1
|
Yang L, Bai X, Liu Y, Zhu S, Li S, Chen Z, Han T, Jin S, Zang M. Angiosome-Guided Perfusion Decellularization of Fasciocutaneous Flaps. J Reconstr Microsurg 2025; 41:405-414. [PMID: 39191422 DOI: 10.1055/a-2404-2608] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/29/2024]
Abstract
BACKGROUND Tissue engineering based on whole-organ perfusion decellularization has successfully generated small-animal organs, including the heart and limbs. Herein, we aimed to use angiosome-guided perfusion decellularization to develop an acellular fasciocutaneous flap matrix with an intact vascular network. METHODS Abdominal flaps of rats were harvested, and the vascular pedicle (iliac artery and vein) was dissected and injected with methylene blue to identify the angiosome region and determine the flap dimension for harvesting. To decellularize flaps, the iliac artery was perfused sequentially with 1% sodium dodecyl sulfate (SDS), deionized water, and 1% Triton-X100. Gross morphology, histology, and DNA quantity of flaps were then obtained. Flaps were also subjected to glycosaminoglycan (GAG) and hydroxyproline content assays and computed tomography angiography. RESULTS Histological assessment indicated that cellular content was completely removed in all flap layers following a 10-hour perfusion in SDS. DNA quantification confirmed 81% DNA removal. Based on biochemical assays, decellularized flaps had hydroxyproline content comparable with that of native flaps, although significantly fewer GAGs (p = 0.0019). Histology and computed tomography angiography illustrated the integrity and perfusability of the vascular system. CONCLUSION The proposed angiosome-guided perfusion decellularization protocol could effectively remove cellular content from rat fasciocutaneous flaps and preserve the integrity of innate vascular networks.
Collapse
Affiliation(s)
- Liya Yang
- Division of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Xueshan Bai
- Division of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Yuanbo Liu
- Division of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Shan Zhu
- Division of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Shanshan Li
- Division of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Zixiang Chen
- Division of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Tinglu Han
- Division of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Shengyang Jin
- Division of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| | - Mengqing Zang
- Division of Plastic Surgery, Plastic Surgery Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, People's Republic of China
| |
Collapse
|
2
|
Tavares DF, Mano JF, Oliveira MB. Advances in abiotic tissue-based biomaterials: A focus on decellularization and devitalization techniques. Mater Today Bio 2025; 32:101735. [PMID: 40275948 PMCID: PMC12020859 DOI: 10.1016/j.mtbio.2025.101735] [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: 01/03/2025] [Revised: 03/14/2025] [Accepted: 04/05/2025] [Indexed: 04/26/2025] Open
Abstract
This Review explores the growing and diversifying field of tissue-derived abiotic constructs for tissue engineering applications, with main focus on decellularization and devitalization techniques and principles. Acellular fractions derived from biological tissues, such as the extracellular matrix (ECM), have long been considered a valuable approach for the generation of numerous scaffolds and more complex constructs. The removal of the cellular content has been considered essential to prevent the development of adverse immunological reactions. Nevertheless, the discovery of promising features of certain cellular components has sparked interest in the use of inactivated or devitalized cellular fractions for several applications, particularly in regenerative medicine and inflammation control. Devitalization has been described for several clinical applications, but remains poorly explored in terms of in vitro constructs compared to decellularization methods currently available. In this review, we present and critically evaluate a spectrum of approaches for the decellularization of whole-organs and in vitro constructs, and the most prevalent devitalization techniques, with a discussion on their implications on scaffolds composition, structure, and potentially therapeutic properties. Processing methodologies to achieve optimal cell-based abiotic materials and approaches for their effective characterization are described and discussed. The application of these materials in healthcare, with most focus on regenerative approaches and including examples of commercially available products, is also addressed.
Collapse
Affiliation(s)
- Diana F. Tavares
- Department of Chemistry, CICECO – Aveiro Institute of Materials. University of Aveiro., Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - João F. Mano
- Department of Chemistry, CICECO – Aveiro Institute of Materials. University of Aveiro., Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| | - Mariana B. Oliveira
- Department of Chemistry, CICECO – Aveiro Institute of Materials. University of Aveiro., Campus Universitário de Santiago, 3810-193, Aveiro, Portugal
| |
Collapse
|
3
|
Sun J, Jia S, Shao C, Dawson MR, Toussaint KC. Emerging Technologies for Multiphoton Writing and Reading of Polymeric Architectures for Biomedical Applications. Annu Rev Biomed Eng 2025; 27:129-155. [PMID: 39874608 DOI: 10.1146/annurev-bioeng-110122-015901] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
The rise in popularity of two-photon polymerization (TPP) as an additive manufacturing technique has impacted many areas of science and engineering, particularly those related to biomedical applications. Compared with other fabrication methods used for biomedical applications, TPP offers 3D, nanometer-scale fabrication dexterity (free-form). Moreover, the existence of turnkey commercial systems has increased accessibility. In this review, we discuss the diversity of biomedical applications that have benefited from the unique features of TPP. We also present the state of the art in approaches for patterning and reading 3D TPP-fabricated structures. The reading process influences the fidelity for both in situ and ex situ characterization methods. We also review efforts to leverage machine learning to facilitate process control for TPP. Finally, we conclude with a discussion of both the current challenges and exciting opportunities for biomedical applications that lie ahead for this intriguing and emerging technology.
Collapse
Affiliation(s)
- Jieliyue Sun
- School of Engineering, Brown University, Providence, Rhode Island, USA;
| | - Sixian Jia
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Chenhui Shao
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, Michigan, USA
| | - Michelle R Dawson
- Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, Rhode Island, USA
- Center for Biomedical Engineering, Brown University, Providence, Rhode Island, USA
| | - Kimani C Toussaint
- School of Engineering, Brown University, Providence, Rhode Island, USA;
- Brown University Center for Digital Health, Providence, Rhode Island, USA
| |
Collapse
|
4
|
Carrascal-Hernández DC, Martínez-Cano JP, Rodríguez Macías JD, Grande-Tovar CD. Evolution in Bone Tissue Regeneration: From Grafts to Innovative Biomaterials. Int J Mol Sci 2025; 26:4242. [PMID: 40362478 PMCID: PMC12072198 DOI: 10.3390/ijms26094242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2025] [Revised: 04/24/2025] [Accepted: 04/27/2025] [Indexed: 05/15/2025] Open
Abstract
Bone defects caused by various traumas and diseases such as osteoporosis, which affects bone density, and osteosarcoma, which affects the integrity of bone structure, are now well known. Given this situation, several innovative research projects have been reported to improve orthopedic methods and technologies that positively contribute to the regeneration of affected bone tissue, representing a significant advance in regenerative medicine. This review article comprehensively analyzes the transition from existing methods and technologies for implants and bone tissue regeneration to innovative biomaterials. These biomaterials have been of great interest in the last decade due to their physicochemical characteristics, which allow them to overcome the most common limitations of traditional grafting methods, such as the availability of biomaterials and the risk of rejection after their application in regenerative medicine. This could be achieved through an exhaustive study of the applications and properties of various materials with potential applications in regenerative medicine, such as using magnetic nanoparticles and hydrogels sensitive to external stimuli, including pH and temperature. In this regard, this review article describes the most relevant compounds used in bone tissue regeneration, promoting the integration of these biomaterials with the affected area's bone structure, thereby allowing for regeneration and preventing amputation. Additionally, the types of interactions between biomaterials and mesenchymal stem cells and their effects on bone tissue are discussed, which is critical for developing biomaterials with optimal regenerative properties. Furthermore, the mechanisms of action of the various biomaterials that enhance osteoconduction and osteoinduction, ensuring the success of orthopedic therapies, are analyzed. This enables the treatment of bone defects tailored to each patient's condition, thereby avoiding limb amputation. Consequently, a promising future for regenerative medicine is emerging, with various therapies that could revolutionize the management of bone defects, offering more efficient and safer solutions.
Collapse
Affiliation(s)
| | - Juan Pablo Martínez-Cano
- Ortopedia y Traumatología, Epidemiología Clínica, Fundación Valle del Lili, Universidad ICESI, Cali 760031, Colombia;
| | | | - Carlos David Grande-Tovar
- Grupo de Investigación en Fotoquímica y Fotobiología, Programa de Química, Universidad del Atlántico, Puerto Colombia 081007, Colombia
| |
Collapse
|
5
|
Suzuki M, Kawauchi K, Machiyama H, Hirata H, Ishiwata S, Fujita H. Dynamic Remodeling of Mechano-Sensing Complexes in Suspended Fibroblast Cell-Sheets Under External Mechanical Stimulus. Biotechnol Bioeng 2025. [PMID: 40270085 DOI: 10.1002/bit.28996] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Revised: 04/02/2025] [Accepted: 04/05/2025] [Indexed: 04/25/2025]
Abstract
Freestanding cell-sheets are valuable bio-materials for use in regenerative medicine and tissue engineering. Because cell-sheets experience various mechanical stimulations during handling, it is important to understand the responses of cells to these stimulations. Here, we demonstrate changes in the localization of various proteins during the stretching of fibroblast cell-sheets. These proteins are known to be involved in mechano-sensing. Upon stretching, actin filaments appear parallel to the stretching direction. At cell-cell junctions, β-catenin forms clusters that co-localize with accumulated vinculin and zyxin as well as the actin filaments. The p130 Crk-associated substrate, known to be present in focal adhesions, is also recruited to these clusters and phosphorylated. Our results suggest that mechano-sensing machinery is formed at cell-cell junctions when the cell-sheets are stretched.
Collapse
Affiliation(s)
- Madoka Suzuki
- Institute for Protein Research, The University of Osaka, Suita, Osaka, Japan
| | - Keiko Kawauchi
- Faculty of Frontiers of Innovative Research in Science and Technology, Konan University, Kobe, Hyogo, Japan
| | - Hiroaki Machiyama
- Department of Immunology, Tokyo Medical University, Shinjuku-ku, Tokyo, Japan
| | - Hiroaki Hirata
- Department of Life Science and Biotechnology, Kanazawa Institute of Technology, Hakusan, Ishikawa, Japan
| | - Shin'ichi Ishiwata
- Department of Physics, Faculty of Science and Engineering, Waseda University, Shinjuku-ku, Tokyo, Japan
| | - Hideaki Fujita
- Department of Stem Cell Biology, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Hiroshima, Japan
| |
Collapse
|
6
|
Shih JH, Chern E. Decellularized Porcine Aorta as a Scaffold for Human Induced Pluripotent Stem Cell-Derived Mesenchymal Stem Cells in Tissue Engineering. Stem Cell Rev Rep 2025:10.1007/s12015-025-10875-y. [PMID: 40227487 DOI: 10.1007/s12015-025-10875-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/02/2025] [Indexed: 04/15/2025]
Abstract
Tissue engineering has been an integral part of regenerative medicine. Functional biomimetic structures were assembled by combining appropriate scaffolds with specific cells. The decellularization of animal tissue preserved the natural biochemical components and structural properties of the extracellular matrix (ECM) of specific organs, thereby providing a suitable niche for tissue-specific cell differentiation and growth. In this study, the extracellular matrix (ECM) of the porcine aorta was obtained through trypsin-based decellularization. The resulting porcine aortic ECM retained a favorable collagen composition, exhibited no cytotoxicity, and demonstrated chemophilic properties for mesenchymal stem cells. Human adipose-derived mesenchymal stem cells (hADSCs) and human induced pluripotent stem cell-derived mesenchymal stem cells (hiMSCs) were transplanted onto the decellularized porcine aortic ECM, where successful differentiation into a mature cartilage layer was observed. These findings suggested that the porcine aortic ECM could serve as a potential scaffold with tubular and linear structures for tissue engineering applications. Functional human iMSCs (induced-mesenchymal stem cells) were generated from human iPSCs (induced-pluripotent stem cells) and analyzed for differences compared to primary MSCs via RNA-seq. The hiMSCs were applied to decellularized porcine aortic ECM (extracellular matrix), demonstrating chondrogenic differentiation and confirming the usability of xenogeneic ECM.
Collapse
Affiliation(s)
- Jheng-Hong Shih
- niChe Lab for Stem Cell and Regenerative Medicine, Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, 10617, Taiwan
| | - Edward Chern
- niChe Lab for Stem Cell and Regenerative Medicine, Department of Biochemical Science and Technology, College of Life Science, National Taiwan University, Taipei, 10617, Taiwan.
- Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, No. 1, Sec. 4 Roosevelt Road, Taipei, 10617, Taiwan.
| |
Collapse
|
7
|
Xia X, Hu M, Zhou W, Jin Y, Yao X. Engineering cardiology with miniature hearts. Mater Today Bio 2025; 31:101505. [PMID: 39911371 PMCID: PMC11795835 DOI: 10.1016/j.mtbio.2025.101505] [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: 11/07/2024] [Revised: 12/28/2024] [Accepted: 01/18/2025] [Indexed: 02/07/2025] Open
Abstract
Cardiac organoids offer sophisticated 3D structures that emulate key aspects of human heart development and function. This review traces the evolution of cardiac organoid technology, from early stem cell differentiation protocols to advanced bioengineering approaches. We discuss the methodologies for creating cardiac organoids, including self-organization techniques, biomaterial-based scaffolds, 3D bioprinting, and organ-on-chip platforms, which have significantly enhanced the structural complexity and physiological relevance of in vitro cardiac models. We examine their applications in fundamental research and medical innovations, highlighting their potential to transform our understanding of cardiac biology and pathology. The integration of multiple cell types, vascularization strategies, and maturation protocols has led to more faithful representations of the adult human heart. However, challenges remain in achieving full functional maturity and scalability. We critically assess the current limitations and outline future directions for advancing cardiac organoid technology. By providing a comprehensive analysis of the field, this review aims to catalyze further innovation in cardiac tissue engineering and facilitate its translation to clinical applications.
Collapse
Affiliation(s)
- Xiaojun Xia
- Department of Cardiology, Center of Regenerative and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Miner Hu
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310000, China
| | - Wenyan Zhou
- School of Medicine, Taizhou University, Taizhou, Zhejiang, 318000, China
| | - Yunpeng Jin
- Department of Cardiology, Center of Regenerative and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Xudong Yao
- Department of Cardiology, Center of Regenerative and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| |
Collapse
|
8
|
Cheng W, Huang Y, Dai J, Zhao M, Wang Y, Turner N, Zhang J. Endotoxin, not DNA, determines the host response and tissue regeneration behavior of acellular biologic scaffolds. Acta Biomater 2025; 195:157-168. [PMID: 39921179 DOI: 10.1016/j.actbio.2025.02.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 01/26/2025] [Accepted: 02/04/2025] [Indexed: 02/10/2025]
Abstract
Established quantitative standards for assessing decellularization of biologic scaffolds based on residual DNA levels have been well-documented and widely acknowledged. However, post-implantation complications, such as fever and seroma, are commonly observed which negatively impact clinical outcomes. The presence of cellular debris following decellularization or using source tissues that are naturally high in endotoxin may contribute to the host response to a biologic scaffold. In the study, several multi-step decellularization methods were used to decellularize small intestinal submucosa (SIS) to obtain materials with three distinct levels of residual DNA, lipid residues, and endogenous endotoxin. The potential influence of these residual components on macrophage and lymphocyte polarization in vitro, as well as on the host inflammatory response in vivo post intra-abdominal implantation or abdominal wall defect repair in rats, was assessed. Urinary bladder matrix (UBM) meeting established decellularization criteria and naturally devoid of endotoxin was utilized as a control. The presence of endogenous endotoxin in SIS-ECM resulted in notable changes in macrophage phenotype. SIS-ECM samples with endotoxin levels below FDA limits still upregulated pro-inflammatory factors in vitro. Conversely, SIS with minimal endotoxin content and UBM controls prompted a shift towards a pro-remodeling M2 phenotype, fostering constructive tissue remodeling in a rodent model of abdominal wall defects, irrespective of DNA content. These findings suggest that endotoxin may be a crucial factor influencing biologic scaffolds that are not fully accounted by current decellularization standards. STATEMENT OF SIGNIFICANCE: Clinically utilized decellularized biologic scaffolds that meet the established quantitative standards still suffer problems in high incidence of inflammatory complications, including fever and seroma. In this study, we confirmed that endotoxin, rather than residual DNA, is the crucial factor influencing host responses and regenerative outcomes. Tissue sources and decellularization processes are critical for reducing endotoxin levels and attenuating immuno-inflammatory complications. These findings enhance the evaluation of ECM scaffold performance for clinical application, thereby facilitating improved preparation and utilization for tissue defect repairs.
