1
|
Zhu J, Du Y, Backman LJ, Chen J, Ouyang H, Zhang W. Cellular Interactions and Biological Effects of Silk Fibroin: Implications for Tissue Engineering and Regenerative Medicine. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2409739. [PMID: 39668424 DOI: 10.1002/smll.202409739] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2024] [Revised: 11/27/2024] [Indexed: 12/14/2024]
Abstract
Silk fibroin (SF), the core structural protein derived from Bombyx mori silk, is extensively employed in tissue engineering and regenerative medicine due to its exceptional mechanical properties, favorable biocompatibility, tunable biodegradability, and versatile processing capabilities. Despite these advantages, current research predominantly focuses on SF biomaterials as structural scaffolds or drug carriers, often overlooking their potential role in modulating cellular behavior and tissue regeneration. This review aims to present a comprehensive overview of the inherent biological effects of SF biomaterials, independent of any exogenous biomolecules, and their implications for various tissue regeneration. It will cover in vitro cellular interactions of SF with various cell types, including stem cells and functional tissue cells such as osteoblasts, chondrocytes, keratinocytes, endothelial cells, fibroblasts, and epithelial cells. Moreover, it will summarize in vivo immune responses, cellular responses, and tissue regeneration following SF implantation, specifically focusing on vascular, bone, skin, cartilage, ocular, and tendon/ligament regeneration. Furthermore, it will address current limitations and future perspectives in the design of bioactive SF biomaterials. A comprehensive understanding of these cellular interactions and the biological effects of SF is crucial for predicting regenerative outcomes with precision and for designing SF-based biomaterials tailored to specific properties, enabling broader applications in regenerative medicine.
Collapse
Affiliation(s)
- Jialin Zhu
- School of Medicine, Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, 210000, China
| | - Yan Du
- School of Medicine, Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, 210000, China
| | - Ludvig J Backman
- Department of Medical and Translational Biology, Anatomy, Umeå University, Umeå, 90187, Sweden
- Department of Community Medicine and Rehabilitation, Section of Physiotherapy, Umeå University, Umeå, 90187, Sweden
| | - Jialin Chen
- School of Medicine, Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, 210000, China
- Department of Ophthalmology, Zhongda Hospital, Southeast University, Nanjing, 210009, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, 310000, China
| | - Hongwei Ouyang
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, 310000, China
- Department of Sports Medicine of the Second Affiliated Hospital, and Liangzhu Laboratory, Zhejiang University School of Medicine, Hangzhou, 310000, China
| | - Wei Zhang
- School of Medicine, Jiangsu Key Laboratory for Biomaterials and Devices, Southeast University, Nanjing, 210000, China
- China Orthopedic Regenerative Medicine Group (CORMed), Hangzhou, 310000, China
| |
Collapse
|
2
|
Chaber P, Andrä-Żmuda S, Śmigiel-Gac N, Zięba M, Dawid K, Martinka Maksymiak M, Adamus G. Enhancing the Potential of PHAs in Tissue Engineering Applications: A Review of Chemical Modification Methods. MATERIALS (BASEL, SWITZERLAND) 2024; 17:5829. [PMID: 39685265 DOI: 10.3390/ma17235829] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2024] [Revised: 11/22/2024] [Accepted: 11/25/2024] [Indexed: 12/18/2024]
Abstract
Polyhydroxyalkanoates (PHAs) are a family of polyesters produced by many microbial species. These naturally occurring polymers are widely used in tissue engineering because of their in vivo degradability and excellent biocompatibility. The best studied among them is poly(3-hydroxybutyrate) (PHB) and its copolymer with 3-hydroxyvaleric acid (PHBV). Despite their superior properties, PHB and PHBV suffer from high crystallinity, poor mechanical properties, a slow resorption rate, and inherent hydrophobicity. Not only are PHB and PHBV hydrophobic, but almost all members of the PHA family struggle because of this characteristic. One can overcome the limitations of microbial polyesters by modifying their bulk or surface chemical composition. Therefore, researchers have put much effort into developing methods for the chemical modification of PHAs. This paper explores a rarely addressed topic in review articles-chemical methods for modifying the structure of PHB and PHBV to enhance their suitability as biomaterials for tissue engineering applications. Different chemical strategies for improving the wettability and mechanical properties of PHA scaffolds are discussed in this review. The properties of PHAs that are important for their applications in tissue engineering are also discussed.
Collapse
Affiliation(s)
- Paweł Chaber
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, ul. M. Curie-Skłodowska 34, 41-819 Zabrze, Poland
| | - Silke Andrä-Żmuda
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, ul. M. Curie-Skłodowska 34, 41-819 Zabrze, Poland
| | - Natalia Śmigiel-Gac
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, ul. M. Curie-Skłodowska 34, 41-819 Zabrze, Poland
| | - Magdalena Zięba
- Department of Optoelectronics, Silesian University of Technology, ul. B. Krzywoustego 2, 44-100 Gliwice, Poland
| | - Kamil Dawid
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, ul. M. Curie-Skłodowska 34, 41-819 Zabrze, Poland
| | - Magdalena Martinka Maksymiak
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, ul. M. Curie-Skłodowska 34, 41-819 Zabrze, Poland
| | - Grażyna Adamus
- Centre of Polymer and Carbon Materials, Polish Academy of Sciences, ul. M. Curie-Skłodowska 34, 41-819 Zabrze, Poland
| |
Collapse
|
3
|
Ben Abdeladhim R, Reis JA, Vieira AM, de Almeida CD. Polyhydroxyalkanoates: Medical Applications and Potential for Use in Dentistry. MATERIALS (BASEL, SWITZERLAND) 2024; 17:5415. [PMID: 39597239 PMCID: PMC11595952 DOI: 10.3390/ma17225415] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/29/2024] [Accepted: 10/31/2024] [Indexed: 11/29/2024]
Abstract
Polyhydroxyalkanoates (PHAs) are promising biopolymers as an alternative to traditional synthetic polymers due to their biodegradability and biocompatibility. The PHA market is blooming in response to the growing demand for biodegradable and environmentally friendly plastics. These biopolyesters are produced and degraded by a variety of microorganisms, making them environmentally friendly, while offering benefits such as biocompatibility (when adequately processed) and biodegradability. Their versatility extends to various areas, from biomedicine to agriculture and composite materials, where they pave the way for significative innovations. In the field of regenerative medicine, some PHAs have key applications, namely in vascular grafts, oral tissue regeneration, and development of self-healing polymers. In addition, PHAs have the potential to be used in the creation of dental implant materials and dental medical devices. PHAs can also be used to encapsulate hydrophobic drugs, providing an approach for more targeted and effective treatments. To summarize, PHAs open new perspectives in the field of medicine by improving drug delivery and offering ecologically biocompatible solutions for medical devices. The aim of this review is to present the medical and dental applications of PHA, their advantages, disadvantages, and indications.
Collapse
Affiliation(s)
- Rim Ben Abdeladhim
- Egas Moniz School of Health & Science, Campus Universitário, Quinta da Granja, 2829-511 Caparica, Portugal; (R.B.A.); (J.A.R.); (A.M.V.)
| | - José Alexandre Reis
- Egas Moniz School of Health & Science, Campus Universitário, Quinta da Granja, 2829-511 Caparica, Portugal; (R.B.A.); (J.A.R.); (A.M.V.)
- Egas Moniz Center for Interdisciplinary Research (CiiEM), Egas Moniz School of Health & Science, Campus Universitário, Quinta da Granja, 2829-511 Caparica, Portugal
| | - Ana Maria Vieira
- Egas Moniz School of Health & Science, Campus Universitário, Quinta da Granja, 2829-511 Caparica, Portugal; (R.B.A.); (J.A.R.); (A.M.V.)
- Egas Moniz Center for Interdisciplinary Research (CiiEM), Egas Moniz School of Health & Science, Campus Universitário, Quinta da Granja, 2829-511 Caparica, Portugal
| | - Catarina Dias de Almeida
- Egas Moniz School of Health & Science, Campus Universitário, Quinta da Granja, 2829-511 Caparica, Portugal; (R.B.A.); (J.A.R.); (A.M.V.)
- Egas Moniz Center for Interdisciplinary Research (CiiEM), Egas Moniz School of Health & Science, Campus Universitário, Quinta da Granja, 2829-511 Caparica, Portugal
| |
Collapse
|
4
|
Xu Y, Saiding Q, Zhou X, Wang J, Cui W, Chen X. Electrospun fiber-based immune engineering in regenerative medicine. SMART MEDICINE 2024; 3:e20230034. [PMID: 39188511 PMCID: PMC11235953 DOI: 10.1002/smmd.20230034] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/13/2023] [Accepted: 01/26/2024] [Indexed: 08/28/2024]
Abstract
Immune engineering, a burgeoning field within regenerative medicine, involves a spectrum of strategies to optimize the intricate interplay between tissue regenerative biomaterials and the host tissue. These strategies are applied across different types of biomaterials and various disease models, which encompasses finely modulating the immune response at the levels of immune cells and factors, aiming to mitigate adverse effects like fibrosis and persistent inflammation that may arise at the injury site and consequently promote tissue regeneration. With the continuous progress in electrospinning technology, the immunoregulatory capabilities of electrospun fibers have gained substantial attention over the years. Electrospun fibers, with their extracellular matrix-like characteristics, high surface-area-to-volume ratio, and reliable pharmaceutical compound capacity, have emerged as key players among tissue engineering materials. This review specifically focuses on the role of electrospun fiber-based immune engineering, emphasizing their unique design strategies. Notably, electrospinning actively engages in immune engineering by modulating immune responses through four essential strategies: (i) surface modification, (ii) drug loading, (iii) physicochemical parameters, and (iv) biological grafting. This review presents a comprehensive overview of the intricate mechanisms of the immune system in injured tissues while unveiling the key strategies adopted by electrospun fibers to orchestrate immune regulation. Furthermore, the review explores the current developmental trends and limitations concerning the immunoregulatory function of electrospun fibers, aiming to drive the advancements in electrospun fiber-based immune engineering to its full potential.
Collapse
Affiliation(s)
- Yiru Xu
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Embryo Original DiseasesShanghaiChina
| | - Qimanguli Saiding
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xue Zhou
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Embryo Original DiseasesShanghaiChina
| | - Juan Wang
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Wenguo Cui
- Department of OrthopaedicsShanghai Key Laboratory for Prevention and Treatment of Bone and Joint DiseasesShanghai Institute of Traumatology and OrthopaedicsRuijin HospitalShanghai Jiao Tong University School of MedicineShanghaiChina
| | - Xinliang Chen
- The International Peace Maternity and Child Health HospitalSchool of MedicineShanghai Jiao Tong UniversityShanghaiChina
- Shanghai Key Laboratory of Embryo Original DiseasesShanghaiChina
| |
Collapse
|
5
|
Aazmi A, Zhang D, Mazzaglia C, Yu M, Wang Z, Yang H, Huang YYS, Ma L. Biofabrication methods for reconstructing extracellular matrix mimetics. Bioact Mater 2024; 31:475-496. [PMID: 37719085 PMCID: PMC10500422 DOI: 10.1016/j.bioactmat.2023.08.018] [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: 05/09/2023] [Revised: 08/23/2023] [Accepted: 08/24/2023] [Indexed: 09/19/2023] Open
Abstract
In the human body, almost all cells interact with extracellular matrices (ECMs), which have tissue and organ-specific compositions and architectures. These ECMs not only function as cellular scaffolds, providing structural support, but also play a crucial role in dynamically regulating various cellular functions. This comprehensive review delves into the examination of biofabrication strategies used to develop bioactive materials that accurately mimic one or more biophysical and biochemical properties of ECMs. We discuss the potential integration of these ECM-mimics into a range of physiological and pathological in vitro models, enhancing our understanding of cellular behavior and tissue organization. Lastly, we propose future research directions for ECM-mimics in the context of tissue engineering and organ-on-a-chip applications, offering potential advancements in therapeutic approaches and improved patient outcomes.
Collapse
Affiliation(s)
- Abdellah Aazmi
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Duo Zhang
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
- School of Medicine, The Chinese University of Hong Kong, Shenzhen, Guangdong, 51817, China
| | - Corrado Mazzaglia
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - Mengfei Yu
- The Affiliated Stomatologic Hospital, School of Medicine, Zhejiang University, Hangzhou, 310003, China
| | - Zhen Wang
- Center for Laboratory Medicine, Allergy Center, Department of Transfusion Medicine, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, Hangzhou, Zhejiang, 310014, China
| | - Huayong Yang
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Yan Yan Shery Huang
- Department of Engineering, University of Cambridge, Cambridge, United Kingdom
| | - Liang Ma
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China
- School of Mechanical Engineering, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
6
|
Okhovatian S, Shakeri A, Huyer LD, Radisic M. Elastomeric Polyesters in Cardiovascular Tissue Engineering and Organs-on-a-Chip. Biomacromolecules 2023; 24:4511-4531. [PMID: 37639715 PMCID: PMC10915885 DOI: 10.1021/acs.biomac.3c00387] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/31/2023]
Abstract
Cardiovascular tissue constructs provide unique design requirements due to their functional responses to substrate mechanical properties and cyclic stretching behavior of cardiac tissue that requires the use of durable elastic materials. Given the diversity of polyester synthesis approaches, an opportunity exists to develop a new class of biocompatible, elastic, and immunomodulatory cardiovascular polymers. Furthermore, elastomeric polyester materials have the capability to provide tailored biomechanical synergy with native tissue and hence reduce inflammatory response in vivo and better support tissue maturation in vitro. In this review, we highlight underlying chemistry and design strategies of polyester elastomers optimized for cardiac tissue scaffolds. The major advantages of these materials such as their tunable elasticity, desirable biodegradation, and potential for incorporation of bioactive compounds are further expanded. Unique fabrication methods using polyester materials such as micromolding, 3D stamping, electrospinning, laser ablation, and 3D printing are discussed. Moreover, applications of these biomaterials in cardiovascular organ-on-a-chip devices and patches are analyzed. Finally, we outline unaddressed challenges in the field that need further study to enable the impactful translation of soft polyesters to clinical applications.
