1
|
Hassannejad Z, Fendereski K, Daryabari SS, Tanourlouee SB, Dehnavi M, Kajbafzadeh AM. Advancing Myocardial Infarction Treatment: Harnessing Multi-Layered Recellularized Cardiac Patches with Fetal Myocardial Scaffolds and Acellular Amniotic Membrane. Cardiovasc Eng Technol 2024; 15:679-690. [PMID: 39133349 DOI: 10.1007/s13239-024-00744-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/04/2023] [Accepted: 07/19/2024] [Indexed: 08/13/2024]
Abstract
PURPOSE Myocardial infarction (MI) is a leading cause of irreversible functional cardiac tissue loss, requiring novel regenerative strategies. This study assessed the potential therapeutic efficacy of recellularized cardiac patches, incorporating fetal myocardial scaffolds with rat fetal cardiomyocytes and acellular human amniotic membrane, in adult Wistar rat models of MI. METHODS Decellularized myocardial tissue was obtained from 14 to 16 week-old human fetuses that had been aborted. Chemical detergents (0.1% EDTA and 0.2% sodium dodecyl sulfate) were used to prepare the fetal extracellular matrix (ECM), which was characterized for bio-scaffold microstructure and biocompatibility via scanning electron microscopy (SEM) and MTT assay, respectively. Neonatal cardiomyocytes were extracted from the ventricles of one-day-old Wistar rats' littermates and characterized through immunostaining against Connexin-43 and α-smooth muscle actin. The isolated cells were seeded onto decellularized tissues and covered with decellularized amniotic membrane. Sixteen healthy adult Wistar rats were systematically allocated to control and MI groups. MI was induced via arterial ligation. Fourteen days post-operation, the MI group was received the engineered patches. Following a two-week post-implantation period, the animals were euthanized, and the hearts were harvested for the graft evaluation. RESULTS Histological analysis, DAPI staining, and ultra-structural examination corroborated the successful depletion of cellular elements, while maintaining the integrity of the fetal ECM and architecture. Subsequent histological and immunohistochemichal (IHC) evaluations confirmed effective cardiomyocyte seeding on the scaffolds. The application of these engineered patches in MI models resulted in increased angiogenesis, reduced fibrosis, and restricted scar tissue formation, with the implanted cardiomyocytes remaining viable at graft sites, indicating prospective in vivo cell viability. CONCLUSIONS This study suggests that multi-layered recellularized cardiac patches are a promising surgical intervention for myocardial infarction, showcasing significant potential by promoting angiogenesis, mitigating fibrosis, and minimizing scar tissue formation in MI models. These features are pivotal for enhancing the therapeutic outcomes in MI patients, focusing on the restoration of the myocardial structure and function post-infarction.
Collapse
Affiliation(s)
- Zahra Hassannejad
- Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center, Tehran University of Medical Sciences, No. 62, Dr. Gharib's Street, Keshavarz Boulevard, Tehran, 1419733151, Iran.
| | - Kiarad Fendereski
- Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center, Tehran University of Medical Sciences, No. 62, Dr. Gharib's Street, Keshavarz Boulevard, Tehran, 1419733151, Iran
| | - Seyedeh Sima Daryabari
- Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center, Tehran University of Medical Sciences, No. 62, Dr. Gharib's Street, Keshavarz Boulevard, Tehran, 1419733151, Iran
| | - Saman Behboodi Tanourlouee
- Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center, Tehran University of Medical Sciences, No. 62, Dr. Gharib's Street, Keshavarz Boulevard, Tehran, 1419733151, Iran
| | - Mehrshad Dehnavi
- Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center, Tehran University of Medical Sciences, No. 62, Dr. Gharib's Street, Keshavarz Boulevard, Tehran, 1419733151, Iran
| | - Abdol-Mohammad Kajbafzadeh
- Pediatric Urology and Regenerative Medicine Research Center, Gene, Cell and Tissue Research Institute, Children's Medical Center, Tehran University of Medical Sciences, No. 62, Dr. Gharib's Street, Keshavarz Boulevard, Tehran, 1419733151, Iran.
| |
Collapse
|
2
|
Malhi A, Padda I, Mahtani A, Fabian D, Karroum P, Mathews AM, Ralhan T, Sethi Y, Emran TB. Bioprinting in cardiovascular medicine: possibilities, challenges, and future perspectives for low and middle-income countries. Int J Surg 2024; 110:6345-6354. [PMID: 38704635 PMCID: PMC11487036 DOI: 10.1097/js9.0000000000001537] [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: 01/17/2024] [Accepted: 04/15/2024] [Indexed: 05/06/2024]
Abstract
Cardiovascular diseases stemming from various factors significantly impact the quality of life and are prevalent with high mortality rates in both developed and developing countries. In cases where pharmacotherapy proves insufficient and end-stage disease ensues, a heart transplant/surgical repair becomes the only feasible treatment option. However, challenges such as a limited supply of heart donors, complications associated with rejection, and issues related to medication compliance introduce an additional burden to the healthcare system and adversely affect patient outcomes. The emergence of bioprinting has facilitated advancements in creating structures, including ventricles, valves, and blood vessels. Notably, the development of myocardial/cardiac patches through bioprinting has offered a promising avenue for revascularizing, strengthening, and regenerating various cardiovascular structures. Employment loss in developing countries as a circumstance of disability or death can severely impact a family's well-being and means for sustainable living. Innovations by means of life sustaining treatment options can provide hope for the impoverished and help reduce disability burden on the economy of low- and middle-income countries (LMICs). Such developments can have a significant impact that can last for generations, especially in these countries. In this review, the authors delve into various types of bioprinting techniques, exploring their possibilities, challenges, and potential future applications in treating various end-stage cardiovascular conditions in LMICs.
Collapse
Affiliation(s)
- Amarveer Malhi
- Department of Medicine, CMU School of Medicine, Netherlands, Antilles
| | - Inderbir Padda
- Department of Internal Medicine, Richmond University Medical Center/Mount Sinai, Staten Island, New York, USA
- PearResearch, Dehradun
| | - Arun Mahtani
- Department of Internal Medicine, Richmond University Medical Center/Mount Sinai, Staten Island, New York, USA
| | - Daniel Fabian
- Department of Internal Medicine, Richmond University Medical Center/Mount Sinai, Staten Island, New York, USA
| | - Paul Karroum
- Department of Internal Medicine, Richmond University Medical Center/Mount Sinai, Staten Island, New York, USA
| | | | - Tushar Ralhan
- School of Medicine, St. George’s University, True Blue, Grenada
| | - Yashendra Sethi
- PearResearch, Dehradun
- Department of Medicine, Government Doon Medical College, Dehradun, India
| | - Talha B. Emran
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka, Bangladesh
| |
Collapse
|
3
|
Wang Z, Lin L, Li X, Zhang Q, Mi X, Xu B, Xu Y, Liu T, Shen Y, Wang Z, Xie N, Wang J. Improving Thermosensitive Bioink Scaffold Fabrication with a Temperature-Regulated Printhead in Robot-Assisted In Situ Bioprinting System. ACS OMEGA 2024; 9:40618-40631. [PMID: 39371970 PMCID: PMC11447728 DOI: 10.1021/acsomega.4c04373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/29/2024] [Accepted: 09/05/2024] [Indexed: 10/08/2024]
Abstract
In situ bioprinting enables precise 3D printing inside the human body using modified bioprinters with thermosensitive bioinks such as gelatin methacrylate (GelMA). However, these devices lack refined temperature-regulated mechanisms essential for ensuring bioink viscosity, as compared to traditional bio-3D printers. Addressing this challenge, this study presents a temperature-regulated printhead designed to improve the fabrication of thermosensitive bioink scaffolds in in situ bioprinting, integrated into a UR5 robotic arm. Featuring a closed-loop system, it achieves a temperature steady error of 1 °C and a response time of approximately 1 min. The effectiveness of the printer was validated by bioprinting multilayer lattice 3D bioscaffolds. Comparisons were made with or without temperature control using different concentrations of GelMA + LAP. The deformation of the bioscaffolds under both conditions was analyzed, and cell culture tests were conducted to verify viability. Additionally, the rheology and mechanical properties of GelMA were tested. A final preliminary in situ bioprinting experiment was conducted on a model of a damaged femur to demonstrate practical application. The fabrication of this printhead is entirely open source, facilitating easy modifications to accommodate various robotic arms. We encourage readers to advance this prototype for application in increasingly complex in situ bioprinting situations, especially those utilizing thermosensitive bioinks.
Collapse
Affiliation(s)
- Zitong Wang
- Department
of Biomedical Engineering, School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Li Lin
- Shanghai
Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Xiangyu Li
- Department
of Mechanical, School of Mechanical & Electrical Engineering, Henan University of Technology, Zhengzhou 450001, Henan Province, China
| | - Quan Zhang
- School
of Pharmacy, Jiangsu Ocean University, Lianyungang 222005, Jiangsu Province, China
| | - Xuelian Mi
- Institute
of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 611756, Sichuan Province, China
| | - Bide Xu
- Innovative
Medical Device Registration Research and Clinical Transformation Service
Center, Institute of Translational Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yuanjing Xu
- Department
of Biomedical Instrument, Institute of Translational
Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Tongyou Liu
- Department
of Biomedical Instrument, Institute of Translational
Medicine, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Yuling Shen
- School
of Future Science and Engineering, Soochow
University, Soochow 215021, Jiangsu Province, China
| | - Zan Wang
- Department
of Mechanical, School of Mechanical & Electrical Engineering, Henan University of Technology, Zhengzhou 450001, Henan Province, China
| | - Neng Xie
- Department
of Biomedical Manufacturing and Engineering, School of Mechanical
Engineering, Shanghai Jiao Tong University, Shanghai 200011, China
| | - Jinwu Wang
- Shanghai
Ninth People’s Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai 200011, China
| |
Collapse
|
4
|
Wei X, Jiang X, Li H. Fundamental characteristics of ultrasonic green formulations using Avena sativa L. extract-mediated gold nanoparticles and electroconductive nanofibers for cardiovascular nursing care. Heliyon 2024; 10:e35018. [PMID: 39170527 PMCID: PMC11336310 DOI: 10.1016/j.heliyon.2024.e35018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 07/18/2024] [Accepted: 07/22/2024] [Indexed: 08/23/2024] Open
Abstract
In the pursuit of novel approaches to address chronic heart failure and enhance cardiovascular nursing care, environmentally sustainable nanomaterials have taken center stage. Recent progress in regenerative medicine has opened doors for the use of biocompatible biomaterials that provide mechanical support to damaged heart tissue and facilitate electrical signaling. This study was dedicated to developing advanced electroconductive nanofibers by incorporating eco-friendly Avena sativa L. extract-mediated gold nanoparticles (AuNPs) into polyaniline to create an intricate cardiac patch. The AuNPs were synthesized through an environmentally friendly chemical process aided by ultrasonic conditions. Comprehensive physicochemical analyses, such as UV-Vis spectroscopy, SEM, TEM, DPPH assay, and XRD, were carried out to characterize the AuNPs. These AuNPs were then blended with a polycaprolactone/gelatin polymeric solution and electrospun to fabricate cardiac patches, which underwent thorough evaluation using various techniques. The resulting cardiac patch demonstrated excellent hemocompatibility, antioxidant properties, and cytocompatibility, offering a promising therapeutic approach for myocardial infarctions and the advancement of cardiovascular nursing care.
Collapse
Affiliation(s)
- Xinfang Wei
- Department of Cardiovascular Medicine CCU, Zhongshan People's Hospital, No. 2 Sunwendong Road, Zhongshan City, Guangdong, 528403, China
| | - Xiaoshan Jiang
- Department of Geriatrics, Qingdao Chengyang District People's Hospital, No. 600, Changcheng Road, Chengyang District, Qingdao, 266109, Shandong Province, China
| | - Hongzan Li
- School of Nursing, Guangdong Medical University, No. 1 Xincheng Road, Songshan Lake Science and Technology Park, Dongguan, Guangdong, 523808, China
| |
Collapse
|
5
|
Saito S, Miyagawa S, Kawamura T, Yoshioka D, Kawamura M, Kawamura A, Misumi Y, Taguchi T, Yamauchi T, Miyagawa S. How should cardiac xenotransplantation be initiated in Japan? Surg Today 2024; 54:829-838. [PMID: 38733536 PMCID: PMC11266268 DOI: 10.1007/s00595-024-02861-7] [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: 02/13/2024] [Accepted: 04/07/2024] [Indexed: 05/13/2024]
Abstract
The world's first clinical cardiac xenotransplantation, using a genetically engineered pig heart with 10 gene modifications, prolonged the life of a 57-year-old man with no other life-saving options, by 60 days. It is foreseeable that xenotransplantation will be introduced in clinical practice in the United States. However, little clinical or regulatory progress has been made in the field of xenotransplantation in Japan in recent years. Japan seems to be heading toward a "device lag", and the over-importation of medical devices and technology in the medical field is becoming problematic. In this review, we discuss the concept of pig-heart xenotransplantation, including the pathobiological aspects related to immune rejection, coagulation dysregulation, and detrimental heart overgrowth, as well as genetic modification strategies in pigs to prevent or minimize these problems. Moreover, we summarize the necessity for and current status of xenotransplantation worldwide, and future prospects in Japan, with the aim of initiating xenotransplantation in Japan using genetically modified pigs without a global delay. It is imperative that this study prompts the initiation of preclinical xenotransplantation research using non-human primates and leads to clinical studies.
