1
|
Kao YC, Yang PC, Lin YP, Chen GH, Liu SW, Ho CH, Huang SC, Lee PY, Chen L, Huang CC. Tailoring the therapeutic potential of stem cell spheroid-derived decellularized ECM through post-decellularization BDNF incorporation to enhance brain repair. Biomaterials 2025; 321:123332. [PMID: 40220567 DOI: 10.1016/j.biomaterials.2025.123332] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 04/06/2025] [Accepted: 04/07/2025] [Indexed: 04/14/2025]
Abstract
Decellularized extracellular matrix (dECM) from tissues has significant therapeutic potential but is limited by its rigid molecular composition and reliance on post-decellularization modifications to tailor its functionality. Harsh decellularization processes often result in substantial glycosaminoglycan (GAG) loss, impairing natural growth factor incorporation and necessitating chemical modifications that complicate processing and limit clinical translation. To address these challenges, we developed mesenchymal stem cell (MSC) spheroid-derived three-dimensional (3D) dECM using gentle decellularization techniques. This study demonstrated a crucial advancement-the retention of endogenous GAGs-enabling direct growth factor incorporation without chemical agents. As a proof-of-concept, brain-derived neurotrophic factor (BDNF) was incorporated into the 3D dECM to enhance its therapeutic potential for brain repair. In vitro, BDNF-loaded 3D dECM enabled sustained growth factor release, significantly enhancing the proneuritogenic, neuroprotective, and proangiogenic effects. In a mouse model of traumatic brain injury, the implantation of BDNF-loaded 3D dECM significantly enhanced motor function and facilitated brain repair. These findings highlight the adaptability of MSC spheroid-derived 3D dECM for tissue-specific customization through straightforward and translatable growth factor incorporation, demonstrating its potential as a pro-regenerative biomaterial for advancing regenerative medicine applications.
Collapse
Affiliation(s)
- Ying-Chi Kao
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Pei-Ching Yang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Yu-Ping Lin
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Grace H Chen
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Shao-Wen Liu
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chia-Hsin Ho
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Shih-Chen Huang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Peng-Ying Lee
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Linyi Chen
- Institute of Molecular Medicine, National Tsing Hua University, Hsinchu, 30013, Taiwan; Department of Medical Science, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Chieh-Cheng Huang
- Institute of Biomedical Engineering, National Tsing Hua University, Hsinchu, 30013, Taiwan.
| |
Collapse
|
2
|
Redi G, Del Piano F, Cappellini S, Paladino M, den Breejen A, Fens MHAM, Caiazzo M. Delivery Systems in Neuronal Direct Cell Reprogramming. Cell Reprogram 2025. [PMID: 40372965 DOI: 10.1089/cell.2025.0008] [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: 05/17/2025] Open
Abstract
Neuronal direct cell reprogramming approach allows direct conversion of somatic cells into neurons via forced expression of neuronal cell-lineage transcription factors (TFs). These so-called induced neuronal cells have significant potential as research tools and for therapeutic applications, such as in cell replacement therapy. However, the optimization of TF delivery strategies is crucial to reach clinical practice. In this review, we outlined the currently explored delivery technologies in neuronal direct cell reprogramming and their limitations and advantages. The first employed delivery strategies were mainly integrating viral systems, such as lentiviruses that exert consistently high transgene expression in most cell types. On the other hand, viral systems cause major safety concerns, including the risk for insertional mutagenesis and inflammation. More recently, several safer nonviral delivery systems have been investigated as well; however, these systems generally exert inferior reprogramming efficiency compared with viral systems. Emerging delivery technologies could provide new opportunities in the achievement of safe and effective delivery for neuronal direct cell reprogramming.
Collapse
Affiliation(s)
- Giulia Redi
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II," Naples, Italy
| | - Filomena Del Piano
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II," Naples, Italy
| | - Sara Cappellini
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Martina Paladino
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II," Naples, Italy
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Anne den Breejen
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Marcel H A M Fens
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | - Massimiliano Caiazzo
- Department of Molecular Medicine and Medical Biotechnology, University of Naples "Federico II," Naples, Italy
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| |
Collapse
|
3
|
Zhao W, Liu Z, Li Z, Qian H, Hu J. Protocol for extraction and characterization of mouse brain-derived extracellular matrix for neuronal cell culture. STAR Protoc 2025; 6:103764. [PMID: 40220305 PMCID: PMC12018537 DOI: 10.1016/j.xpro.2025.103764] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2024] [Revised: 02/26/2025] [Accepted: 03/20/2025] [Indexed: 04/14/2025] Open
Abstract
Neuronal cell cultures are highly sensitive to their microenvironment, particularly the choice of coating substrate. Here, we present a protocol to isolate extracellular matrix (ECM) from mouse brain tissue, providing a native coating material that closely mimics in vivo conditions. We detail decellularization steps, guidelines for measuring ECM quality, and instructions for co-culturing neuronal cells on the resulting substrate. By promoting improved neuronal survival, growth, and differentiation, this protocol has broad implications for in vitro neurobiological research and downstream applications.
Collapse
Affiliation(s)
- Weihao Zhao
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, The Department of Medical Genetics, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610054, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China.
| | - Ziqian Liu
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, The Department of Medical Genetics, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610054, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Zhenghao Li
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, The Department of Medical Genetics, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610054, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China
| | - Hao Qian
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, The Department of Medical Genetics, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610054, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China.
| | - Jing Hu
- The Sichuan Provincial Key Laboratory for Human Disease Gene Study, The Department of Medical Genetics, Sichuan Academy of Medical Sciences & Sichuan Provincial People's Hospital, University of Electronic Science and Technology of China, Chengdu 610054, China; School of Medicine, University of Electronic Science and Technology of China, Chengdu 610054, China.
| |
Collapse
|
4
|
Zhu F, Nie G. Cell reprogramming: methods, mechanisms and applications. CELL REGENERATION (LONDON, ENGLAND) 2025; 14:12. [PMID: 40140235 PMCID: PMC11947411 DOI: 10.1186/s13619-025-00229-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 02/05/2025] [Accepted: 03/09/2025] [Indexed: 03/28/2025]
Abstract
Cell reprogramming represents a powerful approach to achieve the conversion cells of one type into cells of another type of interest, which has substantially changed the landscape in the field of developmental biology, regenerative medicine, disease modeling, drug discovery and cancer immunotherapy. Cell reprogramming is a complex and ordered process that involves the coordination of transcriptional, epigenetic, translational and metabolic changes. Over the past two decades, a range of questions regarding the facilitators/barriers, the trajectories, and the mechanisms of cell reprogramming have been extensively investigated. This review summarizes the recent advances in cell reprogramming mediated by transcription factors or chemical molecules, followed by elaborating on the important roles of biophysical cues in cell reprogramming. Additionally, this review will detail our current understanding of the mechanisms that govern cell reprogramming, including the involvement of the recently discovered biomolecular condensates. Finally, the review discusses the broad applications and future directions of cell reprogramming in developmental biology, disease modeling, drug development, regenerative/rejuvenation therapy, and cancer immunotherapy.
Collapse
Affiliation(s)
- Fei Zhu
- Wisdom Lake Academy of Pharmacy, Xi'an Jiaotong-Liverpool University, Suzhou, 215123, China.
| | - Guangjun Nie
- CAS Key Laboratory for Biomedical Effects of Nanomaterials and Nanosafety, CAS Center of Excellence in Nanoscience National Center for Nanoscience and Technology, Beijing, 100190, China.
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
5
|
Kang N, Hwang J, Jeong D, Choi JH, Thangam R, Min S, Hong H, Kim D, Rha H, Lee S, Jung H, Kim T, Zare I, Jung HJ, Najafabadi AH, Jung HD, Zhang K, Zhao P, Bian L, Kim HK, Kim JS, Song G, Yoon J, Park SG, Jang WY, Kang H. Ligand Inter-Relation Analysis Via Graph Theory Predicts Macrophage Response. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2414356. [PMID: 39719659 DOI: 10.1002/adma.202414356] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/10/2024] [Indexed: 12/26/2024]
Abstract
Graph theory has been widely used to quantitatively analyze complex networks of molecules, materials, and cells. Analyzing the dynamic complex structure of extracellular matrix can predict cell-material interactions but has not yet been demonstrated. In this study, graph theory-based mathematical modeling of RGD ligand graph inter-relation is demonstrated by differentially cutting off RGD-to-RGD interlinkages with flexibly conjugated magnetic nanobars (MNBs) with tunable aspect ratio. The RGD-to-RGD interlinkages are less effectively cut off by MNBs with a lower aspect ratio, which decreases the shortest path while increasing the number of instances thereof, thereby augmenting RGD nano inter-relation. This facilitates integrin recruitment of macrophages and thus actin fiber assembly and vinculin expression, which mediates pro-regenerative polarization, involving myosin II, actin polymerization, and rho-associated protein kinase. Unidirectional pre-aligning or reversibly lifting highly elongated MNBs both increase RGD nano inter-relation, which promotes host macrophage adhesion and switches their polarization from pro-inflammatory to pro-regenerative phenotype. The latter approach produces nano-spaces through which macrophages can penetrate and establish RGD links thereunder. Using graph theory, this study presents the example of mathematically modeling the functionality of extracellular-matrix-mimetic materials, which can help elucidate complex dynamics of the interactions occurring between host cells and materials via versatile geometrical nano-engineering.