Collapse
Affiliation(s)
- Sargol Okhovatian
- Institute of Biomaterials Engineering; University of Toronto; Toronto; Ontario, M5S 3G9; Canada
- Toronto General Research Institute, Toronto; Ontario, M5G 2C4; Canada
| | - Amid Shakeri
- Institute of Biomaterials Engineering; University of Toronto; Toronto; Ontario, M5S 3G9; Canada
- Toronto General Research Institute, Toronto; Ontario, M5G 2C4; Canada
| | - Locke Davenport Huyer
- Department of Applied Oral Sciences, Faculty of Dentistry, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
- School of Biomedical Engineering, Faculties of Medicine and Engineering, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
- Department of Microbiology & Immunology, Faculty of Medicine, Dalhousie University, Halifax, Nova Scotia B3H 4R2, Canada
| | - Milica Radisic
- Institute of Biomaterials Engineering; University of Toronto; Toronto; Ontario, M5S 3G9; Canada
- Toronto General Research Institute, Toronto; Ontario, M5G 2C4; Canada
- Department of Chemical Engineering and Applied Chemistry; University of Toronto; Toronto; Ontario, M5S 3E5; Canada
| |
Collapse
|
7
|
Ren ZW, Wang ZY, Ding YW, Dao JW, Li HR, Ma X, Yang XY, Zhou ZQ, Liu JX, Mi CH, Gao ZC, Pei H, Wei DX. Polyhydroxyalkanoates: the natural biopolyester for future medical innovations. Biomater Sci 2023; 11:6013-6034. [PMID: 37522312 DOI: 10.1039/d3bm01043k] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/01/2023]
Abstract
Polyhydroxyalkanoates (PHAs) are a family of natural microbial biopolyesters with the same basic chemical structure and diverse side chain groups. Based on their excellent biodegradability, biocompatibility, thermoplastic properties and diversity, PHAs are highly promising medical biomaterials and elements of medical devices for applications in tissue engineering and drug delivery. However, due to the high cost of biotechnological production, most PHAs have yet to be applied in the clinic and have only been studied at laboratory scale. This review focuses on the biosynthesis, diversity, physical properties, biodegradability and biosafety of PHAs. We also discuss optimization strategies for improved microbial production of commercial PHAs via novel synthetic biology tools. Moreover, we also systematically summarize various medical devices based on PHAs and related design approaches for medical applications, including tissue repair and drug delivery. The main degradation product of PHAs, 3-hydroxybutyrate (3HB), is recognized as a new functional molecule for cancer therapy and immune regulation. Although PHAs still account for only a small percentage of medical polymers, up-and-coming novel medical PHA devices will enter the clinical translation stage in the next few years.
Collapse
Affiliation(s)
- Zi-Wei Ren
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China.
| | - Ze-Yu Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China.
| | - Yan-Wen Ding
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China.
| | - Jin-Wei Dao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China.
- Dehong Biomedical Engineering Research Center, Dehong Teachers' College, Dehong, 678400, China
| | - Hao-Ru Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China.
| | - Xue Ma
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China.
| | - Xin-Yu Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China.
| | - Zi-Qi Zhou
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China.
| | - Jia-Xuan Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China.
| | - Chen-Hui Mi
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China.
| | - Zhe-Chen Gao
- Department of Orthopaedics, The Second Affiliated Hospital of Anhui Medical University, Hefei, 230601, China
| | - Hua Pei
- Department of Clinical Laboratory, The Second Affiliated Hospital, Hainan Medical University, Haikou, 570311, China.
| | - Dai-Xu Wei
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine, Northwest University, Xi'an, 710069, China.
- Department of Clinical Laboratory, The Second Affiliated Hospital, Hainan Medical University, Haikou, 570311, China.
- Shaanxi Key Laboratory for Carbon Neutral Technology, Xi'an, 710069, China
- Zigong Affiliated Hospital of Southwest Medical University, Zigong Psychiatric Research Center, Zigong Institute of Brain Science, Zigong, 643002, Sichuan, China
| |
Collapse
|
8
|
Perez-Estenaga I, Chevalier MT, Peña E, Abizanda G, Alsharabasy AM, Larequi E, Cilla M, Perez MM, Gurtubay J, Garcia-Yebenes Castro M, Prosper F, Pandit A, Pelacho B. A Multimodal Scaffold for SDF1 Delivery Improves Cardiac Function in a Rat Subacute Myocardial Infarct Model. ACS APPLIED MATERIALS & INTERFACES 2023; 15:50638-50651. [PMID: 37566441 PMCID: PMC10636708 DOI: 10.1021/acsami.3c04245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Accepted: 08/02/2023] [Indexed: 08/12/2023]
Abstract
Ischemic heart disease is one of the leading causes of death worldwide. The efficient delivery of therapeutic growth factors could counteract the adverse prognosis of post-myocardial infarction (post-MI). In this study, a collagen hydrogel that is able to load and appropriately deliver pro-angiogenic stromal cell-derived factor 1 (SDF1) was physically coupled with a compact collagen membrane in order to provide the suture strength required for surgical implantation. This bilayer collagen-on-collagen scaffold (bCS) showed the suitable physicochemical properties that are needed for efficient implantation, and the scaffold was able to deliver therapeutic growth factors after MI. In vitro collagen matrix biodegradation led to a sustained SDF1 release and a lack of cytotoxicity in the relevant cell cultures. In vivo intervention in a rat subacute MI model resulted in the full integration of the scaffold into the heart after implantation and biocompatibility with the tissue, with a prevalence of anti-inflammatory and pro-angiogenic macrophages, as well as evidence of revascularization and improved cardiac function after 60 days. Moreover, the beneficial effect of the released SDF1 on heart remodeling was confirmed by a significant reduction in cardiac tissue stiffness. Our findings demonstrate that this multimodal scaffold is a desirable matrix that can be used as a drug delivery system and a scaffolding material to promote functional recovery after MI.
Collapse
Affiliation(s)
- Iñigo Perez-Estenaga
- Regenerative
Medicine Department, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona 31008, Spain
| | - Merari Tumin Chevalier
- CÚRAM,
SFI Research Center for Medical Devices, University of Galway, Galway H91 TK33, Ireland
| | - Estefania Peña
- Aragon
Institute of Engineering Research, University
of Zaragoza, Zaragoza 50009, Spain
- CIBER-BBN—Centro
de Investigación Biomédica en Red en Bioingeniería
Biomateriales y Nanomedicina, Zaragoza 50018, Spain
| | - Gloria Abizanda
- Regenerative
Medicine Department, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona 31008, Spain
- Instituto
de Investigación Sanitaria de Navarra (IdiSNA), Pamplona 31009, Spain
| | - Amir M. Alsharabasy
- CÚRAM,
SFI Research Center for Medical Devices, University of Galway, Galway H91 TK33, Ireland
| | - Eduardo Larequi
- Regenerative
Medicine Department, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona 31008, Spain
| | - Myriam Cilla
- Aragon
Institute of Engineering Research, University
of Zaragoza, Zaragoza 50009, Spain
- CIBER-BBN—Centro
de Investigación Biomédica en Red en Bioingeniería
Biomateriales y Nanomedicina, Zaragoza 50018, Spain
| | - Marta M. Perez
- Department
of Anatomy, Embryology and Animal Genetics, University of Zaragoza, Zaragoza 50009, Spain
| | - Jon Gurtubay
- Regenerative
Medicine Department, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona 31008, Spain
| | | | - Felipe Prosper
- Regenerative
Medicine Department, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona 31008, Spain
- Instituto
de Investigación Sanitaria de Navarra (IdiSNA), Pamplona 31009, Spain
- Department
of Cell Therapy and Hematology, Clínica
Universidad de Navarra, Pamplona 31008, Spain
- CIBERONC, Madrid 28029, Spain
| | - Abhay Pandit
- CÚRAM,
SFI Research Center for Medical Devices, University of Galway, Galway H91 TK33, Ireland
| | - Beatriz Pelacho
- Regenerative
Medicine Department, Center for Applied Medical Research (CIMA), University of Navarra, Pamplona 31008, Spain
- Instituto
de Investigación Sanitaria de Navarra (IdiSNA), Pamplona 31009, Spain
| |
Collapse
|
9
|
Kalia VC, Patel SKS, Lee JK. Exploiting Polyhydroxyalkanoates for Biomedical Applications. Polymers (Basel) 2023; 15:polym15081937. [PMID: 37112084 PMCID: PMC10144186 DOI: 10.3390/polym15081937] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 04/15/2023] [Accepted: 04/18/2023] [Indexed: 04/29/2023] Open
Abstract
Polyhydroxyalkanoates (PHA) are biodegradable plastic. Numerous bacteria produce PHAs under environmental stress conditions, such as excess carbon-rich organic matter and limitations of other nutritional elements such as potassium, magnesium, oxygen, phosphorus, and nitrogen. In addition to having physicochemical properties similar to fossil-fuel-based plastics, PHAs have unique features that make them ideal for medical devices, such as easy sterilization without damaging the material itself and easy dissolution following use. PHAs can replace traditional plastic materials used in the biomedical sector. PHAs can be used in a variety of biomedical applications, including medical devices, implants, drug delivery devices, wound dressings, artificial ligaments and tendons, and bone grafts. Unlike plastics, PHAs are not manufactured from petroleum products or fossil fuels and are, therefore, environment-friendly. In this review, a recent overview of applications of PHAs with special emphasis on biomedical sectors, including drug delivery, wound healing, tissue engineering, and biocontrols, are discussed.
Collapse
Affiliation(s)
- Vipin Chandra Kalia
- Department of Chemical Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Sanjay K S Patel
- Department of Chemical Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Jung-Kul Lee
- Department of Chemical Engineering, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul 05029, Republic of Korea
| |
Collapse
|
10
|
Novel Production Methods of Polyhydroxyalkanoates and Their Innovative Uses in Biomedicine and Industry. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27238351. [PMID: 36500442 PMCID: PMC9740486 DOI: 10.3390/molecules27238351] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/25/2022] [Accepted: 11/27/2022] [Indexed: 12/03/2022]
Abstract
Polyhydroxyalkanoate (PHA), a biodegradable polymer obtained from microorganisms and plants, have been widely used in biomedical applications and devices, such as sutures, cardiac valves, bone scaffold, and drug delivery of compounds with pharmaceutical interests, as well as in food packaging. This review focuses on the use of polyhydroxyalkanoates beyond the most common uses, aiming to inform about the potential uses of the biopolymer as a biosensor, cosmetics, drug delivery, flame retardancy, and electrospinning, among other interesting uses. The novel applications are based on the production and composition of the polymer, which can be modified by genetic engineering, a semi-synthetic approach, by changing feeding carbon sources and/or supplement addition, among others. The future of PHA is promising, and despite its production costs being higher than petroleum-based plastics, tools given by synthetic biology, bioinformatics, and machine learning, among others, have allowed for great production yields, monomer and polymer functionalization, stability, and versatility, a key feature to increase the uses of this interesting family of polymers.
Collapse
|
11
|
Chen L, Yao Z, Zhang S, Tang K, Yang Q, Wang Y, Li B, Nie Y, Tian X, Sun L. Biomaterial-induced macrophage polarization for bone regeneration. CHINESE CHEM LETT 2022. [DOI: 10.1016/j.cclet.2022.107925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
12
|
Alamdari SG, Alibakhshi A, de la Guardia M, Baradaran B, Mohammadzadeh R, Amini M, Kesharwani P, Mokhtarzadeh A, Oroojalian F, Sahebkar A. Conductive and Semiconductive Nanocomposite-Based Hydrogels for Cardiac Tissue Engineering. Adv Healthc Mater 2022; 11:e2200526. [PMID: 35822350 DOI: 10.1002/adhm.202200526] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2022] [Revised: 05/26/2022] [Indexed: 01/27/2023]
Abstract
Cardiovascular disease is the leading cause of death worldwide and the most common cause is myocardial infarction. Therefore, appropriate approaches should be used to repair damaged heart tissue. Recently, cardiac tissue engineering approaches have been extensively studied. Since the creation of the nature of cardiovascular tissue engineering, many advances have been made in cellular and scaffolding technologies. Due to the hydrated and porous structures of the hydrogel, they are used as a support matrix to deliver cells to the infarct tissue. In heart tissue regeneration, bioactive and biodegradable hydrogels are required by simulating native tissue microenvironments to support myocardial wall stress in addition to preserving cells. Recently, the use of nanostructured hydrogels has increased the use of nanocomposite hydrogels and has revolutionized the field of cardiac tissue engineering. Therefore, to overcome the limitation of the use of hydrogels due to their mechanical fragility, various nanoparticles of polymers, metal, and carbon are used in tissue engineering and create a new opportunity to provide hydrogels with excellent properties. Here, the types of synthetic and natural polymer hydrogels, nanocarbon-based hydrogels, and other nanoparticle-based materials used for cardiac tissue engineering with emphasis on conductive nanostructured hydrogels are briefly introduced.
