101
|
Doryab A, Schmid O. Bioactive Cell-Derived ECM Scaffold Forms a Unique Cellular Microenvironment for Lung Tissue Engineering. Biomedicines 2022; 10:biomedicines10081791. [PMID: 35892691 PMCID: PMC9394345 DOI: 10.3390/biomedicines10081791] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Revised: 07/19/2022] [Accepted: 07/22/2022] [Indexed: 12/11/2022] Open
Abstract
Chronic lung diseases are one of the leading causes of death worldwide. Lung transplantation is currently the only causal therapeutic for lung diseases, which is restricted to end-stage disease and limited by low access to donor lungs. Lung tissue engineering (LTE) is a promising approach to regenerating a replacement for at least a part of the damaged lung tissue. Currently, lung regeneration is limited to a simplified local level (e.g., alveolar−capillary barrier) due to the sophisticated and complex structure and physiology of the lung. Here, we introduce an extracellular matrix (ECM)-integrated scaffold using a cellularization−decellularization−recellularization technique. This ECM-integrated scaffold was developed on our artificial co-polymeric BETA (biphasic elastic thin for air−liquid interface cell culture conditions) scaffold, which were initially populated with human lung fibroblasts (IMR90 cell line), as the main generator of ECM proteins. Due to the interconnected porous structure of the thin (<5 µm) BETA scaffold, the cells can grow on and infiltrate into the scaffold and deposit their own ECM. After a mild decellularization procedure, the ECM proteins remained on the scaffold, which now closely mimicked the cellular microenvironment of pulmonary cells more realistically than the plain artificial scaffolds. We assessed several decellularization methods and found that 20 mM NH4OH and 0.1% Triton X100 with subsequent DNase treatment completely removed the fibroblasts (from the first cellularization) and maintains collagen I and IV as the key ECM proteins on the scaffold. We also showed the repopulation of the primary fibroblast from human (without chronic lung disease (non-CLD) donors) and human bronchial epithelial (16HBE14o−) cells on the ECM-integrated BETA scaffold. With this technique, we developed a biomimetic scaffold that can mimic both the physico-mechanical properties and the native microenvironment of the lung ECM. The results indicate the potential of the presented bioactive scaffold for LTE application.
Collapse
|
102
|
Santra M, Liu YC, Jhanji V, Yam GHF. Human SMILE-Derived Stromal Lenticule Scaffold for Regenerative Therapy: Review and Perspectives. Int J Mol Sci 2022; 23:ijms23147967. [PMID: 35887309 PMCID: PMC9315730 DOI: 10.3390/ijms23147967] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 07/10/2022] [Accepted: 07/18/2022] [Indexed: 12/13/2022] Open
Abstract
A transparent cornea is paramount for vision. Corneal opacity is one of the leading causes of blindness. Although conventional corneal transplantation has been successful in recovering patients’ vision, the outcomes are challenged by a global lack of donor tissue availability. Bioengineered corneal tissues are gaining momentum as a new source for corneal wound healing and scar management. Extracellular matrix (ECM)-scaffold-based engineering offers a new perspective on corneal regenerative medicine. Ultrathin stromal laminar tissues obtained from lenticule-based refractive correction procedures, such as SMall Incision Lenticule Extraction (SMILE), are an accessible and novel source of collagen-rich ECM scaffolds with high mechanical strength, biocompatibility, and transparency. After customization (including decellularization), these lenticules can serve as an acellular scaffold niche to repopulate cells, including stromal keratocytes and stem cells, with functional phenotypes. The intrastromal transplantation of these cell/tissue composites can regenerate native-like corneal stromal tissue and restore corneal transparency. This review highlights the current status of ECM-scaffold-based engineering with cells, along with the development of drug and growth factor delivery systems, and elucidates the potential uses of stromal lenticule scaffolds in regenerative therapeutics.
Collapse
Affiliation(s)
- Mithun Santra
- Corneal Regeneration Laboratory, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (M.S.); (V.J.)
| | - Yu-Chi Liu
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore;
- Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Medical School, Singapore 169857, Singapore
| | - Vishal Jhanji
- Corneal Regeneration Laboratory, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (M.S.); (V.J.)
| | - Gary Hin-Fai Yam
- Corneal Regeneration Laboratory, Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA; (M.S.); (V.J.)
- Tissue Engineering and Cell Therapy Group, Singapore Eye Research Institute, Singapore 169856, Singapore;
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA 15213, USA
- Correspondence:
| |
Collapse
|
103
|
Mahendiran B, Muthusamy S, Sampath S, Jaisankar SN, Selvakumar R, Krishnakumar GS. In vitro and in vivo biocompatibility of decellularized cellulose scaffolds functionalized with chitosan and platelet rich plasma for tissue engineering applications. Int J Biol Macromol 2022; 217:522-535. [PMID: 35841966 DOI: 10.1016/j.ijbiomac.2022.07.052] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 07/07/2022] [Accepted: 07/07/2022] [Indexed: 01/14/2023]
Abstract
This study describes the fabrication of cellulose scaffold (CS) and cellulose-chitosan (CS/CHI) scaffolds from the immature endosperm of Borassus flabellifer (Linn.) (BF) loaded with platelet rich plasma (PRP). Thus, developed scaffolds were evaluated for their physicochemical and mechanical behavior, growth factor release and biological performance. Additionally, in vivo response was assessed in a sub cutaneous rat model to study vascularization, host inflammatory response and macrophage polarization. The results of this study demonstrated that CS and CS/CHI scaffolds with PRP demonstrated favorable physiochemical and morphogical properties. The scaffold groups CS-PRP and CS/CHI-PRP were able to release growth factors in a well sustained manner under physiological conditions. The presence of PRP in cellulosic scaffolds did show significant differences in their behavior when investigated under in vitro studies, where the release of diverse cytokines improved the cellular proliferation and differentiation of osteoblasts. Finally, the PRP enriched scaffolds when studied under in vivo conditions showed increased angiogenesis and re-epithelialization with adequate collagen deposition and tissue remodeling. Our results suggest that besides the conventional carrier systems, this new-generation of plant-based cellulosic scaffolds with/without any modification can serve as a suitable carrier for PRP encapsulation and release, which can be used in numerous tissue regenerative therapies.
Collapse
Affiliation(s)
- Balaji Mahendiran
- Department of Biotechnology, Applied Biomaterials Laboratory, PSG Institute of Advanced Studies, Coimbatore, Tamil Nadu, India
| | - Shalini Muthusamy
- Department of Biotechnology, Applied Biomaterials Laboratory, PSG Institute of Advanced Studies, Coimbatore, Tamil Nadu, India
| | - Sowndarya Sampath
- Department of Polymer Science and Technology, Council of Scientific and Industrial Research-Central Leather Research Institute, Adyar, Chennai, Tamil Nadu, India
| | - S N Jaisankar
- Department of Polymer Science and Technology, Council of Scientific and Industrial Research-Central Leather Research Institute, Adyar, Chennai, Tamil Nadu, India
| | - R Selvakumar
- Department of Nanobiotechnology, Tissue Engineering Laboratory, PSG Institute of Advanced Studies, Coimbatore, Tamil Nadu, India
| | - Gopal Shankar Krishnakumar
- Department of Biotechnology, Applied Biomaterials Laboratory, PSG Institute of Advanced Studies, Coimbatore, Tamil Nadu, India.
| |
Collapse
|
104
|
Amereh M, Seyfoori A, Akbari M. In Vitro Brain Organoids and Computational Models to Study Cell Death in Brain Diseases. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2022; 2515:281-296. [PMID: 35776358 DOI: 10.1007/978-1-0716-2409-8_17] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 10/17/2022]
Abstract
Understanding the mechanisms underlying the formation and progression of brain diseases is challenging due to the vast variety of involved genetic/epigenetic factors and the complexity of the environment of the brain. Current preclinical monolayer culture systems fail to faithfully recapitulate the in vivo complexities of the brain. Organoids are three-dimensional (3D) culture systems that mimic much of the complexities of the brain including cell-cell and cell-matrix interactions. Complemented with a theoretical framework to model the dynamic interactions between different components of the brain, organoids can be used as a potential tool for studying disease progression, transport of therapeutic agents in tissues, drug screening, and toxicity analysis. In this chapter, we first report on the fabrication and use of a novel self-filling microwell arrays (SFMWs) platform that is self-filling and enables the formation of organoids with uniform size distributions. Next, we will introduce a mathematical framework that predicts the organoid growth, cell death, and the therapeutic responses of the organoids to different therapeutic agents. Through systematic investigations, the computational model can identify shortcomings of in vitro assays and reduce the time and effort required to improve preclinical tumor models' design. Lastly, the mathematical model provides new testable hypotheses and encourages mathematically driven experiments.
Collapse
Affiliation(s)
- Meitham Amereh
- Laboratory for Innovations in Microengineering (LiME), University of Victoria, Victoria, BC, Canada.,Department of Mechanical Engineering, University of Victoria, Victoria, BC, Canada
| | - Amir Seyfoori
- Laboratory for Innovations in Microengineering (LiME), University of Victoria, Victoria, BC, Canada.,Center for Advanced Materials and Related Technologies (CAMTEC), University of Victoria, Victoria, BC, Canada
| | - Mohsen Akbari
- Laboratory for Innovations in Microengineering (LiME), University of Victoria, Victoria, BC, Canada. .,Center for Advanced Materials and Related Technologies (CAMTEC), University of Victoria, Victoria, BC, Canada.
| |
Collapse
|
105
|
Allur Subramanian S, Oh S, Mariadoss AVA, Chae S, Dhandapani S, Parasuraman PS, Song SY, Woo C, Dong X, Choi JY, Kim SJ. Tunable mechanical properties of Mo 3Se 3-poly vinyl alcohol-based/silk fibroin-based nanowire ensure the regeneration mechanism in tenocytes derived from human bone marrow stem cells. Int J Biol Macromol 2022; 210:196-207. [PMID: 35513108 DOI: 10.1016/j.ijbiomac.2022.04.211] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Revised: 03/21/2022] [Accepted: 04/28/2022] [Indexed: 02/07/2023]
Abstract
Silk fibroin (SF) and poly vinyl alcohol (PVA)-based nanomaterial has exceptional attention in regenerative medicine. However, the preparation of SF and PVA-based nanomaterials in the desired form is complex due to their poor mechanical strength, brittleness, and compatibility. To this end, Mo3Se3 is chosen as a bio-nanowire to fabricate by combining PVA and SF to improve the mechanical properties. Physicochemical and structural features of the Mo3Se3-PVA-SF nanowire hydrogel (Mo3Se3-PVA-SF-NWH) were characterized by field emission scanning electron microscope (FE-SEM). Mechanical properties, degradation ratio, hydrophilicity, water uptake capacity, biocompatibility, and biological activity of the hydrogel were also studied. Superior interactions were formed between the reinforcing molecules of Mo3Se3 and PVA/SF in the hydrogel network by introducing Mo3Se3 nanowire (NW) into the hydrogel. Conversely, Mo3Se3 NW imparts mechanical stability and robustness to the blends (hydrogel) with predictable long-term degradation characteristics. It was proven by in vitro biodegradable rate, and swelling behaviour was varied depending on the concentration of Mo3Se3 NW. Mo3Se3 reinforced the hydrogels and found high porosity with superior biocompatibility. Excellent cellular adaptation was analyzed by MTT assay, live/dead staining, western blot, and quantitative real-time polymerase chain reaction (qRT-PCR). It revealed moderate toxicity at a concentration of 0.02% among the control samples. There was no discernible difference in 0.01% and 0.005% of Mo3Se3-PVA-SF-NWH in tenocytes derived from human bone marrow mesenchymal stem cells (hBMSC). Hence, this Mo3Se3-PVA-SF-NWH might be considered biocompatible due to its biological activities and appropriate mechanical properties. Overall, the Mo3Se3-PVA-SF-NWH might be considered a biocompatible scaffold for the possible biomedical applications of tendon tissue engineering.
Collapse
Affiliation(s)
- Sivakumar Allur Subramanian
- Department of Orthopaedic Surgery, Dongtan Sacred Heart Hospital, Hallym University, College of Medicine, Hwaseong, Republic of Korea
| | - Seungbae Oh
- School of Advanced Materials Science & Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Arokia Vijaya Anand Mariadoss
- Department of Orthopaedic Surgery, Dongtan Sacred Heart Hospital, Hallym University, College of Medicine, Hwaseong, Republic of Korea
| | - Sudong Chae
- School of Advanced Materials Science & Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Sanjeevram Dhandapani
- Graduate School of Biotechnology, College of Life Science, Kyung Hee University, Yongin-si 17104, Gyeonggi-do, Republic of Korea
| | - Perumalswamy Sekar Parasuraman
- Department of Environmental and Biotechnology, Hallym University, 1 Hallymdeahak-gil, Chuncheon, Gangwon-do 200-702, Republic of Korea
| | - Si Young Song
- Department of Orthopaedic Surgery, Dongtan Sacred Heart Hospital, Hallym University, College of Medicine, Hwaseong, Republic of Korea
| | - Chaeheon Woo
- School of Advanced Materials Science & Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Xue Dong
- SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419, Republic of Korea
| | - Jae-Young Choi
- School of Advanced Materials Science & Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea; SKKU Advanced Institute of Nanotechnology (SAINT), Sungkyunkwan University (SKKU), Suwon, Gyeonggi 16419, Republic of Korea.
| | - Sung Jae Kim
- Department of Orthopaedic Surgery, Dongtan Sacred Heart Hospital, Hallym University, College of Medicine, Hwaseong, Republic of Korea.
| |
Collapse
|
106
|
Guo J, Li Y, Gao Z, Lyu J, Liu W, Duan Y, Zhou L, Gu Q. 3D printed controllable microporous scaffolds support embryonic development in vitro. J Cell Physiol 2022; 237:3408-3420. [PMID: 35699648 PMCID: PMC9545995 DOI: 10.1002/jcp.30810] [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: 05/07/2022] [Revised: 05/21/2022] [Accepted: 05/25/2022] [Indexed: 11/22/2022]
Abstract
Little is known about the complex molecular and cellular events occurring during implantation, which represents a critical step for pregnancy. The conventional 2D culture could not support postimplantation embryos' normal development, and 3D conditions shed light into the “black box”. 3D printing technology has been widely used in recapitulating the structure and function of native tissues in vitro. Here, we 3D printed anisotropic microporous scaffolds to culture embryos by manipulating the advancing angle between printed layers, which affected embryo development. The 30° and 60° scaffolds promote embryo development with moderate embryo‐scaffold attachments. T‐positive cells and FOXA2‐positive cells were observed to appear in the posterior region of the embryo and migrated to the anterior region of the embryo on day 7. These findings demonstrate a 3D printed stand that supports embryonic development in vitro and the critical role of 3D architecture for embryo implantation, in which additive manufacturing is a versatile tool.
