1
|
Jia Y, Xu X, Lu H, Fatima K, Zhang Y, Du H, Yang J, Zhou X, Sui X, Hou L, Pang Y, He C. A super soft thermoplastic biodegradable elastomer with high elasticity for arterial regeneration. Biomaterials 2024; 316:122985. [PMID: 39637585 DOI: 10.1016/j.biomaterials.2024.122985] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2024] [Revised: 10/27/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
Elastomers with innovative performance will provide new opportunities for solving problems in soft tissue repair, such as arterial regeneration. Herein, we present a thermoplastic biodegradable elastomer (PPS) that differs from the rigid, low-elastic traditional ones. It shows super softness (0.41 ± 0.052 MPa), high stretchability (3239 ± 357 %), and viscoelasticity similar to natural soft tissues. In addition, it also has good processability and appropriate degradability, estimated at 4-8 months for complete degradation in vivo. This excellent overall performance makes it a great support material for soft tissue repair and a powerful modifying agent for improving existing materials. For example, introducing it into poly(l-lactide) scaffolds through thermally induced phase separation can create a unique microporous structure with interconnected large pores (diameter >10 μm), demonstrating high efficiency in inducing cell infiltration. Blending it with poly(ε-caprolactone) through electrospinning can produce a composite fibrous film with significantly improved comprehensive performance, displaying artery-matched mechanical properties. Building on the above, we constructed a tri-layer tissue-engineered vascular graft for arterial regeneration, exhibiting promising remodeling outcomes in rabbits.
Collapse
Affiliation(s)
- Yating Jia
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China; Key Laboratory of Science and Technology of Eco-Textile, Ministry of Education, College of Chemistry and Chemical Engineering, Donghua University, Shanghai, 201620, China
| | - Xin Xu
- Department of Cardiology, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China
| | - Hao Lu
- Department of Neurosurgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, China
| | - Kanwal Fatima
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Yali Zhang
- Department of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Haibo Du
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Jin Yang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Xiaojun Zhou
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China
| | - Xiaofeng Sui
- Key Laboratory of Science and Technology of Eco-Textile, Ministry of Education, College of Chemistry and Chemical Engineering, Donghua University, Shanghai, 201620, China
| | - Lei Hou
- Department of Cardiology, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China
| | - Yanan Pang
- Department of Cardiology, Songjiang Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 201600, China; Department of Cardiology, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Chuanglong He
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, College of Biological Science and Medical Engineering, Donghua University, Shanghai, 201620, China.
| |
Collapse
|
2
|
Philips C, Terrie L, Muylle E, Thorrez L. Determination of DNA content as quality control in decellularized tissues: challenges and pitfalls. Regen Biomater 2024; 11:rbae123. [PMID: 39569078 PMCID: PMC11578598 DOI: 10.1093/rb/rbae123] [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: 06/25/2024] [Revised: 09/10/2024] [Accepted: 10/15/2024] [Indexed: 11/22/2024] Open
Abstract
Decellularized organs and tissues are emerging within the field of regenerative medicine to meet the growing demand for organ and tissue transplantation. Quality control of these acellular matrices prior to transplantation is of paramount importance to ensure the absence of an adverse reaction. In particular, thorough evaluation of the DNA content is essential but also poses technical challenges. Therefore, in this study, we compared different methods for quantitative and qualitative evaluation of DNA content in native and decellularized skeletal muscle tissue to identify strengths and weaknesses for each. Histological analysis revealed that Feulgen staining is more sensitive and robust than the commonly used hematoxylin-eosin and 4',6-diamidino-2-phenylindole staining for detection of remaining nuclear material. Furthermore, gel electrophoresis allowed to identify the quality and length of remaining DNA fragments. The results of the quantitative analysis indicated that direct measurement of DNA content in tissue lysates is preferred over silica-based extraction methods, since the latter resulted in the loss of small DNA fragments during extraction. Moreover, a weight loss correction factor should be implemented to take into account the impact of the decellularization on the extracellular matrix. With regard to the detection method, the results revealed that a fluorescence-based approach is more accurate than the use of UV/VIS absorbance. Through combination of the proposed methods, it should be possible to achieve a more standardized evaluation of novel acellular matrices in terms of DNA content and to enhance the predictability of clinical success.
Collapse
Affiliation(s)
- Charlot Philips
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven Campus Kulak, 8500 Kortrijk, Belgium
| | - Lisanne Terrie
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven Campus Kulak, 8500 Kortrijk, Belgium
| | - Ewout Muylle
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven Campus Kulak, 8500 Kortrijk, Belgium
| | - Lieven Thorrez
- Tissue Engineering Lab, Department of Development and Regeneration, KU Leuven Campus Kulak, 8500 Kortrijk, Belgium
| |
Collapse
|
3
|
Liao R, Dewey MJ, Rong J, Johnson SA, D’Angelo WA, Hussey GS, Badylak SF. Matrix-bound nanovesicles alleviate particulate-induced periprosthetic osteolysis. SCIENCE ADVANCES 2024; 10:eadn1852. [PMID: 39423278 PMCID: PMC11488533 DOI: 10.1126/sciadv.adn1852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 09/16/2024] [Indexed: 10/21/2024]
Abstract
Aseptic loosening of orthopedic implants is an inflammatory disease characterized by immune cell activation, chronic inflammation, and destruction of periprosthetic bone, and is one of the leading reasons for prosthetic failure, affecting 12% of total joint arthroplasty patients. Matrix-bound nanovesicles (MBVs) are a subclass of extracellular vesicle recently shown to mitigate inflammation in preclinical models of rheumatoid arthritis and influenza-mediated "cytokine storm." The molecular mechanism of these anti-inflammatory properties is only partially understood. The objective of the present study was to investigate the effects of MBV on RANKL-induced osteoclast formation in vitro and particulate-induced osteolysis in vivo. Results showed that MBV attenuated osteoclast differentiation and activity by suppressing the NF-κB signaling pathway and downstream NFATc1, DC-STAMP, c-Src, and cathepsin K expression. In vivo, local administration of MBV attenuated ultrahigh molecular weight polyethylene particle-induced osteolysis, bone reconstruction, and periosteal inflammation. The results suggest that MBV may be a therapeutic option for preventing periprosthetic loosening.
Collapse
Affiliation(s)
- Runzhi Liao
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Xiangya Hospital, Central South University, Changsha, Hunan 410008, China
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Marley J. Dewey
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Jiayang Rong
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Scott A. Johnson
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - William A. D’Angelo
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - George S. Hussey
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
| | - Stephen F. Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15219, USA
- Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15219, USA
| |
Collapse
|
4
|
Yu Q, Gao Y, Guo J, Wang X, Gao X, Zhao Y, Liu Y, Wen M, Zhang X, An M. Bioactivity and in vitro immunological studies of xenogeneic decellularized extracellular matrix scaffolds for implantable applications. J Mater Chem B 2024; 12:9390-9407. [PMID: 39189732 DOI: 10.1039/d4tb00450g] [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: 08/28/2024]
Abstract
Decellularized scaffolds retain the main bioactive substances of the extracellular matrix, which can better promote cell proliferation and matrix reconstruction at the defect site, and have great potential for morphological and functional restoration in patients with tissue defects. Due to the safety of the material source of allogeneic decellularized scaffolds, there is a great limitation in their clinical application, so the preparation and evaluation of xenodermal acellular scaffolds have attracted much attention. In terms of skin tissue structure and function, porcine skin has a high degree of similarity to human skin and has the advantages of sufficient quantity and no ethical issues. However, there is a risk of immune rejection after xenodermal acellular scaffold transplantation. To address the above problems, this paper focuses on porcine dermal decellularized scaffolds prepared using two common decellularization preparation methods and compares the decellularization efficiency, retention of active components of the extracellular matrix, structural characterization of the decellularized scaffolds, and the effect of porcine dermal decellularized scaffolds on mouse Raw264.7 macrophages, so as to make a functional evaluation of the active components and immune effects of porcine dermal decellularized scaffolds, and to provide a reference for filling trauma-induced defects in humans.
Collapse
Affiliation(s)
- Qing Yu
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi 030024, China.
| | - Yuantao Gao
- School of Medicine, Zhejiang University, Zhejiang 310058, China
| | - Jiqiang Guo
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi 030024, China.
- Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Third Hospital of Shanxi Medical University, Taiyuan 030032, China
| | - Xinyue Wang
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi 030024, China.
| | - Xiang Gao
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi 030024, China.
| | - Yifan Zhao
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi 030024, China.
| | - Yang Liu
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi 030024, China.
| | - Meiling Wen
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi 030024, China.
| | - Xiangyu Zhang
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi 030024, China.
| | - Meiwen An
- Institute of Biomedical Engineering, College of Biomedical Engineering, Taiyuan University of Technology, Shanxi 030024, China.
| |
Collapse
|
5
|
Mozafari M, Barbati ME. Oxygen-generating biomaterials for cardiovascular engineering: unveiling future discoveries. Drug Discov Today 2024; 29:104135. [PMID: 39103145 DOI: 10.1016/j.drudis.2024.104135] [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: 02/15/2024] [Revised: 07/26/2024] [Accepted: 07/31/2024] [Indexed: 08/07/2024]
Abstract
Oxygen-generating biomaterials are emerging as a groundbreaking solution for transforming cardiovascular engineering. These biomaterials generate and release oxygen within various biomedical applications, marking a new frontier in healthcare. Most cardiovascular treatments face a significant challenge, ensuring a consistent oxygen supply to nurture engineered tissues or even implanted devices. Traditional methods relying on passive oxygen diffusion often fall short, hindering functional cardiovascular tissue development. Oxygen-generating biomaterials, incorporating agents like calcium peroxide, provide a controlled oxygen source to the surrounding cells. This innovation potentially enhances cell viability, stimulates growth and boosts metabolic activity crucial for tissue health. Applications include repairing cardiac and vascular tissues, disease modeling, drug testing and personalized medicine, promising tailored treatments. Challenges like material toxicity and oxygen release control need consideration. As research progresses, the use of these innovative biomaterials in clinical translation could reshape cardiovascular healthcare, revolutionizing patient outcomes in heart disease treatment.
Collapse
Affiliation(s)
- Masoud Mozafari
- Research Unit of Health Sciences and Technology, Faculty of Medicine, University of Oulu, Oulu, Finland.
| | - Mohammad E Barbati
- Clinic of Vascular and Endovascular Surgery, RWTH Aachen University Hospital, Aachen, Germany
| |
Collapse
|
6
|
da Costa CS, Jorge SDF, Figueiredo MA, Neves DR, Chagas MA. Use of rainbow trout skin treated with glutaraldehyde as a mesh for abdominal hernioplasty in rats. Acta Cir Bras 2024; 39:e393024. [PMID: 39046040 PMCID: PMC11262756 DOI: 10.1590/acb393024] [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: 12/13/2023] [Accepted: 04/04/2024] [Indexed: 07/25/2024] Open
Abstract
PURPOSE To test the use of rainbow trout skin as a surgical mesh in abdominal hernioplasties in rats. METHODS The experiment involved 20 Wistar rats receiving implants of trout skin processed for disinfection in 0.5% glutaraldehyde and preserved in 100% glycerin. The animals were divided into four groups, divided at 7, 15, 30, and 90 days postoperatively. Clinical and infrared thermography evaluations were performed, and after euthanasia, assessments of adhesion formations and sample collection for histological evaluation were conducted. RESULTS The implant was observed to be intact, ensuring the integrity of the abdominal wall, support for the viscera, and normal mobility for the rats for up to 90 days. Low rates of clinical alterations were observed, with an intense inflammatory reaction up to day 7, chronic inflammation and the onset of angiogenesis at day 15, and a low inflammatory reaction with collagenous infiltrate and fibrosis at day 30. At day 90, the implants showed a collagenous and fibrotic infiltrate with a minimal inflammatory infiltrate. CONCLUSIONS The surgical mesh of trout skin performed well, making it a potential alternative for surgical procedures in muscle aponeurotic corrections in the abdominal wall.
Collapse
Affiliation(s)
- Carolina Seabra da Costa
- Universidade Federal Fluminense – Programa de Pós-graduação em Medicina Veterinária Clínica e Reprodução Animal – Niterói (RJ), Brazil
| | - Siria da Fonseca Jorge
- Centro Universitário Serra dos Órgãos – Curso de Graduação em Medicina Veterinária – Teresópolis (RJ), Brazil
| | - Marcelo Abidu Figueiredo
- Universidade Federal Fluminense – Programa de Pós-graduação em Medicina Veterinária Clínica e Reprodução Animal – Niterói (RJ), Brazil
| | - Danielle Rangel Neves
- Centro Universitário Serra dos Órgãos – Curso de Graduação em Medicina Veterinária – Teresópolis (RJ), Brazil
| | - Maurício Alves Chagas
- Universidade Federal Fluminense – Programa de Pós-graduação em Medicina Veterinária Clínica e Reprodução Animal – Niterói (RJ), Brazil
| |
Collapse
|
7
|
Kim BS, Kim JU, Lee J, Ryu KM, Kim SH, Hwang NS. Decellularized brain extracellular matrix based NGF-releasing cryogel for brain tissue engineering in traumatic brain injury. J Control Release 2024; 368:140-156. [PMID: 38373473 DOI: 10.1016/j.jconrel.2024.02.017] [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: 11/13/2023] [Revised: 02/05/2024] [Accepted: 02/12/2024] [Indexed: 02/21/2024]
Abstract
Traumatic brain injuries(TBI) pose significant challenges to human health, specifically neurological disorders and related motor activities. After TBI, the injured neuronal tissue is known for hardly regenerated and recovered to their normal neuron physiology and tissue compositions. For this reason, tissue engineering strategies that promote neuronal regeneration have gained increasing attention. This study explored the development of a novel neural tissue regeneration cryogel by combining brain-derived decellularized extracellular matrix (ECM) with heparin sulfate crosslinking that can perform nerve growth factor (NGF) release ability. Morphological and mechanical characterizations of the cryogels were performed to assess their suitability as a neural regeneration platform. After that, the heparin concnentration dependent effects of varying NGF concentrations on cryogel were investigated for their controlled release and impact on neuronal cell differentiation. The results revealed a direct correlation between the concentration of released NGF and the heparin sulfate ratio in cryogel, indicating that the cryogel can be tailored to carry higher loads of NGF with heparin concentration in cryogel that induced higher neuronal cell differentiation ratio. Furthermore, the study evaluated the NGF loaded cryogels on neuronal cell proliferation and brain tissue regeneration in vivo. The in vivo results suggested that the NGF loaded brain ECM derived cryogel significantly affects the regeneration of brain tissue. Overall, this research contributes to the development of advanced neural tissue engineering strategies and provides valuable insights into the design of regenerative cryogels that can be customized for specific therapeutic applications.