Collapse
Affiliation(s)
- Nayeon Kang
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Jangsun Hwang
- Department of Orthopedic Surgery, Korea University Anam Hospital, Seoul, 02841, Republic of Korea
| | - Daun Jeong
- Department of Orthopedic Surgery, Korea University Anam Hospital, Seoul, 02841, Republic of Korea
| | - Ji Hye Choi
- Department of Orthopedic Surgery, Korea University Anam Hospital, Seoul, 02841, Republic of Korea
| | - Ramar Thangam
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Sunhong Min
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Hyunsik Hong
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Dahee Kim
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Hyunji Rha
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Sungkyu Lee
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Hwapyung Jung
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Taeeon Kim
- Department of Nano-Bio Convergence, Korea Institute of Materials Science (KIMS), Changwon, Gyeongnam, 51508, Republic of Korea
- Department of Future Convergence Materials, Korea University, Seoul, 02841, Republic of Korea
| | - Iman Zare
- Research and Development Department, Sina Medical Biochemistry Technologies Co. Ltd., Shiraz, 7178795844, Iran
| | - Hee Joon Jung
- Interdisciplinary Materials Measurements Institute, Korea Research Institute of Standards and Science (KRISS), Daejeon, 34113, Republic of Korea
| | | | - Hyun-Do Jung
- Division of Materials Science and Engineering, Hanyang University, Seoul, 04763, Republic of Korea
| | - Kunyu Zhang
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Pengchao Zhao
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Liming Bian
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Hong-Kyu Kim
- Advanced Analysis Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jong Seung Kim
- Department of Chemistry, Korea University, Seoul, 02841, Republic of Korea
| | - Guosheng Song
- State Key Laboratory of Chemo/Biosensing and Chemometrics, College of Chemistry and Chemical Engineering, Hunan University, Changsha, 410082, China
| | - Juyoung Yoon
- Department of Chemistry and Nanoscience, Ewha Womans University, Seoul, 03760, Republic of Korea
| | - Sung-Gyu Park
- Department of Nano-Bio Convergence, Korea Institute of Materials Science (KIMS), Changwon, Gyeongnam, 51508, Republic of Korea
- Department of Future Convergence Materials, Korea University, Seoul, 02841, Republic of Korea
| | - Woo Young Jang
- Department of Orthopedic Surgery, Korea University Anam Hospital, Seoul, 02841, Republic of Korea
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
- Department of Future Convergence Materials, Korea University, Seoul, 02841, Republic of Korea
- College of Medicine, Korea University, Seoul, 02841, Republic of Korea
| |
Collapse
|
6
|
Seo SJ, Jin Y. Enhancing Cardiomyocyte Purity through Lactate-Based Metabolic Selection. Tissue Eng Regen Med 2025; 22:249-260. [PMID: 39820961 PMCID: PMC11794935 DOI: 10.1007/s13770-024-00696-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/20/2024] [Accepted: 12/22/2024] [Indexed: 01/19/2025] Open
Abstract
BACKGROUND Direct reprogramming of fibroblasts into chemically induced cardiomyocyte-like cells (CiCMs) through small molecules presents a promising cell source for cardiac regeneration and therapeutic development. However, the contaminating non-cardiomyocytes, primarily unconverted fibroblasts, reduce the effectiveness of CiCMs in various applications. This study investigated a metabolic selection approach using lactate to enrich CiCMs by exploiting the unique metabolic capability of cardiomyocytes to utilize lactate as an alternative energy source. METHODS Primary mouse embryonic fibroblasts (pMEFs) were reprogrammed into CiCMs and subjected to a glucose-depleted, lactate-supplemented medium for 4 days. Afterward, cell viability was analyzed, and cardiomyocyte efficiency was assessed through the expression of cardiac-specific markers. Additionally, electrophysiological function was evaluated by examining drug-induced responses. RESULTS The lactate treatment led to a significant decrease in the viability of non-cardiomyocytes (pMEF-LAC), while CiCMs (CiCM-LAC) showed minimal cell death. Specifically, the expression of all cardiac-related markers was increased in CiCM-LAC. Metabolically purified CiCMs exhibited enhanced contractile force and increased contraction frequency compared to non-purified CiCMs, as well as an elevated responsiveness to drugs. CONCLUSION This study demonstrates that lactate-based metabolic selection is an effective and practical approach for enriching CiCMs, offering a cost-effective alternative to other purification methods. The application of this strategy could potentially broaden the accessibility and utility of reprogrammed cardiomyocytes in cardiac regeneration and therapeutic development.
Collapse
Affiliation(s)
- Seung Ju Seo
- Department of Physiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea
| | - Yoonhee Jin
- Department of Physiology, Graduate School of Medical Science, Brain Korea 21 Project, Yonsei University College of Medicine, Seoul, 03722, Republic of Korea.
| |
Collapse
|
7
|
Nam Y, Song Y, Seo SJ, Ko GR, Lee SH, Cha E, Kwak SM, Kim S, Shin M, Jin Y, Lee JS. Metabolic reprogramming via mitochondrial delivery for enhanced maturation of chemically induced cardiomyocyte-like cells. MedComm (Beijing) 2024; 5:e70005. [PMID: 39611044 PMCID: PMC11604293 DOI: 10.1002/mco2.70005] [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: 03/18/2024] [Revised: 09/03/2024] [Accepted: 09/03/2024] [Indexed: 11/30/2024] Open
Abstract
Heart degenerative diseases pose a significant challenge due to the limited ability of native heart to restore lost cardiomyocytes. Direct cellular reprogramming technology, particularly the use of small molecules, has emerged as a promising solution to prepare functional cardiomyocyte through faster and safer processes without genetic modification. However, current methods of direct reprogramming often exhibit low conversion efficiencies and immature characteristics of the generated cardiomyocytes, limiting their use in regenerative medicine. This study proposes the use of mitochondrial delivery to metabolically reprogram chemically induced cardiomyocyte-like cells (CiCMs), fostering enhanced maturity and functionality. Our findings show that mitochondria sourced from high-energy-demand organs (liver, brain, and heart) can enhance structural maturation and metabolic functions. Notably, heart-derived mitochondria resulted in CiCMs with a higher oxygen consumption rate capacity, enhanced electrical functionality, and higher sensitivity to hypoxic condition. These results are related to metabolic changes caused by increased number and size of mitochondria and activated mitochondrial fusion after mitochondrial treatment. In conclusion, our study suggests that mitochondrial delivery into CiCMs can be an effective strategy to promote cellular maturation, potentially contributing to the advancement of regenerative medicine and disease modeling.
Collapse
Affiliation(s)
- Yena Nam
- Department of PhysiologyGraduate School of Medical ScienceBrain Korea 21 ProjectYonsei University College of MedicineSeoulRepublic of Korea
| | - Yoonji Song
- Department of Biomedical EngineeringSungkyunkwan University (SKKU)SuwonRepublic of Korea
| | - Seung Ju Seo
- Department of PhysiologyGraduate School of Medical ScienceBrain Korea 21 ProjectYonsei University College of MedicineSeoulRepublic of Korea
| | - Ga Ryang Ko
- Department of Intelligent Precision Healthcare ConvergenceSungkyunkwan University (SKKU)SuwonRepublic of Korea
| | - Seung Hyun Lee
- Department of Intelligent Precision Healthcare ConvergenceSungkyunkwan University (SKKU)SuwonRepublic of Korea
| | - Eunju Cha
- Department of PhysiologyGraduate School of Medical ScienceBrain Korea 21 ProjectYonsei University College of MedicineSeoulRepublic of Korea
| | - Su Min Kwak
- Department of MedicineCollege of MedicineYonsei University Graduate SchoolSeoulRepublic of Korea
| | - Sumin Kim
- Department of Intelligent Precision Healthcare ConvergenceSungkyunkwan University (SKKU)SuwonRepublic of Korea
| | - Mikyung Shin
- Department of Biomedical EngineeringSungkyunkwan University (SKKU)SuwonRepublic of Korea
- Department of Intelligent Precision Healthcare ConvergenceSungkyunkwan University (SKKU)SuwonRepublic of Korea
- Center for Neuroscience Imaging ResearchInstitute for Basic Science (IBS)SuwonRepublic of Korea
| | - Yoonhee Jin
- Department of PhysiologyGraduate School of Medical ScienceBrain Korea 21 ProjectYonsei University College of MedicineSeoulRepublic of Korea
- Department of MedicineCollege of MedicineYonsei University Graduate SchoolSeoulRepublic of Korea
| | - Jung Seung Lee
- Department of Biomedical EngineeringSungkyunkwan University (SKKU)SuwonRepublic of Korea
- Department of Intelligent Precision Healthcare ConvergenceSungkyunkwan University (SKKU)SuwonRepublic of Korea
- Department of MetaBioHealthSungkyunkwan University (SKKU)SuwonRepublic of Korea
| |
Collapse
|
8
|
Li J, Zhou M, Xie J, Chen J, Yang M, Ye C, Cheng S, Liu M, Li R, Tan R. Organoid modeling meets cancers of female reproductive tract. Cell Death Discov 2024; 10:410. [PMID: 39333482 PMCID: PMC11437045 DOI: 10.1038/s41420-024-02186-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 08/13/2024] [Accepted: 09/18/2024] [Indexed: 09/29/2024] Open
Abstract
Diseases of the female reproductive system, especially malignant tumors, pose a serious threat to women's health worldwide. One of the key factors limiting research progress in this area is the lack of representative models. Organoid technology, especially tumor organoids, has been increasingly applied in the study of female reproductive system tumors due to their high heterogeneity, close resemblance to the physiological state, easy acquisition and cultivation advantages. They play a significant role in understanding the origin and causes of tumors, drug screening, and personalized treatment and more. This article reviews the organoid models for the female reproductive system, focusing on the cancer research advancements. It discusses the methods for constructing tumor organoids of the female reproductive tract and summarizes the limitations of current research. The aim is to offer a reference for future development and application of these organoid models, contributing to the advancement of anti-tumor drugs and treatment strategies for female reproductive tract cancer patients.
Collapse
Affiliation(s)
- Jiao Li
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
- West China School of Pharmacy, Sichuan University, Chengdu, China
| | - Mengting Zhou
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jun Xie
- Information Technology Center, West China Hospital of Sichuan University, Sichuan University, Chengdu, China
| | - Jiani Chen
- Chongqing Medical University, Chongqing, China
| | - Mengni Yang
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Changjun Ye
- Rehabilitation Department, Changgeng Yining Hospital, Wenzhou, China
| | - Shihu Cheng
- Geriatric Department, Changgeng Yining Hospital, Wenzhou, China
| | - Miao Liu
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA.
| | - Rui Li
- Department of Radiation Oncology, Radiation Oncology Key Laboratory of Sichuan Province, Sichuan Clinical Research Center for Cancer, Sichuan Cancer Hospital and Institute, Sichuan Cancer Center, Affiliated Cancer Hospital of University of Electronic Science and Technology of China, Chengdu, China.
| | - Ruirong Tan
- Translational Chinese Medicine Key Laboratory of Sichuan, Sichuan-Chongqing Joint Key Laboratory of Innovation of New Drugs of Traditional Chinese Medicine, Sichuan Institute for Translational Chinese Medicine, Sichuan Academy of Chinese Medicine Sciences, Chengdu, China.
| |
Collapse
|
9
|
Bocheng X, França R. Innovative 3D bioprinting approaches for advancing brain science and medicine: a literature review. Biomed Phys Eng Express 2024; 10:062002. [PMID: 39260389 DOI: 10.1088/2057-1976/ad795c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 09/11/2024] [Indexed: 09/13/2024]
Abstract
The rapid advancements in 3D printing technology have revolutionized the field of tissue engineering, particularly in the development of neural tissues for the treatment of nervous system diseases. Brain neural tissue, composed of neurons and glial cells, plays a crucial role in the functioning of the brain, spinal cord, and peripheral nervous system by transmitting nerve impulses and processing information. By leveraging 3D bioprinting and bioinks, researchers can create intricate neural scaffolds that facilitate the proliferation and differentiation of nerve cells, thereby promoting the repair and regeneration of damaged neural tissues. This technology allows for the precise spatial arrangement of various cell types and scaffold materials, enabling the construction of complex neural tissue models that closely mimic the natural architecture of the brain. Human-induced pluripotent stem cells (hiPSCs) have emerged as a groundbreaking tool in neuroscience research and the potential treatment of neurological diseases. These cells can differentiate into diverse cell types within the nervous system, including neurons, astrocytes, microglia, oligodendrocytes, and Schwann cells, providing a versatile platform for studying neural networks, neurodevelopment, and neurodegenerative disorders. The use of hiPSCs also opens new avenues for personalized medicine, allowing researchers to model diseases and develop targeted therapies based on individual patient profiles. Despite the promise of direct hiPSC injections for therapeutic purposes, challenges such as poor localization and limited integration have led to the exploration of biomaterial scaffolds as supportive platforms for cell delivery and tissue regeneration. This paper reviews the integration of 3D bioprinting technologies and bioink materials in neuroscience applications, offering a unique platform to create complex brain and tissue architectures that mimic the mechanical, architectural, and biochemical properties of native tissues. These advancements provide robust tools for modelling, repair, and drug screening applications. The review highlights current research, identifies research gaps, and offers recommendations for future studies on 3D bioprinting in neuroscience. The investigation demonstrates the significant potential of 3D bioprinting to fabricate brain-like tissue constructs, which holds great promise for regenerative medicine and drug testing models. This approach offers new avenues for studying brain diseases and potential treatments.