Collapse
Affiliation(s)
- Jia Guo
- The State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yuanyuan Li
- The State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,Department of Nephrology, Postdoctoral Workstation, Precision Medicine Center of Shanxi Provincial People's Hospital, The Affiliated People's Hospital of Shanxi Medical University, Shanxi Kidney Disease Institute, Taiyuan, China
| | - Zili Gao
- The State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Jiawei Lyu
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Wenli Liu
- The State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Yongchao Duan
- Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China.,The State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Lixun Zhou
- The State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| | - Qi Gu
- The State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China.,Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
107
|
Abstract
The successful transplantation of stem cells has the potential to transform regenerative medicine approaches and open promising avenues to repair, replace, and regenerate diseased, damaged, or aged tissues. However, pre-/post-transplantation issues of poor cell survival, retention, cell fate regulation, and insufficient integration with host tissues constitute significant challenges. The success of stem cell transplantation depends upon the coordinated sequence of stem cell renewal, specific lineage differentiation, assembly, and maintenance of long-term function. Advances in biomaterials can improve pre-/post-transplantation outcomes by integrating biophysiochemical cues and emulating tissue microenvironments. This review highlights leading biomaterials-based approaches for enhancing stem cell transplantation.
Collapse
Affiliation(s)
- Bhushan N Kharbikar
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Priya Mohindra
- UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA 94158, USA
| | - Tejal A Desai
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, San Francisco, CA 94158, USA; UC Berkeley-UCSF Graduate Program in Bioengineering, San Francisco, CA 94158, USA; Department of Bioengineering, University of California, Berkeley, Berkeley, CA 94720, USA; School of Engineering, Brown University, Providence, RI, 02912, USA.
| |
Collapse
|
108
|
Kaba S, Kawai Y, Tanigami Y, Ohnishi H, Kita T, Yoshimatsu M, Omori K, Kishimoto Y. Peroxisome Proliferator-Activated Receptor-γ Agonist Attenuates Vocal Fold Fibrosis in Rats via Regulation of Macrophage Activation. THE AMERICAN JOURNAL OF PATHOLOGY 2022; 192:771-782. [PMID: 35189097 DOI: 10.1016/j.ajpath.2022.02.002] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 01/19/2022] [Accepted: 02/07/2022] [Indexed: 12/30/2022]
Abstract
Macrophages aid in wound healing by changing their phenotype and can be a key driver of fibrosis. However, the contribution of macrophage phenotype to fibrosis following vocal fold injury remains unclear. Peroxisome proliferator-activated receptor-γ (PPARγ) is expressed mainly by macrophages during early wound healing and regulates the macrophage phenotype. This study aimed to evaluate the effects of pioglitazone (PIO), a PPARγ agonist, on the macrophage phenotype and fibrosis following vocal fold injury in rats. PIO was injected into the rat vocal folds on days 1, 3, 5, and 7 after injury, and the vocal fold lamina propria was evaluated on days 4 and 56 after injury. Moreover, THP-1-derived macrophages were treated with PIO, and the expression of proinflammatory cytokines under lipopolysaccharide/interferon-γ stimulation was analyzed. PIO reduced the expression of Ccl2 both in vivo and in vitro. Furthermore, PIO decreased the density of inducible nitric oxide synthase+ CD68+ macrophages and inhibited the expression of fibrosis-related factors on day 4 after injury. On day 56 after injury, PIO inhibited fibrosis, tissue contracture, and hyaluronic acid loss in a PPARγ-dependent manner. These results indicate that PPARγ activation could inhibit accumulation of inflammatory macrophages and improve tissue repair. Taken together, these findings imply that inflammatory macrophages play a key role in vocal fold fibrosis.
Collapse
Affiliation(s)
- Shinji Kaba
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan; Department of Otolaryngology, Kyoto Katsura Hospital, Kyoto, Japan
| | - Yoshitaka Kawai
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yuki Tanigami
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Hiroe Ohnishi
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomoko Kita
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Masayoshi Yoshimatsu
- Department of Otolaryngology, Head and Neck Surgery, Field of Sensory Organology, Graduate School of Medical and Dental Sciences, Kagoshima University, Kagoshima, Japan
| | - Koichi Omori
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Yo Kishimoto
- Department of Otolaryngology-Head and Neck Surgery, Graduate School of Medicine, Kyoto University, Kyoto, Japan.
| |
Collapse
|
109
|
Yan Y, Yao R, Zhao J, Chen K, Duan L, Wang T, Zhang S, Guan J, Zheng Z, Wang X, Liu Z, Li Y, Li G. Implantable nerve guidance conduits: Material combinations, multi-functional strategies and advanced engineering innovations. Bioact Mater 2022; 11:57-76. [PMID: 34938913 PMCID: PMC8665266 DOI: 10.1016/j.bioactmat.2021.09.030] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 09/17/2021] [Accepted: 09/26/2021] [Indexed: 01/15/2023] Open
Abstract
Nerve guidance conduits (NGCs) have attracted much attention due to their great necessity and applicability in clinical use for the peripheral nerve repair. Great efforts in recent years have been devoted to the development of high-performance NGCs using various materials and strategies. The present review provides a comprehensive overview of progress in the material innovation, structural design, advanced engineering technologies and multi functionalization of state-of-the-art nerve guidance conduits NGCs. Abundant advanced engineering technologies including extrusion-based system, laser-based system, and novel textile forming techniques in terms of weaving, knitting, braiding, and electrospinning techniques were also analyzed in detail. Findings arising from this review indicate that the structural mimetic NGCs combined with natural and synthetic materials using advanced manufacturing technologies can make full use of their complementary advantages, acquiring better biomechanical properties, chemical stability and biocompatibility. Finally, the existing challenges and future opportunities of NGCs were put forward aiming for further research and applications of NGCs.
Collapse
Affiliation(s)
- Yixin Yan
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, 215123, China
- Department of Materials, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Ruotong Yao
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, 215123, China
| | - Jingyuan Zhao
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, 215123, China
| | - Kaili Chen
- Department of Materials, Imperial College London, SW7 2AZ, UK
| | - Lirong Duan
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, 215123, China
| | - Tian Wang
- Wilson College of Textiles, North Carolina State University, Raleigh, 27695, USA
| | - Shujun Zhang
- Research Centre of Printed Flexible Electronics, School of Materials Science and Engineering, Harbin Institute of Technology, Shenzhen, 518055, China
| | - Jinping Guan
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, 215123, China
| | - Zhaozhu Zheng
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, 215123, China
| | - Xiaoqin Wang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, 215123, China
| | - Zekun Liu
- Department of Materials, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Yi Li
- Department of Materials, University of Manchester, Oxford Road, Manchester, M13 9PL, UK
| | - Gang Li
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou, 215123, China
| |
Collapse
|
110
|
Nagiah N, El Khoury R, Othman MH, Akimoto J, Ito Y, Roberson DA, Joddar B. Development and Characterization of Furfuryl-Gelatin Electrospun Scaffolds for Cardiac Tissue Engineering. ACS OMEGA 2022; 7:13894-13905. [PMID: 35559153 PMCID: PMC9088935 DOI: 10.1021/acsomega.2c00271] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Accepted: 04/05/2022] [Indexed: 05/14/2023]
Abstract
In this study, three types of electrospun scaffolds, including furfuryl-gelatin (f-gelatin) alone, f-gelatin with polycaprolactone (PCL) in a 1:1 ratio, and coaxial scaffolds with PCL (core) and f-gelatin (sheath), were developed for tissue engineering applications. Scaffolds were developed through single nozzle electrospinning and coaxial electrospinning, respectively, to serve as scaffolds for cardiac tissue engineering. Uniform fibrous structures were revealed in the scaffolds with significantly varying average fiber diameters of 760 ± 80 nm (f-gelatin), 420 ± 110 nm [f-gelatin and PCL (1:1)], and 810 ± 60 nm (coaxial f-gelatin > PCL) via scanning electron microscopy. The distinction between the core and the sheath of the fibers of the coaxial f-gelatin > PCL electrospun fibrous scaffolds was revealed by transmission electron microscopy. Thermal analysis and Fourier transformed infrared (FTIR) spectroscopy revealed no interactions between the polymers in the blended electrospun scaffolds. The varied blending methods led to significant differences in the elastic moduli of the electrospun scaffolds with the coaxial f-gelatin > PCL revealing the highest elastic modulus of all scaffolds (164 ± 3.85 kPa). All scaffolds exhibited excellent biocompatibility by supporting the adhesion and proliferation of human AC16 cardiomyocytes cells. The biocompatibility of the coaxial f-gelatin > PCL scaffolds with superior elastic modulus was assessed further through adhesion and functionality of human-induced pluripotent stem cell (hiPSC)-derived cardiomyocytes, thereby demonstrating the potential of the coaxially spun scaffolds as an ideal platform for developing cardiac tissue-on-a-chip models. Our results demonstrate a facile approach to produce visible light cross-linkable, hybrid, biodegradable nanofibrous scaffold biomaterials, which can serve as platforms for cardiac tissue engineered models.
Collapse
Affiliation(s)
- Naveen Nagiah
- Inspired
Materials & Stem-Cell Based Tissue Engineering Laboratory, Department
of Metallurgical, Materials, and Biomedical Engineering, M201 Engineering, The University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Raven El Khoury
- Inspired
Materials & Stem-Cell Based Tissue Engineering Laboratory, Department
of Metallurgical, Materials, and Biomedical Engineering, M201 Engineering, The University of Texas at El Paso, El Paso, Texas 79968, United States
| | - Mahmoud H. Othman
- Nano
Medical Engineering Laboratory, RIKEN Cluster
for Pioneering Research, Wako, Saitama 351-0198, Japan
| | - Jun Akimoto
- Emergent
Bioengineering Materials Research Team, RIKEN Center for Emergent Matter Science, Wako, Saitama 351-0198, Japan
| | - Yoshihiro Ito
- Nano
Medical Engineering Laboratory, RIKEN Cluster
for Pioneering Research, Wako, Saitama 351-0198, Japan
- Emergent
Bioengineering Materials Research Team, RIKEN Center for Emergent Matter Science, Wako, Saitama 351-0198, Japan
| | - David A. Roberson
- Polymer
Extrusion Lab, Department of Metallurgical, Materials, and Biomedical
Engineering, The University of Texas at
El Paso, El Paso, Texas 79968, United
States
| | - Binata Joddar
- Inspired
Materials & Stem-Cell Based Tissue Engineering Laboratory, Department
of Metallurgical, Materials, and Biomedical Engineering, M201 Engineering, The University of Texas at El Paso, El Paso, Texas 79968, United States
- Border
Biomedical Research Center, The University
of Texas at El Paso, 500 W. University Avenue, El Paso, Texas 79968, United
States
| |
Collapse
|
111
|
Biomechanically and biochemically functional scaffold for recruitment of endogenous stem cells to promote tendon regeneration. NPJ Regen Med 2022; 7:26. [PMID: 35474221 PMCID: PMC9043181 DOI: 10.1038/s41536-022-00220-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Accepted: 03/22/2022] [Indexed: 02/05/2023] Open
Abstract
Tendon regeneration highly relies on biomechanical and biochemical cues in the repair microenvironment. Herein, we combined the decellularized bovine tendon sheet (DBTS) with extracellular matrix (ECM) from tendon-derived stem cells (TDSCs) to fabricate a biomechanically and biochemically functional scaffold (tECM-DBTS), to provide a functional and stem cell ECM-based microenvironment for tendon regeneration. Our prior study showed that DBTS was biomechanically suitable to tendon repair. In this study, the biological function of tECM-DBTS was examined in vitro, and the efficiency of the scaffold for Achilles tendon repair was evaluated using immunofluorescence staining, histological staining, stem cell tracking, biomechanical and functional analyses. It was found that tECM-DBTS increased the content of bioactive factors and had a better performance for the proliferation, migration and tenogenic differentiation of bone marrow-derived stem cells (BMSCs) than DBTS. Furthermore, our results demonstrated that tECM-DBTS promoted tendon regeneration and improved the biomechanical properties of regenerated Achilles tendons in rats by recruiting endogenous stem cells and participating in the functionalization of these stem cells. As a whole, the results of this study demonstrated that the tECM-DBTS can provide a bionic microenvironment for recruiting endogenous stem cells and facilitating in situ regeneration of tendons.
Collapse
|
112
|
Singh S, Vashisth P, Meena VK, Kalyanasundaram D. Cellular studies and sustained drug delivery via nanostructures fabricated on 3D printed porous Neovius lattices of Ti6Al4V ELI. Biomed Mater 2022; 17. [PMID: 35447615 DOI: 10.1088/1748-605x/ac6922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2022] [Accepted: 04/21/2022] [Indexed: 11/11/2022]
Abstract
Site-specific drug delivery has the potential to reduce drug dosage by 3 to 5-folds. Given the propensity of drugs used in the treatment of tuberculosis and cancers, the increased drug dosages via oral ingestion for several months to a few years of medication is often detrimental to the health of patients. In this study, the sustained delivery of drugs with multiscale structured novel Neovius lattices was achieved. 3D Neovius Open Cell Lattices (NOCL) with porosities of 40, 45, and 50 % were fabricated layer-by-layer on the laser bed fusion process. Micron-sized Ti6Al4V Eli powder was used for 3D printing. The Young's modulus achieved from the novel Neovius lattices were in the range of 1.2 to 1.6 GPa, which is comparable to human cortical bone and helps to improve implant failure due to the stress shielding effect. To provide sustained drug delivery, nanotubes (NTs) were fabricated on NOCLs via high-voltage anodisation. The osteogenic agent icariin was loaded onto the NOCL-NT samples and their release profiles were studied for 7 days. A significantly steady and slow release rate of 0.05% per hour of the drug was achieved using NOCL-NT. In addition, the initial burst release of NOCL-NT was 4 fold lower than that of the open-cell lattices without nanotubes. Cellular studies using MG63 human osteoblast-like cells were performed to determine their biocompatibility and osteogenesis which were analysed using Calcein AM staining and Alamar Blue after 1, 5, and 7 days. 3D printed NOCL samples with NTs and with Icariin loaded NTs demonstrated a significant increase in cell proliferation as compared to as printed NOCL samples.
