1
|
Park CH, Park JH, Suh YJ. Perspective of 3D culture in medicine: transforming disease research and therapeutic applications. Front Bioeng Biotechnol 2024; 12:1491669. [PMID: 39749112 PMCID: PMC11693738 DOI: 10.3389/fbioe.2024.1491669] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 12/06/2024] [Indexed: 01/04/2025] Open
Abstract
3D cell culture is gaining momentum in medicine due to its ability to mimic real tissues (in vivo) and provide more accurate biological data compared to traditional methods. This review explores the current state of 3D cell culture in medicine and discusses future directions, including the need for standardization and simpler protocols to facilitate wider use in research. Purpose 3D cell culture develops life sciences by mimicking the natural cellular environment. Cells in 3D cultures grow in three dimensions and interact with a matrix, fostering realistic cell behavior and interactions. This enhanced model offers significant advantages for diverse research areas. Methods By mimicking the cellular organization and functionalities found in human tissues, 3D cultures provide superior platforms for studying complex diseases like cancer and neurodegenerative disorders. This enables researchers to gain deeper insights into disease progression and identify promising therapeutic targets with greater accuracy. 3D cultures also play a crucial role in drug discovery by allowing researchers to effectively assess potential drugs' safety and efficacy. Results 3D cell culture's impact goes beyond disease research. It holds promise for tissue engineering. By replicating the natural tissue environment and providing a scaffold for cell growth, 3D cultures pave the way for regenerating damaged tissues, offering hope for treating burns, organ failure, and musculoskeletal injuries. Additionally, 3D cultures contribute to personalized medicine. Researchers can use patient-derived cells to create personalized disease models and identify the most effective treatment for each individual. Conclusion With ongoing advancements in cell imaging techniques, the development of novel biocompatible scaffolds and bioreactor systems, and a deeper understanding of cellular behavior within 3D environments, 3D cell culture technology stands poised to revolutionize various aspects of healthcare and scientific discovery.
Collapse
Affiliation(s)
- Chan Hum Park
- Nano-Bio Regenerative Medical Institute, College of Medicine, Hallym University, Chuncheon, Republic of Korea
- Departments of Otorhinolaryngology-Head and Neck Surgery, Chuncheon Sacred Heart Hospital, School of Medicine, Hallym University, Chuncheon, Republic of Korea
| | - Jung Ho Park
- Department of Breast and Endocrine Surgery, Hallym University Sacred Heart Hospital, Anyang, Republic of Korea
| | - Yong Joon Suh
- Department of Breast and Endocrine Surgery, Hallym University Sacred Heart Hospital, Anyang, Republic of Korea
| |
Collapse
|
2
|
Zhu J, Luo Q, Yang G, Xiao L. Biofabrication of Tissue-Engineered Cartilage Constructs Through Faraday Wave Bioassembly of Cell-Laden Gelatin Microcarriers. Adv Healthc Mater 2024; 13:e2304541. [PMID: 38762758 DOI: 10.1002/adhm.202304541] [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: 12/19/2023] [Revised: 04/30/2024] [Indexed: 05/20/2024]
Abstract
Acoustic biofabrication is an emerging strategy in tissue engineering due to its mild and fast manufacturing process. Herein, tissue-engineered cartilage constructs with high cell viability are fabricated from cell-laden gelatin microcarriers (GMs) through Faraday wave bioassembly, a typical acoustic "bottom-up" manufacturing process. Assembly modules are first prepared by incorporating cartilage precursor cells, the chondrogenic cell line ATDC5, or bone marrow-derived mesenchymal stem cells (BMSCs), into GMs. Patterned structures are formed by Faraday wave bioassembly of the cell-laden GMs. Due to the gentle and efficient assembly process and the protective effects of microcarriers, cells in the patterned structures maintain high activity. Subsequently, tissue-engineered cartilage constructs are obtained by inducing cell differentiation of the patterned structures. Comprehensive evaluations are conducted to verify chondrocyte differentiation and the formation of cartilage tissue constructs in terms of cell viability, morphological analysis, gene expression, and matrix production. Finally, implantation studies with a rat cartilage defect model demonstrate that these tissue-engineered cartilage constructs are beneficial for the repair of articular cartilage damage in vivo. This study provides the first biofabrication of cartilage tissue constructs using Faraday wave bioassembly, extending its application to engineering tissues with a low cell density.
Collapse
Affiliation(s)
- Jing Zhu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Qiuchen Luo
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| | - Guang Yang
- Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan, 430074, China
| | - Lin Xiao
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, 518107, China
| |
Collapse
|
3
|
Xuan L, Hou Y, Liang L, Wu J, Fan K, Lian L, Qiu J, Miao Y, Ravanbakhsh H, Xu M, Tang G. Microgels for Cell Delivery in Tissue Engineering and Regenerative Medicine. NANO-MICRO LETTERS 2024; 16:218. [PMID: 38884868 PMCID: PMC11183039 DOI: 10.1007/s40820-024-01421-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/11/2024] [Accepted: 04/26/2024] [Indexed: 06/18/2024]
Abstract
Microgels prepared from natural or synthetic hydrogel materials have aroused extensive attention as multifunctional cells or drug carriers, that are promising for tissue engineering and regenerative medicine. Microgels can also be aggregated into microporous scaffolds, promoting cell infiltration and proliferation for tissue repair. This review gives an overview of recent developments in the fabrication techniques and applications of microgels. A series of conventional and novel strategies including emulsification, microfluidic, lithography, electrospray, centrifugation, gas-shearing, three-dimensional bioprinting, etc. are discussed in depth. The characteristics and applications of microgels and microgel-based scaffolds for cell culture and delivery are elaborated with an emphasis on the advantages of these carriers in cell therapy. Additionally, we expound on the ongoing and foreseeable applications and current limitations of microgels and their aggregate in the field of biomedical engineering. Through stimulating innovative ideas, the present review paves new avenues for expanding the application of microgels in cell delivery techniques.
Collapse
Affiliation(s)
- Leyan Xuan
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Yingying Hou
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Lu Liang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Jialin Wu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Kai Fan
- School of Automation, Hangzhou Dianzi University, Hangzhou, 310018, People's Republic of China
| | - Liming Lian
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Jianhua Qiu
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Yingling Miao
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China
| | - Hossein Ravanbakhsh
- Department of Biomedical Engineering, The University of Akron, Akron, OH, 44325, USA.
| | - Mingen Xu
- School of Automation, Hangzhou Dianzi University, Hangzhou, 310018, People's Republic of China.
| | - Guosheng Tang
- Guangzhou Municipal and Guangdong Provincial Key Laboratory of Molecular Target and Clinical Pharmacology, the NMPA and State Key Laboratory of Respiratory Disease, School of Pharmaceutical Sciences and the Fifth Affiliated Hospital, Guangzhou Medical University, Guangzhou, 511436, People's Republic of China.
| |
Collapse
|
4
|
Kwon HC, Jung HS, Kothuri V, Han SG. Current status and challenges for cell-cultured milk technology: a systematic review. J Anim Sci Biotechnol 2024; 15:81. [PMID: 38849927 PMCID: PMC11161985 DOI: 10.1186/s40104-024-01039-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2024] [Accepted: 04/22/2024] [Indexed: 06/09/2024] Open
Abstract
Cellular agriculture is an innovative technology for manufacturing sustainable agricultural products as an alternative to traditional agriculture. While most cellular agriculture is predominantly centered on the production of cultured meat, there is a growing demand for an understanding of the production techniques involved in dairy products within cellular agriculture. This review focuses on the current status of cellular agriculture in the dairy sector and technical challenges for cell-cultured milk production. Cellular agriculture technology in the dairy sector has been classified into fermentation-based and animal cell culture-based cellular agriculture. Currently, various companies synthesize milk components through precision fermentation technology. Nevertheless, several startup companies are pursuing animal cell-based technology, driven by public concerns regarding genetically modified organisms in precision fermentation technology. Hence, this review offers an up-to-date exploration of animal cell-based cellular agriculture to produce milk components, specifically emphasizing the structural, functional, and productive aspects of mammary epithelial cells, providing new information for industry and academia.
Collapse
Affiliation(s)
- Hyuk Cheol Kwon
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul, Republic of Korea
| | - Hyun Su Jung
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul, Republic of Korea
| | - Vahinika Kothuri
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul, Republic of Korea
| | - Sung Gu Han
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul, Republic of Korea.
| |
Collapse
|
5
|
Vaidya G, Pramanik S, Kadi A, Rayshan AR, Abualsoud BM, Ansari MJ, Masood R, Michaelson J. Injecting hope: chitosan hydrogels as bone regeneration innovators. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024; 35:756-797. [PMID: 38300215 DOI: 10.1080/09205063.2024.2304952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2023] [Accepted: 01/08/2024] [Indexed: 02/02/2024]
Abstract
Spontaneous bone regeneration encounters substantial restrictions in cases of bone defects, demanding external intervention to improve the repair and regeneration procedure. The field of bone tissue engineering (BTE), which embraces a range of disciplines, offers compelling replacements for conventional strategies like autografts, allografts, and xenografts. Among the diverse scaffolding materials utilized in BTE applications, hydrogels have demonstrated great promise as templates for the regeneration of bone owing to their resemblance to the innate extracellular matrix. In spite of the advancement of several biomaterials, chitosan (CS), a natural biopolymer, has garnered significant attention in recent years as a beneficial graft material for producing injectable hydrogels. Injectable hydrogels based on CS formulations provide numerous advantages, including their capacity to absorb and preserve a significant amount of water, their minimally invasive character, the existence of porous structures, and their capability to adapt accurately to irregular defects. Moreover, combining CS with other naturally derived or synthetic polymers and bioactive materials has displayed its effectiveness as a feasible substitute for traditional grafts. We aim to spotlight the composition, production, and physicochemical characteristics and practical utilization of CS-based injectable hydrogels, explicitly focusing on their potential implementations in bone regeneration. We consider this review a fundamental resource and a source of inspiration for future research attempts to pioneer the next era of tissue-engineering scaffold materials.
Collapse
Affiliation(s)
- Gayatri Vaidya
- Department of Studies and Research in Food Technology, Davangere University, Davangere, India
| | - Sheersha Pramanik
- Department of Biotechnology, Bhupat and Jyoti Mehta School of Biosciences, Indian Institute of Technology Madras, Chennai, India
| | - Ammar Kadi
- Department of Food and Biotechnology, South Ural State University, Chelyabinsk, Russia
| | - Ahmed Raheem Rayshan
- Department of Physiology, Pharmacology, and Biochemistry, College of Veterinary Medicine, University of Al-Qadisiyah, Al-Diwaniyah, Iraq
| | - Bassam M Abualsoud
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, Al-Ahliyya Amman University, Amman, Jordan
| | - Mohammad Javed Ansari
- Department of Pharmaceutics, College of Pharmacy, Prince Sattam Bin Abdulaziz University, Al-Kharj, Saudi Arabia
| | - Rehana Masood
- Department of Biochemistry, Shaheed Benazir Bhutto Women University, Peshawar, Pakistan
| | - Jacob Michaelson
- Saveetha School of Engineering, Saveetha Institute of Medical and Technical Sciences, Chennai, India
| |
Collapse
|
6
|
Samadi A, Moammeri A, Azimi S, Bustillo-Perez BM, Mohammadi MR. Biomaterial engineering for cell transplantation. BIOMATERIALS ADVANCES 2024; 158:213775. [PMID: 38252986 DOI: 10.1016/j.bioadv.2024.213775] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2023] [Revised: 12/27/2023] [Accepted: 01/12/2024] [Indexed: 01/24/2024]
Abstract
The current paradigm of medicine is mostly designed to block or prevent pathological events. Once the disease-led tissue damage occurs, the limited endogenous regeneration may lead to depletion or loss of function for cells in the tissues. Cell therapy is rapidly evolving and influencing the field of medicine, where in some instances attempts to address cell loss in the body. Due to their biological function, engineerability, and their responsiveness to stimuli, cells are ideal candidates for therapeutic applications in many cases. Such promise is yet to be fully obtained as delivery of cells that functionally integrate with the desired tissues upon transplantation is still a topic of scientific research and development. Main known impediments for cell therapy include mechanical insults, cell viability, host's immune response, and lack of required nutrients for the transplanted cells. These challenges could be divided into three different steps: 1) Prior to, 2) during the and 3) after the transplantation procedure. In this review, we attempt to briefly summarize published approaches employing biomaterials to mitigate the above technical challenges. Biomaterials are offering an engineerable platform that could be tuned for different classes of cell transplantation to potentially enhance and lengthen the pharmacodynamics of cell therapies.