Collapse
Affiliation(s)
- Beom-Seok Kim
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea
| | - Jeong-Uk Kim
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Jaewoo Lee
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Kyung Min Ryu
- School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea
| | - Su-Hwan Kim
- Department of Chemical Engineering (BK21 FOUR), Dong-A University, Busan 49315, Republic of Korea
| | - Nathaniel S Hwang
- Interdisciplinary Program in Bioengineering, Seoul National University, Seoul 08826, Republic of Korea; School of Chemical and Biological Engineering, Institute of Chemical Processes, Seoul National University, Seoul 08826, Republic of Korea; Bio-MAX Institute, Institute of Bio-Engineering, Seoul National University, Seoul 08826, Republic of Korea; Institute of Engineering Research, Seoul National University, Seoul 08826, Republic of Korea.
| |
Collapse
|
8
|
Leng W, Li X, Dong L, Guo Z, Ji X, Cai T, Xu C, Zhu Z, Lin J. The Regenerative Microenvironment of the Tissue Engineering for Urethral Strictures. Stem Cell Rev Rep 2024; 20:672-687. [PMID: 38305981 DOI: 10.1007/s12015-024-10686-7] [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] [Accepted: 01/27/2024] [Indexed: 02/03/2024]
Abstract
Urethral stricture caused by various reasons has threatened the quality of life of patients for decades. Traditional reconstruction methods, especially for long-segment injuries, have shown poor outcomes in treating urethral strictures. Tissue engineering for urethral regeneration is an emerging concept in which special designed scaffolds and seed cells are used to promote local urethral regeneration. The scaffolds, seed cells, various factors and the host interact with each other and form the regenerative microenvironment. Among the various interactions involved, vascularization and fibrosis are the most important biological processes during urethral regeneration. Mesenchymal stem cells and induced pluripotent stem cells play special roles in stricture repair and facilitate long-segment urethral regeneration, but they may also induce carcinogenesis and genomic instability during reconstruction. Nevertheless, current technologies, such as genetic engineering, molecular imaging, and exosome extraction, provide us with opportunities to manage seed cell-related regenerative risks. In this review, we described the interactions among seed cells, scaffolds, factors and the host within the regenerative microenvironment, which may help in determining the exact molecular mechanisms involved in urethral stricture regeneration and promoting clinical trials and the application of urethral tissue engineering in patients suffering from urethral stricture.
Collapse
Affiliation(s)
- Wenyuan Leng
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, No. 8, Street Xishiku, District Xicheng, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Xiaoyu Li
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, No. 8, Street Xishiku, District Xicheng, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Lei Dong
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, No. 8, Street Xishiku, District Xicheng, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Zhenke Guo
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, No. 8, Street Xishiku, District Xicheng, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Xing Ji
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, No. 8, Street Xishiku, District Xicheng, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Tianyu Cai
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, No. 8, Street Xishiku, District Xicheng, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Chunru Xu
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, No. 8, Street Xishiku, District Xicheng, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Zhenpeng Zhu
- Department of Urology, Peking University First Hospital, Beijing, 100034, China
- Institute of Urology, Peking University, Beijing, 100034, China
- National Urological Cancer Center, No. 8, Street Xishiku, District Xicheng, Beijing, 100034, China
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China
| | - Jian Lin
- Department of Urology, Peking University First Hospital, Beijing, 100034, China.
- Institute of Urology, Peking University, Beijing, 100034, China.
- National Urological Cancer Center, No. 8, Street Xishiku, District Xicheng, Beijing, 100034, China.
- Beijing Key Laboratory of Urogenital Diseases (Male) Molecular Diagnosis and Treatment Center, Beijing, 100034, China.
| |
Collapse
|
9
|
Touchet TJ, Horelica M, Gruenbaum R, Lewy K, Hines E, Stranahan L, Saunders WB, Maitland DJ. Fabrication and In Vivo Assessment of Oxidatively Responsive PolyHIPE Scaffolds for Use in Diabetic Orthopedic Applications. Macromol Biosci 2024; 24:e2300393. [PMID: 37904644 DOI: 10.1002/mabi.202300393] [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: 09/23/2023] [Revised: 10/16/2023] [Indexed: 11/01/2023]
Abstract
Achieving surgical success in orthopedic patients with metabolic disease remains a substantial challenge. Diabetic patients exhibit a unique tissue microenvironment consisting of high levels of reactive oxygen species (ROS), which promotes osteoclastic activity and leads to decreased bone healing. Alternative solutions, such as synthetic grafts, incorporating progenitor cells or growth factors, can be costly and have processing constraints. Previously, the potential for thiol-methacrylate networks to sequester ROS while possessing tunable mechanical properties and degradation rates has been demonstrated. In this study, the ability to fabricate thiol-methacrylate interconnected porous scaffolds using emulsion templating to create monoliths with an average porosity of 97.0% is reported. The average pore sizes of the scaffolds range from 27 to 656 µm. The scaffolds can sequester pathologic levels of ROS via hydrogen peroxide consumption and are not impacted by sterilization. Subcutaneous implantation shows no signs of acute toxicity. Finally, in a 6-week bilateral calvarial defect model in Zucker diabetic fatty rats, ROS scaffolds increase new bone volume by 66% over sham defects. Histologic analysis identifies woven bone infiltration throughout the scaffold and neovascularization. Overall, this study suggests that porous thiol-methacrylate scaffolds may improve healing for bone grafting applications where high levels of ROS hinder bone growth.
Collapse
Affiliation(s)
- Tyler J Touchet
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
- Enovis, 727 N. Shepherd Drive Suite 100, Houston, TX, 77007, USA
| | - Madeleine Horelica
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Rachel Gruenbaum
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| | - Keith Lewy
- Department of Comparative Medicine, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Elizabeth Hines
- Department of Veterinary Pathobiology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Lauren Stranahan
- Department of Veterinary Pathobiology, School of Veterinary Medicine and Biomedical Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - W Brian Saunders
- Department of Small Animal Clinical Sciences, School of Veterinary Medicine & Biomedical Sciences, Texas A&M University, College Station, TX, 77843, USA
| | - Duncan J Maitland
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, 77843, USA
| |
Collapse
|
10
|
Maistriaux L, Foulon V, Fievé L, Xhema D, Evrard R, Manon J, Coyette M, Bouzin C, Poumay Y, Gianello P, Behets C, Lengelé B. Reconstruction of the human nipple-areolar complex: a tissue engineering approach. Front Bioeng Biotechnol 2024; 11:1295075. [PMID: 38425730 PMCID: PMC10902434 DOI: 10.3389/fbioe.2023.1295075] [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: 09/15/2023] [Accepted: 12/13/2023] [Indexed: 03/02/2024] Open
Abstract
Introduction: Nipple-areolar complex (NAC) reconstruction after breast cancer surgery is challenging and does not always provide optimal long-term esthetic results. Therefore, generating a NAC using tissue engineering techniques, such as a decellularization-recellularization process, is an alternative option to recreate a specific 3D NAC morphological unit, which is then covered with an in vitro regenerated epidermis and, thereafter, skin-grafted on the reconstructed breast. Materials and methods: Human NACs were harvested from cadaveric donors and decellularized using sequential detergent baths. Cellular clearance and extracellular matrix (ECM) preservation were analyzed by histology, as well as by DNA, ECM proteins, growth factors, and residual sodium dodecyl sulfate (SDS) quantification. In vivo biocompatibility was evaluated 30 days after the subcutaneous implantation of native and decellularized human NACs in rats. In vitro scaffold cytocompatibility was assessed by static seeding of human fibroblasts on their hypodermal side for 7 days, while human keratinocytes were seeded on the scaffold epidermal side for 10 days by using the reconstructed human epidermis (RHE) technique to investigate the regeneration of a new epidermis. Results: The decellularized NAC showed a preserved 3D morphology and appeared white. After decellularization, a DNA reduction of 98.3% and the absence of nuclear and HLA staining in histological sections confirmed complete cellular clearance. The ECM architecture and main ECM proteins were preserved, associated with the detection and decrease in growth factors, while a very low amount of residual SDS was detected after decellularization. The decellularized scaffolds were in vivo biocompatible, fully revascularized, and did not induce the production of rat anti-human antibodies after 30 days of subcutaneous implantation. Scaffold in vitro cytocompatibility was confirmed by the increasing proliferation of seeded human fibroblasts during 7 days of culture, associated with a high number of living cells and a similar viability compared to the control cells after 7 days of static culture. Moreover, the RHE technique allowed us to recreate a keratinized pluristratified epithelium after 10 days of culture. Conclusion: Tissue engineering allowed us to create an acellular and biocompatible NAC with a preserved morphology, microarchitecture, and matrix proteins while maintaining their cell growth potential and ability to regenerate the skin epidermis. Thus, tissue engineering could provide a novel alternative to personalized and natural NAC reconstruction.
Collapse
Affiliation(s)
- Louis Maistriaux
- Pole of Morphology (MORF), Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
- Pole of Experimental Surgery and Transplantation (CHEX), Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
| | - Vincent Foulon
- Pole of Morphology (MORF), Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
| | - Lies Fievé
- Pole of Morphology (MORF), Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
| | - Daela Xhema
- Pole of Experimental Surgery and Transplantation (CHEX), Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
| | - Robin Evrard
- Pole of Experimental Surgery and Transplantation (CHEX), Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
| | - Julie Manon
- Pole of Morphology (MORF), Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
| | - Maude Coyette
- Pole of Morphology (MORF), Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
- Department of Plastic and Reconstructive Surgery, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Caroline Bouzin
- IREC Imaging Platform (2IP), Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
| | - Yves Poumay
- Research Unit for Molecular Physiology (URPhyM), Department of Medicine, Namur Research Institute for Life Sciences (NARILIS), UNamur, Namur, Belgium
| | - Pierre Gianello
- Pole of Experimental Surgery and Transplantation (CHEX), Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
| | - Catherine Behets
- Pole of Morphology (MORF), Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
| | - Benoît Lengelé
- Pole of Morphology (MORF), Institute of Experimental and Clinical Research (IREC), UCLouvain, Brussels, Belgium
- Department of Plastic and Reconstructive Surgery, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
11
|
Zhao P, Yang F, Jia X, Xiao Y, Hua C, Xing M, Lyu G. Extracellular Matrices as Bioactive Materials for In Situ Tissue Regeneration. Pharmaceutics 2023; 15:2771. [PMID: 38140112 PMCID: PMC10747903 DOI: 10.3390/pharmaceutics15122771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2023] [Revised: 10/28/2023] [Accepted: 11/28/2023] [Indexed: 12/24/2023] Open
Abstract
Bioactive materials based on a nature-derived extracellular matrix (NECM) represent a category of biomedical devices with versatile therapeutic applications in the realms of tissue repair and engineering. With advancements in decellularization technique, the inherent bioactive molecules and the innate nano-structural and mechanical properties are preserved in three-dimensional scaffolds mainly composed of collagens. Techniques such as electrospinning, three-dimensional printing, and the intricate fabrication of hydrogels are developed to mimic the physical structures, biosignalling and mechanical cues of ECM. Until now, there has been no approach that can fully account for the multifaceted properties and diverse applications of NECM. In this review, we introduce the main proteins composing NECMs and explicate the importance of them when used as therapeutic devices in tissue repair. Nano-structural features of NECM and their applications regarding tissue repair are summarized. The origins, degradability, and mechanical property of and immune responses to NECM are also introduced. Furthermore, we review their applications, and clinical features thereof, in the repair of acute and chronic wounds, abdominal hernia, breast deformity, etc. Some typical marketed devices based on NECM, their indications, and clinical relevance are summarized.
Collapse
Affiliation(s)
- Peng Zhao
- Burn & Trauma Treatment Center, Affiliated Hospital of Jiangnan University, Wuxi 214122, China
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Affiliated Hospital of Jiangnan University, Wuxi 214000, China; (F.Y.); (Y.X.)
| | - Fengbo Yang
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Affiliated Hospital of Jiangnan University, Wuxi 214000, China; (F.Y.); (Y.X.)
| | - Xiaoli Jia
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Affiliated Hospital of Jiangnan University, Wuxi 214000, China; (F.Y.); (Y.X.)
| | - Yuqin Xiao
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Affiliated Hospital of Jiangnan University, Wuxi 214000, China; (F.Y.); (Y.X.)
| | - Chao Hua
- Burn & Trauma Treatment Center, Affiliated Hospital of Jiangnan University, Wuxi 214122, China
| | - Malcolm Xing
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, MB R3T 2N2, Canada
| | - Guozhong Lyu
- Burn & Trauma Treatment Center, Affiliated Hospital of Jiangnan University, Wuxi 214122, China
- Engineering Research Center of the Ministry of Education for Wound Repair Technology, Jiangnan University, Affiliated Hospital of Jiangnan University, Wuxi 214000, China; (F.Y.); (Y.X.)
| |
Collapse
|
12
|
Zhai X, Tao X, Wu Y, Jin K, Tan H, Zhou T, Chen Y. Injectable and Self-Adaptive Gel Scaffold Based on Heparin Microspheres for Adipogenesis of Human Adipose-Derived Stem Cells. Biomacromolecules 2023; 24:4663-4671. [PMID: 37722066 DOI: 10.1021/acs.biomac.3c00348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/20/2023]
Abstract
An injectable and self-adaptive heparin microsphere-based cell scaffold was developed to achieve adipose regeneration. Simultaneously, the cell scaffold exhibited a dynamic architecture, self-regulated glucose levels, sustained insulin delivery, and steady viscoelastic properties for adipogenesis. The dynamic cell scaffold is cross-linked by the boronate-diol interaction among heparin-based microspheres, which have boronate and maltose groups. Because of the boronate-maltose ester bonds, the gelatinous complex would be partially dismantled and readily display glucose-sensitive performance by free glucose via competitive displacement. The dynamic cross-linking heparin microsphere scaffold can deliver the lipogenic drug insulin to enhance lipid filling, which has an impact on fat tissue enhancement. A 4-week in vitro cell culture demonstrated that the dynamic heparin microsphere-based cell scaffold, through loading with insulin, showed significantly higher efficiency in promoting ASC differentiation compared with traditional 3D culture methods. In vivo histological results further demonstrated that there was a significant increase in adipose in the proposed cell scaffold, which proved to be statistically significant compared with traditional biomaterials. Notable stain expression of the FABP4 and PPAR-γ genes was also observed in the dynamic cell scaffold containing insulin, which was more similar to natural fat.
Collapse
Affiliation(s)
- Xinyue Zhai
- School of Materials Science and Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Xinwei Tao
- School of Materials Science and Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Yuqian Wu
- School of Materials Science and Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Kesun Jin
- School of Materials Science and Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Huaping Tan
- School of Materials Science and Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Tianle Zhou
- School of Materials Science and Engineering, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Yong Chen
- Department of Orthopaedics, Jinling Hospital, Nanjing 210002, China
| |
Collapse
|
13
|
Blaudez F, Ivanovski S, Vaquette C. Harnessing the Native Extracellular Matrix for Periodontal Regeneration Using a Melt Electrowritten Biphasic Scaffold. J Funct Biomater 2023; 14:479. [PMID: 37754893 PMCID: PMC10531993 DOI: 10.3390/jfb14090479] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2023] [Revised: 09/05/2023] [Accepted: 09/13/2023] [Indexed: 09/28/2023] Open
Abstract
Scaffolds have been used to promote periodontal regeneration by providing control over the spacio-temporal healing of the periodontium (cementum, periodontal ligament (PDL) and alveolar bone). This study proposes to enhance the biofunctionality of a biphasic scaffold for periodontal regeneration by means of cell-laid extracellular matrix (ECM) decoration. To this end, a melt electrowritten scaffold was cultured with human osteoblasts for the deposition of bone-specific ECM. In parallel, periodontal ligament cells were used to form a cell sheet, which was later combined with the bone ECM scaffold to form a biphasic PDL-bone construct. The resulting biphasic construct was decellularised to remove all cellular components while preserving the deposited matrix. Decellularisation efficacy was confirmed in vitro, before the regenerative performance of freshly decellularised constructs was compared to that of 3-months stored freeze-dried scaffolds in a rodent periodontal defect model. Four weeks post-surgery, microCT revealed similar bone formation in all groups. Histology showed higher amounts of newly formed cementum and periodontal attachment in the fresh and freeze-dried ECM functionalised scaffolds, although it did not reach statistical significance. This study demonstrated that the positive effect of ECM decoration was preserved after freeze-drying and storing the construct for 3 months, which has important implications for clinical translation.