Collapse
Affiliation(s)
- Sonu Singh
- Indian Institute of Technology Delhi, Centre for Biomedical Engineering, New Delhi, 110016, INDIA
| | - Priya Vashisth
- Mechanical Engineering, Indian Institute of Technology Delhi, II/253, Department of Mechanical Engineering, Indian Institute of Technology Delhi, New Delhi, New Delhi, 110016, INDIA
| | - Vijay Kumar Meena
- Council of Scientific & Industrial Research, CSIR, Chandigarh, New Delhi, 110001, INDIA
| | - Dinesh Kalyanasundaram
- Indian Institute of Technology Delhi, Centre for Biomedical Engineering, New Delhi, 110016, INDIA
| |
Collapse
|
113
|
Nune M, Bhat M, Nagarajan A. Design of ECM Functionalized Polycaprolactone Aligned Nanofibers for Peripheral Nerve Tissue Engineering. J Med Biol Eng 2022. [DOI: 10.1007/s40846-022-00699-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Abstract
Purpose
Peripheral nerve injury (PNI) and its regeneration continue to remain a significant medical burden worldwide. The current treatment strategies used to treat PNI are often associated with multiple complications and yet do not achieve complete motor and sensory functions. Recently, synthetic biodegradable nerve conduits have become one the most commonly used conduits to repair small gaps in nerve injury. But they have not shown better results than nerve grafts possibly because of the lack of biological microenvironment required for axonal growth. Schwann cells play a very crucial role in peripheral nerve regeneration where activated SCs produce multiple neurotrophic factors that help in remyelination and immune modulation during nerve repair. Studies have shown that nanofibrous scaffolds have better bioactivity and more closely mimic the native structure of the extracellular matrix. Therefore, the present study was focused on designing a nanofibrous scaffold that would cover the roles of both structural support for the cells that can provide a microenvironment with biological cues for nerve growth and regeneration.
Methods
Decellularized Schwann cell ECM were spin-coated on polycaprolactone random and aligned nanofibrous scaffolds and their compatibility was evaluated using Schwann cells.
Results
Schwann cells displayed growth in the direction of the aligned PCL nanofibers and ACM treated exhibited appropriate bipolar morphology indicating that these modified fibers could provide directional cues making them highly suitable for neuronal cell growth.
Conclusion
Our results indicate that the fabricated aligned SC-ACM treated PCL scaffolds would be a potential biomaterial to treat peripheral nerve injuries and promote regeneration.
Graphical Abstract
Collapse
|
114
|
Collier CA, Mendiondo C, Raghavan S. Tissue engineering of the gastrointestinal tract: the historic path to translation. J Biol Eng 2022; 16:9. [PMID: 35379299 PMCID: PMC8981633 DOI: 10.1186/s13036-022-00289-6] [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: 01/05/2022] [Accepted: 03/08/2022] [Indexed: 11/15/2022] Open
Abstract
The gastrointestinal (GI) tract is imperative for multiple functions including digestion, nutrient absorption, and timely waste disposal. The central feature of the gut is peristalsis, intestinal motility, which facilitates all of its functions. Disruptions in GI motility lead to sub-optimal GI function, resulting in a lower quality of life in many functional GI disorders. Over the last two decades, tissue engineering research directed towards the intestine has progressed rapidly due to advances in cell and stem-cell biology, integrative physiology, bioengineering and biomaterials. Newer biomedical tools (including optical tools, machine learning, and nuanced regenerative engineering approaches) have expanded our understanding of the complex cellular communication within the GI tract that lead to its orchestrated physiological function. Bioengineering therefore can be utilized towards several translational aspects: (i) regenerative medicine to remedy/restore GI physiological function; (ii) in vitro model building to mimic the complex physiology for drug and pharmacology testing; (iii) tool development to continue to unravel multi-cell communication networks to integrate cell and organ-level physiology. Despite the significant strides made historically in GI tissue engineering, fundamental challenges remain including the quest for identifying autologous human cell sources, enhanced scaffolding biomaterials to increase biocompatibility while matching viscoelastic properties of the underlying tissue, and overall biomanufacturing. This review provides historic perspectives for how bioengineering has advanced over time, highlights newer advances in bioengineering strategies, and provides a realistic perspective on the path to translation.
Collapse
Affiliation(s)
- Claudia A Collier
- Department of Biomedical Engineering, Texas A&M University, Emerging Technologies Building, 3120 TAMU, College Station, TX, 77843, USA
| | - Christian Mendiondo
- Department of Biomedical Engineering, Texas A&M University, Emerging Technologies Building, 3120 TAMU, College Station, TX, 77843, USA
| | - Shreya Raghavan
- Department of Biomedical Engineering, Texas A&M University, Emerging Technologies Building, 3120 TAMU, College Station, TX, 77843, USA.
- Department of Nanomedicine, Houston Methodist Research Institute, Houston, TX, USA.
| |
Collapse
|
115
|
Zhi D, Cheng Q, Midgley AC, Zhang Q, Wei T, Li Y, Wang T, Ma T, Rafique M, Xia S, Cao Y, Li Y, Li J, Che Y, Zhu M, Wang K, Kong D. Mechanically reinforced biotubes for arterial replacement and arteriovenous grafting inspired by architectural engineering. SCIENCE ADVANCES 2022; 8:eabl3888. [PMID: 35294246 PMCID: PMC8926343 DOI: 10.1126/sciadv.abl3888] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/03/2023]
Abstract
There is a lack in clinically-suitable vascular grafts. Biotubes, prepared using in vivo tissue engineering, show potential for vascular regeneration. However, their mechanical strength is typically poor. Inspired by architectural design of steel fiber reinforcement of concrete for tunnel construction, poly(ε-caprolactone) (PCL) fiber skeletons (PSs) were fabricated by melt-spinning and heat treatment. The PSs were subcutaneously embedded to induce the assembly of host cells and extracellular matrix to obtain PS-reinforced biotubes (PBs). Heat-treated medium-fiber-angle PB (hMPB) demonstrated superior performance when evaluated by in vitro mechanical testing and following implantation in rat abdominal artery replacement models. hMPBs were further evaluated in canine peripheral arterial replacement and sheep arteriovenous graft models. Overall, hMPB demonstrated appropriate mechanics, puncture resistance, rapid hemostasis, vascular regeneration, and long-term patency, without incidence of luminal expansion or intimal hyperplasia. These optimized hMPB properties show promise as an alternatives to autologous vessels in clinical applications.
Collapse
Affiliation(s)
- Dengke Zhi
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Quhan Cheng
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Adam C. Midgley
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Qiuying Zhang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Tingting Wei
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Yi Li
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Ting Wang
- Urban Transport Emission Control Research Centre, College of Environmental Science and Engineering, Nankai University, Tianjin 300071, China
| | - Tengzhi Ma
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Muhammad Rafique
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
| | - Shuang Xia
- Department of Radiology, Tianjin Key Disciplines of Radiology, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China
| | - Yuejuan Cao
- Department of Vascular Surgery, Tianjin Union Medical Center, Nankai University, Tianjin 300121, China
| | - Yangchun Li
- Department of Vascular Surgery, Tianjin Union Medical Center, Nankai University, Tianjin 300121, China
| | - Jing Li
- Department of Ultrasound, Tianjin Union Medical Center, Nankai University, Tianjin 300121, China
| | - Yongzhe Che
- Department of Pathology and Anatomy, School of Medicine, Nankai University, Tianjin 300071, China
| | - Meifeng Zhu
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
- Corresponding author. (D.K.); (K.W.); (M.Z.)
| | - Kai Wang
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
- Corresponding author. (D.K.); (K.W.); (M.Z.)
| | - Deling Kong
- Key Laboratory of Bioactive Materials, Ministry of Education, College of Life Sciences, State Key Laboratory of Medicinal Chemical Biology, Nankai University, Tianjin 300071, China
- Institute of Transplant Medicine, Tianjin First Central Hospital, Nankai University, Tianjin 300192, China
- Corresponding author. (D.K.); (K.W.); (M.Z.)
| |
Collapse
|
116
|
Zakharova I, Saaya S, Shevchenko A, Stupnikova A, Zhiven' M, Laktionov P, Stepanova A, Romashchenko A, Yanshole L, Chernonosov A, Volkov A, Kizilova E, Zavjalov E, Chernyavsky A, Romanov A, Karpenko A, Zakian S. Mitomycin-Treated Endothelial and Smooth Muscle Cells Suitable for Safe Tissue Engineering Approaches. Front Bioeng Biotechnol 2022; 10:772981. [PMID: 35360387 PMCID: PMC8963790 DOI: 10.3389/fbioe.2022.772981] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 01/04/2022] [Indexed: 11/13/2022] Open
Abstract
In our previous study, we showed that discarded cardiac tissue from the right atrial appendage and right ventricular myocardium is an available source of functional endothelial and smooth muscle cells for regenerative medicine and tissue engineering. In the study, we aimed to find out what benefits are given by vascular cells from cardiac explants used for seeding on vascular patches engrafted to repair vascular defects in vivo. Additionally, to make the application of these cells safer in regenerative medicine we tested an in vitro approach that arrested mitotic division to avoid the potential tumorigenic effect of dividing cells. A tissue-engineered construction in the form of a patch based on a polycaprolactone-gelatin scaffold and seeded with endothelial and smooth muscle cells was implanted into the abdominal aorta of immunodeficient SCID mice. Aortic patency was assessed using ultrasound, MRI, immunohistochemical and histological staining. Endothelial and smooth muscle cells were treated with mitomycin C at a therapeutic concentration of 10 μg/ml for 2 h with subsequent analysis of cell proliferation and function. The absence of the tumorigenic effect of mitomycin C-treated cells, as well as their angiogenic potential, was examined by injecting them into immunodeficient mice. Cell-containing patches engrafted in the abdominal aorta of immunodeficient mice form the vessel wall loaded with the appropriate cells and extracellular matrix, and do not interfere with normal patency. Endothelial and smooth muscle cells treated with mitomycin C show no tumorigenic effect in the SCID immunodeficient mouse model. During in vitro experiments, we have shown that treatment with mitomycin C does not lead to a decrease in cell viability. Despite the absence of proliferation, mitomycin C-treated vascular cells retain specific cell markers, produce specific extracellular matrix, and demonstrate the ability to stimulate angiogenesis in vivo. We pioneered an approach to arresting cell division with mitomycin C in endothelial and smooth muscle cells from cardiac explant, which prevents the risk of malignancy from dividing cells in vascular surgery. We believe that this approach to the fabrication of tissue-engineered constructs based on mitotically inactivated cells from waste postoperative material may be valuable to bring closer the development of safe cell products for regenerative medicine.
Collapse
Affiliation(s)
- Irina Zakharova
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
- Institute of Chemical Biology and Fundamental Medicine, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- *Correspondence: Irina Zakharova,
| | - Shoraan Saaya
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - Alexander Shevchenko
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
- Institute of Chemical Biology and Fundamental Medicine, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Alena Stupnikova
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- Deparment of Natural Science, Novosibirsk State University, Novosibirsk, Russia
| | - Maria Zhiven'
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Pavel Laktionov
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
- Institute of Chemical Biology and Fundamental Medicine, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Alena Stepanova
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
- Institute of Chemical Biology and Fundamental Medicine, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Alexander Romashchenko
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Lyudmila Yanshole
- International Tomography Center,The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Alexander Chernonosov
- Institute of Chemical Biology and Fundamental Medicine, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Alexander Volkov
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - Elena Kizilova
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- Deparment of Natural Science, Novosibirsk State University, Novosibirsk, Russia
| | - Evgenii Zavjalov
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - Alexander Chernyavsky
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - Alexander Romanov
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - Andrey Karpenko
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
| | - Suren Zakian
- The Federal Research Center Institute of Cytology and Genetics, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
- E.N. Meshalkin National Medical Research Center, Ministry of Health of the Russian Federation, Novosibirsk, Russia
- Institute of Chemical Biology and Fundamental Medicine, The Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
117
|
Wang X, Liu P, Wu Q, Zheng Z, Xie M, Chen G, Yu J, Wang X, Li G, Kaplan D. Sustainable Antibacterial and Anti-Inflammatory Silk Suture with Surface Modification of Combined-Therapy Drugs for Surgical Site Infection. ACS APPLIED MATERIALS & INTERFACES 2022; 14:11177-11191. [PMID: 35192338 DOI: 10.1021/acsami.2c00106] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/16/2023]
Abstract
Silk sutures with antibacterial and anti-inflammatory functions were developed for sustained dual-drug delivery to prevent surgical site infections (SSIs). The silk sutures were prepared with core-shell structures braided from degummed silk filaments and then coated with a silk fibroin (SF) layer loaded with berberine (BB) and artemisinin (ART). Both the rapid release of drugs to prevent initial biofilm formation and the following sustained release to maintain effective concentrations for more than 42 days were demonstrated. In vitro assays using human fibroblasts (Hs 865.Sk) demonstrated cell proliferation on the materials, and hemolysis was 2.4 ± 0.8%, lower than that required by ISO 10993-4 standard. The sutures inhibited platelet adhesion and promoted collagen deposition and blood vessel formation. In vivo assessments using Sprague-Dawley (SD) rats indicated that the coating reduced the expression of pro-inflammatory cytokines interleukin-10 (IL-10) and tumor necrosis factor-α (TNF-α), shortening the inflammatory period and promoting angiogenesis. The results demonstrated that these new sutures exhibited stable structures, favorable biocompatibility, and sustainable antibacterial and anti-inflammatory functions with potential for surgical applications.