Collapse
Affiliation(s)
- Wenyue Cheng
- Department of Colorectal Surgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Yonggang Huang
- Department of General surgery, School of Medicine, Affiliated Hangzhou First People's Hospital, Westlake University, Hangzhou 310030, China
| | - Jing Dai
- Department of Colorectal Surgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Meibiao Zhao
- ZhuoRuan Medical Technology (Suzhou) Co., Ltd, Suzhou 215400, China
| | - Yulu Wang
- Department of Colorectal Surgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, China
| | - Neill Turner
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, USA.
| | - Jian Zhang
- Department of Colorectal Surgery, Changzheng Hospital, Naval Medical University, Shanghai 200003, China.
| |
Collapse
|
9
|
Ahn J, Park JH, Choi HJ, Lee D, Hong HE, Kim OH, Kim SJ. Scaffold implantation vs. intravenous delivery: a comparative preclinical animal study evaluating peroxisome proliferator-activated receptor gamma coactivator 1-alpha adipose-derived stem cells in liver fibrosis treatment. Ann Surg Treat Res 2025; 108:186-197. [PMID: 40083980 PMCID: PMC11896761 DOI: 10.4174/astr.2025.108.3.186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/30/2024] [Accepted: 11/25/2024] [Indexed: 03/16/2025] Open
Abstract
Purpose Regenerative medicine is expected to offer an alternative to liver transplantation for treating liver diseases in the future, with one significant challenge being the establishment of an effective stem cell administration route. This study assessed the antifibrogenic effects of adipose-derived stem cells (ASCs) in a liver fibrosis mouse model, focusing on 2 methods of delivery: intravenous injection and scaffold implantation. Methods An extracellular matrix mimic scaffold was utilized for culturing peroxisome proliferator-activated receptor gamma coactivator 1-alpha-overexpressing ASCs (tASCs). These scaffolds, laden with tASCs, were then implanted subcutaneously in mice exhibiting liver fibrosis. In contrast, the Cell groups received biweekly intravenous injections of tASCs for 4 weeks. After 4 weeks, tissue samples were harvested from the euthanized mice for subsequent analysis. Results Real-time PCR and Western blot analyses on liver tissues, focusing on markers like alpha-smooth muscle actin (α-SMA), matrix metalloproteinase-2, and transforming growth factor-beta 1 (TGF-β1), showed that both delivery routes substantially lowered fibrotic and inflammatory markers compared to controls (P < 0.05), with no significant differences between the routes. Histological examinations, along with immunohistochemical analysis of α-SMA, collagen type I alpha, and TGF-β1, revealed that the scaffold implantation approach resulted in a greater reduction in fibrosis and lower immunoreactivity for fibrotic markers than intravenous delivery (P < 0.05). Conclusion These findings indicate that delivering tASCs via a scaffold could be more effective, or at least similarly effective, in treating liver fibrosis compared to intravenous delivery. Scaffold implantation could offer a beneficial alternative to frequent intravenous treatments, suggesting its potential utility in clinical applications for liver disease treatment.
Collapse
Affiliation(s)
- Joseph Ahn
- Department of Surgery, Bundang Jesaeng General Hospital, Seongnam, Korea
| | - Jung Hyun Park
- Department of Surgery, Eunpyeong St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Catholic Central Laboratory of Surgery, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ho Joong Choi
- Department of Surgery, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Dosang Lee
- Catholic Central Laboratory of Surgery, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Surgery, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
| | - Ha-Eun Hong
- Catholic Central Laboratory of Surgery, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Translational Research Team, Surginex Co., Ltd., Seoul, Korea
| | - Ok-Hee Kim
- Catholic Central Laboratory of Surgery, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Translational Research Team, Surginex Co., Ltd., Seoul, Korea
| | - Say-June Kim
- Catholic Central Laboratory of Surgery, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Department of Surgery, Seoul St. Mary’s Hospital, College of Medicine, The Catholic University of Korea, Seoul, Korea
- Translational Research Team, Surginex Co., Ltd., Seoul, Korea
| |
Collapse
|
10
|
Campo F, Neroni A, Pignatelli C, Pellegrini S, Marzinotto I, Valla L, Manenti F, Policardi M, Lampasona V, Piemonti L, Citro A. Bioengineering of a human iPSC-derived vascularized endocrine pancreas for type 1 diabetes. Cell Rep Med 2025; 6:101938. [PMID: 39922198 PMCID: PMC11866511 DOI: 10.1016/j.xcrm.2025.101938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 11/18/2024] [Accepted: 01/13/2025] [Indexed: 02/10/2025]
Abstract
Intrahepatic islet transplantation in patients with type 1 diabetes is limited by donor availability and lack of engraftment. Alternative β cell sources and transplantation sites are needed. We demonstrate the feasibility to repurpose a decellularized lung as an endocrine pancreas for β cell replacement. We bioengineer an induced pluripotent stem cell (iPSC)-based version, fabricating a human iPSC-based vascularized endocrine pancreas (iVEP) using iPSC-derived β cells (iPSC-derived islets [SC-islets]) and endothelial cells (iECs). SC-islets and iECs are aggregated into vascularized iβ spheroids (ViβeSs), and over 7 days of culture, spheroids integrate into the bioengineered vasculature, generating a functional, perfusable human endocrine organ. In vitro, the vascularized extracellular matrix (ECM) sustained SC-islet engraftment and survival with a significantly preserved β cell mass and a physiologic insulin release. In vivo, iVEP restores normoglycemia in diabetic NSG mice. We report a human iVEP providing a controlled in vitro insulin-secreting phenotype and in vivo function.
Collapse
Affiliation(s)
- Francesco Campo
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy; Università Vita-Salute San Raffaele, Milan, Italy
| | - Alessia Neroni
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy; Università Vita-Salute San Raffaele, Milan, Italy
| | - Cataldo Pignatelli
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Silvia Pellegrini
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Ilaria Marzinotto
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Libera Valla
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy; Chair for Molecular Animal Breeding and Biotechnology, Gene Center and Department of Veterinary Sciences, LMU Munich, 81377 Munich, Germany; Center for Innovative Medical Models (CiMM), LMU Munich, 85764 Oberschleißheim, Germany
| | - Fabio Manenti
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Martina Policardi
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Vito Lampasona
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy
| | - Lorenzo Piemonti
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy; Università Vita-Salute San Raffaele, Milan, Italy
| | - Antonio Citro
- San Raffaele Diabetes Research Institute, IRCCS San Raffaele Scientific Institute, Milan, Italy.
| |
Collapse
|
11
|
Mohapatra SR, Rama E, Werner MP, Call T, Loewenberg T, Loewen A, Apel C, Kiessling F, Jockenhoevel S. Novel Bioreactor Design for Non-invasive Longitudinal Monitoring of Tissue-Engineered Heart Valves in 7T MRI and Ultrasound. Ann Biomed Eng 2025; 53:383-397. [PMID: 39438348 PMCID: PMC11805768 DOI: 10.1007/s10439-024-03632-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 10/02/2024] [Indexed: 10/25/2024]
Abstract
The development of cardiovascular implants is abundant, yet their clinical adoption remains a significant challenge in the treatment of valvular diseases. Tissue-engineered heart valves (TEHV) have emerged as a promising solution due to their remodeling capabilities, which have been extensively studied in recent years. However, ensuring reproducible production and clinical translation of TEHV requires robust longitudinal monitoring methods.Cardiovascular magnetic resonance imaging (MRI) is a non-invasive, radiation-free technique providing detailed valvular imaging and functional assessment. To facilitate this, we designed a state-of-the-art metal-free bioreactor enabling dynamic MRI and ultrasound imaging. Our compact bioreactor, tailored to fit a 72 mm bore 7 T MRI coil, features an integrated backflow design ensuring MRI compatibility. A pneumatic drive system operates the bioreactor, minimizing potential MRI interference. The bioreactor was digitally designed and constructed using polymethyl methacrylate, utilizing only polyether ether ketone screws for secure fastening. Our biohybrid TEHV incorporates a non-degradable polyethylene terephthalate textile scaffold with fibrin matrix hydrogel and human arterial smooth muscle cells.As a result, the bioreactor was successfully proven to be MRI compatible, with no blooming artifacts detected. The dynamic movement of the TEHVs was observed using gated MRI motion artifact compensation and ultrasound imaging techniques. In addition, the conditioning of TEHVs in the bioreactor enhanced ECM production. Immunohistology demonstrated abundant collagen, α-smooth muscle actin, and a monolayer of endothelial cells throughout the valve cusp. Our innovative methodology provides a physiologically relevant environment for TEHV conditioning and development, enabling accurate monitoring and assessment of functionality, thus accelerating clinical acceptance.
Collapse
Affiliation(s)
- Saurav Ranjan Mohapatra
- Department of Biohybrid & Medical Textile (BioTex), Center for Biohybrid Medical Systems (CBMS), Institute for Applied Medical Engineering, RWTH Aachen University, Forckenbeckstr. 55, 52074, Aachen, Germany
| | - Elena Rama
- Institute for Experimental Molecular Imaging, RWTH Aachen University, Forckenbeckstr. 55, 52074, Aachen, Germany
| | - Maximillian P Werner
- Department of Biohybrid & Medical Textile (BioTex), Center for Biohybrid Medical Systems (CBMS), Institute for Applied Medical Engineering, RWTH Aachen University, Forckenbeckstr. 55, 52074, Aachen, Germany
| | - Tobias Call
- Department of Biohybrid & Medical Textile (BioTex), Center for Biohybrid Medical Systems (CBMS), Institute for Applied Medical Engineering, RWTH Aachen University, Forckenbeckstr. 55, 52074, Aachen, Germany
| | - Tanja Loewenberg
- Department of Biohybrid & Medical Textile (BioTex), Center for Biohybrid Medical Systems (CBMS), Institute for Applied Medical Engineering, RWTH Aachen University, Forckenbeckstr. 55, 52074, Aachen, Germany
| | - Alexander Loewen
- Department of Biohybrid & Medical Textile (BioTex), Center for Biohybrid Medical Systems (CBMS), Institute for Applied Medical Engineering, RWTH Aachen University, Forckenbeckstr. 55, 52074, Aachen, Germany
| | - Christian Apel
- Department of Biohybrid & Medical Textile (BioTex), Center for Biohybrid Medical Systems (CBMS), Institute for Applied Medical Engineering, RWTH Aachen University, Forckenbeckstr. 55, 52074, Aachen, Germany
| | - Fabian Kiessling
- Institute for Experimental Molecular Imaging, RWTH Aachen University, Forckenbeckstr. 55, 52074, Aachen, Germany
| | - Stefan Jockenhoevel
- Department of Biohybrid & Medical Textile (BioTex), Center for Biohybrid Medical Systems (CBMS), Institute for Applied Medical Engineering, RWTH Aachen University, Forckenbeckstr. 55, 52074, Aachen, Germany.
| |
Collapse
|
12
|
Liu J, Song Q, Yin W, Li C, An N, Le Y, Wang Q, Feng Y, Hu Y, Wang Y. Bioactive scaffolds for tissue engineering: A review of decellularized extracellular matrix applications and innovations. EXPLORATION (BEIJING, CHINA) 2025; 5:20230078. [PMID: 40040827 PMCID: PMC11875452 DOI: 10.1002/exp.20230078] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/04/2024] [Accepted: 03/12/2024] [Indexed: 03/06/2025]
Abstract
Decellularized extracellular matrix (dECM) offers a three-dimensional, non-immunogenic scaffold, enriched with bioactive components, making it a suitable candidate for tissue regeneration. Although dECM-based scaffolds have been successfully implemented in preclinical and clinical settings within tissue engineering and regenerative medicine, the mechanisms of tissue remodeling and functional restoration are not fully understood. This review critically assesses the state-of-the-art in dECM scaffolds, including decellularization techniques for various tissues, quality control and cross-linking. It highlights the functional properties of dECM components and their latest applications in multiorgan tissue engineering and biomedicine. Additionally, the review addresses current challenges and limitations of decellularized scaffolds and offers perspectives on future directions in the field.
Collapse
Affiliation(s)
- Juan Liu
- Hepato‐Pancreato‐Biliary CenterBeijing Tsinghua Changgung HospitalSchool of Clinical MedicineTsinghua UniversityBeijingChina
- Key Laboratory of Digital Intelligence HepatologyMinistry of EducationSchool of Clinical MedicineTsinghua UniversityBeijingChina
| | - Qingru Song
- Clinical Translational Science CenterBeijing Tsinghua Changgung HospitalTsinghua UniversityBeijingChina
| | - Wenzhen Yin
- Clinical Translational Science CenterBeijing Tsinghua Changgung HospitalTsinghua UniversityBeijingChina
| | - Chen Li
- Hepato‐Pancreato‐Biliary CenterBeijing Tsinghua Changgung HospitalSchool of Clinical MedicineTsinghua UniversityBeijingChina
- College of Chemistry and Life SciencesBeijing University of TechnologyBeijingChina
| | - Ni An
- Clinical Translational Science CenterBeijing Tsinghua Changgung HospitalTsinghua UniversityBeijingChina
| | - Yinpeng Le
- Hepato‐Pancreato‐Biliary CenterBeijing Tsinghua Changgung HospitalSchool of Clinical MedicineTsinghua UniversityBeijingChina
- Institute of Smart Biomedical MaterialsSchool of Materials Science and EngineeringZhejiang Sci‐Tech UniversityHangzhouPeople's Republic of China
| | - Qi Wang
- Hepato‐Pancreato‐Biliary CenterBeijing Tsinghua Changgung HospitalSchool of Clinical MedicineTsinghua UniversityBeijingChina
- Department of Hepatobiliary and Pancreatic SurgeryThe First Hospital of Jilin UniversityJilin UniversityChangchunChina
| | - Yutian Feng
- Hepato‐Pancreato‐Biliary CenterBeijing Tsinghua Changgung HospitalSchool of Clinical MedicineTsinghua UniversityBeijingChina
| | - Yuelei Hu
- Hepato‐Pancreato‐Biliary CenterBeijing Tsinghua Changgung HospitalSchool of Clinical MedicineTsinghua UniversityBeijingChina
- Department of Hepatobiliary and Pancreatic SurgeryThe First Hospital of Jilin UniversityJilin UniversityChangchunChina
| | - Yunfang Wang
- Hepato‐Pancreato‐Biliary CenterBeijing Tsinghua Changgung HospitalSchool of Clinical MedicineTsinghua UniversityBeijingChina
- Key Laboratory of Digital Intelligence HepatologyMinistry of EducationSchool of Clinical MedicineTsinghua UniversityBeijingChina
- Clinical Translational Science CenterBeijing Tsinghua Changgung HospitalTsinghua UniversityBeijingChina
| |
Collapse
|
13
|
Lee YE, Jeong ES, Kim YM, Gong SP. Investigation of scaffold manufacturing conditions for 3-dimensional culture of myogenic cell line derived from black sea bream ( Acanthopagrus schlegelii). Cytotechnology 2025; 77:18. [PMID: 39676766 PMCID: PMC11635079 DOI: 10.1007/s10616-024-00676-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 12/01/2024] [Indexed: 12/17/2024] Open
Abstract
Culturing fish myogenic cells in vitro holds significant potential to revolutionize aquaculture practices and support sustainable food production. However, advancement in in vitro culture technologies for skeletal muscle-derived myogenic cells have predominantly focused on mammals, with limited studies on fish. Scaffold-based three-dimensional (3D) culture systems for fish myogenic cells remain underexplored, highlighting a critical research gap compared to mammalian systems. This study evaluated the effects of scaffold composition and manufacturing methods on cellular growth in the 3D culture of black sea bream (Acanthopagrus schlegelii) myogenic cells. Scaffolds were manufactured using three natural polymers: black sea bream-derived extracellular matrix (ECM), sodium alginate, and gelatin. Two scaffold types were tested: "cell-laden scaffolds" prepared by mixing cells into the pre-scaffold solution followed by gelation, and "cell-seeding scaffolds" produced by freezing, gelation, and lyophilization before cell inoculation. Scaffold characteristics, including pore size, porosity, swelling ratio, and degradation rate, were assessed. Cell-seeding scaffolds exhibited relatively larger pore size, higher porosity, and higher degradation rate, while cell-laden scaffolds had higher swelling ratios. When black sea bream myogenic cells were cultured in these scaffolds, cell-seeding scaffolds supported cellular growth, particularly when composed of 3% sodium alginate and 4% gelatin with any concentration of ECM. In contrast, cell-laden scaffolds did not support cellular growth regardless of their composition. These findings provide fundamental insights for optimizing scaffold properties to develop more optimized conditions for 3D culture of fish muscle lineage cells.