Collapse
Affiliation(s)
- Fanny Blaudez
- School of Dentistry, Centre for Oral Regeneration, Reconstruction and Rehabilitation (COR3), The University of Queensland, Herston, QLD 4006, Australia; (F.B.); (S.I.)
- School of Dentistry and Oral Health, Griffith University, Southport, QLD 4222, Australia
| | - Saso Ivanovski
- School of Dentistry, Centre for Oral Regeneration, Reconstruction and Rehabilitation (COR3), The University of Queensland, Herston, QLD 4006, Australia; (F.B.); (S.I.)
| | - Cedryck Vaquette
- School of Dentistry and Oral Health, Griffith University, Southport, QLD 4222, Australia
| |
Collapse
|
14
|
Blaudez F, Ivanovski S, Fernandez T, Vaquette C. Effect of In Vitro Culture Length on the Bone-Forming Capacity of Osteoblast-Derived Decellularized Extracellular Matrix Melt Electrowritten Scaffolds. Biomacromolecules 2023; 24:3450-3462. [PMID: 37458386 DOI: 10.1021/acs.biomac.2c01504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/15/2023]
Abstract
Recent advancements in decellularization have seen the development of extracellular matrix (ECM)-decorated scaffolds for bone regeneration; however, little is understood of the impact of in vitro culture prior to decellularization on the performances of these constructs. Therefore, this study investigated the effect of in vitro culture on ECM-decorated melt electrowritten polycaprolactone scaffold bioactivity. The scaffolds were seeded with osteoblasts and cultured for 1, 2, or 4 weeks to facilitate bone-specific ECM deposition and subsequently decellularized to form an acellular ECM-decorated scaffold. The utilization of mild chemicals and DNase was highly efficient in removing DNA while preserving ECM structure and composition. ECM decoration of the melt electrowritten fibers was observed within the first week of culture, with increased ECM at 2 and 4 week culture periods. Infiltration of re-seeded cells as well as overall bone regeneration in a rodent calvarial model was impeded by a longer culture period. Thus, it was demonstrated that the length of culture has a key influence on the osteogenic properties of decellularized ECM-decorated scaffolds, with long-term culture (2+ weeks) causing pore obstruction and creating a physical barrier which interfered with bone formation. These findings have important implications for the development of effective ECM-decorated scaffolds for bone regeneration.
Collapse
Affiliation(s)
- Fanny Blaudez
- School of Dentistry and Oral Health, Griffith University, Parklands Dr., Southport QLD 4222, Australia
- The University of Queensland, School of Dentistry, 288 Herston Rd., Herston QLD 4006, Australia
| | - Saso Ivanovski
- The University of Queensland, School of Dentistry, 288 Herston Rd., Herston QLD 4006, Australia
| | - Tulio Fernandez
- The University of Queensland, School of Dentistry, 288 Herston Rd., Herston QLD 4006, Australia
- College of Medicine and Dentistry, James Cook University, Cairns Campus, Cairns 4870, Australia
| | - Cedryck Vaquette
- The University of Queensland, School of Dentistry, 288 Herston Rd., Herston QLD 4006, Australia
| |
Collapse
|
15
|
Ziegler ME, Khabaz K, Khoshab N, Halaseh FF, Chnari E, Chen S, Baldi P, Evans GRD, Widgerow AD. Combining Allograft Adipose and Fascia Matrix as an Off-the-Shelf Scaffold for Adipose Tissue Engineering Stimulates Angiogenic Responses and Activates a Proregenerative Macrophage Profile in a Rodent Model. Ann Plast Surg 2023; 91:294-300. [PMID: 37489973 DOI: 10.1097/sap.0000000000003587] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/26/2023]
Abstract
OBJECTIVE Bioscaffolds for treating soft tissue defects have limitations. As a bioscaffold, allograft adipose matrix (AAM) is a promising approach to treat soft tissue defects. Previously, we revealed that combining superficial adipose fascia matrix with AAM, components of the hypodermis layer of adipose tissue, improved volume retention, adipogenesis, and angiogenesis in rats 8 weeks after it was implanted compared with AAM alone. Here, we modified the fascia matrix and AAM preparation, examined the tissue over 18 weeks, and conducted a deeper molecular investigation. We hypothesized that the combined matrices created a better scaffold by triggering angiogenesis and proregenerative signals. METHODS Human AAM and fascia matrix were implanted (4 [1 mL] implants/animal) into the dorsum of male Fischer rats (6-8 weeks old; ~140 g) randomly as follows: AAM, fascia, 75/25 (AAM/fascia), 50/50, and 50/50 + hyaluronic acid (HA; to improve extrudability) (n = 4/group/time point). After 72 hours, as well as 1, 3, 6, 9, 12, and 18 weeks, graft retention was assessed by a gas pycnometer. Adipogenesis (HE), angiogenesis (CD31), and macrophage infiltration (CD80 and CD163) were evaluated histologically at all time points. The adipose area and M1/M2 macrophage ratio were determined using ImageJ. RNA sequencing (RNA-seq) and bioinformatics were conducted to evaluate pathway enrichments. RESULTS By 18 weeks, the adipose area was 2365% greater for 50/50 HA (281.6 ± 21.6) than AAM (11.4 ± 0.9) (P < 0.001). The M1/M2 macrophage ratio was significantly lower for 50/50 HA (0.8 ± 0.1) than AAM (0.9 ± 0.1) at 6 weeks (16%; P < 0.05). This inversely correlated with adipose area (r = -0.6; P > 0.05). The RNA-seq data revealed that upregulated adipogenesis, angiogenesis, and macrophage-induced tissue regeneration genes were temporally different between the groups. CONCLUSIONS Combining the fascia matrix with AAM creates a bioscaffold with an improved retention volume that supports M2 macrophage-mediated angiogenesis and adipogenesis. This bioscaffold is worthy of further investigation.
Collapse
Affiliation(s)
- Mary E Ziegler
- From the Center for Tissue Engineering, UC Irvine Department of Plastic Surgery, Orange, CA
| | - Kameel Khabaz
- From the Center for Tissue Engineering, UC Irvine Department of Plastic Surgery, Orange, CA
| | - Nima Khoshab
- From the Center for Tissue Engineering, UC Irvine Department of Plastic Surgery, Orange, CA
| | - Faris F Halaseh
- From the Center for Tissue Engineering, UC Irvine Department of Plastic Surgery, Orange, CA
| | | | | | | | - Gregory R D Evans
- From the Center for Tissue Engineering, UC Irvine Department of Plastic Surgery, Orange, CA
| | - Alan D Widgerow
- From the Center for Tissue Engineering, UC Irvine Department of Plastic Surgery, Orange, CA
| |
Collapse
|
16
|
Hasturk O, Sahoo JK, Kaplan DL. Synthesis and characterization of silk-poly(guluronate) hybrid polymers for the fabrication of dual crosslinked, mechanically dynamic hydrogels. POLYMER 2023; 281:126129. [PMID: 37483847 PMCID: PMC10357961 DOI: 10.1016/j.polymer.2023.126129] [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] [Indexed: 07/25/2023]
Abstract
The rapid ionic crosslinking of alginate has been actively studied for biomedical applications including hydrogel scaffolds for tissue engineering, injectable gels, and 3D bioprinting. However, the poor structural stability of ionic crosslinks under physiological conditions limits the widespread applications of these hydrogels. Moreover, the lack of cell adhesion to the material combined with the inability of proteases to degrade alginate further restrict utility as hydrogel scaffolds. Blends of alginate with silk fibroin have been proposed for improved structural and mechanical properties, but potential phase separation between the hydrophobic protein and the hydrophilic polysaccharide remains an issue. In this study, we demonstrated the synthesis of a hybrid biopolymer composed of a silk backbone with side chains of poly(guluronate) isolated from alginate to introduce rapid ionic crosslinking on enzymatically crosslinked silk-based hydrogels for on-demand and reversible stiffening and softening properties. Dual crosslinked macro- and microgels of silk fibroin-poly(guluronate) (SF-PG) hybrid polymers displayed dynamic morphology with reversible shrinking and swelling behavior. SF-PG hydrogel discs demonstrated dynamic mechanics with compressive moduli ranging from less than 5 kPa to over 80 kPa and underwent proteolytic degradation unlike covalently crosslinked alginate controls. SF-PG gels supplemented with gelatin substituted with tyramine or both tyramine and PG also supported the attachment and survival of murine fibroblasts, suggesting potential uses of these new hydrogels in mammalian cell culture to investigate cellular responses to dynamic mechanics or modeling of diseases defined by matrix mechanics, such as fibrosis and cancer.
Collapse
Affiliation(s)
- Onur Hasturk
- Tufts University, Department of Biomedical Engineering, Medford MA, USA
| | | | - David L Kaplan
- Tufts University, Department of Biomedical Engineering, Medford MA, USA
| |
Collapse
|
17
|
Wang D, Charoensombut N, Kawabata K, Kimura T, Kishida A, Ushida T, Furukawa KS. Effect of Pressure Conditions in Uterine Decellularization Using Hydrostatic Pressure on Structural Protein Preservation. Bioengineering (Basel) 2023; 10:814. [PMID: 37508841 PMCID: PMC10376797 DOI: 10.3390/bioengineering10070814] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Revised: 06/26/2023] [Accepted: 07/05/2023] [Indexed: 07/30/2023] Open
Abstract
Uterine regeneration using decellularization scaffolds provides a novel treatment for uterine factor infertility. Decellularized scaffolds require maximal removal of cellular components and minimal damage to the extracellular matrix (ECM). Among many decellularization methods, the hydrostatic pressure (HP) method stands out due to its low cytotoxicity and superior ECM preservation compared to the traditional detergent methods. Conventionally, 980 MPa was utilized in HP decellularization, including the first successful implementation of uterine decellularization previously reported by our team. However, structural protein denaturation caused by exceeding pressure led to a limited regeneration outcome in our previous research. This factor urged the study on the effects of pressure conditions in HP methods on decellularized scaffolds. The authors, therefore, fabricated a decellularized uterine scaffold at varying pressure conditions and evaluated the scaffold qualities from the perspective of cell removal and ECM preservation. The results show that by using lower decellularization pressure conditions of 250 MPa, uterine tissue can be decellularized with more preserved structural protein and mechanical properties, which is considered to be promising for decellularized uterine scaffold fabrication applications.
Collapse
Affiliation(s)
- Dongzhe Wang
- Department of Mechanical Engineering, School of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Narintadeach Charoensombut
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Kinyoshi Kawabata
- Department of Mechanical Engineering, School of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Tsuyoshi Kimura
- Department of Material-Based Medical Engineering, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10, Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| | - Akio Kishida
- Department of Material-Based Medical Engineering, Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10, Kanda-Surugadai, Chiyoda-ku, Tokyo 101-0062, Japan
| | - Takashi Ushida
- Department of Mechanical Engineering, School of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | - Katsuko S Furukawa
- Department of Mechanical Engineering, School of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
- Department of Bioengineering, School of Engineering, The University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| |
Collapse
|
18
|
Makuloluwa AK, Hamill KJ, Rauz S, Bosworth L, Haneef A, Romano V, Williams RL, Dartt DA, Kaye SB. The conjunctival extracellular matrix, related disorders and development of substrates for conjunctival restoration. Ocul Surf 2023; 28:322-335. [PMID: 34102309 DOI: 10.1016/j.jtos.2021.05.011] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2021] [Revised: 05/05/2021] [Accepted: 05/26/2021] [Indexed: 12/15/2022]
Abstract
The conjunctiva can be damaged by numerous diseases with scarring, loss of tissue and dysfunction. Depending on extent of damage, restoration of function may require a conjunctival graft. A wide variety of biological and synthetic substrates have been tested in the search for optimal conditions for ex vivo culture of conjunctival epithelial cells as a route toward tissue grafts. Each substrate has specific advantages but also disadvantages related to their unique physical and biological characteristics, and identification and development of an improved substrate remains a priority. To achieve the goal of mimicking and restoring a biological material, requires information from the material. Specifically, extracellular matrix (ECM) derived from conjunctival tissue. Knowledge of the composition and structure of native ECM and identifying contributions of individual components to its function would enable using or mimicking those components to develop improved biological substrates. ECM is comprised of two components: basement membrane secreted predominantly by epithelial cells containing laminins and type IV collagens, which directly support epithelial and goblet cell adhesion differentiation and growth and, interstitial matrix secreted by fibroblasts in lamina propria, which provides mechanical and structural support. This review presents current knowledge on anatomy, composition of conjunctival ECM and related conjunctival disorders. Requirements of potential substrates for conjunctival tissue engineering and transplantation are discussed. Biological and synthetic substrates and their components are described in an accompanying review.
Collapse
Affiliation(s)
- Aruni K Makuloluwa
- Department of Eye and Vision Science, University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - Kevin J Hamill
- Department of Eye and Vision Science, University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - Saaeha Rauz
- Academic Unit of Ophthalmology, Institute of Inflammation and Ageing, University of Birmingham and Birmingham and Midland Eye Centre, Dudley Road Birmingham, B18 7QU, UK
| | - Lucy Bosworth
- Department of Eye and Vision Science, University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - Atikah Haneef
- Department of Eye and Vision Science, University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - Vito Romano
- Department of Eye and Vision Science, University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - Rachel L Williams
- Department of Eye and Vision Science, University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK
| | - Darlene A Dartt
- Schepens Eye Research Institute, Mass Eye and Ear Infirmary, Harvard Medical School, 20 Staniford St. Boston, MA, 02114, USA
| | - Stephen B Kaye
- Department of Eye and Vision Science, University of Liverpool, William Duncan Building, 6 West Derby Street, Liverpool, L7 8TX, UK.
| |
Collapse
|
19
|
Stearns-Reider KM, Hicks MR, Hammond KG, Reynolds JC, Maity A, Kurmangaliyev YZ, Chin J, Stieg AZ, Geisse NA, Hohlbauch S, Kaemmer S, Schmitt LR, Pham TT, Yamauchi K, Novitch BG, Wollman R, Hansen KC, Pyle AD, Crosbie RH. Myoscaffolds reveal laminin scarring is detrimental for stem cell function while sarcospan induces compensatory fibrosis. NPJ Regen Med 2023; 8:16. [PMID: 36922514 PMCID: PMC10017766 DOI: 10.1038/s41536-023-00287-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2022] [Accepted: 02/22/2023] [Indexed: 03/18/2023] Open
Abstract
We developed an on-slide decellularization approach to generate acellular extracellular matrix (ECM) myoscaffolds that can be repopulated with various cell types to interrogate cell-ECM interactions. Using this platform, we investigated whether fibrotic ECM scarring affected human skeletal muscle progenitor cell (SMPC) functions that are essential for myoregeneration. SMPCs exhibited robust adhesion, motility, and differentiation on healthy muscle-derived myoscaffolds. All SPMC interactions with fibrotic myoscaffolds from dystrophic muscle were severely blunted including reduced motility rate and migration. Furthermore, SMPCs were unable to remodel laminin dense fibrotic scars within diseased myoscaffolds. Proteomics and structural analysis revealed that excessive collagen deposition alone is not pathological, and can be compensatory, as revealed by overexpression of sarcospan and its associated ECM receptors in dystrophic muscle. Our in vivo data also supported that ECM remodeling is important for SMPC engraftment and that fibrotic scars may represent one barrier to efficient cell therapy.