Collapse
Affiliation(s)
- Xuchen Wang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - Peixin Liu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
- Orthopedic Institute, Soochow University, Suzhou 215006, China
| | - Qinting Wu
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - Zhaozhu Zheng
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - Maobin Xie
- Department of Biomedical Engineering, School of Basic Medical Sciences, Guangzhou Medical University, Guangzhou 511436, China
| | - Guoqiang Chen
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - Jia Yu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou 215006, China
- Orthopedic Institute, Soochow University, Suzhou 215006, China
| | - Xiaoqin Wang
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - Gang Li
- National Engineering Laboratory for Modern Silk, College of Textile and Clothing Engineering, Soochow University, Suzhou 215123, China
| | - David Kaplan
- Department of Biomedical Engineering, Tufts University, 4 Colby Street, Medford, Massachusetts 02155, United States
| |
Collapse
|
118
|
Narciso M, Ulldemolins A, Júnior C, Otero J, Navajas D, Farré R, Gavara N, Almendros I. Novel Decellularization Method for Tissue Slices. Front Bioeng Biotechnol 2022; 10:832178. [PMID: 35356779 PMCID: PMC8959585 DOI: 10.3389/fbioe.2022.832178] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Accepted: 02/09/2022] [Indexed: 11/13/2022] Open
Abstract
Decellularization procedures have been developed and optimized for the entire organ or tissue blocks, by either perfusion of decellularizing agents through the tissue’s vasculature or submerging large sections in decellularizing solutions. However, some research aims require the analysis of native as well as decellularized tissue slices side by side, but an optimal protocol has not yet been established to address this need. Thus, the main goal of this work was to develop a fast and efficient decellularization method for tissue slices—with an emphasis on lung—while attached to a glass slide. To this end, different decellularizing agents were compared for their effectiveness in cellular removal while preserving the extracellular matrix. The intensity of DNA staining was taken as an indicator of remaining cells and compared to untreated sections. The presence of collagen, elastin and laminin were quantified using immunostaining and signal quantification. Scaffolds resulting from the optimized protocol were mechanically characterized using atomic force microscopy. Lung scaffolds were recellularized with mesenchymal stromal cells to assess their biocompatibility. Some decellularization agents (CHAPS, triton, and ammonia hydroxide) did not achieve sufficient cell removal. Sodium dodecyl sulfate (SDS) was effective in cell removal (1% remaining DNA signal), but its sharp reduction of elastin signal (only 6% remained) plus lower attachment ratio (32%) singled out sodium deoxycholate (SD) as the optimal treatment for this application (6.5% remaining DNA signal), due to its higher elastin retention (34%) and higher attachment ratio (60%). Laminin and collagen were fully preserved in all treatments. The SD decellularization protocol was also successful for porcine and murine (mice and rat) lungs as well as for other tissues such as the heart, kidney, and bladder. No significant mechanical differences were found before and after sample decellularization. The resulting acellular lung scaffolds were shown to be biocompatible (98% cell survival after 72 h of culture). This novel method to decellularize tissue slices opens up new methodological possibilities to better understand the role of the extracellular matrix in the context of several diseases as well as tissue engineering research and can be easily adapted for scarce samples like clinical biopsies.
Collapse
Affiliation(s)
- Maria Narciso
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
- The Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Anna Ulldemolins
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
| | - Constança Júnior
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
- The Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Jorge Otero
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
- The Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Madrid, Spain
| | - Daniel Navajas
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
- The Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Madrid, Spain
| | - Ramon Farré
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Madrid, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
| | - Núria Gavara
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
- The Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Isaac Almendros
- Unitat de Biofísica i Bioenginyeria, Facultat de Medicina i Ciències de la Salut, Universitat de Barcelona, Barcelona, Spain
- CIBER de Enfermedades Respiratorias, Madrid, Spain
- Institut d’Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- *Correspondence: Isaac Almendros,
| |
Collapse
|
119
|
Miyake K, Miyagawa S, Harada A, Sawa Y. Engineered clustered myoblast cell injection augments angiogenesis and muscle regeneration in peripheral artery disease. Mol Ther 2022; 30:1239-1251. [PMID: 35007760 PMCID: PMC8899600 DOI: 10.1016/j.ymthe.2022.01.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 11/27/2021] [Accepted: 01/05/2022] [Indexed: 11/18/2022] Open
Abstract
The low survival rate of administered cells due to ischemic and inflammatory environments limits the efficacy of the current regenerative cell therapy in peripheral artery disease (PAD). This study aimed to develop a new method to enhance the efficacy of cell therapy in PAD using cell sheet technology. Clustered cells (CCs) from myoblast cell sheets obtained from C57/BL6 mice were administered into ischemic mouse muscles 7 days after induction of ischemia (defined as day 0). Control groups were administered with single myoblast cells (SCs) or saline. Cell survival, blood perfusion of the limb, angiogenesis, muscle regeneration, and inflammation status were evaluated. The survival of administered cells was markedly improved in CCs compared with SCs at days 7 and 28. CCs showed significantly improved blood perfusion, augmented angiogenesis with increased density of CD31+/α-smooth muscle actin+ arterioles, and accelerated muscle regeneration, along with the upregulation of associated genes. Additionally, inflammation status was well regulated by CCs administration. CCs administration increased the number of macrophages and then induced polarization into an anti-inflammatory phenotype (CD11c-/CD206+), along with the increased expression of genes associated with anti-inflammatory cytokines. Our findings suggest clinical potential of rescuing severely damaged limbs in PAD using CCs.
Collapse
Affiliation(s)
- Keisuke Miyake
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Akima Harada
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshiki Sawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan.
| |
Collapse
|
120
|
Samandari M, Quint J, Rodríguez-delaRosa A, Sinha I, Pourquié O, Tamayol A. Bioinks and Bioprinting Strategies for Skeletal Muscle Tissue Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2105883. [PMID: 34773667 PMCID: PMC8957559 DOI: 10.1002/adma.202105883] [Citation(s) in RCA: 60] [Impact Index Per Article: 20.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 10/28/2021] [Indexed: 05/16/2023]
Abstract
Skeletal muscles play important roles in critical body functions and their injury or disease can lead to limitation of mobility and loss of independence. Current treatments result in variable functional recovery, while reconstructive surgery, as the gold-standard approach, is limited due to donor shortage, donor-site morbidity, and limited functional recovery. Skeletal muscle tissue engineering (SMTE) has generated enthusiasm as an alternative solution for treatment of injured tissue and serves as a functional disease model. Recently, bioprinting has emerged as a promising tool for recapitulating the complex and highly organized architecture of skeletal muscles at clinically relevant sizes. Here, skeletal muscle physiology, muscle regeneration following injury, and current treatments following muscle loss are discussed, and then bioprinting strategies implemented for SMTE are critically reviewed. Subsequently, recent advancements that have led to improvement of bioprinting strategies to construct large muscle structures, boost myogenesis in vitro and in vivo, and enhance tissue integration are discussed. Bioinks for muscle bioprinting, as an essential part of any bioprinting strategy, are discussed, and their benefits, limitations, and areas to be improved are highlighted. Finally, the directions the field should expand to make bioprinting strategies more translational and overcome the clinical unmet needs are discussed.
Collapse
Affiliation(s)
- Mohamadmahdi Samandari
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
| | - Jacob Quint
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
| | | | - Indranil Sinha
- Department of Surgery, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02139, USA
| | - Olivier Pourquié
- Department of Genetics, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Ali Tamayol
- Department of Biomedical Engineering, University of Connecticut Health Center, Farmington, CT 06030, USA
| |
Collapse
|
121
|
Yari D, Ebrahimzadeh MH, Movaffagh J, Shahroodi A, Shirzad M, Qujeq D, Moradi A. Biochemical Aspects of Scaffolds for Cartilage Tissue Engineering; from Basic Science to Regenerative Medicine. THE ARCHIVES OF BONE AND JOINT SURGERY 2022; 10:229-244. [PMID: 35514762 PMCID: PMC9034797 DOI: 10.22038/abjs.2022.55549.2766] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Received: 02/18/2021] [Accepted: 01/19/2022] [Indexed: 12/14/2022]
Abstract
Chondral defects are frequent and important causes of pain and disability. Cartilage has limited self-repair and regeneration capacity. The ideal approach for articular cartilage defects is the regeneration of hyaline cartilage with sustainable symptom-free constructs. Tissue engineering provides new strategies for the regeneration of functional cartilage tissue through optimized scaffolds with architectural, mechanical, and biochemical properties similar to the native cartilage tissue. In this review, the basic science of cartilage structure, interactions between proteins, stem cells, as well as biomaterials, scaffold characteristics and fabrication methods, as well as current and potential therapies in regenerative medicine will be discussed mostly from a biochemical point of view. Furthermore, the recent trends in scaffold-based therapies and supplementary factors in cartilage tissue engineering will be considered.
Collapse
Affiliation(s)
- Davood Yari
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran,Department of Clinical Biochemistry, Babol University of Medical Sciences, Babol, Iran,Orthopedic Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | | | - Jebrail Movaffagh
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Azadeh Shahroodi
- Department of Pharmaceutics, School of Pharmacy, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Moein Shirzad
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran,Department of Clinical Biochemistry, Babol University of Medical Sciences, Babol, Iran
| | - Durdi Qujeq
- Cellular and Molecular Biology Research Center, Health Research Institute, Babol University of Medical Sciences, Babol, Iran,Department of Clinical Biochemistry, Babol University of Medical Sciences, Babol, Iran
| | - Ali Moradi
- Orthopedic Research Center, Mashhad University of Medical Sciences, Mashhad, Iran,Clinical Research Development Unit, Ghaem Hospital, Mashhad University of Medical Sciences, Mashhad, Iran
| |
Collapse
|
122
|
Wang S, Yang L, Cai B, Liu F, Hou Y, Zheng H, Cheng F, Zhang H, Wang L, Wang X, Lv Q, Kong L, Lee KB, Zhang Q. Injectable hybrid inorganic nanoscaffold-templated rapid stem cell assembly for cartilage repair. Natl Sci Rev 2022; 9:nwac037. [PMID: 35419207 PMCID: PMC8998491 DOI: 10.1093/nsr/nwac037] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2021] [Revised: 01/13/2022] [Accepted: 01/13/2022] [Indexed: 11/14/2022] Open
Abstract
ABSTRACT
Cartilage injuries are often devastating and most cannot be cured because of the intrinsically low regenerative capacity of cartilage tissues. Although stem cell therapy has shown enormous potential for cartilage repair, the therapeutic outcome has been restricted by low survival rates and poor chondrocyte differentiation in vivo. Here, we report an injectable hybrid inorganic (IHI) nanoscaffold that facilitates fast assembly, enhances survival, and regulates chondrogenic differentiation of stem cells. IHI nanoscaffolds that strongly bind to extracellular matrix (ECM) proteins assemble stem cells through synergistic three-dimensional (3D) cell-cell and cell-matrix interactions, creating a favourable physical microenvironment for stem cell survival and differentiation in vitro and in vivo. Additionally, chondrogenic factors can be loaded into nanoscaffolds with a high capacity, which allows deep, homogenous drug delivery into assembled 3D stem cell-derived tissues for effective control over the soluble microenvironment of stem cells. The developed IHI nanoscaffolds that assemble with stem cells are injectable. They also scavenge reactive oxygen species and timely biodegrade for proper integration into injured cartilage tissues. Implantation of stem cell-assembled IHI nanoscaffolds into injured cartilage results in accelerated tissue regeneration and functional recovery. By establishing our IHI nanoscaffold-templated 3D stem cell assembly method, we provide a promising approach to better overcome the inhibitory microenvironment associated with cartilage injuries and to advance current stem cell-based tissue engineering.
Collapse
Affiliation(s)
- Shenqiang Wang
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi’an 710129, China
- Xi’an Key Laboratory of Functional Organic Porous Materials, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi’an 710072, China
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA
| | - Letao Yang
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA
- Department of Biomedical Engineering, Columbia University, New York, NY 10032, USA
| | - Bolei Cai
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
| | - Fuwei Liu
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
| | - Yannan Hou
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA
| | - Hua Zheng
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi’an 710129, China
| | - Fang Cheng
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi’an 710129, China
| | - Hepeng Zhang
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi’an 710129, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China
- Xi’an Key Laboratory of Functional Organic Porous Materials, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi’an 710072, China
| | - Le Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
| | - Xiaoyi Wang
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
| | - Qianxin Lv
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
| | - Liang Kong
- State Key Laboratory of Military Stomatology & National Clinical Research Center for Oral Diseases & Shaanxi Key Laboratory of Oral Diseases, Department of Oral and Maxillofacial Surgery, School of Stomatology, The Fourth Military Medical University, Xi’an 710032, China
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ 08854, USA
| | - Qiuyu Zhang
- Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi’an 710129, China
- Research & Development Institute of Northwestern Polytechnical University in Shenzhen, Shenzhen 518057, China
- Xi’an Key Laboratory of Functional Organic Porous Materials, School of Chemistry and Chemical Engineering, Northwestern Polytechnical University, Xi’an 710072, China
| |
Collapse
|
123
|
Loreti M, Sacco A. The jam session between muscle stem cells and the extracellular matrix in the tissue microenvironment. NPJ Regen Med 2022; 7:16. [PMID: 35177651 PMCID: PMC8854427 DOI: 10.1038/s41536-022-00204-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 12/14/2021] [Indexed: 12/21/2022] Open
Abstract
Skeletal muscle requires a highly orchestrated coordination between multiple cell types and their microenvironment to exert its function and to maintain its homeostasis and regenerative capacity. Over the past decades, significant advances, including lineage tracing and single-cell RNA sequencing, have contributed to identifying multiple muscle resident cell populations participating in muscle maintenance and repair. Among these populations, muscle stem cells (MuSC), also known as satellite cells, in response to stress or injury, are able to proliferate, fuse, and form new myofibers to repair the damaged tissue. These cells reside adjacent to the myofiber and are surrounded by a specific and complex microenvironment, the stem cell niche. Major components of the niche are extracellular matrix (ECM) proteins, able to instruct MuSC behavior. However, during aging and muscle-associated diseases, muscle progressively loses its regenerative ability, in part due to a dysregulation of ECM components. This review provides an overview of the composition and importance of the MuSC microenvironment. We discuss relevant ECM proteins and how their mutations or dysregulation impact young and aged muscle tissue or contribute to diseases. Recent discoveries have improved our knowledge about the ECM composition of skeletal muscle, which has helped to mimic the architecture of the stem cell niche and improved the regenerative capacity of MuSC. Further understanding about extrinsic signals from the microenvironment controlling MuSC function and innovative technologies are still required to develop new therapies to improve muscle repair.