Collapse
Affiliation(s)
- Shunsuke Saito
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan.
| | - Shuji Miyagawa
- Department of Pediatric Surgery, Osaka University Graduate School of Medicine, Suita, Osaka, 565-0871, Japan
| | - Takuji Kawamura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Daisuke Yoshioka
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Masashi Kawamura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Ai Kawamura
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Yusuke Misumi
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | | | - Takashi Yamauchi
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| | - Shigeru Miyagawa
- Department of Cardiovascular Surgery, Osaka University Graduate School of Medicine, 2-2 Yamada-oka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
6
|
Hao M, Xue L, Wen X, Sun L, Zhang L, Xing K, Hu X, Xu J, Xing D. Advancing bone regeneration: Unveiling the potential of 3D cell models in the evaluation of bone regenerative materials. Acta Biomater 2024; 183:1-29. [PMID: 38815683 DOI: 10.1016/j.actbio.2024.05.041] [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: 02/04/2024] [Revised: 05/23/2024] [Accepted: 05/24/2024] [Indexed: 06/01/2024]
Abstract
Bone, a rigid yet regenerative tissue, has garnered extensive attention for its impressive healing abilities. Despite advancements in understanding bone repair and creating treatments for bone injuries, handling nonunions and large defects remains a major challenge in orthopedics. The rise of bone regenerative materials is transforming the approach to bone repair, offering innovative solutions for nonunions and significant defects, and thus reshaping orthopedic care. Evaluating these materials effectively is key to advancing bone tissue regeneration, especially in difficult healing scenarios, making it a critical research area. Traditional evaluation methods, including two-dimensional cell models and animal models, have limitations in predicting accurately. This has led to exploring alternative methods, like 3D cell models, which provide fresh perspectives for assessing bone materials' regenerative potential. This paper discusses various techniques for constructing 3D cell models, their pros and cons, and crucial factors to consider when using these models to evaluate bone regenerative materials. We also highlight the significance of 3D cell models in the in vitro assessments of these materials, discuss their current drawbacks and limitations, and suggest future research directions. STATEMENT OF SIGNIFICANCE: This work addresses the challenge of evaluating bone regenerative materials (BRMs) crucial for bone tissue engineering. It explores the emerging role of 3D cell models as superior alternatives to traditional methods for assessing these materials. By dissecting the construction, key factors of evaluating, advantages, limitations, and practical considerations of 3D cell models, the paper elucidates their significance in overcoming current evaluation method shortcomings. It highlights how these models offer a more physiologically relevant and ethically preferable platform for the precise assessment of BRMs. This contribution is particularly significant for "Acta Biomaterialia" readership, as it not only synthesizes current knowledge but also propels the discourse forward in the search for advanced solutions in bone tissue engineering and regeneration.
Collapse
Affiliation(s)
- Minglu Hao
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China.
| | - Linyuan Xue
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China
| | - Xiaobo Wen
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China
| | - Li Sun
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China
| | - Lei Zhang
- Department of Chemical Engineering and Waterloo Institute for Nanotechnology, University of Waterloo, Waterloo, Ontario N2L3G1, Canada
| | - Kunyue Xing
- Alliance Manchester Business School, The University of Manchester, Manchester M139PL, UK
| | - Xiaokun Hu
- Department of Interventional Medical Center, Affiliated Hospital of Qingdao University, Qingdao 26600, China
| | - Jiazhen Xu
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China.
| | - Dongming Xing
- The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao 266071, China; Cancer institute, Qingdao University, Qingdao 266071, China; School of Life Sciences, Tsinghua University, Beijing 100084, China.
| |
Collapse
|
7
|
Wang F, Song P, Wang J, Wang S, Liu Y, Bai L, Su J. Organoid bioinks: construction and application. Biofabrication 2024; 16:032006. [PMID: 38697093 DOI: 10.1088/1758-5090/ad467c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 05/02/2024] [Indexed: 05/04/2024]
Abstract
Organoids have emerged as crucial platforms in tissue engineering and regenerative medicine but confront challenges in faithfully mimicking native tissue structures and functions. Bioprinting technologies offer a significant advancement, especially when combined with organoid bioinks-engineered formulations designed to encapsulate both the architectural and functional elements of specific tissues. This review provides a rigorous, focused examination of the evolution and impact of organoid bioprinting. It emphasizes the role of organoid bioinks that integrate key cellular components and microenvironmental cues to more accurately replicate native tissue complexity. Furthermore, this review anticipates a transformative landscape invigorated by the integration of artificial intelligence with bioprinting techniques. Such fusion promises to refine organoid bioink formulations and optimize bioprinting parameters, thus catalyzing unprecedented advancements in regenerative medicine. In summary, this review accentuates the pivotal role and transformative potential of organoid bioinks and bioprinting in advancing regenerative therapies, deepening our understanding of organ development, and clarifying disease mechanisms.
Collapse
Affiliation(s)
- Fuxiao Wang
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, People's Republic of China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, People's Republic of China
- These authors contributed equally
| | - Peiran Song
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, People's Republic of China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, People's Republic of China
- These authors contributed equally
| | - Jian Wang
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, People's Republic of China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, People's Republic of China
- These authors contributed equally
| | - Sicheng Wang
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, People's Republic of China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, People's Republic of China
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai 200444, People's Republic of China
| | - Yuanyuan Liu
- School of Mechatronic Engineering and Automation, Shanghai University, Shanghai 200444, People's Republic of China
| | - Long Bai
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, People's Republic of China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, People's Republic of China
- Wenzhou Institute of Shanghai University, Wenzhou 325000, People's Republic of China
| | - Jiacan Su
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai 200444, People's Republic of China
- Department of Orthopedics, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200092, People's Republic of China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai 200444, People's Republic of China
| |
Collapse
|
8
|
Vento V, Kuntz S, Lejay A, Chakfe N. Evolutionary trends and innovations in cardiovascular intervention. FRONTIERS IN MEDICAL TECHNOLOGY 2024; 6:1384008. [PMID: 38756327 PMCID: PMC11098563 DOI: 10.3389/fmedt.2024.1384008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Accepted: 04/12/2024] [Indexed: 05/18/2024] Open
Abstract
Cardiovascular diseases remain a global health challenge, prompting continuous innovation in medical technology, particularly in Cardiovascular MedTech. This article provides a comprehensive exploration of the transformative landscape of Cardiovascular MedTech in the 21st century, focusing on interventions. The escalating prevalence of cardiovascular diseases and the demand for personalized care drive the evolving landscape, with technologies like wearables and AI reshaping patient-centric healthcare. Wearable devices offer real-time monitoring, enhancing procedural precision and patient outcomes. AI facilitates risk assessment and personalized treatment strategies, revolutionizing intervention precision. Minimally invasive procedures, aided by robotics and novel materials, minimize patient impact and improve outcomes. 3D printing enables patient-specific implants, while regenerative medicine promises cardiac regeneration. Augmented reality headsets empower surgeons during procedures, enhancing precision and awareness. Novel materials and radiation reduction techniques further optimize interventions, prioritizing patient safety. Data security measures ensure patient privacy in the era of connected healthcare. Modern technologies enhance traditional surgeries, refining outcomes. The integration of these innovations promises to shape a healthier future for cardiovascular procedures, emphasizing collaboration and research to maximize their transformative potential.
Collapse
Affiliation(s)
- Vincenzo Vento
- Vascular Surgery Department, Lancisi Cardiovascular Center, Ancona, Italy
- Department of Vascular Surgery and Kidney Transplantation, University Hospital of Strasbourg, Strasbourg, France
| | - Salomé Kuntz
- Department of Vascular Surgery and Kidney Transplantation, University Hospital of Strasbourg, Strasbourg, France
- Department of Vascular Surgery, Kidney Transplantation and Innovation, University Hospital of Strasbourg, Strasbourg, France
| | - Anne Lejay
- Department of Vascular Surgery and Kidney Transplantation, University Hospital of Strasbourg, Strasbourg, France
- Department of Vascular Surgery, Kidney Transplantation and Innovation, University Hospital of Strasbourg, Strasbourg, France
| | - Nabil Chakfe
- Department of Vascular Surgery and Kidney Transplantation, University Hospital of Strasbourg, Strasbourg, France
- Department of Vascular Surgery, Kidney Transplantation and Innovation, University Hospital of Strasbourg, Strasbourg, France
| |
Collapse
|
9
|
Zheng Z, Tang W, Li Y, Ai Y, Tu Z, Yang J, Fan C. Advancing cardiac regeneration through 3D bioprinting: methods, applications, and future directions. Heart Fail Rev 2024; 29:599-613. [PMID: 37943420 DOI: 10.1007/s10741-023-10367-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/29/2023] [Indexed: 11/10/2023]
Abstract
Cardiovascular diseases (CVDs) represent a paramount global mortality concern, and their prevalence is on a relentless ascent. Despite the effectiveness of contemporary medical interventions in mitigating CVD-related fatality rates and complications, their efficacy remains curtailed by an array of limitations. These include the suboptimal efficiency of direct cell injection and an inherent disequilibrium between the demand and availability of heart transplantations. Consequently, the imperative to formulate innovative strategies for cardiac regeneration therapy becomes unmistakable. Within this context, 3D bioprinting technology emerges as a vanguard contender, occupying a pivotal niche in the realm of tissue engineering and regenerative medicine. This state-of-the-art methodology holds the potential to fabricate intricate heart tissues endowed with multifaceted structures and functionalities, thereby engendering substantial promise. By harnessing the prowess of 3D bioprinting, it becomes plausible to synthesize functional cardiac architectures seamlessly enmeshed with the host tissue, affording a viable avenue for the restitution of infarcted domains and, by extension, mitigating the onerous yoke of CVDs. In this review, we encapsulate the myriad applications of 3D bioprinting technology in the domain of heart tissue regeneration. Furthermore, we usher in the latest advancements in printing methodologies and bioinks, culminating in an exploration of the extant challenges and the vista of possibilities inherent to a diverse array of approaches.
Collapse
Affiliation(s)
- Zilong Zheng
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China
| | - Weijie Tang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China
| | - Yichen Li
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China
| | - Yinze Ai
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China
| | - Zhi Tu
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China
| | - Jinfu Yang
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China
| | - Chengming Fan
- Department of Cardiovascular Surgery, The Second Xiangya Hospital, Central South University, Middle Renmin Road 139, Changsha, 410011, China.
| |
Collapse
|
10
|
Kennedy SM, K A, J JJB, V E, Rb JR. Transformative applications of additive manufacturing in biomedical engineering: bioprinting to surgical innovations. J Med Eng Technol 2024; 48:151-168. [PMID: 39282861 DOI: 10.1080/03091902.2024.2399017] [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: 09/04/2023] [Revised: 08/17/2024] [Accepted: 08/24/2024] [Indexed: 10/10/2024]
Abstract
This paper delves into the diverse applications and transformative impact of additive manufacturing (AM) in biomedical engineering. A detailed analysis of various AM technologies showcases their distinct capabilities and specific applications within the medical field. Special emphasis is placed on bioprinting of organs and tissues, a revolutionary area where AM has the potential to revolutionize organ transplantation and regenerative medicine by fabricating functional tissues and organs. The review further explores the customization of implants and prosthetics, demonstrating how tailored medical devices enhance patient comfort and performance. Additionally, the utility of AM in surgical planning is examined, highlighting how printed models contribute to increased surgical precision, reduced operating times, and minimized complications. The discussion extends to the 3D printing of surgical instruments, showcasing how these bespoke tools can improve surgical outcomes. Moreover, the integration of AM in drug delivery systems, including the development of innovative drug-loaded implants, underscores its potential to enhance therapeutic efficacy and reduce side effects. It also addresses personalized prosthetic implants, regulatory frameworks, biocompatibility concerns, and the future potential of AM in global health and sustainable practices.
Collapse
Affiliation(s)
- Senthil Maharaj Kennedy
- Department of Mechanical Engineering, AAA College of Engineering and Technology, Sivakasi, India
| | - Amudhan K
- Department of Mechanical Engineering, Mepco Schlenk Engineering College, Sivakasi, India
| | - Jerold John Britto J
- Department of Mechanical Engineering, Ramco Institute of Technology, Rajapalayam, India
| | - Ezhilmaran V
- Department of Manufacturing Engineering, Anna University, Chennai, India
| | - Jeen Robert Rb
- Department of Mechanical Engineering, Sri Krishna College of Technology, Coimbatore, India
| |
Collapse
|
11
|
Asl SK, Rahimzadegan M, Asl AK. Progress in cardiac tissue engineering and regeneration: Implications of gelatin-based hybrid scaffolds. Int J Biol Macromol 2024; 261:129924. [PMID: 38311143 DOI: 10.1016/j.ijbiomac.2024.129924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 01/06/2024] [Accepted: 01/31/2024] [Indexed: 02/09/2024]
Abstract
Cardiovascular diseases, particularly myocardial infarction (MI), remain a leading cause of morbidity and mortality worldwide. Current treatments for MI, more palliative than curative, have limitations in reversing the disease completely. Tissue engineering (TE) has emerged as a promising strategy to address this challenge and may lead to improved therapeutic approaches for MI. Gelatin-based scaffolds, including gelatin and its derivative, gelatin methacrylate (GelMA), have attracted significant attention in cardiac tissue engineering (CTE) due to their optimal physical and biochemical properties and capacity to mimic the native extracellular matrix (ECM). CTE mainly recruits two classes of gelatin/GelMA-based scaffolds: hydrogels and nanofibrous. This article reviews state-of-the-art gelatin/GelMA-based hybrid scaffolds currently applied for CTE and regenerative therapy. Hybrid scaffolds, fabricated by combining gelatin/GelMA hydrogel or nanofibrous scaffolds with other materials such as natural/synthetic polymers, nanoparticles, protein-based biomaterials, etc., are explored for enhanced cardiac tissue regeneration functionality. The engraftment of stem/cardiac cells, bioactive molecules, or drugs into these hybrid systems shows great promise in cardiac tissue repair and regeneration. Finally, the role of gelatin/GelMA scaffolds combined with the 3D bioprinting strategy in CTE will also be briefly highlighted.
