, PR China.
| |
Collapse
|
29
|
A Novel Window into Angiogenesis-Intravital Microscopy in the AV-Loop-Model. Cells 2023; 12:cells12020261. [PMID: 36672196 PMCID: PMC9857023 DOI: 10.3390/cells12020261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2022] [Revised: 01/04/2023] [Accepted: 01/06/2023] [Indexed: 01/11/2023] Open
Abstract
Due to the limitations of current in vivo experimental designs, our comprehensive knowledge of vascular development and its implications for the development of large-scale engineered tissue constructs is very limited. Therefore, the purpose of this study was to develop unique in vivo imaging chambers that allow the live visualization of cellular processes in the arteriovenous (AV) loop model in rats. We have developed two different types of chambers. Chamber A is installed in the skin using the purse sting fixing method, while chamber B is installed subcutaneously under the skin. Both chambers are filled with modified gelatin hydrogel as a matrix. Intravital microscopy (IVM) was performed after the injection of fluorescein isothiocyanate (FITC)-labeled dextran and rhodamine 6G dye. The AV loop was functional for two weeks in chamber A and allowed visualization of the leukocyte trafficking. In chamber B, microvascular development in the AV loop could be examined for 21 days. Quantification of the microvascular outgrowth was performed using Fiji-ImageJ. Overall, by combining these two IVM chambers, we can comprehensively understand vascular development in the AV loop tissue engineering model¯.
Collapse
|
30
|
Pazarçeviren AE, Evis Z, Dikmen T, Altunbaş K, Yaprakçı MV, Keskin D, Tezcaner A. Alginate/gelatin/boron-doped hydroxyapatite-coated Ti implants: in vitro and in vivo evaluation of osseointegration. Biodes Manuf 2023. [DOI: 10.1007/s42242-022-00218-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
|
31
|
Xiong Y, Xu Y, Zhou F, Hu Y, Zhao J, Liu Z, Zhai Q, Qi S, Zhang Z, Chen L. Bio-functional hydrogel with antibacterial and anti-inflammatory dual properties to combat with burn wound infection. Bioeng Transl Med 2023; 8:e10373. [PMID: 36684072 PMCID: PMC9842067 DOI: 10.1002/btm2.10373] [Citation(s) in RCA: 32] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2022] [Revised: 05/07/2022] [Accepted: 06/12/2022] [Indexed: 01/25/2023] Open
Abstract
Burn infection delays wound healing and increases the burn patient mortality. Consequently, a new dressing with antibacterial and anti-inflammatory dual properties is urgently required for wound healing. In this study, we propose a combination of methacrylate gelatin (GelMA) hydrogel system with silver nanoparticles embed in γ-cyclodextrin metal-organic frameworks (Ag@MOF) and hyaluronic acid-epigallocatechin gallate (HA-E) for the burn wound infection treatment. Ag@MOF is used as an antibacterial agent and epigallocatechin gallate (EGCG) has exhibited biological properties of anti-inflammation and antibacterial. The GelMA/HA-E/Ag@MOF hydrogel enjoys suitable physical properties and sustained release of Ag+. Meanwhile, the hydrogel has excellent biocompatibility and could promote macrophage polarization from M1 to M2. In vivo wound healing evaluations further demonstrate that the GelMA/HA-E/Ag@MOF hydrogel reduces the number of the bacterium efficiently, accelerates wound healing, promotes early angiogenesis, and regulates immune reaction. A further evaluation indicates that the noncanonical Wnt signal pathway is significantly activated in the GelMA/HA-E/Ag@MOF hydrogel treated group. In conclusion, the GelMA/HA-E/Ag@MOF hydrogel could serve as a promising multifunctional dressing for the burn wound healing.