Collapse
Affiliation(s)
- Mafalda Loreti
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901N Torrey Pines Rd, La Jolla, CA, 92037, USA
| | - Alessandra Sacco
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, 10901N Torrey Pines Rd, La Jolla, CA, 92037, USA.
| |
Collapse
|
124
|
Liu C, Pei M, Li Q, Zhang Y. Decellularized extracellular matrix mediates tissue construction and regeneration. Front Med 2022; 16:56-82. [PMID: 34962624 PMCID: PMC8976706 DOI: 10.1007/s11684-021-0900-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 09/23/2021] [Indexed: 02/05/2023]
Abstract
Contributing to organ formation and tissue regeneration, extracellular matrix (ECM) constituents provide tissue with three-dimensional (3D) structural integrity and cellular-function regulation. Containing the crucial traits of the cellular microenvironment, ECM substitutes mediate cell-matrix interactions to prompt stem-cell proliferation and differentiation for 3D organoid construction in vitro or tissue regeneration in vivo. However, these ECMs are often applied generically and have yet to be extensively developed for specific cell types in 3D cultures. Cultured cells also produce rich ECM, particularly stromal cells. Cellular ECM improves 3D culture development in vitro and tissue remodeling during wound healing after implantation into the host as well. Gaining better insight into ECM derived from either tissue or cells that regulate 3D tissue reconstruction or organ regeneration helps us to select, produce, and implant the most suitable ECM and thus promote 3D organoid culture and tissue remodeling for in vivo regeneration. Overall, the decellularization methodologies and tissue/cell-derived ECM as scaffolds or cellular-growth supplements used in cell propagation and differentiation for 3D tissue culture in vitro are discussed. Moreover, current preclinical applications by which ECM components modulate the wound-healing process are reviewed.
Collapse
Affiliation(s)
- Chuanqi Liu
- Department of Plastic and Burn Surgery, West China Hospital, Sichuan University, Chengdu, 610041, China
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China
| | - Ming Pei
- Stem Cell and Tissue Engineering Laboratory, Department of Orthopaedics, West Virginia University, Morgantown, WV, 26506, USA
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200011, China.
| | - Yuanyuan Zhang
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, 27109, USA.
| |
Collapse
|
125
|
Yi X, Liu F, Gao K, Chen F, Wang Y, Li H, Wang X, Huang Y, Fu H, Zhou W, Fan JB, Wang S, Gao Y. Reconstructable Uterus-Derived Materials for Uterus Recovery toward Efficient Live Births. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2106510. [PMID: 34854148 DOI: 10.1002/adma.202106510] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/18/2021] [Revised: 11/17/2021] [Indexed: 06/13/2023]
Abstract
Uterine factor infertility is increasingly common in modern society and has severely affected human life and health. However, the existing biomaterial scaffold-mediated systems remain limited in efficient uterus recovery, leading to low pregnancy rate and live births. Here, reconstructable uterus-derived materials (RUMs) are demonstrated by combining uterus-derived extracellular matrix and seeded chorionic villi mesenchymal stem cells for uterus recovery, achieving highly efficient live births in rats with severe uterine injury. The RUMs can be designed into different states (such as, liquid RUMs and solid RUMs) and shapes (such as, cuboid, triangular-prism, and cube) in terms of requirements. The RUMs can effectively prevent intrauterine adhesion, and promote endometrial regeneration and muscle collagen reconstruction, as well as, accelerate wound healing by constructing a physical barrier and secreting cytokines, allowing efficient uterus recovery. The injured uterus nearly achieves complete recovery after treating with the RUMs and has normal pregnancies for supporting fetal development and live births, similar to the normal rats. The study provides a regenerative medicine therapeutics for uterine factor infertility.
Collapse
Affiliation(s)
- Xiao Yi
- Research Centre for Artificial Organ and Tissue Engineering & Institute of Regenerative Medicine, Department of Hepatobiliary Surgery II, Zhujiang Hospital, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, 510280, P. R. China
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, P. R. China
| | - Fan Liu
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, P. R. China
| | - Kunjie Gao
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, P. R. China
| | - Feng Chen
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, P. R. China
| | - Yifeng Wang
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, P. R. China
| | - Huayan Li
- Department of Gynecology, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong Province, P. R. China
| | - Xuefeng Wang
- Department of Gynecology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Yi Huang
- Department of Gynecology, Nanhai People's Hospital, Southern Medical University, Guangzhou, Guangdong Province, P. R. China
| | - Huijiao Fu
- Department of Gynecology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, Guangdong, P. R. China
| | - Weijie Zhou
- Department of Pathology, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong, 510515, P. R. China
| | - Jun-Bing Fan
- Research Centre for Artificial Organ and Tissue Engineering & Institute of Regenerative Medicine, Department of Hepatobiliary Surgery II, Zhujiang Hospital, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, 510280, P. R. China
| | - Shutao Wang
- CAS Key Laboratory of Bio-inspired Materials and Interfacial Science, CAS Center for Excellence in Nanoscience, Technical Institute of Physics and Chemistry, Chinese Academy of Sciences, Beijing, 100190, P. R. China
| | - Yi Gao
- Research Centre for Artificial Organ and Tissue Engineering & Institute of Regenerative Medicine, Department of Hepatobiliary Surgery II, Zhujiang Hospital, Cancer Research Institute, School of Basic Medical Sciences, Southern Medical University, Guangzhou, Guangdong Province, 510280, P. R. China
| |
Collapse
|
126
|
Ye P, Fu Z, Chung JYF, Cao X, Ko H, Tian XY, Tang PMK, Lui KO. Endothelial Agrin Is Dispensable for Normal and Tumor Angiogenesis. Front Cardiovasc Med 2022; 8:810477. [PMID: 35174224 PMCID: PMC8841877 DOI: 10.3389/fcvm.2021.810477] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Accepted: 12/30/2021] [Indexed: 11/13/2022] Open
Abstract
Recently, the extracellular matrix protein agrin has been reported to promote tumor angiogenesis that supports tumorigenesis and metastasis; however, there is a lack of in vivo genetic evidence to prove whether agrin derived from the tumors or endothelial cells (ECs) systemically should be the therapeutic target. To date, the physiological role of endothelial agrin has also not been investigated. In the EC-specific agrin knockout mice, we observed normal endothelial and haematopoietic cell development during embryogenesis. Moreover, these mice develop normal vascular barrier integrity and vasoreactivity at the adult stage. Importantly, the growth of localized or metastatic cancer cells was not affected after implantation into endothelial agrin depleted mice. Mechanistically, agrin did not regulate endothelial ERK1/2, YAP or p53 activation in vivo that is central to support endothelial proliferation, survival and invasion. Cumulatively, our findings may suggest that agrin could play a redundant role in endothelial development during physiological and tumor angiogenesis. Targeting the endothelial derived agrin might not be effective in inhibiting tumor angiogenesis.
Collapse
Affiliation(s)
- Peng Ye
- Department of Chemical Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Zelong Fu
- Department of Chemical Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Jeff Yat-Fai Chung
- State Key Laboratory of Translational Oncology, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiaoyun Cao
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Ho Ko
- Department of Medicine and Therapeutics, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Xiao Yu Tian
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Patrick Ming-Kuen Tang
- State Key Laboratory of Translational Oncology, Department of Anatomical and Cellular Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
| | - Kathy O. Lui
- Department of Chemical Pathology, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- Li Ka Shing Institute of Health Sciences, Prince of Wales Hospital, The Chinese University of Hong Kong, Hong Kong, China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, China
- *Correspondence: Kathy O. Lui
| |
Collapse
|
127
|
Wang X, Zhao R, Wang J, Li X, Jin L, Liu W, Yang L, Zhu Y, Tan Z. 3D-printed tissue repair patch combining mechanical support and magnetism for controlled skeletal muscle regeneration. Biodes Manuf 2022. [DOI: 10.1007/s42242-021-00180-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
128
|
Redenski I, Guo S, Machour M, Szklanny A, Landau S, Egozi D, Gabet Y, Levenberg S. Microcomputed Tomography-Based Analysis of Neovascularization within Bioengineered Vascularized Tissues. ACS Biomater Sci Eng 2022; 8:232-241. [PMID: 34905338 DOI: 10.1021/acsbiomaterials.1c01401] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
In the field of tissue engineering, evaluating newly formed vascular networks is considered a fundamental step in deciphering the processes underlying tissue development. Several common modalities exist to study vessel network formation and function. However, a proper methodology that allows through three-dimensional visualization of neovessels in a reproducible manner is required. Here, we describe in-depth exploration, visualization, and analysis of vessels within newly formed tissues by utilizing a contrast agent perfusion protocol and high-resolution microcomputed tomography. Bioengineered constructs consisting of porous, biocompatible, and biodegradable scaffolds are loaded with cocultures of adipose-derived microvascular endothelial cells (HAMECs) and dental pulp stem cells (DPSCs) and implanted in a rat femoral bundle model. After 14 days of in vivo maturation, we performed the optimized perfusion protocol to allow host penetrating vascular visualization and assessment within neotissues. Following high-resolution microCT scanning of DPSC:HAMEC explants, we performed the volumetric and spatial analysis of neovasculature. Eventually, the process was repeated with a previously published coculture system for prevascularization based on adipose-derived mesenchymal stromal cells (MSCs) and HAMECs. Overall, our approach allows a comprehensive understanding of vessel organization during engraftment and development of neotissues.
Collapse
Affiliation(s)
- Idan Redenski
- Department of Biomedical Engineering, Technion─Israel Institute of Technology, Haifa 3200003, Israel
| | - Shaowei Guo
- Department of Biomedical Engineering, Technion─Israel Institute of Technology, Haifa 3200003, Israel
- The First Affiliated Hospital, Shantou University Medical College, Shantou 515000, China
| | - Majd Machour
- Department of Biomedical Engineering, Technion─Israel Institute of Technology, Haifa 3200003, Israel
| | - Ariel Szklanny
- Department of Biomedical Engineering, Technion─Israel Institute of Technology, Haifa 3200003, Israel
| | - Shira Landau
- Department of Biomedical Engineering, Technion─Israel Institute of Technology, Haifa 3200003, Israel
| | - Dana Egozi
- Department of Plastic and Reconstructive Surgery, Kaplan Hospital, Rehovot and the Hebrew University, Jerusalem 9190401, Israel
| | - Yankel Gabet
- Department of Anatomy and Anthropology, Sackler Faculty of Medicine, Tel-Aviv University, Tel-Aviv 6997801, Israel
| | - Shulamit Levenberg
- Department of Biomedical Engineering, Technion─Israel Institute of Technology, Haifa 3200003, Israel
| |
Collapse
|
129
|
Liang W, Li Y, Cuellar-Camacho JL, Yu L, Zhou S, Li W, Haag R. Chemically defined stem cell microniche engineering by microfluidics compatible with iPSCs' growth in 3D culture. Biomaterials 2022; 280:121253. [PMID: 34801253 DOI: 10.1016/j.biomaterials.2021.121253] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 09/13/2021] [Accepted: 11/10/2021] [Indexed: 11/02/2022]
Abstract
The development of induced pluripotent stem cell (iPSCs) has opened unprecedented opportunities for biomedical applications, but poorly defined animal-derived matrices yield cells with limited therapeutic value. Considerable challenges remain in improving cell-culturing approaches to create the conditions for iPSCs' reliable expansion. Herein we report the development of a chemically defined, artificial three-dimensional (3D) microniche for iPSCs' growth and reliable expansion, constructed with degradable polyethyleneglycol-co-polycaprolactone and RGDfk-functionalized dendritic polyglycerol precursors according to bioorthogonal strain-promoted azide-alkyne cycloaddition by droplet-based microfluidics. This compatible microniche can allow for the robust production of iPSCs that maintain high pluripotency expression and excellent viability without pathogen or immunogen transfer risks. This microniche technology shows great promise in enabling iPSCs to achieve their full therapeutic potential.
Collapse
Affiliation(s)
- Wanjun Liang
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 3, 14195, Berlin, Germany
| | - Yan Li
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 3, 14195, Berlin, Germany
| | - Jose Luis Cuellar-Camacho
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 3, 14195, Berlin, Germany
| | - Leixiao Yu
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 3, 14195, Berlin, Germany
| | - Suqiong Zhou
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 3, 14195, Berlin, Germany
| | - Wenzhong Li
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 3, 14195, Berlin, Germany.
| | - Rainer Haag
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, Takustrasse 3, 14195, Berlin, Germany.
| |
Collapse
|
130
|
Piras CC, Kay AG, Genever PG, Fitremann J, Smith DK. Self-assembled gel tubes, filaments and 3D-printing with in situ metal nanoparticle formation and enhanced stem cell growth. Chem Sci 2022; 13:1972-1981. [PMID: 35308847 PMCID: PMC8848986 DOI: 10.1039/d1sc06062g] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2021] [Accepted: 01/16/2022] [Indexed: 12/18/2022] Open
Abstract
This paper reports simple strategies to fabricate self-assembled artificial tubular and filamentous systems from a low molecular weight gelator (LMWG). In the first strategy, tubular ‘core–shell’ gel structures based on the dibenzylidenesorbitol-based LMWG DBS-CONHNH2 were made in combination with the polymer gelator (PG) calcium alginate. In the second approach, gel filaments based on DBS-CONHNH2 alone were prepared by wet spinning at elevated concentrations using a ‘solvent-switch’ approach. The higher concentrations used in wet-spinning prevent the need for a supporting PG. Furthermore, this can be extended into a 3D-printing method, with the printed LMWG objects showing excellent stability for at least a week in water. The LMWG retains its unique ability for in situ precious metal reduction, yielding Au nanoparticles (AuNPs) within the tubes and filaments when they are exposed to AuCl3 solutions. Since the gel filaments have a higher loading of DBS-CONHNH2, they can be loaded with significantly more AuNPs. Cytotoxicity and viability studies on human mesenchymal stem cells show that the DBS-CONHNH2 and DBS-CONHNH2/alginate hybrid gels loaded with AuNPs are biocompatible, with the presence of AuNPs enhancing stem cell metabolism. Taken together, these results indicate that DBS-CONHNH2 can be shaped and 3D-printed, and has considerable potential for use in tissue engineering applications. Simple fabrication and 3D-printing methods are used to generate tubes and filaments from self-assembled gels, which can be loaded in situ with gold nanoparticles, with the resulting gels encouraging stem cell proliferation.![]()
Collapse
Affiliation(s)
- Carmen C. Piras
- Department of Chemistry, University of York, Heslington, York, YO10 5DD, UK
| | - Alasdair G. Kay
- Department of Biology, University of York, Heslington, York, YO10 5DD, UK
| | - Paul G. Genever
- Department of Biology, University of York, Heslington, York, YO10 5DD, UK
| | - Juliette Fitremann
- IMRCP, UMR 5623, CNRS, Université de Toulouse, 118 Route de Narbonne, F-31062 Toulouse, France
| | - David K. Smith
- Department of Chemistry, University of York, Heslington, York, YO10 5DD, UK
| |
Collapse
|
131
|
Md Fadilah NI, Mohd Abdul Kader Jailani MS, Badrul Hisham MAI, Sunthar Raj N, Shamsuddin SA, Ng MH, Fauzi MB, Maarof M. Cell secretomes for wound healing and tissue regeneration: Next generation acellular based tissue engineered products. J Tissue Eng 2022; 13:20417314221114273. [PMID: 35923177 PMCID: PMC9340325 DOI: 10.1177/20417314221114273] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 07/01/2022] [Indexed: 12/20/2022] Open
Abstract
Wound represents a significant socioeconomic burden for both affected individuals and as a whole healthcare system. Accordingly, stem cells have garnered attention due to their differentiation capacity and ability to aid tissue regeneration by releasing biologically active molecules, found in the cells' cultivated medium which known as conditioned medium (CM) or secretomes. This acellular approach provides a huge advantage over conventional treatment options, which are mainly used cellular treatment at wound closure. Interestingly, the secretomes contained the cell-secreted proteins such as growth factors, cytokines, chemokines, extracellular matrix (ECM), and small molecules including metabolites, microvesicles, and exosomes. This review aims to provide a general view on secretomes and how it is proven to have great potential in accelerating wound healing. Utilizing the use of secretomes with its secreted proteins and suitable biomaterials for fabrications of acellular skin substitutes can be promising in treating skin loss and accelerate the healing process.