Collapse
Affiliation(s)
- Siamak Kazemi Asl
- Deputy of Education, Ministry of Health and Medical Education, Tehran, Iran.
| | - Milad Rahimzadegan
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Kazemi Asl
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
12
|
He W, Deng J, Ma B, Tao K, Zhang Z, Ramakrishna S, Yuan W, Ye T. Recent Advancements of Bioinks for 3D Bioprinting of Human Tissues and Organs. ACS APPLIED BIO MATERIALS 2024; 7:17-43. [PMID: 38091514 DOI: 10.1021/acsabm.3c00806] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2024]
Abstract
3D bioprinting is recognized as a promising biomanufacturing technology that enables the reproducible and high-throughput production of tissues and organs through the deposition of different bioinks. Especially, bioinks based on loaded cells allow for immediate cellularity upon printing, providing opportunities for enhanced cell differentiation for organ manufacturing and regeneration. Thus, extensive applications have been found in the field of tissue engineering. The performance of the bioinks determines the functionality of the entire printed construct throughout the bioprinting process. It is generally expected that bioinks should support the encapsulated cells to achieve their respective cellular functions and withstand normal physiological pressure exerted on the printed constructs. The bioinks should also exhibit a suitable printability for precise deposition of the constructs. These characteristics are essential for the functional development of tissues and organs in bioprinting and are often achieved through the combination of different biomaterials. In this review, we have discussed the cutting-edge outstanding performance of different bioinks for printing various human tissues and organs in recent years. We have also examined the current status of 3D bioprinting and discussed its future prospects in relieving or curing human health problems.
Collapse
Affiliation(s)
- Wen He
- Ministry of Education Key Laboratory of Micro/Nano Systems for Aerospace, Northwestern Polytechnical University, Xi'an 710072, China
| | - Jinjun Deng
- Ministry of Education Key Laboratory of Micro/Nano Systems for Aerospace, Northwestern Polytechnical University, Xi'an 710072, China
| | - Binghe Ma
- Ministry of Education Key Laboratory of Micro/Nano Systems for Aerospace, Northwestern Polytechnical University, Xi'an 710072, China
| | - Kai Tao
- Ministry of Education Key Laboratory of Micro/Nano Systems for Aerospace, Northwestern Polytechnical University, Xi'an 710072, China
| | - Zhi Zhang
- State Key Laboratory of Oral Diseases and National Center for Stomatology and National Clinical Research Center for Oral Diseases, Department of Oral Maxillofacial Surgery, West China Hospital of Stomatology, Sichuan University, Chengdu 610041, Sichuan, China
| | - Seeram Ramakrishna
- Centre for Nanofibers and Nanotechnology, National University of Singapore, Singapore 117576, Singapore
| | - Weizheng Yuan
- Ministry of Education Key Laboratory of Micro/Nano Systems for Aerospace, Northwestern Polytechnical University, Xi'an 710072, China
| | - Tao Ye
- Ministry of Education Key Laboratory of Micro/Nano Systems for Aerospace, Northwestern Polytechnical University, Xi'an 710072, China
| |
Collapse
|
13
|
Narkhede M, Pardeshi A, Bhagat R, Dharme G. Review on Emerging Therapeutic Strategies for Managing Cardiovascular Disease. Curr Cardiol Rev 2024; 20:e160424228949. [PMID: 38629366 PMCID: PMC11327830 DOI: 10.2174/011573403x299265240405080030] [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: 12/18/2023] [Revised: 03/04/2024] [Accepted: 03/18/2024] [Indexed: 08/07/2024] Open
Abstract
Cardiovascular disease (CVD) remains a foremost global health concern, necessitating ongoing exploration of innovative therapeutic strategies. This review surveys the latest developments in cardiovascular therapeutics, offering a comprehensive overview of emerging approaches poised to transform disease management. The examination begins by elucidating the current epidemiological landscape of CVD and the economic challenges it poses to healthcare systems. It proceeds to scrutinize the limitations of traditional therapies, emphasizing the need for progressive interventions. The core focus is on novel pharmacological interventions, including advancements in drug development, targeted therapies, and repurposing existing medications. The burgeoning field of gene therapy and its potential in addressing genetic predispositions to cardiovascular disorders are explored, alongside the integration of artificial intelligence and machine learning in risk assessment and treatment optimization. Non-pharmacological interventions take center stage, with an exploration of digital health technologies, wearable devices, and telemedicine as transformative tools in CVD management. Regenerative medicine and stem cell therapies, offering promises of tissue repair and functional recovery, are investigated for their potential impact on cardiac health. This review also delves into the interplay of lifestyle modifications, diet, exercise, and behavioral changes, emphasizing their pivotal role in cardiovascular health and disease prevention. As precision medicine gains prominence, this synthesis of emerging therapeutic modalities aims to guide clinicians and researchers in navigating the dynamic landscape of cardiovascular disease management, fostering a collective effort to alleviate the global burden of CVD and promote a healthier future.
Collapse
Affiliation(s)
- Minal Narkhede
- SMBT College of Pharmacy, Nandi Hills Dhamangaon Taluka Igatpuri, Nashik 422403, India
| | - Avinash Pardeshi
- SMBT College of Pharmacy, Nandi Hills Dhamangaon Taluka Igatpuri, Nashik 422403, India
| | - Rahul Bhagat
- SMBT College of Pharmacy, Nandi Hills Dhamangaon Taluka Igatpuri, Nashik 422403, India
| | - Gajanan Dharme
- SMBT College of Pharmacy, Nandi Hills Dhamangaon Taluka Igatpuri, Nashik 422403, India
| |
Collapse
|
14
|
Shariati L, Esmaeili Y, Rahimmanesh I, Babolmorad S, Ziaei G, Hasan A, Boshtam M, Makvandi P. Advances in nanobased platforms for cardiovascular diseases: Early diagnosis, imaging, treatment, and tissue engineering. ENVIRONMENTAL RESEARCH 2023; 238:116933. [PMID: 37652218 DOI: 10.1016/j.envres.2023.116933] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2023] [Revised: 08/15/2023] [Accepted: 08/18/2023] [Indexed: 09/02/2023]
Abstract
Cardiovascular diseases (CVDs) present a significant threat to health, with traditional therapeutics based treatment being hindered by inefficiencies, limited biological effects, and resistance to conventional drug. Addressing these challenges requires advanced approaches for early disease diagnosis and therapy. Nanotechnology and nanomedicine have emerged as promising avenues for personalized CVD diagnosis and treatment through theranostic agents. Nanoparticles serve as nanodevices or nanocarriers, efficiently transporting drugs to injury sites. These nanocarriers offer the potential for precise drug and gene delivery, overcoming issues like bioavailability and solubility. By attaching specific target molecules to nanoparticle surfaces, controlled drug release to targeted areas becomes feasible. In the field of cardiology, nanoplatforms have gained popularity due to their attributes, such as passive or active targeting of cardiac tissues, enhanced sensitivity and specificity, and easy penetration into heart and artery tissues due to their small size. However, concerns persist about the immunogenicity and cytotoxicity of nanomaterials, necessitating careful consideration. Nanoparticles also hold promise for CVD diagnosis and imaging, enabling straightforward diagnostic procedures and real-time tracking during therapy. Nanotechnology has revolutionized cardiovascular imaging, yielding multimodal and multifunctional vehicles that outperform traditional methods. The paper provides an overview of nanomaterial delivery routes, targeting techniques, and recent advances in treating, diagnosing, and engineering tissues for CVDs. It also discusses the future potential of nanomaterials in CVDs, including theranostics, aiming to enhance cardiovascular treatment in clinical practice. Ultimately, refining nanocarriers and delivery methods has the potential to enhance treatment effectiveness, minimize side effects, and improve patients' well-being and outcomes.
Collapse
Affiliation(s)
- Laleh Shariati
- Department of Biomaterials, Nanotechnology, and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan, Iran; Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Yasaman Esmaeili
- Biosensor Research Center, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Ilnaz Rahimmanesh
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shahrzad Babolmorad
- Faculty of Veterinary Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ghazal Ziaei
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, Qatar University, Doha, 2713, Qatar; Biomedical Research Center, Qatar University, Doha, 2713, Qatar
| | - Maryam Boshtam
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, Iran.
| | - Pooyan Makvandi
- The Quzhou Affiliated Hospital of Wenzhou Medical University, Quzhou People's Hospital, Quzhou, 324000, Zhejiang, China; School of Engineering, Institute for Bioengineering, The University of Edinburgh, Edinburgh, EH9 3JL, UK.
| |
Collapse
|
15
|
Abstract
Bioprinting, as a groundbreaking technology, enables the fabrication of biomimetic tissues and organs with highly complex structures, multiple cell types, mechanical heterogeneity, and diverse functional gradients. With the growing demand for organ transplantation and the limited number of organ donors, bioprinting holds great promise for addressing the organ shortage by manufacturing completely functional organs. While the bioprinting of complete organs remains a distant goal, there has been considerable progress in the development of bioprinted transplantable tissues and organs for regenerative medicine. This review article recapitulates the current achievements of organ 3D bioprinting, primarily encompassing five important organs in the human body (i.e., the heart, kidneys, liver, pancreas, and lungs). Challenges from cellular techniques, biomanufacturing technologies, and organ maturation techniques are also deliberated for the broad application of organ bioprinting. In addition, the integration of bioprinting with other cutting-edge technologies including machine learning, organoids, and microfluidics is envisioned, which strives to offer the reader the prospect of bioprinting in constructing functional organs.
Collapse
Affiliation(s)
- Yang Wu
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen 518055, China.
| | - Minghao Qin
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen 518055, China.
| | - Xue Yang
- School of Mechanical Engineering and Automation, Harbin Institute of Technology, Shenzhen 518055, China.
| |
Collapse
|
16
|
Sun L, Bian F, Xu D, Luo Y, Wang Y, Zhao Y. Tailoring biomaterials for biomimetic organs-on-chips. MATERIALS HORIZONS 2023; 10:4724-4745. [PMID: 37697735 DOI: 10.1039/d3mh00755c] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/13/2023]
Abstract
Organs-on-chips are microengineered microfluidic living cell culture devices with continuously perfused chambers penetrating to cells. By mimicking the biological features of the multicellular constructions, interactions among organs, vascular perfusion, physicochemical microenvironments, and so on, these devices are imparted with some key pathophysiological function levels of living organs that are difficult to be achieved in conventional 2D or 3D culture systems. In this technology, biomaterials are extremely important because they affect the microstructures and functionalities of the organ cells and the development of the organs-on-chip functions. Thus, herein, we provide an overview on the advances of biomaterials for the construction of organs-on-chips. After introducing the general components, structures, and fabrication techniques of the biomaterials, we focus on the studies of the functions and applications of these biomaterials in the organs-on-chips systems. Applications of the biomaterial-based organs-on-chips as alternative animal models for pharmaceutical, chemical, and environmental tests are described and highlighted. The prospects for exciting future directions and the challenges of biomaterials for realizing the further functionalization of organs-on-chips are also presented.
Collapse
Affiliation(s)
- Lingyu Sun
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Feika Bian
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Dongyu Xu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Yuan Luo
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| | - Yongan Wang
- State Key Laboratory of Toxicology and Medical Countermeasures, Beijing Institute of Pharmacology and Toxicology, Beijing 100850, China.
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
- Southeast University Shenzhen Research Institute, Shenzhen 518071, China
| |
Collapse
|
17
|
Sang S, Wang X, Duan J, Cao Y, Shen Z, Sun L, Duan Q, Liu Z. 3D printing to construct in vitro multicellular models of melanoma. Biotechnol Bioeng 2023; 120:2853-2864. [PMID: 37227037 DOI: 10.1002/bit.28429] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2022] [Revised: 04/15/2023] [Accepted: 05/02/2023] [Indexed: 05/26/2023]
Abstract
Currently, there is a lack of suitable models for in-vitro studies of malignant melanoma and traditional single cell culture models no longer reproduce tumor structure and physiological complexity well. The tumor microenvironment is closely related to carcinogenesis and it is particularly important to understand how tumor cells interact and communicate with surrounding nonmalignant cells. Three-dimensional (3D) in vitro multicellular culture models can better simulate the tumor microenvironment due to their excellent physicochemical properties. In this study, 3D composite hydrogel scaffolds were prepared from gelatin methacrylate and polyethylene glycol diacrylate hydrogels by 3D printing and light curing techniques, and 3D multicellular in vitro tumor culture models were established by inoculating human melanoma cells (A375) and human fibroblasts cells on them. The cell proliferation, migration, invasion, and drug resistance of the 3D multicellular in vitro model was evaluated. Compared with the single-cell model, the cells in the multicellular model had higher proliferation activity and migration ability, and were easy to form dense structures. Several tumor cell markers, such as matrix metalloproteinase-9 (MMP-9), MMP-2, and vascular endothelial growth factor, were highly expressed in the multicellular culture model, which were more favorable for tumor development. In addition, higher cell survival rate was observed after exposure to luteolin. The anticancer drug resistance result of the malignant melanoma cells in the 3D bioprinted construct demonstrated physiological properties, suggesting the promising potential of current 3D printed tumor model in the development of personalized therapy, especially for discovery of more conducive targeted drugs.