Collapse
Affiliation(s)
- Ye-Eun Lee
- Department of Fisheries Biology, Pukyong National University, Busan, 48513 Korea
| | - Eun Soo Jeong
- Department of Fisheries Biology, Pukyong National University, Busan, 48513 Korea
| | - Young-Mog Kim
- Department of Food Science and Technology, Pukyong National University, Busan, 48513 Korea
| | - Seung Pyo Gong
- Department of Fisheries Biology, Pukyong National University, Busan, 48513 Korea
- Major in Aquaculture and Applied Life Science, Division of Fisheries Life Science, Pukyong National University, 45 Yongso-ro, Nam-Gu, Busan, 48513 Korea
| |
Collapse
|
14
|
Gionet-Gonzales M, Gathman G, Rosas J, Kunisaki KY, Inocencio DGP, Hakami N, Milburn GN, Pitenis AA, Campbell KS, Pruitt BL, Stowers RS. Stress relaxation rates of myocardium from failing and non-failing hearts. Biomech Model Mechanobiol 2025; 24:265-280. [PMID: 39741200 PMCID: PMC11846740 DOI: 10.1007/s10237-024-01909-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 11/06/2024] [Indexed: 01/02/2025]
Abstract
The heart is a dynamic pump whose function is influenced by its mechanical properties. The viscoelastic properties of the heart, i.e., its ability to exhibit both elastic and viscous characteristics upon deformation, influence cardiac function. Viscoelastic properties change during heart failure (HF), but direct measurements of failing and non-failing myocardial tissue stress relaxation under constant displacement are lacking. Further, how consequences of tissue remodeling, such as fibrosis and fat accumulation, alter the stress relaxation remains unknown. To address this gap, we conducted stress relaxation tests on porcine myocardial tissue to establish baseline properties of cardiac tissue. We found porcine myocardial tissue to be fast relaxing, characterized by stress relaxation tests on both a rheometer and microindenter. We then measured human left ventricle (LV) epicardium and endocardium tissue from non-failing, ischemic HF and non-ischemic HF patients by microindentation. Analyzing by patient groups, we found that ischemic HF samples had slower stress relaxation than non-failing endocardium. Categorizing the data by stress relaxation times, we found that slower stress relaxing tissues were correlated with increased collagen deposition and increased α-smooth muscle actin (α-SMA) stress fibers, a marker of fibrosis and cardiac fibroblast activation, respectively. In the epicardium, analyzing by patient groups, we found that ischemic HF had faster stress relaxation than non-ischemic HF and non-failing. When categorizing by stress relaxation times, we found that faster stress relaxation correlated with Oil Red O staining, a marker for adipose tissue. These data show that changes in stress relaxation vary across the different layers of the heart during ischemic versus non-ischemic HF. These findings reveal how the viscoelasticity of the heart changes, which will lead to better modeling of cardiac mechanics for in vitro and in silico HF models.
Collapse
Affiliation(s)
- Marissa Gionet-Gonzales
- Bioengineering, University of California, Santa Barbara, Santa Barbara, United States
- Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, United States
| | - Gianna Gathman
- Bioengineering, University of California, Santa Barbara, Santa Barbara, United States
| | - Jonah Rosas
- Materials, University of California, Santa Barbara, Santa Barbara, United States
| | - Kyle Y Kunisaki
- Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, United States
| | | | - Niki Hakami
- Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, United States
| | | | - Angela A Pitenis
- Materials, University of California, Santa Barbara, Santa Barbara, United States
| | | | - Beth L Pruitt
- Bioengineering, University of California, Santa Barbara, Santa Barbara, United States.
- Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, United States.
| | - Ryan S Stowers
- Bioengineering, University of California, Santa Barbara, Santa Barbara, United States.
- Mechanical Engineering, University of California, Santa Barbara, Santa Barbara, United States.
| |
Collapse
|
15
|
Morawski M, Krasnodębski M, Rochoń J, Kubiszewski H, Marzęcki M, Topyła D, Murat K, Staszewski M, Szczytko J, Maleszewski M, Grąt M. Decellularized Liver Matrices for Expanding the Donor Pool-An Evaluation of Existing Protocols and Future Trends. Biomolecules 2025; 15:98. [PMID: 39858491 PMCID: PMC11762870 DOI: 10.3390/biom15010098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2024] [Revised: 01/07/2025] [Accepted: 01/08/2025] [Indexed: 01/27/2025] Open
Abstract
Liver transplantation is the only curative option for end-stage liver disease and is necessary for an increasing number of patients with advanced primary or secondary liver cancer. Many patient groups can benefit from this treatment, however the shortage of liver grafts remains an unsolved problem. Liver bioengineering offers a promising method for expanding the donor pool through the production of acellular scaffolds that can be seeded with recipient cells. Decellularization protocols involve the removal of cells using various chemical, physical, and enzymatic steps to create a collagenous network that provides support for introduced cells and future vascular and biliary beds. However, the removal of the cells causes varying degrees of matrix damage, that can affect cell seeding and future organ performance. The main objective of this review is to present the existing techniques of producing decellularized livers, with an emphasis on the assessment and definition of acellularity. Decellularization agents are discussed, and the standard process of acellular matrix production is evaluated. We also introduce the concept of the stepwise assessment of the matrix during decellularization through decellularization cycles. This method may lead to shorter detergent exposure times and less scaffold damage. The introduction of apoptosis induction in the field of organ engineering may provide a valuable alternative to existing long perfusion protocols, which lead to significant matrix damage. A thorough understanding of the decellularization process and the action of the various factors influencing the final composition of the scaffold is essential to produce a biocompatible matrix, which can be the basis for further studies regarding recellularization and retransplantation.
Collapse
Affiliation(s)
- Marcin Morawski
- Department of General, Transplant, and Liver Surgery, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.K.); (J.R.); (H.K.); (M.S.); (M.G.)
| | - Maciej Krasnodębski
- Department of General, Transplant, and Liver Surgery, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.K.); (J.R.); (H.K.); (M.S.); (M.G.)
| | - Jakub Rochoń
- Department of General, Transplant, and Liver Surgery, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.K.); (J.R.); (H.K.); (M.S.); (M.G.)
| | - Hubert Kubiszewski
- Department of General, Transplant, and Liver Surgery, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.K.); (J.R.); (H.K.); (M.S.); (M.G.)
| | - Michał Marzęcki
- Institute of Telecommunications, Warsaw University of Technology, 00-665 Warsaw, Poland; (M.M.); (D.T.); (K.M.)
| | - Dominik Topyła
- Institute of Telecommunications, Warsaw University of Technology, 00-665 Warsaw, Poland; (M.M.); (D.T.); (K.M.)
| | - Kacper Murat
- Institute of Telecommunications, Warsaw University of Technology, 00-665 Warsaw, Poland; (M.M.); (D.T.); (K.M.)
| | - Mikołaj Staszewski
- Department of General, Transplant, and Liver Surgery, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.K.); (J.R.); (H.K.); (M.S.); (M.G.)
| | - Jacek Szczytko
- Institute of Experimental Physics, Faculty of Physics, University of Warsaw, 02-093 Warsaw, Poland;
| | - Marek Maleszewski
- Department of Embryology, Institute of Developmental Biology and Biomedical Sciences, Faculty of Biology, University of Warsaw, 02-096 Warsaw, Poland;
| | - Michał Grąt
- Department of General, Transplant, and Liver Surgery, Medical University of Warsaw, 02-091 Warsaw, Poland; (M.K.); (J.R.); (H.K.); (M.S.); (M.G.)
| |
Collapse
|
16
|
Martínez-López C, Martínez-Cáceres CM, Cortina-Navarro M, Izquierdo-Rico MJ, García-Vázquez FA. Characterization of decellularized porcine oviduct- and uterine-derived scaffolds evaluated by spermatozoa-based biocompatibility and biotoxicity. Theriogenology 2025; 231:36-51. [PMID: 39405946 DOI: 10.1016/j.theriogenology.2024.10.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2024] [Revised: 10/07/2024] [Accepted: 10/07/2024] [Indexed: 11/15/2024]
Abstract
Decellularized extracellular matrix (dECM) are widely utilized in regenerative medicine and tissue engineering due to their ability to promote cell growth, proliferation, and differentiation. In reproduction, research is focused on the utilization of these scaffolds to treat pathologies causing reproductive dysfunction or to improve assisted reproduction technologies (ARTs). We developed an efficient protocol employing the immersion-agitation technique to decellularize porcine oviductal and uterine sections, comparing the efficacy of fresh versus frozen treatments. Both methods successfully generated acellular matrices with less than 3 % residual DNA, effectively preserving structural and protein integrity. Scanning and transmission electron microscopy confirmed the ultrastructural integrity, whereas Masson's Trichrome staining highlighted better collagen preservation in frozen treatments. Proteomic analysis of decellularized scaffolds revealed collagen and key macromolecules such as laminin, filamin, dermatopontin, and fibronectin, which are essential for extracellular matrix structure and cell functions such as adhesion and migration. Innovatively, we assessed the biocompatibility and cytotoxicity of the scaffolds using spermatozoa, demonstrating that thorough washing ensures the scaffold biocompatibility without compromising sperm viability or motility. Our findings not only contribute to the standardization of decellularization protocols for female reproductive organs but also emphasize the importance of evaluating sperm biocompatibility to ensure the safety of dECM scaffolds.
Collapse
Affiliation(s)
- Cristina Martínez-López
- Departamento de Fisiología, Facultad de Veterinaria, Universidad de Murcia, 30100, Murcia, Spain; Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Murcia, 30120, Murcia, Spain.
| | - Carlos Manuel Martínez-Cáceres
- Instituto Murciano de Investigación Biosanitaria Pascual Parrilla (IMIB), Campus de Ciencias de la Salud, 30120, Murcia, Spain.
| | - María Cortina-Navarro
- Departamento de Fisiología, Facultad de Veterinaria, Universidad de Murcia, 30100, Murcia, Spain.
| | - Mª José Izquierdo-Rico
- Departamento de Biología Celular e Histología, Facultad de Medicina, Universidad de Murcia, 30120, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria Pascual Parrilla (IMIB), Campus de Ciencias de la Salud, 30120, Murcia, Spain.
| | - Francisco Alberto García-Vázquez
- Departamento de Fisiología, Facultad de Veterinaria, Universidad de Murcia, 30100, Murcia, Spain; Instituto Murciano de Investigación Biosanitaria Pascual Parrilla (IMIB), Campus de Ciencias de la Salud, 30120, Murcia, Spain.
| |
Collapse
|
17
|
Brouki Milan P, Masoumi F, Biazar E, Zare Jalise S, Mehrabi A. Exploiting the Potential of Decellularized Extracellular Matrix (ECM) in Tissue Engineering: A Review Study. Macromol Biosci 2025; 25:e2400322. [PMID: 39412772 DOI: 10.1002/mabi.202400322] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/03/2024] [Indexed: 01/14/2025]
Abstract
While significant progress has been made in creating polymeric structures for tissue engineering, the therapeutic application of these scaffolds remains challenging owing to the intricate nature of replicating the conditions of native organs and tissues. The use of human-derived biomaterials for therapeutic purposes closely imitates the properties of natural tissue, thereby assisting in tissue regeneration. Decellularized extracellular matrix (dECM) scaffolds derived from natural tissues have become popular because of their unique biomimetic properties. These dECM scaffolds can enhance the body's ability to heal itself or be used to generate new tissues for restoration, expanding beyond traditional tissue transfers and transplants. Enhanced knowledge of how ECM scaffold materials affect the microenvironment at the injury site is expected to improve clinical outcomes. In this review, recent advancements in dECM scaffolds are explored and relevant perspectives are offered, highlighting the development and application of these scaffolds in tissue engineering for various organs, such as the skin, nerve, bone, heart, liver, lung, and kidney.
Collapse
Affiliation(s)
- Peiman Brouki Milan
- Department of Tissue Engineering & Regenerative Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, 144-961-4535, Iran
| | - Farimah Masoumi
- School of Medicine, Tonekabon Branch, Islamic Azad University, Tonekabon, 468-416-1167, Iran
| | - Esmaeil Biazar
- Department of Biomedical Engineering, Tonekabon Branch, Islamic Azad University, Tonekabon, 468-416-1167, Iran
| | - Saeedeh Zare Jalise
- Department of Tissue Engineering and Applied Cell Sciences, School of Medicine, Qom University of Medical Sciences, Qom, 371-364-9373, Iran
| | - Arezou Mehrabi
- School of Medicine, Tonekabon Branch, Islamic Azad University, Tonekabon, 468-416-1167, Iran
| |
Collapse
|
18
|
Torkashvand M, Rezakhani L, Habibi Z, Mikaeili A, Rahmati S. Innovative approaches in lung tissue engineering: the role of exosome-loaded bioscaffolds in regenerative medicine. Front Bioeng Biotechnol 2024; 12:1502155. [PMID: 39758953 PMCID: PMC11695380 DOI: 10.3389/fbioe.2024.1502155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 12/04/2024] [Indexed: 01/07/2025] Open
Abstract
Lung diseases account for over four million premature deaths every year, and experts predict that this number will increase in the future. The top cause of death globally is diseases which include conditions like lung cancer asthma and COPD. Treating severe acute lung injury is a complex task because lungs struggle to heal themselves in the presence of swelling inflammation and scarring caused by damage, to the lung tissues. Though achieving lung regeneration, in controlled environments is still an ambition; ongoing studies are concentrating on notable progress, in the field of lung tissue engineering and methods for repairing lung damage. This review delves into methods, for regenerating lungs with a focus on exosome carry bioscaffolds and mesenchymal stem cells among others. It talks about how these new techniques can help repair lung tissue and improve lung function in cases of damage. Also noted is the significance of ex vivo lung perfusion (EVLP), for rejuvenating donor lungs and the healing properties of exosomes in supporting lung regeneration.