Collapse
Affiliation(s)
- Amirmasoud Samadi
- Department of Chemical and Biomolecular Engineering, 6000 Interdisciplinary Science & Engineering Building (ISEB), Irvine, CA 92617, USA
| | - Ali Moammeri
- Department of Chemical Engineering, Virginia Tech, Blacksburg, VA 24061, USA
| | - Shamim Azimi
- Department of Chemical Engineering, Queen's University, Kingston, ON K7L 3N6, Canada
| | - Bexi M Bustillo-Perez
- Department of Chemical Engineering, Queen's University, Kingston, ON K7L 3N6, Canada
| | - M Rezaa Mohammadi
- Dale E. and Sarah Ann Fowler School of Engineering, Chapman University, Orange, CA 92866, USA.
| |
Collapse
|
7
|
Teryek M, Jadhav P, Bento R, Parekkadan B. 3D microcapsules for human bone marrow-derived mesenchymal stem cell biomanufacturing in a vertical-wheel bioreactor. BIOTECHNOL BIOPROC E 2024. [DOI: 10.1007/s12257-024-00069-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/01/2023] [Accepted: 05/28/2023] [Indexed: 01/06/2025]
|
8
|
Song X, Man J, Qiu Y, Wang J, Liu J, Li R, Zhang Y, Li J, Li J, Chen Y. Design, preparation, and characterization of lubricating polymer brushes for biomedical applications. Acta Biomater 2024; 175:76-105. [PMID: 38128641 DOI: 10.1016/j.actbio.2023.12.024] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/21/2023] [Accepted: 12/14/2023] [Indexed: 12/23/2023]
Abstract
The lubrication modification of biomedical devices significantly enhances the functionality of implanted interventional medical devices, thereby providing additional benefits for patients. Polymer brush coating provides a convenient and efficient method for surface modification while ensuring the preservation of the substrate's original properties. The current research has focused on a "trial and error" method to finding polymer brushes with superior lubricity qualities, which is time-consuming and expensive, as obtaining effective and long-lasting lubricity properties for polymer brushes is difficult. This review summarizes recent research advances in the biomedical field in the design, material selection, preparation, and characterization of lubricating and antifouling polymer brushes, which follow the polymer brush development process. This review begins by examining various approaches to polymer brush design, including molecular dynamics simulation and machine learning, from the fundamentals of polymer brush lubrication. Recent advancements in polymer brush design are then synthesized and potential avenues for future research are explored. Emphasis is placed on the burgeoning field of zwitterionic polymer brushes, and highlighting the broad prospects of supramolecular polymer brushes based on host-guest interactions in the field of self-repairing polymer brush applications. The review culminates by providing a summary of methodologies for characterizing the structural and functional attributes of polymer brushes. It is believed that a development approach for polymer brushes based on "design-material selection-preparation-characterization" can be created, easing the challenge of creating polymer brushes with high-performance lubricating qualities and enabling the on-demand creation of coatings. STATEMENT OF SIGNIFICANCE: Biomedical devices have severe lubrication modification needs, and surface lubrication modification by polymer brush coating is currently the most promising means. However, the design and preparation of polymer brushes often involves "iterative testing" to find polymer brushes with excellent lubrication properties, which is both time-consuming and expensive. This review proposes a polymer brush development process based on the "design-material selection-preparation-characterization" strategy and summarizes recent research advances and trends in the design, material selection, preparation, and characterization of polymer brushes. This review will help polymer brush researchers by alleviating the challenges of creating polymer brushes with high-performance lubricity and promises to enable the on-demand construction of polymer brush lubrication coatings.
Collapse
Affiliation(s)
- Xinzhong Song
- Key Laboratory of High Efficiency and Clean Mechanicalanufacture of Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan 250061, PR China; Key National Demonstration Center for Experimental Mechanical Engineering Education, Shandong University, Jinan 250061, PR China
| | - Jia Man
- Key Laboratory of High Efficiency and Clean Mechanicalanufacture of Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan 250061, PR China; Key National Demonstration Center for Experimental Mechanical Engineering Education, Shandong University, Jinan 250061, PR China.
| | - Yinghua Qiu
- Key Laboratory of High Efficiency and Clean Mechanicalanufacture of Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan 250061, PR China; Key National Demonstration Center for Experimental Mechanical Engineering Education, Shandong University, Jinan 250061, PR China
| | - Jiali Wang
- Qilu Hospital of Shandong University, Jinan 250012, PR China
| | - Jianing Liu
- Qilu Hospital of Shandong University, Jinan 250012, PR China
| | - Ruijian Li
- Qilu Hospital of Shandong University, Jinan 250012, PR China
| | - Yongqi Zhang
- Key Laboratory of High Efficiency and Clean Mechanicalanufacture of Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan 250061, PR China; Key National Demonstration Center for Experimental Mechanical Engineering Education, Shandong University, Jinan 250061, PR China
| | - Jianyong Li
- Key Laboratory of High Efficiency and Clean Mechanicalanufacture of Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan 250061, PR China; Key National Demonstration Center for Experimental Mechanical Engineering Education, Shandong University, Jinan 250061, PR China
| | - Jianfeng Li
- Key Laboratory of High Efficiency and Clean Mechanicalanufacture of Ministry of Education, School of Mechanical Engineering, Shandong University, Jinan 250061, PR China; Key National Demonstration Center for Experimental Mechanical Engineering Education, Shandong University, Jinan 250061, PR China
| | - Yuguo Chen
- Qilu Hospital of Shandong University, Jinan 250012, PR China
| |
Collapse
|
9
|
Ye Q, Wang J, Wang B, Zhao M, Wu Z, Liu X. Establishment of an in Vitro Three-Dimensional Vascularized Micro-Tumor Model and Screening of Chemotherapeutic Drugs. Technol Cancer Res Treat 2024; 23:15330338241286755. [PMID: 39311637 PMCID: PMC11425739 DOI: 10.1177/15330338241286755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/28/2024] Open
Abstract
Breast cancer is the most common malignancy in women worldwide, and major challenges in its treatment include drug resistance and metastasis. Three-dimensional cell culture systems have received widespread attention in drug discovery studies but existing models have limitations, that warrant the development of a simple and repeatable three-dimensional culture model for high-throughput screening. In this study, we designed a simple, reproducible, and highly efficient microencapsulated device to co-culture MCF-7 cells and HUVECs in microcapsules to establish an in vitro vascularized micro-tumor model for chemotherapeutic drug screening. First, to construct a three-dimensional micro-tumor model, cell encapsulation devices were created using three different sizes of flat-mouthed needles. Immunohistochemistry and immunofluorescence assays were conducted to determine vascular lumen formation. Cell proliferation was detected using the Cell Counting Kit-8 assay. Finally, to observe the drug reactions between the models, anticancer drugs (doxorubicin or paclitaxel) were added 12 h after the two-dimensional cultured cells were plated or 7 days after cell growth in the core-shell microcapsules. Vascularized micro-tumors were obtained after 14 days of three-dimensional culture. The proliferation rate in the three-dimensional cultured cells was slower than that of two-dimensional cultured cells. Three-dimensional cultured cells were more resistant to anticancer drugs than two-dimensional cultured cells. This novel sample encapsulation device formed core-shell microcapsules and can be used to successfully construct 3D vascularized micro-tumors in vitro. The three-dimensional culture model may provide a platform for drug screening and is valuable for studying tumor development and angiogenesis.
Collapse
Affiliation(s)
- Qian Ye
- Department of Pathological Anatomy, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Juanru Wang
- Department of Pathology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Boyang Wang
- Department of Pathological Anatomy, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| | - Min Zhao
- The Second People's Hospital of Hefei, Hefei Hospital Affiliated to Anhui Medical University, Hefei, Anhui, China
| | - Zhengsheng Wu
- Department of Pathological Anatomy, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
- Department of Pathology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xiaoli Liu
- Department of Pathological Anatomy, School of Basic Medical Sciences, Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
10
|
Luo Q, Shang K, Zhu J, Wu Z, Cao T, Ahmed AAQ, Huang C, Xiao L. Biomimetic cell culture for cell adhesive propagation for tissue engineering strategies. MATERIALS HORIZONS 2023; 10:4662-4685. [PMID: 37705440 DOI: 10.1039/d3mh00849e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/15/2023]
Abstract
Biomimetic cell culture, which involves creating a biomimetic microenvironment for cells in vitro by engineering approaches, has aroused increasing interest given that it maintains the normal cellular phenotype, genotype and functions displayed in vivo. Therefore, it can provide a more precise platform for disease modelling, drug development and regenerative medicine than the conventional plate cell culture. In this review, initially, we discuss the principle of biomimetic cell culture in terms of the spatial microenvironment, chemical microenvironment, and physical microenvironment. Then, the main strategies of biomimetic cell culture and their state-of-the-art progress are summarized. To create a biomimetic microenvironment for cells, a variety of strategies has been developed, ranging from conventional scaffold strategies, such as macroscopic scaffolds, microcarriers, and microgels, to emerging scaffold-free strategies, such as spheroids, organoids, and assembloids, to simulate the native cellular microenvironment. Recently, 3D bioprinting and microfluidic chip technology have been applied as integrative platforms to obtain more complex biomimetic structures. Finally, the challenges in this area are discussed and future directions are discussed to shed some light on the community.
Collapse
Affiliation(s)
- Qiuchen Luo
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China.
| | - Keyuan Shang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China.
| | - Jing Zhu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China.
| | - Zhaoying Wu
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China.
| | - Tiefeng Cao
- Department of Gynaecology, First Affiliated Hospital of Sun Yat-Sen University, Guangzhou 510070, China
| | - Abeer Ahmed Qaed Ahmed
- Department of Molecular Medicine, Biochemistry Unit, University of Pavia, 27100 Pavia, Italy
| | - Chixiang Huang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China.
| | - Lin Xiao
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-Sen University, Shenzhen 518107, China.
| |
Collapse
|
11
|
Huang L, Jiang Y, Chen X, Zhang W, Luo Q, Chen S, Wang S, Weng F, Xiao L. Supramolecular Responsive Chitosan Microcarriers for Cell Detachment Triggered by Adamantane. Polymers (Basel) 2023; 15:4024. [PMID: 37836073 PMCID: PMC10574836 DOI: 10.3390/polym15194024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2023] [Revised: 09/30/2023] [Accepted: 10/03/2023] [Indexed: 10/15/2023] Open
Abstract
Supramolecular responsive microcarriers based on chitosan microspheres were prepared and applied for nonenzymatic cell detachment. Briefly, chitosan microspheres (CSMs) were first prepared by an emulsion crosslinking approach, the surface of which was then modified with β-cyclodextrin (β-CD) by chemical grafting. Subsequently, gelatin was attached onto the surface of the CSMs via the host-guest interaction between β-CD groups and aromatic residues in gelatin. The resultant microspheres were denoted CSM-g-CD-Gel. Due to their superior biocompatibility and gelatin niches, CSM-g-CD-Gel microspheres can be used as effective microcarriers for cell attachment and expansion. L-02, a human fetal hepatocyte line, was used to evaluate cell attachment and expansion with these microcarriers. After incubation for 48 h, the cells attached and expanded to cover the entire surface of microcarriers. Moreover, with the addition of adamantane (AD), cells can be detached from the microcarriers together with gelatin because of the competitive binding between β-CD and AD. Overall, these supramolecular responsive microcarriers could effectively support cell expansion and achieve nonenzymatic cell detachment and may be potentially reusable with a new cycle of gelatin attachment and detachment.
Collapse
Affiliation(s)
- Lixia Huang
- Hubei Key Laboratory of Purification and Application of Plant Anti-Cancer Active Ingredients, School of Chemistry and Life Sciences, Hubei University of Education, Wuhan 430205, China; (L.H.); (Y.J.); (S.C.); (F.W.)
| | - Yifei Jiang
- Hubei Key Laboratory of Purification and Application of Plant Anti-Cancer Active Ingredients, School of Chemistry and Life Sciences, Hubei University of Education, Wuhan 430205, China; (L.H.); (Y.J.); (S.C.); (F.W.)
| | - Xinying Chen
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (W.Z.); (Q.L.)
| | - Wenqi Zhang
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (W.Z.); (Q.L.)
| | - Qiuchen Luo
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (W.Z.); (Q.L.)
| | - Siyan Chen
- Hubei Key Laboratory of Purification and Application of Plant Anti-Cancer Active Ingredients, School of Chemistry and Life Sciences, Hubei University of Education, Wuhan 430205, China; (L.H.); (Y.J.); (S.C.); (F.W.)
| | - Shuhan Wang
- Shenzhen Institute for Drug Control, Shenzhen Testing Center of Medical Devices, Shenzhen 518057, China;
| | - Fangqing Weng
- Hubei Key Laboratory of Purification and Application of Plant Anti-Cancer Active Ingredients, School of Chemistry and Life Sciences, Hubei University of Education, Wuhan 430205, China; (L.H.); (Y.J.); (S.C.); (F.W.)
| | - Lin Xiao
- School of Biomedical Engineering, Shenzhen Campus of Sun Yat-sen University, Shenzhen 518107, China; (W.Z.); (Q.L.)
| |
Collapse
|
12
|
Teryek M, Jadhav P, Bento R, Parekkadan B. High-Throughput Production of Microcapsules for Human Bone Marrow Derived Mesenchymal Stem Cell Biomanufacturing in a Vertical-Wheel Bioreactor. BIOTECHNOL BIOPROC E 2023; 28:528-544. [DOI: 10.1007/s12257-023-0020-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Revised: 05/02/2023] [Accepted: 05/28/2023] [Indexed: 01/06/2025]
|
13
|
Aitova A, Scherbina S, Berezhnoy A, Slotvitsky M, Tsvelaya V, Sergeeva T, Turchaninova E, Rybkina E, Bakumenko S, Sidorov I, Popov MA, Dontsov V, Agafonov EG, Efimov AE, Agapov I, Zybin D, Shumakov D, Agladze K. Novel Molecular Vehicle-Based Approach for Cardiac Cell Transplantation Leads to Rapid Electromechanical Graft-Host Coupling. Int J Mol Sci 2023; 24:10406. [PMID: 37373555 DOI: 10.3390/ijms241210406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2023] [Revised: 06/09/2023] [Accepted: 06/19/2023] [Indexed: 06/29/2023] Open
Abstract
Myocardial remodeling is an inevitable risk factor for cardiac arrhythmias and can potentially be corrected with cell therapy. Although the generation of cardiac cells ex vivo is possible, specific approaches to cell replacement therapy remain unclear. On the one hand, adhesive myocyte cells must be viable and conjugated with the electromechanical syncytium of the recipient tissue, which is unattainable without an external scaffold substrate. On the other hand, the outer scaffold may hinder cell delivery, for example, making intramyocardial injection difficult. To resolve this contradiction, we developed molecular vehicles that combine a wrapped (rather than outer) polymer scaffold that is enveloped by the cell and provides excitability restoration (lost when cells were harvested) before engraftment. It also provides a coating with human fibronectin, which initiates the process of graft adhesion into the recipient tissue and can carry fluorescent markers for the external control of the non-invasive cell position. In this work, we used a type of scaffold that allowed us to use the advantages of a scaffold-free cell suspension for cell delivery. Fragmented nanofibers (0.85 µm ± 0.18 µm in diameter) with fluorescent labels were used, with solitary cells seeded on them. Cell implantation experiments were performed in vivo. The proposed molecular vehicles made it possible to establish rapid (30 min) electromechanical contact between excitable grafts and the recipient heart. Excitable grafts were visualized with optical mapping on a rat heart with Langendorff perfusion at a 0.72 ± 0.32 Hz heart rate. Thus, the pre-restored grafts' excitability (with the help of a wrapped polymer scaffold) allowed rapid electromechanical coupling with the recipient tissue. This information could provide a basis for the reduction of engraftment arrhythmias in the first days after cell therapy.