Collapse
Affiliation(s)
- Shengbo Sang
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Information and Computer, Taiyuan University of Technology, Taiyuan, China
- Key Lab of Advanced Transducers and Intelligent Control System of the Ministry of Education, Taiyuan University of Technology, Taiyuan, China
| | - Xiaoyuan Wang
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Information and Computer, Taiyuan University of Technology, Taiyuan, China
- Shanxi Institute of 6D Artificial Intelligence Biomedical Science, Taiyuan, China
| | - Jiahui Duan
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Information and Computer, Taiyuan University of Technology, Taiyuan, China
- Key Lab of Advanced Transducers and Intelligent Control System of the Ministry of Education, Taiyuan University of Technology, Taiyuan, China
| | - Yanyan Cao
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Information and Computer, Taiyuan University of Technology, Taiyuan, China
- Shanxi Institute of 6D Artificial Intelligence Biomedical Science, Taiyuan, China
| | - Zhizhong Shen
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Information and Computer, Taiyuan University of Technology, Taiyuan, China
- Shanxi Institute of 6D Artificial Intelligence Biomedical Science, Taiyuan, China
| | - Lei Sun
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Information and Computer, Taiyuan University of Technology, Taiyuan, China
- Key Lab of Advanced Transducers and Intelligent Control System of the Ministry of Education, Taiyuan University of Technology, Taiyuan, China
| | - Qianqian Duan
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Information and Computer, Taiyuan University of Technology, Taiyuan, China
- Key Lab of Advanced Transducers and Intelligent Control System of the Ministry of Education, Taiyuan University of Technology, Taiyuan, China
| | - Zixian Liu
- Shanxi Key Laboratory of Micro Nano Sensors & Artificial Intelligence Perception, College of Information and Computer, Taiyuan University of Technology, Taiyuan, China
- Key Lab of Advanced Transducers and Intelligent Control System of the Ministry of Education, Taiyuan University of Technology, Taiyuan, China
| |
Collapse
|
18
|
Xie M, Zhu S, Liu G, Wu Y, Zhou W, Yu D, Wan J, Xing S, Wang S, Gan L, Li G, Chang D, Lai H, Liu N, Zhu P. A Novel Quantitative Electrocardiography Strategy Reveals the Electroinhibitory Effect of Tamoxifen on the Mouse Heart. J Cardiovasc Transl Res 2023; 16:1232-1248. [PMID: 37155136 DOI: 10.1007/s12265-023-10395-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2023] [Accepted: 04/26/2023] [Indexed: 05/10/2023]
Abstract
Tamoxifen, a selective estrogen receptor modulator, was initially used to treat cancer in women and more recently to induce conditional gene editing in rodent hearts. However, little is known about the baseline biological effects of tamoxifen on the myocardium. In order to clarify the short-term effects of tamoxifen on cardiac electrophysiology of myocardium, we applied a single-chest-lead quantitative method and analyzed the short-term electrocardiographic phenotypes induced by tamoxifen in the heart of adult female mice. We found that tamoxifen prolonged the PP interval and caused a decreased heartbeat, and further induced atrioventricular block by gradually prolonging the PR interval. Further correlation analysis suggested that tamoxifen had a synergistic and dose-independent inhibition on the time course of the PP interval and PR interval. This prolongation of the critical time course may represent a tamoxifen-specific ECG excitatory-inhibitory mechanism, leading to a reduction in the number of supraventricular action potentials and thus bradycardia. Segmental reconstructions showed that tamoxifen induced a decrease in the conduction velocity of action potentials throughout the atria and parts of the ventricles, resulting in a flattening of the P wave and R wave. In addition, we detected the previously reported prolongation of the QT interval, which may be due to a prolonged duration of the ventricular repolarizing T wave rather than the depolarizing QRS complex. Our study highlights that tamoxifen can produce patterning alternations in the cardiac conduction system, including the formation of inhibitory electrical signals with reduced conduction velocity, implying its involvement in the regulation of myocardial ion transport and the mediation of arrhythmias. A Novel Quantitative Electrocardiography Strategy Reveals the Electroinhibitory Effect of Tamoxifen on the Mouse Heart(Figure 9). A working model of tamoxifen producing acute electrical disturbances in the myocardium. SN, sinus node; AVN, atrioventricular node; RA, right atrium; LA, left atrium; RV, right ventricle; LV, left ventricle.
Collapse
Affiliation(s)
- Ming Xie
- Department of Cardiac Surgery, School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510100, Guangdong, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, and Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, 510100, Guangdong, China
| | - Shuoji Zhu
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510100, Guangdong, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, and Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, 510100, Guangdong, China
- University of Tokyo, Tokyo, 113-8666, Japan
| | - Gang Liu
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Yijin Wu
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510100, Guangdong, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, and Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, 510100, Guangdong, China
| | - Wenkai Zhou
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510100, Guangdong, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, and Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, 510100, Guangdong, China
| | - Dingdang Yu
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510100, Guangdong, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, and Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, 510100, Guangdong, China
| | - Jinkai Wan
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Shenghui Xing
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Siqing Wang
- Institutes of Biomedical Sciences, Fudan University, Shanghai, 200032, China
| | - Lin Gan
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510100, Guangdong, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, and Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, 510100, Guangdong, China
| | - Ge Li
- Department of Cardiac Surgery, School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510100, Guangdong, China
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, and Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, 510100, Guangdong, China
| | - Dehua Chang
- University of Tokyo Hospital Department of Cell Therapy in Regenerative Medicine, Tokyo, 113-8666, Japan.
| | - Hao Lai
- Department of Cardiac Surgery, Zhongshan Hospital, Fudan University, Shanghai, 200032, China.
| | - Nanbo Liu
- Department of Cardiac Surgery, School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China.
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510100, Guangdong, China.
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, and Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, 510100, Guangdong, China.
| | - Ping Zhu
- Department of Cardiac Surgery, School of Medicine, South China University of Technology, Guangzhou, 510006, Guangdong, China.
- Department of Cardiac Surgery, Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, 510100, Guangdong, China.
- Guangdong Provincial Key Laboratory of Pathogenesis, Targeted Prevention and Treatment of Heart Disease, and Guangzhou Key Laboratory of Cardiac Pathogenesis and Prevention, Guangzhou, 510100, Guangdong, China.
| |
Collapse
|
19
|
Ibi Y, Nishinakamura R. Kidney Bioengineering for Transplantation. Transplantation 2023; 107:1883-1894. [PMID: 36717963 DOI: 10.1097/tp.0000000000004526] [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: 02/01/2023]
Abstract
The kidney is an important organ for maintenance of homeostasis in the human body. As renal failure progresses, renal replacement therapy becomes necessary. However, there is a chronic shortage of kidney donors, creating a major problem for transplantation. To solve this problem, many strategies for the generation of transplantable kidneys are under investigation. Since the first reports describing that nephron progenitors could be induced from human induced pluripotent stem cells, kidney organoids have been attracting attention as tools for studying human kidney development and diseases. Because the kidney is formed through the interactions of multiple renal progenitors, current studies are investigating ways to combine these progenitors derived from human induced pluripotent stem cells for the generation of transplantable kidney organoids. Other bioengineering strategies, such as decellularization and recellularization of scaffolds, 3-dimensional bioprinting, interspecies blastocyst complementation and progenitor replacement, and xenotransplantation, also have the potential to generate whole kidneys, although each of these strategies has its own challenges. Combinations of these approaches will lead to the generation of bioengineered kidneys that are transplantable into humans.
Collapse
Affiliation(s)
- Yutaro Ibi
- Department of Kidney Development, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | | |
Collapse
|
20
|
Khan A, Kumari P, Kumari N, Shaikh U, Ekhator C, Halappa Nagaraj R, Yadav V, Khan AW, Lazarevic S, Bharati B, Lakshmipriya Vetrivendan G, Mulmi A, Mohamed H, Ullah A, Kadel B, Bellegarde SB, Rehman A. Biomimetic Approaches in Cardiac Tissue Engineering: Replicating the Native Heart Microenvironment. Cureus 2023; 15:e43431. [PMID: 37581196 PMCID: PMC10423641 DOI: 10.7759/cureus.43431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/13/2023] [Indexed: 08/16/2023] Open
Abstract
Cardiovascular diseases, including heart failure, pose significant challenges in medical practice, necessitating innovative approaches for cardiac repair and regeneration. Cardiac tissue engineering has emerged as a promising solution, aiming to develop functional and physiologically relevant cardiac tissue constructs. Replicating the native heart microenvironment, with its complex and dynamic milieu necessary for cardiac tissue growth and function, is crucial in tissue engineering. Biomimetic strategies that closely mimic the natural heart microenvironment have gained significant interest due to their potential to enhance synthetic cardiac tissue functionality and therapeutic applicability. Biomimetic approaches focus on mimicking biochemical cues, mechanical stimuli, coordinated electrical signaling, and cell-cell/cell-matrix interactions of cardiac tissue. By combining bioactive ligands, controlled delivery systems, appropriate biomaterial characteristics, electrical signals, and strategies to enhance cell interactions, biomimetic approaches provide a more physiologically relevant environment for tissue growth. The replication of the native cardiac microenvironment enables precise regulation of cellular responses, tissue remodeling, and the development of functional cardiac tissue constructs. Challenges and future directions include refining complex biochemical signaling networks, paracrine signaling, synchronized electrical networks, and cell-cell/cell-matrix interactions. Advancements in biomimetic approaches hold great promise for cardiovascular regenerative medicine, offering potential therapeutic strategies and revolutionizing cardiac disease modeling. These approaches contribute to the development of more effective treatments, personalized medicine, and improved patient outcomes. Ongoing research and innovation in biomimetic approaches have the potential to revolutionize regenerative medicine and cardiac disease modeling by replicating the native heart microenvironment, advancing functional cardiac tissue engineering, and improving patient outcomes.
Collapse
Affiliation(s)
- Anoosha Khan
- Medicine, Dow University of Health Sciences, Karachi, PAK
| | - Priya Kumari
- Medicine, Jinnah Postgraduate Medical Centre, Karachi, PAK
| | - Naina Kumari
- Dow Medical College, Dow University of Health Sciences, Karachi, PAK
| | - Usman Shaikh
- Medicine, Dow University of Health Sciences, Karachi, PAK
| | - Chukwuyem Ekhator
- Neuro-Oncology, New York Institute of Technology, College of Osteopathic Medicine, Old Westbury, USA
| | | | - Vikas Yadav
- Internal Medicine, Pandit Bhagwat Dayal Sharma Post Graduate Institute of Medical Sciences, Rohtak, IND
| | | | | | - Bishal Bharati
- Internal Medicine, Nepal Medical College, Kathmandu, NPL
| | | | | | - Hana Mohamed
- Medicine, United Nations Study & Understanding, The International Academy, Khartoum, SDN
- Medicine, Elrazi University, Khartoum, SDN
| | | | - Bijan Kadel
- Internal Medicine, Nepal Medical College and Teaching Hospital, Kathmandu, NPL
| | - Sophia B Bellegarde
- Pathology and Laboratory Medicine, American University of Antigua, St. John's, ATG
| | | |
Collapse
|
21
|
Sun Z, Zhao J, Leung E, Flandes-Iparraguirre M, Vernon M, Silberstein J, De-Juan-Pardo EM, Jansen S. Three-Dimensional Bioprinting in Cardiovascular Disease: Current Status and Future Directions. Biomolecules 2023; 13:1180. [PMID: 37627245 PMCID: PMC10452258 DOI: 10.3390/biom13081180] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2023] [Revised: 07/24/2023] [Accepted: 07/26/2023] [Indexed: 08/27/2023] Open
Abstract
Three-dimensional (3D) printing plays an important role in cardiovascular disease through the use of personalised models that replicate the normal anatomy and its pathology with high accuracy and reliability. While 3D printed heart and vascular models have been shown to improve medical education, preoperative planning and simulation of cardiac procedures, as well as to enhance communication with patients, 3D bioprinting represents a potential advancement of 3D printing technology by allowing the printing of cellular or biological components, functional tissues and organs that can be used in a variety of applications in cardiovascular disease. Recent advances in bioprinting technology have shown the ability to support vascularisation of large-scale constructs with enhanced biocompatibility and structural stability, thus creating opportunities to replace damaged tissues or organs. In this review, we provide an overview of the use of 3D bioprinting in cardiovascular disease with a focus on technologies and applications in cardiac tissues, vascular constructs and grafts, heart valves and myocardium. Limitations and future research directions are highlighted.
Collapse
Affiliation(s)
- Zhonghua Sun
- Discipline of Medical Radiation Science, Curtin Medical School, Curtin University, Perth, WA 6102, Australia;
- Curtin Health Innovation Research Institute (CHIRI), Curtin University, Perth, WA 6102, Australia
| | - Jack Zhao
- School of Medicine, Faculty of Health Sciences, The University of Western Australia, Perth, WA 6009, Australia; (J.Z.); (E.L.)
| | - Emily Leung
- School of Medicine, Faculty of Health Sciences, The University of Western Australia, Perth, WA 6009, Australia; (J.Z.); (E.L.)
| | - Maria Flandes-Iparraguirre
- Regenerative Medicine Program, Cima Universidad de Navarra, 31008 Pamplona, Spain;
- T3mPLATE, Harry Perkins Institute of Medical Research, QEII Medical Centre and UWA Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia; (M.V.); (E.M.D.-J.-P.)
- School of Engineering, The University of Western Australia, Perth, WA 6009, Australia
| | - Michael Vernon
- T3mPLATE, Harry Perkins Institute of Medical Research, QEII Medical Centre and UWA Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia; (M.V.); (E.M.D.-J.-P.)