Collapse
Affiliation(s)
- Sania Ghobadi Alamdari
- Department of Cell and Molecular Biology, Faculty of Basic Science, University of Maragheh, Maragheh, 83111-55181, Iran.,Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, 5165665931, Iran
| | - Abbas Alibakhshi
- Molecular Medicine Research Center, Hamadan University of Medical Sciences, Hamadan, 6517838736, Iran
| | - Miguel de la Guardia
- Department of Analytical Chemistry, University of Valencia, Dr. Moliner 50, Burjassot, Valencia, 46100, Spain
| | - Behzad Baradaran
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, 5165665931, Iran
| | - Reza Mohammadzadeh
- Department of Cell and Molecular Biology, Faculty of Basic Science, University of Maragheh, Maragheh, 83111-55181, Iran
| | - Mohammad Amini
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, 5165665931, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, 110062, India
| | - Ahad Mokhtarzadeh
- Immunology Research Center, Tabriz University of Medical Sciences, Tabriz, 5165665931, Iran
| | - Fatemeh Oroojalian
- Department of Advanced Technologies, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, 94149-75516, Iran.,Natural Products and Medicinal Plants Research Center, North Khorasan University of Medical Sciences, Bojnurd, 94149-75516, Iran
| | - Amirhossein Sahebkar
- Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, 9177899191, Iran.,Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, 9177899191, Iran.,Department of Biotechnology, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, 9177899191, Iran
| |
Collapse
|
13
|
Chernozem RV, Pariy IO, Pryadko A, Bonartsev AP, Voinova VV, Zhuikov VA, Makhina TK, Bonartseva GA, Shaitan KV, Shvartsman VV, Lupascu DC, Romanyuk KN, Kholkin AL, Surmenev RA, Surmeneva MA. A comprehensive study of the structure and piezoelectric response of biodegradable polyhydroxybutyrate-based films for tissue engineering applications. Polym J 2022. [DOI: 10.1038/s41428-022-00662-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
14
|
Gomes MR, Castelo Ferreira F, Sanjuan-Alberte P. Electrospun piezoelectric scaffolds for cardiac tissue engineering. BIOMATERIALS ADVANCES 2022; 137:212808. [PMID: 35929248 DOI: 10.1016/j.bioadv.2022.212808] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Revised: 03/29/2022] [Accepted: 04/13/2022] [Indexed: 06/15/2023]
Abstract
The use of smart materials in tissue engineering is becoming increasingly appealing to provide additional functionalities and control over cell fate. The stages of tissue development and regeneration often require various electrical and electromechanical cues supported by the extracellular matrix, which is often neglected in most tissue engineering approaches. Particularly, in cardiac cells, electrical signals modulate cell activity and are responsible for the maintenance of the excitation-contraction coupling. Addition of electroconductive and topographical cues improves the biomimicry of cardiac tissues and plays an important role in driving cells towards the desired phenotype. Current platforms used to apply electrical stimulation to cells in vitro often require large external equipment and wires and electrodes immersed in the culture media, limiting the scalability and applicability of this process. Piezoelectric materials represent a shift in paradigm in materials and methods aimed at providing electrical stimulation to cardiac cells since they can produce and deliver electrical signals to cells and tissues by mechanoelectrical transduction. Despite the ability of piezoelectric materials to mimic the mechanoelectrical transduction of the heart, the use of these materials is limited in cardiac tissue engineering and methods to characterise piezoelectricity are often built in-house, which poses an additional difficulty when comparing results from the literature. In this work, we aim at providing an overview of the main challenges in cardiac tissue engineering and how piezoelectric materials could offer a solution to them. A revision on the existing literature in electrospun piezoelectric materials applied to cardiac tissue engineering is performed for the first time, as electrospinning plays an important role in the manufacturing of scaffolds with enhanced piezoelectricity and extracellular matrix native-like morphology. Finally, an overview of the current techniques used to evaluate piezoelectricity and their limitations is provided.
Collapse
Affiliation(s)
- Mariana Ramalho Gomes
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Frederico Castelo Ferreira
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Paola Sanjuan-Alberte
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; Associate Laboratory i4HB, Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal.
| |
Collapse
|
15
|
Pulingam T, Appaturi JN, Parumasivam T, Ahmad A, Sudesh K. Biomedical Applications of Polyhydroxyalkanoate in Tissue Engineering. Polymers (Basel) 2022; 14:2141. [PMID: 35683815 PMCID: PMC9182786 DOI: 10.3390/polym14112141] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2022] [Revised: 05/19/2022] [Accepted: 05/23/2022] [Indexed: 02/06/2023] Open
Abstract
Tissue engineering technology aids in the regeneration of new tissue to replace damaged or wounded tissue. Three-dimensional biodegradable and porous scaffolds are often utilized in this area to mimic the structure and function of the extracellular matrix. Scaffold material and design are significant areas of biomaterial research and the most favorable material for seeding of in vitro and in vivo cells. Polyhydroxyalkanoates (PHAs) are biopolyesters (thermoplastic) that are appropriate for this application due to their biodegradability, thermo-processability, enhanced biocompatibility, mechanical properties, non-toxicity, and environmental origin. Additionally, they offer enormous potential for modification through biological, chemical and physical alteration, including blending with various other materials. PHAs are produced by bacterial fermentation under nutrient-limiting circumstances and have been reported to offer new perspectives for devices in biological applications. The present review discusses PHAs in the applications of conventional medical devices, especially for soft tissue (sutures, wound dressings, cardiac patches and blood vessels) and hard tissue (bone and cartilage scaffolds) regeneration applications. The paper also addresses a recent advance highlighting the usage of PHAs in implantable devices, such as heart valves, stents, nerve guidance conduits and nanoparticles, including drug delivery. This review summarizes the in vivo and in vitro biodegradability of PHAs and conducts an overview of current scientific research and achievements in the development of PHAs in the biomedical sector. In the future, PHAs may replace synthetic plastics as the material of choice for medical researchers and practitioners.
Collapse
Affiliation(s)
- Thiruchelvi Pulingam
- School of Biological Sciences, Universiti Sains Malaysia, Penang 11800, Malaysia; (T.P.); (A.A.)
| | | | | | - Azura Ahmad
- School of Biological Sciences, Universiti Sains Malaysia, Penang 11800, Malaysia; (T.P.); (A.A.)
| | - Kumar Sudesh
- School of Biological Sciences, Universiti Sains Malaysia, Penang 11800, Malaysia; (T.P.); (A.A.)
| |
Collapse
|
16
|
Dhania S, Bernela M, Rani R, Parsad M, Grewal S, Kumari S, Thakur R. Scaffolds the backbone of tissue engineering: Advancements in use of polyhydroxyalkanoates (PHA). Int J Biol Macromol 2022; 208:243-259. [PMID: 35278518 DOI: 10.1016/j.ijbiomac.2022.03.030] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 03/05/2022] [Accepted: 03/07/2022] [Indexed: 12/11/2022]
Abstract
Our body is built to heal from inside out naturally but wide-ranging medical conditions necessitate the need for artificial assistance, and therefore, something that can assist the body to heal wounds and damaged tissues quickly and efficiently is of utmost importance. Tissue engineering technology helps to regenerate new tissue to replace the diseased or injured one. The technology uses biodegradable porous three-dimensional scaffolds for mimicking the structure and functions of the natural extracellular matrix. The material and design of scaffolds are critical areas of biomaterial research. Biomaterial-based three-dimensional structures have been the most promising material to serve as scaffolds for seeding cells, both in vivo and in vitro. One such material is polyhydroxyalkanoates (PHAs) which are thermoplastic biopolyesters that are highly suitable for this purpose due to their enhanced biocompatibility, biodegradability, thermo-processability, diverse mechanical properties, non-toxicity and natural origin. Moreover, they have tremendous possibilities of customization through biological physical and chemical modification as well as blending with other materials. They are being used for several tissue engineering applications such as bone graft substitute, cardiovascular patches, stents, for nerve repair and in implantology as valves and sutures. The present review overviews usage of a multitude of PHA-based biomaterials for a wide range of tissue engineering applications, based on their properties suitable for the specific applications.
Collapse
Affiliation(s)
- Sunena Dhania
- Department of Bio & Nanotechnology, Guru Jambheshwar University of Science and Technology, Hisar 125001, Haryana, India
| | - Manju Bernela
- Department of Biotechnology, Guru Nanak Dev University, Amritsar 143005, Punjab, India
| | - Ruma Rani
- ICAR-National Research Centre on Equines, Hisar 125001, Haryana, India
| | - Minakshi Parsad
- Department of Animal Biotechnology, LUVAS, Hisar 125001, Haryana, India
| | - Sapna Grewal
- Department of Bio & Nanotechnology, Guru Jambheshwar University of Science and Technology, Hisar 125001, Haryana, India
| | - Santosh Kumari
- Department of Bio & Nanotechnology, Guru Jambheshwar University of Science and Technology, Hisar 125001, Haryana, India
| | - Rajesh Thakur
- Department of Bio & Nanotechnology, Guru Jambheshwar University of Science and Technology, Hisar 125001, Haryana, India.
| |
Collapse
|
17
|
Al-Ghadban S, Artiles M, Bunnell BA. Adipose Stem Cells in Regenerative Medicine: Looking Forward. Front Bioeng Biotechnol 2022; 9:837464. [PMID: 35096804 PMCID: PMC8792599 DOI: 10.3389/fbioe.2021.837464] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 12/27/2021] [Indexed: 12/16/2022] Open
Abstract
Over the last decade, stem cell-based regenerative medicine has progressed to clinical testing and therapeutic applications. The applications range from infusions of autologous and allogeneic stem cells to stem cell-derived products. Adult stem cells from adipose tissue (ASCs) show significant promise in treating autoimmune and neurodegenerative diseases, vascular and metabolic diseases, bone and cartilage regeneration and wound defects. The regenerative capabilities of ASCs in vivo are primarily orchestrated by their secretome of paracrine factors and cell-matrix interactions. More recent developments are focused on creating more complex structures such as 3D organoids, tissue elements and eventually fully functional tissues and organs to replace or repair diseased or damaged tissues. The current and future applications for ASCs in regenerative medicine are discussed here.
Collapse
Affiliation(s)
| | | | - Bruce A. Bunnell
- Department of Microbiology Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX, United States
| |
Collapse
|
18
|
Conductive polycaprolactone/gelatin/polyaniline nanofibres as functional scaffolds for cardiac tissue regeneration. REACT FUNCT POLYM 2022. [DOI: 10.1016/j.reactfunctpolym.2021.105064] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
19
|
Guo W, Yang K, Qin X, Luo R, Wang H, Huang R. Polyhydroxyalkanoates in tissue repair and regeneration. ENGINEERED REGENERATION 2022. [DOI: 10.1016/j.engreg.2022.01.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
|
20
|
Multiscale-Engineered Muscle Constructs: PEG Hydrogel Micro-Patterning on an Electrospun PCL Mat Functionalized with Gold Nanoparticles. Int J Mol Sci 2021; 23:ijms23010260. [PMID: 35008686 PMCID: PMC8745500 DOI: 10.3390/ijms23010260] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/10/2021] [Accepted: 12/20/2021] [Indexed: 12/25/2022] Open
Abstract
The development of new, viable, and functional engineered tissue is a complex and challenging task. Skeletal muscle constructs have specific requirements as cells are sensitive to the stiffness, geometry of the materials, and biological micro-environment. The aim of this study was thus to design and characterize a multi-scale scaffold and to evaluate it regarding the differentiation process of C2C12 skeletal myoblasts. The significance of the work lies in the microfabrication of lines of polyethylene glycol, on poly(ε-caprolactone) nanofiber sheets obtained using the electrospinning process, coated or not with gold nanoparticles to act as a potential substrate for electrical stimulation. The differentiation of C2C12 cells was studied over a period of seven days and quantified through both expression of specific genes, and analysis of the myotubes’ alignment and length using confocal microscopy. We demonstrated that our multiscale bio-construct presented tunable mechanical properties and supported the different stages skeletal muscle, as well as improving the parallel orientation of the myotubes with a variation of less than 15°. These scaffolds showed the ability of sustained myogenic differentiation by enhancing the organization of reconstructed skeletal muscle. Moreover, they may be suitable for applications in mechanical and electrical stimulation to mimic the muscle’s physiological functions.