Collapse
Affiliation(s)
- Aleria Aitova
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, Institutskiy Lane 9, 141700 Dolgoprudny, Russia
| | - Serafima Scherbina
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, Institutskiy Lane 9, 141700 Dolgoprudny, Russia
| | - Andrey Berezhnoy
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, Institutskiy Lane 9, 141700 Dolgoprudny, Russia
- M.F. Vladimirsky Moscow Regional Clinical Research Institute, Schepkina St. 61/2, 129110 Moscow, Russia
- Almetyevsk State Oil Institute, 2 Lenina St., 423450 Almetyevsk, Tatarstan, Russia
| | - Mikhail Slotvitsky
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, Institutskiy Lane 9, 141700 Dolgoprudny, Russia
- M.F. Vladimirsky Moscow Regional Clinical Research Institute, Schepkina St. 61/2, 129110 Moscow, Russia
- Almetyevsk State Oil Institute, 2 Lenina St., 423450 Almetyevsk, Tatarstan, Russia
| | - Valeriya Tsvelaya
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, Institutskiy Lane 9, 141700 Dolgoprudny, Russia
- M.F. Vladimirsky Moscow Regional Clinical Research Institute, Schepkina St. 61/2, 129110 Moscow, Russia
- Almetyevsk State Oil Institute, 2 Lenina St., 423450 Almetyevsk, Tatarstan, Russia
| | - Tatyana Sergeeva
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, Institutskiy Lane 9, 141700 Dolgoprudny, Russia
- Almetyevsk State Oil Institute, 2 Lenina St., 423450 Almetyevsk, Tatarstan, Russia
| | - Elena Turchaninova
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, Institutskiy Lane 9, 141700 Dolgoprudny, Russia
| | - Elizaveta Rybkina
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, Institutskiy Lane 9, 141700 Dolgoprudny, Russia
| | - Sergey Bakumenko
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, Institutskiy Lane 9, 141700 Dolgoprudny, Russia
- Almetyevsk State Oil Institute, 2 Lenina St., 423450 Almetyevsk, Tatarstan, Russia
| | - Ilya Sidorov
- Nanobiomedicine Division, Sirius University of Science and Technology, 1 Olympic Ave, 354340 Sochi, Russia
| | - Mikhail A Popov
- M.F. Vladimirsky Moscow Regional Clinical Research Institute, Schepkina St. 61/2, 129110 Moscow, Russia
| | - Vladislav Dontsov
- M.F. Vladimirsky Moscow Regional Clinical Research Institute, Schepkina St. 61/2, 129110 Moscow, Russia
| | - Evgeniy G Agafonov
- M.F. Vladimirsky Moscow Regional Clinical Research Institute, Schepkina St. 61/2, 129110 Moscow, Russia
| | - Anton E Efimov
- Academician V.I. Shumakov National Medical Research Center of Transplantology and Artificial Organs, Ministry of Health of the Russian Federation, 1 Schukinskaya St., 123182 Moscow, Russia
| | - Igor Agapov
- Academician V.I. Shumakov National Medical Research Center of Transplantology and Artificial Organs, Ministry of Health of the Russian Federation, 1 Schukinskaya St., 123182 Moscow, Russia
| | - Dmitriy Zybin
- M.F. Vladimirsky Moscow Regional Clinical Research Institute, Schepkina St. 61/2, 129110 Moscow, Russia
| | - Dmitriy Shumakov
- M.F. Vladimirsky Moscow Regional Clinical Research Institute, Schepkina St. 61/2, 129110 Moscow, Russia
| | - Konstantin Agladze
- Laboratory of Experimental and Cellular Medicine, Moscow Institute of Physics and Technology, Institutskiy Lane 9, 141700 Dolgoprudny, Russia
- M.F. Vladimirsky Moscow Regional Clinical Research Institute, Schepkina St. 61/2, 129110 Moscow, Russia
| |
Collapse
|
14
|
Guo C, Zhang T, Tang J, Gao C, Zhou Z, Li C. Construction of PLGA Porous Microsphere-Based Artificial Pancreatic Islets Assisted by the Cell Centrifugation Perfusion Technique. ACS OMEGA 2023; 8:15288-15297. [PMID: 37151553 PMCID: PMC10157690 DOI: 10.1021/acsomega.3c00424] [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: 01/20/2023] [Accepted: 04/06/2023] [Indexed: 05/09/2023]
Abstract
Pancreatic islet transplantation is a promising treatment that could potentially reverse diabetes, but its clinical applicability is severely limited by a shortage of organ donors. Various cell loading approaches using polymeric porous microspheres (PMs) have been developed for tissue regeneration; however, PM-based multicellular artificial pancreatic islets' construction has been scarcely reported. In this study, MIN6 (a mouse insulinoma cell line) and MS1 (a mouse pancreatic islet endothelial cell line) cells were seeded into poly(lactic-co-glycolic acid) (PLGA) PMs via an upgraded centrifugation-based cell perfusion seeding technique invented and patented by our group. Cell morphology, distribution, viability, migration, and proliferation were all evaluated. Results from glucose-stimulated insulin secretion (GSIS) assay and RNA-seq analysis suggested that MIN6 and MS1-loaded PLGA PMs exhibited better glucose responsiveness, which is partly attributable to vascular formation during PM-dependent islet construction. The present study suggests that the PLGA PM-based artificial pancreatic islets may provide an alternative strategy for the potential treatment of diabetes in the future.
Collapse
Affiliation(s)
- Chuanjia Guo
- Biomedical
Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union
Medical College, Tianjin Key Laboratory of Biomedical Materials, Tianjin 300192, China
| | - Tong Zhang
- Clinical
Laboratory, Tianjin Hospital, Tianjin 300211, China
| | - Jianghai Tang
- Biomedical
Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union
Medical College, Tianjin Key Laboratory of Biomedical Materials, Tianjin 300192, China
| | - Chang Gao
- Biomedical
Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union
Medical College, Tianjin Key Laboratory of Biomedical Materials, Tianjin 300192, China
| | - Zhimin Zhou
- Biomedical
Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union
Medical College, Tianjin Key Laboratory of Biomedical Materials, Tianjin 300192, China
- ,
| | - Chen Li
- Biomedical
Barriers Research Center, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences & Peking Union
Medical College, Tianjin Key Laboratory of Biomedical Materials, Tianjin 300192, China
| |
Collapse
|
15
|
Man K, Eisenstein NM, Hoey DA, Cox SC. Bioengineering extracellular vesicles: smart nanomaterials for bone regeneration. J Nanobiotechnology 2023; 21:137. [PMID: 37106449 PMCID: PMC10134574 DOI: 10.1186/s12951-023-01895-2] [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: 01/12/2023] [Accepted: 04/13/2023] [Indexed: 04/29/2023] Open
Abstract
In the past decade, extracellular vesicles (EVs) have emerged as key regulators of bone development, homeostasis and repair. EV-based therapies have the potential to circumnavigate key issues hindering the translation of cell-based therapies including functional tissue engraftment, uncontrolled differentiation and immunogenicity issues. Due to EVs' innate biocompatibility, low immunogenicity, and high physiochemical stability, these naturally-derived nanoparticles have garnered growing interest as potential acellular nanoscale therapeutics for a variety of diseases. Our increasing knowledge of the roles these cell-derived nanoparticles play, has made them an exciting focus in the development of novel pro-regenerative therapies for bone repair. Although these nano-sized vesicles have shown promise, their clinical translation is hindered due to several challenges in the EV supply chain, ultimately impacting therapeutic efficacy and yield. From the biochemical and biophysical stimulation of parental cells to the transition to scalable manufacture or maximising vesicles therapeutic response in vivo, a multitude of techniques have been employed to improve the clinical efficacy of EVs. This review explores state of the art bioengineering strategies to promote the therapeutic utility of vesicles beyond their native capacity, thus maximising the clinical potential of these pro-regenerative nanoscale therapeutics for bone repair.
Collapse
Affiliation(s)
- Kenny Man
- School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK
| | - Neil M Eisenstein
- Research and Clinical Innovation, Royal Centre for Defence Medicine, ICT Centre, Vincent Drive, Birmingham, B15 2SQ, UK
- Institute of Translational Medicine, University of Birmingham, Heritage Building, Mindelsohn Way, Birmingham, B15 2TH, UK
| | - David A Hoey
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College, Dublin, D02 R590, Ireland
- Dept. of Mechanical, Manufacturing, and Biomedical Engineering, School of Engineering, Trinity College, Dublin 2, D02 DK07, Dublin, Ireland
- Advanced Materials and Bioengineering Research Centre, Trinity College Dublin & RCSI, Dublin 2, D02 VN51, Dublin, Ireland
| | - Sophie C Cox
- School of Chemical Engineering, University of Birmingham, Birmingham, B15 2TT, UK.
| |
Collapse
|
16
|
Ma M, Zou F, Abudureheman B, Han F, Xu G, Xie Y, Qiao K, Peng J, Guan Y, Meng H, Zheng Y. Magnetic Microcarriers with Accurate Localization and Proliferation of Mesenchymal Stem Cell for Cartilage Defects Repairing. ACS NANO 2023; 17:6373-6386. [PMID: 36961738 DOI: 10.1021/acsnano.2c10995] [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] [Indexed: 06/18/2023]
Abstract
Magnetic biomaterials are widely used in the field of tissue engineering because of their functions such as drug delivery and targeted therapy. In this study, a magnetically responsive composite microcarrier was prepared through in situ polymerization of dopamine with Fe3O4 (MS) to form a complex. The magnetic composite microcarriers are paramagnetic and have certain magnetic responsiveness, suitable pore size porosity for cell growth, and good blood compatibility and biocompatibility. The bone marrow mesenchyml stem cells (BMSCs) were cultured on magnetic composite microcarriers, and a static magnetic field (SMF) was applied. The results showed that BMSCs adhered to the microcarriers proliferated under the action of horizontal and vertical forces. Magnetic composite microcarriers loaded with BMSCs were implanted into the SD rat model of cartilage defect, and a magnet was added to the operative side. After 12 weeks, cartilage regeneration was observed. The results of gross observation and histological immunostaining 1 month, 2 months, and 3 mounths after operation showed that the magnetic composite microcarriers of loaded cells promoted the early maturation of cartilage and collagen secretion, and the effect of cartilage repair was significantly better than that of the control group. Gait analysis showed that implanting magnetic composite microcarriers loaded with stem cells can reduce postoperative pain and promote limb recovery in SD rats. In conclusion, this study suggests that magnetic composite microcarriers are promising tissue-engineered scaffolds for cartilage regeneration and repair.