Collapse
Affiliation(s)
- Kristen M Stearns-Reider
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Orthopaedic Surgery, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Michael R Hicks
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Physiology and Biophysics, University of California, Irvine, Irvine, CA, 92697, USA
| | - Katherine G Hammond
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Joseph C Reynolds
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Alok Maity
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Institute for Quantitative and Computational Biology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Yerbol Z Kurmangaliyev
- Institute for Quantitative and Computational Biology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Biological Chemistry, HHMI, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Jesse Chin
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Adam Z Stieg
- California NanoSystems Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | | | - Sophia Hohlbauch
- Asylum Research, An Oxford Instruments Company, Santa Barbara, CA, 93117, USA
| | - Stefan Kaemmer
- Park Systems, 3040 Olcott St, Santa Clara, CA, 95054, USA
| | - Lauren R Schmitt
- Department of Biochemistry and Molecular Genetics, University of Colorado, Denver, Aurora, CO, 80045, USA
| | - Thanh T Pham
- Department of Biochemistry and Molecular Genetics, University of Colorado, Denver, Aurora, CO, 80045, USA
| | - Ken Yamauchi
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Bennett G Novitch
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Intellectual and Developmental Disabilities Research Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Roy Wollman
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Department of Chemistry and Biochemistry, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Institute for Quantitative and Computational Biology, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Kirk C Hansen
- Department of Biochemistry and Molecular Genetics, University of Colorado, Denver, Aurora, CO, 80045, USA
| | - April D Pyle
- Department of Microbiology, Immunology, and Molecular Genetics, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA
| | - Rachelle H Crosbie
- Department of Integrative Biology and Physiology, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Eli and Edythe Broad Center of Regenerative Medicine and Stem Cell Research, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Molecular Biology Institute, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Neurology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
20
|
Intravaia JT, Graham T, Kim HS, Nanda HS, Kumbar SG, Nukavarapu SP. Smart Orthopedic Biomaterials and Implants. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2023; 25:100439. [PMID: 36642994 PMCID: PMC9835562 DOI: 10.1016/j.cobme.2022.100439] [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] [Indexed: 12/24/2022]
Abstract
Musculoskeletal injuries including bone defects continue to present a significant challenge in orthopedic surgery due to suboptimal healing. Bone reconstruction strategies focused on the use of biological grafts and bone graft substitutes in the form of biomaterials-based 3D structures in fracture repair. Recent advances in biomaterials science and engineering have resulted in the creation of intricate 3D bone-mimicking structures that are mechanically stable, biodegradable, and bioactive to support bone regeneration. Current efforts are focused on improving the biomaterial and implant physicochemical properties to promote interactions with the host tissue and osteogenesis. The "smart" biomaterials and their 3D structures are designed to actively interact with stem/progenitor cells and the extracellular matrix (ECM) to influence the local environment towards osteogenesis and de novo tissue formation. This article will summarize such smart biomaterials and the methodologies to apply either internal or external stimuli to control the tissue healing microenvironment. A particular emphasis is also made on the use of smart biomaterials and strategies to create functional bioactive implants for bone defect repair and regeneration.
Collapse
Affiliation(s)
| | - Trevon Graham
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
| | - Hyun S. Kim
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
| | - Himansu S. Nanda
- Terasaki Institute, University of California, Los Angeles, CA, USA
- Mechanical Engineering, IIITDM, Jabalpur, MP, India
| | - Sangamesh G. Kumbar
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
- Department of Materials Science & Engineering, University of Connecticut, Storrs, CT, USA
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, USA
| | - Syam P. Nukavarapu
- Department of Biomedical Engineering, University of Connecticut, Storrs, CT, USA
- Department of Materials Science & Engineering, University of Connecticut, Storrs, CT, USA
- Department of Orthopaedic Surgery, University of Connecticut Health, Farmington, CT, USA
| |
Collapse
|
21
|
Wei Z, Zhang J, Guo Z, Wu Z, Sun Y, Wang K, Duan R. Study on the preparation and properties of acellular matrix from the skin of silver carp (Hypophthalmichthys molitrix). J Biomed Mater Res B Appl Biomater 2023; 111:1328-1335. [PMID: 36811266 DOI: 10.1002/jbm.b.35236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2022] [Revised: 12/29/2022] [Accepted: 01/30/2023] [Indexed: 02/24/2023]
Abstract
Acellular matrices are mainly composed of mammalian tissues, and aquatic tissues with lower biological risks and less religious restrictions are considered alternatives to mammalian tissues. The acellular fish skin matrix (AFSM) has been commercially available. Silver carp has the advantages of farmability, high yield and low price, but there are few studies on the silver carp acellular fish skin matrix (SC-AFSM). In this study, an acellular matrix with low DNA and endotoxin was prepared from the skin of silver carp. After treatment with trypsin/sodium dodecyl sulfate and Triton X-100 solutions, the DNA content in SC-AFSM reached 11.03 ± 0.85 ng/mg, and the endotoxin removal rate was 96.8%. The porosity of SC-AFSM was 79.64% ± 0.17%, which is favorable for cell infiltration and proliferation. The relative cell proliferation rate of SC-AFSM extract was 117.79% ± 15.26%. The wound healing experiment showed that SC-AFSM had no adverse acute pro-inflammatory response, which had a similar effect as commercial products in promoting tissue repair. Therefore, SC-AFSM has great application potential in biomaterials.
Collapse
Affiliation(s)
- Zeyu Wei
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang, China.,School of Food Science and Engineering, Jiangsu Ocean University, Lianyungang, China
| | - Junjie Zhang
- School of Food Science and Engineering, Jiangsu Ocean University, Lianyungang, China.,Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang, China.,Jiangsu Institute of Marine Resources Development, Jiangsu Ocean University, Lianyungang, China
| | - Zhiwen Guo
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang, China.,Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang, China.,College of Marine Science and Fisheries, Jiangsu Ocean University, Lianyungang, China
| | - Zhiming Wu
- School of Food Science and Engineering, Jiangsu Ocean University, Lianyungang, China
| | - Yaru Sun
- School of Food Science and Engineering, Jiangsu Ocean University, Lianyungang, China
| | - Ke Wang
- School of Food Science and Engineering, Jiangsu Ocean University, Lianyungang, China
| | - Rui Duan
- Jiangsu Key Laboratory of Marine Bioresources and Environment, Jiangsu Ocean University, Lianyungang, China.,Co-Innovation Center of Jiangsu Marine Bio-Industry Technology, Jiangsu Ocean University, Lianyungang, China.,College of Marine Science and Fisheries, Jiangsu Ocean University, Lianyungang, China
| |
Collapse
|
22
|
Long J, Qin Z, Chen G, Song B, Zhang Z. Decellularized extracellular matrix (d-ECM): the key role of the inflammatory process in pre-regeneration after implantation. Biomater Sci 2023; 11:1215-1235. [PMID: 36625281 DOI: 10.1039/d2bm01204a] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Clinical medicine is encountering the challenge of repairing soft-tissue defects. Currently, natural and synthetic materials have been developed as natural scaffolds. Among them, the decellularized extracellular matrix (d-ECM) can achieve tissue remodeling following injury and, thus, replace defects due to its advantages of the extensiveness of the source and excellent biological and mechanical properties. However, by analyzing the existing decellularization techniques, we found that different preparation methods directly affect the residual components of the d-ECM, and further have different effects on inflammation and regeneration of soft tissues. Therefore, we analyzed the role of different residual components of the d-ECM after decellularization. Then, we explored the inflammatory process and immune cells in an attempt to understand the mechanisms and causes of tissue degeneration and regeneration after transplantation. In this paper, we summarize the current studies related to updated protocols for the preparation of the d-ECM, biogenic and exogenous residual substances, inflammation, and immune cells influencing the fate of the d-ECM.
Collapse
Affiliation(s)
- Jie Long
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Zijin Qin
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Guo Chen
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Baoqiang Song
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| | - Ziang Zhang
- Department of Plastic and Reconstructive Surgery, Xijing Hospital, Fourth Military Medical University, Xi'an, Shaanxi, China.
| |
Collapse
|
23
|
McKenna E, Futrega K, Klein TJ, Altalhi TA, Popat A, Kumeria T, Doran MR. Spray nebulization enables polycaprolactone nanofiber production in a manner suitable for generation of scaffolds or direct deposition of nanofibers onto cells. Biofabrication 2023; 15:025003. [PMID: 36595260 DOI: 10.1088/1758-5090/aca5b7] [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: 09/24/2021] [Accepted: 11/24/2022] [Indexed: 11/27/2022]
Abstract
Spray nebulization is an elegant, but relatively unstudied, technique for scaffold production. Herein we fabricated mesh scaffolds of polycaprolactone (PCL) nanofibers via spray nebulization of 8% PCL in dichloromethane (DCM) using a 55.2 kPa compressed air stream and 17 ml h-1polymer solution flow rate. Using a refined protocol, we tested the hypothesis that spray nebulization would simultaneously generate nanofibers and eliminate solvent, yielding a benign environment at the point of fiber deposition that enabled the direct deposition of nanofibers onto cell monolayers. Nanofibers were collected onto a rotating plate 20 cm from the spray nozzle, but could be collected onto any static or moving surface. Scaffolds exhibited a mean nanofiber diameter of 910 ± 190 nm, ultimate tensile strength of 2.1 ± 0.3 MPa, elastic modulus of 3.3 ± 0.4 MPa, and failure strain of 62 ± 6%.In vitro, scaffolds supported growth of human keratinocyte cell epithelial-like layers, consistent with potential utility as a dermal scaffold. Fourier-transform infrared spectroscopy demonstrated that DCM had vaporized and was undetectable in scaffolds immediately following production. Exploiting the rapid elimination of DCM during fiber production, we demonstrated that nanofibers could be directly deposited on to cell monolayers, without compromising cell viability. This is the first description of spray nebulization generating nanofibers using PCL in DCM. Using this method, it is possible to rapidly produce nanofiber scaffolds, without need for high temperatures or voltages, yielding a method that could potentially be used to deposit nanofibers onto cell cultures or wound sites.
Collapse
Affiliation(s)
- Eamonn McKenna
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Queensland, Australia
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, Queensland, Australia
- Translational Research Institute, Brisbane, Queensland, Australia
| | - Kathryn Futrega
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Queensland, Australia
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, Queensland, Australia
- Translational Research Institute, Brisbane, Queensland, Australia
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States of America
| | - Travis J Klein
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Queensland, Australia
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology, Brisbane, Queensland, Australia
| | - Tariq A Altalhi
- Department of Chemistry, Faculty of Science, Taif University, Taif, Saudi Arabia
| | - Amirali Popat
- School of Pharmacy, University of Queensland, Brisbane, Queensland, Australia
| | - Tushar Kumeria
- School of Pharmacy, University of Queensland, Brisbane, Queensland, Australia
- School of Materials Science and Engineering, University of New South Wales, Sydney, Australia
- Australian Centre for Nanomedicine, University of New South Wales, Sydney, Australia
| | - Michael R Doran
- Centre for Biomedical Technologies, Queensland University of Technology, Brisbane, Queensland, Australia
- School of Biomedical Science, Queensland University of Technology, Brisbane, Queensland, Australia
- Translational Research Institute, Brisbane, Queensland, Australia
- National Institute of Dental and Craniofacial Research, National Institutes of Health, Bethesda, MD, United States of America
- Mater Research Institute-University of Queensland, Brisbane, Queensland, Australia
| |
Collapse
|
24
|
Crum RJ, Capella-Monsonís H, Chang J, Dewey MJ, Kolich BD, Hall KT, El-Mossier SO, Nascari DG, Hussey GS, Badylak SF. Biocompatibility and biodistribution of matrix-bound nanovesicles in vitro and in vivo. Acta Biomater 2023; 155:113-122. [PMID: 36423817 DOI: 10.1016/j.actbio.2022.11.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 11/11/2022] [Accepted: 11/15/2022] [Indexed: 11/23/2022]
Abstract
Matrix-bound nanovesicles (MBV) are a distinct subtype of extracellular vesicles that are firmly embedded within biomaterials composed of extracellular matrix (ECM). MBV both store and transport a diverse, tissue specific portfolio of signaling molecules including proteins, miRNAs, and bioactive lipids. MBV function as a key mediator in ECM-mediated control of the local tissue microenvironment. One of the most important mechanisms by which MBV in ECM bioscaffolds support constructive tissue remodeling following injury is immunomodulation and, specifically, the promotion of an anti-inflammatory, pro-remodeling immune cell activation state. Recent in vivo studies have shown that isolated MBV have therapeutic efficacy in rodent models of both retinal damage and rheumatoid arthritis through the targeted immunomodulation of pro-inflammatory macrophages towards an anti-inflammatory activation state. While these results show the therapeutic potential of MBV administered independent of the rest of the ECM, the in vitro and in vivo safety and biodistribution profile of MBV remain uncharacterized. The purpose of the present study was to thoroughly characterize the pre-clinical safety profile of MBV through a combination of in vitro cytotoxicity and MBV uptake studies and in vivo toxicity, immunotoxicity, and imaging studies. The results showed that MBV isolated from porcine urinary bladder are well-tolerated and are not cytotoxic in cell culture, are non-toxic to the whole organism, and are not immunosuppressive compared to the potent immunosuppressive drug cyclophosphamide. Furthermore, this safety profile was sustained across a wide range of MBV doses. STATEMENT OF SIGNIFICANCE: Matrix-bound nanovesicles (MBV) are a distinct subtype of bioactive extracellular vesicles that are embedded within biomaterials composed of extracellular matrix (ECM). Recent studies have shown therapeutic efficacy of MBV in models of both retinal damage and rheumatoid arthritis through the targeted immunomodulation of pro-inflammatory macrophages towards an anti-inflammatory activation state. While these results show the therapeutic potential of MBV, the in vitro and in vivo biocompatibility and biodistribution profile of MBV remain uncharacterized. The results of the present study showed that MBV are a well-tolerated ECM-derived therapy that are not cytotoxic in cell culture, are non-toxic to the whole organism, and are not immunosuppressive. Collectively, these data highlight the translational feasibility of MBV therapeutics across a wide variety of clinical applications.
Collapse
Affiliation(s)
- Raphael J Crum
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Héctor Capella-Monsonís
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, USA; Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Jordan Chang
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Marley J Dewey
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, USA; Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Brian D Kolich
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Kelsey T Hall
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, USA; Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Salma O El-Mossier
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, USA; Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, USA
| | - David G Nascari
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, USA
| | - George S Hussey
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, USA; Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Stephen F Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, USA; Department of Surgery, School of Medicine, University of Pittsburgh, Pittsburgh, USA; Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, USA; Department of Bioengineering, University of Pittsburgh, Pittsburgh, USA.
| |
Collapse
|
25
|
Abstract
The low regenerative potential of the human body hinders proper regeneration of dysfunctional or lost tissues and organs due to trauma, congenital defects, and diseases. Tissue or organ transplantation has hence been a major conventional option for replacing the diseased or dysfunctional body parts of the patients. In fact, a great number of patients on waiting lists would benefit tremendously if tissue and organs could be replaced with biomimetic spare parts on demand. Herein, regenerative medicine and advanced biomaterials strive to reach this distant goal. Tissue engineering aims to create new biological tissue or organ substitutes, and promote regeneration of damaged or diseased tissue and organs. This approach has been jointly evolving with the major advances in biomaterials, stem cells, and additive manufacturing technologies. In particular, three-dimensional (3D) bioprinting utilizes 3D printing to fabricate viable tissue-like structures (perhaps organs in the future) using bioinks composed of special hydrogels, cells, growth factors, and other bioactive contents. A third generation of multifunctional biomaterials could also show opportunities for building biomimetic scaffolds, upon which to regenerate stem cells in vivo. Besides, decellularization technology based on isolation of extracellular matrix of tissue and organs from their inhabiting cells is presented as an alternative to synthetic biomaterials. Today, the gained knowledge of functional microtissue engineering and biointerfaces, along with the remarkable advances in pluripotent stem cell technology, seems to be instrumental for the development of more realistic microphysiological 3D in vitro tissue models, which can be utilized for personalized disease modeling and drug development. This chapter will discuss the recent advances in the field of regenerative medicine and biomaterials, alongside challenges, limitations, and potentials of the current technologies.