Collapse
Affiliation(s)
| | - Leila Rezakhani
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zahra Habibi
- Clinical Research Development Unit, Hajar Hospital, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Abdolhamid Mikaeili
- Medical Biology Research Center, Health Technology Institute, University of Medical Sciences, Kermanshah, Iran
| | - Shima Rahmati
- Cancer Research Center, Shahrekord University of Medical Sciences, Shahrekord, Iran
| |
Collapse
|
19
|
He X, Good A, Kalou W, Ahmad W, Dutta S, Chen S, Lin CN, Chella Krishnan K, Fan Y, Huang W, Liang J, Wang Y. Current Advances and Future Directions of Pluripotent Stem Cells-Derived Engineered Heart Tissue for Treatment of Cardiovascular Diseases. Cells 2024; 13:2098. [PMID: 39768189 PMCID: PMC11674482 DOI: 10.3390/cells13242098] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 12/11/2024] [Accepted: 12/17/2024] [Indexed: 01/11/2025] Open
Abstract
Cardiovascular diseases resulting from myocardial infarction (MI) remain a leading cause of death worldwide, imposing a substantial burden on global health systems. Current MI treatments, primarily pharmacological and surgical, do not regenerate lost myocardium, leaving patients at high risk for heart failure. Engineered heart tissue (EHT) offers a promising solution for MI and related cardiac conditions by replenishing myocardial loss. However, challenges like immune rejection, inadequate vascularization, limited mechanical strength, and incomplete tissue maturation hinder clinical application. The discovery of human-induced pluripotent stem cells (hiPSCs) has transformed the EHT field, enabling new bioengineering innovations. This review explores recent advancements and future directions in hiPSC-derived EHTs, focusing on innovative materials and fabrication methods like bioprinting and decellularization, and assessing their therapeutic potential through preclinical and clinical studies. Achieving functional integration of EHTs in the heart remains challenging due to the need for synchronized contraction, sufficient vascularization, and mechanical compatibility. Solutions such as genome editing, personalized medicine, and AI technologies offer promising strategies to address these translational barriers. Beyond MI, EHTs also show potential in treating ischemic cardiomyopathy, heart valve engineering, and drug screening, underscoring their promise in cardiovascular regenerative medicine.
Collapse
Affiliation(s)
- Xingyu He
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA; (X.H.)
| | - Angela Good
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA; (X.H.)
| | - Wael Kalou
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA; (X.H.)
| | - Waqas Ahmad
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA; (X.H.)
| | - Suchandrima Dutta
- Department of Internal Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Sophie Chen
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA; (X.H.)
| | - Charles Noah Lin
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA; (X.H.)
| | - Karthickeyan Chella Krishnan
- Department of Pharmacology and Systems Physiology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Yanbo Fan
- Department of Cancer Biology, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Wei Huang
- Department of Internal Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA
| | - Jialiang Liang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA; (X.H.)
| | - Yigang Wang
- Department of Pathology and Laboratory Medicine, College of Medicine, University of Cincinnati, Cincinnati, OH 45267, USA; (X.H.)
| |
Collapse
|
20
|
Asadian E, Abbaszadeh S, Ghorbani-Bidkorpeh F, Rezaei S, Xiao B, Santos HA, Shahbazi MA. Hijacking plant skeletons for biomedical applications: from regenerative medicine and drug delivery to biosensing. Biomater Sci 2024; 13:9-92. [PMID: 39534968 DOI: 10.1039/d4bm00982g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
The field of biomedical engineering continually seeks innovative technologies to address complex healthcare challenges, ranging from tissue regeneration to drug delivery and biosensing. Plant skeletons offer promising opportunities for these applications due to their unique hierarchical structures, desirable porosity, inherent biocompatibility, and adjustable mechanical properties. This review comprehensively discusses chemical principles underlying the utilization of plant-based scaffolds in biomedical engineering. Highlighting their structural integrity, tunable properties, and possibility of chemical modification, the review explores diverse preparation strategies to tailor plant skeleton properties for bone, neural, cardiovascular, skeletal muscle, and tendon tissue engineering. Such applications stem from the cellulosic three-dimensional structure of different parts of plants, which can mimic the complexity of native tissues and extracellular matrices, providing an ideal environment for cell adhesion, proliferation, and differentiation. We also discuss the application of plant skeletons as carriers for drug delivery due to their structural diversity and versatility in encapsulating and releasing therapeutic agents with controlled kinetics. Furthermore, we present the emerging role played by plant-derived materials in biosensor development for diagnostic and monitoring purposes. Challenges and future directions in the field are also discussed, offering insights into the opportunities for future translation of sustainable plant-based technologies to address critical healthcare needs.
Collapse
Affiliation(s)
- Elham Asadian
- Medical Nanotechnology and Tissue Engineering Research Center, Shahid Beheshti University of Medical Sciences, 19689-17313, Tehran, Iran
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, 19689-17313, Tehran, Iran
| | - Samin Abbaszadeh
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Fatemeh Ghorbani-Bidkorpeh
- Department of Pharmaceutics and Pharmaceutical Nanotechnology, School of Pharmacy, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Saman Rezaei
- Department of Pharmacology and Toxicology, School of Pharmacy, Urmia University of Medical Sciences, Urmia, Iran
| | - Bo Xiao
- Personalized Drug Therapy Key Laboratory of Sichuan Province, Department of Pharmacy, Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610072, China.
| | - Hélder A Santos
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands.
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, University of Helsinki, Helsinki FI-00014, Finland.
| | - Mohammad-Ali Shahbazi
- Department of Biomaterials and Biomedical Technology, The Personalized Medicine Research Institute (PRECISION), University Medical Center Groningen, University of Groningen, Antonius Deusinglaan 1, 9713 AV Groningen, Netherlands.
| |
Collapse
|
21
|
Jiang Y, Jin L, Liu W, Liu H, Liu X, Tan Z. Construction of 3D tumor in vitro models with an immune microenvironment exhibiting similar tumor properties and biomimetic physiological functionality. Biomater Sci 2024; 13:223-235. [PMID: 39526532 DOI: 10.1039/d4bm00754a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2024]
Abstract
Tumors pose a serious threat to people's lives and health, and the complex tumor microenvironment is the biggest obstacle to their treatment. In contrast to the basic protein matrices typically employed in 2D or 3D cell culture systems, decellularized extracellular matrix (dECM) can create complex microenvironments. In this study, a combination of physicochemical methods was established to obtain liver decellularized extracellular matrix scaffolds (dLECMs) to provide mechanical support and cell adhesion sites. By co-culturing tumor cells, tumor-associated stromal cells and immune cells, an in vitro 3D tumor model with a biomimetic immune microenvironment was constructed. By utilizing microenvironment data obtained from human liver tumor tissues and refining the double seeding modeling process, 3D in vitro liver tumor-like tissues with a tumor immune microenvironment (TIME) were obtained and designated as reconstructed human liver cancer (RHLC). These tissues demonstrated similar tumor characteristics and exhibited satisfactory physiological functionality. The results of metabolic characterisation and mouse tumorigenicity testing verified that the constructed RHLC significantly increased in vitro drug resistance while also closely mimicking in vivo tissue metabolism. This opens up new possibilities for creating effective in vitro models for screening chemotherapy drugs.
Collapse
Affiliation(s)
- Yuhong Jiang
- College of Biology, Hunan University, Changsha, 410082, China.
- Institute of Shenzhen, Hunan University Shenzhen, 518000, China
| | - Lijuan Jin
- Greater Bay Area Institute for Innovation, Hunan University, Guangzhou, 511300, China
| | - Wenyu Liu
- College of Biology, Hunan University, Changsha, 410082, China.
| | - Hui Liu
- College of Biology, Hunan University, Changsha, 410082, China.
- Institute of Shenzhen, Hunan University Shenzhen, 518000, China
| | - Xiao Liu
- Greater Bay Area Institute for Innovation, Hunan University, Guangzhou, 511300, China
| | - Zhikai Tan
- College of Biology, Hunan University, Changsha, 410082, China.
- Institute of Shenzhen, Hunan University Shenzhen, 518000, China
- Greater Bay Area Institute for Innovation, Hunan University, Guangzhou, 511300, China
| |
Collapse
|
22
|
Razavi ZS, Farokhi S, Mahmoudvand G, Karimi-Rouzbahani A, Farasati-Far B, Tahmasebi-Ghorabi S, Pazoki-Toroudi H, Saadat-Fakhr M, Afkhami H. Stem cells and bio scaffolds for the treatment of cardiovascular diseases: new insights. Front Cell Dev Biol 2024; 12:1472103. [PMID: 39726717 PMCID: PMC11669526 DOI: 10.3389/fcell.2024.1472103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2024] [Accepted: 10/01/2024] [Indexed: 12/28/2024] Open
Abstract
Mortality and morbidity from cardiovascular diseases are common worldwide. In order to improve survival and quality of life for this patient population, extensive efforts are being made to establish effective therapeutic modalities. New treatment options are needed, it seems. In addition to treating cardiovascular diseases, cell therapy is one of the most promising medical platforms. One of the most effective therapeutic approaches in this area is stem cell therapy. In stem cell biology, multipotent stem cells and pluripotent stem cells are divided into two types. There is evidence that stem cell therapy could be used as a therapeutic approach for cardiovascular diseases based on multiple lines of evidence. The effectiveness of stem cell therapies in humans has been studied in several clinical trials. In spite of the challenges associated with stem cell therapy, it appears that resolving them may lead to stem cells being used in cardiovascular disease patients. This may be an effective therapeutic approach. By mounting these stem cells on biological scaffolds, their effect can be enhanced.
Collapse
Affiliation(s)
- Zahra Sadat Razavi
- Physiology Research Center, Iran University of Medical Sciences, Tehran, Iran
| | - Simin Farokhi
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Golnaz Mahmoudvand
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Arian Karimi-Rouzbahani
- Student Research Committee, USERN Office, Lorestan University of Medical Sciences, Khorramabad, Iran
| | - Bahareh Farasati-Far
- Department of Chemistry, Iran University of Science and Technology, Tehran, Iran
| | - Samaneh Tahmasebi-Ghorabi
- Master of Health Education, Research Expert, Clinical Research Development Unit, Emam Khomeini Hospital, Ilam University of Medical Sciences, Ilam, Iran
| | | | - Masoud Saadat-Fakhr
- Faculty of Medicine, Tehran Medical Sciences Branch, Islamic Azad University, Tehran, Iran
| | - Hamed Afkhami
- Cellular and Molecular Research Center, Qom University of Medical Sciences, Qom, Iran
- Nervous System Stem Cells Research Center, Semnan University of Medical Sciences, Semnan, Iran
- Department of Medical Microbiology, Faculty of Medicine, Shahed University, Tehran, Iran
| |
Collapse
|
23
|
Ozudogru E, Kurt T, Derkus B, Cengiz U, Arslan YE. Supercritical CO 2-Mediated Decellularization of Bovine Spinal Cord Meninges: A Comparative Study for Decellularization Performance. ACS OMEGA 2024; 9:48781-48790. [PMID: 39676980 PMCID: PMC11635505 DOI: 10.1021/acsomega.4c08684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 11/07/2024] [Accepted: 11/15/2024] [Indexed: 12/17/2024]
Abstract
The extracellular matrix (ECM) of spinal meninge tissue closely resembles the wealthy ECM content of the brain and spinal cord. The ECM is typically acquired through the process of decellularizing tissues. Nevertheless, the decellularization process of the brain and spinal cord is challenging due to their high-fat content, in contrast to the spinal meninges. Hence, bovine spinal cord meninges offer a promising source to produce ECM-based scaffolds, thanks to their abundance, accessibility, and ease of decellularization for neural tissue engineering. However, most decellularization techniques involve disruptive chemicals and repetitive rinsing processes, which could lead to drastic modifications in the tissue ultrastructure and a loss of mechanical stability. Over the past decade, supercritical fluid technology has experienced considerable advancements in fabricating biomaterials with its applications spreading out to tissue engineering to tackle the complications mentioned above. Supercritical carbon-dioxide (scCO2)-based decellularization procedures especially offer a significant advantage over classical decellularization techniques, enabling the preservation of extracellular matrix components and structures. In this study, we decellularized the bovine spinal cord meninges by seven different methods. To identify the most effective approach, the decellularized matrices were characterized by dsDNA, collagen, and glycosaminoglycan contents and histological analyses. Moreover, the mechanical properties of the hydrogels produced from the decellularized matrices were evaluated. The novel scCO2-based treatment was completed in a shorter time than the conventional method (3 versus 7 days) while maintaining the structural and mechanical integrity of the tissue. Additionally, all hydrogels derived from scCO2-decellularized matrices demonstrated high cell viability and biocompatibility in a cell culture. The current study suggests a rapid, effective, and detergent-free scCO2-assisting decellularization protocol for clinical tissue engineering applications.
Collapse
Affiliation(s)
- Eren Ozudogru
- Regenerative
Biomaterials Laboratory, Department of Bioengineering, Faculty of
Engineering, Canakkale Onsekiz Mart University, Canakkale 17100, Turkey
| | - Tugce Kurt
- Regenerative
Biomaterials Laboratory, Department of Bioengineering, Faculty of
Engineering, Canakkale Onsekiz Mart University, Canakkale 17100, Turkey
| | - Burak Derkus
- Stem Cell
Research Laboratory, Department of Chemistry, Faculty of Science, Ankara University, Ankara 06560, Turkey
| | - Ugur Cengiz
- Surface Science
Research Laboratory, Department of Chemical Engineering, Faculty of
Engineering, Canakkale Onsekiz Mart University, Canakkale 17020, Turkey
| | - Yavuz Emre Arslan
- Regenerative
Biomaterials Laboratory, Department of Bioengineering, Faculty of
Engineering, Canakkale Onsekiz Mart University, Canakkale 17100, Turkey
| |
Collapse
|
24
|
Blazeski A, Garcia-Cardena G, Kamm RD. Advancing Cardiac Organoid Engineering Through Application of Biophysical Forces. IEEE Rev Biomed Eng 2024; PP:211-230. [PMID: 40030454 DOI: 10.1109/rbme.2024.3514378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
Cardiac organoids represent an important bioengineering opportunity in the development of models to study human heart pathophysiology. By incorporating multiple cardiac cell types in three-dimensional culture and developmentally-guided biochemical signaling, cardiac organoids recapitulate numerous features of heart tissue. However, cardiac tissue also experiences a variety of mechanical forces as the heart develops and over the course of each contraction cycle. It is now clear that these forces impact cellular specification, phenotype, and function, and should be incorporated into the engineering of cardiac organoids in order to generate better models. In this review, we discuss strategies for engineering cardiac organoids and report the effects of organoid design on the function of cardiac cells. We then discuss the mechanical environment of the heart, including forces arising from tissue elasticity, contraction, blood flow, and stretch, and report on efforts to mimic these biophysical cues in cardiac organoids. Finally, we review emerging areas of cardiac organoid research, for the study of cardiac development, the formation of multi-organ models, and the simulation of the effects of spaceflight on cardiac tissue and consider how these investigations might benefit from the inclusion of mechanical cues.