Collapse
Affiliation(s)
- Mengjiao Ma
- School of Material Science and Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Faxing Zou
- School of Material Science and Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Bahatibieke Abudureheman
- School of Material Science and Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Feng Han
- Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma &War Injuries, PLA Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China
| | - Guoli Xu
- School of Material Science and Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - YaJie Xie
- School of Material Science and Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Kun Qiao
- Beijing Gerecov Technology Company Ltd., Beijing 100142, China
| | - Jiang Peng
- Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma &War Injuries, PLA Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China
| | - Yueping Guan
- School of Material Science and Engineering, University of Science and Technology Beijing, Beijing 100083, China
| | - Haoye Meng
- School of Material Science and Engineering, University of Science and Technology Beijing, Beijing 100083, China
- Beijing Key Lab of Regenerative Medicine in Orthopaedics, Key Laboratory of Musculoskeletal Trauma &War Injuries, PLA Institute of Orthopedics, Chinese PLA General Hospital, Beijing 100853, China
| | - Yudong Zheng
- School of Material Science and Engineering, University of Science and Technology Beijing, Beijing 100083, China
| |
Collapse
|
17
|
Gruhn T, Monsalve CO, Müller C, Heid S, Boccaccini AR, Salehi S. Fabrication of Hydrogel-Based Composite Fibers and Computer Simulation of the Filler Dynamics in the Composite Flow. Bioengineering (Basel) 2023; 10:bioengineering10040448. [PMID: 37106635 PMCID: PMC10135958 DOI: 10.3390/bioengineering10040448] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/21/2023] [Accepted: 04/02/2023] [Indexed: 04/29/2023] Open
Abstract
Fibrous structures with anisotropic fillers as composites have found increasing interest in the field of biofabrication since they can mimic the extracellular matrix of anisotropic tissues such as skeletal muscle or nerve tissue. In the present work, the inclusion of anisotropic fillers in hydrogel-based filaments with an interpenetrating polymeric network (IPN) was evaluated and the dynamics of such fillers in the composite flow were analyzed using computational simulations. In the experimental part, microfabricated rods (200 and 400 μm length, 50 μm width) were used as anisotropic fillers in extrusion of composite filaments using two techniques of wet spinning and 3D printing. Hydrogels such as oxidized alginate (ADA) and methacrylated gelatin (GelMA) were used as matrices. In the computational simulation, a combination of computational fluid dynamics and coarse-grained molecular dynamics was used to study the dynamics of rod-like fillers in the flow field of a syringe. It showed that, during the extrusion process, microrods are far from being well aligned. Instead, many of them tumble on their way through the needle leading to a random orientation in the fiber which was confirmed experimentally.
Collapse
Affiliation(s)
- Thomas Gruhn
- Department of Biomaterials, Faculty of Engineering Science, University of Bayreuth, Prof.-Rüdiger-Bormann Str. 1, 95447 Bayreuth, Germany
| | - Camilo Ortiz Monsalve
- Department of Biomaterials, Faculty of Engineering Science, University of Bayreuth, Prof.-Rüdiger-Bormann Str. 1, 95447 Bayreuth, Germany
- Invertec-eV, Gottlieb-Keim-Straße 60, 95448 Bayreuth, Germany
| | - Claudia Müller
- Department of Biomaterials, Faculty of Engineering Science, University of Bayreuth, Prof.-Rüdiger-Bormann Str. 1, 95447 Bayreuth, Germany
| | - Susanne Heid
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstraße 6, 91058 Erlangen, Germany
| | - Aldo R Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, Cauerstraße 6, 91058 Erlangen, Germany
| | - Sahar Salehi
- Department of Biomaterials, Faculty of Engineering Science, University of Bayreuth, Prof.-Rüdiger-Bormann Str. 1, 95447 Bayreuth, Germany
| |
Collapse
|
18
|
Samrot AV, Sathiyasree M, Rahim SBA, Renitta RE, Kasipandian K, Krithika Shree S, Rajalakshmi D, Shobana N, Dhiva S, Abirami S, Visvanathan S, Mohanty BK, Sabesan GS, Chinni SV. Scaffold Using Chitosan, Agarose, Cellulose, Dextran and Protein for Tissue Engineering-A Review. Polymers (Basel) 2023; 15:polym15061525. [PMID: 36987305 PMCID: PMC10054888 DOI: 10.3390/polym15061525] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2022] [Revised: 01/27/2023] [Accepted: 01/27/2023] [Indexed: 03/30/2023] Open
Abstract
Biological macromolecules like polysaccharides/proteins/glycoproteins have been widely used in the field of tissue engineering due to their ability to mimic the extracellular matrix of tissue. In addition to this, these macromolecules are found to have higher biocompatibility and no/lesser toxicity when compared to synthetic polymers. In recent years, scaffolds made up of proteins, polysaccharides, or glycoproteins have been highly used due to their tensile strength, biodegradability, and flexibility. This review is about the fabrication methods and applications of scaffolds made using various biological macromolecules, including polysaccharides like chitosan, agarose, cellulose, and dextran and proteins like soy proteins, zein proteins, etc. Biopolymer-based nanocomposite production and its application and limitations are also discussed in this review. This review also emphasizes the importance of using natural polymers rather than synthetic ones for developing scaffolds, as natural polymers have unique properties, like high biocompatibility, biodegradability, accessibility, stability, absence of toxicity, and low cost.
Collapse
Affiliation(s)
- Antony V Samrot
- School of Bioscience, Faculty of Medicine, Bioscience and Nursing, MAHSA University, Jalan SP2, Bandar Saujana Putra, Jenjarom 42610, Selangor, Malaysia
| | - Mahendran Sathiyasree
- Department of Biotechnology, School of Bio and Chemical Engineering Sathyabama Institute of Science and Technology, Chennai 600119, Tamil Nadu, India
| | - Sadiq Batcha Abdul Rahim
- Faculty of Engineering, Built Environment and IT, MAHSA University, Jalan SP2, Bandar Saujana Putra, Jenjarom 42610, Selangor, Malaysia
| | - Robinson Emilin Renitta
- Department of Food Processing, Karunya Institute of Technology and Science, Coimbatore 641114, Tamil Nadu, India
| | - Kasirajan Kasipandian
- Faculty of Engineering, Built Environment and IT, MAHSA University, Jalan SP2, Bandar Saujana Putra, Jenjarom 42610, Selangor, Malaysia
| | - Sivasuriyan Krithika Shree
- Department of Biotechnology, School of Bio and Chemical Engineering Sathyabama Institute of Science and Technology, Chennai 600119, Tamil Nadu, India
| | - Deenadhayalan Rajalakshmi
- Department of Biotechnology, School of Bio and Chemical Engineering Sathyabama Institute of Science and Technology, Chennai 600119, Tamil Nadu, India
| | - Nagarajan Shobana
- Department of Biotechnology, School of Bio and Chemical Engineering Sathyabama Institute of Science and Technology, Chennai 600119, Tamil Nadu, India
| | - Shanmugaboopathi Dhiva
- Department of Microbiology, Sree Narayana College, Alathur, Palakkad 678682, Kerala, India
| | - Sasi Abirami
- Department of Microbiology, Kamaraj College, Thoothukudi, Affiliated to Manonmaniam Sundaranar University, Thoothukudi 628003, Tamil Nadu, India
| | - Sridevi Visvanathan
- Unit of Biochemistry, Faculty of Medicine, AIMST University, Semeling, Bedong 08100, Kedah Darul Aman, Malaysia
| | - Basanta Kumar Mohanty
- Faculty of Medicine, Manipal University College Malaysia (MUCM), Jalan Padang Jambu, Bukit Baru 75150, Melaka, Malaysia
| | - Gokul Shankar Sabesan
- Faculty of Medicine, Manipal University College Malaysia (MUCM), Jalan Padang Jambu, Bukit Baru 75150, Melaka, Malaysia
| | - Suresh V Chinni
- Department of Biochemistry, Faculty of Medicine, Bioscience and Nursing, MAHSA University, Jalan SP2, Bandar Saujana Putra, Jenjarom 42610, Selangor, Malaysia
- Department of Periodontics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Chennai 602105, Tamil Nadu, India
| |
Collapse
|
19
|
Xiang Y, Yan J, Bao X, Gleadall A, Roach P, Sun T. Evaluation of Polymeric Particles for Modular Tissue Cultures in Developmental Engineering. Int J Mol Sci 2023; 24:ijms24065234. [PMID: 36982306 PMCID: PMC10049291 DOI: 10.3390/ijms24065234] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/03/2023] [Accepted: 03/07/2023] [Indexed: 03/30/2023] Open
Abstract
Developmental engineering (DE) aims to culture mammalian cells on corresponding modular scaffolds (scale: micron to millimeter), then assemble these into functional tissues imitating natural developmental biology processes. This research intended to investigate the influences of polymeric particles on modular tissue cultures. When poly(methyl methacrylate) (PMMA), poly(lactic acid) (PLA) and polystyrene (PS) particles (diameter: 5-100 µm) were fabricated and submerged in culture medium in tissue culture plastics (TCPs) for modular tissue cultures, the majority of adjacent PMMA, some PLA but no PS particles aggregated. Human dermal fibroblasts (HDFs) could be directly seeded onto large (diameter: 30-100 µm) PMMA particles, but not small (diameter: 5-20 µm) PMMA, nor all the PLA and PS particles. During tissue cultures, HDFs migrated from the TCPs surfaces onto all the particles, while the clustered PMMA or PLA particles were colonized by HDFs into modular tissues with varying sizes. Further comparisons revealed that HDFs utilized the same cell bridging and stacking strategies to colonize single or clustered polymeric particles, and the finely controlled open pores, corners and gaps on 3D-printed PLA discs. These observed cell-scaffold interactions, which were then used to evaluate the adaptation of microcarrier-based cell expansion technologies for modular tissue manufacturing in DE.
Collapse
Affiliation(s)
- Yu Xiang
- Department of Materials, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK
| | - Jiongyi Yan
- Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK
| | - Xujin Bao
- Department of Materials, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK
| | - Andrew Gleadall
- Wolfson School of Mechanical, Electrical and Manufacturing Engineering, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK
| | - Paul Roach
- Department of Chemistry, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK
| | - Tao Sun
- Department of Chemical Engineering, Loughborough University, Epinal Way, Loughborough LE11 3TU, UK
| |
Collapse
|
20
|
Teryek M, Jadhav P, Bento R, Parekkadan B. 3D Microcapsules for Human Bone Marrow Derived Mesenchymal Stem Cell Biomanufacturing in a Vertical-Wheel Bioreactor. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.16.528656. [PMID: 36824906 PMCID: PMC9949076 DOI: 10.1101/2023.02.16.528656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
Abstract
Microencapsulation of human mesenchymal stromal cells (MSCs) via electrospraying has been well documented in tissue engineering and regenerative medicine. Herein, we report the use of microencapsulation, via electrospraying, for MSC expansion using a commercially available hydrogel that is durable, optimized to MSC culture, and enzymatically degradable for cell recovery. Critical parameters of the electrospraying encapsulation process such as seeding density, correlation of microcapsule output with hydrogel volume, and applied voltage were characterized to consistently fabricate cell-laden microcapsules of uniform size. Upon encapsulation, we then verified ~ 10x expansion of encapsulated MSCs within a vertical-wheel bioreactor and the preservation of critical quality attributes such as immunophenotype and multipotency after expansion and cell recovery. Finally, we highlight the genetic manipulation of encapsulated MSCs as an example of incorporating bioactive agents in the capsule material to create new compositions of MSCs with altered phenotypes.
Collapse
Affiliation(s)
- Matthew Teryek
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Pankaj Jadhav
- Department of Chemical and Biochemical Engineering, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Raphaela Bento
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey 08854, USA
| | - Biju Parekkadan
- Department of Biomedical Engineering, Rutgers University, Piscataway, New Jersey 08854, USA
| |
Collapse
|
21
|
Casajuana Ester M, Day RM. Production and Utility of Extracellular Vesicles with 3D Culture Methods. Pharmaceutics 2023; 15:pharmaceutics15020663. [PMID: 36839984 PMCID: PMC9961751 DOI: 10.3390/pharmaceutics15020663] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 02/13/2023] [Accepted: 02/13/2023] [Indexed: 02/18/2023] Open
Abstract
In recent years, extracellular vesicles (EVs) have emerged as promising biomarkers, cell-free therapeutic agents, and drug delivery carriers. Despite their great clinical potential, poor yield and unscalable production of EVs remain significant challenges. When using 3D culture methods, such as scaffolds and bioreactors, large numbers of cells can be expanded and the cell environment can be manipulated to control the cell phenotype. This has been employed to successfully increase the production of EVs as well as to enhance their therapeutic effects. The physiological relevance of 3D cultures, such as spheroids, has also provided a strategy for understanding the role of EVs in the pathogenesis of several diseases and to evaluate their role as tools to deliver drugs. Additionally, 3D culture methods can encapsulate EVs to achieve more sustained therapeutic effects as well as prevent premature clearance of EVs to enable more localised delivery and concentrated exosome dosage. This review highlights the opportunities and drawbacks of different 3D culture methods and their use in EV research.
Collapse
|
22
|
Assessing Polysaccharides/Aloe Vera-Based Hydrogels for Tumor Spheroid Formation. Gels 2023; 9:gels9010051. [PMID: 36661817 PMCID: PMC9858450 DOI: 10.3390/gels9010051] [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: 12/10/2022] [Revised: 01/03/2023] [Accepted: 01/05/2023] [Indexed: 01/11/2023] Open
Abstract
In vitro tumor spheroids have proven to be useful 3D tumor culture models for drug testing, and determining the molecular mechanism of tumor progression and cellular interactions. Therefore, there is a continuous search for their industrial scalability and routine preparation. Considering that hydrogels are promising systems that can favor the formation of tumor spheroids, our study aimed to investigate and develop less expensive and easy-to-use amorphous and crosslinked hydrogels, based on natural compounds such as sodium alginate (NaAlg), aloe vera (AV) gel powder, and chitosan (CS) for tumor spheroid formation. The ability of the developed hydrogels to be a potential spheroid-forming system was evaluated using MDA-MB-231 and U87MG cancer cells. Spheroid abilities were influenced by pH, viscosity, and crosslinking of the hydrogel. Addition of either AV or chitosan to sodium alginate increased the viscosity at pH 5, resulting in amorphous hydrogels with a strong gel texture, as shown by rheologic analysis. Only the chitosan-based gel allowed formation of spheroids at pH 5. Among the variants of AV-based amorphous hydrogels tested, only hydrogels at pH 12 and with low viscosity promoted the formation of spheroids. The crosslinked NaAlg/AV, NaAlg/AV/glucose, and NaAlg/CS hydrogel variants favored more efficient spheroid formation. Additional studies would be needed to use AV in other physical forms and other formulations of hydrogels, as the current study is an initiation, in evaluating the potential use of AV gel in tumor spheroid formation systems.