Collapse
Affiliation(s)
- Xu Bocheng
- Department of Biomedical Engineering, Price Faculty of Engineering, University of Manitoba, Winnipeg, R3E 0W2, Canada
| | - Rodrigo França
- Department of Biomedical Engineering, Price Faculty of Engineering, University of Manitoba, Winnipeg, R3E 0W2, Canada
- Rady Faculty of Health Science, Dental Biomaterials Research Lab, University of Manitoba, Winnipeg, R3E 0W2, Canada
| |
Collapse
|
10
|
Kong JS, Kim JJ, Riva L, Ginestra PS, Cho DW. In vitrothree-dimensional volumetric printing of vitreous body models using decellularized extracellular matrix bioink. Biofabrication 2024; 16:045030. [PMID: 39142325 DOI: 10.1088/1758-5090/ad6f46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Accepted: 08/14/2024] [Indexed: 08/16/2024]
Abstract
Hyalocytes, which are considered to originate from the monocyte/macrophage lineage, play active roles in vitreous collagen and hyaluronic acid synthesis. Obtaining a hyalocyte-compatible bioink during the 3D bioprinting of eye models is challenging. In this study, we investigated the suitability of a cartilage-decellularized extracellular matrix (dECM)-based bioink for printing a vitreous body model. Given that achieving a 3D structure and environment identical to those of the vitreous body necessitates good printability and biocompatibility, we examined the mechanical and biological properties of the developed dECM-based bioink. Furthermore, we proposed a 3D bioprinting strategy for volumetric vitreous body fabrication that supports cell viability, transparency, and self-sustainability. The construction of a 3D structure composed of bioink microfibers resulted in improved transparency and hyalocyte-like macrophage activity in volumetric vitreous mimetics, mimicking real vitreous bodies. The results indicate that our 3D structure could serve as a platform for drug testing in disease models and demonstrate that the proposed printing technology, utilizing a dECM-based bioink and volumetric vitreous body, has the potential to facilitate the development of advanced eye models for future studies on floater formation and visual disorders.
Collapse
Affiliation(s)
- Jeong Sik Kong
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk 37673, Republic of Korea
- POSTECH-Catholic Biomedical Engineering Institute, POSTECH, Pohang, Kyungbuk 37673, Republic of Korea
| | - Joeng Ju Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk 37673, Republic of Korea
- POSTECH-Catholic Biomedical Engineering Institute, POSTECH, Pohang, Kyungbuk 37673, Republic of Korea
| | - Leonardo Riva
- Department of Industrial and Mechanical Engineering, University of Brescia, Via Branze 38, 25125 Brescia, Italy
| | - Paola Serena Ginestra
- Department of Industrial and Mechanical Engineering, University of Brescia, Via Branze 38, 25125 Brescia, Italy
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk 37673, Republic of Korea
- POSTECH-Catholic Biomedical Engineering Institute, POSTECH, Pohang, Kyungbuk 37673, Republic of Korea
| |
Collapse
|
11
|
Kim D, Lee MJ, Arai Y, Ahn J, Lee GW, Lee SH. Ultrasound-triggered three dimensional hyaluronic acid hydrogel promotes in vitro and in vivo reprogramming into induced pluripotent stem cells. Bioact Mater 2024; 38:331-345. [PMID: 38764447 PMCID: PMC11101682 DOI: 10.1016/j.bioactmat.2024.05.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2024] [Revised: 04/12/2024] [Accepted: 05/05/2024] [Indexed: 05/21/2024] Open
Abstract
Cellular reprogramming technologies have been developed with different physicochemical factors to improve the reprogramming efficiencies of induced pluripotent stem cells (iPSCs). Ultrasound is a clinically applied noncontact biophysical factor known for regulating various cellular behaviors but remains uninvestigated for cellular reprogramming. Here, we present a new reprogramming strategy using low-intensity ultrasound (LIUS) to improve cellular reprogramming of iPSCs in vitro and in vivo. Under 3D microenvironment conditions, increased LIUS stimulation shows enhanced cellular reprogramming of the iPSCs. The cellular reprogramming process facilitated by LIUS is accompanied by increased mesenchymal to epithelial transition and histone modification. LIUS stimulation transiently modulates the cytoskeletal rearrangement, along with increased membrane fluidity and mobility to increase HA/CD44 interactions. Furthermore, LIUS stimulation with HA hydrogel can be utilized in application of both human cells and in vivo environment, for enhanced reprogrammed cells into iPSCs. Thus, LIUS stimulation with a combinatorial 3D microenvironment system can improve cellular reprogramming in vitro and in vivo environments, which can be applied in various biomedical fields.
Collapse
Affiliation(s)
| | | | - Yoshie Arai
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620, Seoul, South Korea
| | - Jinsung Ahn
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620, Seoul, South Korea
| | - Gun Woo Lee
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620, Seoul, South Korea
| | - Soo-Hong Lee
- Department of Biomedical Engineering, Dongguk University-Seoul, 04620, Seoul, South Korea
| |
Collapse
|
12
|
Yang M, Cai X, Wang C, Li P, Chen S, Liu C, Wang Y, Qian K, Dong Q, Xue F, Chu C, Bai J, Liu Q, Ni X. Humidity-Responsive Amorphous Calcium-Magnesium Pyrophosphate/Cassava Starch Scaffold for Enhanced Neurovascular Bone Regeneration. ACS APPLIED MATERIALS & INTERFACES 2024; 16:35964-35984. [PMID: 38968558 DOI: 10.1021/acsami.4c03204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/07/2024]
Abstract
Developing a neurovascular bone repair scaffold with an appropriate mechanical strength remains a challenge. Calcium phosphate (CaP) is similar to human bone, but its scaffolds are inherently brittle and inactive, which require recombination with active ions and polymers for bioactivity and suitable strength. This work discussed the synthesis of amorphous magnesium-calcium pyrophosphate (AMCP) and the subsequent development of a humidity-responsive AMCP/cassava starch (CS) scaffold. The scaffold demonstrated enhanced mechanical properties by strengthening the intermolecular hydrogen bonds and ionic bonds between AMCP and CS during the gelatinization and freeze-thawing processes. The release of active ions was rapid initially and stabilized into a long-term stable release after 3 days, which is well-matched with new bone growth. The release of pyrophosphate ions endowed the scaffold with antibacterial properties. At the cellular level, the released active ions simultaneously promoted the proliferation and mineralization of osteoblasts, the proliferation and migration of endothelial cells, and the proliferation of Schwann cells. At the animal level, the scaffold was demonstrated to promote vascular growth and peripheral nerve regeneration in a rat skull defect experiment, ultimately resulting in the significant and rapid repair of bone defects. The construction of the AMCP/CS scaffold offers practical suggestions and references for neurovascular bone repair.
Collapse
Affiliation(s)
- Mengmeng Yang
- School of Materials Science and Engineering, Southeast University, Nanjing 211189, Jiangsu, China
- Jiangsu Key Laboratory for Advanced Metallic Materials, Nanjing 211189, Jiangsu, China
- Institute of Biomedical Devices (Suzhou), Southeast University, Suzhou 215163, China
| | - Xiang Cai
- School of Materials Science and Engineering, Southeast University, Nanjing 211189, Jiangsu, China
- Jiangsu Key Laboratory for Advanced Metallic Materials, Nanjing 211189, Jiangsu, China
| | - Cheng Wang
- School of Materials Science and Engineering, Southeast University, Nanjing 211189, Jiangsu, China
| | - Pengyin Li
- Center of Medical Physics, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213003, Jiangsu, China
| | - Shaoqing Chen
- Center of Medical Physics, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213003, Jiangsu, China
| | - Chun Liu
- Center of Medical Physics, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213003, Jiangsu, China
| | - Yao Wang
- Department of Emergency, The First Affiliated Hospital of Nanjing Medical University, Nanjing 210029, China
| | - Kun Qian
- School of Materials Science and Engineering, Southeast University, Nanjing 211189, Jiangsu, China
| | - Qiangsheng Dong
- School of Materials Science and Engineering, Nanjing Institute of Technology, Nanjing 211167, China
| | - Feng Xue
- School of Materials Science and Engineering, Southeast University, Nanjing 211189, Jiangsu, China
- Jiangsu Key Laboratory for Advanced Metallic Materials, Nanjing 211189, Jiangsu, China
- Institute of Biomedical Devices (Suzhou), Southeast University, Suzhou 215163, China
| | - Chenglin Chu
- School of Materials Science and Engineering, Southeast University, Nanjing 211189, Jiangsu, China
- Jiangsu Key Laboratory for Advanced Metallic Materials, Nanjing 211189, Jiangsu, China
| | - Jing Bai
- School of Materials Science and Engineering, Southeast University, Nanjing 211189, Jiangsu, China
- Jiangsu Key Laboratory for Advanced Metallic Materials, Nanjing 211189, Jiangsu, China
- Institute of Biomedical Devices (Suzhou), Southeast University, Suzhou 215163, China
- Jiangsu Key Laboratory for Light Metal Alloys, Nanjing 211212, China
| | - Qizhan Liu
- School of Materials Science and Engineering, Southeast University, Nanjing 211189, Jiangsu, China
- Center for Global Health, The Key Laboratory of Modern Toxicology, Ministry of Education, School of Public Health, Nanjing Medical University, Nanjing 211166, Jiangsu, China
| | - Xinye Ni
- Center of Medical Physics, The Affiliated Changzhou No. 2 People's Hospital of Nanjing Medical University, Changzhou 213003, Jiangsu, China
| |
Collapse
|
13
|
Bae M, Ngo H, Kang YJ, Lee SJ, Park W, Jo Y, Choi YM, Kim JJ, Yi HG, Kim HS, Jang J, Cho DW, Cho H. Laminin-Augmented Decellularized Extracellular Matrix Ameliorating Neural Differentiation and Neuroinflammation in Human Mini-Brains. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2308815. [PMID: 38161254 DOI: 10.1002/smll.202308815] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/03/2023] [Revised: 11/21/2023] [Indexed: 01/03/2024]
Abstract
Non-neural extracellular matrix (ECM) has limited application in humanized physiological neural modeling due to insufficient brain-specificity and safety concerns. Although brain-derived ECM contains enriched neural components, certain essential components are partially lost during the decellularization process, necessitating augmentation. Here, it is demonstrated that the laminin-augmented porcine brain-decellularized ECM (P-BdECM) is xenogeneic factor-depleted as well as favorable for the regulation of human neurons, astrocytes, and microglia. P-BdECM composition is comparable to human BdECM regarding brain-specificity through the matrisome and gene ontology-biological process analysis. As augmenting strategy, laminin 111 supplement promotes neural function by synergic effect with laminin 521 in P-BdECM. Annexin A1(ANXA1) and Peroxiredoxin(PRDX) in P-BdECM stabilized microglial and astrocytic behavior under normal while promoting active neuroinflammation in response to neuropathological factors. Further, supplementation of the brain-specific molecule to non-neural matrix also ameliorated glial cell inflammation as in P-BdECM. In conclusion, P-BdECM-augmentation strategy can be used to recapitulate humanized pathophysiological cerebral environments for neurological study.