- School of Engineering, The University of Western Australia, Perth, WA 6009, Australia
- Vascular Engineering Laboratory, Harry Perkins Institute of Medical Research, QEII Medical Centre and UWA Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia
| | - Jenna Silberstein
- Discipline of Medical Radiation Science, Curtin Medical School, Curtin University, Perth, WA 6102, Australia;
| | - Elena M. De-Juan-Pardo
- T3mPLATE, Harry Perkins Institute of Medical Research, QEII Medical Centre and UWA Centre for Medical Research, The University of Western Australia, Perth, WA 6009, Australia; (M.V.); (E.M.D.-J.-P.)
- School of Engineering, The University of Western Australia, Perth, WA 6009, Australia
- Curtin Medical School, Curtin University, Perth, WA 6102, Australia;
| | - Shirley Jansen
- Curtin Medical School, Curtin University, Perth, WA 6102, Australia;
- Department of Vascular and Endovascular Surgery, Sir Charles Gairdner Hospital, Perth, WA 6009, Australia
- Heart and Vascular Research Institute, Harry Perkins Medical Research Institute, Perth, WA 6009, Australia
- School of Medicine, The University of Western Australia, Perth, WA 6009, Australia
| |
Collapse
|
22
|
Muniyandi P, O’Hern C, Popa MA, Aguirre A. Biotechnological advances and applications of human pluripotent stem cell-derived heart models. Front Bioeng Biotechnol 2023; 11:1214431. [PMID: 37560538 PMCID: PMC10407810 DOI: 10.3389/fbioe.2023.1214431] [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: 04/29/2023] [Accepted: 07/12/2023] [Indexed: 08/11/2023] Open
Abstract
In recent years, significant biotechnological advancements have been made in engineering human cardiac tissues and organ-like models. This field of research is crucial for both basic and translational research due to cardiovascular disease being the leading cause of death in the developed world. Additionally, drug-associated cardiotoxicity poses a major challenge for drug development in the pharmaceutical and biotechnological industries. Progress in three-dimensional cell culture and microfluidic devices has enabled the generation of human cardiac models that faithfully recapitulate key aspects of human physiology. In this review, we will discuss 3D pluripotent stem cell (PSC)-models of the human heart, such as engineered heart tissues and organoids, and their applications in disease modeling and drug screening.
Collapse
Affiliation(s)
- Priyadharshni Muniyandi
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University, East Lansing, MI, United States
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, United States
| | - Colin O’Hern
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University, East Lansing, MI, United States
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, United States
| | - Mirel Adrian Popa
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University, East Lansing, MI, United States
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, United States
- Institute of Cellular Biology and Pathology Nicolae Simionescu, Bucharest, Romania
| | - Aitor Aguirre
- Institute for Quantitative Health Science and Engineering, Division of Developmental and Stem Cell Biology, Michigan State University, East Lansing, MI, United States
- Department of Biomedical Engineering, College of Engineering, Michigan State University, East Lansing, MI, United States
| |
Collapse
|
23
|
Wu CA, Zhu Y, Woo YJ. Advances in 3D Bioprinting: Techniques, Applications, and Future Directions for Cardiac Tissue Engineering. Bioengineering (Basel) 2023; 10:842. [PMID: 37508869 PMCID: PMC10376421 DOI: 10.3390/bioengineering10070842] [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: 06/16/2023] [Revised: 07/11/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
Cardiovascular diseases are the leading cause of morbidity and mortality in the United States. Cardiac tissue engineering is a direction in regenerative medicine that aims to repair various heart defects with the long-term goal of artificially rebuilding a full-scale organ that matches its native structure and function. Three-dimensional (3D) bioprinting offers promising applications through its layer-by-layer biomaterial deposition using different techniques and bio-inks. In this review, we will introduce cardiac tissue engineering, 3D bioprinting processes, bioprinting techniques, bio-ink materials, areas of limitation, and the latest applications of this technology, alongside its future directions for further innovation.
Collapse
Affiliation(s)
- Catherine A Wu
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA
| | - Yuanjia Zhu
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Y Joseph Woo
- Department of Cardiothoracic Surgery, Stanford University, Stanford, CA 94305, USA
- Department of Bioengineering, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
24
|
de Paula AP, de Lima JD, Bastos TSB, Czaikovski AP, dos Santos Luz RB, Yuasa BS, Smanioto CCS, Robert AW, Braga TT. Decellularized Extracellular Matrix: The Role of This Complex Biomaterial in Regeneration. ACS OMEGA 2023; 8:22256-22267. [PMID: 37396215 PMCID: PMC10308580 DOI: 10.1021/acsomega.2c06216] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Accepted: 01/12/2023] [Indexed: 07/04/2023]
Abstract
Organ transplantation is understood as a technique where an organ from a donor patient is transferred to a recipient patient. This practice gained strength in the 20th century and ensured advances in areas of knowledge such as immunology and tissue engineering. The main problems that comprise the practice of transplants involve the demand for viable organs and immunological aspects related to organ rejection. In this review, we address advances in tissue engineering for reversing the current challenges of transplants, focusing on the possible use of decellularized tissues in tissue engineering. We address the interaction of acellular tissues with immune cells, especially macrophages and stem cells, due to their potential use in regenerative medicine. Our goal is to exhibit data that demonstrate the use of decellularized tissues as alternative biomaterials that can be applied clinically as partial or complete organ substitutes.
Collapse
Affiliation(s)
| | - Jordana Dinorá de Lima
- Department
of Pathology, Federal University of Parana, Curitiba, Parana 80060-000, Brazil
| | | | | | | | - Bruna Sadae Yuasa
- Department
of Pathology, Federal University of Parana, Curitiba, Parana 80060-000, Brazil
| | | | - Anny Waloski Robert
- Stem
Cells Basic Biology Laboratory, Carlos Chagas
Institute − FIOCRUZ/PR, Curitiba, Parana 81350-010, Brazil
| | - Tárcio Teodoro Braga
- Department
of Pathology, Federal University of Parana, Curitiba, Parana 80060-000, Brazil
- Graduate
Program in Biosciences and Biotechnology, Institute Carlos Chagas, Fiocruz, Parana 81310-020, Brazil
| |
Collapse
|
25
|
Abstract
PURPOSE OF REVIEW Bioengineering of functional cardiac tissue composed of primary cardiomyocytes has great potential for myocardial regeneration and in vitro tissue modeling. 3D bioprinting was developed to create cardiac tissue in hydrogels that can mimic the structural, physiological, and functional features of native myocardium. Through a detailed review of the 3D printing technologies and bioink materials used in the creation of a heart tissue, this article discusses the potential of engineered heart tissues in biomedical applications. RECENT FINDINGS In this review, we discussed the recent progress in 3D bioprinting strategies for cardiac tissue engineering, including bioink and 3D bioprinting methods as well as examples of engineered cardiac tissue such as in vitro cardiac models and vascular channels. 3D printing is a powerful tool for creating in vitro cardiac tissues that are structurally and functionally similar to real tissues. The use of human-induced pluripotent stem cell-derived cardiomyocytes (iPSC-CM) enables the generation of patient-specific tissues. These tissues have the potential to be used for regenerative therapies, disease modeling, and drug testing.
Collapse
Affiliation(s)
- Ting-Yu Lu
- Materials Science and Engineering Program, University of California, 9500 Gilman Dr. San Diego, 92093 La Jolla, CA USA
| | - Yi Xiang
- Department of NanoEngineering, University of California, 9500 Gilman Dr. San Diego, 92093 La Jolla, CA USA
| | - Min Tang
- Department of NanoEngineering, University of California, 9500 Gilman Dr. San Diego, 92093 La Jolla, CA USA
| | - Shaochen Chen
- Materials Science and Engineering Program, University of California, 9500 Gilman Dr. San Diego, 92093 La Jolla, CA USA
- Department of NanoEngineering, University of California, 9500 Gilman Dr. San Diego, 92093 La Jolla, CA USA
- Department of Bioengineering, University of California, 9500 Gilman Dr. San Diego, 92093 La Jolla, CA USA
| |
Collapse
|
26
|
Mir A, Lee E, Shih W, Koljaka S, Wang A, Jorgensen C, Hurr R, Dave A, Sudheendra K, Hibino N. 3D Bioprinting for Vascularization. Bioengineering (Basel) 2023; 10:bioengineering10050606. [PMID: 37237676 DOI: 10.3390/bioengineering10050606] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2023] [Revised: 04/27/2023] [Accepted: 05/07/2023] [Indexed: 05/28/2023] Open
Abstract
In the world of clinic treatments, 3D-printed tissue constructs have emerged as a less invasive treatment method for various ailments. Printing processes, scaffold and scaffold free materials, cells used, and imaging for analysis are all factors that must be observed in order to develop successful 3D tissue constructs for clinical applications. However, current research in 3D bioprinting model development lacks diverse methods of successful vascularization as a result of issues with scaling, size, and variations in printing method. This study analyzes the methods of printing, bioinks used, and analysis techniques in 3D bioprinting for vascularization. These methods are discussed and evaluated to determine the most optimal strategies of 3D bioprinting for successful vascularization. Integrating stem and endothelial cells in prints, selecting the type of bioink according to its physical properties, and choosing a printing method according to physical properties of the desired printed tissue are steps that will aid in the successful development of a bioprinted tissue and its vascularization.
Collapse
Affiliation(s)
- Amatullah Mir
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA
| | - Eugenia Lee
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA
| | - Wesley Shih
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA
| | - Sarah Koljaka
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA
| | - Anya Wang
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA
| | - Caitlin Jorgensen
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA
| | - Riley Hurr
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA
| | - Amartya Dave
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA
| | - Krupa Sudheendra
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA
| | - Narutoshi Hibino
- Section of Cardiac Surgery, Department of Surgery, University of Chicago, 5841 S. Maryland Ave., Chicago, IL 60637, USA
- Pediatric Cardiac Surgery, Advocate Children's Hospital, 4440 W 95th St. Oak Lawn, IL 60453, USA
| |
Collapse
|
27
|
Sharma A, Kaur I, Dheer D, Nagpal M, Kumar P, Venkatesh DN, Puri V, Singh I. A propitious role of marine sourced polysaccharides: Drug delivery and biomedical applications. Carbohydr Polym 2023; 308:120448. [PMID: 36813329 DOI: 10.1016/j.carbpol.2022.120448] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2022] [Revised: 11/06/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
Numerous compounds, with extensive applications in biomedical and biotechnological fields, are present in the oceans, which serve as a prime renewable source of natural substances, further promoting the development of novel medical systems and devices. Polysaccharides are present in the marine ecosystem in abundance, promoting minimal extraction costs, in addition to their solubility in extraction media, and an aqueous solvent, along with their interactions with biological compounds. Certain algae-derived polysaccharides include fucoidan, alginate, and carrageenan, while animal-derived polysaccharides comprise hyaluronan, chitosan and many others. Furthermore, these compounds can be modified to facilitate their processing into multiple shapes and sizes, as well as exhibit response dependence to external conditions like temperature and pH. All these properties have promoted the use of these biomaterials as raw materials for the development of drug delivery carrier systems (hydrogels, particles, capsules). The present review enlightens marine polysaccharides providing its sources, structures, biological properties, and its biomedical applications. In addition to this, their role as nanomaterials is also portrayed by the authors, along with the methods employed to develop them and associated biological and physicochemical properties designed to develop suitable drug delivery systems.
Collapse
Affiliation(s)
- Ameya Sharma
- Chitkara School of Pharmacy, Chitkara University, Himachal Pradesh, India
| | - Ishnoor Kaur
- Chitkara College of Pharmacy, Chitkara University, Punjab, India; University of Glasgow, College of Medical, Veterinary and Life Sciences, Glasgow, United Kingdom, G12 8QQ
| | - Divya Dheer
- Chitkara School of Pharmacy, Chitkara University, Himachal Pradesh, India
| | - Manju Nagpal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Pradeep Kumar
- Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, Johannesburg, South Africa
| | - D Nagasamy Venkatesh
- JSS College of Pharmacy, JSS Academy of Higher Education & Research, Ooty, Tamil Nadu, India
| | - Vivek Puri
- Chitkara School of Pharmacy, Chitkara University, Himachal Pradesh, India.
| | - Inderbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| |
Collapse
|
28
|
Aziz R, Falanga M, Purenovic J, Mancini S, Lamberti P, Guida M. A Review on the Applications of Natural Biodegradable Nano Polymers in Cardiac Tissue Engineering. NANOMATERIALS (BASEL, SWITZERLAND) 2023; 13:1374. [PMID: 37110959 PMCID: PMC10145986 DOI: 10.3390/nano13081374] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Revised: 04/12/2023] [Accepted: 04/13/2023] [Indexed: 06/19/2023]
Abstract
As cardiac diseases, which mostly result in heart failure, are increasing rapidly worldwide, heart transplantation seems the only solution for saving lives. However, this practice is not always possible due to several reasons, such as scarcity of donors, rejection of organs from recipient bodies, or costly medical procedures. In the framework of nanotechnology, nanomaterials greatly contribute to the development of these cardiovascular scaffolds as they provide an easy regeneration of the tissues. Currently, functional nanofibers can be used in the production of stem cells and in the regeneration of cells and tissues. The small size of nanomaterials, however, leads to changes in their chemical and physical characteristics that could alter their interaction and exposure to stem cells with cells and tissues. This article aims to review the naturally occurring biodegradable nanomaterials that are used in cardiovascular tissue engineering for the development of cardiac patches, vessels, and tissues. Moreover, this article also provides an overview of cell sources used for cardiac tissue engineering, explains the anatomy and physiology of the human heart, and explores the regeneration of cardiac cells and the nanofabrication approaches used in cardiac tissue engineering as well as scaffolds.