Collapse
|
21
|
Sarıkaya B, Gümüşderelioğlu M. Aligned silk fibroin/poly-3-hydroxybutyrate nanofibrous scaffolds seeded with adipose-derived stem cells for tendon tissue engineering. Int J Biol Macromol 2021; 193:276-286. [PMID: 34687764 DOI: 10.1016/j.ijbiomac.2021.10.104] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 09/28/2021] [Accepted: 10/14/2021] [Indexed: 11/18/2022]
Abstract
In this work we investigated tenogenic differentiation of adipose-derived mesenchymal stem cells (AdMSCs), which were seeded onto silk fibroin/poly-3-hydroxybutyrate (SF/P3HB) scaffolds with aligned topography, and high mechanical strength. The electrospinning process was optimized by using the response surface method (RSM) and SF/P3HB nanofibrous matrices with a total polymer concentration of 5% (SF: PHB = 3: 1), flow rate 1 mL/h, collector rotation speed 2000 rpm, applied voltage 14 kV, and collector distance 25 cm were obtained. The average fiber diameter was 699 ± 203 nm and 80% of the nanofibers were aligned within the ±15o range. SF reinforcement reduced the crystallinity of P3HB, and the elastic modulus was found to be 197.0 ± 7.7 MPa. The scaffolds showed bacteriostatic effect. A 21-day of cell culture study was performed with rat rAdMSCs in the absence and presence of tenogenic differentiation factor-5 (GDF-5). The results demonstrated that SF/P3HB scaffolds allow the cells to proliferate and differentiate to the tenocytes. However, no significant effect of GDF-5 on the differentiation of cells was observed. These findings indicated that our aligned SF/P3HB scaffolds have a significant potential to be used for tendon tissue engineering.
Collapse
Affiliation(s)
- Burcu Sarıkaya
- Hacettepe University, Graduate School of Science and Engineering, Bioengineering Division, Ankara, Turkey
| | - Menemşe Gümüşderelioğlu
- Hacettepe University, Graduate School of Science and Engineering, Bioengineering Division, Ankara, Turkey.
| |
Collapse
|
22
|
Konuk Tokak E, Çetin Altındal D, Akdere ÖE, Gümüşderelioğlu M. In-vitro effectiveness of poly-β-alanine reinforced poly(3-hydroxybutyrate) fibrous scaffolds for skeletal muscle regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 131:112528. [PMID: 34857307 DOI: 10.1016/j.msec.2021.112528] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/15/2021] [Revised: 10/16/2021] [Accepted: 10/25/2021] [Indexed: 12/13/2022]
Abstract
In skeletal muscle tissue engineering, success has not been achieved yet, since the properties of the tissue cannot be fully mimicked. The aim of this study is to investigate the potential use of poly-3-hydroxybutyrate (P3HB)/poly-β-alanine (PBA) fibrous tissue scaffolds with piezoelectric properties for skeletal muscle regeneration. Random and aligned P3HB/PBA (5:1) fibrous matrices were prepared by electrospinning with average diameters of 951 ± 153 nm and 891 ± 247 nm, respectively. X-ray diffraction (XRD) analysis showed that PBA reinforcement and aligned orientation of fibers reduced the crystallinity and brittleness of P3HB matrix. While tensile strength and elastic modulus of random fibrous matrices were determined as 3.9 ± 1.0 MPa and 86.2 ± 10.6 MPa, respectively, in the case of aligned fibers they increased to 8.5 ± 1.8 MPa and 378.2 ± 4.2 MPa, respectively. Aligned matrices exhibited a soft and an elastic behaviour with ~70% elongation in similar to the natural tissue. For the first time, d33 piezoelectric modulus of P3HB/PBA matrices were measured as 5 pC/N and 5.3 pC/N, for random and aligned matrices, respectively. Cell culture studies were performed with C2C12 myoblastic cell line. Both of random and aligned P3HB/PBA fibrous matrices supported attachment and proliferation of myoblasts, but cells cultured on aligned fibers formed regular and thick myofibril structures similar to the native muscle tissue. Reverse transcription polymerase chain reaction (RT-qPCR) analysis indicated that MyoD gene was expressed in the cells cultured on both fiber orientation, however, on the aligned fibers significant increase was determined in Myogenin and Myosin Heavy Chain (MHC) gene expressions, which indicate functional tubular structures. The results of RT-qPCR analysis were also supported with immunohistochemistry for myogenic markers. These in vitro studies have shown that piezoelectric P3HB/PBA aligned fibrous scaffolds can successfully mimic skeletal muscle tissue with its superior chemical, morphological, mechanical, and electroactive properties.
Collapse
Affiliation(s)
- Elvan Konuk Tokak
- Nanotechnology and Nanomedicine Division, Hacettepe University, Graduate School of Science and Engineering, Beytepe, Ankara, Turkey
| | - Damla Çetin Altındal
- Bioengineering Division, Hacettepe University, Graduate School of Science and Engineering, Beytepe, Ankara, Turkey
| | - Özge Ekin Akdere
- Bioengineering Division, Hacettepe University, Graduate School of Science and Engineering, Beytepe, Ankara, Turkey
| | - Menemşe Gümüşderelioğlu
- Nanotechnology and Nanomedicine Division, Hacettepe University, Graduate School of Science and Engineering, Beytepe, Ankara, Turkey; Bioengineering Division, Hacettepe University, Graduate School of Science and Engineering, Beytepe, Ankara, Turkey.
| |
Collapse
|
23
|
An YH, Kim SH. Facile Fabrication of Three-Dimensional Hydrogel Film with Complex Tissue Morphology. Bioengineering (Basel) 2021; 8:bioengineering8110164. [PMID: 34821730 PMCID: PMC8614799 DOI: 10.3390/bioengineering8110164] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2021] [Revised: 10/22/2021] [Accepted: 10/24/2021] [Indexed: 11/16/2022] Open
Abstract
In this study, we proposed a simple and easy method for fabricating a three-dimensional (3D) structure that can recapitulate the morphology of a tissue surface and deliver biological molecules into complex-shaped target tissues. To fabricate the 3D hydrogel film structure, we utilized a direct tissue casting method that can recapitulate tissue structure in micro-/macroscale using polydimethylsiloxane (PDMS). A replica 3D negative mold was manufactured by a polyurethane acrylate (PUA)-based master mold. Then, we poured the catechol-conjugated alginate (ALG-C) solution into the mold and evaporated it to form a dried film, followed by crosslinking the film using calcium chloride. The ALG-C hydrogel film had a tensile modulus of 725.2 ± 123.4 kPa and maintained over 95% of initial weight after 1 week without significant degradation. The ALG-C film captured over 4.5 times as much macromolecule (FITC-dextran) compared to alginate film (ALG). The cardiomyoblast cells exhibited high cell viability over 95% on ALG-C film. Moreover, the ALG-C film had about 70% of surface-bound lentivirus (1% in ALG film), which finally exhibited much higher viral transfection efficiency of GFP protein to C2C12 cells on the film than ALG film. In conclusion, we demonstrated a 3D film structure of biofunctionalized hydrogel for substrate-mediated drug delivery, and this approach could be utilized to recapitulate the complex-shaped tissues.
Collapse
Affiliation(s)
- Young-Hyeon An
- BioMax/N-Bio Institute, Seoul National University, Seoul 08826, Korea;
| | - Su-Hwan Kim
- Department of Chemical Engineering (BK 21 FOUR), Dong-A University, Busan 49315, Korea
- Correspondence:
| |
Collapse
|
24
|
López-Díaz de Cerio A, Perez-Estenaga I, Inoges S, Abizanda G, Gavira JJ, Larequi E, Andreu E, Rodriguez S, Gil AG, Crisostomo V, Sanchez-Margallo FM, Bermejo J, Jauregui B, Quintana L, Fernández-Avilés F, Pelacho B, Prósper F. Preclinical Evaluation of the Safety and Immunological Action of Allogeneic ADSC-Collagen Scaffolds in the Treatment of Chronic Ischemic Cardiomyopathy. Pharmaceutics 2021; 13:pharmaceutics13081269. [PMID: 34452230 PMCID: PMC8399291 DOI: 10.3390/pharmaceutics13081269] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2021] [Revised: 07/31/2021] [Accepted: 08/11/2021] [Indexed: 12/11/2022] Open
Abstract
The use of allogeneic adipose-derived mesenchymal stromal cells (alloADSCs) represents an attractive approach for treating myocardial infarction (MI). Furthermore, adding a natural support improves alloADSCs engraftment and survival in heart tissues, leading to a greater therapeutic effect. We aimed to examine the safety and immunological reaction induced by epicardial implantation of a clinical-grade collagen scaffold (CS) seeded with alloADSCs for its future application in humans. Thus, cellularized scaffolds were myocardially or subcutaneously implanted in immunosuppressed rodent models. The toxicological parameters were not significantly altered, and tumor formation was not found over the short or long term. Furthermore, biodistribution analyses in the infarcted immunocompetent rats displayed cell engraftment in the myocardium but no migration to other organs. The immunogenicity of alloADSC-CS was also evaluated in a preclinical porcine model of chronic MI; no significant humoral or cellular alloreactive responses were found. Moreover, CS cellularized with human ADSCs cocultured with human allogeneic immune cells produced no alloreactive response. Interestingly, alloADSC-CS significantly inhibited lymphocyte responses, confirming its immunomodulatory action. Thus, alloADSC-CS is likely safe and does not elicit any alloreactive immunological response in the host. Moreover, it exerts an immunomodulatory action, which supports its translation to a clinical setting.
Collapse
Affiliation(s)
- Ascensión López-Díaz de Cerio
- Department of Cell Therapy and Hematology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (A.L.-D.d.C.); (S.I.); (E.A.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (G.A.); (J.J.G.)
| | - Iñigo Perez-Estenaga
- Center for Applied Medical Research (CIMA), Regenerative Medicine Department, 31008 Pamplona, Spain; (I.P.-E.); (E.L.); (S.R.)
| | - Susana Inoges
- Department of Cell Therapy and Hematology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (A.L.-D.d.C.); (S.I.); (E.A.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (G.A.); (J.J.G.)
| | - Gloria Abizanda
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (G.A.); (J.J.G.)
- Center for Applied Medical Research (CIMA), Regenerative Medicine Department, 31008 Pamplona, Spain; (I.P.-E.); (E.L.); (S.R.)
| | - Juan José Gavira
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (G.A.); (J.J.G.)
- Department of Cardiology, Clínica Universidad de Navarra, 31008 Pamplona, Spain
| | - Eduardo Larequi
- Center for Applied Medical Research (CIMA), Regenerative Medicine Department, 31008 Pamplona, Spain; (I.P.-E.); (E.L.); (S.R.)
| | - Enrique Andreu
- Department of Cell Therapy and Hematology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (A.L.-D.d.C.); (S.I.); (E.A.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (G.A.); (J.J.G.)
| | - Saray Rodriguez
- Center for Applied Medical Research (CIMA), Regenerative Medicine Department, 31008 Pamplona, Spain; (I.P.-E.); (E.L.); (S.R.)
| | - Ana Gloria Gil
- Department of Pharmacology and Toxicology, University of Navarra, 31009 Pamplona, Spain;
| | - Verónica Crisostomo
- Jesús Usón Minimally Invasive Surgery Centre (CCMIJU), Ctra. N-521, Km. 41.8, 10071 Cáceres, Spain; (V.C.); (F.M.S.-M.)
- CIBERCV, Instituto de Salud Carlos III, 28026 Madrid, Spain; (J.B.); (F.F.-A.)
| | - Francisco Miguel Sanchez-Margallo
- Jesús Usón Minimally Invasive Surgery Centre (CCMIJU), Ctra. N-521, Km. 41.8, 10071 Cáceres, Spain; (V.C.); (F.M.S.-M.)
- CIBERCV, Instituto de Salud Carlos III, 28026 Madrid, Spain; (J.B.); (F.F.-A.)
| | - Javier Bermejo
- CIBERCV, Instituto de Salud Carlos III, 28026 Madrid, Spain; (J.B.); (F.F.-A.)
- Department of Cardiology, Hospital Gregorio Marañón and Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
- Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | | | | | - Francisco Fernández-Avilés
- CIBERCV, Instituto de Salud Carlos III, 28026 Madrid, Spain; (J.B.); (F.F.-A.)
- Department of Cardiology, Hospital Gregorio Marañón and Instituto de Investigación Sanitaria Gregorio Marañón, 28007 Madrid, Spain
- Facultad de Medicina, Universidad Complutense de Madrid, 28040 Madrid, Spain
| | - Beatriz Pelacho
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (G.A.); (J.J.G.)
- Center for Applied Medical Research (CIMA), Regenerative Medicine Department, 31008 Pamplona, Spain; (I.P.-E.); (E.L.); (S.R.)
- Correspondence: (B.P.); (F.P.); Tel.: +34-948194700 (B.P.); +34-948255400 (F.P.)
| | - Felipe Prósper
- Department of Cell Therapy and Hematology, Clínica Universidad de Navarra, 31008 Pamplona, Spain; (A.L.-D.d.C.); (S.I.); (E.A.)
- Instituto de Investigación Sanitaria de Navarra (IdiSNA), 31008 Pamplona, Spain; (G.A.); (J.J.G.)
- Center for Applied Medical Research (CIMA), Regenerative Medicine Department, 31008 Pamplona, Spain; (I.P.-E.); (E.L.); (S.R.)