Collapse
Affiliation(s)
- Mihyeon Bae
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk, 37673, South Korea
| | - Huyen Ngo
- Department of Biophysics, Institute of Quantum Biophysics, Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Gyeonggi, 16419, South Korea
| | - You Jung Kang
- Department of Biophysics, Institute of Quantum Biophysics, Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Gyeonggi, 16419, South Korea
| | - Su-Jin Lee
- Biomedical Research Institute, Chonnam National University Hospital, Gwangju, 61469, South Korea
| | - Wonbin Park
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk, 37673, South Korea
| | - Yeonggwon Jo
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk, 37673, South Korea
| | - Yoo-Mi Choi
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk, 37673, South Korea
| | - Joeng Ju Kim
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk, 37673, South Korea
| | - Hee-Gyeong Yi
- Department of Convergence Biosystems Engineering, College of Agriculture and Life Sciences, Chonnam National University, Gwangju, 61186, South Korea
| | - Hyung-Seok Kim
- Department of Forensic medicine, Chonnam National University Medical School & Research Institute of Medical Sciences, Gwangju, 61469, South Korea
| | - Jinah Jang
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk, 37673, South Korea
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk, 37673, South Korea
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Kyungbuk, 37673, South Korea
| | - Hansang Cho
- Department of Biophysics, Institute of Quantum Biophysics, Department of Intelligent Precision Healthcare Convergence, Sungkyunkwan University, Suwon, Gyeonggi, 16419, South Korea
| |
Collapse
|
14
|
Wang J, Yang X, Xu M, Liu H, Liu L, Tan Z. Distinct cellular microenvironment with cytotypic effects regulates orderly regeneration of vascular tissues. Mater Today Bio 2024; 26:101033. [PMID: 38533377 PMCID: PMC10963652 DOI: 10.1016/j.mtbio.2024.101033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2023] [Revised: 02/26/2024] [Accepted: 03/15/2024] [Indexed: 03/28/2024] Open
Abstract
Regeneration of the architecturally complex blood vascular system requires precise temporal and spatial control of cell behaviours. Additional components must be integrated into the structure to achieve clinical success for in situ tissue engineering. Consequently, this study proposed a universal method for including any substrate type in vascular cell extracellular matrices (VCEM) via regulating selective adhesion to promote vascular tissue regeneration. The results uncovered that the VCEM worked as cell adhesion substrates, exhibited cell type specificity, and functioned as an address signal for recognition by vascular cells, which resulted in matching with the determined cells. The qPCR and immunofluorescence results revealed that a cell type-specific VCEM could be designed to promote or inhibit cell adhesion, consistenting with the expression patterns of eyes absent 3 (Eya3). In addition, a 3D vascular graft combined with VCEM which could recapitulate the vascular cell-like microenvironment was fabricated. The vascular graft revealed a prospective role for cellular microenvironment in the establishment of vascular cell distribution and tissue architecture, and potentiated the orderly regeneration and functional recovery of vascular tissues in vivo. The findings demonstrate that differential adhesion between cell types due to the cellular microenvironment is sufficient to drive the complex assembly of engineered blood vessel functional units, and underlies hierarchical organization during vascular regeneration.
Collapse
Affiliation(s)
- Jian Wang
- College of Biology, Hunan University, Changsha, 410082, China
- Institute of Shenzhen, Hunan University Shenzhen, 518000, China
| | - Xun Yang
- Department of Traumatic Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital, Shenzhen University), Shenzhen, 518028, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Miaomiao Xu
- College of Biology, Hunan University, Changsha, 410082, China
- Greater Bay Area Institute for Innovation, Hunan University, Guangzhou, 511300, China
| | - Hui Liu
- College of Biology, Hunan University, Changsha, 410082, China
- Greater Bay Area Institute for Innovation, Hunan University, Guangzhou, 511300, China
| | - Lijun Liu
- Department of Traumatic Orthopedics, Shenzhen Second People's Hospital (The First Affiliated Hospital, Shenzhen University), Shenzhen, 518028, China
- Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, National-Regional Key Technology Engineering Laboratory for Medical Ultrasound, School of Biomedical Engineering, Shenzhen University Medical School, Shenzhen, 518060, China
| | - Zhikai Tan
- College of Biology, Hunan University, Changsha, 410082, China
- Institute of Shenzhen, Hunan University Shenzhen, 518000, China
- Greater Bay Area Institute for Innovation, Hunan University, Guangzhou, 511300, China
| |
Collapse
|
15
|
Yoon H, Kang JH, Cho SW, Park CG, Kim DW, Park TE. Brain-Decellularized ECM-Based 3D Myeloid Sarcoma Platform: Mimicking Adaptive Phenotypic Alterations in the Brain. Adv Healthc Mater 2024; 13:e2304371. [PMID: 38320209 DOI: 10.1002/adhm.202304371] [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: 12/09/2023] [Revised: 01/31/2024] [Indexed: 02/08/2024]
Abstract
Leukemia circulates in the bloodstream and induces various symptoms and complications. Occasionally, these cells accumulate in non-marrow tissues, forming a tumor-like myeloid sarcoma (MS). When the blast-stage leukemia cells invade the brain parenchyma, intracranial MS occurs, leading to a challenging prognosis owing to the limited penetration of cytostatic drugs into the brain and the development of drug resistance. The scarcity of tissue samples from MS makes understanding the phenotypic changes occurring in leukemia cells within the brain environment challenging, thereby hindering development of effective treatment strategies for intracranial MS. This study presents a novel 3D in vitro model mimicking intracranial MS, employing a hydrogel scaffold derived from the brain-decellularized extracellular matrix in which suspended leukemia cells are embedded, simulating the formation of tumor masses in the brain parenchyma. This model reveals marked phenotypic changes in leukemia cells, including altered survival, proliferation, differentiation, and cell cycle regulation. Notably, proportion of dormant leukemia stem cells increases and expression of multidrug resistance genes is upregulated, leading to imatinib resistance, mirroring the pathological features of in vivo MS tissue. Furthermore, suppression of ferroptosis is identified as an important characteristic of intracranial MS, providing valuable insights for the development of targeted therapeutic strategies.
Collapse
Affiliation(s)
- Heejeong Yoon
- Department of Biomedical Engineering, College of Information and Biotechnology, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Joo H Kang
- Department of Biomedical Engineering, College of Information and Biotechnology, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Seung Woo Cho
- Department of Biomedical Engineering, College of Information and Biotechnology, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Chun Gwon Park
- Department of Biomedical Engineering, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
- Department of Intelligent Precision Healthcare Convergence, SKKU Institute for Convergence, Sungkyunkwan University (SKKU), Suwon, 16419, Republic of Korea
| | - Dong-Wook Kim
- Department of Hematology, Hematology Center, Uijeongbu Eulji Medical Center, Eulji University, Uijeongbu, 11750, Republic of Korea
- Leukemia Omics Research Institute, Eulji University, Uijeongbu, 11750, Republic of Korea
| | - Tae-Eun Park
- Department of Biomedical Engineering, College of Information and Biotechnology, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| |
Collapse
|
16
|
Ortega JA, Soares de Aguiar GP, Chandravanshi P, Levy N, Engel E, Álvarez Z. Exploring the properties and potential of the neural extracellular matrix for next-generation regenerative therapies. WILEY INTERDISCIPLINARY REVIEWS. NANOMEDICINE AND NANOBIOTECHNOLOGY 2024; 16:e1962. [PMID: 38723788 DOI: 10.1002/wnan.1962] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/20/2023] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 05/24/2024]
Abstract
The extracellular matrix (ECM) is a dynamic and complex network of proteins and molecules that surrounds cells and tissues in the nervous system and orchestrates a myriad of biological functions. This review carefully examines the diverse interactions between cells and the ECM, as well as the transformative chemical and physical changes that the ECM undergoes during neural development, aging, and disease. These transformations play a pivotal role in shaping tissue morphogenesis and neural activity, thereby influencing the functionality of the central nervous system (CNS). In our comprehensive review, we describe the diverse behaviors of the CNS ECM in different physiological and pathological scenarios and explore the unique properties that make ECM-based strategies attractive for CNS repair and regeneration. Addressing the challenges of scalability, variability, and integration with host tissues, we review how advanced natural, synthetic, and combinatorial matrix approaches enhance biocompatibility, mechanical properties, and functional recovery. Overall, this review highlights the potential of decellularized ECM as a powerful tool for CNS modeling and regenerative purposes and sets the stage for future research in this exciting field. This article is categorized under: Implantable Materials and Surgical Technologies > Nanotechnology in Tissue Repair and Replacement Therapeutic Approaches and Drug Discovery > Nanomedicine for Neurological Disease Implantable Materials and Surgical Technologies > Nanomaterials and Implants.
Collapse
Affiliation(s)
- J Alberto Ortega
- Department of Pathology and Experimental Therapeutics, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
- Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet del Llobregat, Spain
| | - Gisele P Soares de Aguiar
- Department of Pathology and Experimental Therapeutics, Institute of Neurosciences, University of Barcelona, L'Hospitalet de Llobregat, Barcelona, Spain
- Institut d'Investigació Biomèdica de Bellvitge (IDIBELL), L'Hospitalet del Llobregat, Spain
| | - Palash Chandravanshi
- Biomaterials for Neural Regeneration Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Natacha Levy
- Biomaterials for Neural Regeneration Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Elisabeth Engel
- IMEM-BRT Group, Department of Materials Science and Engineering, EEBE, Technical University of Catalonia (UPC), Barcelona, Spain
- Biomaterials for Regenerative Therapies Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
| | - Zaida Álvarez
- Biomaterials for Neural Regeneration Group, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- CIBER en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid, Spain
- Simpson Querrey Institute for BioNanotechnology, Northwestern University, Chicago, Illinois, USA
| |
Collapse
|
17
|
Arai K, Saito F, Miyazaki M, Kushige H, Izu Y, Maeta N, Yamazoe K. Small Molecules Temporarily Induce Neuronal Features in Adult Canine Dermal Fibroblasts. Int J Mol Sci 2023; 24:15804. [PMID: 37958789 PMCID: PMC10648228 DOI: 10.3390/ijms242115804] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2023] [Revised: 10/28/2023] [Accepted: 10/30/2023] [Indexed: 11/15/2023] Open
Abstract
Several methods have been developed to generate neurons from other cell types for performing regeneration therapy and in vitro studies of central nerve disease. Small molecules (SMs) can efficiently induce neuronal features in human and rodent fibroblasts without transgenes. Although canines have been used as a spontaneous disease model of human central nerve, efficient neuronal reprogramming method of canine cells have not been well established. We aimed to induce neuronal features in adult canine dermal fibroblasts (ACDFs) by SMs and assess the permanency of these changes. ACDFs treated with eight SMs developed a round-shaped cell body with branching processes and expressed neuronal proteins, including βIII-tubulin, microtubule-associated protein 2 (MAP2), and neurofilament-medium. Transcriptome profiling revealed the upregulation of neuron-related genes, such as SNAP25 and GRIA4, and downregulation of fibroblast-related genes, such as COL12A1 and CCN5. Calcium fluorescent imaging demonstrated an increase in intracellular Ca2+ concentration upon stimulation with glutamate and KCl. Although neuronal features were induced similarly in basement membrane extract droplet culture, they diminished after culturing without SMs or in vivo transplantation into an injured spinal cord. In conclusion, SMs temporarily induce neuronal features in ACDFs. However, the analysis of bottlenecks in the neuronal induction is crucial for optimizing the process.