Collapse
Affiliation(s)
- Şükran Şeker
- Ankara University Faculty of Science, and Ankara University Stem Cell Institute, Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara, Turkey
| | - Ayşe Eser Elçin
- Ankara University Faculty of Science, and Ankara University Stem Cell Institute, Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara, Turkey
| | - Yaşar Murat Elçin
- Ankara University Faculty of Science, Tissue Engineering, Biomaterials and Nanobiotechnology Laboratory, Ankara, Turkey.
- Biovalda Health Technologies, Inc., Ankara, Turkey.
| |
Collapse
|
26
|
Xuan Y, Li L, Zhang C, Zhang M, Cao J, Zhang Z. The 3D-Printed Ordered Bredigite Scaffold Promotes Pro-Healing of Critical-Sized Bone Defects by Regulating Macrophage Polarization. Int J Nanomedicine 2023; 18:917-932. [PMID: 36844434 PMCID: PMC9951604 DOI: 10.2147/ijn.s393080] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Accepted: 01/29/2023] [Indexed: 02/22/2023] Open
Abstract
Background Repairing critical-sized bone defects secondary to traumatic or tumorous damage is a complex conundrum in clinical practice; in this case, artificial scaffolds exhibited preferable outcomes. Bredigite (BRT, Ca7MgSi4O16) bioceramic possesses excellent physicochemical properties and biological activity as a promising candidate for bone tissue engineering. Methods Structurally ordered BRT (BRT-O) scaffolds were fabricated by a three-dimensional (3D) printing technique, and the random BRT (BRT-R) scaffolds and clinically available β-tricalcium phosphate (β-TCP) scaffolds were compared as control groups. Their physicochemical properties were characterized, and RAW 264.7 cells, bone marrow mesenchymal stem cells (BMSCs), and rat cranial critical-sized bone defect models were utilized for evaluating macrophage polarization and bone regeneration. Results The BRT-O scaffolds exhibited regular morphology and homogeneous porosity. In addition, the BRT-O scaffolds released higher concentrations of ionic products based on coordinated biodegradability than the β-TCP scaffolds. In vitro, the BRT-O scaffolds facilitated RWA264.7 cells polarization to pro-healing M2 macrophage phenotype, whereas the BRT-R and β-TCP scaffolds stimulated more pro-inflammatory M1-type macrophages. A conditioned medium derived from macrophages seeding on the BRT-O scaffolds notably promoted the osteogenic lineage differentiation of BMSCs in vitro. The cell migration ability of BMSCs was significantly enhanced under the BRT-O-induced immune microenvironment. Moreover, in rat cranial critical-sized bone defect models, the BRT-O scaffolds group promoted new bone formation with a higher proportion of M2-type macrophage infiltration and expression of osteogenesis-related markers. Therefore, in vivo, BRT-O scaffolds play immunomodulatory roles in promoting critical-sized bone defects by enhancing the polarization of M2 macrophages. Conclusion 3D-printed BRT-O scaffolds can be a promising option for bone tissue engineering, at least partly through macrophage polarization and osteoimmunomodulation.
Collapse
Affiliation(s)
- Yaowei Xuan
- Department of Stomatology, The First Medical Centre, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Lin Li
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Clinical Research Center for Oral Diseases, Department of Periodontology, School of Stomatology, Fourth Military Medical University, Xi'an, People's Republic of China
| | - Chenping Zhang
- Department of Oral Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China
| | - Min Zhang
- Department of Stomatology, The First Medical Centre, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Junkai Cao
- Department of Stomatology, The First Medical Centre, Chinese PLA General Hospital, Beijing, People's Republic of China
| | - Zhen Zhang
- Department of Oral Maxillofacial-Head Neck Oncology, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People's Republic of China.,College of Stomatology, Shanghai Jiao Tong University, National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai, People's Republic of China
| |
Collapse
|
27
|
Muacevic A, Adler JR, Ajmal M, Nawaz G. Organ Regeneration Through Stem Cells and Tissue Engineering. Cureus 2023; 15:e34336. [PMID: 36865965 PMCID: PMC9973391 DOI: 10.7759/cureus.34336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/29/2023] [Indexed: 01/30/2023] Open
Abstract
Loss of organ and tissue due to injuries or diseases led to the development of regenerative therapies to decrease reliance on organ transplantations. It deals with employing the self-renewal ability of stem cells to differentiate into numerous lineages to assist in providing effective treatment for a range of various injuries and diseases. Regenerative engineering of organs or tissues represents an ever-expanding field that is aimed at developing biological replacements for dysfunctional organs or injured tissues. The critical issue, however, with the engineering of organs outside the human body is the insufficient availability of human cells, the absence of a suitable matrix with the same architecture and composition as the target tissue, and the maintenance of organ viability in the absence of the blood supply. The issue regarding the maintenance of the engineered organ viability can be solved using bioreactors consisting of mediums with defined chemical composition, i.e., nutrients, cofactors, and growth factors that can successively sustain the target cell's viability. Engineered extracellular matrices and stem cells to regenerate organs outside the human body are also being used. Clinically, various adult stem cell therapies are readily under practice. This review will focus on the regeneration of organs through various types of stem cells and tissue engineering techniques.
Collapse
|
28
|
Xie X, Wu Q, Liu Y, Chen C, Chen Z, Xie C, Song M, Jiang Z, Qi X, Liu S, Tang Z, Wu Z. Vascular endothelial growth factor attenuates neointimal hyperplasia of decellularized small-diameter vascular grafts by modulating the local inflammatory response. Front Bioeng Biotechnol 2022; 10:1066266. [PMID: 36605251 PMCID: PMC9808043 DOI: 10.3389/fbioe.2022.1066266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2022] [Accepted: 12/09/2022] [Indexed: 12/24/2022] Open
Abstract
Small-diameter vascular grafts (diameter <6 mm) are in high demand in clinical practice. Neointimal hyperplasia, a common complication after implantation of small-diameter vascular grafts, is one of the common causes of graft failure. Modulation of local inflammatory responses is a promising strategy to attenuates neointimal hyperplasia. Vascular endothelial growth factor (VEGF) is an angiogenesis stimulator that also induces macrophage polarization and modulates inflammatory responses. In the present study, we evaluated the effect of VEGF on the neointima hyperplasia and local inflammatory responses of decellularized vascular grafts. In the presence of rhVEGF-165 in RAW264.6 macrophage culture, rhVEGF-165 induces RAW264.6 macrophage polarization to M2 phenotype. Decellularized bovine internal mammary arteries were implanted into the subcutaneous and infrarenal abdominal aorta of New Zealand rabbits, with rhVEGF-165 applied locally to the adventitial of the grafts. The vascular grafts were removed en-bloc and submitted to histological and immunofluorescence analyses on days 7 and 28 following implantation. The thickness of the fibrous capsule and neointima was thinner in the VEGF group than that in the control group. In the immunofluorescence analysis, the number of M2 macrophages and the ratio of M2/M1 macrophages in vascular grafts in the VEGF group were higher than those in the control group, and the proinflammatory factor IL-1 was expressed less than in the control group, but the anti-inflammatory factor IL-10 was expressed more. In conclusion, local VEGF administration attenuates neointimal hyperplasia in decellularized small-diameter vascular grafts by inducing macrophage M2 polarization and modulating the inflammatory response.
Collapse
Affiliation(s)
- Xinlong Xie
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China,Department of Cardiothoracic Surgery, The First Affiliated Hospital, Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Qiying Wu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuhong Liu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chunyang Chen
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zeguo Chen
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Chao Xie
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Mingzhe Song
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhenlin Jiang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Xiaoke Qi
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Sixi Liu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Zhenjie Tang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China,Engineering Laboratory of Hunan Province for Cardiovascular Biomaterials, Changsha, Hunan, China
| | - Zhongshi Wu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China,Engineering Laboratory of Hunan Province for Cardiovascular Biomaterials, Changsha, Hunan, China,*Correspondence: Zhongshi Wu,
| |
Collapse
|
29
|
Mendes C, Thirupathi A, Corrêa MEAB, Gu Y, Silveira PCL. The Use of Metallic Nanoparticles in Wound Healing: New Perspectives. Int J Mol Sci 2022; 23:15376. [PMID: 36499707 PMCID: PMC9740811 DOI: 10.3390/ijms232315376] [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] [Received: 10/27/2022] [Revised: 11/19/2022] [Accepted: 12/02/2022] [Indexed: 12/12/2022] Open
Abstract
Chronic wounds represent a challenge for the health area, as they directly impact patients' quality of life and represent a threat to public health and the global economy due to their high cost of treatment. Alternative strategies must be developed for cost-effective and targeted treatment. In this scenario, the emerging field of nanobiotechnology may provide an alternative platform to develop new therapeutic agents for the chronic wound healing process. This manuscript aims to demonstrate that the application of metallic nanoparticles (gold, silver, copper, and zinc oxide) opened a new chapter in the treatment of wounds, as they have different properties such as drug delivery, antimicrobial activity, and healing acceleration. Furthermore, metallic nanoparticles (NPs) produced through green synthesis ensure less toxicity in biological tissues, and greater safety of applicability, other than adding the effects of NPs with those of extracts.
Collapse
Affiliation(s)
- Carolini Mendes
- Faculty of Sports Science, Ningbo University, Ningbo 315211, China
- Laboratory of Experimental Phisiopatology, Program of Postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Criciúma 88806-000, Brazil
| | - Anand Thirupathi
- Faculty of Sports Science, Ningbo University, Ningbo 315211, China
| | - Maria E A B Corrêa
- Laboratory of Experimental Phisiopatology, Program of Postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Criciúma 88806-000, Brazil
| | - Yaodong Gu
- Faculty of Sports Science, Ningbo University, Ningbo 315211, China
| | - Paulo C L Silveira
- Faculty of Sports Science, Ningbo University, Ningbo 315211, China
- Laboratory of Experimental Phisiopatology, Program of Postgraduate in Science of Health, Universidade do Extremo Sul Catarinense, Criciúma 88806-000, Brazil
| |
Collapse
|
30
|
Bate TSR, Shanahan W, Casillo JP, Grant R, Forbes SJ, Callanan A. Rat liver ECM incorporated into electrospun polycaprolactone scaffolds as a platform for hepatocyte culture. J Biomed Mater Res B Appl Biomater 2022; 110:2612-2623. [PMID: 35734943 PMCID: PMC9796056 DOI: 10.1002/jbm.b.35115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Revised: 05/13/2022] [Accepted: 06/08/2022] [Indexed: 12/30/2022]
Abstract
Liver disease is expanding across the globe; however, health-care systems still lack approved pharmaceutical treatment strategies to mitigate potential liver failures. Organ transplantation is the only treatment for liver failure and with increasing cases of liver disease, transplant programs increasingly cannot provide timely transplant availability for all patients. The development of pharmaceutical mitigation strategies is clearly necessary and methods to improve drug development processes are considered vital for this purpose. Herein, we present a methodology for incorporating whole organ decellularised rat liver ECM (rLECM) into polycaprolactone (PCL) electrospun scaffolds with the aim of producing biologically relevant liver tissue models. rLECM PCL scaffolds have been produced with 5 w/w% and 10 w/w% rLECM:PCL and were analyzed by SEM imaging, tensile mechanical analyses and FTIR spectroscopy. The hepatocellular carcinoma cell line, HepG2, was cultured upon the scaffolds for 14 days and were analyzed through cell viability assay, DNA quantification, albumin quantification, immunohistochemistry, and RT-qPCR gene expression analysis. Results showed significant increases in proliferative activity of HepG2 on rLECM containing scaffolds alongside maintained key gene expression. This study confirms that rLECM can be utilized to modulate the bioactivity of electrospun PCL scaffolds and has the potential to produce electrospun scaffolds suitable for enhanced hepatocyte cultures and in-vitro liver tissue models.
Collapse
Affiliation(s)
- Thomas S. R. Bate
- School of EngineeringInstitute for Bioengineering, University of EdinburghEdinburghUK
| | - William Shanahan
- School of EngineeringInstitute for Bioengineering, University of EdinburghEdinburghUK
| | - Joseph P. Casillo
- School of GeoSciencesUniversity of Edinburgh, Grant InstituteEdinburghUK
| | - Rhiannon Grant
- MERLN InstituteMaastricht UniversityMaastrichtThe Netherlands
| | - Stuart J. Forbes
- Centre for Regenerative MedicineUniversity of EdinburghEdinburghUK
| | - Anthony Callanan
- School of EngineeringInstitute for Bioengineering, University of EdinburghEdinburghUK
| |
Collapse
|
31
|
Wang B, Sierad LN, Mercuri JJ, Simionescu A, Simionescu DT, Williams LN, Vela R, Bajona P, Peltz M, Ramaswamy S, Hong Y, Liao J. Structural and biomechanical characterizations of acellular porcine mitral valve scaffolds: anterior leaflets, posterior leaflets, and chordae tendineae. ENGINEERED REGENERATION 2022; 3:374-386. [PMID: 38362305 PMCID: PMC10869114 DOI: 10.1016/j.engreg.2022.08.003] [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] [Indexed: 11/15/2022] Open
Abstract
Mitral valve (MV) tissue engineering is still in its early stage, and one major challenge in MV tissue engineering is to identify appropriate scaffold materials. With the potential of acellular MV scaffolds being demonstrated recently, it is important to have a full understanding of the biomechanics of the native MV components and their acellular scaffolds. In this study, we have successfully characterized the structural and mechanical properties of porcine MV components, including anterior leaflet (AL), posterior leaflet (PL), strut chordae, and basal chordae, before and after decellularization. Quantitative DNA assay showed more than 90% reduction in DNA content, and Griffonia simplicifolia (GS) lectin immunohistochemistry confirmed the complete lack of porcine α-Gal antigen in the acellular MV components. In the acellular AL and PL, the atrialis, spongiosa, and fibrosa trilayered structure, along with its ECM constitutes, i.e., collagen fibers, elastin fibers, and portion of GAGs, were preserved. Nevertheless, the ECM of both AL and PL experienced a certain degree of disruption, exhibiting a less dense, porous ECM morphology. The overall anatomical morphology of the strut and basal chordae were also maintained after decellularization, with longitudinal morphology experiencing minimum disruption, but the cross-sectional morphology exhibiting evenly-distributed porous structure. In the acellular AL and PL, the nonlinear anisotropic biaxial mechanical behavior was overall preserved; however, uniaxial tensile tests showed that the removal of cellular content and the disruption of structural ECM did result in small decreases in maximum tensile modulus, tissue extensibility, failure stress, and failure strain for both MV leaflets and chordae.