Collapse
|
23
|
Tutty MA, Prina-Mello A. Three-Dimensional Spheroids for Cancer Research. Methods Mol Biol 2023; 2645:65-103. [PMID: 37202612 DOI: 10.1007/978-1-0716-3056-3_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
In vitro cell culture is one of the most widely used tools used today for increasing our understanding of various things such as protein production, mechanisms of drug action, tissue engineering, and overall cellular biology. For the past decades, however, cancer researchers have relied heavily on conventional two-dimensional (2D) monolayer culture techniques to test a variety of aspects of cancer research ranging from the cytotoxic effects of antitumor drugs to the toxicity of diagnostic dyes and contact tracers. However, many promising cancer therapies have either weak or no efficacy in real-life conditions, therefore delaying or stopping altogether their translating to the clinic. This is, in part, due to the reductionist 2D cultures used to test these materials, which lack appropriate cell-cell contacts, have altered signaling, do not represent the natural tumor microenvironment, and have different drug responses, due to their reduced malignant phenotype when compared to real in vivo tumors. With the most recent advances, cancer research has moved into 3D biological investigation. Three-dimensional (3D) cultures of cancer cells not only recapitulate the in vivo environment better than their 2D counterparts, but they have, in recent years, emerged as a relatively low-cost and scientifically accurate methodology for studying cancer. In this chapter, we highlight the importance of 3D culture, specifically 3D spheroid culture, reviewing some key methodologies for forming 3D spheroids, discussing the experimental tools that can be used in conjunction with 3D spheroids and finally their applications in cancer research.
Collapse
Affiliation(s)
- Melissa Anne Tutty
- Laboratory for Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, Dublin, Ireland.
| | - Adriele Prina-Mello
- Laboratory for Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, Dublin, Ireland
- Nanomedicine and Molecular Imaging Group, Trinity Translational Medicine Institute, (TTMI), School of Medicine, Trinity College Dublin, Dublin, Ireland
- Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, CRANN Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
24
|
Ding SL, Liu X, Zhao XY, Wang KT, Xiong W, Gao ZL, Sun CY, Jia MX, Li C, Gu Q, Zhang MZ. Microcarriers in application for cartilage tissue engineering: Recent progress and challenges. Bioact Mater 2022; 17:81-108. [PMID: 35386447 PMCID: PMC8958326 DOI: 10.1016/j.bioactmat.2022.01.033] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 01/18/2022] [Accepted: 01/19/2022] [Indexed: 12/11/2022] Open
Abstract
Successful regeneration of cartilage tissue at a clinical scale has been a tremendous challenge in the past decades. Microcarriers (MCs), usually used for cell and drug delivery, have been studied broadly across a wide range of medical fields, especially the cartilage tissue engineering (TE). Notably, microcarrier systems provide an attractive method for regulating cell phenotype and microtissue maturations, they also serve as powerful injectable carriers and are combined with new technologies for cartilage regeneration. In this review, we introduced the typical methods to fabricate various types of microcarriers and discussed the appropriate materials for microcarriers. Furthermore, we highlighted recent progress of applications and general design principle for microcarriers. Finally, we summarized the current challenges and promising prospects of microcarrier-based systems for medical applications. Overall, this review provides comprehensive and systematic guidelines for the rational design and applications of microcarriers in cartilage TE.
Collapse
Affiliation(s)
- Sheng-Long Ding
- Center of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Xin Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Xi-Yuan Zhao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Ke-Tao Wang
- Center of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Wei Xiong
- Center of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Zi-Li Gao
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Cheng-Yi Sun
- Center of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| | - Min-Xuan Jia
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Cheng Li
- Beijing Advanced Innovation Center for Big Data-Based Precision Medicine, School of Engineering Medicine, Beihang University, Beijing, 100083, China
| | - Qi Gu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
- Beijing Institute for Stem Cell and Regeneration, University of Chinese Academy of Sciences, Beijing, 100101, China
| | - Ming-Zhu Zhang
- Center of Foot and Ankle Surgery, Beijing Tongren Hospital, Capital Medical University, Beijing, 100730, China
| |
Collapse
|
25
|
Cruz-Maya I, Guarino V. 3D Scaffolds Fabrication via Bicomponent Microgels Assembly: Process Optimization and In Vitro Characterization. MICROMACHINES 2022; 13:1726. [PMID: 36296078 PMCID: PMC9607065 DOI: 10.3390/mi13101726] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Revised: 10/06/2022] [Accepted: 10/07/2022] [Indexed: 06/16/2023]
Abstract
In the last decade, different technological approaches have been proposed for the fabrication of 3D models suitable to evaluate in vitro cell response. Among them, electro fluid dynamic atomization (EFDA) belonging to the family of electro-assisted technologies allows for the dropping of polysaccharides and/or proteins solutions to produce micro-scaled hydrogels or microgels with the peculiar features of hydrogel-like materials (i.e., biocompatibility, wettability, swelling). In this work, a method to fabricate 3D scaffolds by the assembly of bicomponent microgels made of sodium alginate and gelatin was proposed. As first step, optical and scanning electron microscopy with the support of image analysis enabled to explore the basic properties of single blocks in terms of correlation between particle morphology and process parameters (i.e., voltage, flow rate, electrode gap, and needle diameter). Chemical analysis via ninhydrin essays and FTIR analysis confirmed the presence of gelatin, mostly retained by physical interactions into the alginate network mediated by electrostatic forces. In vitro tests confirmed the effect of biochemical signals exerted by the protein on the biological response of hMSCs cultured onto the microgels surface. Hence, it is concluded that alginate/gelatin microgels assemblies can efficiently work as 3D scaffolds able to support in vitro cells functions, thus providing a friendly microenvironment to investigate in vitro cell interactions.
Collapse
|
26
|
Patrick MD, Keys JF, Suresh Kumar H, Annamalai RT. Injectable nanoporous microgels generate vascularized constructs and support bone regeneration in critical-sized defects. Sci Rep 2022; 12:15811. [PMID: 36138042 PMCID: PMC9499928 DOI: 10.1038/s41598-022-19968-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2022] [Accepted: 09/07/2022] [Indexed: 11/09/2022] Open
Abstract
Large and aberrant bone fractures require ossification and concomitant vascularization for proper healing. Evidence indicates that osteogenesis and vessel growth are coupled in bone fractures. Although the synergistic role of endothelial cells has been recognized, vascularizing large bone grafts remains a challenge and has apprehended the clinical translation of engineered bone constructs. Here, we describe a facile method to fabricate vascularized constructs using chitosan and gelatin-based microgels that promote osteogenesis of human mesenchymal stromal cells (MSC) while supporting endothelial sprouting and network formation. The microgels are enzymatically degradable and had a high hydration rate with a volume swelling ratio of ~ 493% and a polymer density of ~ 431 mg/cm3, which is comparable to that of native skeletal tissues. AFM indentation of the surface showed an average Young's modulus of 189 kPa, falling in a range that is conducive to both osteogenesis and vasculogenesis. The osteogenic microgel containing chitosan, gelatin, and hydroxyapatite, mimicking the bone matrix, supported robust attachment, proliferation, and differentiation of MSC. On the other hand, the vasculogenic microgels containing only gelatin, enriched endothelial phenotype and enabled vascular networks formation when embedded in 3D matrices. Combining the two types of microgels created a hybrid construct that sustained the functions of both osteogenic and vasculogenic microgels and enhanced one another. Using a murine model, we also show that the osteogenic microgels regenerate bone in a critical-sized defect with > 95% defect closure by week 12. These multifunctional microgels can be administered minimally invasively and can conformally fill large bone defects. This work lays the foundation to establish principles of designing multiphasic scaffolds with tissue-specific biophysical and biochemical properties for regenerating vascularized and interfacial tissues.
Collapse
Affiliation(s)
- Matthew D Patrick
- Department of Biomedical Engineering, University of Kentucky, 760 Press Avenue, 138 Healthy Kentucky Research Building, Lexington, KY, 40536, USA
| | - Jeremy F Keys
- Department of Biomedical Engineering, University of Kentucky, 760 Press Avenue, 138 Healthy Kentucky Research Building, Lexington, KY, 40536, USA
| | - Harshini Suresh Kumar
- Department of Biomedical Engineering, University of Kentucky, 760 Press Avenue, 138 Healthy Kentucky Research Building, Lexington, KY, 40536, USA
| | - Ramkumar T Annamalai
- Department of Biomedical Engineering, University of Kentucky, 760 Press Avenue, 138 Healthy Kentucky Research Building, Lexington, KY, 40536, USA.
| |
Collapse
|
27
|
Fernandes S, Vyas C, Lim P, Pereira RF, Virós A, Bártolo P. 3D Bioprinting: An Enabling Technology to Understand Melanoma. Cancers (Basel) 2022; 14:cancers14143535. [PMID: 35884596 PMCID: PMC9318274 DOI: 10.3390/cancers14143535] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2022] [Revised: 07/04/2022] [Accepted: 07/12/2022] [Indexed: 02/06/2023] Open
Abstract
Melanoma is a potentially fatal cancer with rising incidence over the last 50 years, associated with enhanced sun exposure and ultraviolet radiation. Its incidence is highest in people of European descent and the ageing population. There are multiple clinical and epidemiological variables affecting melanoma incidence and mortality, such as sex, ethnicity, UV exposure, anatomic site, and age. Although survival has improved in recent years due to advances in targeted and immunotherapies, new understanding of melanoma biology and disease progression is vital to improving clinical outcomes. Efforts to develop three-dimensional human skin equivalent models using biofabrication techniques, such as bioprinting, promise to deliver a better understanding of the complexity of melanoma and associated risk factors. These 3D skin models can be used as a platform for patient specific models and testing therapeutics.
Collapse
Affiliation(s)
- Samantha Fernandes
- Department of Mechanical, Aerospace and Civil Engineering, University of Manchester, Oxford Road, Manchester M13 9PL, UK; (S.F.); (C.V.); (P.L.)
| | - Cian Vyas
- Department of Mechanical, Aerospace and Civil Engineering, University of Manchester, Oxford Road, Manchester M13 9PL, UK; (S.F.); (C.V.); (P.L.)
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore 639798, Singapore
| | - Peggy Lim
- Department of Mechanical, Aerospace and Civil Engineering, University of Manchester, Oxford Road, Manchester M13 9PL, UK; (S.F.); (C.V.); (P.L.)
| | - Rúben F. Pereira
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal;
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal
- INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
| | - Amaya Virós
- Skin Cancer and Ageing Laboratory, Cancer Research UK Manchester Institute, University of Manchester, Oxford Road, Manchester M13 9PL, UK;
| | - Paulo Bártolo
- Department of Mechanical, Aerospace and Civil Engineering, University of Manchester, Oxford Road, Manchester M13 9PL, UK; (S.F.); (C.V.); (P.L.)
- Singapore Centre for 3D Printing, School of Mechanical and Aerospace Engineering, Nanyang Technological University, Singapore 639798, Singapore
- Correspondence: or
| |
Collapse
|
28
|
Filippi M, Buchner T, Yasa O, Weirich S, Katzschmann RK. Microfluidic Tissue Engineering and Bio-Actuation. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2108427. [PMID: 35194852 DOI: 10.1002/adma.202108427] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 02/07/2022] [Indexed: 06/14/2023]
Abstract
Bio-hybrid technologies aim to replicate the unique capabilities of biological systems that could surpass advanced artificial technologies. Soft bio-hybrid robots consist of synthetic and living materials and have the potential to self-assemble, regenerate, work autonomously, and interact safely with other species and the environment. Cells require a sufficient exchange of nutrients and gases, which is guaranteed by convection and diffusive transport through liquid media. The functional development and long-term survival of biological tissues in vitro can be improved by dynamic flow culture, but only microfluidic flow control can develop tissue with fine structuring and regulation at the microscale. Full control of tissue growth at the microscale will eventually lead to functional macroscale constructs, which are needed as the biological component of soft bio-hybrid technologies. This review summarizes recent progress in microfluidic techniques to engineer biological tissues, focusing on the use of muscle cells for robotic bio-actuation. Moreover, the instances in which bio-actuation technologies greatly benefit from fusion with microfluidics are highlighted, which include: the microfabrication of matrices, biomimicry of cell microenvironments, tissue maturation, perfusion, and vascularization.