- Correspondence: (B.P.); (F.P.); Tel.: +34-948194700 (B.P.); +34-948255400 (F.P.)
| |
Collapse
|
25
|
Zhao X, Niu Y, Mi C, Gong H, Yang X, Cheng J, Zhou Z, Liu J, Peng X, Wei D. Electrospinning nanofibers of microbial polyhydroxyalkanoates for applications in medical tissue engineering. JOURNAL OF POLYMER SCIENCE 2021. [DOI: 10.1002/pol.20210418] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Affiliation(s)
- Xiao‐Hong Zhao
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine Northwest University Xi'an China
| | - Yi‐Nuo Niu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine Northwest University Xi'an China
| | - Chen‐Hui Mi
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine Northwest University Xi'an China
| | - Hai‐Lun Gong
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine Northwest University Xi'an China
| | - Xin‐Yu Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine Northwest University Xi'an China
| | - Ji‐Si‐Yu Cheng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine Northwest University Xi'an China
| | - Zi‐Qi Zhou
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine Northwest University Xi'an China
| | - Jia‐Xuan Liu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine Northwest University Xi'an China
| | - Xue‐Liang Peng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine Northwest University Xi'an China
| | - Dai‐Xu Wei
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, School of Medicine, Department of Life Sciences and Medicine Northwest University Xi'an China
| |
Collapse
|
26
|
Hemalatha T, Aarthy M, Pandurangan S, Kamini NR, Ayyadurai N. A deep dive into the darning effects of biomaterials in infarct myocardium: current advances and future perspectives. Heart Fail Rev 2021; 27:1443-1467. [PMID: 34342769 DOI: 10.1007/s10741-021-10144-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 07/07/2021] [Indexed: 12/21/2022]
Abstract
Myocardial infarction (MI) occurs due to the obstruction of coronary arteries, a major crux that restricts blood flow and thereby oxygen to the distal part of the myocardium, leading to loss of cardiomyocytes and eventually, if left untreated, leads to heart failure. MI, a potent cardiovascular disorder, requires intense therapeutic interventions and thereby presents towering challenges. Despite the concerted efforts, the treatment strategies for MI are still demanding, which has paved the way for the genesis of biomaterial applications. Biomaterials exhibit immense potentials for cardiac repair and regeneration, wherein they act as extracellular matrix replacing scaffolds or as delivery vehicles for stem cells, protein, plasmids, etc. This review concentrates on natural, synthetic, and hybrid biomaterials; their function; and interaction with the body, mechanisms of repair by which they are able to improve cardiac function in a MI milieu. We also provide focus on future perspectives that need attention. The cognizance provided by the research results certainly indicates that biomaterials could revolutionize the treatment paradigms for MI with a positive impact on clinical translation.
Collapse
Affiliation(s)
- Thiagarajan Hemalatha
- Department of Biochemistry and Biotechnology, CSIR- Central Leather Research Institute, Chennai, 600020, India
| | - Mayilvahanan Aarthy
- Department of Biochemistry and Biotechnology, CSIR- Central Leather Research Institute, Chennai, 600020, India
| | - Suryalakshmi Pandurangan
- Department of Biochemistry and Biotechnology, CSIR- Central Leather Research Institute, Chennai, 600020, India
| | - Numbi Ramudu Kamini
- Department of Biochemistry and Biotechnology, CSIR- Central Leather Research Institute, Chennai, 600020, India
| | - Niraikulam Ayyadurai
- Department of Biochemistry and Biotechnology, CSIR- Central Leather Research Institute, Chennai, 600020, India.
| |
Collapse
|
27
|
Kalia VC, Singh Patel SK, Shanmugam R, Lee JK. Polyhydroxyalkanoates: Trends and advances toward biotechnological applications. BIORESOURCE TECHNOLOGY 2021; 326:124737. [PMID: 33515915 DOI: 10.1016/j.biortech.2021.124737] [Citation(s) in RCA: 92] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 01/10/2021] [Accepted: 01/13/2021] [Indexed: 06/12/2023]
Abstract
Plastics are an integral part of most of the daily requirements. Indiscriminate usage and disposal have led to the accumulation of massive quantities of waste. Their non-biodegradable nature makes it increasingly difficult to manage and dispose them. To counter this impending disaster, biodegradable polymers, especially polyhydroxy-alkanoates (PHAs), have been envisaged as potential alternatives. Owing to their unique physicochemical characteristics, PHAs are gaining importance for versatile applications in the agricultural and medical sectors. Applications in the medical sector are more promising because of their commercial viability and sustainability. Despite such potential, their production and commercialization are significant challenges. The major limitations are their poor mechanical strength, production in small quantities, costly feed, and lack of facilities for industrial production. This article provides an overview of the contemporary progress in the field, to attract researchers and stakeholders to further exploit these renewable resources to produce biodegradable plastics on a commercial scale.
Collapse
Affiliation(s)
- Vipin Chandra Kalia
- Department of Chemical Engineering, Konkuk University, Seoul 05029, Republic of Korea
| | | | - Ramasamy Shanmugam
- Department of Chemical Engineering, Konkuk University, Seoul 05029, Republic of Korea
| | - Jung-Kul Lee
- Department of Chemical Engineering, Konkuk University, Seoul 05029, Republic of Korea.
| |
Collapse
|
28
|
Song Y, Wang H, Yue F, Lv Q, Cai B, Dong N, Wang Z, Wang L. Silk-Based Biomaterials for Cardiac Tissue Engineering. Adv Healthc Mater 2020; 9:e2000735. [PMID: 32939999 DOI: 10.1002/adhm.202000735] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 08/29/2020] [Indexed: 12/18/2022]
Abstract
Cardiovascular diseases are one of the leading causes of death globally. Among various cardiovascular diseases, myocardial infarction is an important one. Compared with conventional treatments, cardiac tissue engineering provides an alternative to repair and regenerate the injured tissue. Among various types of materials used for tissue engineering applications, silk biomaterials have been increasingly utilized due to their biocompatibility, biological functions, and many favorable physical/chemical properties. Silk biomaterials are often used alone or in combination with other materials in the forms of patches or hydrogels, and serve as promising delivery systems for bioactive compounds in tissue engineering repair scenarios. This review focuses primarily on the promising characteristics of silk biomaterials and their recent advances in cardiac tissue engineering.
Collapse
Affiliation(s)
- Yu Song
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Huifang Wang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Feifei Yue
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qiying Lv
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bo Cai
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Zheng Wang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Department of Gastrointestinal Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Lin Wang
- Department of Clinical Laboratory, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
- Research Center for Tissue Engineering and Regenerative Medicine, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
29
|
Majid QA, Fricker ATR, Gregory DA, Davidenko N, Hernandez Cruz O, Jabbour RJ, Owen TJ, Basnett P, Lukasiewicz B, Stevens M, Best S, Cameron R, Sinha S, Harding SE, Roy I. Natural Biomaterials for Cardiac Tissue Engineering: A Highly Biocompatible Solution. Front Cardiovasc Med 2020; 7:554597. [PMID: 33195451 PMCID: PMC7644890 DOI: 10.3389/fcvm.2020.554597] [Citation(s) in RCA: 53] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Accepted: 09/10/2020] [Indexed: 02/06/2023] Open
Abstract
Cardiovascular diseases (CVD) constitute a major fraction of the current major global diseases and lead to about 30% of the deaths, i.e., 17.9 million deaths per year. CVD include coronary artery disease (CAD), myocardial infarction (MI), arrhythmias, heart failure, heart valve diseases, congenital heart disease, and cardiomyopathy. Cardiac Tissue Engineering (CTE) aims to address these conditions, the overall goal being the efficient regeneration of diseased cardiac tissue using an ideal combination of biomaterials and cells. Various cells have thus far been utilized in pre-clinical studies for CTE. These include adult stem cell populations (mesenchymal stem cells) and pluripotent stem cells (including autologous human induced pluripotent stem cells or allogenic human embryonic stem cells) with the latter undergoing differentiation to form functional cardiac cells. The ideal biomaterial for cardiac tissue engineering needs to have suitable material properties with the ability to support efficient attachment, growth, and differentiation of the cardiac cells, leading to the formation of functional cardiac tissue. In this review, we have focused on the use of biomaterials of natural origin for CTE. Natural biomaterials are generally known to be highly biocompatible and in addition are sustainable in nature. We have focused on those that have been widely explored in CTE and describe the original work and the current state of art. These include fibrinogen (in the context of Engineered Heart Tissue, EHT), collagen, alginate, silk, and Polyhydroxyalkanoates (PHAs). Amongst these, fibrinogen, collagen, alginate, and silk are isolated from natural sources whereas PHAs are produced via bacterial fermentation. Overall, these biomaterials have proven to be highly promising, displaying robust biocompatibility and, when combined with cells, an ability to enhance post-MI cardiac function in pre-clinical models. As such, CTE has great potential for future clinical solutions and hence can lead to a considerable reduction in mortality rates due to CVD.
Collapse
Affiliation(s)
- Qasim A. Majid
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Annabelle T. R. Fricker
- Department of Material Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
| | - David A. Gregory
- Department of Material Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
| | - Natalia Davidenko
- Department of Materials Science and Metallurgy, Cambridge Centre for Medical Materials, University of Cambridge, Cambridge, United Kingdom
| | - Olivia Hernandez Cruz
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Bioengineering, Department of Materials, IBME, Faculty of Engineering, Imperial College London, United Kingdom
| | - Richard J. Jabbour
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Thomas J. Owen
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Pooja Basnett
- Applied Biotechnology Research Group, School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, London, United Kingdom
| | - Barbara Lukasiewicz
- Applied Biotechnology Research Group, School of Life Sciences, College of Liberal Arts and Sciences, University of Westminster, London, United Kingdom
| | - Molly Stevens
- Department of Bioengineering, Department of Materials, IBME, Faculty of Engineering, Imperial College London, United Kingdom
| | - Serena Best
- Department of Materials Science and Metallurgy, Cambridge Centre for Medical Materials, University of Cambridge, Cambridge, United Kingdom
| | - Ruth Cameron
- Department of Materials Science and Metallurgy, Cambridge Centre for Medical Materials, University of Cambridge, Cambridge, United Kingdom
| | - Sanjay Sinha
- Wellcome-MRC Cambridge Stem Cell Institute, University of Cambridge, Cambridge, United Kingdom
| | - Sian E. Harding
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
| | - Ipsita Roy
- Faculty of Medicine, National Heart and Lung Institute, Imperial College London, London, United Kingdom
- Department of Material Science and Engineering, Faculty of Engineering, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
30
|
Ahuja R, Kumari N, Srivastava A, Bhati P, Vashisth P, Yadav PK, Jacob T, Narang R, Bhatnagar N. Biocompatibility analysis of PLA based candidate materials for cardiovascular stents in a rat subcutaneous implant model. Acta Histochem 2020; 122:151615. [PMID: 33066837 DOI: 10.1016/j.acthis.2020.151615] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 08/08/2020] [Accepted: 08/11/2020] [Indexed: 01/18/2023]
Abstract
Modification of Polylactic acid (PLA), a biopolymer, is a strategy still to be fully explored for the next generation of bioresorbable vascular stent (BVS) biomaterials. With this focus, inclusions upto 5% of Polycaprolactone (PCL) and Magnesium in PLA were tested in the rat subcutaneous model and their cellular and tissue interactions characterized, specifically with respect to inflammatory response, angiogenesis and capsularization. The cytokines IL6, TNF Alpha and IL-1Beta were estimated in the peri-implant tissue, all of which showed a non-significant difference between the non-implanted animals and those containing PLA by 8 weeks, speaking to the benign nature of PLA as an implant biomaterial. Both modified materials, had increased macrophage counts and cytokine levels, except IL6 at 8 weeks. Vascularization only at 8 weeks in PLA PCL containing tissue was significantly higher than pure PLA, which may be more carefully controlled along with the material hydrophobicity for possible efforts towards therapeutic angiogenesis. Capsule thickness, measured by staining with both Hematoxylin & Eosin and Masson's Trichome did not show any differences between materials, including PLA.