Collapse
Affiliation(s)
- Nur Izzah Md Fadilah
- Centre for Tissue Engineering and Regenerative
Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur,
Malaysia
| | | | - Muhd Aliff Iqmal Badrul Hisham
- Centre for Tissue Engineering and Regenerative
Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur,
Malaysia
| | - Nithiaraj Sunthar Raj
- Centre for Tissue Engineering and Regenerative
Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur,
Malaysia
| | - Sharen Aini Shamsuddin
- Centre for Tissue Engineering and Regenerative
Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur,
Malaysia
| | - Min Hwei Ng
- Centre for Tissue Engineering and Regenerative
Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur,
Malaysia
| | - Mh Busra Fauzi
- Centre for Tissue Engineering and Regenerative
Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur,
Malaysia
| | - Manira Maarof
- Centre for Tissue Engineering and Regenerative
Medicine, Faculty of Medicine, Universiti Kebangsaan Malaysia, Kuala Lumpur,
Malaysia
| |
Collapse
|
132
|
Chai Y, Zhao H, Yang S, Gao X, Cao Z, Lu J, Sun Q, Liu W, Zhang Z, Yang J, Wang X, Chen T, Kong X, Mikos AG, Zhang X, Zhang Y, Wang X. Structural alignment guides oriented migration and differentiation of endogenous neural stem cells for neurogenesis in brain injury treatment. Biomaterials 2021; 280:121310. [PMID: 34890972 DOI: 10.1016/j.biomaterials.2021.121310] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 11/23/2021] [Accepted: 12/02/2021] [Indexed: 02/04/2023]
Abstract
Radial glia (RG) cells that align in parallel in the embryonic brain are found to be able to guide the directed migration of neurons in response to brain injury. Therefore, biomaterials with aligned architectures are supposed to have positive effects on neural migration and neurogenic differentiation for brain injury repair that are rarely addressed, although they have been widely demonstrated in spinal cord and peripheral nerve system. Here, we present a highly biomimetic scaffold of aligned fibrin hydrogel (AFG) that mimics the oriented structure of RG fibers. Through a combination of histological, behavioral, imaging, and transcriptomic analyses, we demonstrated that transplanting the AFG scaffold into injured cortical brains promotes effective migration, differentiation, and maturation of endogenous neural stem cells, resulting in neurological functional recovery. Therefore, this study will light up a new perspective on applying an aligned scaffold to promote cortical regeneration after injury by inducing endogenous neurogenesis.
Collapse
Affiliation(s)
- Yi Chai
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China; Department of Neurosurgery, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 100040, China; Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, No. 160, Pujian Road, District Pudong, Shanghai, 200127, China
| | - He Zhao
- Department of orthopacdic III, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Shuhui Yang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
| | - Xiaohan Gao
- Department of Neurosurgery, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 100040, China
| | - Zheng Cao
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
| | - Jiaju Lu
- School of Materials Science and Engineering, Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Qingling Sun
- Department of orthopacdic III, Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, 100700, China
| | - Wei Liu
- Department of Neurosurgery, The First Hospital of Hebei Medical University, Hebei, 050000, China
| | - Zhe Zhang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
| | - Junyi Yang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
| | - Xuelin Wang
- School of Medical Science and Engineering, Beihang University, Beijing, 100191, China
| | - Tuoyu Chen
- Department of Neurosurgery, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 100040, China
| | - Xiangdong Kong
- School of Materials Science and Engineering, Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Antonios G Mikos
- Department of Bioengineering, Rice University, Houston, TX, 77030, USA
| | - Xiaohua Zhang
- Department of Neurosurgery, Renji Hospital, School of Medicine, Shanghai Jiaotong University, No. 160, Pujian Road, District Pudong, Shanghai, 200127, China
| | - Yuqi Zhang
- Department of Neurosurgery, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing, 100040, China.
| | - Xiumei Wang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials of Ministry of Education, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
133
|
Sun Q, Shen Z, Liang X, He Y, Kong D, Midgley AC, Wang K. Progress and Current Limitations of Materials for Artificial Bile Duct Engineering. MATERIALS 2021; 14:ma14237468. [PMID: 34885623 PMCID: PMC8658964 DOI: 10.3390/ma14237468] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/19/2021] [Accepted: 10/25/2021] [Indexed: 01/30/2023]
Abstract
Bile duct injury (BDI) and bile tract diseases are regarded as prominent challenges in hepatobiliary surgery due to the risk of severe complications. Hepatobiliary, pancreatic, and gastrointestinal surgery can inadvertently cause iatrogenic BDI. The commonly utilized clinical treatment of BDI is biliary-enteric anastomosis. However, removal of the Oddi sphincter, which serves as a valve control over the unidirectional flow of bile to the intestine, can result in complications such as reflux cholangitis, restenosis of the bile duct, and cholangiocarcinoma. Tissue engineering and biomaterials offer alternative approaches for BDI treatment. Reconstruction of mechanically functional and biomimetic structures to replace bile ducts aims to promote the ingrowth of bile duct cells and realize tissue regeneration of bile ducts. Current research on artificial bile ducts has remained within preclinical animal model experiments. As more research shows artificial bile duct replacements achieving effective mechanical and functional prevention of biliary peritonitis caused by bile leakage or obstructive jaundice after bile duct reconstruction, clinical translation of tissue-engineered bile ducts has become a theoretical possibility. This literature review provides a comprehensive collection of published works in relation to three tissue engineering approaches for biomimetic bile duct construction: mechanical support from scaffold materials, cell seeding methods, and the incorporation of biologically active factors to identify the advancements and current limitations of materials and methods for the development of effective artificial bile ducts that promote tissue regeneration.
Collapse
Affiliation(s)
- Qiqi Sun
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China; (Q.S.); (D.K.)
| | - Zefeng Shen
- Department of General Surgery, Sir Run-Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China; (Z.S.); (X.L.)
| | - Xiao Liang
- Department of General Surgery, Sir Run-Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou 310016, China; (Z.S.); (X.L.)
| | - Yingxu He
- School of Computing, National University of Singapore, Singapore 119077, Singapore;
| | - Deling Kong
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China; (Q.S.); (D.K.)
| | - Adam C. Midgley
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China; (Q.S.); (D.K.)
- Correspondence: (A.C.M.); (K.W.)
| | - Kai Wang
- Key Laboratory of Bioactive Materials for the Ministry of Education, College of Life Sciences, Nankai University, Tianjin 300071, China; (Q.S.); (D.K.)
- Correspondence: (A.C.M.); (K.W.)
| |
Collapse
|
134
|
Development of 3D culture scaffolds for directional neuronal growth using 2-photon lithography. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 131:112502. [PMID: 34857288 DOI: 10.1016/j.msec.2021.112502] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 10/13/2021] [Accepted: 10/16/2021] [Indexed: 01/02/2023]
Abstract
Conventional applications of transplant technology, applied to severe traumatic injuries of the nervous system, have met limited success in the clinics due to the complexity of restoring function to the damaged tissue. Neural tissue engineering aims to deploy scaffolds mimicking the physiological properties of the extracellular matrix to facilitate the elongation of axons and the repair of damaged nerves. However, the fabrication of ideal scaffolds with precisely controlled thickness, texture, porosity, alignment, and with the required mechanical strength, features needed for effective clinical applications, remains technically challenging. We took advantage of state-of-the-art 2-photon photolithography to fabricate highly ordered and biocompatible 3D nanogrid structures to enhance neuronal directional growth. First, we characterized the physical and chemical properties and proved the biocompatibility of said scaffolds by successfully culturing primary sensory and motor neurons on their surface. Interestingly, axons extended along the fibers with a high degree of alignment to the pattern of the nanogrid, as opposed to the lack of directionality observed on flat glass or polymeric surfaces, and could grow in 3D between different layers of the scaffold. The axonal growth pattern observed is highly desirable for the treatment of traumatic nerve damage occurring during peripheral and spinal cord injuries. Thus, our findings provide a proof of concept and explore the possibility of deploying aligned fibrous 3D scaffold/implants for the directed growth of axons, and could be used in the design of scaffolds targeted towards the restoration and repair of lost neuronal connections.
Collapse
|
135
|
Yoo YI, Ko KW, Cha SG, Park SY, Woo J, Han DK. Highly effective induction of cell-derived extracellular matrix by macromolecular crowding for osteogenic differentiation of mesenchymal stem cells. J IND ENG CHEM 2021. [DOI: 10.1016/j.jiec.2021.12.008] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
|
136
|
Kasper M, Ellenbogen B, Hardy R, Cydis M, Mojica-Santiago J, Afridi A, Spearman BS, Singh I, Kuliasha CA, Atkinson E, Otto KJ, Judy JW, Rinaldi-Ramos C, Schmidt CE. Development of a magnetically aligned regenerative tissue-engineered electronic nerve interface for peripheral nerve applications. Biomaterials 2021; 279:121212. [PMID: 34717196 PMCID: PMC9036633 DOI: 10.1016/j.biomaterials.2021.121212] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2021] [Revised: 10/11/2021] [Accepted: 10/20/2021] [Indexed: 01/18/2023]
Abstract
Peripheral nerve injuries can be debilitating to motor and sensory function, with severe cases often resulting in complete limb amputation. Over the past two decades, prosthetic limb technology has rapidly advanced to provide users with crude motor control of up to 20° of freedom; however, the nerve-interfacing technology required to provide high movement selectivity has not progressed at the same rate. The work presented here focuses on the development of a magnetically aligned regenerative tissue-engineered electronic nerve interface (MARTEENI) that combines polyimide "threads" encapsulated within a magnetically aligned hydrogel scaffold. The technology exploits tissue-engineered strategies to address concerns over traditional peripheral nerve interfaces including poor axonal sampling through the nerve and rigid substrates. A magnetically templated hydrogel is used to physically support the polyimide threads while also promoting regeneration in close proximity to the electrode sites on the polyimide. This work demonstrates the utility of magnetic templating for use in tuning the mechanical properties of hydrogel scaffolds to match the stiffness of native nerve tissue while providing an aligned substrate for Schwann cell migration in vitro. MARTEENI devices were fabricated and implanted within a 5-mm-long rat sciatic-nerve transection model to assess regeneration at 6 and 12 weeks. MARTEENI devices do not disrupt tissue remodeling and show axon densities equivalent to fresh tissue controls around the polyimide substrates. Devices are observed to have attenuated foreign-body responses around the polyimide threads. It is expected that future studies with functional MARTEENI devices will be able to record and stimulate single axons with high selectivity and low stimulation regimes.
Collapse
Affiliation(s)
- Mary Kasper
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Dr. JG56, P.O. Box 116131, Gainesville, FL, 32611, USA
| | - Bret Ellenbogen
- Department of Chemistry, University of Florida, P.O. Box 117200, Gainesville, FL, 32611, USA
| | - Ryan Hardy
- Department of Materials Science and Engineering, University of Florida, 549 Gale Lemerand Dr., P.O. Box 116400, Gainesville, FL, 32611, USA
| | - Madison Cydis
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Dr. JG56, P.O. Box 116131, Gainesville, FL, 32611, USA
| | - Jorge Mojica-Santiago
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Dr. JG56, P.O. Box 116131, Gainesville, FL, 32611, USA
| | - Abdullah Afridi
- Department of Chemistry, University of Florida, P.O. Box 117200, Gainesville, FL, 32611, USA
| | - Benjamin S Spearman
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Dr. JG56, P.O. Box 116131, Gainesville, FL, 32611, USA
| | - Ishita Singh
- Department of Chemical Engineering, University of Florida, 1030 Center Dr., P.O. Box 116005, Gainesville, FL, 32611, USA
| | - Cary A Kuliasha
- Department of Electrical and Computer Engineering, University of Florida, 968 Center Dr., Gainesville, FL, 32611, USA
| | - Eric Atkinson
- Department of Neuroscience, 1149 Newell Dr. L1-100, P.O. Box 100244, University of Florida, Gainesville, FL, 32610, USA
| | - Kevin J Otto
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Dr. JG56, P.O. Box 116131, Gainesville, FL, 32611, USA; Department of Materials Science and Engineering, University of Florida, 549 Gale Lemerand Dr., P.O. Box 116400, Gainesville, FL, 32611, USA; Department of Electrical and Computer Engineering, University of Florida, 968 Center Dr., Gainesville, FL, 32611, USA; Department of Neuroscience, 1149 Newell Dr. L1-100, P.O. Box 100244, University of Florida, Gainesville, FL, 32610, USA; Department of Neurology, 1149 Newell Dr. L3-100, P.O. Box 100236, University of Florida, Gainesville, FL, 32610, USA
| | - Jack W Judy
- Department of Electrical and Computer Engineering, University of Florida, 968 Center Dr., Gainesville, FL, 32611, USA
| | - Carlos Rinaldi-Ramos
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Dr. JG56, P.O. Box 116131, Gainesville, FL, 32611, USA; Department of Chemistry, University of Florida, P.O. Box 117200, Gainesville, FL, 32611, USA
| | - Christine E Schmidt
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Dr. JG56, P.O. Box 116131, Gainesville, FL, 32611, USA.