Collapse
Affiliation(s)
- Rabia Aziz
- Department of Information and Electrical Engineering and Applied Mathematics (DIEM), University of Salerno, 84084 Fisciano, Italy; (M.F.); (S.M.); (P.L.); (M.G.)
- Consiglio Nazionale Delle Ricerche (CNR)-Istituto Officina dei Materiali (IOM), Area Science Park Basovizza S.S. 14-Km. 163, 5-34149 Trieste, Italy
| | - Mariarosaria Falanga
- Department of Information and Electrical Engineering and Applied Mathematics (DIEM), University of Salerno, 84084 Fisciano, Italy; (M.F.); (S.M.); (P.L.); (M.G.)
| | - Jelena Purenovic
- Department of Physics and Materials, Faculty of Sciences at Cacak, University of Kragujevac, 32000 Cacak, Serbia;
| | - Simona Mancini
- Department of Information and Electrical Engineering and Applied Mathematics (DIEM), University of Salerno, 84084 Fisciano, Italy; (M.F.); (S.M.); (P.L.); (M.G.)
| | - Patrizia Lamberti
- Department of Information and Electrical Engineering and Applied Mathematics (DIEM), University of Salerno, 84084 Fisciano, Italy; (M.F.); (S.M.); (P.L.); (M.G.)
- Italian Interuniversity Research Center on Interaction between Electromagnetic Fields and Biosystems (ICEmB), Università Degli Studi di Genova, DITEN, Via all’Opera Pia 11/a, 16145 Genova, Italy
- Interdepartmental Research Centre for Nanomaterials and Nanotechnology at the University of Salerno (NanoMates), Department of Physics, University of Salerno, Via Giovanni Paolo II 132, 84084 Fisciano, Italy
| | - Michele Guida
- Department of Information and Electrical Engineering and Applied Mathematics (DIEM), University of Salerno, 84084 Fisciano, Italy; (M.F.); (S.M.); (P.L.); (M.G.)
- Italian Interuniversity Research Center on Interaction between Electromagnetic Fields and Biosystems (ICEmB), Università Degli Studi di Genova, DITEN, Via all’Opera Pia 11/a, 16145 Genova, Italy
| |
Collapse
|
29
|
Zhou Q, Su X, Wu J, Zhang X, Su R, Ma L, Sun Q, He R. Additive Manufacturing of Bioceramic Implants for Restoration Bone Engineering: Technologies, Advances, and Future Perspectives. ACS Biomater Sci Eng 2023; 9:1164-1189. [PMID: 36786214 DOI: 10.1021/acsbiomaterials.2c01164] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/15/2023]
Abstract
Treating bone defects is highly challenging because they do not heal on their own inside the patients, so implants are needed to assist in the reconstruction of the bone. Bioceramic implants based on additive manufacturing (AM) are currently emerging as promising treatment options for restoration bone engineering. On the one hand, additively manufactured bioceramic implants have excellent mechanical properties and biocompatibility, which are suitable for bone regeneration. On the other hand, the designable structure and adjustable pores of additively manufactured bioceramic implants allow them to promote suitable cell growth and tissue climbing. Herein, this review unfolds to introduce several frequently employed AM technologies for bioceramic implants. After that, advances in commonly used additively manufactured bioceramic implants, including bioinert ceramic implants, bioactive ceramic implants, and bioceramic/organic composite implants, are categorized and summarized. Finally, the future perspectives of additively manufactured bioceramic implants, in terms of mechanical performance improvement, innovative structural design, biological property enhancement, and other functionalization approaches, are proposed and forecasted. This review is believed to provide some fundamental understanding and cutting-edge knowledge for the additive manufacturing of bioceramic implants for restoration bone engineering.
Collapse
Affiliation(s)
- Qing Zhou
- Institute of Advanced Structure Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Xiaonan Su
- Beijing Scrianen Pharmaceutical Co. Ltd., Beijing 102699, China
| | - Jianqin Wu
- Institute of Advanced Structure Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Xueqin Zhang
- Institute of Advanced Structure Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Ruyue Su
- Institute of Advanced Structure Technology, Beijing Institute of Technology, Beijing 100081, China
| | - Lili Ma
- Center of Dental Medicine, China-Japan Friendship Hospital, Beijing 100029, China
| | - Qiang Sun
- Center of Dental Medicine, China-Japan Friendship Hospital, Beijing 100029, China
| | - Rujie He
- Institute of Advanced Structure Technology, Beijing Institute of Technology, Beijing 100081, China
| |
Collapse
|
30
|
Singh BN, Yucel D, Garay BI, Tolkacheva EG, Kyba M, Perlingeiro RCR, van Berlo JH, Ogle BM. Proliferation and Maturation: Janus and the Art of Cardiac Tissue Engineering. Circ Res 2023; 132:519-540. [PMID: 36795845 PMCID: PMC9943541 DOI: 10.1161/circresaha.122.321770] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
During cardiac development and morphogenesis, cardiac progenitor cells differentiate into cardiomyocytes that expand in number and size to generate the fully formed heart. Much is known about the factors that regulate initial differentiation of cardiomyocytes, and there is ongoing research to identify how these fetal and immature cardiomyocytes develop into fully functioning, mature cells. Accumulating evidence indicates that maturation limits proliferation and conversely proliferation occurs rarely in cardiomyocytes of the adult myocardium. We term this oppositional interplay the proliferation-maturation dichotomy. Here we review the factors that are involved in this interplay and discuss how a better understanding of the proliferation-maturation dichotomy could advance the utility of human induced pluripotent stem cell-derived cardiomyocytes for modeling in 3-dimensional engineered cardiac tissues to obtain truly adult-level function.
Collapse
Affiliation(s)
- Bhairab N. Singh
- Department of Pediatrics, University of Minnesota, MN, USA
- Department of Biomedical Engineering, University of Minnesota, MN, USA
- Stem Cell Institute, University of Minnesota, MN, USA
| | - Dogacan Yucel
- Stem Cell Institute, University of Minnesota, MN, USA
- Department of Medicine, Cardiovascular Division, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
| | - Bayardo I. Garay
- Stem Cell Institute, University of Minnesota, MN, USA
- Department of Medicine, Cardiovascular Division, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
- Medical Scientist Training Program, University of Minnesota Medical School, MN, USA
| | - Elena G. Tolkacheva
- Department of Biomedical Engineering, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
- Institute for Engineering in Medicine, University of Minnesota, MN, USA
| | - Michael Kyba
- Department of Pediatrics, University of Minnesota, MN, USA
- Stem Cell Institute, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
| | - Rita C. R. Perlingeiro
- Stem Cell Institute, University of Minnesota, MN, USA
- Department of Medicine, Cardiovascular Division, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
| | - Jop H. van Berlo
- Stem Cell Institute, University of Minnesota, MN, USA
- Department of Medicine, Cardiovascular Division, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
| | - Brenda M. Ogle
- Department of Pediatrics, University of Minnesota, MN, USA
- Department of Biomedical Engineering, University of Minnesota, MN, USA
- Stem Cell Institute, University of Minnesota, MN, USA
- Lillehei Heart Institute, University of Minnesota, MN, USA
- Institute for Engineering in Medicine, University of Minnesota, MN, USA
- Masonic Cancer Center, University of Minnesota, MN, USA
| |
Collapse
|
31
|
Nanocomposite Hydrogels as Functional Extracellular Matrices. Gels 2023; 9:gels9020153. [PMID: 36826323 PMCID: PMC9957407 DOI: 10.3390/gels9020153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/31/2023] [Accepted: 02/08/2023] [Indexed: 02/16/2023] Open
Abstract
Over recent years, nano-engineered materials have become an important component of artificial extracellular matrices. On one hand, these materials enable static enhancement of the bulk properties of cell scaffolds, for instance, they can alter mechanical properties or electrical conductivity, in order to better mimic the in vivo cell environment. Yet, many nanomaterials also exhibit dynamic, remotely tunable optical, electrical, magnetic, or acoustic properties, and therefore, can be used to non-invasively deliver localized, dynamic stimuli to cells cultured in artificial ECMs in three dimensions. Vice versa, the same, functional nanomaterials, can also report changing environmental conditions-whether or not, as a result of a dynamically applied stimulus-and as such provide means for wireless, long-term monitoring of the cell status inside the culture. In this review article, we present an overview of the technological advances regarding the incorporation of functional nanomaterials in artificial extracellular matrices, highlighting both passive and dynamically tunable nano-engineered components.
Collapse
|
32
|
The Exciting Realities and Possibilities of iPS-Derived Cardiomyocytes. Bioengineering (Basel) 2023; 10:bioengineering10020237. [PMID: 36829731 PMCID: PMC9952364 DOI: 10.3390/bioengineering10020237] [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: 01/16/2023] [Revised: 02/03/2023] [Accepted: 02/09/2023] [Indexed: 02/12/2023] Open
Abstract
Induced pluripotent stem cells (iPSCs) have become a prevalent topic after their discovery, advertised as an ethical alternative to embryonic stem cells (ESCs). Due to their ability to differentiate into several kinds of cells, including cardiomyocytes, researchers quickly realized the potential for differentiated cardiomyocytes to be used in the treatment of heart failure, a research area with few alternatives. This paper discusses the differentiation process for human iPSC-derived cardiomyocytes and the possible applications of said cells while answering some questions regarding ethical issues.
Collapse
|
33
|
Mao X, Wang Z. Research Progress of Three-Dimensional Bioprinting Artificial Cardiac Tissue. Tissue Eng Regen Med 2023; 20:1-9. [PMID: 36401767 PMCID: PMC9852375 DOI: 10.1007/s13770-022-00495-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2022] [Revised: 09/17/2022] [Accepted: 09/19/2022] [Indexed: 11/21/2022] Open
Abstract
Cardiovascular disease is one of the main diseases that endanger human life and health, and heart failure often occurs when the cardiovascular disease develops to the end-stage. Heart transplantation is the most effective treatment. However, there has always been a shortage of living heart organs. With the development of regenerative medicine, researchers have turned to bioprinting technology that can build tissues and organs in vitro. A large number of relevant literature on three-dimensional (3D) bioprinted hearts were searched and screened in Google Scholar. 3D bioprinting technology can accurately print biomaterials containing living cells into 3D functional living tissues, providing a feasible solution to the shortage of transplantable organs. As one of the most important organs in the human body, the research on 3D bioprinting of the heart has currently become a hot topic. This paper briefly overviews 3D bioprinting technology and the progress in bioprinting cardiac tissue. It is believed that in the future, bio-printed hearts will become a reality, making a new way of providing artificial organs for heart transplantation.
Collapse
Affiliation(s)
- Xin Mao
- School of Chemistry and Pharmaceutical Engineering, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271016, People's Republic of China
| | - Zhehui Wang
- School of Chemistry and Pharmaceutical Engineering, Shandong First Medical University & Shandong Academy of Medical Sciences, Taian, 271016, People's Republic of China.
| |
Collapse
|
34
|
Khajehmohammadi M, Azizi Tafti R, Nikukar H. Effect of porosity on mechanical and biological properties of bioprinted scaffolds. J Biomed Mater Res A 2023; 111:245-260. [PMID: 36205372 DOI: 10.1002/jbm.a.37455] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2022] [Revised: 09/25/2022] [Accepted: 09/27/2022] [Indexed: 01/10/2023]
Abstract
Treatment of tissue defects commonly represents a major problem in clinics due to difficulties involving a shortage of donors, inappropriate sizes, abnormal shapes, and immunological rejection. While many scaffold parameters such as pore shape, porosity percentage, and pore connectivity could be adjusted to achieve desired mechanical and biological properties. These parameters are crucial scaffold parameters that can be accurately produced by 3D bioprinting technology based on the damaged tissue. In the present research, the effect of porosity percentage (40%, 50%, and 60%) and different pore shapes (square, star, and gyroid) on the mechanical (e.g., stiffness, compressive and tensile behavior) and biological (e.g., biodegradation, and cell viability) properties of porous polycaprolactone (PCL) scaffolds coated with gelatin have been investigated. Moreover, human foreskin fibroblast cells were cultured on the scaffolds in the in-vitro procedures. MTT assay (4, 7, and 14 days) was utilized to determine the cytotoxicity of the porous scaffolds. It is revealed that the porous scaffolds produced by the bioprinter did not produce a cytotoxic effect. Among all the porous scaffolds, scaffolds with a pore size of about 500 μm and porosity of 50% showed the best cell proliferation compared to the controls after 14 days. The results demonstrated that the pore shape, porosity percentage, and pore connectivity have an important role in improving the mechanical and biological properties of porous scaffolds. These 3D bioprinted biodegradable scaffolds exhibit potential for future application as polymeric scaffolds in hard tissue engineering applications.
Collapse
Affiliation(s)
| | | | - Habib Nikukar
- Department of Advanced Medical Sciences and Technologies, School of Paramedicine, Shahid Sadoughi University of Medical Sciences, Yazd, Iran.,Medical Nanotechnology and Tissue Engineering Research Center, Yazd Reproductive Sciences Institute, Shahid Sadoughi University of Medical Sciences, Yazd, Iran
| |
Collapse
|
35
|
Sun L, Wang Y, Xu D, Zhao Y. Emerging technologies for cardiac tissue engineering and artificial hearts. SMART MEDICINE 2023; 2:e20220040. [PMID: 39188557 PMCID: PMC11235648 DOI: 10.1002/smmd.20220040] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/14/2022] [Accepted: 12/24/2022] [Indexed: 08/28/2024]
Abstract
Heart diseases, especially cardiovascular diseases, have brought heavy burden on society for their high morbidity and mortality. In clinical, heart transplantation is recognized as an effective strategy to rescue the lives of patients, while it may suffer from lack of donors and possible immune responses. In view of this, tremendous efforts have been devoted to developing alternative strategies to recover the function and promote the regeneration of cardiac tissues. As an emerging field blending cell biology and material science, tissue engineering technique allows the construction of biomimetic living complexes as organ substitutes for heart repair. In this review, we will present the recent progress in cardiac tissue engineering and artificial hearts. After introducing the critical elements in cardiac tissue engineering, we will present advanced fabrication methods to achieve scaffolds with desired micro/nanostructure design as well as the applications of these bioinspired scaffolds. We will also discuss the current dilemma and possible development direction from a biomedical perspective.