Collapse
|
25
|
Lunde IG, Rypdal KB, Van Linthout S, Diez J, González A. Myocardial fibrosis from the perspective of the extracellular matrix: Mechanisms to clinical impact. Matrix Biol 2024; 134:1-22. [PMID: 39214156 DOI: 10.1016/j.matbio.2024.08.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 08/08/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Fibrosis is defined by the excessive accumulation of extracellular matrix (ECM) and constitutes a central pathophysiological process that underlies tissue dysfunction, across organs, in multiple chronic diseases and during aging. Myocardial fibrosis is a key contributor to dysfunction and failure in numerous diseases of the heart and is a strong predictor of poor clinical outcome and mortality. The excess structural and matricellular ECM proteins deposited by cardiac fibroblasts, is found between cardiomyocytes (interstitial fibrosis), in focal areas where cardiomyocytes have died (replacement fibrosis), and around vessels (perivascular fibrosis). Although myocardial fibrosis has important clinical prognostic value, access to cardiac tissue biopsies for histological evaluation is limited. Despite challenges with sensitivity and specificity, cardiac magnetic resonance imaging (CMR) is the most applicable diagnostic tool in the clinic, and the scientific community is currently actively searching for blood biomarkers reflecting myocardial fibrosis, to complement the imaging techniques. The lack of mechanistic insights into specific pro- and anti-fibrotic molecular pathways has hampered the development of effective treatments to prevent or reverse myocardial fibrosis. Development and implementation of anti-fibrotic therapies is expected to improve patient outcomes and is an urgent medical need. Here, we discuss the importance of the ECM in the heart, the central role of fibrosis in heart disease, and mechanistic pathways likely to impact clinical practice with regards to diagnostics of myocardial fibrosis, risk stratification of patients, and anti-fibrotic therapy.
Collapse
Affiliation(s)
- Ida G Lunde
- Oslo Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Oslo, Norway; KG Jebsen Center for Cardiac Biomarkers, Campus Ahus, University of Oslo, Oslo, Norway.
| | - Karoline B Rypdal
- Oslo Center for Clinical Heart Research, Department of Cardiology, Oslo University Hospital Ullevaal, Oslo, Norway; KG Jebsen Center for Cardiac Biomarkers, Campus Ahus, University of Oslo, Oslo, Norway
| | - Sophie Van Linthout
- Berlin Institute of Health at Charité - Universitätsmedizin Berlin, BIH Center for Regenerative Therapies (BCRT), Berlin, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Berlin, Berlin, Germany
| | - Javier Diez
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Department of Cardiology, Clínica Universidad de Navarra and IdiSNA Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| | - Arantxa González
- Program of Cardiovascular Diseases, CIMA Universidad de Navarra, Department of Cardiology, Clínica Universidad de Navarra and IdiSNA Pamplona, Spain; CIBERCV, Carlos III Institute of Health, Madrid, Spain
| |
Collapse
|
26
|
Tabatabai TS, Salehi M, Rezakhani L, Arabpour Z, Djalilian AR, Alizadeh M. Decellularization of various tissues and organs through chemical methods. Tissue Cell 2024; 91:102573. [PMID: 39393204 PMCID: PMC11993266 DOI: 10.1016/j.tice.2024.102573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 09/20/2024] [Accepted: 09/23/2024] [Indexed: 10/13/2024]
Abstract
Due to the increase in demand for donor organs and tissues during the past 20 years, new approaches have been created. These methods include, for example, tissue engineering in vitro and the production of regenerative biomaterials for transplantation. Applying the natural extracellular matrix (ECM) as a bioactive biomaterial for clinical applications is a unique approach known as decellularization technology. Decellularization is the process of eliminating cells from an extracellular matrix while preserving its natural components including its structural and functional proteins and glycosaminoglycan. This can be achieved by physical, chemical, or biological processes. A naturally formed three-dimensional structure with a biocompatible and regenerative structure is the result of the decellularization process. Decreasing the biological factors and antigens at the transplant site reduces the risk of adverse effects including inflammatory responses and immunological rejection. Regenerative medicine and tissue engineering applications can benefit from the use of decellularization, a promising approach that provides a biomaterial that preserves its extracellular matrix.
Collapse
Affiliation(s)
- Tayebeh Sadat Tabatabai
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Majid Salehi
- Tissue Engineering and Stem Cells Research Center, Shahroud University of Medical Sciences, Shahroud, Iran; Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Leila Rezakhani
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran; Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Zohreh Arabpour
- Department of Ophthalmology and Visual Sciences, University of Illinois, Chicago, IL 60612, USA
| | - Ali R Djalilian
- Department of Ophthalmology and Visual Sciences, University of Illinois, Chicago, IL 60612, USA
| | - Morteza Alizadeh
- Department of Tissue Engineering and Biomaterials, School of Advanced Medical Sciences and Technologies, Hamadan University of Medical Sciences, Hamadan, Iran.
| |
Collapse
|
27
|
Rahaman MS, Arin A, Farwa U, Park M, Bae SH, Lee BT. ECM derivatized alginate augmenting bio-functionalities of lyophilized mat for skin and liver wound treatment. Biomaterials 2024; 311:122698. [PMID: 38968688 DOI: 10.1016/j.biomaterials.2024.122698] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 06/27/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024]
Abstract
Peptides and molecular residues sourced from the fragmentation of the extracellular matrix (ECM) can exacerbate a plethora of cellular functions. We selected a natural ECM-derived complex peptide mixture to functionalize sodium alginate. Three alginate derivatives (sodium alginate conjugated with ECM) SALE-1, SALE-2, and SALE-3 were synthesized using the lowest (10 % w/w), moderate (50 % w/w), and highest (100 % w/w) concentrations of ECM. Thereafter, they were used to fabricate three groups of mat scaffolds EMAT-1 (ECM derivatized alginate thrombin-mat), EMAT-2, and EMAT-3, respectively by the freeze-drying process. To enhance the hemostatic activity, thrombin was loaded onto the scaffolds. Another group, AT, without any derivatized alginate was additionally included in order to comparative analysis. Physical characteristics revealed that the porous mat scaffold showed enhancement in degradation and swelling ability with the increase in ECM content. The higher cell proliferation, migration, and cell viability were noticed in the higher ECM-containing samples EMAT-2 and EMAT-3. In vivo studies using rodent hepatic and rabbit ear models were carried out to ensure the hemostatic ability of the scaffolds. EMAT-2 and EMAT-3 demonstrate excellent liver regeneration ability in rat models. Moreover, the rat cutaneous wound model depicted that EMAT-3 dramatically elevated the skin's healing ability, thereby rendering it an excellent candidate for future clinical application in wound healing.
Collapse
Affiliation(s)
- Md Sohanur Rahaman
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Asuva Arin
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Ume Farwa
- Institute of Tissue Regeneration, Soonchunhyang University, Cheonan-31151, Republic of Korea
| | - Myeongki Park
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea
| | - Sang Ho Bae
- Department of Surgery, Soonchunhyang University Cheonan Hospital, Cheonan 31151, Republic of Korea
| | - Byong-Taek Lee
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, 31151, Republic of Korea; Institute of Tissue Regeneration, Soonchunhyang University, Cheonan-31151, Republic of Korea.
| |
Collapse
|
28
|
Luo X, Pang Z, Li J, Anh M, Kim BS, Gao G. Bioengineered human arterial equivalent and its applications from vascular graft to in vitro disease modeling. iScience 2024; 27:111215. [PMID: 39555400 PMCID: PMC11565542 DOI: 10.1016/j.isci.2024.111215] [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] [Indexed: 11/19/2024] Open
Abstract
Arterial disorders such as atherosclerosis, thrombosis, and aneurysm pose significant health risks, necessitating advanced interventions. Despite progress in artificial blood vessels and animal models aimed at understanding pathogenesis and developing therapies, limitations in graft functionality and species discrepancies restrict their clinical and research utility. Addressing these issues, bioengineered arterial equivalents (AEs) with enhanced vascular functions have been developed, incorporating innovative technologies that improve clinical outcomes and enhance disease progression modeling. This review offers a comprehensive overview of recent advancements in bioengineered AEs, systematically summarizing the bioengineered technologies used to construct these AEs, and discussing their implications for clinical application and pathogenesis understanding. Highlighting current breakthroughs and future perspectives, this review aims to inform and inspire ongoing research in the field, potentially transforming vascular medicine and offering new avenues for preclinical and clinical advances.
Collapse
Affiliation(s)
- Xi Luo
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Zherui Pang
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Jinhua Li
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- School of Medical Technology, Beijing Institute of Technology, Zhengzhou Academy of Intelligent Technology, Zhengzhou 450000, China
- Beijing Institute of Technology, Zhuhai, Beijing Institute of Technology, Zhuhai 519088, China
| | - Minjun Anh
- Medical Research Institute, Pusan National University, Yangsan 50612, Republic of Korea
| | - Byoung Soo Kim
- Medical Research Institute, Pusan National University, Yangsan 50612, Republic of Korea
- School of Biomedical Convergence Engineering, Pusan National University, Yangsan 50612, Republic of Korea
| | - Ge Gao
- School of Medical Technology, Beijing Institute of Technology, Beijing 100081, China
- School of Medical Technology, Beijing Institute of Technology, Zhengzhou Academy of Intelligent Technology, Zhengzhou 450000, China
| |
Collapse
|
29
|
Fazel Anvari Yazdi A, Tahermanesh K, Ejlali M, Babaei-Ghazvini A, Acharya B, Badea I, MacPhee DJ, Chen X. Comparative analysis of porcine-uterine decellularization for bioactive-molecule preservation and DNA removal. Front Bioeng Biotechnol 2024; 12:1418034. [PMID: 39416283 PMCID: PMC11480021 DOI: 10.3389/fbioe.2024.1418034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Accepted: 08/27/2024] [Indexed: 10/19/2024] Open
Abstract
Introduction Decellularized uterine extracellular matrix has emerged as a pivotal focus in the realm of biomaterials, offering a promising source in uterine tissue regeneration, research on disease diagnosis and treatments, and ultimately uterine transplantation. In this study, we examined various protocols for decellularizing porcine uterine tissues, aimed to unravel the intricate dynamics of DNA removal, bioactive molecules preservation, and microstructural alterations. Methods Porcine uterine tissues were treated with 6 different, yet rigorously selected and designed, protocols with sodium dodecyl sulfate (SDS), Triton® X-100, peracetic acid + ethanol, and DNase I. After decellularization, we examined DNA quantification, histological staining (H&E and DAPI), glycosaminoglycans (GAG) assay, scanning electron microscopy (SEM), Fourier-transform infrared spectroscopy (FT-IR), X-ray photoelectron spectroscopy (XPS), and Thermogravimetric Analysis (TGA). Results A comparative analysis among all 6 protocols was conducted with the results demonstrating that all protocols achieved decellularization; while 0.1% SDS + 1% Triton® X-100, coupled with agitation, demonstrated the highest efficiency in DNA removal. Also, it was found that DNase I played a key role in enhancing the efficiency of the decellularization process by underscoring its significance in digesting cellular contents and eliminating cell debris by 99.79% (19.63 ± 3.92 ng/mg dry weight). Conclusions Our findings enhance the nuanced understanding of DNA removal, GAG preservation, microstructural alteration, and protein decomposition in decellularized uterine extracellular matrix, while highlighting the importance of decellularization protocols designed for intended applications. This study along with our findings represents meaningful progress for advancing the field of uterine transplantation and related tissue engineering/regenerative medicine.
Collapse
Affiliation(s)
| | - Kobra Tahermanesh
- Department of Obstetrics and Gynecology, School of Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Maryam Ejlali
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - Amin Babaei-Ghazvini
- Department of Chemical and Biological Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - Bishnu Acharya
- Department of Chemical and Biological Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| | - Ildiko Badea
- College of Pharmacy and Nutrition, University of Saskatchewan, Saskatoon, SK, Canada
| | - Daniel J. MacPhee
- Department of Veterinary Biomedical Sciences, Western College of Veterinary Medicine, University of Saskatchewan, Saskatoon, SK, Canada
| | - Xiongbiao Chen
- Division of Biomedical Engineering, University of Saskatchewan, Saskatoon, Canada
- Department of Mechanical Engineering, University of Saskatchewan, Saskatoon, SK, Canada
| |
Collapse
|
30
|
Shang L, Wang S, Mao Y. Recent advances in plant-derived polysaccharide scaffolds in tissue engineering: A review. Int J Biol Macromol 2024; 277:133830. [PMID: 39002914 DOI: 10.1016/j.ijbiomac.2024.133830] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 06/13/2024] [Accepted: 07/10/2024] [Indexed: 07/15/2024]
Abstract
As a natural three-dimensional biopolymer, decellularized plant-derived scaffolds usually comprise various polysaccharides, mostly cellulose, pectin, and hemicellulose. They are characterized by natural biocompatibility and porous structures. The emergence of decellularized purified polysaccharide scaffolds provides an attractive method to overcome the challenges associated with nutrient delivery and biocompatibility, as they serve as optimal non-immune environments for stem cell adhesion and proliferation. To date, limited corresponding literature is available to systemically summarize the development and potential of these scaffolds in tissue engineering. Therefore, the current review summarized the biomimetic properties of plant-derived polysaccharide scaffolds and the latest progress in tissue engineering applications. This review first discusses the advantages of decellularized plant-derived polysaccharide scaffolds by briefly introducing their features and current limitations in clinical applications. Subsequently, the latest progress in emerging applications of regenerative biomaterials is reviewed, followed by a discussion of the studies on the interactions of biomaterials with cells and tissues. Finally, challenges in obtaining reliable scaffolds and possible future directions are discussed.
Collapse
Affiliation(s)
- Lijun Shang
- School of Life Sciences, Bengbu Medical University, Bengbu, China
| | - Shan Wang
- School of Life Sciences, Bengbu Medical University, Bengbu, China
| | - Yingji Mao
- School of Life Sciences, Bengbu Medical University, Bengbu, China.
| |
Collapse
|
31
|
Evrard R, Manon J, Maistriaux L, Fievé L, Darius T, Cornu O, Lengelé B, Schubert T. Enhancing the biological integration of massive bone allografts: A porcine preclinical in vivo pilot-study. Bone 2024; 187:117213. [PMID: 39084545 DOI: 10.1016/j.bone.2024.117213] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 07/02/2024] [Accepted: 07/23/2024] [Indexed: 08/02/2024]
Abstract
Critical bone loss can have several origins: infections, tumors or trauma. Therefore, massive bone allograft can be a solution for limb salvage. Such a biological reconstruction should have the ideal biomechanical qualities. However, their complication rate remains too high. Perfusion-decellularization of massive allografts could promote the vitality of these grafts, thereby improving their integration and bone remodeling. Three perfusion-decellularized massive bone allografts were compared to 3 fresh frozen massive bone allografts in a preclinical in vivo porcine study using an orthopedic surgery model. Three pigs each underwent a critical diaphyseal femoral defects followed by an allogeneic intercalary femoral graft on their both femurs (one decellularized and one conventional fresh frozen as "native") to reconstruct the defect. Clinical imaging was performed over 3 months of follow-up. The grafts were then explanted and examined by non-decalcified histology, fluoroscopic microscopy and immunohistochemistry. Bone consolidation was achieved in both groups at the same time. However, the volume of bone callus appeared to be greater in the decellularized group. Histology demonstrated a superior bone remodeling in the decellularized group, with a higher number of osteoclasts (p < 0.001) and larger areas of osteoid matrix and newly formed bone as compared to the "native" group. Immunohistochemistry showed a superior vitality and remodeling in both the cortical and medullary cavities for osteocalcin (p < 0.001), Ki67 (p < 0.001), CD3 (p < 0.001) and α-SMA (p < 0.001) as compared the "native" group. Three months after implantation, the decellularized grafts were proven to be biologically more active compared to native grafts. Fluoroscopic microscopy revealed more ossification fronts in the depth of the decellularized grafts (p = 0.021). This pilot study provides the first in vivo demonstration on the enhanced biological capacities of massive bone allograft decellularized by perfusion as compared to conventional massive bone allografts.