| |
Collapse
|
137
|
Geng G, Xiao Y, Shang Y, Zhang Y, Zhu F, Tang L, Peng F, Shen W, Jin Y, Yang Z, Li Q, Chen X. Naphthalenephenylalanine-phenylalanine-glycine-arginine-glycine-aspartic promotes self-assembly of nephron progenitor cells in decellularized scaffolds to construct bioengineered kidneys. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 134:112590. [DOI: 10.1016/j.msec.2021.112590] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2021] [Revised: 11/09/2021] [Accepted: 11/29/2021] [Indexed: 10/19/2022]
|
138
|
Ge J, Li Y, Wang M, Gao C, Yang S, Lei B. Engineering conductive antioxidative antibacterial nanocomposite hydrogel scaffolds with oriented channels promotes structure-functional skeletal muscle regeneration. CHEMICAL ENGINEERING JOURNAL 2021; 425:130333. [DOI: 10.1016/j.cej.2021.130333] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/27/2025]
|
139
|
Oliveira CS, Leeuwenburgh S, Mano JF. New insights into the biomimetic design and biomedical applications of bioengineered bone microenvironments. APL Bioeng 2021; 5:041507. [PMID: 34765857 PMCID: PMC8568480 DOI: 10.1063/5.0065152] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 10/06/2021] [Indexed: 12/31/2022] Open
Abstract
The bone microenvironment is characterized by an intricate interplay between cellular and noncellular components, which controls bone remodeling and repair. Its highly hierarchical architecture and dynamic composition provide a unique microenvironment as source of inspiration for the design of a wide variety of bone tissue engineering strategies. To overcome current limitations associated with the gold standard for the treatment of bone fractures and defects, bioengineered bone microenvironments have the potential to orchestrate the process of bone regeneration in a self-regulated manner. However, successful approaches require a strategic combination of osteogenic, vasculogenic, and immunomodulatory factors through a synergic coordination between bone cells, bone-forming factors, and biomaterials. Herein, we provide an overview of (i) current three-dimensional strategies that mimic the bone microenvironment and (ii) potential applications of bioengineered microenvironments. These strategies range from simple to highly complex, aiming to recreate the architecture and spatial organization of cell-cell, cell-matrix, and cell-soluble factor interactions resembling the in vivo microenvironment. While several bone microenvironment-mimicking strategies with biophysical and biochemical cues have been proposed, approaches that exploit the ability of the cells to self-organize into microenvironments with a high regenerative capacity should become a top priority in the design of strategies toward bone regeneration. These miniaturized bone platforms may recapitulate key characteristics of the bone regenerative process and hold great promise to provide new treatment concepts for the next generation of bone implants.
Collapse
Affiliation(s)
- Cláudia S. Oliveira
- Department of Chemistry, CICECO–Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| | - Sander Leeuwenburgh
- Department of Dentistry-Regenerative Biomaterials, Radboud University Medical Center, Radboud Institute for Molecular Life Sciences, Philips van Leydenlaan 25, 6525 EX Nijmegen, The Netherlands
| | - João F. Mano
- Department of Chemistry, CICECO–Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, 3810-193 Aveiro, Portugal
| |
Collapse
|
140
|
Wang S, Zhu C, Zhang B, Hu J, Xu J, Xue C, Bao S, Gu X, Ding F, Yang Y, Gu X, Gu Y. BMSC-derived extracellular matrix better optimizes the microenvironment to support nerve regeneration. Biomaterials 2021; 280:121251. [PMID: 34810037 DOI: 10.1016/j.biomaterials.2021.121251] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2021] [Revised: 11/02/2021] [Accepted: 11/08/2021] [Indexed: 12/26/2022]
Abstract
A favorable microenvironment plays an important role in nerve regeneration. Extracellular matrix (ECM) derived from cultured cells or natural tissues can facilitate nerve regeneration in the presence of various microenvironmental cues, including biochemical, spatial, and biomechanical factors. This study, through proteomics and three-dimensional image analysis, determines that the components and spatial organization of the ECM secreted by bone marrow mesenchymal cells (BMSCs) are more similar to acellular nerves than those of the ECMs derived from Schwann cells (SCs), skin-derived precursor Schwann cells (SKP-SCs), or fibroblasts (FBs). ECM-modified nerve grafts (ECM-NGs) are engineered by co-cultivating BMSCs, SCs, FBs, SKP-SCs with well-designed nerve grafts used to bridge nerve defects. BMSC-ECM-NGs exhibit the most promising nerve repair properties based on the histology, neurophysiology, and behavioral analyses. The regeneration microenvironment formed by the ECM-NGs is also characterized by proteomics, and the advantages of BMSC-ECM-NGs are evidenced by the enhanced expression of factors related to neural regeneration and reduced immune response. Together, these findings indicate that BMSC-derived ECMs create a more superior microenvironment for nerve regeneration than that by the other ECMs and may, therefore, represent a potential alternative for the clinical repair of peripheral nerve defects.
Collapse
Affiliation(s)
- Shengran Wang
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Changlai Zhu
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Bin Zhang
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Junxia Hu
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Jinghui Xu
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Chengbin Xue
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Shuangxi Bao
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Xiaokun Gu
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Fei Ding
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Yumin Yang
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China
| | - Xiaosong Gu
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China.
| | - Yun Gu
- Jiangsu Key Laboratory of Neuroregeneration, Jiangsu Clinical Medicine Center of Tissue Engineering and Nerve Injury Repair, Co-innovation Center of Neuroregeneration, NMPA Key Laboratory for Research and Evaluation of Tissue Engineering Technology Products, Nantong University, 19 Qixiu Road, Nantong, JS, 226001, PR China.
| |
Collapse
|
141
|
Jana S, Morse D, Lerman A. Leaflet Tissue Generation from Microfibrous Heart Valve Leaflet Scaffolds with Native Characteristics. ACS APPLIED BIO MATERIALS 2021; 4:7836-7847. [PMID: 35006765 DOI: 10.1021/acsabm.1c00768] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Mechanical and bioprosthetic valves that are currently applied for replacing diseased heart valves are not fully efficient. Heart valve tissue engineering may solve the issues faced by the prosthetic valves in heart valve replacement. The leaflets of native heart valves have a trilayered structure with layer-specific orientations; thus, it is imperative to develop functional leaflet tissue constructs with a native trilayered, oriented structure. Its key solution is to develop leaflet scaffolds with a native morphology and structure. In this study, microfibrous leaflet scaffolds with a native trilayered and oriented structure were developed in an electrospinning system. The scaffolds were implanted for 3 months in rats subcutaneously to study the scaffold efficiencies in generating functional tissue-engineered leaflet constructs. These in vivo tissue-engineered leaflet constructs had a trilayered, oriented structure similar to native leaflets. The tensile properties of constructs indicated that they were able to endure the hydrodynamic load of the native heart valve. Collagen, glycosaminoglycans, and elastin─the predominant extracellular matrix components of native leaflets─were found sufficiently in the leaflet tissue constructs. The residing cells in the leaflet tissue constructs showed vimentin and α-smooth muscle actin expression, i.e., the constructs were in a growing state. Thus, the trilayered, oriented fibrous leaflet scaffolds produced in this study could be useful to develop heart valve scaffolds for successful heart valve replacements.
Collapse
Affiliation(s)
- Soumen Jana
- Department of Bioengineering, University of Missouri, Columbia, Missouri 65211, United States.,Division of Cardiovascular Diseases, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905, United States
| | - David Morse
- Division of Cardiovascular Diseases, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905, United States
| | - Amir Lerman
- Division of Cardiovascular Diseases, Mayo Clinic, 200 First Street SW, Rochester, Minnesota 55905, United States
| |
Collapse
|
142
|
Shen J, Wang J, Liu X, Sun Y, Yin A, Chai Y, Zhang K, Wang C, Zheng X. In Situ Prevascularization Strategy with Three-Dimensional Porous Conduits for Neural Tissue Engineering. ACS APPLIED MATERIALS & INTERFACES 2021; 13:50785-50801. [PMID: 34664947 DOI: 10.1021/acsami.1c16138] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/13/2023]
Abstract
Neovascularization is crucial for peripheral nerve regeneration and long-term functional restoration. Previous studies have emphasized strategies that enhance axonal repair over vascularization. Here, we describe the development and application of an in situ prevascularization strategy that uses 3D porous nerve guidance conduits (NGCs) to achieve angiogenesis-mediated neural regeneration. The optimal porosity of the NGC is a critical feature for achieving neovascularization and nerve growth patency. Hollow silk fibroin/poly(l-lactic acid-co-ε-caprolactone) NGCs with 3D sponge-like walls were fabricated using electrospinning and freeze-drying. In vitro results showed that 3D porous NGC favored cell biocompatibility had neuroregeneration potential and, most importantly, had angiogenic activity. Results from our mechanistic studies suggest that activation of HIF-1α signaling might be associated with this process. We also tested in situ prevascularized 3D porous NGCs in vivo by transplanting them into a 10 mm rat sciatic nerve defect model with the aim of regenerating the severed nerve. The prevascularized 3D porous NGCs greatly enhanced intraneural angiogenesis, resulting in demonstrable neurogenesis. Eight weeks after transplantation, the performance of the prevascularized 3D NGCs was similar to that of traditional autografts in terms of improved anatomical structure, morphology, and neural function. In conclusion, combining a reasonably fabricated 3D-pore conduit structure with in situ prevascularization promoted functional nerve regeneration, suggesting an alternative strategy for achieving functional recovery after peripheral nerve trauma.
Collapse
Affiliation(s)
- Junjie Shen
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Yishan Road 600, Shanghai 200233, PR China
- Haikou Orthopedic and Diabetes Hospital of Shanghai Sixth People's Hospital, Hainan 570300, PR China
| | - Jiayan Wang
- College of Materials and Textile Engineering, Nanotechnology Research Institute, Jiaxing University, Zhejiang 314001, PR China
- Key Laboratory of Yarn Materials Forming and Composite Processing Technology, Zhejiang 314001, PR China
| | - Xuanzhe Liu
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Yishan Road 600, Shanghai 200233, PR China
| | - Yi Sun
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Yishan Road 600, Shanghai 200233, PR China
| | - Anlin Yin
- College of Materials and Textile Engineering, Nanotechnology Research Institute, Jiaxing University, Zhejiang 314001, PR China
- Key Laboratory of Yarn Materials Forming and Composite Processing Technology, Zhejiang 314001, PR China
| | - Yimin Chai
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Yishan Road 600, Shanghai 200233, PR China
| | - Kuihua Zhang
- College of Materials and Textile Engineering, Nanotechnology Research Institute, Jiaxing University, Zhejiang 314001, PR China
- Key Laboratory of Yarn Materials Forming and Composite Processing Technology, Zhejiang 314001, PR China
| | - Chunyang Wang
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Yishan Road 600, Shanghai 200233, PR China
- Haikou Orthopedic and Diabetes Hospital of Shanghai Sixth People's Hospital, Hainan 570300, PR China
| | - Xianyou Zheng
- Department of Orthopedic Surgery, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Yishan Road 600, Shanghai 200233, PR China
| |
Collapse
|
143
|
Sun T, Meng C, Ding Q, Yu K, Zhang X, Zhang W, Tian W, Zhang Q, Guo X, Wu B, Xiong Z. In situ bone regeneration with sequential delivery of aptamer and BMP2 from an ECM-based scaffold fabricated by cryogenic free-form extrusion. Bioact Mater 2021; 6:4163-4175. [PMID: 33997500 PMCID: PMC8099605 DOI: 10.1016/j.bioactmat.2021.04.013] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 04/09/2021] [Accepted: 04/12/2021] [Indexed: 12/11/2022] Open
Abstract
In situ tissue engineering is a powerful strategy for the treatment of bone defects. It could overcome the limitations of traditional bone tissue engineering, which typically involves extensive cell expansion steps, low cell survival rates upon transplantation, and a risk of immuno-rejection. Here, a porous scaffold polycaprolactone (PCL)/decellularized small intestine submucosa (SIS) was fabricated via cryogenic free-form extrusion, followed by surface modification with aptamer and PlGF-2123-144*-fused BMP2 (pBMP2). The two bioactive molecules were delivered sequentially. The aptamer Apt19s, which exhibited binding affinity to bone marrow-derived mesenchymal stem cells (BMSCs), was quickly released, facilitating the mobilization and recruitment of host BMSCs. BMP2 fused with a PlGF-2123-144 peptide, which showed "super-affinity" to the ECM matrix, was released in a slow and sustained manner, inducing BMSC osteogenic differentiation. In vitro results showed that the sequential release of PCL/SIS-pBMP2-Apt19s promoted cell migration, proliferation, alkaline phosphatase activity, and mRNA expression of osteogenesis-related genes. The in vivo results demonstrated that the sequential release system of PCL/SIS-pBMP2-Apt19s evidently increased bone formation in rat calvarial critical-sized defects compared to the sequential release system of PCL/SIS-BMP2-Apt19s. Thus, the novel delivery system shows potential as an ideal alternative for achieving cell-free scaffold-based bone regeneration in situ.
Collapse
Key Words
- 3D, three-dimensional
- Apt19s, aptamer 19s
- Aptamer
- BMD, bone mineral density
- BMP2
- BMP2, bone morphogenic protein 2
- BMSC, bone marrow-derived mesenchymal stem cell
- Bone regeneration in situ
- CLSM, confocal laser scanning microscopy
- CSD, critical-sized calvarial defect
- Cell recruitment
- Controlled delivery
- ECM, decellularized matrix
- FBS, fetal bovine serum
- FDA, US Food and Drug Administration
- FITC, fluorescein isothiocyanate
- FTIR, Fourier transform infrared
- H&E, hematoxylin and eosin
- HA, hydroxyapatite
- PCL, polycaprolactone
- PVDF, polyvinylidene difluoride
- Rh6G, rhodamine 6G
- SIS, small intestine submucosa
- pBMP2, PlGF-2123-144*-fused BMP2
- ssDNA, single-stranded DNA
Collapse
Affiliation(s)
- Tingfang Sun
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Chunqing Meng
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Qiuyue Ding
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Keda Yu
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Xianglin Zhang
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Wancheng Zhang
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Wenqing Tian
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Qi Zhang
- Wuhan Hi-tech Medical Tissue Research Center, Wuhan, 430206, China
| | - Xiaodong Guo
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| | - Bin Wu
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Zekang Xiong
- Department of Orthopaedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, China
| |
Collapse
|
144
|
Xue W, Shi W, Kong Y, Kuss M, Duan B. Anisotropic scaffolds for peripheral nerve and spinal cord regeneration. Bioact Mater 2021; 6:4141-4160. [PMID: 33997498 PMCID: PMC8099454 DOI: 10.1016/j.bioactmat.2021.04.019] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/05/2021] [Accepted: 04/13/2021] [Indexed: 12/12/2022] Open
Abstract
The treatment of long-gap (>10 mm) peripheral nerve injury (PNI) and spinal cord injury (SCI) remains a continuous challenge due to limited native tissue regeneration capabilities. The current clinical strategy of using autografts for PNI suffers from a source shortage, while the pharmacological treatment for SCI presents dissatisfactory results. Tissue engineering, as an alternative, is a promising approach for regenerating peripheral nerves and spinal cords. Through providing a beneficial environment, a scaffold is the primary element in tissue engineering. In particular, scaffolds with anisotropic structures resembling the native extracellular matrix (ECM) can effectively guide neural outgrowth and reconnection. In this review, the anatomy of peripheral nerves and spinal cords, as well as current clinical treatments for PNI and SCI, is first summarized. An overview of the critical components in peripheral nerve and spinal cord tissue engineering and the current status of regeneration approaches are also discussed. Recent advances in the fabrication of anisotropic surface patterns, aligned fibrous substrates, and 3D hydrogel scaffolds, as well as their in vitro and in vivo effects are highlighted. Finally, we summarize potential mechanisms underlying the anisotropic architectures in orienting axonal and glial cell growth, along with their challenges and prospects.