Collapse
Affiliation(s)
- Miriam Filippi
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Thomas Buchner
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Oncay Yasa
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Stefan Weirich
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Robert K Katzschmann
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| |
Collapse
|
29
|
Ladeira BMF, Gomes MC, Custódio CA, Mano JF. High-Throughput Production of Microsponges from Platelet Lysate for Tissue Engineering Applications. Tissue Eng Part C Methods 2022; 28:325-334. [PMID: 35343236 DOI: 10.1089/ten.tec.2022.0029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Cell-based therapies require a large number of cells, as well as appropriate methods to deliver the cells to damaged tissue. Microcarriers provide an optimal platform for large-scale cell culture while also improving cell retention during cell delivery. However, this technology still presents significant challenges due to low-throughput fabrication methods and an inability of the microcarriers to recreate the properties of human tissue. This work proposes, for the first time, the use of methacryloyl platelet lysates (PLMA), a photocrosslinkable material derived from human platelet lysates, to produce porous microcarriers. Initially, high quantities of PLMA/alginate core-shell microcapsules are produced using coaxial electrospray. Subsequently, the microcapsules are collected, irradiated with ultraviolet light, washed, and freeze dried yielding PLMA microsponges. These microsponges are able to support the adhesion and proliferation of human adipose-derived stem cells, while also displaying potential in the assembly of autologous microtissues. Cell-laden microsponges were shown to self-organize into aggregates, suggesting possible applications in bottom-up tissue engineering applications. Impact Statement Microcarriers have increasingly been used as delivery platforms in cell therapy. Herein, the encapsulation of human-derived proteins in alginate microcapsules is proposed as a method to produce microcarriers from photopolymerizable materials. The capsules function as a template structure, which is then processed into spherical microparticles, which can be used in cell culture, cell delivery, and bottom-up assembly. As a proof of concept, this method was combined with lyophilization to process methacryloyl platelet lysates into injectable microsponges for cell delivery.
Collapse
Affiliation(s)
- Bruno M F Ladeira
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | - Maria C Gomes
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | - Catarina A Custódio
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| | - João F Mano
- Department of Chemistry, CICECO-Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
30
|
Jang W, Kim DY, Mun SJ, Choi JH, Roh YH, Bong KW. Direct functionalization of cell‐adhesion promoters to hydrogel microparticles synthesized by stop‐flow lithography. JOURNAL OF POLYMER SCIENCE 2022. [DOI: 10.1002/pol.20210934] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Affiliation(s)
- Wookyoung Jang
- Department of Chemical and Biological Engineering Korea University Seoul Republic of Korea
| | - Do Yeon Kim
- Department of Chemical and Biological Engineering Korea University Seoul Republic of Korea
| | - Seok Joon Mun
- Department of Chemical and Biological Engineering Korea University Seoul Republic of Korea
| | - Jun Hee Choi
- Department of Chemical and Biological Engineering Korea University Seoul Republic of Korea
| | - Yoon Ho Roh
- Department of Chemical and Biological Engineering Korea University Seoul Republic of Korea
| | - Ki Wan Bong
- Department of Chemical and Biological Engineering Korea University Seoul Republic of Korea
| |
Collapse
|
31
|
Bjørge IM, Correia CR, Mano JF. Hipster microcarriers: exploring geometrical and topographical cues of non-spherical microcarriers in biomedical applications. MATERIALS HORIZONS 2022; 9:908-933. [PMID: 34908074 DOI: 10.1039/d1mh01694f] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Structure and organisation are key aspects of the native tissue environment, which ultimately condition cell fate via a myriad of processes, including the activation of mechanotransduction pathways. By modulating the formation of integrin-mediated adhesions and consequently impacting cell contractility, engineered geometrical and topographical cues may be introduced to activate downstream signalling and ultimately control cell morphology, proliferation, and differentiation. Microcarriers appear as attractive vehicles for cell-based tissue engineering strategies aiming to modulate this 3D environment, but also as vehicles for cell-free applications, given the ease in tuning their chemical and physical properties. In this review, geometry and topography are highlighted as two preponderant features in actively regulating interactions between cells and the extracellular matrix. While most studies focus on the 2D environment, we focus on how the incorporation of these strategies in 3D systems could be beneficial. The techniques applied to design 3D microcarriers with unique geometries and surface topographical cues are covered, as well as specific tissue engineering approaches employing these microcarriers. In fact, successfully achieving a functional histoarchitecture may depend on a combination of fine-tuned geometrically shaped microcarriers presenting intricately tailored topographical cues. Lastly, we pinpoint microcarrier geometry as a key player in cell-free biomaterial-based strategies, and its impact on drug release kinetics, the production of steerable microcarriers to target tumour cells, and as protein or antibody biosensors.
Collapse
Affiliation(s)
- Isabel M Bjørge
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal.
| | - Clara R Correia
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal.
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Aveiro, Portugal.
| |
Collapse
|
32
|
Maji S, Lee H. Engineering Hydrogels for the Development of Three-Dimensional In Vitro Models. Int J Mol Sci 2022; 23:2662. [PMID: 35269803 PMCID: PMC8910155 DOI: 10.3390/ijms23052662] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/25/2022] [Accepted: 02/26/2022] [Indexed: 02/06/2023] Open
Abstract
The superiority of in vitro 3D cultures over conventional 2D cell cultures is well recognized by the scientific community for its relevance in mimicking the native tissue architecture and functionality. The recent paradigm shift in the field of tissue engineering toward the development of 3D in vitro models can be realized with its myriad of applications, including drug screening, developing alternative diagnostics, and regenerative medicine. Hydrogels are considered the most suitable biomaterial for developing an in vitro model owing to their similarity in features to the extracellular microenvironment of native tissue. In this review article, recent progress in the use of hydrogel-based biomaterial for the development of 3D in vitro biomimetic tissue models is highlighted. Discussions of hydrogel sources and the latest hybrid system with different combinations of biopolymers are also presented. The hydrogel crosslinking mechanism and design consideration are summarized, followed by different types of available hydrogel module systems along with recent microfabrication technologies. We also present the latest developments in engineering hydrogel-based 3D in vitro models targeting specific tissues. Finally, we discuss the challenges surrounding current in vitro platforms and 3D models in the light of future perspectives for an improved biomimetic in vitro organ system.
Collapse
Affiliation(s)
- Somnath Maji
- Department of Mechanical and Biomedical Engineering, Kangwon National University (KNU), Chuncheon 24341, Korea;
| | - Hyungseok Lee
- Department of Mechanical and Biomedical Engineering, Kangwon National University (KNU), Chuncheon 24341, Korea;
- Department of Smart Health Science and Technology, Kangwon National University (KNU), Chuncheon 24341, Korea
| |
Collapse
|
33
|
Jabbari E, Sepahvandi A. Decellularized Articular Cartilage Microgels as Microcarriers for Expansion of Mesenchymal Stem Cells. Gels 2022; 8:gels8030148. [PMID: 35323261 PMCID: PMC8949257 DOI: 10.3390/gels8030148] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2022] [Revised: 02/17/2022] [Accepted: 02/23/2022] [Indexed: 11/16/2022] Open
Abstract
Conventional microcarriers used for expansion of human mesenchymal stem cells (hMSCs) require detachment and separation of the cells from the carrier prior to use in clinical applications for regeneration of articular cartilage, and the carrier can cause undesirable phenotypic changes in the expanded cells. This work describes a novel approach to expand hMSCs on biomimetic carriers based on adult or fetal decellularized bovine articular cartilage that supports tissue regeneration without the need to detach the expanded cells from the carrier. In this approach, the fetal or adult bovine articular cartilage was minced, decellularized, freeze-dried, ground, and sieved to produce articular cartilage microgels (CMGs) in a specified size range. Next, the hMSCs were expanded on CMGs in a bioreactor in basal medium to generate hMSC-loaded CMG microgels (CMG-MSCs). Then, the CMG-MSCs were suspended in sodium alginate, injected in a mold, crosslinked with calcium chloride, and incubated in chondrogenic medium as an injectable cellular construct for regeneration of articular cartilage. The expression of chondrogenic markers and compressive moduli of the injectable CMG-MSCs/alginate hydrogels incubated in chondrogenic medium were higher compared to the hMSCs directly encapsulated in alginate hydrogels.
Collapse
|
34
|
Mohajeri M, Eskandari M, Ghazali ZS, Ghazali HS. Cell encapsulation in alginate-based microgels using droplet microfluidics; a review on gelation methods and applications. Biomed Phys Eng Express 2022; 8. [PMID: 35073537 DOI: 10.1088/2057-1976/ac4e2d] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 01/24/2022] [Indexed: 11/12/2022]
Abstract
Cell encapsulation within the microspheres using a semi-permeable polymer allows the two-way transfer of molecules such as oxygen, nutrients, and growth factors. The main advantages of cell encapsulation technology include controlling the problems involved in transplanting rejection in tissue engineering applications and reducing the long-term need for immunosuppressive drugs following organ transplantation to eliminate the side effects. Cell-laden microgels can also be used in 3D cell cultures, wound healing, and cancerous clusters for drug testing. Since cell encapsulation is used for different purposes, several techniques have been developed to encapsulate cells. Droplet-based microfluidics is one of the most valuable techniques in cell encapsulating. This study aimed to review the geometries and the mechanisms proposed in microfluidic systems to precisely control cell-laden microgels production with different biopolymers. We also focused on alginate gelation techniques due to their essential role in cell encapsulation applications. Finally, some applications of these microgels and researches will be explored.
Collapse
Affiliation(s)
- Mohammad Mohajeri
- Biomedical Engineering Department, Amirkabir University of Technology, Department of Biomedical Engineering No. 350, Hafez Ave, Valiasr Square, Tehran, Iran, Tehran, 159163-4311, Iran (the Islamic Republic of)
| | - Mahnaz Eskandari
- Biomedical Engineering Department, Amirkabir University of Technology, Department of Biomedical Engineering No. 350, Hafez Ave, Valiasr Square, Tehran, Iran, Tehran, 159163-4311, Iran (the Islamic Republic of)
| | - Zahra Sadat Ghazali
- Biomedical Engineering Department, Amirkabir University of Technology, No. 350, Hafez Ave, Valiasr Square, Tehran, Iran, Tehran, 159163-4311, Iran (the Islamic Republic of)
| | - Hanieh Sadat Ghazali
- Department of Nanobiotechnology, Tarbiat Modares University, Jalal Aleahmad-Tehran-Iran, Tehran, 14115-111, Iran (the Islamic Republic of)
| |
Collapse
|
35
|
Wei L, Tan J, Li L, Wang H, Liu S, Chen J, Weng Y, Liu T. Chitosan/Alginate Hydrogel Dressing Loaded FGF/VE-Cadherin to Accelerate Full-Thickness Skin Regeneration and More Normal Skin Repairs. Int J Mol Sci 2022; 23:ijms23031249. [PMID: 35163172 PMCID: PMC8835731 DOI: 10.3390/ijms23031249] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2021] [Revised: 01/21/2022] [Accepted: 01/21/2022] [Indexed: 02/04/2023] Open
Abstract
The process of full-thickness skin regeneration is complex and has many parameters involved, which makes it difficult to use a single dressing to meet the various requirements of the complete regeneration at the same time. Therefore, developing hydrogel dressings with multifunction, including tunable rheological properties and aperture, hemostatic, antibacterial and super cytocompatibility, is a desirable candidate in wound healing. In this study, a series of complex hydrogels were developed via the hydrogen bond and covalent bond between chitosan (CS) and alginate (SA). These hydrogels exhibited suitable pore size and tunable rheological properties for cell adhesion. Chitosan endowed hemostatic, antibacterial properties and great cytocompatibility and thus solved two primary problems in the early stage of the wound healing process. Moreover, the sustained cytocompatibility of the hydrogels was further investigated after adding FGF and VE-cadherin via the co-culture of L929 and EC for 12 days. The confocal 3D fluorescent images showed that the cells were spherical and tended to form multicellular spheroids, which distributed in about 40-60 μm thick hydrogels. Furthermore, the hydrogel dressings significantly accelerate defected skin turn to normal skin with proper epithelial thickness and new blood vessels and hair follicles through the histological analysis of in vivo wound healing. The findings mentioned above demonstrated that the CS/SA hydrogels with growth factors have great potential as multifunctional hydrogel dressings for full-thickness skin regeneration incorporated with hemostatic, antibacterial, sustained cytocompatibility for 3D cell culture and normal skin repairing.
Collapse
Affiliation(s)
| | | | | | | | | | - Junying Chen
- Correspondence: ; Tel.: +86-028-87634148; Fax: +86-028-87600625
| | | | | |
Collapse
|
36
|
Yap JX, Leo CP, Mohd Yasin NH, Show PL, Chu DT, Singh V, Derek CJC. Recent advances of natural biopolymeric culture scaffold: synthesis and modification. Bioengineered 2022; 13:2226-2247. [PMID: 35030968 PMCID: PMC8974151 DOI: 10.1080/21655979.2021.2024322] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Traditionally existing 2D culture scaffold has been inappropriately validated due to the failure in generating the precise therapeutic response. Therefore, this leads to the fabrication of 3D culture scaffold resolving the limitations in the in vivo environment. In recent years, tissue engineering played an important role in the field of bio-medical engineering. Biopolymer material, a novel natural material with excellent properties of nontoxic and biodegradable merits can be served as culture scaffold. This review summarizes the modifications of natural biopolymeric culture scaffold with different crosslinkers and their application. In addition, this review provides the recent progress of natural biopolymeric culture scaffold mainly focusing on their properties, synthesizing and modification and application.