Collapse
Affiliation(s)
- Kiyotaka Arai
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Okayama University of Science, 1-3 Ikoi-no-oka, Imabari 794-8555, Japan; (M.M.); (H.K.); (N.M.); (K.Y.)
| | - Fumiyo Saito
- Department of Toxicology, Faculty of Veterinary Medicine, Okayama University of Science, 1-3 Ikoi-no-oka, Imabari 794-8555, Japan;
| | - Masashi Miyazaki
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Okayama University of Science, 1-3 Ikoi-no-oka, Imabari 794-8555, Japan; (M.M.); (H.K.); (N.M.); (K.Y.)
| | - Haruto Kushige
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Okayama University of Science, 1-3 Ikoi-no-oka, Imabari 794-8555, Japan; (M.M.); (H.K.); (N.M.); (K.Y.)
| | - Yayoi Izu
- Department of Laboratory Animal Science, Faculty of Veterinary Medicine, Okayama University of Science, 1-3 Ikoi-no-oka, Imabari 794-8555, Japan;
| | - Noritaka Maeta
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Okayama University of Science, 1-3 Ikoi-no-oka, Imabari 794-8555, Japan; (M.M.); (H.K.); (N.M.); (K.Y.)
| | - Kazuaki Yamazoe
- Department of Veterinary Surgery, Faculty of Veterinary Medicine, Okayama University of Science, 1-3 Ikoi-no-oka, Imabari 794-8555, Japan; (M.M.); (H.K.); (N.M.); (K.Y.)
| |
Collapse
|
18
|
Roh EJ, Kim DS, Kim JH, Lim CS, Choi H, Kwon SY, Park SY, Kim JY, Kim HM, Hwang DY, Han DK, Han I. Multimodal therapy strategy based on a bioactive hydrogel for repair of spinal cord injury. Biomaterials 2023; 299:122160. [PMID: 37209541 DOI: 10.1016/j.biomaterials.2023.122160] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2022] [Revised: 03/13/2023] [Accepted: 05/10/2023] [Indexed: 05/22/2023]
Abstract
Traumatic spinal cord injury results in permanent and serious neurological impairment, but there is no effective treatment yet. Tissue engineering approaches offer great potential for the treatment of SCI, but spinal cord complexity poses great challenges. In this study, the composite scaffold consists of a hyaluronic acid-based hydrogel, decellularized brain matrix (DBM), and bioactive compounds such as polydeoxyribonucleotide (PDRN), tumor necrosis factor-α/interferon-γ primed mesenchymal stem cell-derived extracellular vesicles (TI-EVs), and human embryonic stem cell-derived neural progenitor cells (NPC). The composite scaffold showed significant effects on regenerative prosses including angiogenesis, anti-inflammation, anti-apoptosis, and neural differentiation. In addition, the composite scaffold (DBM/PDRN/TI-EV/NPC@Gel) induced an effective spinal cord regeneration in a rat spinal cord transection model. Therefore, this multimodal approach using an integrated bioactive scaffold coupled with biochemical cues from PDRN and TI-EVs could be used as an advanced tissue engineering platform for spinal cord regeneration.
Collapse
Affiliation(s)
- Eun Ji Roh
- Department of Neurosurgery CHA University School of Medicine, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea; Department of Biomedical Science CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - Da-Seul Kim
- Department of Biomedical Science CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea; School of Integrative Engineering Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Jun Hyuk Kim
- Department of Biomedical Science CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - Chang Su Lim
- Department of Neurosurgery CHA University School of Medicine, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - Hyemin Choi
- Department of Neurosurgery CHA University School of Medicine, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - Su Yeon Kwon
- Department of Neurosurgery CHA University School of Medicine, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - So-Yeon Park
- Department of Biomedical Science CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea; Division of Biotechnology College of Life Sciences and Biotechnology Korea University, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Jun Yong Kim
- Department of Biomedical Science CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - Hyun-Mun Kim
- Department of Biomedical Science CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - Dong-Youn Hwang
- Department of Biomedical Science CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea
| | - Dong Keun Han
- Department of Biomedical Science CHA University, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea.
| | - Inbo Han
- Department of Neurosurgery CHA University School of Medicine, 335 Pangyo-ro Bundang-gu, Seongnam-si, 13488, Republic of Korea.
| |
Collapse
|
19
|
Liu Y, Zhang W, Hu C, Zheng C, Zhang F, Yang L, Li Z, Wang Y. A composite hydrogel improves the survival and differentiation of human iPSC-derived neural stem cells after ischemic stroke. COMPOSITES PART B: ENGINEERING 2023; 259:110711. [DOI: 10.1016/j.compositesb.2023.110711] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
20
|
Yan M, Wang L, Wu Y, Wang L, Lu Y. Three-dimensional highly porous hydrogel scaffold for neural circuit dissection and modulation. Acta Biomater 2023; 157:252-262. [PMID: 36521677 DOI: 10.1016/j.actbio.2022.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2022] [Revised: 12/03/2022] [Accepted: 12/06/2022] [Indexed: 12/14/2022]
Abstract
Biomimetic brain structures and artificial neural networks have provided a simplified strategy for quantitatively investigating the complex structural and functional characteristics of highly interconnected neural networks. To achieve this, three-dimensional (3D) cell culture approaches have attracted much attention, which can mimic cell-cell interactions at the organism level and help better understand the function of specific neurons and neuronal networks than traditional two-dimensional cell culture methods. However, 3D scaffolds similar to the natural extracellular matrix to support the culturing, recording, and manipulation of neurons have long been an unresolved challenge. To resolve this, 3D hydrogel scaffolds can be fabricated via an innovative thermal treatment followed by an esterification process. A highly porous microstructure was formed within the bulk hydrogel scaffold, which showed a high porosity of 91% and a low Young's modulus of 6.11 kPa. Due to the merits of the fabricated hydrogel scaffolds, we constructed 3D neural networks and detected spontaneous action potentials in vitro. We successfully induced seizure-like waveforms in 3D cultured neurons and suppressed hyperactivated discharges by selectively activating γ-aminobutyric acid-ergic (GABAergic) interneurons. These results prove the advantages of our hydrogel scaffolds and demonstrate their application potential in the accurate dissection of neural circuits, which may help develop effective treatments for various neurological disorders. STATEMENT OF SIGNIFICANCE: While 3D cell culture approaches have attracted much attention and offer more advantages than two-dimensional cell culture methods, 3D scaffolds similar to the natural extracellular matrix to support the culturing, recording, and manipulation of neurons have long been an unresolved challenge. Herein, we developed a simplified and low-cost strategy for fabricating highly porous and cytocompatible hydrogel scaffolds for the construction of three-dimensional (3D) neural networks in vitro. The cultured 3D neural networks can mimic the in vivo connection among different neuron subgroups and help accurately dissect and manipulate the structure and function of specific neural circuits.
Collapse
Affiliation(s)
- Mengying Yan
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Lulu Wang
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Yiyong Wu
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China
| | - Liping Wang
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China.
| | - Yi Lu
- CAS Key Laboratory of Brain Connectome and Manipulation, the Brain Cognition and Brain Disease Institute, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, Shenzhen 518055, China.
| |
Collapse
|
21
|
Jeong E, Choi S, Cho SW. Recent Advances in Brain Organoid Technology for Human Brain Research. ACS APPLIED MATERIALS & INTERFACES 2023; 15:200-219. [PMID: 36468535 DOI: 10.1021/acsami.2c17467] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Brain organoids are self-assembled three-dimensional aggregates with brain-like cell types and structures and have emerged as new model systems that can be used to investigate human neurodevelopment and neurological disorders. However, brain organoids are not as mature and functional as real human brains due to limitations of the culture system with insufficient developmental patterning signals and a lack of components that are important for brain development and function, such as the non-neural population and vasculature. In addition, establishing the desired brain-like environment and monitoring the complex neural networks and physiological functions of the brain organoids remain challenging. The current protocols to generate brain organoids also have problems with heterogeneity and batch variation due to spontaneous self-organization of brain organoids into complex architectures of the brain. To address these limitations of current brain organoid technologies, various engineering platforms, such as extracellular matrices, fluidic devices, three-dimensional bioprinting, bioreactors, polymeric scaffolds, microelectrodes, and biochemical sensors, have been employed to improve neuronal development and maturation, reduce structural heterogeneity, and facilitate functional analysis and monitoring. In this review, we provide an overview of the latest engineering techniques that overcome these limitations in the production and application of brain organoids.
Collapse
Affiliation(s)
- Eunseon Jeong
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Suah Choi
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
22
|
Min S, Cho SW. Engineered human cardiac tissues for modeling heart diseases. BMB Rep 2023; 56:32-42. [PMID: 36443005 PMCID: PMC9887099 DOI: 10.5483/bmbrep.2022-0185] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/28/2022] [Accepted: 11/28/2022] [Indexed: 07/30/2023] Open
Abstract
Heart disease is one of the major life-threatening diseases with high mortality and incidence worldwide. Several model systems, such as primary cells and animals, have been used to understand heart diseases and establish appropriate treatments. However, they have limitations in accuracy and reproducibility in recapitulating disease pathophysiology and evaluating drug responses. In recent years, three-dimensional (3D) cardiac tissue models produced using tissue engineering technology and human cells have outperformed conventional models. In particular, the integration of cell reprogramming techniques with bioengineering platforms (e.g., microfluidics, scaffolds, bioprinting, and biophysical stimuli) has facilitated the development of heart-ona- chip, cardiac spheroid/organoid, and engineered heart tissue (EHT) to recapitulate the structural and functional features of the native human heart. These cardiac models have improved heart disease modeling and toxicological evaluation. In this review, we summarize the cell types for the fabrication of cardiac tissue models, introduce diverse 3D human cardiac tissue models, and discuss the strategies to enhance their complexity and maturity. Finally, recent studies in the modeling of various heart diseases are reviewed. [BMB Reports 2023; 56(1): 32-42].