Collapse
Affiliation(s)
- Lingyu Sun
- Department of Rheumatology and ImmunologyInstitute of Translational MedicineNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| | - Yu Wang
- Department of Rheumatology and ImmunologyInstitute of Translational MedicineNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| | - Dongyu Xu
- Department of Rheumatology and ImmunologyInstitute of Translational MedicineNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| | - Yuanjin Zhao
- Department of Rheumatology and ImmunologyInstitute of Translational MedicineNanjing Drum Tower HospitalSchool of Biological Science and Medical EngineeringSoutheast UniversityNanjingChina
| |
Collapse
|
36
|
McIvor MJ, Ó Maolmhuaidh F, Meenagh A, Hussain S, Bhattacharya G, Fishlock S, Ward J, McFerran A, Acheson JG, Cahill PA, Forster R, McEneaney DJ, Boyd AR, Meenan BJ. 3D Fabrication and Characterisation of Electrically Receptive PCL-Graphene Scaffolds for Bioengineered In Vitro Tissue Models. MATERIALS (BASEL, SWITZERLAND) 2022; 15:9030. [PMID: 36556835 PMCID: PMC9783119 DOI: 10.3390/ma15249030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/16/2022] [Revised: 12/11/2022] [Accepted: 12/14/2022] [Indexed: 06/17/2023]
Abstract
Polycaprolactone (PCL) is a well-established biomaterial, offering extensive mechanical attributes along with low cost, biocompatibility, and biodegradability; however, it lacks hydrophilicity, bioactivity, and electrical conductivity. Advances in 3D fabrication technologies allow for these sought-after attributes to be incorporated into the scaffolds during fabrication. In this study, solvent-free Fused Deposition Modelling was employed to fabricate 3D scaffolds from PCL with increasing amounts of graphene (G), in the concentrations of 0.75, 1.5, 3, and 6% (w/w). The PCL+G scaffolds created were characterised physico-chemically, electrically, and biologically. Raman spectroscopy demonstrated that the scaffold outer surface contained both PCL and G, with the G component relatively uniformly distributed. Water contact angle measurement demonstrated that as the amount of G in the scaffold increases (0.75-6% w/w), hydrophobicity decreases; mean contact angle for pure PCL was recorded as 107.22 ± 9.39°, and that with 6% G (PCL+6G) as 77.56 ± 6.75°. Electrochemical Impedance Spectroscopy demonstrated a marked increase in electroactivity potential with increasing G concentration. Cell viability results indicated that even the smallest addition of G (0.75%) resulted in a significant improvement in electroactivity potential and bioactivity compared with that for pure PCL, with 1.5 and 3% exhibiting the highest statistically significant increases in cell proliferation.
Collapse
Affiliation(s)
- Mary Josephine McIvor
- Nanotechnology and Integrated Bioengineering Centre (NIBEC), School of Engineering, Ulster University, 2-24 York Street, Belfast BT15 1AP, UK
| | - Fionn Ó Maolmhuaidh
- The National Centre for Sensor Research, School of Chemical Sciences, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Aidan Meenagh
- Nanotechnology and Integrated Bioengineering Centre (NIBEC), School of Engineering, Ulster University, 2-24 York Street, Belfast BT15 1AP, UK
| | - Shahzad Hussain
- Nanotechnology and Integrated Bioengineering Centre (NIBEC), School of Engineering, Ulster University, 2-24 York Street, Belfast BT15 1AP, UK
| | - Gourav Bhattacharya
- Nanotechnology and Integrated Bioengineering Centre (NIBEC), School of Engineering, Ulster University, 2-24 York Street, Belfast BT15 1AP, UK
| | - Sam Fishlock
- Nanotechnology and Integrated Bioengineering Centre (NIBEC), School of Engineering, Ulster University, 2-24 York Street, Belfast BT15 1AP, UK
| | - Joanna Ward
- Nanotechnology and Integrated Bioengineering Centre (NIBEC), School of Engineering, Ulster University, 2-24 York Street, Belfast BT15 1AP, UK
| | - Aoife McFerran
- Nanotechnology and Integrated Bioengineering Centre (NIBEC), School of Engineering, Ulster University, 2-24 York Street, Belfast BT15 1AP, UK
| | - Jonathan G. Acheson
- Nanotechnology and Integrated Bioengineering Centre (NIBEC), School of Engineering, Ulster University, 2-24 York Street, Belfast BT15 1AP, UK
| | - Paul A. Cahill
- School of Biotechnology, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - Robert Forster
- The National Centre for Sensor Research, School of Chemical Sciences, Dublin City University, Glasnevin, Dublin 9, Ireland
| | - David J. McEneaney
- Cardiovascular Research Unit, Craigavon Area Hospital, 68 Lurgan Road, Portadown, Co., Armagh BT63 5QQ, UK
| | - Adrian R. Boyd
- Nanotechnology and Integrated Bioengineering Centre (NIBEC), School of Engineering, Ulster University, 2-24 York Street, Belfast BT15 1AP, UK
| | - Brian J. Meenan
- Nanotechnology and Integrated Bioengineering Centre (NIBEC), School of Engineering, Ulster University, 2-24 York Street, Belfast BT15 1AP, UK
| |
Collapse
|
37
|
Ngomi N, Khayeka-Wandabwa C, Egondi T, Marinda PA, Haregu TN. Determinants of inequality in health care seeking for childhood illnesses: insights from Nairobi informal settlements. GLOBAL HEALTH JOURNAL 2022. [DOI: 10.1016/j.glohj.2022.12.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
|
38
|
Majidansari S, Vahedi N, Rekabgardan M, Ganjoury C, Najmoddin N, Tabatabaei M, Sigaroodi F, Naraghi‐Bagherpour P, Taheri SAA, Khani M. Enhancing endothelial differentiation of human mesenchymal stem cells by culture on a nanofibrous polycaprolactone/(poly‐glycerol sebacate)/gelatin scaffold. POLYM ADVAN TECHNOL 2022. [DOI: 10.1002/pat.5925] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Affiliation(s)
- Shima Majidansari
- Department of Tissue Engineering Science and Research branch, Islamic Azad University Tehran Iran
| | - Negin Vahedi
- Department of Life Science Engineering Faculty of New Sciences and Technologies, University of Tehran Tehran Iran
| | - Mahmood Rekabgardan
- Department of Tissue Engineering and Applied Cell Sciences School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Camellia Ganjoury
- Medical Nanotechnology and Tissue Engineering Research Center Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Najmeh Najmoddin
- Department of Biomedical Engineering Science and Research Branch, Islamic Azad University Tehran Iran
| | - Mohammad Tabatabaei
- Cell Engineering and Biomicrofluidics Systems Lab Department of Biomedical Engineering, Amirkabir University of Technology Tehran Iran
| | - Faraz Sigaroodi
- Department of Tissue Engineering and Applied Cell Sciences School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Paniz Naraghi‐Bagherpour
- Medical Nanotechnology and Tissue Engineering Research Center Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Seyed Amir Ali Taheri
- Medical Nanotechnology and Tissue Engineering Research Center Shahid Beheshti University of Medical Sciences Tehran Iran
| | - Mohammad‐Mehdi Khani
- Department of Tissue Engineering and Applied Cell Sciences School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences Tehran Iran
| |
Collapse
|
39
|
3D Bioprinting Technology and Hydrogels Used in the Process. J Funct Biomater 2022; 13:jfb13040214. [PMID: 36412855 PMCID: PMC9680466 DOI: 10.3390/jfb13040214] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Revised: 10/21/2022] [Accepted: 10/23/2022] [Indexed: 11/06/2022] Open
Abstract
3D bioprinting has gained visibility in regenerative medicine and tissue engineering due to its applicability. Over time, this technology has been optimized and adapted to ensure a better printability of bioinks and biomaterial inks, contributing to developing structures that mimic human anatomy. Therefore, cross-linked polymeric materials, such as hydrogels, have been highly targeted for the elaboration of bioinks, as they guarantee cell proliferation and adhesion. Thus, this short review offers a brief evolution of the 3D bioprinting technology and elucidates the main hydrogels used in the process.
Collapse
|
40
|
Javaid M, Haleem A, Singh RP, Suman R. 3D printing applications for healthcare research and development. GLOBAL HEALTH JOURNAL 2022. [DOI: 10.1016/j.glohj.2022.11.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
|
41
|
Luo Y, Xu X, Ye Z, Xu Q, Li J, Liu N, Du Y. 3D bioprinted mesenchymal stromal cells in skin wound repair. Front Surg 2022; 9:988843. [PMID: 36311952 PMCID: PMC9614372 DOI: 10.3389/fsurg.2022.988843] [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: 07/07/2022] [Accepted: 09/20/2022] [Indexed: 11/07/2022] Open
Abstract
Skin tissue regeneration and repair is a complex process involving multiple cell types, and current therapies are limited to promoting skin wound healing. Mesenchymal stromal cells (MSCs) have been proven to enhance skin tissue repair through their multidifferentiation and paracrine effects. However, there are still difficulties, such as the limited proliferative potential and the biological processes that need to be strengthened for MSCs in wound healing. Recently, three-dimensional (3D) bioprinting has been applied as a promising technology for tissue regeneration. 3D-bioprinted MSCs could maintain a better cell ability for proliferation and expression of biological factors to promote skin wound healing. It has been reported that 3D-bioprinted MSCs could enhance skin tissue repair through anti-inflammatory, cell proliferation and migration, angiogenesis, and extracellular matrix remodeling. In this review, we will discuss the progress on the effect of MSCs and 3D bioprinting on the treatment of skin tissue regeneration, as well as the perspective and limitations of current research.
Collapse
|
42
|
Kim W, Kim GH. Highly Bioactive Cell-laden Hydrogel Constructs Bioprinted Using an Emulsion Bioink for Tissue Engineering Applications. Biofabrication 2022; 14. [PMID: 36067738 DOI: 10.1088/1758-5090/ac8fb8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Accepted: 09/06/2022] [Indexed: 11/11/2022]
Abstract
The insufficient pore structure of cell-laden hydrogel scaffolds has limited their application in various tissue regeneration applications owing to low cell-to-cell/matrix interactions and low transfer of nutrients and metabolic wastes. Herein, we designed a highly porous cell-laden hydrogel scaffold fabricated using an emulsion bioink consisting of methacrylated collagen (CMA), mineral oil (MO), and human adipose stem cells (hASCs) to induce efficient cell infiltration and cellular activities. By selecting the most appropriate concentration of CMA and MO, the emulsion bioink can be successfully formulated with proper yield stress and printability. The cell-laden scaffold exhibited significantly greater cell growth and cytoskeletal reorganization than the normally printed cell-laden CMA scaffold. Furthermore, two bioactive components (kartogenin and bone morphogenetic protein 2) were physically encapsulated in the oil droplets of the cell construct, and the molecules in the cell constructs enhanced chondrogenic or osteogenic differentiation of hASCs in the printed structure. Based on these results, the cell-printed structure using an emulsion bioink can not only provide a good cellular microenvironment but also be a new potential method to accelerate stem cell differentiation by combining bioactive molecules and cell-laden scaffolds.
Collapse
Affiliation(s)
- WonJin Kim
- Sungkyunkwan University - Suwon Campus, 2066 Seobu-Ro, Suwon, Gyeonggi-do, 440-746, Korea (the Republic of)
| | - Geun Hyung Kim
- Biomechatronic Eng., Sungkyunkwan University - Natural Sciences Campus, 2066 Seobu-ro, Suwon, Gyeonggi-do, 16419, Korea (the Republic of)
| |
Collapse
|
43
|
Basara G, Bahcecioglu G, Ozcebe SG, Ellis BW, Ronan G, Zorlutuna P. Myocardial infarction from a tissue engineering and regenerative medicine point of view: A comprehensive review on models and treatments. BIOPHYSICS REVIEWS 2022; 3:031305. [PMID: 36091931 PMCID: PMC9447372 DOI: 10.1063/5.0093399] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/08/2022] [Indexed: 05/12/2023]
Abstract
In the modern world, myocardial infarction is one of the most common cardiovascular diseases, which are responsible for around 18 million deaths every year or almost 32% of all deaths. Due to the detrimental effects of COVID-19 on the cardiovascular system, this rate is expected to increase in the coming years. Although there has been some progress in myocardial infarction treatment, translating pre-clinical findings to the clinic remains a major challenge. One reason for this is the lack of reliable and human representative healthy and fibrotic cardiac tissue models that can be used to understand the fundamentals of ischemic/reperfusion injury caused by myocardial infarction and to test new drugs and therapeutic strategies. In this review, we first present an overview of the anatomy of the heart and the pathophysiology of myocardial infarction, and then discuss the recent developments on pre-clinical infarct models, focusing mainly on the engineered three-dimensional cardiac ischemic/reperfusion injury and fibrosis models developed using different engineering methods such as organoids, microfluidic devices, and bioprinted constructs. We also present the benefits and limitations of emerging and promising regenerative therapy treatments for myocardial infarction such as cell therapies, extracellular vesicles, and cardiac patches. This review aims to overview recent advances in three-dimensional engineered infarct models and current regenerative therapeutic options, which can be used as a guide for developing new models and treatment strategies.