Collapse
Affiliation(s)
- Jia Xin Yap
- School of Chemical Engineering, Engineering Campus, Universiti Sains Malaysia, Nibong Tebal, Malaysia
| | - C P Leo
- School of Chemical Engineering, Engineering Campus, Universiti Sains Malaysia, Nibong Tebal, Malaysia
| | - Nazlina Haiza Mohd Yasin
- Department of Biological Sciences and Biotechnology, Faculty of Science and Technology, Universiti Kebangsaan Malaysia, Bangi, Malaysia
| | - Pau Loke Show
- Department of Chemical and Environmental Engineering, Faculty of Science and Engineering, University of Nottingham Malaysia, Semenyih, Malaysia
| | - Dinh-Toi Chu
- Center for Biomedicine and Community Health, International School, Vietnam National University, Hanoi, Vietnam
| | - Vijai Singh
- Department of Biosciences, School of Science, Indrashil University, Rajpur, Mehsana, India
| | - C J C Derek
- School of Chemical Engineering, Engineering Campus, Universiti Sains Malaysia, Nibong Tebal, Malaysia
| |
Collapse
|
37
|
Engineering Biological Tissues from the Bottom-Up: Recent Advances and Future Prospects. MICROMACHINES 2021; 13:mi13010075. [PMID: 35056239 PMCID: PMC8780533 DOI: 10.3390/mi13010075] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/13/2021] [Revised: 12/24/2021] [Accepted: 12/28/2021] [Indexed: 02/06/2023]
Abstract
Tissue engineering provides a powerful solution for current organ shortages, and researchers have cultured blood vessels, heart tissues, and bone tissues in vitro. However, traditional top-down tissue engineering has suffered two challenges: vascularization and reconfigurability of functional units. With the continuous development of micro-nano technology and biomaterial technology, bottom-up tissue engineering as a promising approach for organ and tissue modular reconstruction has gradually developed. In this article, relevant advances in living blocks fabrication and assembly techniques for creation of higher-order bioarchitectures are described. After a critical overview of this technology, a discussion of practical challenges is provided, and future development prospects are proposed.
Collapse
|
38
|
Wang C, Wang Q, Shang L. Microfluidic single-cell coating with defined chemomechanical cues for cell therapy. Sci Bull (Beijing) 2021; 66:2434-2436. [PMID: 36654197 DOI: 10.1016/j.scib.2021.08.015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Affiliation(s)
- Chong Wang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Qiao Wang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China
| | - Luoran Shang
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, the Shanghai Key Laboratory of Medical Epigenetics, International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences, Fudan University, Shanghai 200032, China.
| |
Collapse
|
39
|
Poongodi R, Chen YL, Yang TH, Huang YH, Yang KD, Lin HC, Cheng JK. Bio-Scaffolds as Cell or Exosome Carriers for Nerve Injury Repair. Int J Mol Sci 2021; 22:13347. [PMID: 34948144 PMCID: PMC8707664 DOI: 10.3390/ijms222413347] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 12/08/2021] [Accepted: 12/10/2021] [Indexed: 12/12/2022] Open
Abstract
Central and peripheral nerve injuries can lead to permanent paralysis and organ dysfunction. In recent years, many cell and exosome implantation techniques have been developed in an attempt to restore function after nerve injury with promising but generally unsatisfactory clinical results. Clinical outcome may be enhanced by bio-scaffolds specifically fabricated to provide the appropriate three-dimensional (3D) conduit, growth-permissive substrate, and trophic factor support required for cell survival and regeneration. In rodents, these scaffolds have been shown to promote axonal regrowth and restore limb motor function following experimental spinal cord or sciatic nerve injury. Combining the appropriate cell/exosome and scaffold type may thus achieve tissue repair and regeneration with safety and efficacy sufficient for routine clinical application. In this review, we describe the efficacies of bio-scaffolds composed of various natural polysaccharides (alginate, chitin, chitosan, and hyaluronic acid), protein polymers (gelatin, collagen, silk fibroin, fibrin, and keratin), and self-assembling peptides for repair of nerve injury. In addition, we review the capacities of these constructs for supporting in vitro cell-adhesion, mechano-transduction, proliferation, and differentiation as well as the in vivo properties critical for a successful clinical outcome, including controlled degradation and re-absorption. Finally, we describe recent advances in 3D bio-printing for nerve regeneration.
Collapse
Affiliation(s)
- Raju Poongodi
- Department of Medical Research, Mackay Memorial Hospital, Taipei 10449, Taiwan; (R.P.); (T.-H.Y.)
| | - Ying-Lun Chen
- Department of Anesthesiology, Mackay Memorial Hospital, Taipei 10449, Taiwan; (Y.-L.C.); (Y.-H.H.)
- Department of Medicine, Mackay Medical College, New Taipei City 25245, Taiwan
| | - Tao-Hsiang Yang
- Department of Medical Research, Mackay Memorial Hospital, Taipei 10449, Taiwan; (R.P.); (T.-H.Y.)
| | - Ya-Hsien Huang
- Department of Anesthesiology, Mackay Memorial Hospital, Taipei 10449, Taiwan; (Y.-L.C.); (Y.-H.H.)
- Department of Medicine, Mackay Medical College, New Taipei City 25245, Taiwan
| | - Kuender D. Yang
- Institute of Biomedical Science, Mackay Medical College, New Taipei City 25245, Taiwan;
- Department of Pediatrics, Mackay Memorial Hospital, Taipei 10449, Taiwan
- Institute of Clinical Medicine, National Yang Ming Chiao Tung University, Taipei 11221, Taiwan
| | - Hsin-Chieh Lin
- Department of Materials Science and Engineering, National Yang Ming Chiao Tung University, Hsinchu 30010, Taiwan;
| | - Jen-Kun Cheng
- Department of Medical Research, Mackay Memorial Hospital, Taipei 10449, Taiwan; (R.P.); (T.-H.Y.)
- Department of Anesthesiology, Mackay Memorial Hospital, Taipei 10449, Taiwan; (Y.-L.C.); (Y.-H.H.)
- Department of Medicine, Mackay Medical College, New Taipei City 25245, Taiwan
| |
Collapse
|
40
|
Rosen O, Jayson A, Natan N, Epstein E. A Simple Method for in situ Quantification of Cells on Carriers. Bio Protoc 2021; 11:e4254. [PMID: 35005097 DOI: 10.21769/bioprotoc.4254] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/23/2021] [Accepted: 09/29/2021] [Indexed: 11/02/2022] Open
Abstract
The technology of cell carriers was developed as a response to the need for high cell density to enable higher production levels in cell-based production processes. To follow the production process, quantifying the number of cells on these carriers is required, as well as tracking their viability and proliferation. However, owing to various carriers' unique structures, tracking the cells is challenging using current traditional assays that were originally developed for monolayers of adherent cells. The current "gold standard" method is counting cell nuclei, which is tedious and counts both live and dead cells. A few other techniques have been developed, but they are all specific to a carrier type and involve specialized equipment. Here, we describe a broad ranging method for counting cells on carriers. The method is based on the Alamar blue dye, a well-known, common marker for cell activity. No separation of the cells from the carriers is needed, nor is any specialized equipment. The method is simple and rapid, and provides comprehensive details necessary for control of production processes in cells. This method can be easily implemented in any cell-based process and other unique platforms for measuring growth of cells. Graphic abstract: Schematic of the in situ quantification method.
Collapse
Affiliation(s)
- Osnat Rosen
- Department of Biotechnology, Israel Institute for Biological Research, Ness Ziona 7410001, Israel
| | - Avital Jayson
- Department of Biotechnology, Israel Institute for Biological Research, Ness Ziona 7410001, Israel
| | - Niva Natan
- Department of Biotechnology, Israel Institute for Biological Research, Ness Ziona 7410001, Israel
| | - Eyal Epstein
- Department of Biotechnology, Israel Institute for Biological Research, Ness Ziona 7410001, Israel
| |
Collapse
|
41
|
Chizhov AO. Complex Carbohydrates and Glycoconjugates: Structure, Functions and Applications. Int J Mol Sci 2021; 22:ijms222212219. [PMID: 34830101 PMCID: PMC8618160 DOI: 10.3390/ijms222212219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Accepted: 10/21/2021] [Indexed: 11/29/2022] Open
Affiliation(s)
- Alexander O Chizhov
- N.D. Zelinsky Institute of Organic Chemistry, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
42
|
Adrian E, Treľová D, Filová E, Kumorek M, Lobaz V, Poreba R, Janoušková O, Pop-Georgievski O, Lacík I, Kubies D. Complexation of CXCL12, FGF-2 and VEGF with Heparin Modulates the Protein Release from Alginate Microbeads. Int J Mol Sci 2021; 22:11666. [PMID: 34769095 PMCID: PMC8583835 DOI: 10.3390/ijms222111666] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2021] [Revised: 10/20/2021] [Accepted: 10/25/2021] [Indexed: 12/19/2022] Open
Abstract
Long-term delivery of growth factors and immunomodulatory agents is highly required to support the integrity of tissue in engineering constructs, e.g., formation of vasculature, and to minimize immune response in a recipient. However, for proteins with a net positive charge at the physiological pH, controlled delivery from negatively charged alginate (Alg) platforms is challenging due to electrostatic interactions that can hamper the protein release. In order to regulate such interactions between proteins and the Alg matrix, we propose to complex proteins of interest in this study - CXCL12, FGF-2, VEGF - with polyanionic heparin prior to their encapsulation into Alg microbeads of high content of α-L-guluronic acid units (high-G). This strategy effectively reduced protein interactions with Alg (as shown by model ITC and SPR experiments) and, depending on the protein type, afforded control over the protein release for at least one month. The released proteins retained their in vitro bioactivity: CXCL12 stimulated the migration of Jurkat cells, and FGF-2 and VEGF induced proliferation and maturation of HUVECs. The presence of heparin also intensified protein biological efficiency. The proposed approach for encapsulation of proteins with a positive net charge into high-G Alg hydrogels is promising for controlled long-term protein delivery under in vivo conditions.
Collapse
Affiliation(s)
- Edyta Adrian
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovsky sq.2, 162 06 Prague, Czech Republic; (E.A.); (M.K.); (V.L.); (R.P.); (O.J.); (O.P.-G.)
- Department of Chemical Engineering, University of Chemistry and Technology, Technicka 5, 166 28 Prague, Czech Republic
| | - Dušana Treľová
- Polymer Institute of the Slovak Academy of Sciences, Dubravska cesta 9, 845 41 Bratislava, Slovakia; (D.T.); (I.L.)
| | - Elena Filová
- Department of Biomaterials and Tissue Engineering, Institute of Physiology of the Czech Academy of Sciences, Videnska 1083, 142 20 Prague, Czech Republic;
| | - Marta Kumorek
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovsky sq.2, 162 06 Prague, Czech Republic; (E.A.); (M.K.); (V.L.); (R.P.); (O.J.); (O.P.-G.)
| | - Volodymyr Lobaz
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovsky sq.2, 162 06 Prague, Czech Republic; (E.A.); (M.K.); (V.L.); (R.P.); (O.J.); (O.P.-G.)
| | - Rafal Poreba
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovsky sq.2, 162 06 Prague, Czech Republic; (E.A.); (M.K.); (V.L.); (R.P.); (O.J.); (O.P.-G.)
| | - Olga Janoušková
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovsky sq.2, 162 06 Prague, Czech Republic; (E.A.); (M.K.); (V.L.); (R.P.); (O.J.); (O.P.-G.)
| | - Ognen Pop-Georgievski
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovsky sq.2, 162 06 Prague, Czech Republic; (E.A.); (M.K.); (V.L.); (R.P.); (O.J.); (O.P.-G.)
| | - Igor Lacík
- Polymer Institute of the Slovak Academy of Sciences, Dubravska cesta 9, 845 41 Bratislava, Slovakia; (D.T.); (I.L.)
- Centre for Advanced Materials Application of the Slovak Academy of Sciences, Dubravska cesta 9, 845 11 Bratislava, Slovakia
| | - Dana Kubies
- Institute of Macromolecular Chemistry, Czech Academy of Sciences, Heyrovsky sq.2, 162 06 Prague, Czech Republic; (E.A.); (M.K.); (V.L.); (R.P.); (O.J.); (O.P.-G.)
| |
Collapse
|
43
|
Natural Polymers for the Maintenance of Oral Health: Review of Recent Advances and Perspectives. Int J Mol Sci 2021; 22:ijms221910337. [PMID: 34638678 PMCID: PMC8508910 DOI: 10.3390/ijms221910337] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2021] [Revised: 09/18/2021] [Accepted: 09/21/2021] [Indexed: 12/14/2022] Open
Abstract
The success of modern dental treatment is strongly dependent on the materials used both temporarily and permanently. Among all dental materials, polymers are a very important class with a wide spectrum of applications. This review aims to provide a state-of-the-art overview of the recent advances in the field of natural polymers used to maintain or restore oral health. It focuses on the properties of the most common proteins and polysaccharides of natural origin in terms of meeting the specific biological requirements in the increasingly demanding field of modern dentistry. The use of naturally derived polymers in different dental specialties for preventive and therapeutic purposes has been discussed. The major fields of application cover caries and the management of periodontal diseases, the fabrication of membranes and scaffolds for the regeneration of dental structures, the manufacturing of oral appliances and dentures as well as providing systems for oral drug delivery. This paper also includes a comparative characteristic of natural and synthetic dental polymers. Finally, the current review highlights new perspectives, possible future advancements, as well as challenges that may be encountered by researchers in the field of dental applications of polymers of natural origin.