Collapse
Affiliation(s)
- Sungjin Min
- Department of Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Korea
| |
Collapse
|
23
|
Min S, Cho SW. Engineered human cardiac tissues for modeling heart diseases. BMB Rep 2023; 56:32-42. [PMID: 36443005 PMCID: PMC9887099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2022] [Revised: 11/28/2022] [Accepted: 11/28/2022] [Indexed: 01/28/2023] Open
Abstract
Heart disease is one of the major life-threatening diseases with high mortality and incidence worldwide. Several model systems, such as primary cells and animals, have been used to understand heart diseases and establish appropriate treatments. However, they have limitations in accuracy and reproducibility in recapitulating disease pathophysiology and evaluating drug responses. In recent years, three-dimensional (3D) cardiac tissue models produced using tissue engineering technology and human cells have outperformed conventional models. In particular, the integration of cell reprogramming techniques with bioengineering platforms (e.g., microfluidics, scaffolds, bioprinting, and biophysical stimuli) has facilitated the development of heart-ona- chip, cardiac spheroid/organoid, and engineered heart tissue (EHT) to recapitulate the structural and functional features of the native human heart. These cardiac models have improved heart disease modeling and toxicological evaluation. In this review, we summarize the cell types for the fabrication of cardiac tissue models, introduce diverse 3D human cardiac tissue models, and discuss the strategies to enhance their complexity and maturity. Finally, recent studies in the modeling of various heart diseases are reviewed. [BMB Reports 2023; 56(1): 32-42].
Collapse
Affiliation(s)
- Sungjin Min
- Department of Biotechnology, Yonsei University, Seoul 03722, Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Korea
| |
Collapse
|
24
|
Prospects for hypoxia-based drug delivery platforms for the elimination of advanced metastatic tumors: From 3D modeling to clinical concepts. J Control Release 2023; 353:1002-1022. [PMID: 36516901 DOI: 10.1016/j.jconrel.2022.12.009] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Revised: 11/30/2022] [Accepted: 12/03/2022] [Indexed: 12/24/2022]
Abstract
Hypoxia is a unique characteristic of the solid tumor microenvironment. Hypoxia contributes to multi-drug resistance, metastasis and cancer relapse through numerous molecular pathways, but at the same time provides an opportunity for the development of novel drugs or modalities specifically targeting hypoxic tumor regions. Given the high significance of tumor hypoxia in therapeutic results, we here discuss a variety of hypoxia-adopted strategies, and their potential and utility in the treatment of deep-seated hypoxic tumor cells. We discuss the merits and demerits of these approaches, as well as their combination with other approaches such as photodynamic therapy. We also survey the currently available 3D hypoxia modeling systems, in particular organoid-based microfluidics. Finally, we discuss the potential and the current status of preclinical tumor hypoxia approaches in clinical trials for advanced cancer. We believe that multi-modal imaging and therapeutic hypoxia adopted drug delivery platforms could provide better efficacy and safety profiles, and more importantly personalized therapy. Determining the hypoxia status of tumors could offer a second chance for the clinical translation of hypoxia-based agents, such as hypoxia activated prodrugs (HAPs) from bench to bedside.
Collapse
|
25
|
Sellahewa SG, Li JY, Xiao Q. Updated Perspectives on Direct Vascular Cellular Reprogramming and Their Potential Applications in Tissue Engineered Vascular Grafts. J Funct Biomater 2022; 14:21. [PMID: 36662068 PMCID: PMC9866165 DOI: 10.3390/jfb14010021] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2022] [Revised: 12/25/2022] [Accepted: 12/27/2022] [Indexed: 01/03/2023] Open
Abstract
Cardiovascular disease is a globally prevalent disease with far-reaching medical and socio-economic consequences. Although improvements in treatment pathways and revascularisation therapies have slowed disease progression, contemporary management fails to modulate the underlying atherosclerotic process and sustainably replace damaged arterial tissue. Direct cellular reprogramming is a rapidly evolving and innovative tissue regenerative approach that holds promise to restore functional vasculature and restore blood perfusion. The approach utilises cell plasticity to directly convert somatic cells to another cell fate without a pluripotent stage. In this narrative literature review, we comprehensively analyse and compare direct reprogramming protocols to generate endothelial cells, vascular smooth muscle cells and vascular progenitors. Specifically, we carefully examine the reprogramming factors, their molecular mechanisms, conversion efficacies and therapeutic benefits for each induced vascular cell. Attention is given to the application of these novel approaches with tissue engineered vascular grafts as a therapeutic and disease-modelling platform for cardiovascular diseases. We conclude with a discussion on the ethics of direct reprogramming, its current challenges, and future perspectives.
Collapse
Affiliation(s)
- Saneth Gavishka Sellahewa
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Jojo Yijiao Li
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
| | - Qingzhong Xiao
- William Harvey Research Institute, Faculty of Medicine and Dentistry, Queen Mary University of London, London EC1M 6BQ, UK
- Key Laboratory of Cardiovascular Diseases, School of Basic Medical Sciences, Guangzhou Institute of Cardiovascular Disease, The Second Affiliated Hospital, Guangzhou Medical University, Guangzhou 511436, China
| |
Collapse
|
26
|
Jin Y, Kim H, Min S, Choi YS, Seo SJ, Jeong E, Kim SK, Lee HA, Jo SH, Park JH, Park BW, Sim WS, Kim JJ, Ban K, Kim YG, Park HJ, Cho SW. Three-dimensional heart extracellular matrix enhances chemically induced direct cardiac reprogramming. SCIENCE ADVANCES 2022; 8:eabn5768. [PMID: 36516259 PMCID: PMC9750148 DOI: 10.1126/sciadv.abn5768] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 11/12/2022] [Indexed: 06/17/2023]
Abstract
Direct cardiac reprogramming has emerged as a promising therapeutic approach for cardiac regeneration. Full chemical reprogramming with small molecules to generate cardiomyocytes may be more amenable than genetic reprogramming for clinical applications as it avoids safety concerns associated with genetic manipulations. However, challenges remain regarding low conversion efficiency and incomplete cardiomyocyte maturation. Furthermore, the therapeutic potential of chemically induced cardiomyocytes (CiCMs) has not been investigated. Here, we report that a three-dimensional microenvironment reconstituted with decellularized heart extracellular matrix can enhance chemical reprogramming and cardiac maturation of fibroblasts to cardiomyocytes. The resultant CiCMs exhibit elevated cardiac marker expression, sarcomeric organization, and improved electrophysiological features and drug responses. We investigated the therapeutic potential of CiCMs reprogrammed in three-dimensional heart extracellular matrix in a rat model of myocardial infarction. Our platform can facilitate the use of CiCMs for regenerative medicine, disease modeling, and drug screening.
Collapse
Affiliation(s)
- Yoonhee Jin
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Hyeok Kim
- Department of Biomedicine and Health Sciences, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sungjin Min
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Yi Sun Choi
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Seung Ju Seo
- Department of Physiology, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Eunseon Jeong
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Su Kyeom Kim
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Hyang-Ae Lee
- Korea Institute of Toxicology, Korea Research Institute of Chemical Technology, Daejeon 34114, Republic of Korea
| | - Sung-Hyun Jo
- Department of Chemical Engineering, Soongsil University, Seoul 06978, Republic of Korea
| | - Jae-Hyun Park
- Department of Biomedicine and Health Sciences, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Bong-Woo Park
- Department of Biomedicine and Health Sciences, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Woo-Sup Sim
- Department of Biomedicine and Health Sciences, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Jin-Ju Kim
- Department of Biomedicine and Health Sciences, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Kiwon Ban
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon 999077, Hong Kong
| | - Yun-Gon Kim
- Department of Chemical Engineering, Soongsil University, Seoul 06978, Republic of Korea
| | - Hun-Jun Park
- Department of Biomedicine and Health Sciences, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The Catholic University of Korea, Seoul 06591, Republic of Korea
- Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
27
|
Caballero D, Reis RL, Kundu SC. Boosting the Clinical Translation of Organ-on-a-Chip Technology. Bioengineering (Basel) 2022; 9:549. [PMID: 36290517 PMCID: PMC9598310 DOI: 10.3390/bioengineering9100549] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2022] [Revised: 10/02/2022] [Accepted: 10/11/2022] [Indexed: 11/05/2022] Open
Abstract
Organ-on-a-chip devices have become a viable option for investigating critical physiological events and responses; this technology has matured substantially, and many systems have been reported for disease modeling or drug screening over the last decade. Despite the wide acceptance in the academic community, their adoption by clinical end-users is still a non-accomplished promise. The reasons behind this difficulty can be very diverse but most likely are related to the lack of predictive power, physiological relevance, and reliability necessary for being utilized in the clinical area. In this Perspective, we briefly discuss the main attributes of organ-on-a-chip platforms in academia and how these characteristics impede their easy translation to the clinic. We also discuss how academia, in conjunction with the industry, can contribute to boosting their adoption by proposing novel design concepts, fabrication methods, processes, and manufacturing materials, improving their standardization and versatility, and simplifying their manipulation and reusability.
Collapse
Affiliation(s)
- David Caballero
- 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, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4704-553 Braga, Portugal
| | - 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, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4704-553 Braga, Portugal
| | - Subhas C. Kundu
- 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, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Portugal
- ICVS/3B’s—PT Government Associate Laboratory, 4704-553 Braga, Portugal
| |
Collapse
|
28
|
Habibey R, Rojo Arias JE, Striebel J, Busskamp V. Microfluidics for Neuronal Cell and Circuit Engineering. Chem Rev 2022; 122:14842-14880. [PMID: 36070858 PMCID: PMC9523714 DOI: 10.1021/acs.chemrev.2c00212] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Indexed: 02/07/2023]
Abstract
The widespread adoption of microfluidic devices among the neuroscience and neurobiology communities has enabled addressing a broad range of questions at the molecular, cellular, circuit, and system levels. Here, we review biomedical engineering approaches that harness the power of microfluidics for bottom-up generation of neuronal cell types and for the assembly and analysis of neural circuits. Microfluidics-based approaches are instrumental to generate the knowledge necessary for the derivation of diverse neuronal cell types from human pluripotent stem cells, as they enable the isolation and subsequent examination of individual neurons of interest. Moreover, microfluidic devices allow to engineer neural circuits with specific orientations and directionality by providing control over neuronal cell polarity and permitting the isolation of axons in individual microchannels. Similarly, the use of microfluidic chips enables the construction not only of 2D but also of 3D brain, retinal, and peripheral nervous system model circuits. Such brain-on-a-chip and organoid-on-a-chip technologies are promising platforms for studying these organs as they closely recapitulate some aspects of in vivo biological processes. Microfluidic 3D neuronal models, together with 2D in vitro systems, are widely used in many applications ranging from drug development and toxicology studies to neurological disease modeling and personalized medicine. Altogether, microfluidics provide researchers with powerful systems that complement and partially replace animal models.