Collapse
Affiliation(s)
- Yahui Xiong
- Department of Burns, Laboratory of General SurgeryThe First Affiliated Hospital, SunYat‐Sen UniversityGuangzhouChina
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of ProductsSunYat‐Sen UniversityGuangzhouChina
- Institute of Precision MedicineThe First Affiliated Hospital, SunYat‐Sen UniversityGuangzhouChina
| | - Yingbin Xu
- Department of Burns, Laboratory of General SurgeryThe First Affiliated Hospital, SunYat‐Sen UniversityGuangzhouChina
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of ProductsSunYat‐Sen UniversityGuangzhouChina
- Institute of Precision MedicineThe First Affiliated Hospital, SunYat‐Sen UniversityGuangzhouChina
| | - Fei Zhou
- Department of Burns, Laboratory of General SurgeryThe First Affiliated Hospital, SunYat‐Sen UniversityGuangzhouChina
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of ProductsSunYat‐Sen UniversityGuangzhouChina
- Institute of Precision MedicineThe First Affiliated Hospital, SunYat‐Sen UniversityGuangzhouChina
| | - Yanke Hu
- Department of Burns, Laboratory of General SurgeryThe First Affiliated Hospital, SunYat‐Sen UniversityGuangzhouChina
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of ProductsSunYat‐Sen UniversityGuangzhouChina
- Institute of Precision MedicineThe First Affiliated Hospital, SunYat‐Sen UniversityGuangzhouChina
| | - Jingling Zhao
- Department of Burns, Laboratory of General SurgeryThe First Affiliated Hospital, SunYat‐Sen UniversityGuangzhouChina
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of ProductsSunYat‐Sen UniversityGuangzhouChina
- Institute of Precision MedicineThe First Affiliated Hospital, SunYat‐Sen UniversityGuangzhouChina
| | - Zhonghua Liu
- South China Agricultural UniversityGuangzhouChina
| | - Qiyi Zhai
- ZhuJiang HospitalSouthern Medical UniversityGuangzhouChina
| | - Shaohai Qi
- Department of Burns, Laboratory of General SurgeryThe First Affiliated Hospital, SunYat‐Sen UniversityGuangzhouChina
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of ProductsSunYat‐Sen UniversityGuangzhouChina
- Institute of Precision MedicineThe First Affiliated Hospital, SunYat‐Sen UniversityGuangzhouChina
| | - Zhaoqiang Zhang
- Department of Oral and Maxillofacial SurgeryStomatological Hospital, Southern Medical UniversityGuangzhouChina
| | - Lei Chen
- Department of Burns, Laboratory of General SurgeryThe First Affiliated Hospital, SunYat‐Sen UniversityGuangzhouChina
- Guangdong Provincial Engineering Technology Research Center of Burn and Wound Accurate Diagnosis and Treatment Key Technology and Series of ProductsSunYat‐Sen UniversityGuangzhouChina
- Institute of Precision MedicineThe First Affiliated Hospital, SunYat‐Sen UniversityGuangzhouChina
| |
Collapse
|
32
|
Vascularization of Poly-ε-Caprolactone-Collagen I-Nanofibers with or without Sacrificial Fibers in the Neurotized Arteriovenous Loop Model. Cells 2022; 11:cells11233774. [PMID: 36497034 PMCID: PMC9736129 DOI: 10.3390/cells11233774] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2022] [Revised: 11/18/2022] [Accepted: 11/22/2022] [Indexed: 11/29/2022] Open
Abstract
Electrospun nanofibers represent an ideal matrix for the purpose of skeletal muscle tissue engineering due to their highly aligned structure in the nanoscale, mimicking the extracellular matrix of skeletal muscle. However, they often consist of high-density packed fibers, which might impair vascularization. The integration of polyethylene oxide (PEO) sacrificial fibers, which dissolve in water, enables the creation of less dense structures. This study examines potential benefits of poly-ε-caprolactone-collagen I-PEO-nanoscaffolds (PCP) in terms of neovascularization and distribution of newly formed vessels compared to poly-ε-caprolactone -collagen I-nanoscaffolds (PC) in a modified arteriovenous loop model in the rat. For this purpose, the superficial inferior epigastric artery and vein as well as a motor nerve branch were integrated into a multilayer three-dimensional nanofiber scaffold construct, which was enclosed by an isolation chamber. Numbers and spatial distribution of sprouting vessels as well as macrophages were analyzed via immunohistochemistry after two and four weeks of implantation. After four weeks, aligned PC showed a higher number of newly formed vessels, regardless of the compartments formed in PCP by the removal of sacrificial fibers. Both groups showed cell influx and no difference in macrophage invasion. In this study, a model of combined axial vascularization and neurotization of a PCL-collagen I-nanofiber construct could be established for the first time. These results provide a foundation for the in vivo implantation of cells, taking a major step towards the generation of functional skeletal muscle tissue.