Collapse
|
44
|
Argentiere S, Siciliano PA, Blasi L. How Microgels Can Improve the Impact of Organ-on-Chip and Microfluidic Devices for 3D Culture: Compartmentalization, Single Cell Encapsulation and Control on Cell Fate. Polymers (Basel) 2021; 13:3216. [PMID: 34641032 PMCID: PMC8512905 DOI: 10.3390/polym13193216] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 08/27/2021] [Accepted: 08/30/2021] [Indexed: 12/13/2022] Open
Abstract
The Organ-on-chip (OOC) devices represent the new frontier in biomedical research to produce micro-organoids and tissues for drug testing and regenerative medicine. The development of such miniaturized models requires the 3D culture of multiple cell types in a highly controlled microenvironment, opening new challenges in reproducing the extracellular matrix (ECM) experienced by cells in vivo. In this regard, cell-laden microgels (CLMs) represent a promising tool for 3D cell culturing and on-chip generation of micro-organs. The engineering of hydrogel matrix with properly balanced biochemical and biophysical cues enables the formation of tunable 3D cellular microenvironments and long-term in vitro cultures. This focused review provides an overview of the most recent applications of CLMs in microfluidic devices for organoids formation, highlighting microgels' roles in OOC development as well as insights into future research.
Collapse
Affiliation(s)
| | | | - Laura Blasi
- Institute for Microelectronics and Microsystems IMM-CNR, Via Monteroni, University Campus, 73100 Lecce, Italy; (S.A.); (P.A.S.)
| |
Collapse
|
45
|
Ebhodaghe SO. Natural Polymeric Scaffolds for Tissue Engineering Applications. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2021; 32:2144-2194. [PMID: 34328068 DOI: 10.1080/09205063.2021.1958185] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
Natural polymeric scaffolds can be used for tissue engineering applications such as cell delivery and cell-free supporting of native tissues. This is because of their desirable properties such as; high biocompatibility, tunable mechanical strength and conductivity, large surface area, porous- and extracellular matrix (ECM)-mimicked structures. Specifically, their less toxicity and biocompatibility makes them suitable for several tissue engineering applications. For these reasons, several biopolymeric scaffolds are currently being explored for numerous tissue engineering applications. To date, research on the nature, chemistry, and properties of nanocomposite biopolymers are been reported, while the need for a comprehensive research note on more tissue engineering application of these biopolymers remains. As a result, this present study comprehensively reviews the development of common natural biopolymers as scaffolds for tissue engineering applications such as cartilage tissue engineering, cornea repairs, osteochondral defect repairs, and nerve regeneration. More so, the implications of research findings for further studies are presented, while the impact of research advances on future research and other specific recommendations are added as well.
Collapse
|
46
|
Darge HF, Chuang SH, Lai JY, Lin SY, Tsai HC. Preparation of thermosensitive PNIPAm-based copolymer coated cytodex 3 microcarriers for efficient nonenzymatic cell harvesting during 3D culturing. Biotechnol Bioeng 2021; 118:4076-4091. [PMID: 34251680 DOI: 10.1002/bit.27885] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Revised: 06/09/2021] [Accepted: 07/09/2021] [Indexed: 12/28/2022]
Abstract
Enzymatic detachment of cells might damage important features and functions of cells and could affect subsequent cell-based applications. Therefore, nonenzymatic cell detachment using thermosensitive polymer matrix is necessary for maintaining cell quality after harvesting. In this study, we prepared thermosensitive PNIPAm-co-AAc-b-PS and PNIPAm-co-AAm-b-PS copolymers and low critical solution temperature (LCST) was tuned near to body temperature. Then, spin coated polymer films were prepared for cell adhesion and thermal-induced cell detachment. The alpha-step analysis and scanning electron microscope image of the films suggested that the thickness of the films depends on the molecular weight and concentration which ranged from 206 to 1330 nm for PNIPAm-co-AAc-b-PS and 97.5-497 nm for PNIPAm-co-AAm-b-PS. The contact angles of the films verified that the polymer surface was moderately hydrophilic at 37°C. Importantly, RAW264.7 cells were convincingly proliferated on the films to a confluent of >80% within 48 h and abled to detach by reducing the temperature. However, relatively more cells were grown on PNIPAm-co-AAm-b-PS (5%w/v) films and thermal-induced cell detachment was more abundant in this formulation. As a result, PNIPAm-co-AAm-b-PS (5%w/v) was further used to coat commercial cytodex 3 microcarriers for 3D cell culturing and interestingly enhanced cell detachment with preserved potential of recovery was observed at a temperature of below LCST. Thus, surface modification of microcarriers with thermosensitive PNIPAm-co-AAm-b-PS could be vital strategy for nonenzymatic cell detachment and to achieve adequate number of cells with maximum cell viability and functionality.
Collapse
Affiliation(s)
- Haile F Darge
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei, Taiwan.,Advanced Membrane Materials Center, National Taiwan University of Science and Technology, Taipei, Taiwan.,College of Medicine and Health Science, Bahir Dar University, Bahir Dar, Ethiopia
| | - Shun-Hao Chuang
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei, Taiwan
| | - Juin-Yih Lai
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei, Taiwan.,Advanced Membrane Materials Center, National Taiwan University of Science and Technology, Taipei, Taiwan.,R&D Center for Membrane Technology, Chung Yuan Christian University, Chungli, Taiwan
| | - Shuian-Yin Lin
- Biomedical Technology and Device Research Center, Industrial Technology Research Institute, Hsinchu, Taiwan
| | - Hsieh-Chih Tsai
- Graduate Institute of Applied Science and Technology, National Taiwan University of Science and Technology, Taipei, Taiwan.,Advanced Membrane Materials Center, National Taiwan University of Science and Technology, Taipei, Taiwan.,R&D Center for Membrane Technology, Chung Yuan Christian University, Chungli, Taiwan
| |
Collapse
|
47
|
Rosendahl J, Svanström A, Berglin M, Petronis S, Bogestål Y, Stenlund P, Standoft S, Ståhlberg A, Landberg G, Chinga-Carrasco G, Håkansson J. 3D Printed Nanocellulose Scaffolds as a Cancer Cell Culture Model System. Bioengineering (Basel) 2021; 8:97. [PMID: 34356204 PMCID: PMC8301137 DOI: 10.3390/bioengineering8070097] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 06/28/2021] [Accepted: 06/28/2021] [Indexed: 12/12/2022] Open
Abstract
Current conventional cancer drug screening models based on two-dimensional (2D) cell culture have several flaws and there is a large need of more in vivo mimicking preclinical drug screening platforms. The microenvironment is crucial for the cells to adapt relevant in vivo characteristics and here we introduce a new cell culture system based on three-dimensional (3D) printed scaffolds using cellulose nanofibrils (CNF) pre-treated with 2,2,6,6-tetramethylpyperidine-1-oxyl (TEMPO) as the structural material component. Breast cancer cell lines, MCF7 and MDA-MB-231, were cultured in 3D TEMPO-CNF scaffolds and were shown by scanning electron microscopy (SEM) and histochemistry to grow in multiple layers as a heterogenous cell population with different morphologies, contrasting 2D cultured mono-layered cells with a morphologically homogenous cell population. Gene expression analysis demonstrated that 3D TEMPO-CNF scaffolds induced elevation of the stemness marker CD44 and the migration markers VIM and SNAI1 in MCF7 cells relative to 2D control. T47D cells confirmed the increased level of the stemness marker CD44 and migration marker VIM which was further supported by increased capacity of holoclone formation for 3D cultured cells. Therefore, TEMPO-CNF was shown to represent a promising material for 3D cell culture model systems for cancer cell applications such as drug screening.
Collapse
Affiliation(s)
- Jennifer Rosendahl
- Unit of Biological Function, Division Materials and Production, RISE Research Institutes of Sweden, Box 857, SE-50115 Borås, Sweden; (J.R.); (M.B.); (S.P.); (Y.B.); (P.S.); (S.S.)
| | - Andreas Svanström
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 425, Medicinaregatan 1G, SE-41390 Gothenburg, Sweden; (A.S.); (A.S.); (G.L.)
| | - Mattias Berglin
- Unit of Biological Function, Division Materials and Production, RISE Research Institutes of Sweden, Box 857, SE-50115 Borås, Sweden; (J.R.); (M.B.); (S.P.); (Y.B.); (P.S.); (S.S.)
| | - Sarunas Petronis
- Unit of Biological Function, Division Materials and Production, RISE Research Institutes of Sweden, Box 857, SE-50115 Borås, Sweden; (J.R.); (M.B.); (S.P.); (Y.B.); (P.S.); (S.S.)
| | - Yalda Bogestål
- Unit of Biological Function, Division Materials and Production, RISE Research Institutes of Sweden, Box 857, SE-50115 Borås, Sweden; (J.R.); (M.B.); (S.P.); (Y.B.); (P.S.); (S.S.)
| | - Patrik Stenlund
- Unit of Biological Function, Division Materials and Production, RISE Research Institutes of Sweden, Box 857, SE-50115 Borås, Sweden; (J.R.); (M.B.); (S.P.); (Y.B.); (P.S.); (S.S.)
| | - Simon Standoft
- Unit of Biological Function, Division Materials and Production, RISE Research Institutes of Sweden, Box 857, SE-50115 Borås, Sweden; (J.R.); (M.B.); (S.P.); (Y.B.); (P.S.); (S.S.)
| | - Anders Ståhlberg
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 425, Medicinaregatan 1G, SE-41390 Gothenburg, Sweden; (A.S.); (A.S.); (G.L.)
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, SE-40530 Gothenburg, Sweden
- Department of Clinical Genetics and Genomics, Region Västra Götaland, Sahlgrenska University Hospital, SE-40530 Gothenburg, Sweden
| | - Göran Landberg
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Box 425, Medicinaregatan 1G, SE-41390 Gothenburg, Sweden; (A.S.); (A.S.); (G.L.)
- Department of Clinical Pathology, Sahlgrenska University Hospital, SE-41345 Gothenburg, Sweden
| | | | - Joakim Håkansson
- Unit of Biological Function, Division Materials and Production, RISE Research Institutes of Sweden, Box 857, SE-50115 Borås, Sweden; (J.R.); (M.B.); (S.P.); (Y.B.); (P.S.); (S.S.)
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, P.O. Box 440, SE-40530 Gothenburg, Sweden
| |
Collapse
|
48
|
Decante G, Costa JB, Silva-Correia J, Collins MN, Reis RL, Oliveira JM. Engineering bioinks for 3D bioprinting. Biofabrication 2021; 13. [PMID: 33662949 DOI: 10.1088/1758-5090/abec2c] [Citation(s) in RCA: 105] [Impact Index Per Article: 26.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Accepted: 03/04/2021] [Indexed: 02/07/2023]
Abstract
In recent years, three-dimensional (3D) bioprinting has attracted wide research interest in biomedical engineering and clinical applications. This technology allows for unparalleled architecture control, adaptability and repeatability that can overcome the limits of conventional biofabrication techniques. Along with the emergence of a variety of 3D bioprinting methods, bioinks have also come a long way. From their first developments to support bioprinting requirements, they are now engineered to specific injury sites requirements to mimic native tissue characteristics and to support biofunctionality. Current strategies involve the use of bioinks loaded with cells and biomolecules of interest, without altering their functions, to deliverin situthe elements required to enhance healing/regeneration. The current research and trends in bioink development for 3D bioprinting purposes is overviewed herein.
Collapse
Affiliation(s)
- Guy Decante
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - João B Costa
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Joana Silva-Correia
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Maurice N Collins
- Bernal Institute, School of Engineering, University of Limerick, Limerick, Ireland.,Health Research Institute, University of Limerick, Limerick, Ireland
| | - Rui L Reis
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - J Miguel Oliveira
- 3B's Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, 4805-017 Barco GMR, Portugal.,ICVS/3B's-PT Government Associate Laboratory, Braga/Guimarães, Portugal
| |
Collapse
|
49
|
Design of alginate based micro‐gels via electro fluid dynamics to construct microphysiological cell culture systems. POLYM ADVAN TECHNOL 2021. [DOI: 10.1002/pat.5310] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
50
|
The Marine Polysaccharide Ulvan Confers Potent Osteoinductive Capacity to PCL-Based Scaffolds for Bone Tissue Engineering Applications. Int J Mol Sci 2021; 22:ijms22063086. [PMID: 33802984 PMCID: PMC8002638 DOI: 10.3390/ijms22063086] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2021] [Revised: 03/08/2021] [Accepted: 03/15/2021] [Indexed: 12/11/2022] Open
Abstract
Hybrid composites of synthetic and natural polymers represent materials of choice for bone tissue engineering. Ulvan, a biologically active marine sulfated polysaccharide, is attracting great interest in the development of novel biomedical scaffolds due to recent reports on its osteoinductive properties. Herein, a series of hybrid polycaprolactone scaffolds containing ulvan either alone or in blends with κ-carrageenan and chondroitin sulfate was prepared and characterized. The impact of the preparation methodology and the polysaccharide composition on their morphology, as well as on their mechanical, thermal, water uptake and porosity properties was determined, while their osteoinductive potential was investigated through the evaluation of cell adhesion, viability, and osteogenic differentiation of seeded human adipose-derived mesenchymal stem cells. The results verified the osteoinductive ability of ulvan, showing that its incorporation into the polycaprolactone matrix efficiently promoted cell attachment and viability, thus confirming its potential in the development of biomedical scaffolds for bone tissue regeneration applications.
Collapse
|