Collapse
Affiliation(s)
- Rouhollah Habibey
- Department
of Ophthalmology, Universitäts-Augenklinik
Bonn, University of Bonn, Ernst-Abbe-Straße 2, D-53127 Bonn, Germany
| | - Jesús Eduardo Rojo Arias
- Wellcome—MRC
Cambridge Stem Cell Institute, Jeffrey Cheah Biomedical Centre, Cambridge
Biomedical Campus, University of Cambridge, Cambridge CB2 0AW, United Kingdom
| | - Johannes Striebel
- Department
of Ophthalmology, Universitäts-Augenklinik
Bonn, University of Bonn, Ernst-Abbe-Straße 2, D-53127 Bonn, Germany
| | - Volker Busskamp
- Department
of Ophthalmology, Universitäts-Augenklinik
Bonn, University of Bonn, Ernst-Abbe-Straße 2, D-53127 Bonn, Germany
| |
Collapse
|
29
|
Ko MJ, Hong H, Choi H, Kang H, Kim D. Multifunctional Magnetic Nanoparticles for Dynamic Imaging and Therapy. ADVANCED NANOBIOMED RESEARCH 2022. [DOI: 10.1002/anbr.202200053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022] Open
Affiliation(s)
- Min Jun Ko
- Department of Radiology Feinberg School of Medicine Northwestern University Chicago IL 60611 USA
| | - Hyunsik Hong
- Department of Materials Science and Engineering Korea University Seoul 02841 Republic of Korea
| | - Hyunjun Choi
- Department of Radiology Feinberg School of Medicine Northwestern University Chicago IL 60611 USA
- Department of Bioengineering University of Illinois at Chicago Chicago IL 60607 USA
| | - Heemin Kang
- Department of Materials Science and Engineering Korea University Seoul 02841 Republic of Korea
- College of Medicine Korea University Seoul 02841 Republic of Korea
| | - Dong‐Hyun Kim
- Department of Radiology Feinberg School of Medicine Northwestern University Chicago IL 60611 USA
- Department of Bioengineering University of Illinois at Chicago Chicago IL 60607 USA
- Department of Biomedical Engineering McCormick School of Engineering Northwestern University Evanston IL 60208 USA
- Robert H. Lurie Comprehensive Cancer Center Northwestern University Chicago Illinois 60611 USA
| |
Collapse
|
30
|
Trinh VQH, Roland JT, Wong J, Revetta F, Patel K, Shi C, DelGiorno KE, Carter BD, Tan MCB. Peak density of immature nerve cells occurs with high-grade dysplasia in intraductal papillary mucinous neoplasms of the pancreas. J Pathol 2022; 258:69-82. [PMID: 35686747 PMCID: PMC9378585 DOI: 10.1002/path.5978] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 05/10/2022] [Accepted: 06/07/2022] [Indexed: 11/11/2022]
Abstract
The development of neural structures within tumors is now considered vital for carcinogenesis. However, the time course of this development in human pre-invasive neoplasia has been incompletely described. Therefore, we performed a detailed analysis of nerves across the neoplastic spectrum in resected intraductal papillary mucinous neoplasms (IPMNs) of the pancreas. Histology and multiplexed immunochemistry demonstrated that nerve density increased from low-grade (LG) to high-grade dysplasia (HG) but did not further increase once invasive IPMN (INV IPMN) was present. Higher nerve density correlated with increasing expression of nerve growth factor (NGF) by the tumor cells. Intra-tumoral nerves were immature and lacked markers of sympathetic, parasympathetic, and sensory lineages. Here, we show for the first time the presence of neural precursor cells (NPCs) within the stroma of pancreatic tumors. The density of these doublecortin (DCX)-positive NPCs increased from LG to HG, but not from HG to INV IPMN. We conclude that peak neural density of tumors is reached in high-grade dysplasia (often termed carcinoma in situ) rather than after invasion. These findings suggest that nerve-tumor interactions are important in IPMN progression and may serve as the basis for future mechanistic studies and novel therapeutic modalities. © 2022 The Pathological Society of Great Britain and Ireland.
Collapse
Affiliation(s)
- Vincent Quoc-Huy Trinh
- Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Joseph Thomas Roland
- Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, TN, USA
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Jahg Wong
- Département de pathologie et biologie cellulaire, Université de Montréal, Montréal, Québec, Canada
| | - Frank Revetta
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Krutika Patel
- Department of Pathology, Microbiology and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Chanjuan Shi
- Department of Pathology, Duke University School of Medicine, Durham, NC, USA
| | - Kathleen E. DelGiorno
- Epithelial Biology Center, Vanderbilt University School of Medicine, Nashville, TN, USA
- Cell and Developmental Biology, Vanderbilt University, Nashville TN, USA
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bruce D. Carter
- Department of Biochemistry, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Marcus Chuan Beng Tan
- Section of Surgical Sciences, Vanderbilt University School of Medicine, Nashville, TN, USA
- Division of Surgical Oncology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Ingram Cancer Center, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
31
|
Xu Z, Li Y, Li P, Sun Y, Lv S, Wang Y, He X, Xu J, Xu Z, Li L, Li Y. Soft substrates promote direct chemical reprogramming of fibroblasts into neurons. Acta Biomater 2022; 152:255-272. [PMID: 36041647 DOI: 10.1016/j.actbio.2022.08.049] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Revised: 08/13/2022] [Accepted: 08/23/2022] [Indexed: 11/01/2022]
Abstract
Fibroblasts can be directly reprogrammed via a combination of small molecules to generate induced neurons (iNs), bypassing intermediate stages. This method holds great promise for regenerative medicine; however, it remains inefficient. Recently, studies have suggested that physical cues may improve the direct reprogramming of fibroblasts into neurons, but the underlying mechanisms remain to be further explored, and the physical factors reported to date do not exhibit the full properties of the extracellular matrix (ECM). Previous in vitro studies mainly used rigid polystyrene dishes, while one of the characteristics of the native in-vivo environment of neurons is the soft nature of brain ECM. The reported stiffness of brain tissue is very soft ranging between 100 Pa and 3 kPa, and the effect of substrate stiffness on direct neuronal reprogramming has not been explored. Here, we show for the first time that soft substrates substantially improved the production efficiency and quality of iNs, without needing to co-culture with glial cells during reprogramming, producing more glutamatergic neurons with electrophysiological functions in a shorter time. Transcriptome sequencing indicated that soft substrates might promote glutamatergic neuron reprogramming through integrins, actin cytoskeleton, Hippo signalling pathway, and regulation of mesenchymal-to-epithelial transition, and competing endogenous RNA network analysis provided new targets for neuronal reprogramming. We demonstrated that soft substrates may promote neuronal reprogramming by inhibiting microRNA-615-3p-targeting integrin subunit beta 4. Our findings can aid the development of regenerative therapies and help improve our understanding of neuronal reprogramming. STATEMENT OF SIGNIFICANCE: : First, we have shown that low stiffness promotes direct reprogramming on the basis of small molecule combinations. To the best of our knowledge, this is the first report on this type of method, which may greatly promote the progress of neural reprogramming. Second, we found that miR-615-3p may interact with ITGB4, and the soft substrates may promote neural reprogramming by inhibiting microRNA (miR)-615-3p targeting integrin subunit beta 4 (ITGB4). We are the first to report on this mechanism. Our findings will provide more functional neurons for subsequent basic and clinical research in neurological regenerative medicine, and will help to improve the overall understanding of neural reprogramming. This work also provides new ideas for the design of medical biomaterials for nerve regeneration.
Collapse
Affiliation(s)
- Ziran Xu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Yan Li
- Division of Orthopedics and Biotechnology, Department for Clinical Intervention and Technology (CLINTEC), Karolinska Institute, Stockholm, Sweden.
| | - Pengdong Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; The Sixth Affiliated Hospital of Guangzhou Medical University, Qingyuan People's Hospital, Qingyuan 511518, Guangdong, China.
| | - Yingying Sun
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; Department of Stomatology, The First Hospital of Jilin University, Changchun 130021, China.
| | - Shuang Lv
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Yin Wang
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Xia He
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; Department of Pathology, Shanxi Bethune Hospital, Taiyuan 030032, China.
| | - Jinying Xu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China; Department of Burns Surgery, The First Hospital of Jilin University, Changchun 130000, China.
| | - Zhixiang Xu
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Lisha Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| | - Yulin Li
- The Key Laboratory of Pathobiology, Ministry of Education, College of Basic Medical Sciences, Jilin University, Changchun 130021, China.
| |
Collapse
|
32
|
Elkhoury K, Chen M, Koçak P, Enciso-Martínez E, Bassous NJ, Lee MC, Byambaa B, Rezaei Z, Li Y, Urbina M, Gurian M, Sobahi N, Hussain MA, Sanchez-Gonzalez L, Leijten J, Hassan S, Arab-Tehrany E, Ward JE, Shin SR. Hybrid extracellular vesicles-liposome incorporated advanced bioink to deliver microRNA. Biofabrication 2022; 14:10.1088/1758-5090/ac8621. [PMID: 35917808 PMCID: PMC9594995 DOI: 10.1088/1758-5090/ac8621] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2022] [Accepted: 08/02/2022] [Indexed: 11/12/2022]
Abstract
In additive manufacturing, bioink formulations govern strategies to engineer 3D living tissues that mimic the complex architectures and functions of native tissues for successful tissue regeneration. Conventional 3D-printed tissues are limited in their ability to alter the fate of laden cells. Specifically, the efficient delivery of gene expression regulators (i.e. microRNAs (miRNAs)) to cells in bioprinted tissues has remained largely elusive. In this study, we explored the inclusion of extracellular vesicles (EVs), naturally occurring nanovesicles (NVs), into bioinks to resolve this challenge. EVs show excellent biocompatibility, rapid endocytosis, and low immunogenicity, which lead to the efficient delivery of miRNAs without measurable cytotoxicity. EVs were fused with liposomes to prolong and control their release by altering their physical interaction with the bioink. Hybrid EVs-liposome (hEL) NVs were embedded in gelatin-based hydrogels to create bioinks that could efficiently encapsulate and deliver miRNAs at the target site in a controlled and sustained manner. The regulation of cells' gene expression in a 3D bioprinted matrix was achieved using the hELs-laden bioink as a precursor for excellent shape fidelity and high cell viability constructs. Novel regulatory factors-loaded bioinks will expedite the translation of new bioprinting applications in the tissue engineering field.