Collapse
Affiliation(s)
- Ramya Ahuja
- Department of Mechanical Engineering, Indian Institute of Technology, Delhi, India
| | - Nisha Kumari
- Department of Mechanical Engineering, Indian Institute of Technology, Delhi, India
| | - Alok Srivastava
- Department of Mechanical Engineering, Indian Institute of Technology, Delhi, India
| | - Pooja Bhati
- Department of Mechanical & Automation Engineering, Indira Gandhi Delhi Technical University for Women, India
| | - Priya Vashisth
- Department of Mechanical Engineering, Indian Institute of Technology, Delhi, India
| | - P K Yadav
- Central Animal Facility, All India Institute of Medical Sciences, Delhi, India
| | - Tony Jacob
- Department of Anatomy, All India Institute of Medical Sciences, Delhi, India
| | - Rajiv Narang
- Department of Cardiology, All India Institute of Medical Sciences, Delhi, India
| | - Naresh Bhatnagar
- Department of Mechanical Engineering, Indian Institute of Technology, Delhi, India.
| |
Collapse
|
31
|
Kuncorojakti S, Rodprasert W, Yodmuang S, Osathanon T, Pavasant P, Srisuwatanasagul S, Sawangmake C. Alginate/Pluronic F127-based encapsulation supports viability and functionality of human dental pulp stem cell-derived insulin-producing cells. J Biol Eng 2020; 14:23. [PMID: 32855655 PMCID: PMC7446208 DOI: 10.1186/s13036-020-00246-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2020] [Accepted: 08/13/2020] [Indexed: 11/16/2022] Open
Abstract
BACKGROUND Current approach for diabetes treatment remained several adverse events varied from gastrointestinal to life-threatening symptoms. Regenerative therapy regarding Edmonton protocol has been facing serious limitations involving protocol efficiency and safety. This led to the study for alternative insulin-producing cell (IPC) resource and transplantation platform. In this study, evaluation of encapsulated human dental pulp-derived stem cell (hDPSC)-derived IPCs by alginate (ALG) and pluronic F127-coated alginate (ALGPA) was performed. RESULTS The results showed that ALG and ALGPA preserved hDPSC viability and allowed glucose and insulin diffusion in and out. ALG and ALGPA-encapsulated hDPSC-derived IPCs maintained viability for at least 336 h and sustained pancreatic endoderm marker (NGN3), pancreatic islet markers (NKX6.1, MAF-A, ISL-1, GLUT-2 and INSULIN), and intracellular pro-insulin and insulin expressions for at least 14 days. Functional analysis revealed a glucose-responsive C-peptide secretion of ALG- and ALGPA-encapsulated hDPSC-derived IPCs at 14 days post-encapsulation. CONCLUSION ALG and ALGPA encapsulations efficiently preserved the viability and functionality of hDPSC-derived IPCs in vitro and could be the potential transplantation platform for further clinical application.
Collapse
Affiliation(s)
- Suryo Kuncorojakti
- International Graduate Course in Veterinary Science and Technology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330 Thailand
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Veterinary Pharmacology and Stem Cell Research Laboratory, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330 Thailand
| | - Watchareewan Rodprasert
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Veterinary Pharmacology and Stem Cell Research Laboratory, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330 Thailand
| | - Supansa Yodmuang
- Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, 10330 Thailand
- Excellence Center for Advanced Therapy Medicinal Products, King Chulalongkorn Memorial Hospital, Bangkok, 10330 Thailand
| | - Thanaphum Osathanon
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330 Thailand
- Center of Excellence in Regenerative Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330 Thailand
| | - Prasit Pavasant
- Department of Anatomy, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330 Thailand
- Center of Excellence in Regenerative Dentistry, Faculty of Dentistry, Chulalongkorn University, Bangkok, 10330 Thailand
| | - Sayamon Srisuwatanasagul
- Department of Veterinary Anatomy, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330 Thailand
| | - Chenphop Sawangmake
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Veterinary Pharmacology and Stem Cell Research Laboratory, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330 Thailand
- Veterinary Clinical Stem Cell and Bioengineering Research Unit, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330 Thailand
- Department of Pharmacology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, 10330 Thailand
| |
Collapse
|
32
|
Nazarnezhad S, Baino F, Kim HW, Webster TJ, Kargozar S. Electrospun Nanofibers for Improved Angiogenesis: Promises for Tissue Engineering Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2020; 10:E1609. [PMID: 32824491 PMCID: PMC7466668 DOI: 10.3390/nano10081609] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/26/2020] [Revised: 08/13/2020] [Accepted: 08/14/2020] [Indexed: 12/27/2022]
Abstract
Angiogenesis (or the development of new blood vessels) is a key event in tissue engineering and regenerative medicine; thus, a number of biomaterials have been developed and combined with stem cells and/or bioactive molecules to produce three-dimensional (3D) pro-angiogenic constructs. Among the various biomaterials, electrospun nanofibrous scaffolds offer great opportunities for pro-angiogenic approaches in tissue repair and regeneration. Nanofibers made of natural and synthetic polymers are often used to incorporate bioactive components (e.g., bioactive glasses (BGs)) and load biomolecules (e.g., vascular endothelial growth factor (VEGF)) that exert pro-angiogenic activity. Furthermore, seeding of specific types of stem cells (e.g., endothelial progenitor cells) onto nanofibrous scaffolds is considered as a valuable alternative for inducing angiogenesis. The effectiveness of these strategies has been extensively examined both in vitro and in vivo and the outcomes have shown promise in the reconstruction of hard and soft tissues (mainly bone and skin, respectively). However, the translational of electrospun scaffolds with pro-angiogenic molecules or cells is only at its beginning, requiring more research to prove their usefulness in the repair and regeneration of other highly-vascularized vital tissues and organs. This review will cover the latest progress in designing and developing pro-angiogenic electrospun nanofibers and evaluate their usefulness in a tissue engineering and regenerative medicine setting.
Collapse
Affiliation(s)
- Simin Nazarnezhad
- Tissue Engineering Research Group (TERG), Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad 917794-8564, Iran;
| | - Francesco Baino
- Institute of Materials Physics and Engineering, Applied Science and Technology Department, Politecnico di Torino, Corso Duca degli Abruzzi 24, 10129 Torino, Italy
| | - Hae-Won Kim
- Department of Biomaterials Science, School of Dentistry, Dankook University, Cheonan 31116, Korea;
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan 31116, Korea
- Department of Nanobiomedical Science & BK21 PLUS NBM Global Research Center for Regenerative Medicine Research Center, Dankook University, Cheonan 31116, Korea
| | - Thomas J. Webster
- Department of Chemical Engineering, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA;
| | - Saeid Kargozar
- Tissue Engineering Research Group (TERG), Department of Anatomy and Cell Biology, School of Medicine, Mashhad University of Medical Sciences, Mashhad 917794-8564, Iran;
| |
Collapse
|
33
|
Nanoengineering in Cardiac Regeneration: Looking Back and Going Forward. NANOMATERIALS 2020; 10:nano10081587. [PMID: 32806691 PMCID: PMC7466652 DOI: 10.3390/nano10081587] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/04/2020] [Revised: 08/07/2020] [Accepted: 08/10/2020] [Indexed: 12/19/2022]
Abstract
To deliver on the promise of cardiac regeneration, an integration process between an emerging field, nanomedicine, and a more consolidated one, tissue engineering, has begun. Our work aims at summarizing some of the most relevant prevailing cases of nanotechnological approaches applied to tissue engineering with a specific interest in cardiac regenerative medicine, as well as delineating some of the most compelling forthcoming orientations. Specifically, this review starts with a brief statement on the relevant clinical need, and then debates how nanotechnology can be combined with tissue engineering in the scope of mimicking a complex tissue like the myocardium and its natural extracellular matrix (ECM). The interaction of relevant stem, precursor, and differentiated cardiac cells with nanoengineered scaffolds is thoroughly presented. Another correspondingly relevant area of experimental study enclosing both nanotechnology and cardiac regeneration, e.g., nanoparticle applications in cardiac tissue engineering, is also discussed.
Collapse
|
34
|
Talebi A, Labbaf S, Karimzadeh F, Masaeli E, Nasr Esfahani MH. Electroconductive Graphene-Containing Polymeric Patch: A Promising Platform for Future Cardiac Repair. ACS Biomater Sci Eng 2020; 6:4214-4224. [DOI: 10.1021/acsbiomaterials.0c00266] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Affiliation(s)
- Alireza Talebi
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Sheyda Labbaf
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Fathallah Karimzadeh
- Department of Materials Engineering, Isfahan University of Technology, Isfahan 84156-83111, Iran
| | - Elahe Masaeli
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Mohammad-Hossein Nasr Esfahani
- Department of Cellular Biotechnology, Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| |
Collapse
|
35
|
Pryjmaková J, Kaimlová M, Hubáček T, Švorčík V, Siegel J. Nanostructured Materials for Artificial Tissue Replacements. Int J Mol Sci 2020; 21:E2521. [PMID: 32260477 PMCID: PMC7178059 DOI: 10.3390/ijms21072521] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Revised: 03/26/2020] [Accepted: 04/01/2020] [Indexed: 02/04/2023] Open
Abstract
This paper review current trends in applications of nanomaterials in tissue engineering. Nanomaterials applicable in this area can be divided into two groups: organic and inorganic. Organic nanomaterials are especially used for the preparation of highly porous scaffolds for cell cultivation and are represented by polymeric nanofibers. Inorganic nanomaterials are implemented as they stand or dispersed in matrices promoting their functional properties while preserving high level of biocompatibility. They are used in various forms (e.g., nano- particles, -tubes and -fibers)-and when forming the composites with organic matrices-are able to enhance many resulting properties (biologic, mechanical, electrical and/or antibacterial). For this reason, this contribution points especially to such type of composite nanomaterials. Basic information on classification, properties and application potential of single nanostructures, as well as complex scaffolds suitable for 3D tissues reconstruction is provided. Examples of practical usage of these structures are demonstrated on cartilage, bone, neural, cardiac and skin tissue regeneration and replacements. Nanomaterials open up new ways of treatments in almost all areas of current tissue regeneration, especially in tissue support or cell proliferation and growth. They significantly promote tissue rebuilding by direct replacement of damaged tissues.
Collapse
Affiliation(s)
- Jana Pryjmaková
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague, Czech Republic; (J.P.); (M.K.); (V.Š.)
| | - Markéta Kaimlová
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague, Czech Republic; (J.P.); (M.K.); (V.Š.)
| | - Tomáš Hubáček
- Soil & Water Research Infrastructure, Biology Centre CAS, Na Sádkách 7, 370 05 České Budějovice, Czech Republic;
| | - Václav Švorčík
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague, Czech Republic; (J.P.); (M.K.); (V.Š.)
| | - Jakub Siegel
- Department of Solid State Engineering, University of Chemistry and Technology Prague, Technická 5, 166 28 Prague, Czech Republic; (J.P.); (M.K.); (V.Š.)
| |
Collapse
|
36
|
Kuncorojakti S, Srisuwatanasagul S, Kradangnga K, Sawangmake C. Insulin-Producing Cell Transplantation Platform for Veterinary Practice. Front Vet Sci 2020; 7:4. [PMID: 32118053 PMCID: PMC7028771 DOI: 10.3389/fvets.2020.00004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2019] [Accepted: 01/06/2020] [Indexed: 12/14/2022] Open
Abstract
Diabetes mellitus (DM) remains a global concern in both human and veterinary medicine. Type I DM requires prolonged and consistent exogenous insulin administration to address hyperglycemia, which can increase the risk of diabetes complications such as retinopathy, nephropathy, neuropathy, and heart disorders. Cell-based therapies have been successful in human medicine using the Edmonton protocol. These therapies help maintain the production of endogenous insulin and stabilize blood glucose levels and may possibly be adapted to veterinary clinical practice. The limited number of cadaveric pancreas donors and the long-term use of immunosuppressive agents are the main obstacles for this protocol. Over the past decade, the development of potential therapies for DM has mainly focused on the generation of effective insulin-producing cells (IPCs) from various sources of stem cells that can be transplanted into the body. Another successful application of stem cells in type I DM therapies is transplanting generated IPCs. Encapsulation can be an alternative strategy to protect IPCs from rejection by the body due to their immunoisolation properties. This review summarizes current concepts of IPCs and encapsulation technology for veterinary clinical application and proposes a potential stem-cell-based platform for veterinary diabetic regenerative therapy.
Collapse
Affiliation(s)
- Suryo Kuncorojakti
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Veterinary Pharmacology and Stem Cell Research Laboratory, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand.,Department of Veterinary Anatomy, Faculty of Veterinary Medicine, Universitas Airlangga, Surabaya, Indonesia
| | - Sayamon Srisuwatanasagul
- Department of Anatomy, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Krishaporn Kradangnga
- Department of Surgery, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| | - Chenphop Sawangmake
- Veterinary Stem Cell and Bioengineering Innovation Center (VSCBIC), Veterinary Pharmacology and Stem Cell Research Laboratory, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand.,Veterinary Clinical Stem Cell and Bioengineering Research Unit, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand.,Department of Pharmacology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
37
|
Chouhan D, Mandal BB. Silk biomaterials in wound healing and skin regeneration therapeutics: From bench to bedside. Acta Biomater 2020; 103:24-51. [PMID: 31805409 DOI: 10.1016/j.actbio.2019.11.050] [Citation(s) in RCA: 167] [Impact Index Per Article: 33.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 11/25/2019] [Accepted: 11/26/2019] [Indexed: 02/08/2023]
Abstract
Silk biomaterials are known for biomedical and tissue engineering applications including drug delivery and implantable devices owing to their biocompatible and a wide range of ideal physico-chemical properties. Herein, we present a critical overview of the progress of silk-based matrices in skin regeneration therapeutics with an emphasis on recent innovations and scientific findings. Beginning with a brief description of numerous varieties of silks, the review summarizes our current understanding of the biological properties of silk that help in the wound healing process. Various silk varieties such as silkworm silk fibroin, silk sericin, native spider silk and recombinant silk materials have been explored for cutaneous wound healing applications from the past few decades. With an aim to harness the regenerative properties of silk, numerous strategies have been applied to develop functional bioactive wound dressings and viable bio-artificial skin grafts in recent times. The review examines multiple inherent properties of silk that aid in the critical events of the healing process such as cell migration, cell proliferation, angiogenesis, and re-epithelialization. A detailed insight into the progress of silk-based cellular skin grafts is also provided that discusses various co-culture strategies and development of bilayer and tri-layer human skin equivalent under in vitro conditions. In addition, functionalized silk matrices loaded with bioactive molecules and antibacterial compounds are discussed, which have shown great potential in treating hard-to-heal wounds. Finally, clinical studies performed using silk-based translational products are reviewed that validate their regenerative properties and future applications in this area. STATEMENT OF SIGNIFICANCE: The review article discusses the recent advances in silk-based technologies for wound healing applications, covering various types of silk biomaterials and their properties suitable for wound repair and regeneration. The article demonstrates the progress of silk-based matrices with an update on the patented technologies and clinical advancements over the years. The rationale behind this review is to highlight numerous properties of silk biomaterials that aid in all the critical events of the wound healing process towards skin regeneration. Functionalization strategies to fabricate silk dressings containing bioactive molecules and antimicrobial compounds for drug delivery to the wound bed are discussed. In addition, a separate section describes the approaches taken to generate living human skin equivalent that have recently contributed in the field of skin tissue engineering.