Collapse
|
33
|
Stumpfe MC, Horch RE, Arkudas A, Cai A, Müller-Seubert W, Hauck T, Ludolph I. The Value of Negative-Pressure Wound Therapy and Flap Surgery in Hidradenitis Suppurativa – A Single Center Analysis of Different Treatment Options. Front Surg 2022; 9:867487. [PMID: 35836613 PMCID: PMC9273893 DOI: 10.3389/fsurg.2022.867487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2022] [Accepted: 05/31/2022] [Indexed: 11/30/2022] Open
Abstract
Background Hidradenitis suppurativa is manifested by painful abscesses and scarring of sweat glands. Axillary, inguinal and genital regions are mostly affected. Multiple options exist in the treatment of hidradenitis suppurativa. The aim of this retrospective, mono-center cohort study was to analyze the outcome of different treatment methods after radical excision of hidradenitis suppurativa. Methods We retrospectively evaluated the treatment strategy and recurrence rate of hidradenitis suppurativa. We included all eligible patients of legal age between February 2003 and October 2021, with the diagnosis of Hidradenitis suppurativa and the necessity for surgical treatment. All patients with surgical treatment and direct wound closure by suture were excluded. Bacterial load and flora were analyzed for primary and secondary reconstruction in combination with negative-pressure wound therapy. Patient data were analyzed for recurrence rate and remission time according to different reconstructive techniques. Results In 44 affected anatomical sites (n = 23 patients) we treated 15 patients with negative-pressure wound therapy. Bacterial load and flora were lower in the last wound swab of patients with multi-surgical procedures (22 localizations) compared to the first wound swab independent of the use of negative-pressure wound therapy. Wound closure, independent of a direct and multi-stage procedure was achieved by local fasciocutaneous flaps (n = 12), secondary intention healing (n = 7), secondary intention healing with buried chip skin grafts (n = 10), or split-thickness skin grafts (n = 15). Radical excision combined with split-thickness skin grafts showed the lowest recurrence rate in the follow-up (16%; n = 4). Conclusion Radical excision of hidradenitis suppurativa as gold standard for surgical treatment combined with negative-pressure wound therapy as multi-stage procedures ultimately reduced bacterial load and flora in our study. The use of split-thickness skin grafts showed the lowest recurrence rate.
Collapse
|
34
|
Steiner D, Reinhardt L, Fischer L, Popp V, Körner C, Geppert CI, Bäuerle T, Horch RE, Arkudas A. Impact of Endothelial Progenitor Cells in the Vascularization of Osteogenic Scaffolds. Cells 2022; 11:cells11060926. [PMID: 35326377 PMCID: PMC8946714 DOI: 10.3390/cells11060926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 03/01/2022] [Accepted: 03/04/2022] [Indexed: 02/06/2023] Open
Abstract
The microvascular endothelial network plays an important role in osteogenesis, bone regeneration and bone tissue engineering. Endothelial progenitor cells (EPCs) display a high angiogenic and vasculogenic potential. The endothelialization of scaffolds with endothelial progenitor cells supports vascularization and tissue formation. In addition, EPCs enhance the osteogenic differentiation and bone formation of mesenchymal stem cells (MSCs). This study aimed to investigate the impact of EPCs on vascularization and bone formation of a hydroxyapatite (HA) and beta-tricalcium phosphate (ß-TCP)–fibrin scaffold. Three groups were designed: a scaffold-only group (A), a scaffold and EPC group (B), and a scaffold and EPC/MSC group (C). The HA/ß–TCP–fibrin scaffolds were placed in a porous titanium chamber permitting extrinsic vascularization from the surrounding tissue. Additionally, intrinsic vascularization was achieved by means of an arteriovenous loop (AV loop). After 12 weeks, the specimens were explanted and investigated by histology and CT. We were able to prove a strong scaffold vascularization in all groups. No differences regarding the vessel number and density were detected between the groups. Moreover, we were able to prove bone formation in the coimplantation group. Taken together, the AV loop is a powerful tool for vascularization which is independent from scaffold cellularization with endothelial progenitor cells’ prior implantation.
Collapse
Affiliation(s)
- Dominik Steiner
- Laboratory for Tissue Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (L.R.); (L.F.); (R.E.H.); (A.A.)
- Correspondence:
| | - Lea Reinhardt
- Laboratory for Tissue Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (L.R.); (L.F.); (R.E.H.); (A.A.)
| | - Laura Fischer
- Laboratory for Tissue Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (L.R.); (L.F.); (R.E.H.); (A.A.)
| | - Vanessa Popp
- Preclinical Imaging Platform Erlangen (PIPE), Institute of Radiology, University Hospital of Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (V.P.); (T.B.)
| | - Carolin Körner
- Department of Materials Science and Engineering, Institute of Science and Technology of Metals, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91058 Erlangen, Germany;
| | - Carol I. Geppert
- Institute of Pathology, University Hospital of Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany;
- Comprehensive Cancer Center Erlangen-EMN (CCC ER-EMN), 91054 Erlangen, Germany
| | - Tobias Bäuerle
- Preclinical Imaging Platform Erlangen (PIPE), Institute of Radiology, University Hospital of Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (V.P.); (T.B.)
| | - Raymund E. Horch
- Laboratory for Tissue Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (L.R.); (L.F.); (R.E.H.); (A.A.)
| | - Andreas Arkudas
- Laboratory for Tissue Engineering and Regenerative Medicine, Department of Plastic and Hand Surgery, University Hospital of Erlangen, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054 Erlangen, Germany; (L.R.); (L.F.); (R.E.H.); (A.A.)
| |
Collapse
|