Collapse
Affiliation(s)
- Robin Evrard
- Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain, Avenue E. Mounier, 52-B1.52.04 - 1200, Bruxelles, Belgium; Institut de Recherche Expérimentale et Clinique, Pôle Chirurgie Expérimentale et Transplantation, Université Catholique de Louvain, Avenue E. Mounier, 52-B1.52.04 - 1200, Bruxelles, Belgium; Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10-1200, Bruxelles, Belgium; Unité de Thérapie Tissulaire et Cellulaire de l'Appareil Locomoteur, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10-1200, Bruxelles, Belgium.
| | - Julie Manon
- Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain, Avenue E. Mounier, 52-B1.52.04 - 1200, Bruxelles, Belgium; Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10-1200, Bruxelles, Belgium; Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, Université Catholique de Louvain, Avenue E. Mounier, 52-B1.52.04 - 1200, Bruxelles, Belgium; Unité de Thérapie Tissulaire et Cellulaire de l'Appareil Locomoteur, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10-1200, Bruxelles, Belgium
| | - Louis Maistriaux
- Institut de Recherche Expérimentale et Clinique, Pôle Chirurgie Expérimentale et Transplantation, Université Catholique de Louvain, Avenue E. Mounier, 52-B1.52.04 - 1200, Bruxelles, Belgium; Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, Université Catholique de Louvain, Avenue E. Mounier, 52-B1.52.04 - 1200, Bruxelles, Belgium
| | - Lies Fievé
- Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, Université Catholique de Louvain, Avenue E. Mounier, 52-B1.52.04 - 1200, Bruxelles, Belgium
| | - Tom Darius
- Institut de Recherche Expérimentale et Clinique, Pôle Chirurgie Expérimentale et Transplantation, Université Catholique de Louvain, Avenue E. Mounier, 52-B1.52.04 - 1200, Bruxelles, Belgium; Département de Chirurgie, Chirurgie abdominale et unité de transplantation, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10-1200, Bruxelles, Belgium
| | - Olivier Cornu
- Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain, Avenue E. Mounier, 52-B1.52.04 - 1200, Bruxelles, Belgium; Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10-1200, Bruxelles, Belgium; Unité de Thérapie Tissulaire et Cellulaire de l'Appareil Locomoteur, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10-1200, Bruxelles, Belgium
| | - Benoit Lengelé
- Institut de Recherche Expérimentale et Clinique, Pôle Morphologie, Université Catholique de Louvain, Avenue E. Mounier, 52-B1.52.04 - 1200, Bruxelles, Belgium; Service de Chirurgie Plastique, Reconstructrice et Esthétique, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10-1200, Bruxelles, Belgium
| | - Thomas Schubert
- Institut de Recherche Expérimentale et Clinique, Neuro Musculo-Skeletal Lab, Université Catholique de Louvain, Avenue E. Mounier, 52-B1.52.04 - 1200, Bruxelles, Belgium; Service de Chirurgie Orthopédique et Traumatologique, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10-1200, Bruxelles, Belgium; Unité de Thérapie Tissulaire et Cellulaire de l'Appareil Locomoteur, Cliniques Universitaires Saint-Luc, Avenue Hippocrate 10-1200, Bruxelles, Belgium
| |
Collapse
|
32
|
Hussein KH, Ahmadzada B, Correa JC, Sultan A, Wilken S, Amiot B, Nyberg SL. Liver tissue engineering using decellularized scaffolds: Current progress, challenges, and opportunities. Bioact Mater 2024; 40:280-305. [PMID: 38973992 PMCID: PMC11226731 DOI: 10.1016/j.bioactmat.2024.06.001] [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: 04/03/2024] [Revised: 05/30/2024] [Accepted: 06/01/2024] [Indexed: 07/09/2024] Open
Abstract
Liver transplantation represents the only definitive treatment for patients with end-stage liver disease. However, the shortage of liver donors provokes a dramatic gap between available grafts and patients on the waiting list. Whole liver bioengineering, an emerging field of tissue engineering, holds great potential to overcome this gap. This approach involves two main steps; the first is liver decellularization and the second is recellularization. Liver decellularization aims to remove cellular and nuclear materials from the organ, leaving behind extracellular matrices containing different structural proteins and growth factors while retaining both the vascular and biliary networks. Recellularization involves repopulating the decellularized liver with appropriate cells, theoretically from the recipient patient, to reconstruct the parenchyma, vascular tree, and biliary network. The aim of this review is to identify the major advances in decellularization and recellularization strategies and investigate obstacles for the clinical application of bioengineered liver, including immunogenicity of the designed liver extracellular matrices, the need for standardization of scaffold fabrication techniques, selection of suitable cell sources for parenchymal repopulation, vascular, and biliary tree reconstruction. In vivo transplantation models are also summarized for evaluating the functionality of bioengineered livers. Finally, the regulatory measures and future directions for confirming the safety and efficacy of bioengineered liver are also discussed. Addressing these challenges in whole liver bioengineering may offer new solutions to meet the demand for liver transplantation and improve patient outcomes.
Collapse
Affiliation(s)
- Kamal H. Hussein
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
- Department of Surgery, Anesthesiology, and Radiology, College of Veterinary Medicine, Assiut University, Assiut, Egypt
| | - Boyukkhanim Ahmadzada
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
| | - Julio Cisneros Correa
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
| | - Ahmer Sultan
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
| | - Silvana Wilken
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
| | - Bruce Amiot
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
| | - Scott L. Nyberg
- Department of Surgery, Mayo Clinic, Rochester, MN, United States
- William J. von Liebig Center for Transplantation and Clinical Regeneration, Mayo Clinic, Rochester, MN, United States
| |
Collapse
|
33
|
Du X, Jia H, Chang Y, Zhao Y, Song J. Progress of organoid platform in cardiovascular research. Bioact Mater 2024; 40:88-103. [PMID: 38962658 PMCID: PMC11220467 DOI: 10.1016/j.bioactmat.2024.05.043] [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: 03/04/2024] [Revised: 05/28/2024] [Accepted: 05/28/2024] [Indexed: 07/05/2024] Open
Abstract
Cardiovascular disease is a significant cause of death in humans. Various models are necessary for the study of cardiovascular diseases, but once cellular and animal models have some defects, such as insufficient fidelity. As a new technology, organoid has certain advantages and has been used in many applications in the study of cardiovascular diseases. This article aims to summarize the application of organoid platforms in cardiovascular diseases, including organoid construction schemes, modeling, and application of cardiovascular organoids. Advances in cardiovascular organoid research have provided many models for different cardiovascular diseases in a variety of areas, including myocardium, blood vessels, and valves. Physiological and pathological models of different diseases, drug research models, and methods for evaluating and promoting the maturation of different kinds of organ tissues are provided for various cardiovascular diseases, including cardiomyopathy, myocardial infarction, and atherosclerosis. This article provides a comprehensive overview of the latest research progress in cardiovascular organ tissues, including construction protocols for cardiovascular organoid tissues and their evaluation system, different types of disease models, and applications of cardiovascular organoid models in various studies. The problems and possible solutions in organoid development are summarized.
Collapse
Affiliation(s)
- Xingchao Du
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Hao Jia
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Yuan Chang
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Yiqi Zhao
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| | - Jiangping Song
- Beijing Key Laboratory of Preclinical Research and Evaluation for Cardiovascular Implant Materials, Animal Experimental Centre, National Centre for Cardiovascular Disease, Department of Cardiac Surgery, Fuwai Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, Chinese Academy of Medical Science, PUMC, 167 Beilishi Road, Xicheng District, Beijing, 100037, China
| |
Collapse
|
34
|
Birla RK. State of the art in Purkinje bioengineering. Tissue Cell 2024; 90:102467. [PMID: 39053130 DOI: 10.1016/j.tice.2024.102467] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 06/09/2024] [Accepted: 07/08/2024] [Indexed: 07/27/2024]
Abstract
This review article will cover the recent developments in the new evolving field of Purkinje bioengineering and the development of human Purkinje networks. Recent work has progressed to the point of a methodological and systematic process to bioengineer Purkinje networks. This involves the development of 3D models based on human anatomy, followed by the development of tunable biomaterials, and strategies to reprogram stem cells to Purkinje cells. Subsequently, the reprogrammed cells and the biomaterials are coupled to bioengineer Purkinje networks, which are then tested using a small animal injury model. In this article, we discuss this process as a whole and then each step separately. We then describe potential applications of bioengineered Purkinje networks and challenges in the field that need to be overcome to move this field forward. Although the field of Purkinje bioengineering is new and in a state of infancy, it holds tremendous potential, both for therapeutic applications and to develop tools that can be used for disease modeling.
Collapse
Affiliation(s)
- Ravi K Birla
- Laboratory for Regenerative Tissue Repair, Texas Children's Hospital, Houston, TX, USA; Center for Congenital Cardiac Research, Texas Children's Hospital, Houston, TX, USA; Division of Congenital Heart Surgery, Texas Children's Hospital, Houston, TX, USA; Department of Surgery, Baylor College of Medicine, Houston, TX, USA; Division of Pediatric Surgery, Department of Surgery, Texas Children's Hospital, Houston, TX, USA.
| |
Collapse
|
35
|
Li C, An N, Song Q, Hu Y, Yin W, Wang Q, Le Y, Pan W, Yan X, Wang Y, Liu J. Enhancing organoid culture: harnessing the potential of decellularized extracellular matrix hydrogels for mimicking microenvironments. J Biomed Sci 2024; 31:96. [PMID: 39334251 PMCID: PMC11429032 DOI: 10.1186/s12929-024-01086-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2024] [Accepted: 09/18/2024] [Indexed: 09/30/2024] Open
Abstract
Over the past decade, organoids have emerged as a prevalent and promising research tool, mirroring the physiological architecture of the human body. However, as the field advances, the traditional use of animal or tumor-derived extracellular matrix (ECM) as scaffolds has become increasingly inadequate. This shift has led to a focus on developing synthetic scaffolds, particularly hydrogels, that more accurately mimic three-dimensional (3D) tissue structures and dynamics in vitro. The ECM-cell interaction is crucial for organoid growth, necessitating hydrogels that meet organoid-specific requirements through modifiable physical and compositional properties. Advanced composite hydrogels have been engineered to more effectively replicate in vivo conditions, offering a more accurate representation of human organs compared to traditional matrices. This review explores the evolution and current uses of decellularized ECM scaffolds, emphasizing the application of decellularized ECM hydrogels in organoid culture. It also explores the fabrication of composite hydrogels and the prospects for their future use in organoid systems.
Collapse
Affiliation(s)
- Chen Li
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Hepato-Pancreato-Biliary Center, Tsinghua University, Beijing, 102218, China
| | - Ni An
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Clinical Translational Science Center, Tsinghua University, Beijing, 102218, China
| | - Qingru Song
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Hepato-Pancreato-Biliary Center, Tsinghua University, Beijing, 102218, China
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Clinical Translational Science Center, Tsinghua University, Beijing, 102218, China
| | - Yuelei Hu
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Hepato-Pancreato-Biliary Center, Tsinghua University, Beijing, 102218, China
- Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 100084, China
| | - Wenzhen Yin
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Clinical Translational Science Center, Tsinghua University, Beijing, 102218, China
| | - Qi Wang
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Hepato-Pancreato-Biliary Center, Tsinghua University, Beijing, 102218, China
- Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 100084, China
| | - Yinpeng Le
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Hepato-Pancreato-Biliary Center, Tsinghua University, Beijing, 102218, China
- School of Materials Science and Engineering, Institute of Smart Biomedical Materials, Zhejiang Sci-Tech University, Hangzhou, 310018, China
| | - Wenting Pan
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China
| | - Xinlong Yan
- Beijing International Science and Technology Cooperation Base for Antiviral Drugs, Beijing Key Laboratory of Environmental and Viral Oncology, College of Chemistry and Life Science, Beijing University of Technology, Beijing, 100124, China.
| | - Yunfang Wang
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Hepato-Pancreato-Biliary Center, Tsinghua University, Beijing, 102218, China.
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Clinical Translational Science Center, Tsinghua University, Beijing, 102218, China.
- Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 100084, China.
| | - Juan Liu
- School of Clinical Medicine, Beijing Tsinghua Changgung Hospital, Hepato-Pancreato-Biliary Center, Tsinghua University, Beijing, 102218, China.
- Key Laboratory of Digital Intelligence Hepatology (Ministry of Education/Beijing), School of Clinical Medicine, Tsinghua University, Beijing, 100084, China.
| |
Collapse
|
36
|
Stoian A, Adil A, Biniazan F, Haykal S. Two Decades of Advances and Limitations in Organ Recellularization. Curr Issues Mol Biol 2024; 46:9179-9214. [PMID: 39194760 DOI: 10.3390/cimb46080543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 08/14/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024] Open
Abstract
The recellularization of tissues after decellularization is a relatively new technology in the field of tissue engineering (TE). Decellularization involves removing cells from a tissue or organ, leaving only the extracellular matrix (ECM). This can then be recellularized with new cells to create functional tissues or organs. The first significant mention of recellularization in decellularized tissues can be traced to research conducted in the early 2000s. One of the landmark studies in this field was published in 2008 by Ott, where researchers demonstrated the recellularization of a decellularized rat heart with cardiac cells, resulting in a functional organ capable of contraction. Since then, other important studies have been published. These studies paved the way for the widespread application of recellularization in TE, demonstrating the potential of decellularized ECM to serve as a scaffold for regenerating functional tissues. Thus, although the concept of recellularization was initially explored in previous decades, these studies from the 2000s marked a major turning point in the development and practical application of the technology for the recellularization of decellularized tissues. The article reviews the historical advances and limitations in organ recellularization in TE over the last two decades.
Collapse
Affiliation(s)
- Alina Stoian
- Latner Thoracic Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Aisha Adil
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
- Translational Biology and Engineering Program, Ted Rogers Centre for Heart Research, Toronto, ON M5G 1M1, Canada
| | - Felor Biniazan
- Latner Thoracic Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
| | - Siba Haykal
- Latner Thoracic Research Laboratories, Division of Thoracic Surgery, Toronto General Hospital Research Institute, University Health Network, Toronto, ON M5G 1L7, Canada
- Reconstructive Oncology, Division of Plastic and Reconstructive Surgery, Smilow Cancer Hospital, Yale, New Haven, CT 06519, USA
| |
Collapse
|
37
|
Shi W, Zhang Z, Wang X. The Prospect of Hepatic Decellularized Extracellular Matrix as a Bioink for Liver 3D Bioprinting. Biomolecules 2024; 14:1019. [PMID: 39199406 PMCID: PMC11352484 DOI: 10.3390/biom14081019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Revised: 07/24/2024] [Accepted: 07/26/2024] [Indexed: 09/01/2024] Open
Abstract
The incidence of liver diseases is high worldwide. Many factors can cause liver fibrosis, which in turn can lead to liver cirrhosis and even liver cancer. Due to the shortage of donor organs, immunosuppression, and other factors, only a few patients are able to undergo liver transplantation. Therefore, how to construct a bioartificial liver that can be transplanted has become a global research hotspot. With the rapid development of three-dimensional (3D) bioprinting in the field of tissue engineering and regenerative medicine, researchers have tried to use various 3D bioprinting technologies to construct bioartificial livers in vitro. In terms of the choice of bioinks, liver decellularized extracellular matrix (dECM) has many advantages over other materials for cell-laden hydrogel in 3D bioprinting. This review mainly summarizes the acquisition of liver dECM and its application in liver 3D bioprinting as a bioink with respect to availability, printability, and biocompatibility in many aspects and puts forward the current challenges and prospects.