Collapse
|
38
|
McMahan S, Taylor A, Copeland KM, Pan Z, Liao J, Hong Y. Current advances in biodegradable synthetic polymer based cardiac patches. J Biomed Mater Res A 2020; 108:972-983. [PMID: 31895482 DOI: 10.1002/jbm.a.36874] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 12/19/2019] [Accepted: 12/26/2019] [Indexed: 12/21/2022]
Abstract
The number of people affected by heart disease such as coronary artery disease and myocardial infarction increases at an alarming rate each year. Currently, the methods to treat these diseases are restricted to lifestyle change, pharmaceuticals, and eventually heart transplant if the condition is severe enough. While these treatment options are the standard for caring for patients who suffer from heart disease, limited regenerative ability of the heart restricts the effectiveness of treatment and may lead to other heart-related health problems in the future. Because of the increasing need for more effective therapeutic technologies for treating diseased heart tissue, cardiac patches are now a large focus for researchers. The cardiac patches are designed to be integrated into the patients' natural tissue to introduce mechanical support and healing to the damaged areas. As a promising alternative, synthetic biodegradable polymer based biomaterials can be easily manipulated to customize material properties, as well as possess certain desired characteristics for cardiac patch use. This comprehensive review summarizes recent works on synthetic biodegradable cardiac patches implanted into infarcted animal models. In addition, this review describes the basic requirements that should be met for cardiac patch development, and discusses the inspirations to designing new biomaterials and technologies for cardiac patches.
Collapse
Affiliation(s)
- Sara McMahan
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas
| | - Alan Taylor
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas
| | - Katherine M Copeland
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas
| | - Zui Pan
- College of Nursing and Health Innovation, University of Texas at Arlington, Arlington, Texas
| | - Jun Liao
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas
| |
Collapse
|
39
|
Polycaprolactone/gelatin-based scaffolds with tailored performance: in vitro and in vivo validation. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2019; 107:110296. [PMID: 31761169 DOI: 10.1016/j.msec.2019.110296] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/29/2019] [Revised: 09/16/2019] [Accepted: 10/07/2019] [Indexed: 12/16/2022]
Abstract
Nanofibrous scaffolds composed of polycaprolactone (PCL) and gelatin (Ge) were obtained through a hydrolytic assisted electrospinning process. The PCL-to-Ge proportion (100/0 to 20/80), as well as the dissolution time (24, 48, 72, 96, 120 h) into a 1:1 formic/acetic acid solvent before electrospinning were modified to obtain the different samples. A strong influence of these factors on the physicochemical properties of the scaffolds was observed. Higher Ge percentage reduced crystallinity, allowed a uniform morphology and increased water contact angle. The increase in the dissolution time considerably reduced the molar mass and, subsequently, fibre diameter and crystallinity were affected. During in vitro biocompatibility tests, higher cell adhesion and proliferation were found for the 60/40, 50/50 and 40/60 PCL/Ge compositions that was corroborated by MTT assay, fluorescence and microscopy. A weakened structure, more labile to the in vitro degradation in physiologic conditions was found for these compositions with higher dissolution times (72 and 96 h). Particularly, the 40/60 PCL/Ge scaffolds revealed an interesting progressive degradation behaviour as a function of the dissolution time. Moreover, these scaffolds were non-inflammatory, as revealed by the pyrogen test and after the 15-day subcutaneous in vivo implantation in mice. Finally, a reduction of the scar tissue area after infarction was found for the 40/60 PCL/Ge scaffolds electrospun after 72 h implanted in rat hearts. These results are especially interesting and represent a feasible way to avoid undesired inflammatory reactions during the scaffold assimilation.
Collapse
|
40
|
Janani G, Kumar M, Chouhan D, Moses JC, Gangrade A, Bhattacharjee S, Mandal BB. Insight into Silk-Based Biomaterials: From Physicochemical Attributes to Recent Biomedical Applications. ACS APPLIED BIO MATERIALS 2019; 2:5460-5491. [DOI: 10.1021/acsabm.9b00576] [Citation(s) in RCA: 54] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
41
|
Gil-Castell O, Badia JD, Bou J, Ribes-Greus A. Performance of Polyester-Based Electrospun Scaffolds under In Vitro Hydrolytic Conditions: From Short-Term to Long-Term Applications. NANOMATERIALS (BASEL, SWITZERLAND) 2019; 9:E786. [PMID: 31121950 PMCID: PMC6566282 DOI: 10.3390/nano9050786] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/15/2019] [Accepted: 05/20/2019] [Indexed: 12/15/2022]
Abstract
The evaluation of the performance of polyesters under in vitro physiologic conditions is essential to design scaffolds with an adequate lifespan for a given application. In this line, the degradation-durability patterns of poly(lactide-co-glycolide) (PLGA), polydioxanone (PDO), polycaprolactone (PCL) and polyhydroxybutyrate (PHB) scaffolds were monitored and compared giving, as a result, a basis for the specific design of scaffolds from short-term to long-term applications. For this purpose, they were immersed in ultra-pure water and phosphate buffer solution (PBS) at 37 °C. The scaffolds for short-time applications were PLGA and PDO, in which the molar mass diminished down to 20% in a 20-30 days lifespan. While PDO developed crystallinity that prevented the geometry of the fibres, those of PLGA coalesced and collapsed. The scaffolds for long-term applications were PCL and PHB, in which the molar mass followed a progressive decrease, reaching values of 10% for PCL and almost 50% for PHB after 650 days of immersion. This resistant pattern was mainly ascribed to the stability of the crystalline domains of the fibres, in which the diameters remained almost unaffected. From the perspective of an adequate balance between the durability and degradation, this study may serve technologists as a reference point to design polyester-based scaffolds for biomedical applications.
Collapse
Affiliation(s)
- Oscar Gil-Castell
- Instituto de Tecnología de Materiales (ITM), Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain.
- Departament d'Enginyeria Química, Escola Tècnica Superior d'Enginyeria, Universitat de València, Av. de la Universitat s/n, 46100 Burjassot, Spain.
| | - José David Badia
- Instituto de Tecnología de Materiales (ITM), Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain.
- Departament d'Enginyeria Química, Escola Tècnica Superior d'Enginyeria, Universitat de València, Av. de la Universitat s/n, 46100 Burjassot, Spain.
| | - Jordi Bou
- Departament d'Enginyeria Química, Universitat Politècnica de Catalunya, Av. Diagonal 647 (ETSEIB), 08028 Barcelona, Spain.
| | - Amparo Ribes-Greus
- Instituto de Tecnología de Materiales (ITM), Universitat Politècnica de València, Camino de Vera s/n, 46022 Valencia, Spain.
| |
Collapse
|
42
|
Bonartsev AP, Bonartseva GA, Reshetov IV, Kirpichnikov MP, Shaitan KV. Application of Polyhydroxyalkanoates in Medicine and the Biological Activity of Natural Poly(3-Hydroxybutyrate). Acta Naturae 2019; 11:4-16. [PMID: 31413875 PMCID: PMC6643351 DOI: 10.32607/20758251-2019-11-2-4-16] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2019] [Indexed: 12/18/2022] Open
Abstract
Biodegradable and biocompatible polymers, polyhydroxyalkanoates (PHAs), are actively used in medicine to produce a wide range of medical devices and dosage formulations. The medical industry mainly utilizes PHAs obtained by chemical synthesis, but interest in the medical application of natural PHAs obtained biotechnologically is also growing. Synthetic PHAs are the biomimetic analogs of bacterial poly(3-hydroxybutyrate) (PHB) and other natural PHAs. This paper addresses the issue of the presence of biological activity in synthetic and natural PHAs (stimulation of cell proliferation and differentiation, tissue regeneration) and their possible association with various biological functions of PHB in bacteria and eukaryotes, including humans.
Collapse
Affiliation(s)
- A. P. Bonartsev
- Faculty of Biology, M.V. Lomonosov Moscow State University, Leninskie Gory 1, bldg. 12, Moscow, 119234, Russia
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Ave. 33, bldg. 2, Moscow, 119071, Russia
| | - G. A. Bonartseva
- A.N. Bach Institute of Biochemistry, Research Center of Biotechnology of the Russian Academy of Sciences, Leninsky Ave. 33, bldg. 2, Moscow, 119071, Russia
| | - I. V. Reshetov
- Sechenov First Moscow State University, Trubetskaya Str. 8, bldg. 2, Moscow, 119991, Russia
| | - M. P. Kirpichnikov
- Faculty of Biology, M.V. Lomonosov Moscow State University, Leninskie Gory 1, bldg. 12, Moscow, 119234, Russia
| | - K. V. Shaitan
- Faculty of Biology, M.V. Lomonosov Moscow State University, Leninskie Gory 1, bldg. 12, Moscow, 119234, Russia
| |
Collapse
|
43
|
Xu JY, Xiong YY, Lu XT, Yang YJ. Regulation of Type 2 Immunity in Myocardial Infarction. Front Immunol 2019; 10:62. [PMID: 30761134 PMCID: PMC6362944 DOI: 10.3389/fimmu.2019.00062] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2018] [Accepted: 01/11/2019] [Indexed: 12/12/2022] Open
Abstract
Type 2 immunity participates in the pathogeneses of helminth infection and allergic diseases. Emerging evidence indicates that the components of type 2 immunity are also involved in maintaining metabolic hemostasis and facilitating the healing process after tissue injury. Numerous preclinical studies have suggested regulation of type 2 immunity-related cytokines, such as interleukin-4, -13, and -33, and cell types, such as M2 macrophages, mast cells, and eosinophils, affects cardiac functions after myocardial infarction (MI), providing new insights into the importance of immune modulation in the infarcted heart. This review provides an overview of the functions of these cytokines and cells in the setting of MI as well as their potential to predict the severity and prognosis of MI.
Collapse
Affiliation(s)
- Jun-Yan Xu
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Yu-Yan Xiong
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| | - Xiao-Tong Lu
- National Cancer Center/National Clinical Research Center for Cancer/Cancer Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yue-Jin Yang
- State Key Laboratory of Cardiovascular Disease, Department of Cardiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Science and Peking Union Medical College, Beijing, China
| |
Collapse
|
44
|
Chen WL, Kan CD. Using cell-seeded electrospun patch for myocardial injury: in-vitro and in rat model. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2018; 2018:5338-5341. [PMID: 30441542 DOI: 10.1109/embc.2018.8513557] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Electrospinning has been widely used to fabricate scaffolds and commonly used biodegradable polymers. Cellular cardiomyoplasty is a type of regenerative medicine that has potential use for treatment of myocardial infarction or terminal cardiac failure. The aims of this study are to use electrospinning to create cardiovascular patches and to assess their potential therapeutic use by transplantation into the hearts of rats. Tissue engineering scaffolds were generated by use of electrospinning, in which the fibers consist of nanoscale-to-microscale fibers whose diameters are comparable to those of essential components of the extracellular matrix. A polymer solution was pumped at a constant rate through a syringe with a small-diameter needle that is connected to a high-voltage source, so that an electric field is created between the needle and a metallic collecting plate. The final product is a mat composed of individual continuous nanofibers. Cell survival, cell characteristics, and growth factors of electrospun patches of different thicknesses using bone marrow and human cardiac stem cells were tested. The results demonstrated that the cells can survive in Poly-caprolactone (PCL) patches, even deep within these patches. The PCL patches are nontoxic and do not alter cell properties. Transplantation of these patches into the hearts of a rat model of myocardial infarction led to strong compliance and good survival.The use of PCL cellular patches is feasible method for cellular transplantation. Future studies should attempt to use orientated electrospun cellular patches to improve overall cell survival within deeper layers of these patches.