Collapse
Affiliation(s)
- Bo Wang
- Joint Department of Biomedical Engineering, Medical College of Wisconsin and Marquette University, Milwaukee, WI 53226, United States
| | - Leslie N. Sierad
- Department of Bioengineering, Clemson University, Clemson, SC 29634, United States
| | - Jeremy J. Mercuri
- Department of Bioengineering, Clemson University, Clemson, SC 29634, United States
| | - Agneta Simionescu
- Department of Bioengineering, Clemson University, Clemson, SC 29634, United States
| | - Dan T. Simionescu
- Department of Bioengineering, Clemson University, Clemson, SC 29634, United States
| | - Lakiesha N. Williams
- Department of Biomedical Engineering, University of Florida, Gainesville, FL 32611, United States
| | - Ryan Vela
- Department of Cardiovascular and Thoracic Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Pietro Bajona
- Department of Cardiovascular and Thoracic Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
- Allegheny Health Network-Drexel University College of Medicine, Pittsburgh, PA 15212, United States
| | - Matthias Peltz
- Department of Cardiovascular and Thoracic Surgery, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States
| | - Sharan Ramaswamy
- Department of Biomedical Engineering, Florida International University, Miami, FL 33174, United States
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76010, United States
| | - Jun Liao
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76010, United States
| |
Collapse
|
32
|
Outcome of a novel porcine-derived UBM/SIS composite biological mesh in a rabbit vaginal defect model. Int Urogynecol J 2022:10.1007/s00192-022-05400-5. [DOI: 10.1007/s00192-022-05400-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 10/10/2022] [Indexed: 12/13/2022]
|
33
|
Peshkova M, Kosheleva N, Shpichka A, Radenska-Lopovok S, Telyshev D, Lychagin A, Li F, Timashev P, Liang XJ. Targeting Inflammation and Regeneration: Scaffolds, Extracellular Vesicles, and Nanotechnologies as Cell-Free Dual-Target Therapeutic Strategies. Int J Mol Sci 2022; 23:13796. [PMID: 36430272 PMCID: PMC9694395 DOI: 10.3390/ijms232213796] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Revised: 10/25/2022] [Accepted: 11/02/2022] [Indexed: 11/11/2022] Open
Abstract
Osteoarthritis (OA) affects over 250 million people worldwide and despite various existing treatment strategies still has no cure. It is a multifactorial disease characterized by cartilage loss and low-grade synovial inflammation. Focusing on these two targets together could be the key to developing currently missing disease-modifying OA drugs (DMOADs). This review aims to discuss the latest cell-free techniques applied in cartilage tissue regeneration, since they can provide a more controllable approach to inflammation management than the cell-based ones. Scaffolds, extracellular vesicles, and nanocarriers can be used to suppress inflammation, but they can also act as immunomodulatory agents. This is consistent with the latest tissue engineering paradigm, postulating a moderate, controllable inflammatory reaction to be beneficial for tissue remodeling and successful regeneration.
Collapse
Affiliation(s)
- Maria Peshkova
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov University, 119991 Moscow, Russia
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
| | - Nastasia Kosheleva
- Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- FSBSI Institute of General Pathology and Pathophysiology, 125315 Moscow, Russia
| | - Anastasia Shpichka
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov University, 119991 Moscow, Russia
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- Chemistry Department, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Stefka Radenska-Lopovok
- Institute for Clinical Morphology and Digital Pathology, Sechenov University, 119991 Moscow, Russia
| | - Dmitry Telyshev
- Institute of Biomedical Systems, National Research University of Electronic Technology, 124498 Moscow, Russia
- Institute of Bionic Technologies and Engineering, Sechenov University, 119991 Moscow, Russia
| | - Alexey Lychagin
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- Department of Traumatology, Orthopedics and Disaster Surgery, Sechenov University, 119991 Moscow, Russia
| | - Fangzhou Li
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
| | - Peter Timashev
- World-Class Research Center “Digital Biodesign and Personalized Healthcare”, Sechenov University, 119991 Moscow, Russia
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- Chemistry Department, Lomonosov Moscow State University, 119991 Moscow, Russia
| | - Xing-Jie Liang
- Laboratory of Clinical Smart Nanotechnologies, Institute for Regenerative Medicine, Sechenov University, 119991 Moscow, Russia
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center for Excellence in Nanoscience, National Center for Nanoscience and Technology of China, Beijing 100190, China
- University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
34
|
Barbulescu GI, Bojin FM, Ordodi VL, Goje ID, Barbulescu AS, Paunescu V. Decellularized Extracellular Matrix Scaffolds for Cardiovascular Tissue Engineering: Current Techniques and Challenges. Int J Mol Sci 2022; 23:13040. [PMID: 36361824 PMCID: PMC9658138 DOI: 10.3390/ijms232113040] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Revised: 10/18/2022] [Accepted: 10/26/2022] [Indexed: 08/13/2023] Open
Abstract
Cardiovascular diseases are the leading cause of global mortality. Over the past two decades, researchers have tried to provide novel solutions for end-stage heart failure to address cardiac transplantation hurdles such as donor organ shortage, chronic rejection, and life-long immunosuppression. Cardiac decellularized extracellular matrix (dECM) has been widely explored as a promising approach in tissue-regenerative medicine because of its remarkable similarity to the original tissue. Optimized decellularization protocols combining physical, chemical, and enzymatic agents have been developed to obtain the perfect balance between cell removal, ECM composition, and function maintenance. However, proper assessment of decellularized tissue composition is still needed before clinical translation. Recellularizing the acellular scaffold with organ-specific cells and evaluating the extent of cardiomyocyte repopulation is also challenging. This review aims to discuss the existing literature on decellularized cardiac scaffolds, especially on the advantages and methods of preparation, pointing out areas for improvement. Finally, an overview of the state of research regarding the application of cardiac dECM and future challenges in bioengineering a human heart suitable for transplantation is provided.
Collapse
Affiliation(s)
- Greta Ionela Barbulescu
- Immuno-Physiology and Biotechnologies Center (CIFBIOTEH), Department of Functional Sciences, “Victor Babes” University of Medicine and Pharmacy, No 2 Eftimie Murgu Square, 300041 Timisoara, Romania
- Department of Clinical Practical Skills, “Victor Babes” University of Medicine and Pharmacy, No 2 Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Florina Maria Bojin
- Immuno-Physiology and Biotechnologies Center (CIFBIOTEH), Department of Functional Sciences, “Victor Babes” University of Medicine and Pharmacy, No 2 Eftimie Murgu Square, 300041 Timisoara, Romania
- Clinical Emergency County Hospital “Pius Brinzeu” Timisoara, Center for Gene and Cellular Therapies in the Treatment of Cancer Timisoara-OncoGen, No 156 Liviu Rebreanu, 300723 Timisoara, Romania
| | - Valentin Laurentiu Ordodi
- Clinical Emergency County Hospital “Pius Brinzeu” Timisoara, Center for Gene and Cellular Therapies in the Treatment of Cancer Timisoara-OncoGen, No 156 Liviu Rebreanu, 300723 Timisoara, Romania
- Faculty of Industrial Chemistry and Environmental Engineering, “Politehnica” University Timisoara, No 2 Victoriei Square, 300006 Timisoara, Romania
| | - Iacob Daniel Goje
- Department of Medical Semiology I, “Victor Babes” University of Medicine and Pharmacy, No 2 Eftimie Murgu Square, 300041 Timisoara, Romania
- Advanced Cardiology and Hemostaseology Research Center, “Victor Babes” University of Medicine and Pharmacy, No 2 Eftimie Murgu Square, 300041 Timisoara, Romania
| | - Andreea Severina Barbulescu
- Center for Advanced Research in Gastroenterology and Hepatology, Department of Internal Medicine II, Division of Gastroenterology and Hepatology, “Victor Babes” University of Medicine and Pharmacy, 300041 Timisoara, Romania
| | - Virgil Paunescu
- Immuno-Physiology and Biotechnologies Center (CIFBIOTEH), Department of Functional Sciences, “Victor Babes” University of Medicine and Pharmacy, No 2 Eftimie Murgu Square, 300041 Timisoara, Romania
- Clinical Emergency County Hospital “Pius Brinzeu” Timisoara, Center for Gene and Cellular Therapies in the Treatment of Cancer Timisoara-OncoGen, No 156 Liviu Rebreanu, 300723 Timisoara, Romania
| |
Collapse
|
35
|
Haghdel M, Imanieh MH, Hosseinpour H, Ghasemi Y, Alizadeh AA. Development of Bio-artificial Esophageal Tissue Engineering Utilization for Circumferential Lesion Transplantation: A Narrative Review. IRANIAN JOURNAL OF MEDICAL SCIENCES 2022; 47:406-421. [PMID: 36117582 PMCID: PMC9445863 DOI: 10.30476/ijms.2021.89194.1991] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 02/12/2021] [Accepted: 04/06/2021] [Indexed: 11/19/2022]
Abstract
The esophagus is the gastrointestinal tract's primary organ that transfers bolus into the stomach with peristaltic motion. Therefore, its lesions cause a significant disturbance in the nutrition and digestive system. Esophageal disease treatment sometimes requires surgical procedures that involve removal and circumferential full-thickness replacement. Unlike other organs, the esophagus has a limited regeneration ability and cannot be transplanted from donors. There are various methods of restoring the esophageal continuity; however, they are associated with certain flaws that lead to a non-functional recovery. As an exponentially growing science, tissue engineering has become a leading technique for the development of tissue replacement to repair damaged esophageal segments. Scaffold plays a significant role in the process of tissue engineering, as it acts as a template for the regeneration of growing tissue. A variety of scaffolds have been studied to replace the esophagus. Due to the many tissue quality challenges, the results are still inadequate and need to be improved. The success of esophageal tissue regeneration will finally depend on the scaffold's capability to mimic natural tissue properties and provide a qualified environment for regeneration. Thereby, scaffold fabrication techniques are fundamental. This article reviews the recent developments in esophageal tissue engineering for the treatment of circumferential lesions based on scaffold biomaterial engineering approaches.
Collapse
Affiliation(s)
- Mobin Haghdel
- Department of Tissue Engineering, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hadi Imanieh
- Department of Pediatrics Gastroenterology, School of Medicine, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Hamidreza Hosseinpour
- Department of Surgery, Shiraz Laparoscopic Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Younes Ghasemi
- Department of Pharmaceutical Biotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Akbar Alizadeh
- Department of Tissue Engineering, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
36
|
Inflammation-mediated matrix remodeling of extracellular matrix-mimicking biomaterials in tissue engineering and regenerative medicine. Acta Biomater 2022; 151:106-117. [PMID: 35970482 DOI: 10.1016/j.actbio.2022.08.015] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2022] [Revised: 06/30/2022] [Accepted: 08/08/2022] [Indexed: 12/12/2022]
Abstract
Extracellular matrix (ECM)-mimicking biomaterials are considered effective tissue-engineered scaffolds for regenerative medicine because of their biocompatibility, biodegradability, and bioactivity. ECM-mimicking biomaterials preserve natural microstructures and matrix-related bioactive components and undergo continuous matrix remodeling upon transplantation. The interaction between host immune cells and transplanted ECM-mimicking biomaterials has attracted considerable attention in recent years. Transplantation of biomaterials may initiate injuries and early pro-inflammation reactions characterized by infiltration of neutrophils and M1 macrophages. Pro-inflammation reactions may lead to degradation of the transplanted biomaterial and drive the matrix into a fetal-like state. ECM degradation leads to the release of matrix-related bioactive components that act as signals for cell migration, proliferation, and differentiation. In late stages, pro-inflammatory cells fade away, and anti-inflammatory cells emerge, which involves macrophage polarization to the M2 phenotype and leukocyte activation to T helper 2 (Th2) cells. These anti-inflammatory cells interact with each other to facilitate matrix deposition and tissue reconstruction. Deposited ECM molecules serve as vital components of the mature tissue and influence tissue homeostasis. However, dysregulation of matrix remodeling results in several pathological conditions, such as aggressive inflammation, difficult healing, and non-functional fibrosis. In this review, we summarize the characteristics of inflammatory responses in matrix remodeling after transplantation of ECM-mimicking biomaterials. Additionally, we discuss the intrinsic linkages between matrix remodeling and tissue regeneration. STATEMENT OF SIGNIFICANCE: Extracellular matrix (ECM)-mimicking biomaterials are effectively used as scaffolds in tissue engineering and regenerative medicine. However, dysregulation of matrix remodeling can cause various pathological conditions. Here, the review describes the characteristics of inflammatory responses in matrix remodeling after transplantation of ECM-mimicking biomaterials. Additionally, we discuss the intrinsic linkages between matrix remodeling and tissue regeneration. We believe that understanding host immune responses to matrix remodeling of transplanted biomaterials is important for directing effective tissue regeneration of ECM-mimicking biomaterials. Considering the close relationship between immune response and matrix remodeling results, we highlight the need for studies of the effects of clinical characteristics on matrix remodeling of transplanted biomaterials.
Collapse
|
37
|
Wu H, Yin G, Pu X, Wang J, Liao X, Huang Z. Preliminary Study on the Antigen-Removal from Extracellular Matrix via Different Decellularization. Tissue Eng Part C Methods 2022; 28:250-263. [PMID: 35596569 DOI: 10.1089/ten.tec.2022.0025] [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] [Indexed: 12/12/2022] Open
Abstract
Due to the abundance of bioactive components, surficial decoration with cell-derived extracellular matrix (ECM) is a promising strategy to improve the biological functionality of the tissue engineering scaffolds. However, decellularization is necessary to remove antigenic components in the ECM that may trigger adverse immune response. Freeze-thaw (FT) cycles and treatment with Triton X-100/ammonium hydroxide (TN) are two commonly used decellularization methods for ECM, but their effects on both growth factor retention and antigen removal are still controversial. The objectives of this study are to compare the preservation of ECM texture and beneficial ingredients and the removal of cellular antigens by these two methods. First, the constructs combined bone marrow mesenchymal stem cell-derived ECM and poly(lactic-co-glycolic acid) (PLGA) membrane are prepared and decellularized using FT and TN treatments. Moreover, the effects of decellularization on the ultrastructure and the composition of ECM-decorated PLGA membrane are compared by scanning electron microscope observation and protein quantification. Furthermore, the ECM deposited on PLGA is stripped off and then implanted subcutaneously in rats, and the host macrophage and local lymphocyte responses were investigated. Finally, ECM-decorated porous PLGA scaffolds are implanted into rat calvarial defects, and the new bone formation is evaluated. Our results showed that both methods effectively removed DNA. TN treatment partially retained collagen, glycosaminoglycan, bone morphogenetic protein-2, and vascular endothelial growth factor, and better preserved structural integrity than FT treatment. ECM implants decellularized by both methods induced a mild host response after subcutaneous implantation. Although the total content of residual DNA in the two ECMs digested by the DNA enzyme seemed to be similar and very low, the interfaces between implanted materials and natural tissues in the TN group recruited lower numbers of CD68+ macrophages, CD68+CD86+ (M1) macrophages, and CD4+ T lymphocytes than that in FT group, implying that there exist other ECM antigens to influence immune response besides DNA. Furthermore, ECM-decorated scaffolds decellularized by TN treatment induced greater bone formation than that of bare scaffolds in vivo, demonstrating the effective retention of ECM bioactive components after decellularization. This study showed that TN treatment was a more effective and safer decellularization method than FT cycles. Impact statement Decellularization is a prerequisite for extracellular matrix (ECM) application, but there is still no standard for its selection. This study demonstrated that detergent treatment was more effective than freeze-thaw (FT) cycles in removing ECM antigens besides DNA, and the prepared ECM elicited a milder allogenic immune response, which ensured the safety of ECM. Moreover, detergent better preserved the ECM integrity than FT cycles, and effectively retained growth factors, and the decellularized ECM-decorated scaffolds significantly promoted bone repair, which ensured the effectiveness of ECM. This study provides the theoretical and experimental bases for the decellularization strategy of ECM-modified tissue engineering scaffolds.