Collapse
Affiliation(s)
- Wen Xue
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Wen Shi
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Yunfan Kong
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Mitchell Kuss
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program, University of Nebraska Medical Center, Omaha, NE, USA
- Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Surgery, University of Nebraska Medical Center, Omaha, NE, USA
- Department of Mechanical Engineering, University of Nebraska-Lincoln, Lincoln, NE, USA
| |
Collapse
|
145
|
Sthijns MMJPE, van Blitterswijk CA, LaPointe VLS. Synthetic Materials that Affect the Extracellular Matrix via Cellular Metabolism and Responses to a Metabolic State. Front Bioeng Biotechnol 2021; 9:742132. [PMID: 34708025 PMCID: PMC8542861 DOI: 10.3389/fbioe.2021.742132] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 09/28/2021] [Indexed: 11/18/2022] Open
Abstract
In regenerative medicine and tissue engineering, many materials are developed to mimic the extracellular matrix (ECM). However, these ECM-mimicking materials do not yet completely recapitulate the diversity and complexity of biological tissue-specific ECM. In this review, an alternative strategy is proposed to generate ECM, namely synthesizing a material that functions as a drug delivery system, releasing molecules that target cellular metabolic pathways and thereby stimulate the local cells to create their own ECM. This is based on the fact that ECM synthesis, modification, composition, signaling, stiffness, and degradation are modulated by cellular metabolism. Metabolism can be targeted at different levels, ranging from modulating the availability of substrates or co-factors to regulating the activity of essential transcription factors. Depending on the drug of interest, its characteristics, mechanism of action, cellular target, and application, a different drug delivery system should be designed. Metabolic drugs modulating the ECM require cellular uptake for their function, therefore reversible linkers are recommended. Preferably the metabolic modulators are only released when needed, which will be upon a specific metabolic state, a change in ECM stiffness, or ECM remodeling. Therefore, reversible linkers that respond to an environmental stimulus could be incorporated. All in all, a novel strategy is suggested to develop a tissue-specific ECM by generating a synthetic material that releases metabolic molecules modulating the ECM. Various ways to modulate the ECM properties via the metabolism are reviewed and guidelines for the development of these materials are provided.
Collapse
Affiliation(s)
- Mireille M J P E Sthijns
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands.,Department of Food Innovation and Health at the Centre of Healthy Eating and Food Innovation, Maastricht University, Maastricht, Netherlands
| | - Clemens A van Blitterswijk
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| | - Vanessa L S LaPointe
- Department of Cell Biology-Inspired Tissue Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, Maastricht, Netherlands
| |
Collapse
|
146
|
Redenski I, Guo S, Machour M, Szklanny A, Landau S, Kaplan B, Lock RI, Gabet Y, Egozi D, Vunjak‐Novakovic G, Levenberg S. Engineered Vascularized Flaps, Composed of Polymeric Soft Tissue and Live Bone, Repair Complex Tibial Defects. ADVANCED FUNCTIONAL MATERIALS 2021; 31:2008687. [DOI: 10.1002/adfm.202008687] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Indexed: 02/05/2023]
Affiliation(s)
- Idan Redenski
- Department of Biomedical Engineering Technion—Israel Institute of Technology Haifa 32000 Israel
| | - Shaowei Guo
- Department of Biomedical Engineering Technion—Israel Institute of Technology Haifa 32000 Israel
- The First Affiliated Hospital Shantou University Medical College Shantou 515000 China
| | - Majd Machour
- Department of Biomedical Engineering Technion—Israel Institute of Technology Haifa 32000 Israel
| | - Ariel Szklanny
- Department of Biomedical Engineering Technion—Israel Institute of Technology Haifa 32000 Israel
| | - Shira Landau
- Department of Biomedical Engineering Technion—Israel Institute of Technology Haifa 32000 Israel
| | - Ben Kaplan
- Department of Biomedical Engineering Technion—Israel Institute of Technology Haifa 32000 Israel
| | - Roberta I. Lock
- Department of Biomedical Engineering Columbia University New York NY 10032 USA
| | - Yankel Gabet
- Department of Anatomy and Anthropology Sackler Faculty of Medicine Tel‐Aviv University Tel‐Aviv 6997801 Israel
| | - Dana Egozi
- Department of Plastic and Reconstructive Surgery Kaplan Hospital Rehovot and the Hebrew University Jerusalem 7661041 Israel
| | | | - Shulamit Levenberg
- Department of Biomedical Engineering Technion—Israel Institute of Technology Haifa 32000 Israel
| |
Collapse
|
147
|
Jana S, Franchi F, Lerman A. Fibrous heart valve leaflet substrate with native-mimicked morphology. APPLIED MATERIALS TODAY 2021; 24:101112. [PMID: 34485682 PMCID: PMC8415466 DOI: 10.1016/j.apmt.2021.101112] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/26/2023]
Abstract
Tissue-engineered heart valves are a promising alternative solution to prosthetic valves. However, long-term functionalities of tissue-engineered heart valves depend on the ability to mimic the trilayered, oriented structure of native heart valve leaflets. In this study, using electrospinning, we developed trilayered microfibrous leaflet substrates with morphological characteristics similar to native leaflets. The substrates were implanted subcutaneously in rats to study the effect of their trilayered oriented structure on in vivo tissue engineering. The tissue constructs showed a well-defined structure, with a circumferentially oriented layer, a randomly oriented layer and a radially oriented layer. The extracellular matrix, produced during in vivo tissue engineering, consisted of collagen, glycosaminoglycans, and elastin, all major components of native leaflets. Moreover, the anisotropic tensile properties of the constructs were sufficient to bear the valvular physiological load. Finally, the expression of vimentin and α-smooth muscle actin, at the gene and protein level, was detected in the residing cells, revealing their growing state and their transdifferentiation to myofibroblasts. Our data support a critical role for the trilayered structure and anisotropic properties in functional leaflet tissue constructs, and indicate that the leaflet substrates have the potential for the development of valve scaffolds for heart valve replacements.
Collapse
Affiliation(s)
- Soumen Jana
- Department of Bioengineering, University of Missouri,
Columbia, MO 65211, USA
- Department of Cardiovascular Medicine, Mayo Clinic, 200
First Street SW, Rochester, MN 55905, USA
| | - Federico Franchi
- Department of Cardiovascular Medicine, Mayo Clinic, 200
First Street SW, Rochester, MN 55905, USA
| | - Amir Lerman
- Department of Cardiovascular Medicine, Mayo Clinic, 200
First Street SW, Rochester, MN 55905, USA
| |
Collapse
|
148
|
Blake C, Massey O, Boyd-Moss M, Firipis K, Rifai A, Franks S, Quigley A, Kapsa R, Nisbet DR, Williams RJ. Replace and repair: Biomimetic bioprinting for effective muscle engineering. APL Bioeng 2021; 5:031502. [PMID: 34258499 PMCID: PMC8270648 DOI: 10.1063/5.0040764] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Accepted: 05/10/2021] [Indexed: 12/24/2022] Open
Abstract
The debilitating effects of muscle damage, either through ischemic injury or volumetric muscle loss (VML), can have significant impacts on patients, and yet there are few effective treatments. This challenge arises when function is degraded due to significant amounts of skeletal muscle loss, beyond the regenerative ability of endogenous repair mechanisms. Currently available surgical interventions for VML are quite invasive and cannot typically restore function adequately. In response to this, many new bioengineering studies implicate 3D bioprinting as a viable option. Bioprinting for VML repair includes three distinct phases: printing and seeding, growth and maturation, and implantation and application. Although this 3D bioprinting technology has existed for several decades, the advent of more advanced and novel printing techniques has brought us closer to clinical applications. Recent studies have overcome previous limitations in diffusion distance with novel microchannel construct architectures and improved myotubule alignment with highly biomimetic nanostructures. These structures may also enhance angiogenic and nervous ingrowth post-implantation, though further research to improve these parameters has been limited. Inclusion of neural cells has also shown to improve myoblast maturation and development of neuromuscular junctions, bringing us one step closer to functional, implantable skeletal muscle constructs. Given the current state of skeletal muscle 3D bioprinting, the most pressing future avenues of research include furthering our understanding of the physical and biochemical mechanisms of myotube development and expanding our control over macroscopic and microscopic construct structures. Further to this, current investigation needs to be expanded from immunocompromised rodent and murine myoblast models to more clinically applicable human cell lines as we move closer to viable therapeutic implementation.
Collapse
Affiliation(s)
- Cooper Blake
- Institute of Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia
| | - Oliver Massey
- Institute of Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia
| | | | | | | | - Stephanie Franks
- Laboratory of Advanced Biomaterials, The Australian National University, Canberra, ACT 2601, Australia
| | | | | | | | | |
Collapse
|
149
|
Safdari M, Bibak B, Soltani H, Hashemi J. Recent advancements in decellularized matrix technology for bone tissue engineering. Differentiation 2021; 121:25-34. [PMID: 34454348 DOI: 10.1016/j.diff.2021.08.004] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 08/17/2021] [Accepted: 08/20/2021] [Indexed: 12/11/2022]
Abstract
The native extracellular matrix (ECM) provides a matrix to hold tissue/organ, defines the cellular fate and function, and retains growth factors. Such a matrix is considered as a most biomimetic scaffold for tissue engineering due to the biochemical and biological components, 3D hierarchical structure, and physicomechanical properties. Several attempts have been performed to decellularize allo- or xeno-graft tissues and used them for bone repairing and regeneration. Decellularized ECM (dECM) technology has been developed to create an in vivo-like microenvironment to promote cell adhesion, growth, and differentiation for tissue repair and regeneration. Decellularization is mediated through physical, chemical, and enzymatic methods. In this review, we describe the recent progress in bone decellularization and their applications as a scaffold, hydrogel, bioink, or particles in bone tissue engineering. Furthermore, we address the native dECM limitations and the potential of non-bone dECM, cell-based ECM, and engineered ECM (eECM) for in vitro osteogenic differentiation and in vivo bone regeneration.
Collapse
Affiliation(s)
- Mohammadreza Safdari
- Department of Surgery, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Bahram Bibak
- Department of Physiology and Pharmacology, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran; Research Center of Natural Products Safety and Medicinal Plants, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Hoseinali Soltani
- Department of General Surgery, Imam Ali Hospital, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran
| | - Javad Hashemi
- Research Center of Natural Products Safety and Medicinal Plants, North Khorasan University of Medical Sciences, Bojnurd, Iran; Department of Pathobiology and Laboratory Sciences, School of Medicine, North Khorasan University of Medical Sciences, Bojnurd, Iran.
| |
Collapse
|
150
|
Mallis P, Sokolis DP, Katsimpoulas M, Kostakis A, Stavropoulos-Giokas C, Michalopoulos E. Improved Repopulation Efficacy of Decellularized Small Diameter Vascular Grafts Utilizing the Cord Blood Platelet Lysate. Bioengineering (Basel) 2021; 8:118. [PMID: 34562940 PMCID: PMC8467559 DOI: 10.3390/bioengineering8090118] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2021] [Revised: 08/18/2021] [Accepted: 08/24/2021] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND The development of functional bioengineered small-diameter vascular grafts (SDVGs), represents a major challenge of tissue engineering. This study aimed to evaluate the repopulation efficacy of biological vessels, utilizing the cord blood platelet lysate (CBPL). METHODS Human umbilical arteries (hUAs, n = 10) were submitted to decellularization. Then, an evaluation of decellularized hUAs, involving histological, biochemical and biomechanical analysis, was performed. Wharton's Jelly (WJ) Mesenchymal Stromal Cells (MSCs) were isolated and characterized for their properties. Then, WJ-MSCs (1.5 × 106 cells) were seeded on decellularized hUAs (n = 5) and cultivated with (Group A) or without the presence of the CBPL, (Group B) for 30 days. Histological analysis involving immunohistochemistry (against Ki67, for determination of cell proliferation) and indirect immunofluorescence (against activated MAP kinase, additional marker for cell growth and proliferation) was performed. RESULTS The decellularized hUAs retained their initial vessel's properties, in terms of key-specific proteins, the biochemical and biomechanical characteristics were preserved. The evaluation of the repopulation process indicated a more uniform distribution of WJ-MSCs in group A compared to group B. The repopulated vascular grafts of group B were characterized by greater Ki67 and MAP kinase expression compared to group A. CONCLUSION The results of this study indicated that the CBPL may improve the repopulation efficacy, thus bringing the biological SDVGs one step closer to clinical application.
Collapse
Affiliation(s)
- Panagiotis Mallis
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece; (C.S.-G.); (E.M.)
| | - Dimitrios P. Sokolis
- Laboratory of Biomechanics, Center for Experimental Surgery, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece;
| | - Michalis Katsimpoulas
- Center of Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece; (M.K.); (A.K.)
| | - Alkiviadis Kostakis
- Center of Experimental Surgery and Translational Research, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece; (M.K.); (A.K.)
| | - Catherine Stavropoulos-Giokas
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece; (C.S.-G.); (E.M.)
| | - Efstathios Michalopoulos
- Hellenic Cord Blood Bank, Biomedical Research Foundation Academy of Athens, 4 Soranou Ephessiou Street, 115 27 Athens, Greece; (C.S.-G.); (E.M.)
| |
Collapse
|