Collapse
|
45
|
Bonartsev AP, Voinova VV, Bonartseva GA. Poly(3-hydroxybutyrate) and Human Microbiota (Review). APPL BIOCHEM MICRO+ 2018; 54:547-568. [DOI: 10.1134/s0003683818060066] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2018] [Indexed: 01/11/2025]
|
46
|
Timin AS, Muslimov AR, Zyuzin MV, Peltek OO, Karpov TE, Sergeev IS, Dotsenko AI, Goncharenko AA, Yolshin ND, Sinelnik A, Krause B, Baumbach T, Surmeneva MA, Chernozem RV, Sukhorukov GB, Surmenev RA. Multifunctional Scaffolds with Improved Antimicrobial Properties and Osteogenicity Based on Piezoelectric Electrospun Fibers Decorated with Bioactive Composite Microcapsules. ACS APPLIED MATERIALS & INTERFACES 2018; 10:34849-34868. [PMID: 30230807 DOI: 10.1021/acsami.8b09810] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2023]
Abstract
The incorporation of bioactive compounds onto polymer fibrous scaffolds with further control of drug release kinetics is essential to improve the functionality of scaffolds for personalized drug therapy and regenerative medicine. In this study, polymer and hybrid microcapsules were prepared and used as drug carriers, which are further deposited onto polymer microfiber scaffolds [polycaprolactone (PCL), poly(3-hydroxybutyrate) (PHB), and PHB doping with the conductive polyaniline (PANi) of 2 wt % (PHB-PANi)]. The number of immobilized microcapsules decreased with increase in their ζ-potential due to electrostatic repulsion with the negatively charged fiber surface, depending on the polymer used for the scaffold's fabrication. Additionally, the immobilization of the capsules in dynamic mechanical conditions at a frequency of 10 Hz resulted in an increase in the number of the capsules on the fibers with increase in the scaffold piezoelectric response in the order PCL < PHB < PHB-PANi, depending on the chemical composition of the capsules. The immobilization of microcapsules loaded with different bioactive molecules onto the scaffold surface enabled multimodal triggering by physical (ultrasound, laser radiation) and biological (enzymatic treatment) stimuli, providing controllable release of the cargo from scaffolds. Importantly, the microcapsules immobilized onto the surface of the scaffolds did not influence the cell growth, viability, and cell proliferation on the scaffolds. Moreover, the attachment of human mesenchymal stem cells (hMSCs) on the scaffolds revealed that the PHB and PHB-PANi scaffolds promoted adhesion of hMSCs compared to that of the PCL scaffolds. Two bioactive compounds, antibiotic ceftriaxone sodium (CS) and osteogenic factor dexamethasone (DEXA), were chosen to load the microcapsules and demonstrate the antimicrobial properties and osteogenesis of the scaffolds. The modified scaffolds had prolonged release of CS or DEXA, which provided an improved antimicrobial effect, as well as enhanced osteogenic differentiation and mineralization of the scaffolds modified with capsules compared to that of individual scaffolds soaked in CS solution or incubated in an osteogenic medium. Thus, the immobilization of microcapsules provides a simple, convenient way to incorporate bioactive compounds onto polymer scaffolds, which makes these multimodal materials suitable for personalized drug therapy and bone tissue engineering.
Collapse
Affiliation(s)
- Alexander S Timin
- First I. P. Pavlov State Medical University of St. Petersburg , Lev Tolstoy Street, 6/8 , 197022 St. Petersburg , Russian Federation
- Physical Materials Science and Composite Materials Centre , National Research Tomsk Polytechnic University , Lenin Avenue, 30 , 634050 Tomsk , Russian Federation
| | - Albert R Muslimov
- First I. P. Pavlov State Medical University of St. Petersburg , Lev Tolstoy Street, 6/8 , 197022 St. Petersburg , Russian Federation
- Peter the Great St. Petersburg Polytechnic University , Polytechnicheskaya, 29 , 195251 St. Petersburg , Russian Federation
- Smorodintsev Influenza Research Institute , Ministry of Healthcare of the Russian Federation , Prof. Popova Street, 15/17 , 197376 St. Petersburg , Russian Federation
| | | | - Oleksii O Peltek
- Peter the Great St. Petersburg Polytechnic University , Polytechnicheskaya, 29 , 195251 St. Petersburg , Russian Federation
| | - Timofey E Karpov
- Peter the Great St. Petersburg Polytechnic University , Polytechnicheskaya, 29 , 195251 St. Petersburg , Russian Federation
| | - Igor S Sergeev
- Peter the Great St. Petersburg Polytechnic University , Polytechnicheskaya, 29 , 195251 St. Petersburg , Russian Federation
| | - Anna I Dotsenko
- First I. P. Pavlov State Medical University of St. Petersburg , Lev Tolstoy Street, 6/8 , 197022 St. Petersburg , Russian Federation
| | - Alexander A Goncharenko
- Peter the Great St. Petersburg Polytechnic University , Polytechnicheskaya, 29 , 195251 St. Petersburg , Russian Federation
| | - Nikita D Yolshin
- Smorodintsev Influenza Research Institute , Ministry of Healthcare of the Russian Federation , Prof. Popova Street, 15/17 , 197376 St. Petersburg , Russian Federation
| | | | - Bärbel Krause
- Institute for Photon Science and Synchrotron Radiation , Karlsruhe Institute of Technology , 76344 Eggenstein-Leopoldshafen , Germany
| | - Tilo Baumbach
- Institute for Photon Science and Synchrotron Radiation , Karlsruhe Institute of Technology , 76344 Eggenstein-Leopoldshafen , Germany
- Laboratory for Applications of Synchrotron Radiation (LAS) , Karlsruhe Institute of Technology (KIT) , 76049 Karlsruhe , Germany
| | - Maria A Surmeneva
- Physical Materials Science and Composite Materials Centre , National Research Tomsk Polytechnic University , Lenin Avenue, 30 , 634050 Tomsk , Russian Federation
| | - Roman V Chernozem
- Physical Materials Science and Composite Materials Centre , National Research Tomsk Polytechnic University , Lenin Avenue, 30 , 634050 Tomsk , Russian Federation
| | - Gleb B Sukhorukov
- Physical Materials Science and Composite Materials Centre , National Research Tomsk Polytechnic University , Lenin Avenue, 30 , 634050 Tomsk , Russian Federation
- Peter the Great St. Petersburg Polytechnic University , Polytechnicheskaya, 29 , 195251 St. Petersburg , Russian Federation
- School of Engineering and Materials Science , Queen Mary University of London , Mile End Road , London E1 4NS , United Kingdom
| | - Roman A Surmenev
- Physical Materials Science and Composite Materials Centre , National Research Tomsk Polytechnic University , Lenin Avenue, 30 , 634050 Tomsk , Russian Federation
| |
Collapse
|
47
|
Das S, Jang J. 3D bioprinting and decellularized ECM-based biomaterials for in vitro CV tissue engineering. ACTA ACUST UNITED AC 2018. [DOI: 10.2217/3dp-2018-0002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
Advanced extrusion-based 3D printing strategies allow the rapid fabrication of complex anatomically relevant architectures. Moreover, they have the potential to fabricate 3D-bioprinted cardiac constructs by depositing cardiac cells with appropriate biomaterials. Heart-derived decellularized extracellular matrices containing a complex mixture of various extracellular molecules provide a comprehensive microenvironmental niche similar to native cardiac tissue. Nonetheless, a major concern persists pertaining to insufficient vascularization and mimicking of the complex 3D architectural features, which can be tackled using 3D printing approaches. In this review, we discuss the advantage and application of decellularized extracellular matrix-based hydrogels for the 3D printing of engineered cardiac tissues. We also briefly talk about the integration of electroactive materials within cardiac patches to improve the myocardium's electrophysiological properties.
Collapse
Affiliation(s)
- Sanskrita Das
- Department of Creative IT Engineering, Pohang University of Science & Technology, Pohang, 37673, Republic of Korea
| | - Jinah Jang
- Department of Creative IT Engineering, Pohang University of Science & Technology, Pohang, 37673, Republic of Korea
- School of Interdisciplinary Bioscience and Bioengineering (IBIO), Pohang University of Science & Technology, Pohang, 37673, Republic of Korea
| |
Collapse
|
48
|
Zhuravleva M, Gilazieva Z, Grigoriev TE, Shepelev AD, Kh. Tenchurin T, Kamyshinsky R, Krasheninnikov SV, Orlov S, Caralogli G, Archipova S, Holterman MJ, Mavlikeev M, Deev RV, Chvalun SN, Macchiarini P. In vitroassessment of electrospun polyamide-6 scaffolds for esophageal tissue engineering. J Biomed Mater Res B Appl Biomater 2018; 107:253-268. [DOI: 10.1002/jbm.b.34116] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2017] [Revised: 02/08/2018] [Accepted: 02/26/2018] [Indexed: 12/11/2022]
Affiliation(s)
- Margarita Zhuravleva
- Laboratory of Bioengineering and Regenerative Medicine (BioReM); Kazan Federal University; Kazan Russia
| | - Zarema Gilazieva
- Laboratory of Bioengineering and Regenerative Medicine (BioReM); Kazan Federal University; Kazan Russia
| | | | | | | | | | | | - Sergei Orlov
- Scientific Research Institute of Medical Primatology; Sochi Russia
| | - Gina Caralogli
- Scientific Research Institute of Medical Primatology; Sochi Russia
| | - Svetlana Archipova
- Laboratory of Bioengineering and Regenerative Medicine (BioReM); Kazan Federal University; Kazan Russia
| | | | - Mikhail Mavlikeev
- Laboratory of Bioengineering and Regenerative Medicine (BioReM); Kazan Federal University; Kazan Russia
| | - Roman V. Deev
- Human Stem Cells Institute, Moscow, Russia; Ryazan State Medical University; Ryazan Russia
| | | | - Paolo Macchiarini
- Laboratory of Bioengineering and Regenerative Medicine (BioReM); Kazan Federal University; Kazan Russia
| |
Collapse
|
49
|
Becker M, Maring JA, Schneider M, Herrera Martin AX, Seifert M, Klein O, Braun T, Falk V, Stamm C. Towards a Novel Patch Material for Cardiac Applications: Tissue-Specific Extracellular Matrix Introduces Essential Key Features to Decellularized Amniotic Membrane. Int J Mol Sci 2018; 19:E1032. [PMID: 29596384 PMCID: PMC5979550 DOI: 10.3390/ijms19041032] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2018] [Revised: 03/26/2018] [Accepted: 03/27/2018] [Indexed: 12/18/2022] Open
Abstract
There is a growing need for scaffold material with tissue-specific bioactivity for use in regenerative medicine, tissue engineering, and for surgical repair of structural defects. We developed a novel composite biomaterial by processing human cardiac extracellular matrix (ECM) into a hydrogel and combining it with cell-free amniotic membrane via a dry-coating procedure. Cardiac biocompatibility and immunogenicity were tested in vitro using human cardiac fibroblasts, epicardial progenitor cells, murine HL-1 cells, and human immune cells derived from buffy coat. Processing of the ECM preserved important matrix proteins as demonstrated by mass spectrometry. ECM coating did not alter the mechanical characteristics of decellularized amniotic membrane but did cause a clear increase in adhesion capacity, cell proliferation and viability. Activated monocytes secreted less pro-inflammatory cytokines, and both macrophage polarization towards the pro-inflammatory M1 type and T cell proliferation were prevented. We conclude that the incorporation of human cardiac ECM hydrogel shifts and enhances the bioactivity of decellularized amniotic membrane, facilitating its use in future cardiac applications.
Collapse
Affiliation(s)
- Matthias Becker
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany.
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353 Berlin, Germany.
| | - Janita A Maring
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany.
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353 Berlin, Germany.
| | - Maria Schneider
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany.
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353 Berlin, Germany.
| | - Aarón X Herrera Martin
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany.
- Julius Wolff Institute for Biomechanics and Musculoskeletal Regeneration, 13353 Berlin, Germany.
| | - Martina Seifert
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany.
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353 Berlin, Germany.
| | - Oliver Klein
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany.
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353 Berlin, Germany.
| | - Thorsten Braun
- Department of Obstetrics and Gynecology, Charite Medical University, 13353 Berlin, Germany.
| | - Volkmar Falk
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353 Berlin, Germany.
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, 13316 Berlin, Germany.
- Deutsches Herzzentrum Berlin (DHZB), Augustenburger Platz 1, 13353 Berlin, Germany.
| | - Christof Stamm
- Charité-Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin, Humboldt-Universität zu Berlin, and Berlin Institute of Health, 13353 Berlin, Germany.
- Berlin-Brandenburg Center for Regenerative Therapies (BCRT), 13353 Berlin, Germany.
- German Centre for Cardiovascular Research (DZHK), Partner Site Berlin, 13316 Berlin, Germany.
- Deutsches Herzzentrum Berlin (DHZB), Augustenburger Platz 1, 13353 Berlin, Germany.
| |
Collapse
|
50
|
Lakshmanan R, Maulik N. Development of next generation cardiovascular therapeutics through bio-assisted nanotechnology. J Biomed Mater Res B Appl Biomater 2017; 106:2072-2083. [DOI: 10.1002/jbm.b.34000] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2017] [Revised: 08/14/2017] [Accepted: 09/01/2017] [Indexed: 12/14/2022]
Affiliation(s)
- Rajesh Lakshmanan
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery; UConn Health; Farmington Connecticut
| | - Nilanjana Maulik
- Molecular Cardiology and Angiogenesis Laboratory, Department of Surgery; UConn Health; Farmington Connecticut
| |
Collapse
|