Collapse
Affiliation(s)
- Huan Wu
- College of Biomedical Engineering, Sichuan University, Chengdu, P.R. China
| | - Guangfu Yin
- College of Biomedical Engineering, Sichuan University, Chengdu, P.R. China
| | - Ximing Pu
- College of Biomedical Engineering, Sichuan University, Chengdu, P.R. China
| | - Juan Wang
- College of Biomedical Engineering, Sichuan University, Chengdu, P.R. China
| | - Xiaoming Liao
- College of Biomedical Engineering, Sichuan University, Chengdu, P.R. China
| | - Zhongbing Huang
- College of Biomedical Engineering, Sichuan University, Chengdu, P.R. China
| |
Collapse
|
38
|
Boehm AK, Hillebrandt KH, Dziodzio T, Krenzien F, Neudecker J, Spuler S, Pratschke J, Sauer IM, Andreas MN. Tissue engineering for the diaphragm and its various therapeutic possibilities – A Systematic Review. ADVANCED THERAPEUTICS 2022. [DOI: 10.1002/adtp.202100247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/09/2022]
Affiliation(s)
- Agnes K Boehm
- Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt‐Universität zu Berlin Department of Surgery Augustenburger Platz 1 Berlin 13353 Germany
| | - Karl H Hillebrandt
- Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt‐Universität zu Berlin Department of Surgery Augustenburger Platz 1 Berlin 13353 Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin Charitéplatz 1 Berlin 10117 Germany
| | - Tomasz Dziodzio
- Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt‐Universität zu Berlin Department of Surgery Augustenburger Platz 1 Berlin 13353 Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin Charitéplatz 1 Berlin 10117 Germany
| | - Felix Krenzien
- Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt‐Universität zu Berlin Department of Surgery Augustenburger Platz 1 Berlin 13353 Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin Charitéplatz 1 Berlin 10117 Germany
| | - Jens Neudecker
- Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt‐Universität zu Berlin Department of Surgery Augustenburger Platz 1 Berlin 13353 Germany
| | - Simone Spuler
- Muscle Research Unit Experimental and Clinical Research Center Charité Universitätsmedizin Berlin and Max‐Delbrück‐Centrum für Molekulare Medizin in der Helmholtz‐Gemeinschaft Lindenberger Weg 80 Berlin 13125 Germany
| | - Johann Pratschke
- Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt‐Universität zu Berlin Department of Surgery Augustenburger Platz 1 Berlin 13353 Germany
- Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin Cluster of Excellence Matters of Activity. Image Space Material funded by the Deutsche Forschungsgemeinschaft (DFG German Research Foundation) under Germany's Excellence Strategy Berlin EXC 2025 Germany
| | - Igor M Sauer
- Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt‐Universität zu Berlin Department of Surgery Augustenburger Platz 1 Berlin 13353 Germany
- Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt Universität zu Berlin Cluster of Excellence Matters of Activity. Image Space Material funded by the Deutsche Forschungsgemeinschaft (DFG German Research Foundation) under Germany's Excellence Strategy Berlin EXC 2025 Germany
| | - Marco N Andreas
- Charité – Universitätsmedizin Berlin corporate member of Freie Universität Berlin and Humboldt‐Universität zu Berlin Department of Surgery Augustenburger Platz 1 Berlin 13353 Germany
| |
Collapse
|
39
|
Woodard DA, Kim G, Nilsson KR. Risk profiles and outcomes of patients receiving antibacterial cardiovascular implantable electronic device envelopes: A retrospective analysis. World J Cardiol 2022; 14:177-186. [PMID: 35432770 PMCID: PMC8968457 DOI: 10.4330/wjc.v14.i3.177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Revised: 02/08/2022] [Accepted: 03/17/2022] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Cardiovascular implantable electronic devices (CIEDs) are implanted in an increasing number of patients each year, which has led to an increase in the risk of CIED infection. Antibacterial CIED envelopes locally deliver antibiotics to the implant site over a short-term period and have been shown to reduce the risk of implant site infection. These envelopes are derived from either biologic or non-biologic materials. There is a paucity of data examining patient risk profiles and outcomes from using these envelope materials in the clinical setting and comparing these results to patients receiving no envelope with their CIED implantation.
AIM To evaluate risk profiles and outcomes of patients who underwent CIED procedures with an antibacterial envelope or no envelope.
METHODS After obtaining Internal Review Board approval, the records of consecutive patients who underwent a CIED implantation procedure by a single physician between March 2017 and December 2019 were retrospectively collected from our hospital. A total of 248 patients within this period were identified and reviewed through 12 mo of follow up. The CIED procedures used either no envelope (n = 57), a biologic envelope (CanGaroo®, Aziyo Biologics) that was pre-hydrated by the physician with vancomycin and gentamicin (n = 89), or a non-biologic envelope (Tyrx™, Medtronic) that was coated with a resorbable polymer containing the drug substances rifampin and minocycline by the manufacturer (n = 102). Patient selection for receiving either no envelope or an envelope (and which envelope to use) was determined by the treating physician. Statistical analyses were performed between the 3 groups (CanGaroo, Tyrx, and no envelope), and also between the No Envelope and Any Envelope groups by an independent, experienced biostatistician.
RESULTS On average, patients who received any envelope (biologic or non-biologic) were younger (70.7 ± 14.0 vs 74.9 ± 10.6, P = 0.017), had a greater number of infection risk factors (81.2% vs 49.1%, P < 0.001), received more high-powered devices (37.2% vs 5.8%, P = 0.004), and were undergoing more reoperative procedures (47.1% vs 0.0%, P < 0.001) than patients who received no envelope. Between the two envelopes, biologic envelopes tended to be used more often in higher risk patients (84.3% vs 78.4%) and reoperative procedures (62.9% vs 33.3%) than non-biologic envelopes. The rate of CIED implant site pocket infection was low (any envelope 0.5% vs no envelope 0.0%) and was statistically equivalent between the two envelope groups. Other reported adverse events (lead dislodgement, lead or pocket revision, device migration or erosion, twiddler’s syndrome, and erythema/fever) were low and statistically equivalent between groups (biologic 2.2%, non-biologic 3.9%, no envelope 1.8%).
CONCLUSION CIED infection rates for biologic and non-biologic antibacterial envelopes are similar. Antibacterial envelopes may benefit patients who are higher risk for infection, however additional studies are warranted to confirm this.
Collapse
Affiliation(s)
- David A Woodard
- Department of Cardiology, Piedmont Heart Institute, Athens, GA 30606, United States
| | - Grace Kim
- Department of Medicine, Augusta University-University of Georgia Medical Partnership, Athens, GA 30606, United States
| | - Kent R Nilsson
- Department of Cardiology, Piedmont Heart Institute, Athens, GA 30606, United States
- Department of Medicine, Augusta University-University of Georgia Medical Partnership, Athens, GA 30606, United States
| |
Collapse
|
40
|
Failure Analysis of TEVG’s II: Late Failure and Entering the Regeneration Pathway. Cells 2022; 11:cells11060939. [PMID: 35326390 PMCID: PMC8946846 DOI: 10.3390/cells11060939] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2021] [Revised: 02/03/2022] [Accepted: 03/01/2022] [Indexed: 12/21/2022] Open
Abstract
Tissue-engineered vascular grafts (TEVGs) are a promising alternative to treat vascular disease under complex hemodynamic conditions. However, despite efforts from the tissue engineering and regenerative medicine fields, the interactions between the material and the biological and hemodynamic environment are still to be understood, and optimization of the rational design of vascular grafts is an open challenge. This is of special importance as TEVGs not only have to overcome the surgical requirements upon implantation, they also need to withhold the inflammatory response and sustain remodeling of the tissue. This work aims to analyze and evaluate the bio-molecular interactions and hemodynamic phenomena between blood components, cells and materials that have been reported to be related to the failure of the TEVGs during the regeneration process once the initial stages of preimplantation have been resolved, in order to tailor and refine the needed criteria for the optimal design of TEVGs.
Collapse
|
41
|
Crum RJ, Hall K, Molina CP, Hussey GS, Graham E, Li H, Badylak SF. Immunomodulatory matrix-bound nanovesicles mitigate acute and chronic pristane-induced rheumatoid arthritis. NPJ Regen Med 2022; 7:13. [PMID: 35110573 PMCID: PMC8810774 DOI: 10.1038/s41536-022-00208-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2021] [Accepted: 12/20/2021] [Indexed: 12/13/2022] Open
Abstract
Rheumatoid arthritis (RA) is an autoimmune disease characterized by chronic inflammation and destruction of synovial joints affecting ~7.5 million people worldwide. Disease pathology is driven by an imbalance in the ratio of pro-inflammatory vs. anti-inflammatory immune cells, especially macrophages. Modulation of macrophage phenotype, specifically an M1 to M2, pro- to anti-inflammatory transition, can be induced by biologic scaffold materials composed of extracellular matrix (ECM). The ECM-based immunomodulatory effect is thought to be mediated in part through recently identified matrix-bound nanovesicles (MBV) embedded within ECM. Isolated MBV was delivered via intravenous (i.v.) or peri-articular (p.a.) injection to rats with pristane-induced arthritis (PIA). The results of MBV administration were compared to intraperitoneal (i.p.) administration of methotrexate (MTX), the clinical standard of care. Relative to the diseased animals, i.p. MTX, i.v. MBV, and p.a. MBV reduced arthritis scores in both acute and chronic pristane-induced arthritis, decreased synovial inflammation, decreased adverse joint remodeling, and reduced the ratio of synovial and splenic M1 to M2 macrophages (p < 0.05). Both p.a. and i.v. MBV reduced the serum concentration of RA and PIA biomarkers CXCL10 and MCP-3 in the acute and chronic phases of disease (p < 0.05). Flow-cytometry revealed the presence of a systemic CD43hi/His48lo/CD206+, immunoregulatory monocyte population unique to p.a. and i.v. MBV treatment associated with disease resolution. The results show that the therapeutic efficacy of MBV is equal to that of MTX for the management of acute and chronic pristane-induced arthritis and, further, this effect is associated with modulation of local synovial macrophages and systemic myeloid populations.
Collapse
Affiliation(s)
- Raphael J Crum
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, 15219, USA
| | - Kelsey Hall
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, 15219, USA
| | - Catalina Pineda Molina
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, 15219, USA.,Department of Surgery, School of Medicine, University of Pittsburgh, University of Pittsburgh Medical Center Presbyterian Hospital, 200 Lothrop Street, Pittsburgh, PA, 15213, USA
| | - George S Hussey
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, 15219, USA.,Department of Surgery, School of Medicine, University of Pittsburgh, University of Pittsburgh Medical Center Presbyterian Hospital, 200 Lothrop Street, Pittsburgh, PA, 15213, USA.,ECM Therapeutics, Inc., 118 Marshall Dr., Warrendale, PA, 15086, USA
| | - Emma Graham
- Musculoskeletal Growth and Regeneration Laboratory, Department of Orthopaedic Surgery, University of Pittsburgh, 450 Technology Drive, Suite 206, Pittsburgh, PA, 15219, USA
| | - Hongshuai Li
- Department of Orthopedics and Rehabilitation, University of Iowa, 25 Grand Ave, Iowa City, IA, 52246, USA
| | - Stephen F Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Suite 300, Pittsburgh, PA, 15219, USA. .,Department of Surgery, School of Medicine, University of Pittsburgh, University of Pittsburgh Medical Center Presbyterian Hospital, 200 Lothrop Street, Pittsburgh, PA, 15213, USA. .,ECM Therapeutics, Inc., 118 Marshall Dr., Warrendale, PA, 15086, USA. .,Department of Bioengineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA, 15261, USA.
| |
Collapse
|
42
|
Antibacterial and Immunomodulatory Properties of Acellular Wharton’s Jelly Matrix. Biomedicines 2022; 10:biomedicines10020227. [PMID: 35203437 PMCID: PMC8869352 DOI: 10.3390/biomedicines10020227] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/10/2022] [Accepted: 01/17/2022] [Indexed: 12/13/2022] Open
Abstract
Of all biologic matrices, decellularized tissues have emerged as a promising tool in the field of regenerative medicine. Few empirical clinical studies have shown that Wharton’s jelly (WJ) of the human umbilical cord promotes wound closure and reduces wound-related infections. In this scope, we herein investigated whether decellularized (DC)-WJ could be used as an engineered biomaterial. In comparison with devitalized (DV)-WJ, our results showed an inherent effect of DC-WJ on Gram positive (S. aureus and S. epidermidis) and Gram negative (E. coli and P. aeruginosa) growth and adhesion. Although DC-WJ activated the neutrophils and monocytes in a comparable magnitude to DV-WJ, macrophages modulated their phenotypes and polarization states from the resting M0 phenotype to the hybrid M1/M2 phenotype in the presence of DC-WJ. M1 phenotype was predominant in the presence of DV-WJ. Finally, the subcutaneous implantation of DC-WJ showed total resorption after three weeks of implantation without any sign of foreign body reaction. These significant data shed light on the potential regenerative application of DC-WJ in providing a suitable biomaterial for tissue regenerative medicine and an ideal strategy to prevent wound-associated infections.
Collapse
|
43
|
Modulation of the Immune System Promotes Tissue Regeneration. Mol Biotechnol 2022; 64:599-610. [PMID: 35022994 DOI: 10.1007/s12033-021-00430-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Accepted: 11/22/2021] [Indexed: 10/19/2022]
Abstract
The immune system plays an essential role in the angiogenesis, repair, and regeneration of damaged tissues. Therefore, the design of scaffolds that manipulate immune cells and factors in such a way that could accelerate the repair of damaged tissues, following implantation, is one of the main goals of regenerative medicine. However, before manipulating the immune system, the function of the various components of the immune system during the repair process should be well understood and the fabrication conditions of the manipulated scaffolds should be brought closer to the physiological state of the body. In this article, we first review the studies aimed at the role of distinct immune cell populations in angiogenesis and support of damaged tissue repair. In the second part, we discuss the use of strategies that promote tissue regeneration by modulating the immune system. Given that various studies have shown an increase in tissue repair rate with the addition of stem cells and growth factors to the scaffolds, and regarding the limited resources of stem cells, we suggest the design of scaffolds that are capable to develop repair of damaged tissue by manipulating the immune system and create an alternative for repair strategies that use stem cells or growth factors.