Collapse
Affiliation(s)
- Wen Shi
- Center of 3D Printing & Organ Manufacturing, School of Intelligent Medicine, China Medical University, Shenyang 110122, China;
- Department of Ultrasound, The First Hospital of China Medical University, Shenyang 110001, China
| | - Zhe Zhang
- Department of Thoracic Surgery, The First Hospital of China Medical University, Shenyang 110001, China;
| | - Xiaohong Wang
- Center of 3D Printing & Organ Manufacturing, School of Intelligent Medicine, China Medical University, Shenyang 110122, China;
| |
Collapse
|
38
|
Zhao N, Pessell AF, Zhu N, Searson PC. Tissue-Engineered Microvessels: A Review of Current Engineering Strategies and Applications. Adv Healthc Mater 2024; 13:e2303419. [PMID: 38686434 PMCID: PMC11338730 DOI: 10.1002/adhm.202303419] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2023] [Revised: 04/10/2024] [Indexed: 05/02/2024]
Abstract
Microvessels, including arterioles, capillaries, and venules, play an important role in regulating blood flow, enabling nutrient and waste exchange, and facilitating immune surveillance. Due to their important roles in maintaining normal function in human tissues, a substantial effort has been devoted to developing tissue-engineered models to study endothelium-related biology and pathology. Various engineering strategies have been developed to recapitulate the structural, cellular, and molecular hallmarks of native human microvessels in vitro. In this review, recent progress in engineering approaches, key components, and culture platforms for tissue-engineered human microvessel models is summarized. Then, tissue-specific models, and the major applications of tissue-engineered microvessels in development, disease modeling, drug screening and delivery, and vascularization in tissue engineering, are reviewed. Finally, future research directions for the field are discussed.
Collapse
Affiliation(s)
- Nan Zhao
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Alexander F Pessell
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Ninghao Zhu
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Peter C Searson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| |
Collapse
|
39
|
Zamora Alvarado JE, McCloskey KE, Gopinathan A. Migration and proliferation drive the emergence of patterns in co-cultures of differentiating vascular progenitor cells. MATHEMATICAL BIOSCIENCES AND ENGINEERING : MBE 2024; 21:6731-6757. [PMID: 39483091 PMCID: PMC11556463 DOI: 10.3934/mbe.2024295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/03/2024]
Abstract
Vascular cells self-organize into unique structures guided by cell proliferation, migration, and/or differentiation from neighboring cells, mechanical factors, and/or soluble signals. However, the relative contribution of each of these factors remains unclear. Our objective was to develop a computational model to explore the different factors affecting the emerging micropatterns in 2D. This was accomplished by developing a stochastic on-lattice population-based model starting with vascular progenitor cells with the potential to proliferate, migrate, and/or differentiate into either endothelial cells or smooth muscle cells. The simulation results yielded patterns that were qualitatively and quantitatively consistent with experimental observations. Our results suggested that post-differentiation cell migration and proliferation when balanced could generate between 30-70% of each cell type enabling the formation of vascular patterns. Moreover, the cell-to-cell sensing could enhance the robustness of this patterning. These findings computationally supported that 2D patterning is mechanistically similar to current microfluidic platforms that take advantage of the migration-directed self-assembly of mature endothelial and mural cells to generate perfusable 3D vasculature in permissible hydrogel environments and suggest that stem or progenitor cells may not be fully necessary components in many tissue formations like those formed by vasculogenesis.
Collapse
Affiliation(s)
- Jose E. Zamora Alvarado
- School of Engineering, University of California Merced, Merced, CA 95343, USA
- Graduate Program in Materials and Biomaterials Science and Engineering, University of California Merced, Merced, CA 95343, USA
| | - Kara E. McCloskey
- School of Engineering, University of California Merced, Merced, CA 95343, USA
- Graduate Program in Materials and Biomaterials Science and Engineering, University of California Merced, Merced, CA 95343, USA
| | - Ajay Gopinathan
- Graduate Program in Materials and Biomaterials Science and Engineering, University of California Merced, Merced, CA 95343, USA
- Department of Physics, University of California Merced, Merced, CA 95343, USA
| |
Collapse
|
40
|
Capella-Monsonís H, Crum RJ, Hussey GS, Badylak SF. Advances, challenges, and future directions in the clinical translation of ECM biomaterials for regenerative medicine applications. Adv Drug Deliv Rev 2024; 211:115347. [PMID: 38844005 DOI: 10.1016/j.addr.2024.115347] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 05/29/2024] [Accepted: 06/03/2024] [Indexed: 06/11/2024]
Abstract
Extracellular Matrix (ECM) scaffolds and biomaterials have been widely used for decades across a variety of diverse clinical applications and have been implanted in millions of patients worldwide. ECM-based biomaterials have been especially successful in soft tissue repair applications but their utility in other clinical applications such as for regeneration of bone or neural tissue is less well understood. The beneficial healing outcome with the use of ECM biomaterials is the result of their biocompatibility, their biophysical properties and their ability to modify cell behavior after injury. As a consequence of successful clinical outcomes, there has been motivation for the development of next-generation formulations of ECM materials ranging from hydrogels, bioinks, powders, to whole organ or tissue scaffolds. The continued development of novel ECM formulations as well as active research interest in these materials ensures a wealth of possibilities for future clinical translation and innovation in regenerative medicine. The clinical translation of next generation formulations ECM scaffolds faces predictable challenges such as manufacturing, manageable regulatory pathways, surgical implantation, and the cost required to address these challenges. The current status of ECM-based biomaterials, including clinical translation, novel formulations and therapies currently under development, and the challenges that limit clinical translation of ECM biomaterials are reviewed herein.
Collapse
Affiliation(s)
- Héctor Capella-Monsonís
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, USA; Department of Surgery, School of Medicine, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA; Viscus Biologics LLC, 2603 Miles Road, Cleveland, OH 44128, USA
| | - Raphael J Crum
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, USA; Department of Surgery, School of Medicine, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - George S Hussey
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, USA; Department of Pathology, School of Medicine, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA
| | - Stephen F Badylak
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, 450 Technology Drive, Pittsburgh, PA 15219, USA; Department of Surgery, School of Medicine, University of Pittsburgh, 200 Lothrop Street, Pittsburgh, PA 15213, USA; Department of Bioengineering, University of Pittsburgh, 3700 O'Hara Street, Pittsburgh, PA 15261, USA.
| |
Collapse
|
41
|
Wang C, Chen Q, Wang H, Gang H, Zhou Y, Gu S, Zhang R, Xu W, Yang H. Biomechanical Scaffolds of Decellularized Heart Valves Modified by Electrospun Polylactic Acid. Appl Biochem Biotechnol 2024; 196:4256-4272. [PMID: 37922030 DOI: 10.1007/s12010-023-04756-8] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2023] [Indexed: 11/05/2023]
Abstract
Enhancing the mechanical properties and cytocompatibility of decellularized heart valves is the key to promote the application of biological heart valves. In order to further improve the mechanical properties, the electrospinning and non-woven processing methods are combined to prepare the polylactic acid (PLA)/decellularized heart valve nanofiber-reinforced sandwich structure electrospun scaffold. The effect of electrospinning time on the performance of decellularized heart valve is investigated from the aspects of morphology, mechanical properties, softness, and biocompatibility of decellularized heart valve. Results of the mechanical tests show that compared with the pure decellularized heart valve, the mechanical properties of the composite heart valve were significantly improved with the tensile strength increasing by 108% and tensile strain increased by 571% when the electrospinning time exceeded 2 h. In addition, with this electrospinning time, the composite heart valve has a certain promoting effect on the human umbilical vein endothelial cells proliferation behavior. This work provides a promising foundation for tissue heart valve reendothelialization to lay the groundwork for organoid.
Collapse
Affiliation(s)
- Chaorong Wang
- College of Materials Science and Engineering, Wuhan Textile University, No.1 Yangguang Road, Wuhan, 430200, Hubei Province, China
| | - Qingqing Chen
- College of Materials Science and Engineering, Wuhan Textile University, No.1 Yangguang Road, Wuhan, 430200, Hubei Province, China
| | - Han Wang
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan, 430200, China
- Institute for Frontier Materials, Deakin University, Geelong, Victoria, 3216, Australia
| | - Hanlin Gang
- College of Materials Science and Engineering, Wuhan Textile University, No.1 Yangguang Road, Wuhan, 430200, Hubei Province, China
| | - Yingshan Zhou
- College of Materials Science and Engineering, Wuhan Textile University, No.1 Yangguang Road, Wuhan, 430200, Hubei Province, China
| | - Shaojin Gu
- College of Materials Science and Engineering, Wuhan Textile University, No.1 Yangguang Road, Wuhan, 430200, Hubei Province, China
| | - Ruoyun Zhang
- College of Materials Science and Engineering, Wuhan Textile University, No.1 Yangguang Road, Wuhan, 430200, Hubei Province, China.
| | - Weilin Xu
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan, 430200, China
| | - Hongjun Yang
- College of Materials Science and Engineering, Wuhan Textile University, No.1 Yangguang Road, Wuhan, 430200, Hubei Province, China.
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, Wuhan Textile University, Wuhan, 430200, China.
| |
Collapse
|
42
|
Yamaguchi A, Hashimoto Y, Negishi J. Fabrication of a cell culture scaffold that mimics the composition and structure of bone marrow extracellular matrix. J Biosci Bioeng 2024; 138:83-88. [PMID: 38643031 DOI: 10.1016/j.jbiosc.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 03/26/2024] [Accepted: 03/28/2024] [Indexed: 04/22/2024]
Abstract
Cell culture models that mimic tissue environments are useful for cell and extracellular matrix (ECM) function analysis. Decellularized tissues with tissue-specific ECM are expected to be applied as cell culture scaffolds, however, it is often difficult for seeded cells to permeate their structures. In this study, we evaluated the adhesion and proliferation of mouse fibroblasts seeded onto decellularized bone marrow scaffolds that we fabricated from adult and fetal porcine. Decellularized fetal bone marrow displays more cell attachment and faster cell proliferation than decellularized adult bone marrow. Our findings suggest that decellularized fetal bone marrow is useful as a cell culture scaffold with bone marrow ECM and structure.
Collapse
Affiliation(s)
- Ayana Yamaguchi
- Department of Textile Science and Technology, Graduate School of Science and Technology, Shinshu University, 3-15-1 Tokida, Ueda, Nagano 386-8567, Japan
| | - Yoshihide Hashimoto
- Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-006, Japan
| | - Jun Negishi
- Department of Textile Science and Technology, Graduate School of Science and Technology, Shinshu University, 3-15-1 Tokida, Ueda, Nagano 386-8567, Japan; Institute of Biomaterials and Bioengineering, Tokyo Medical and Dental University, 2-3-10 Kanda-Surugadai, Chiyoda-ku, Tokyo 101-006, Japan.
| |
Collapse
|
43
|
Patel R, Patel D. Injectable Hydrogels in Cardiovascular Tissue Engineering. Polymers (Basel) 2024; 16:1878. [PMID: 39000733 PMCID: PMC11244148 DOI: 10.3390/polym16131878] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 06/28/2024] [Accepted: 06/29/2024] [Indexed: 07/17/2024] Open
Abstract
Heart problems are quite prevalent worldwide. Cardiomyocytes and stem cells are two examples of the cells and supporting matrix that are used in the integrated process of cardiac tissue regeneration. The objective is to create innovative materials that can effectively replace or repair damaged cardiac muscle. One of the most effective and appealing 3D/4D scaffolds for creating an appropriate milieu for damaged tissue growth and healing is hydrogel. In order to successfully regenerate heart tissue, bioactive and biocompatible hydrogels are required to preserve cells in the infarcted region and to bid support for the restoration of myocardial wall stress, cell survival and function. Heart tissue engineering uses a variety of hydrogels, such as natural or synthetic polymeric hydrogels. This article provides a quick overview of the various hydrogel types employed in cardiac tissue engineering. Their benefits and drawbacks are discussed. Hydrogel-based techniques for heart regeneration are also addressed, along with their clinical application and future in cardiac tissue engineering.
Collapse
Affiliation(s)
- Raj Patel
- Banas Medical College and Research Institute, Palanpur 385001, India;
| | - Dhruvi Patel
- School of Civil and Environmental Engineering, Cornell University, Ithaca, NY 14850, USA
| |
Collapse
|
44
|
Dong G, Wang J, Chen Z, Wang F, Xia B, Chen G. Regulatory effects of stress release from decellularized periosteum on proliferation, migration, and osteogenic differentiation of periosteum-derived cells. Biomater Sci 2024; 12:3360-3373. [PMID: 38771565 DOI: 10.1039/d4bm00358f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024]
Abstract
Bone injury is often associated with tears in the periosteum and changes in the internal stress microenvironment of the periosteum. In this study, we investigated the biological effects of periosteal prestress release on periosteum-derived cells (PDCs) and the potential mechanisms of endogenous stem cell recruitment. Decellularized periosteum with natural extracellular matrix (ECM) components was obtained by a combination of physical, chemical, and enzymatic decellularization. The decellularized periosteum removed immunogenicity while retaining the natural network structure and composition of the ECM. The Young's modulus has no significant difference between the periosteum before and after decellularization. The extracted PDCs were further composited with the decellularized periosteum and subjected to 20% stress release. It was found that the proliferative capacity of PDCs seeded on decellularized periosteum was significantly enhanced 6 h after stress release of the periosteum. The cell culture supernatant obtained after periosteal prestress release was able to significantly promote the migration ability of PDCs within 24 h. Enzyme-linked immunosorbnent assay (ELISA) experiments showed that the expression of stroma-derived factor-1α (SDF-1α) and vascular endothelial growth factor (VEGF) in the supernatant increased significantly after 3 h and 12 h of stress release, respectively. Furthermore, periosteal stress release promoted the high expression of osteogenic markers osteocalcin (OCN), osteopontin (OPN), and collagen type I of PDCs. The change in stress environment caused by the release of periosteal prestress was sensed by integrin β1, a mechanoreceptor on the membrane of PDCs, which further stimulated the expression of YAP in the nucleus. These investigations provided a novel method to evaluate the importance of mechanical stimulation in periosteum, which is also of great significance for the design and fabrication of artificial periosteum with mechanical regulation function.