Collapse
Affiliation(s)
- Gozde Basara
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Gokhan Bahcecioglu
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - S. Gulberk Ozcebe
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Bradley W Ellis
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - George Ronan
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Pinar Zorlutuna
- Present address: 143 Multidisciplinary Research Building, University of Notre Dame, Notre Dame, IN 46556. Author to whom correspondence should be addressed:. Tel.: +1 574 631 8543. Fax: +1 574 631 8341
| |
Collapse
|
44
|
Raza A, Mumtaz M, Hayat U, Hussain N, Ghauri MA, Bilal M, Iqbal HM. Recent advancements in extrudable gel-based bioinks for biomedical settings. J Drug Deliv Sci Technol 2022. [DOI: 10.1016/j.jddst.2022.103697] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
45
|
Prabhakaran P, Palaniyandi T, Kanagavalli B, Ram Kumar V, Hari R, Sandhiya V, Baskar G, Rajendran BK, Sivaji A. Prospect and retrospect of 3D bio-printing. Acta Histochem 2022; 124:151932. [PMID: 36027838 DOI: 10.1016/j.acthis.2022.151932] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 07/23/2022] [Accepted: 07/23/2022] [Indexed: 11/01/2022]
Abstract
3D bioprinting has become a popular medical technique in recent years. The most compelling rationale for the development of 3D bioprinting is the paucity of biological structures required for the rehabilitation of missing organs and tissues. They're useful in a multitude of domains, including disease modelling, regenerative medicine, tissue engineering, drug discovery with testing, personalised medicine, organ development, toxicity studies, and implants. Bioprinting requires a range of bioprinting technologies and bioinks to finish their procedure, that Inkjet-based bioprinting, extrusion-based bioprinting, laser-assisted bioprinting, stereolithography-based bioprinting, and in situ bioprinting are some of the technologies listed here. Bioink is a 3D printing material that is used to construct engineered artificial living tissue. It can be constructed solely for cells, but it usually includes a carrier substance that envelops the cells, then there's Agarose-based bioinks, alginate-based bioinks, collagen-based bioinks, and hyaluronic acid-based bioinks, to name a few. Here we presented about the different bioprinting methods with the use of bioinks in it and then Prospected over various applications in different fields.
Collapse
Affiliation(s)
- Pranav Prabhakaran
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to University, Chennai, India
| | - Thirunavukkarsu Palaniyandi
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to University, Chennai, India; Department of Anatomy, Biomedical Reseach Unit and Laboratory Animal Centre, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India.
| | - B Kanagavalli
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to University, Chennai, India
| | - V Ram Kumar
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to University, Chennai, India
| | - Rajeswari Hari
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to University, Chennai, India
| | - V Sandhiya
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to University, Chennai, India
| | - Gomathy Baskar
- Department of Biotechnology, Dr. M.G.R Educational and Research Institute, Deemed to University, Chennai, India
| | | | - Asha Sivaji
- Department of Biochemistry, DKM College for Women, Vellore, India
| |
Collapse
|
46
|
Recent advances in 3D-printed polylactide and polycaprolactone-based biomaterials for tissue engineering applications. Int J Biol Macromol 2022; 218:930-968. [PMID: 35896130 DOI: 10.1016/j.ijbiomac.2022.07.140] [Citation(s) in RCA: 121] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Revised: 07/13/2022] [Accepted: 07/18/2022] [Indexed: 01/10/2023]
Abstract
The three-dimensional printing (3DP) also known as the additive manufacturing (AM), a novel and futuristic technology that facilitates the printing of multiscale, biomimetic, intricate cytoarchitecture, function-structure hierarchy, multi-cellular tissues in the complicated micro-environment, patient-specific scaffolds, and medical devices. There is an increasing demand for developing 3D-printed products that can be utilized for organ transplantations due to the organ shortage. Nowadays, the 3DP has gained considerable interest in the tissue engineering (TE) field. Polylactide (PLA) and polycaprolactone (PCL) are exemplary biomaterials with excellent physicochemical properties and biocompatibility, which have drawn notable attraction in tissue regeneration. Herein, the recent advancements in the PLA and PCL biodegradable polymer-based composites as well as their reinforcement with hydrogels and bio-ceramics scaffolds manufactured through 3DP are systematically summarized and the applications of bone, cardiac, neural, vascularized and skin tissue regeneration are thoroughly elucidated. The interaction between implanted biodegradable polymers, in-vivo and in-vitro testing models for possible evaluation of degradation and biological properties are also illustrated. The final section of this review incorporates the current challenges and future opportunities in the 3DP of PCL- and PLA-based composites that will prove helpful for biomedical engineers to fulfill the demands of the clinical field.
Collapse
|
47
|
Li Y, Ye Z, Zhang J, Zhao Y, Zhu T, Song J, Xu F, Li F. In Situ and Quantitative Monitoring of Cardiac Tissues Using Programmable Scanning Electrochemical Microscopy. Anal Chem 2022; 94:10515-10523. [PMID: 35822575 DOI: 10.1021/acs.analchem.2c01919] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In vitro cardiac tissue model holds great potential as a powerful platform for drug screening. Respiratory activity, contraction frequency, and extracellular H2O2 levels are the three key parameters for determining the physiological functions of cardiac tissues, which are technically challenging to be monitored in an in situ and quantitative manner. Herein, we constructed an in vitro cardiac tissue model on polyacrylamide gels and applied a pulsatile electrical field to promote the maturation of the cardiac tissue. Then, we built a scanning electrochemical microscopy (SECM) platform with programmable pulse potentials to in situ characterize the dynamic changes in the respiratory activity, contraction frequency, and extracellular H2O2 level of cardiac tissues under both normal physiological and drug (isoproterenol and propranolol) treatment conditions using oxygen, ferrocenecarboxylic acid (FcCOOH), and H2O2 as the corresponding redox mediators. The SECM results showed that isoproterenol treatment induced enhanced oxygen consumption, accelerated contractile frequency, and increased released H2O2 level, while propranolol treatment induced dynamically decreased oxygen consumption and contractile frequency and no obvious change in H2O2 levels, suggesting the effects of activation and inhibition of β-adrenoceptor on the metabolic and electrophysiological activities of cardiac tissues. Our work realizes the in situ and quantitative monitoring of respiratory activity, contraction frequency, and secreted H2O2 level of living cardiac tissues using SECM for the first time. The programmable SECM methodology can also be used to real-time and quantitatively monitor electrochemical and electrophysiological parameters of cardiac tissues for future drug screening studies.
Collapse
Affiliation(s)
- Yabei Li
- School of Chemistry, Xi'an Jiaotong University, Xi'an 710049, P. R. China.,Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Zhaoyang Ye
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P. R. China.,The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Junjie Zhang
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P. R. China.,The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Yuxiang Zhao
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P. R. China.,The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Tong Zhu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P. R. China.,The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China.,Department of Cardiovasology, Xidian Group Hospital, Xi'an, Shaanxi Province 710077, P. R. China
| | - Jingjing Song
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P. R. China.,The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Feng Xu
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P. R. China.,The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| | - Fei Li
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an 710049, P. R. China.,The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an 710049, P. R. China
| |
Collapse
|
48
|
Kit O, Shikhlyarova A, Frantsiyants E, Neskubina I, Kaplieva I, Zhukova G, Trepitaki L, Pogorelova Y, Bandovkina V, Surikova E, Popov I, Voronina T, Bykadorova O, Serdyukova E. Mitochondrial therapy: direct visual assessment of the possibility of preventing myocardial infarction under chronic neurogenic pain and B16 melanoma growth in the experiment. CARDIOMETRY 2022. [DOI: 10.18137/cardiometry.2022.22.3849] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
On models of chronic neurogenic pain (CNP) and the growth of a malignant tumor (metastasizing B16 melanoma) in male mice, we studied an effect produced by mitochondrial therapy (MCT) on the state of the myocardium. Some structural correlates of the compensatory-restorative effect by mitochondria transplanted from healthy recipient rats were revealed. It has been identified that MCT contributes to the preservation of the structural integrity of the myocardial tissue, the inclusion of an auxiliary link in the cellular mechanisms of tissue restoration: fibroblasts, histiocytes, lymphocytes, eosinophils and other connective tissue elements, which implement the intercellular mechanism of information transfer that provides the external regulatory function of MCT. The ability of mitochondria to prevent the DNA decay determines the possibility of initiation of the operation of the nuclear mechanisms of the cardiomyocyte division, which is characteristic of a population of young cells and which indicates the determining position of exogenous mitochondria.
Collapse
|
49
|
Hu Y, Jia Y, Wang H, Cao Q, Yang Y, Zhou Y, Tan T, Huang X, Zhou Q. Low-intensity pulsed ultrasound promotes cell viability and inhibits apoptosis of H9C2 cardiomyocytes in 3D bioprinting scaffolds via PI3K-Akt and ERK1/2 pathways. J Biomater Appl 2022; 37:402-414. [PMID: 35574901 DOI: 10.1177/08853282221102669] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The aim of this study was to investigate whether low-intensity pulsed ultrasound (LIPUS) promotes myocardial cell viability in three-dimensional (3D) cell-laden gelatin methacryloyl (GelMA) scaffolds. Cardiomyoblasts (H9C2s) were mixed in 6% (w/v) GelMA bio-inks and printed using an extrusion-based 3D bioprinter. These scaffolds were exposed to LIPUS with different parameters or sham-irradiated to optimize the LIPUS treatment. The viability of H9C2s was measured using Cell Counting Kit-8 (CCK8), cell cycle, and live and dead cell double-staining assays. Western blot analysis was performed to determine the protein expression levels. We successfully fabricated 3D bio-printed cell-laden GelMA scaffolds. CCK8 and live and dead cell double-staining assays indicated that the optimal conditions for LIPUS were a frequency of 0.5 MHz and an exposure time of 10 min. Cell cycle analysis showed that LIPUS promoted the entry of cells into the S and G2/M phases from the G0/G1 phase. Western blot analysis revealed that LIPUS promoted the phosphorylation and activation of ERK1/2 and PI3K-Akt. The ERK1/2 inhibitor (U0126) and PI3K inhibitor (LY294002) significantly reduced LIPUS-induced phosphorylation of ERK1/2 and PI3K-Akt, respectively, which in turn reduced the LIPUS-induced viability of H9C2s in 3D bio-printed cell-laden GelMA scaffolds. A frequency of 0.5 MHz and exposure time of 10 min for LIPUS exposure can be adapted to achieve optimized culture effects on myocardial cells in 3D bio-printed cell-laden GelMA scaffolds via the ERK1/2 and PI3K-Akt signaling pathways.
Collapse
Affiliation(s)
- Yugang Hu
- Department of Ultrasound Imaging, 117921Renmin Hospital of Wuhan University, Wuhan, China
| | - Yan Jia
- Department of Ultrasound Imaging, 117921Renmin Hospital of Wuhan University, Wuhan, China
| | - Hao Wang
- Department of Ultrasound Imaging, 117921Renmin Hospital of Wuhan University, Wuhan, China
| | - Quan Cao
- Department of Ultrasound Imaging, 117921Renmin Hospital of Wuhan University, Wuhan, China
| | - Yuanting Yang
- Department of Ultrasound Imaging, 117921Renmin Hospital of Wuhan University, Wuhan, China
| | - Yanxiang Zhou
- Department of Ultrasound Imaging, 117921Renmin Hospital of Wuhan University, Wuhan, China
| | - Tuantuan Tan
- Department of Ultrasound Imaging, 117921Renmin Hospital of Wuhan University, Wuhan, China
| | - Xin Huang
- Department of Ultrasound Imaging, 117921Renmin Hospital of Wuhan University, Wuhan, China
| | - Qing Zhou
- Department of Ultrasound Imaging, 117921Renmin Hospital of Wuhan University, Wuhan, China
| |
Collapse
|
50
|
Zhu L, Liu K, Feng Q, Liao Y. Cardiac Organoids: A 3D Technology for Modeling Heart Development and Disease. Stem Cell Rev Rep 2022; 18:2593-2605. [PMID: 35525908 DOI: 10.1007/s12015-022-10385-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 04/29/2022] [Indexed: 12/14/2022]
Abstract
Cardiac organoids (COs) are miniaturized and simplified organ structures that can be used in heart development biology, drug screening, disease modeling, and regenerative medicine. This cardiac organoid (CO) model is revolutionizing our perspective on answering major cardiac physiology and pathology issues. Recently, many research groups have reported various methods for modeling the heart in vitro. However, there are differences in methodologies and concepts. In this review, we discuss the recent advances in cardiac organoid technologies derived from human embryonic stem cells (hESCs) and human-induced pluripotent stem cells (hiPSCs), with a focus on the summary of methods for organoid generation. In addition, we introduce CO applications in modeling heart development and cardiovascular diseases and discuss the prospects for and common challenges of CO that still need to be addressed. A detailed understanding of the development of CO will help us design better methods, explore and expand its application in the cardiovascular field.
Collapse
Affiliation(s)
- Liyuan Zhu
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Kui Liu
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China
| | - Qi Feng
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | - Yingnan Liao
- Xiamen Key Laboratory of Cardiovascular Disease, Xiamen Cardiovascular Hospital of Xiamen University, School of Medicine, Xiamen University, Xiamen, China.
| |
Collapse
|