Collapse
|
44
|
de Sá Schiavo Matias G, da Silva Nunes Barreto R, Carreira ACO, Junior MYN, Fratini P, Ferreira CR, Miglino MA. Proteomic profile of extracellular matrix from native and decellularized chorionic canine placenta. J Proteomics 2022; 256:104497. [DOI: 10.1016/j.jprot.2022.104497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 01/06/2022] [Accepted: 01/21/2022] [Indexed: 10/19/2022]
|
45
|
Edwards JH, Jones GL, Herbert A, Fisher J, Ingham E. Integration and functional performance of a decellularised porcine superflexor tendon graft in an ovine model of anterior cruciate ligament reconstruction. Biomaterials 2021; 279:121204. [PMID: 34736146 PMCID: PMC8683753 DOI: 10.1016/j.biomaterials.2021.121204] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2020] [Revised: 10/10/2021] [Accepted: 10/20/2021] [Indexed: 12/30/2022]
Abstract
The objective was to evaluate the performance of decellularised porcine superflexor tendon (pSFT) as an anterior cruciate ligament (ACL) reconstruction device. The ACL of adult sheep was reconstructed with decellularised pSFT or ovine allograft SFT and animals sacrificed at 4, 12 and 26 weeks (n = 4 per group) for biological evaluation and 26 weeks (n = 6) for biomechanical evaluation of the grafts. Both grafts showed good in vivo performance with no major differences at macroscopic evaluation post euthanasia. Histopathology revealed an inflammatory reaction to both grafts at 4 weeks, which reduced by 26 weeks. There was advanced cellular ingrowth from 12 weeks, ligamentisation of intra-articular grafts, ossification and formation of Sharpey's fibers at the graft/bone junctions. Immunohistochemistry showed that at 4 and 12 weeks, the host response was dominated by CD163+ M2 macrophages and a cell infiltrate comprising α-SMA + myofibroblasts, CD34+ and CD271+ progenitor cells. At 26 weeks the biomechanical properties of decellularised pSFT and oSFT grafts were comparable, with all grafts failing in the intra-articular region. This study provides new insight into constructive remodelling of tendons used for ACL replacement and evidence of integration and functional performance of a decellularised xenogeneic tendon with potential as an alternative for ACL reconstruction.
Collapse
Affiliation(s)
- Jennifer Helen Edwards
- Institute of Medical and Biological Engineering, School of Biomedical Sciences, University of Leeds, Leeds, LS2 9JT, UK; Institute of Medical and Biological Engineering, School of Mechanical Engineering, University of Leeds, Leeds, LS2 9JT, UK.
| | - Gemma Louise Jones
- Institute of Medical and Biological Engineering, School of Biomedical Sciences, University of Leeds, Leeds, LS2 9JT, UK.
| | - Anthony Herbert
- Institute of Medical and Biological Engineering, School of Mechanical Engineering, University of Leeds, Leeds, LS2 9JT, UK.
| | - John Fisher
- Institute of Medical and Biological Engineering, School of Mechanical Engineering, University of Leeds, Leeds, LS2 9JT, UK.
| | - Eileen Ingham
- Institute of Medical and Biological Engineering, School of Biomedical Sciences, University of Leeds, Leeds, LS2 9JT, UK.
| |
Collapse
|
46
|
Wang X, Chen Y, Fan Z, Hua K. Evaluating tissue-engineered repair material for pelvic floor dysfunction: a comparison of in vivo response to meshes implanted in rats. Int Urogynecol J 2021; 33:2143-2150. [PMID: 34741620 DOI: 10.1007/s00192-021-05008-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 10/04/2021] [Indexed: 11/26/2022]
Abstract
INTRODUCTION AND HYPOTHESIS Achieving better anatomic restoration and decreasing the associated complications are necessary for material repair of pelvic floor dysfunction (PFD). This study was aimed to investigate host response to tissue-engineered repair material (TERM) in rat models by comparing different materials and study the changes in biomechanical properties over time. METHODS TERM was constructed by seeding adipose-derived stem cells (ADSCs) on electrospun poly(L-lactide)-trimethylene carbonate-glycolide (PLTG) terpolymers. The TERM, PLTG, porcine small intestine submucosa mesh (SIS), and polypropylene (PP) (n = 6 / group per time point) were implanted in rats for 7, 30, 60, and 90 days. Hematoxylin-eosin and Masson's trichrome staining were used to assess the host response, and mechanical testing was used to evaluate the changes in biomechanical properties. RESULTS In vivo imaging showed that the ADSCs were confined to the abdominal wall and did not migrate to other organs or tissues. The TERM was encapsulated by a thicker layer of connective tissue and was associated with less reduced inflammatory scores compared with PLTG and PP over time. The vascularization of the TERM was greater than that with PP and PLTG over time (p < 0.05) and was greater than that with SIS on day 90. The ultimate tensile strain and Young's modulus of the PP group showed the greatest increases, and the TERM group followed on day 90. CONCLUSIONS This TERM achieved better host integration in rat models and better biomechanical properties, and it may be an alternative material for PFD.
Collapse
Affiliation(s)
- Xiaojuan Wang
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, 128 Shenyang Road, Shanghai, 200090, People's Republic of China
| | - Yisong Chen
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, 128 Shenyang Road, Shanghai, 200090, People's Republic of China
| | - Zhongyong Fan
- Department of Materials Science, Fudan University, No. 220 Handan Road, Shanghai, 200433, People's Republic of China
| | - Keqin Hua
- Department of Gynecology, Obstetrics and Gynecology Hospital, Fudan University, 128 Shenyang Road, Shanghai, 200090, People's Republic of China.
| |
Collapse
|
47
|
Weis J, Geiger R, Kilo J, Zimpfer D. Cormatrix® for vessel reconstruction in paediatric cardiac surgery-a word of caution. Interact Cardiovasc Thorac Surg 2021; 34:597-603. [PMID: 34687545 PMCID: PMC8972283 DOI: 10.1093/icvts/ivab264] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 04/19/2021] [Accepted: 08/27/2021] [Indexed: 11/13/2022] Open
Abstract
OBJECTIVES The aim of this retrospective study was to determine if Cormatrix® (CM) represents a safe alternative to conventional patch materials used in congenital heart surgery. METHODS A total of 57 paediatric patients who underwent cardiac surgery using an Extracellular Matrix Bioscaffold (CM) were categorized into 4 groups according to the patch implant location. Patch-related complications and reintervention rates were analysed. A subgroup of 18 patients was subsequently compared to a matched group of 36 patients who underwent similar surgical procedures with autologous pericardium as patch material. RESULTS No patient died during hospitalization. There were 2 late deaths, not related to the implanted CM patch. Fourteen (66.7%) out of 21 patients with arterial patch plasty developed progressive vessel/right ventricular outflow tract stenosis or aneurysm. All 3 patients with a valved CM conduit developed haemodynamically relevant valve stenosis or regurgitation. A total of 18 (31.5%) patients needed reintervention and 12 (21.1%) related to CM. Four (7%) patients needed surgical treatment with operative removal of the stenosis. Redo valve replacement was performed on 2 (3.5%) patients. Six (10.5%) patients required an interventional cardiology procedure at a median interval of 5 months from surgery. The subgroup analysis revealed a significantly lower patch-related reintervention rate in patients treated with autologous pericardium when compared to CM (P = 0.006). CONCLUSIONS CM is safe for atrial and ventricular defect closure. The use of CM for arterial vessel reconstruction is associated with higher reintervention rates when compared to autologous pericardium.
Collapse
Affiliation(s)
- Johanna Weis
- Pediatrics III (Cardiopulmonary Unit), Department of Child and Adolescent Health, Medical University Innsbruck, Innsbruck, Austria
| | - Ralf Geiger
- Pediatrics III (Cardiopulmonary Unit), Department of Child and Adolescent Health, Medical University Innsbruck, Innsbruck, Austria
| | - Juliane Kilo
- Division of Cardiac Surgery, Department of Surgery, Medical University of Innsbruck, Innsbruck, Austria
| | - Daniel Zimpfer
- Division of Cardiac Surgery, Department of Surgery, Pediatric Heart Center Vienna, Medical University Vienna, Vienna, Austria
| |
Collapse
|
48
|
Al-Qurayshi Z, Wafa EI, Rossi Meyer MK, Owen S, Salem AK. Tissue Engineering the Pinna: Comparison and Characterization of Human Decellularized Auricular Biological Scaffolds. ACS APPLIED BIO MATERIALS 2021; 4:7234-7242. [PMID: 34568774 PMCID: PMC8456428 DOI: 10.1021/acsabm.1c00766] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2021] [Accepted: 08/17/2021] [Indexed: 12/03/2022]
Abstract
Decellularization is one of the promising techniques in tissue engineering used to create a biological scaffold for subsequent repopulation with the patient's own cells. This study aims to compare two different decellularization protocols to optimize the process of auricle decellularization by assessing and characterizing the decellularization effects on human auricular cartilage. Herein, 12 pairs (8 females, 4 males) of freshly frozen adult human cadaveric auricles were de-epithelialized and defatted leaving only the cartilaginous framework. An auricle from each pair was randomly assigned to either protocol A (latrunculin B-based decellularization) or protocol B (trypsin-based decellularization). Gross examination of the generated scaffolds demonstrated preservation of the auricles' contours and a change in color from pinkish-white to yellowish-white. Hematoxylin and eosin staining demonstrated empty cartilaginous lacunae in both study groups, which confirms the depletion of cells. However, there was greater preservation of the extracellular matrix in auricles decellularized with protocol A as compared to protocol B. Comparing protocol A to protocol B, Masson's trichrome and Safranin-O stains also demonstrated noticeable preservation of collagen and proteoglycans, respectively. Additionally, scanning electron micrographs demonstrated preservation of the cartilaginous microtopography in both study groups. Biomechanical testing demonstrated a substantial decrease in Young's modulus after decellularization using protocol B (1.3 MPa), albeit not significant (P-value > 0.05) when compared to Young's modulus prior to decellularization (2.6 MPa) or after decellularization with protocol A (2.7 MPa). A DNA quantification assay demonstrated a significant drop (P-value < 0.05) in the DNA content after decellularization with protocol A (111.0 ng/mg) and protocol B (127.6 ng/mg) in comparison to before decellularization (865.3 ng/mg). Overall, this study demonstrated effective decellularization of human auricular cartilage, and it is concluded that protocol A provided greater preservation of the extracellular matrix and biomechanical characteristics. These findings warrant proceeding with the assessment of inflammation and cell migration in a decellularized scaffold using an animal model.
Collapse
Affiliation(s)
- Zaid Al-Qurayshi
- Department
of Otolaryngology − Head & Neck Surgery, University of Iowa Hospitals and Clinics, Iowa City, Iowa 52242, United States
| | - Emad I. Wafa
- Department
of Pharmaceutical Sciences and Experimental Therapeutics, College
of Pharmacy, University of Iowa, Iowa City, Iowa 52242, United States
| | - Monica K. Rossi Meyer
- Department
of Otolaryngology − Head & Neck Surgery, University of Iowa Hospitals and Clinics, Iowa City, Iowa 52242, United States
| | - Scott Owen
- Department
of Otolaryngology − Head & Neck Surgery, University of Iowa Hospitals and Clinics, Iowa City, Iowa 52242, United States
| | - Aliasger K. Salem
- Department
of Pharmaceutical Sciences and Experimental Therapeutics, College
of Pharmacy, University of Iowa, Iowa City, Iowa 52242, United States
- Holden
Comprehensive Cancer Center, University
of Iowa, Iowa City, Iowa 52242, United States
| |
Collapse
|
49
|
Gębczak K, Wiatrak B, Fortuna W. Evaluation of PC12 Cells' Proliferation, Adhesion and Migration with the Use of an Extracellular Matrix (CorMatrix) for Application in Neural Tissue Engineering. MATERIALS (BASEL, SWITZERLAND) 2021; 14:3858. [PMID: 34300779 PMCID: PMC8307728 DOI: 10.3390/ma14143858] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/03/2021] [Revised: 07/02/2021] [Accepted: 07/06/2021] [Indexed: 01/19/2023]
Abstract
The use of extracellular matrix (ECM) biomaterials for soft tissue repair has proved extremely successful in animal models and in some clinical settings. The aim of the study was to investigate the effect of the commercially obtained CorMatrix bioscaffold on the viability, proliferation and migration of rat pheochromocytoma cell line PC12. PC12 cells were plated directly onto a CorMatrix flake or the well surface of a 12-well plate and cultured in RPMI-1640 medium and a medium supplemented with the nerve growth factor (NGF). The surface of the culture plates was modified with collagen type I (Col I). The number of PC12 cells was counted at four time points and then analysed for apoptosis using a staining kit containing annexin V conjugate with fluorescein and propidium iodide (PI). The effect of CorMatrix bioscaffold on the proliferation and migration of PC12 cells was tested by staining the cells with Hoechst 33258 solution for analysis using fluorescence microscopy. The research showed that the percentage of apoptotic and necrotic cells was low (less than 7%). CorMatrix stimulates the proliferation and possibly migration of PC12 cells that populate all levels of the three-dimensional architecture of the biomaterial. Further research on the mechanical and biochemical capabilities of CorMatrix offers prospects for the use of this material in neuro-regenerative applications.
Collapse
Affiliation(s)
- Katarzyna Gębczak
- Department of Basic Medical Sciences, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland;
| | - Benita Wiatrak
- Department of Basic Medical Sciences, Wroclaw Medical University, Borowska 211, 50-556 Wroclaw, Poland;
- Department of Pharmacology, Faculty of Medicine, Wroclaw Medical University, Mikulicza-Radeckiego 2, 50-345 Wroclaw, Poland
| | - Wojciech Fortuna
- Department of Neurosurgery, Wroclaw Medical University, Borowska 213, 50-556 Wroclaw, Poland;
| |
Collapse
|
50
|
Das P, Mishra R, Devi B, Rajesh K, Basak P, Roy M, Roy P, Lahiri D, Nandi SK. Decellularized xenogenic cartilage extracellular matrix (ECM) scaffolds for the reconstruction of osteochondral defects in rabbits. J Mater Chem B 2021; 9:4873-4894. [PMID: 34095925 DOI: 10.1039/d1tb00314c] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
The use of decellularized native allogenic or xenogenic cartilaginous extracellular matrix (ECM) biomaterials is widely expanding in the fields of tissue engineering and regenerative medicine. In this study, we aimed to develop an acellular, affordable, biodegradable, easily available goat conchal cartilaginous ECM derived scaffolding biomaterial for repair and regeneration of osteochondral defects in rabbits. Cartilages harvested from freshly collected goat ears were decellularized using chemical agents, namely, hypotonic-hypertonic (HH) buffer and Triton X-100 solution, separately. The morphologies and ultrastructure orientations of the decellularized cartilages remained unaltered in spite of complete cellular loss. Furthermore, when the acellular cartilaginous ECMs were cultured with murine mesenchymal stem cells (MSCs) (C3H10T1/2 cells), cellular infiltration and proliferation were thoroughly monitored using SEM, DAPI and FDA stained images, whereas the MTT assay proved the biocompatibility of the matrices. The increasing amounts of secreted ECM proteins (collagen and sGAG) indicated successful chondrogenic differentiation of the MSCs in the presence of the treated cartilage samples. In vivo biocompatibility studies showed no significant immune response or tissue rejection in the treated samples but tissue necrosis in control samples after 3 months. Upon implantation of the constructs in rabbits' osteochondral defects for 3 months, the histological and micro-CT evaluation revealed significant enhancement and regeneration of neocartilage and subchondral bony tissues. The IGF-1 loaded cartilaginous constructs showed comparatively better healing response after 3 months. Our results showed that decellularized xenogenic cartilaginous biomaterials preserved the bioactivity and integrity of the matrices that also favored in vitro stem cell proliferation and chondrogenic differentiation and enabled osteochondral regeneration, thus paving a new way for articular cartilage reconstruction.
Collapse
Affiliation(s)
- Piyali Das
- School of Bioscience and Engineering, Jadavpur University, Kolkata, India
| | | | | | | | | | | | | | | | | |
Collapse
|