Collapse
Affiliation(s)
- Gangli Dong
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, P. R. China.
| | - Jinsong Wang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, P. R. China.
| | - Zhongmin Chen
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, P. R. China.
| | - Fuping Wang
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, P. R. China.
| | - Bin Xia
- Engineering Research Center for Waste Oil Recovery Technology and Equipment, Ministry of Education, Chongqing Technology and Business University, Chongqing 400067, P. R. China.
| | - Guobao Chen
- School of Pharmacy and Bioengineering, Chongqing University of Technology, Chongqing 400054, P. R. China.
| |
Collapse
|
45
|
Karabıyık Acar Ö, Bozdağ G, Hacıhasanoğlu E, Tuncer AA, Aysan E, Torun Köse G. Optimizing decellularization protocols for human thyroid tissues: a step towards tissue engineering and transplantation. Biomed Mater 2024; 19:045034. [PMID: 38857607 DOI: 10.1088/1748-605x/ad565e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Accepted: 06/10/2024] [Indexed: 06/12/2024]
Abstract
Hypothyroidism is caused by insufficient stimulation or disruption of the thyroid. However, the drawbacks of thyroid transplantation have led to the search for new treatments. Decellularization allows tissue transplants to maintain their biomimetic structures while preserving cell adhesion, proliferation, and differentiation. This study aimed to decellularize human thyroid tissues using a structure-preserving optimization strategy and present preliminary data on recellularization. Nine methods were used for physical and chemical decellularization. Quantitative and immunohistochemical analyses were performed to investigate the DNA and extracellular matrix components of the tissues. Biomechanical properties were determined by compression test, and cell viability was examined after seeding MDA-T32 papillary thyroid cancer (PTC) cells onto the decellularized tissues. Decellularized tissues exhibited a notable decrease (<50 ng mg-1DNA, except for Groups 2 and 7) compared to the native thyroid tissue. Nonetheless, collagen and glycosaminoglycans were shown to be conserved in all decellularized tissues. Laminin and fibronectin were preserved at comparatively higher levels, and Young's modulus was elevated when decellularization included SDS. It was observed that the strain value in Group 1 (1.63 ± 0.14 MPa) was significantly greater than that in the decellularized tissues between Groups 2-9, ranging from 0.13 ± 0.03-0.72 ± 0.29 MPa. Finally, viability assessment demonstrated that PTC cells within the recellularized tissue groups successfully attached to the 3D scaffolds and sustained metabolic activity throughout the incubation period. We successfully established a decellularization optimization for human thyroid tissues, which has potential applications in tissue engineering and transplantation research. Our next goal is to conduct recellularization using the methods utilized in Group 1 and transplant the primary thyroid follicular cell-seeded tissues into anin vivoanimal model, particularly due to their remarkable 3D structural preservation and cell adhesion-promoting properties.
Collapse
Affiliation(s)
- Özge Karabıyık Acar
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul, Türkiye
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Türkiye
| | - Gülnihal Bozdağ
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Türkiye
| | - Ezgi Hacıhasanoğlu
- Department of Pathology, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - A Alperen Tuncer
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Türkiye
| | - Erhan Aysan
- Department of General Surgery, Faculty of Medicine, Yeditepe University, Istanbul, Türkiye
| | - Gamze Torun Köse
- Department of Genetics and Bioengineering, Faculty of Engineering, Yeditepe University, Istanbul, Türkiye
| |
Collapse
|
46
|
Wu J, Fu J. Toward developing human organs via embryo models and chimeras. Cell 2024; 187:3194-3219. [PMID: 38906095 PMCID: PMC11239105 DOI: 10.1016/j.cell.2024.05.027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2023] [Revised: 02/02/2024] [Accepted: 05/14/2024] [Indexed: 06/23/2024]
Abstract
Developing functional organs from stem cells remains a challenging goal in regenerative medicine. Existing methodologies, such as tissue engineering, bioprinting, and organoids, only offer partial solutions. This perspective focuses on two promising approaches emerging for engineering human organs from stem cells: stem cell-based embryo models and interspecies organogenesis. Both approaches exploit the premise of guiding stem cells to mimic natural development. We begin by summarizing what is known about early human development as a blueprint for recapitulating organogenesis in both embryo models and interspecies chimeras. The latest advances in both fields are discussed before highlighting the technological and knowledge gaps to be addressed before the goal of developing human organs could be achieved using the two approaches. We conclude by discussing challenges facing embryo modeling and interspecies organogenesis and outlining future prospects for advancing both fields toward the generation of human tissues and organs for basic research and translational applications.
Collapse
Affiliation(s)
- Jun Wu
- Department of Molecular Biology, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Hamon Center for Regenerative Science and Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA; Cecil H. and Ida Green Center for Reproductive Biology Sciences, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| | - Jianping Fu
- Department of Mechanical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI 48109, USA; Department of Cell & Developmental Biology, University of Michigan Medical School, Ann Arbor, MI 48109, USA.
| |
Collapse
|
47
|
Khodayari H, Khodayari S, Rezaee M, Rezaeiani S, Alipour Choshali M, Erfanian S, Muhammadnejad A, Nili F, Pourmehran Y, Pirjani R, Rajabi S, Aghdami N, Nebigil-Désaubry C, Wang K, Mahmoodzadeh H, Pahlavan S. Promotion of cardiac microtissue assembly within G-CSF-enriched collagen I-cardiogel hybrid hydrogel. Regen Biomater 2024; 11:rbae072. [PMID: 38974665 PMCID: PMC11226883 DOI: 10.1093/rb/rbae072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2024] [Revised: 05/20/2024] [Accepted: 06/10/2024] [Indexed: 07/09/2024] Open
Abstract
Tissue engineering as an interdisciplinary field of biomedical sciences has raised many hopes in the treatment of cardiovascular diseases as well as development of in vitro three-dimensional (3D) cardiac models. This study aimed to engineer a cardiac microtissue using a natural hybrid hydrogel enriched by granulocyte colony-stimulating factor (G-CSF), a bone marrow-derived growth factor. Cardiac ECM hydrogel (Cardiogel: CG) was mixed with collagen type I (ColI) to form the hybrid hydrogel, which was tested for mechanical and biological properties. Three cell types (cardiac progenitor cells, endothelial cells and cardiac fibroblasts) were co-cultured in the G-CSF-enriched hybrid hydrogel to form a 3D microtissue. ColI markedly improved the mechanical properties of CG in the hybrid form with a ratio of 1:1. The hybrid hydrogel demonstrated acceptable biocompatibility and improved retention of encapsulated human foreskin fibroblasts. Co-culture of three cell types in G-CSF enriched hybrid hydrogel, resulted in a faster 3D structure shaping and a well-cellularized microtissue with higher angiogenesis compared to growth factor-free hybrid hydrogel (control). Immunostaining confirmed the presence of CD31+ tube-like structures as well as vimentin+ cardiac fibroblasts and cTNT+ human pluripotent stem cells-derived cardiomyocytes. Bioinformatics analysis of signaling pathways related to the G-CSF receptor in cardiovascular lineage cells, identified target molecules. The in silico-identified STAT3, as one of the major molecules involved in G-CSF signaling of cardiac tissue, was upregulated in G-CSF compared to control. The G-CSF-enriched hybrid hydrogel could be a promising candidate for cardiac tissue engineering, as it facilitates tissue formation and angiogenesis.
Collapse
Affiliation(s)
- Hamid Khodayari
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 19395-4644, Iran
- Department of Developmental Biology, University of Science and Culture, Tehran 13145-871, Iran
| | - Saeed Khodayari
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran 1419733141, Iran
| | - Malihe Rezaee
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 19395-4644, Iran
| | - Siamak Rezaeiani
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 19395-4644, Iran
| | - Mahmoud Alipour Choshali
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 19395-4644, Iran
| | - Saiedeh Erfanian
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 19395-4644, Iran
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 19395-4644, Iran
| | - Ahad Muhammadnejad
- Cancer Biology Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran 1419733141, Iran
| | - Fatemeh Nili
- Department of Pathology, Cancer Institute, Imam Khomeini Hospital Complex, Tehran University of Medical Sciences, Tehran 1419733141, Iran
| | - Yasaman Pourmehran
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 19395-4644, Iran
- Department of Developmental Biology, University of Science and Culture, Tehran 13145-871, Iran
| | - Reihaneh Pirjani
- Obstetrics and Gynecology Department, Arash Women’s Hospital, Tehran University of Medical Sciences, Tehran 1653915981, Iran
| | - Sarah Rajabi
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 19395-4644, Iran
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 19395-4644, Iran
| | - Naser Aghdami
- Department of Regenerative Medicine, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, Tehran 19395-4644, Iran
| | - Canan Nebigil-Désaubry
- Institute National de le santé et de la recherce médicale, INSERM, University of Strasbourg, UMR 1260-Regenerative Nanomedicine, CRBS, Central of Research in biomedicine of Strasbourg, Strasbourg 90032, France
| | - Kai Wang
- Department of Physiology and Pathophysiology, School of Basic Medical Sciences, State Key Laboratory of Vascular Homeostasis and Remodeling, Peking University, Beijing 100191, China
| | - Habibollah Mahmoodzadeh
- Cancer Research Center, Cancer Institute of Iran, Tehran University of Medical Sciences, Tehran 1419733141, Iran
| | - Sara Pahlavan
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran 19395-4644, Iran
| |
Collapse
|
48
|
Goto T, Nakamura Y, Ito Y, Miyagawa S. Regenerative medicine in cardiovascular disease. Regen Ther 2024; 26:859-866. [PMID: 39430582 PMCID: PMC11490749 DOI: 10.1016/j.reth.2024.09.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Revised: 09/04/2024] [Accepted: 09/11/2024] [Indexed: 10/22/2024] Open
Abstract
Owing to the rapid increase in the number of people with severe heart failure, regenerative medicine is anticipated to play a role in overcoming the limitations inherent in existing surgical interventions. There are essentially two types of cardiac regenerative therapies for a failing heart. Cellular regenerative therapies using various stem cells improve the functional recovery of the heart mainly by cytokine paracrine effects. The implantation of induced pluripotent stem cell-derived cardiomyocytes can contribute not only to the inhibition of adverse heart remodeling by paracrine effects but also to the supply of newly born functional myocytes with the recipient myocardium as "mechanically working cells." Cell transplantation, including autologous myoblast transplantation, reduces heart failure exacerbations and benefits patients without the need for other treatment options. Although cellular therapy is currently the mainstream approach, it requires an in-house cell-processing center with an aseptic environment. In addition, these stem cells are usually introduced via several invasive delivery methods, including intracoronary administration, and cellular sheet implantation. Simplifying the culture methods for these cells is a crucial problem that needs to be resolved. Drug-induced regenerative therapy is another option that enhances self-endogenous regenerative systems in the human body and does not require invasive methods or cell cultures. Therefore, drug-induced regenerative therapies may overcome the disadvantages of these cellular therapies. The purpose of this report is to summarize cell transplantation therapy in the cardiovascular system and regenerative therapy for heart failure using an autologous endogenous regenerative system.
Collapse
Affiliation(s)
- Takasumi Goto
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
- Department of Cardiovascular Surgery, Toyonaka Municipal Hospital, Osaka, Japan
| | - Yuki Nakamura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, Osaka, Japan
| | - Yoshito Ito
- 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
| |
Collapse
|
49
|
Bjorgvinsdottir O, Ferguson SJ, Snorradottir BS, Gudjonsson T, Wuertz-Kozak K. The influence of physical and spatial substrate characteristics on endothelial cells. Mater Today Bio 2024; 26:101060. [PMID: 38711934 PMCID: PMC11070711 DOI: 10.1016/j.mtbio.2024.101060] [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: 12/30/2023] [Revised: 03/10/2024] [Accepted: 04/13/2024] [Indexed: 05/08/2024] Open
Abstract
Cardiovascular diseases are a main cause of death worldwide, leading to a growing demand for medical devices to treat this patient group. Central to the engineering of such devices is a good understanding of the biology and physics of cell-surface interactions. In existing blood-contacting devices, such as vascular grafts, the interaction between blood, cells, and material is one of the main limiting factors for their long-term durability. An improved understanding of the material's chemical- and physical properties as well as its structure all play a role in how endothelial cells interact with the material surface. This review provides an overview of how different surface structures influence endothelial cell responses and what is currently known about the underlying mechanisms that guide this behavior. The structures reviewed include decellularized matrices, electrospun fibers, pillars, pits, and grated surfaces.
Collapse
Affiliation(s)
- Oddny Bjorgvinsdottir
- Faculty of Pharmaceutical Sciences, University of Iceland, Hofsvallagata 53, 107 Reykjavik, Iceland
| | - Stephen J. Ferguson
- Institute for Biomechanics, ETH Zurich, Gloriastrasse 37 / 39, 8092, Zurich, Switzerland
| | | | - Thorarinn Gudjonsson
- Faculty of Medicine, University of Iceland, Vatnsmyrarvegur 16, 101 Reykjavik, Iceland
| | - Karin Wuertz-Kozak
- Department of Biomedical Engineering, Rochester Institute of Technology (RIT), 160 Lomb Memorial Drive Bldg. 73, Rochester, NY, 14623, USA
| |
Collapse
|
50
|
Shu Z, Liu E, Huang Y, Luo Q, Wang T, Li X, Mequanint K, Yang S, Xing M, Fan C. Analyzing Mushroom Structural Patterns of a Highly Compressible and Expandable Hemostatic Foam for Gastric Perforation Repair. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2306917. [PMID: 38439601 DOI: 10.1002/advs.202306917] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 12/10/2023] [Indexed: 03/06/2024]
Abstract
Nature presents the most beautiful patterns through evolving. Here, a layered porous pattern in golden ratio (0.618) is reported from a type of mushroom -Dictyophora Rubrovalvata stipe (DRS). The hierarchical structure shows a mathematical correlation with the golden ratio. This unique structure leads to superior mechanical properties. The gradient porous structure from outside to innermost endows it with asymmetrical hydrophilicity. A mathematical model is then developed to predict and apply to 3D printed structures. The mushroom is then explored to repair gastric perforation because the stomach is a continuous peristaltic organ, and the perforated site is subject to repeated mechanical movements and pressure changes. At present, endoscopic clipping is ineffective in treating ulcerative perforation with fragile surrounding tissues. Although endoscopic implant occlusion provides a new direction for the treatment of gastric ulcers, but the metal or plastic occluder needs to be removed, requiring a second intervention. Decellularized DRS (DDRS) is found with asymmetric water absorption rate, super-compressive elasticity, shape memory, and biocompatibility, making it a suitable occluder for the gastric perforation. The efficacy in blocking gastric perforation and promoting healing is confirmed by endoscopic observation and tissue analysis during a 2-month study.
Collapse
Affiliation(s)
- Zhenzhen Shu
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, NO.183, Xinqiao Street, Chongqing, 400037, China
| | - En Liu
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, NO.183, Xinqiao Street, Chongqing, 400037, China
| | - Yu Huang
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, NO.183, Xinqiao Street, Chongqing, 400037, China
| | - Qiang Luo
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, NO.183, Xinqiao Street, Chongqing, 400037, China
| | - Tongchuan Wang
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, NO.183, Xinqiao Street, Chongqing, 400037, China
| | - Xin Li
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, NO.183, Xinqiao Street, Chongqing, 400037, China
| | - Kibret Mequanint
- Department of Chemical and Biochemical Engineering, and School of Biomedical Engineering, The University of Western Ontario, London, Ontario, N6A 5B9, Canada
| | - Shiming Yang
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, NO.183, Xinqiao Street, Chongqing, 400037, China
- Chongqing Municipality Clinical Research Center for Gastroenterology, Chongqing, 400037, China
| | - Malcolm Xing
- Department of Mechanical Engineering, University of Manitoba, Winnipeg, Manitoba, R3T 2N2, Canada
| | - Chaoqiang Fan
- Department of Gastroenterology, Xinqiao Hospital, Army Medical University, NO.183, Xinqiao Street, Chongqing, 400037, China
- Chongqing Municipality Clinical Research Center for Gastroenterology, Chongqing, 400037, China
| |
Collapse
|