Collapse
Affiliation(s)
- Kamil Elkhoury
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA, 02139 USA
- LIBio, Université de Lorraine, F-54000 Nancy, France
- These authors contributed equally to this work
| | - Mo Chen
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA, 02139 USA
- Department of Gynecology, Obstetrics & Gynecology Hospital, Fudan University, Shanghai 200011, China
- These authors contributed equally to this work
| | - Polen Koçak
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA, 02139 USA
- Department of Biomedical Engineering, Faculty of Engineering, İstinye University, 34396 Sariyer/Istanbul, Trukey
| | - Eduardo Enciso-Martínez
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA, 02139 USA
| | - Nicole Joy Bassous
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA, 02139 USA
| | - Myung Chul Lee
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA, 02139 USA
| | | | - Zahra Rezaei
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA, 02139 USA
| | - Yang Li
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA, 02139 USA
| | - Mariely Urbina
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA, 02139 USA
| | - Melvin Gurian
- Department of Developmental BioEngineering, University of Twente, Enschede, Overijssel 7522 NB, The Netherlands
| | - Nebras Sobahi
- Department of Electrical and Computer Engineering, King Abdulaziz University, Jeddah 21569, Saudi Arabia
| | - Mohammad Asif Hussain
- Department of Electrical and Computer Engineering, King Abdulaziz University, Jeddah 21569, Saudi Arabia
| | | | - Jeroen Leijten
- Department of Developmental BioEngineering, University of Twente, Enschede, Overijssel 7522 NB, The Netherlands
| | - Shabir Hassan
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA, 02139 USA
- Department of Biology, Khalifa University, 127788, Abu Dhabi, UAE Division of Genetics
| | | | - Jennifer Ellis Ward
- Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Boston, MA, 02115 USA
| | - Su Ryon Shin
- Division of Engineering in Medicine, Department of Medicine, Harvard Medical School, Brigham and Women’s Hospital, Cambridge, MA, 02139 USA
| |
Collapse
|
33
|
Jagiełło A, Castillo U, Botvinick E. Cell mediated remodeling of stiffness matched collagen and fibrin scaffolds. Sci Rep 2022; 12:11736. [PMID: 35817812 PMCID: PMC9273755 DOI: 10.1038/s41598-022-14953-w] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2022] [Accepted: 06/15/2022] [Indexed: 02/07/2023] Open
Abstract
Cells are known to continuously remodel their local extracellular matrix (ECM) and in a reciprocal way, they can also respond to mechanical and biochemical properties of their fibrous environment. In this study, we measured how stiffness around dermal fibroblasts (DFs) and human fibrosarcoma HT1080 cells differs with concentration of rat tail type 1 collagen (T1C) and type of ECM. Peri-cellular stiffness was probed in four directions using multi-axes optical tweezers active microrheology (AMR). First, we found that neither cell type significantly altered local stiffness landscape at different concentrations of T1C. Next, rat tail T1C, bovine skin T1C and fibrin cell-free hydrogels were polymerized at concentrations formulated to match median stiffness value. Each of these hydrogels exhibited distinct fiber architecture. Stiffness landscape and fibronectin secretion, but not nuclear/cytoplasmic YAP ratio differed with ECM type. Further, cell response to Y27632 or BB94 treatments, inhibiting cell contractility and activity of matrix metalloproteinases, respectively, was also dependent on ECM type. Given differential effect of tested ECMs on peri-cellular stiffness landscape, treatment effect and cell properties, this study underscores the need for peri-cellular and not bulk stiffness measurements in studies on cellular mechanotransduction.
Collapse
Affiliation(s)
- Alicja Jagiełło
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697-2715, USA
| | - Ulysses Castillo
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697-2715, USA
| | - Elliot Botvinick
- Department of Biomedical Engineering, University of California, Irvine, CA, 92697-2715, USA.
- Beckman Laser Institute and Medical Clinic, University of California, Irvine, CA, 92612, USA.
- Department of Surgery, University of California Irvine, 333 City Boulevard, Suite 700, Orange, CA, 92868, USA.
- The Edwards Lifesciences Foundation Cardiovascular Innovation and Research Center, University of California, Irvine, CA, 92697-2730, USA.
| |
Collapse
|
34
|
Yang L, Conley BM, Rathnam C, Cho HY, Pongkulapa T, Conklin B, Lee KB. Predictive Biophysical Cue Mapping for Direct Cell Reprogramming Using Combinatorial Nanoarrays. ACS NANO 2022; 16:5577-5586. [PMID: 35301847 DOI: 10.1021/acsnano.1c10344] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Biophysical cues, such as nanotopographies of extracellular matrix (ECM), are key cell regulators for direct cell reprogramming. Therefore, high-throughput methods capable of systematically screening a wide range of biophysical cue-regulated cell reprogramming are increasingly needed for tissue engineering and regenerative medicine. Here, we report the development of a dynamic laser interference lithography (DIL) to generate large-scale combinatorial biophysical cue (CBC) arrays with diverse micro/nanostructures at higher complexities than most current arrays. Using CBC arrays, a high-throughput cell mapping method is further demonstrated for the systematic investigation of biophysical cue-mediated direct cell reprogramming. This CBC array-based high-throughput cell screening approach facilitates the rapid identification of unconventional hierarchical nanopatterns that induce the direct reprogramming of human fibroblasts into neurons through epigenetic modulation mechanisms. In this way, we successfully demonstrate DIL for generating highly complex CBC arrays and establish CBC array-based cell screening as a valuable strategy for systematically investigating the role of biophysical cues in cell reprogramming.
Collapse
Affiliation(s)
- Letao Yang
- Department of Chemistry and Chemical Biology, Rutgers University, the State University of New Jersey, 123 Bevier Road, Piscataway, New Jersey 08854, United States
| | - Brian M Conley
- Department of Chemistry and Chemical Biology, Rutgers University, the State University of New Jersey, 123 Bevier Road, Piscataway, New Jersey 08854, United States
| | - Christopher Rathnam
- Department of Chemistry and Chemical Biology, Rutgers University, the State University of New Jersey, 123 Bevier Road, Piscataway, New Jersey 08854, United States
| | - Hyeon-Yeol Cho
- Department of Chemistry and Chemical Biology, Rutgers University, the State University of New Jersey, 123 Bevier Road, Piscataway, New Jersey 08854, United States
| | - Thanapat Pongkulapa
- Department of Chemistry and Chemical Biology, Rutgers University, the State University of New Jersey, 123 Bevier Road, Piscataway, New Jersey 08854, United States
| | - Brandon Conklin
- Department of Chemistry and Chemical Biology, Rutgers University, the State University of New Jersey, 123 Bevier Road, Piscataway, New Jersey 08854, United States
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers University, the State University of New Jersey, 123 Bevier Road, Piscataway, New Jersey 08854, United States
| |
Collapse
|
35
|
Tissue extracellular matrix hydrogels as alternatives to Matrigel for culturing gastrointestinal organoids. Nat Commun 2022; 13:1692. [PMID: 35354790 PMCID: PMC8967832 DOI: 10.1038/s41467-022-29279-4] [Citation(s) in RCA: 174] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2021] [Accepted: 03/04/2022] [Indexed: 12/16/2022] Open
Abstract
Matrigel, a mouse tumor extracellular matrix protein mixture, is an indispensable component of most organoid tissue culture. However, it has limited the utility of organoids for drug development and regenerative medicine due to its tumor-derived origin, batch-to-batch variation, high cost, and safety issues. Here, we demonstrate that gastrointestinal tissue-derived extracellular matrix hydrogels are suitable substitutes for Matrigel in gastrointestinal organoid culture. We found that the development and function of gastric or intestinal organoids grown in tissue extracellular matrix hydrogels are comparable or often superior to those in Matrigel. In addition, gastrointestinal extracellular matrix hydrogels enabled long-term subculture and transplantation of organoids by providing gastrointestinal tissue-mimetic microenvironments. Tissue-specific and age-related extracellular matrix profiles that affect organoid development were also elucidated through proteomic analysis. Together, our results suggest that extracellular matrix hydrogels derived from decellularized gastrointestinal tissues are effective alternatives to the current gold standard, Matrigel, and produce organoids suitable for gastrointestinal disease modeling, drug development, and tissue regeneration.
Collapse
|
36
|
Intestinal extracellular matrix hydrogels to generate intestinal organoids for translational applications. J IND ENG CHEM 2022. [DOI: 10.1016/j.jiec.2021.11.044] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
37
|
Decellularised extracellular matrix-based biomaterials for repair and regeneration of central nervous system. Expert Rev Mol Med 2022; 23:e25. [PMID: 34994341 PMCID: PMC9884794 DOI: 10.1017/erm.2021.22] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
The central nervous system (CNS), consisting of the brain and spinal cord, regulates the mind and functions of the organs. CNS diseases, leading to changes in neurological functions in corresponding sites and causing long-term disability, represent one of the major public health issues with significant clinical and economic burdens worldwide. In particular, the abnormal changes in the extracellular matrix under various disease conditions have been demonstrated as one of the main factors that can alter normal cell function and reduce the neuroregeneration potential in damaged tissue. Decellularised extracellular matrix (dECM)-based biomaterials have been recently utilised for CNS applications, closely mimicking the native tissue. dECM retains tissue-specific components, including proteoglycan as well as structural and functional proteins. Due to their unique composition, these biomaterials can stimulate sensitive repair mechanisms associated with CNS damages. Herein, we discuss the decellularisation of the brain and spinal cord as well as recellularisation of acellular matrix and the recent progress in the utilisation of brain and spinal cord dECM.
Collapse
|
38
|
Hsu Y, Huang K, Cheng K. Resuscitating the Field of Cardiac Regeneration: Seeking Answers from Basic Biology. Adv Biol (Weinh) 2021; 6:e2101133. [PMID: 34939372 DOI: 10.1002/adbi.202101133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 12/02/2021] [Indexed: 11/09/2022]
Abstract
Heart failure (HF) is one of the leading causes for hospital admissions worldwide. HF patients are classified based on the chronic changes in left ventricular ejection fraction (LVEF) as preserved (LVEF ≥ 50%), reduced (LVEF ≤ 40%), or mid-ranged (40% < LVEF < 50%) HFs. Treatments nowadays can prevent HFrEF progress, whereas only a few of the treatments have been proven to be effective in improving the survival of HFpEF. In this review, numerous mediators involved in the pathogenesis of HF are summarized. The regional upstream signaling and their diagnostic and therapeutic potential are also discussed. Additionally, the recent challenges and development in cardiac regenerative therapy that hold opportunities for future research and clinical translation are discussed.
Collapse
Affiliation(s)
- Yaching Hsu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, NC, 27607, USA
| | - Ke Huang
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, NC, 27607, USA
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, NC, 27607, USA.,Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill & North Carolina State University, Raleigh, NC, 27607, USA
| |
Collapse
|
39
|
Josan C, Kakar S, Raha S. Matrigel® enhances 3T3-L1 cell differentiation. Adipocyte 2021; 10:361-377. [PMID: 34288778 PMCID: PMC8296963 DOI: 10.1080/21623945.2021.1951985] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Revised: 06/28/2021] [Accepted: 07/01/2021] [Indexed: 12/24/2022] Open
Abstract
Culturing cells on bio-gels are believed to provide a more in vivo-like extracellular matrix. 3T3-L1 cells cultured on Matrigel® significantly alteregd their proliferation and differentiation as compared to growth on tissue culture-coated polystyrene surfaces. Growth on a 250-μm thick layer of Matrigel® facilitated the formation of cellular aggregates of 3T3-L1 cells. Differentiation of 3T3-L1 cells cultured on Matrigel® demonstrated increased levels of mRNA levels for key adipogenic transcription factors (PPARγ, C/EBPα, SREBP1), lipogenic markers (FAS, FABP4, LPL, PLIN1) and markers of adipocyte maturity (LEP), compared to cells cultured directly on a polystyrene tissue culture surface. The gene expression of extracellular matrix proteins (FN1, COL1A1, COL4A1, COL6, LAM) was decreased in 3T3-L1 cells cultured on Matrigel®. Furthermore, growth on Matrigel® increased lipid accumulation in 3T3-L1 cells in the presence and absence of rosiglitazone, a thiazolidinedione routinely used to optimize differentiation in these cells. These changes in adipocyte gene expression and lipid accumulation patterns may be a result of the increased cell-cell and cell-ECM interactions occurring on the Matrigel®, a scenario that is more reflective of an in vivo model. Taken together, our data advance the understanding of the value of culturing 3T3-L1 cells on Matrigel®.
Collapse
Affiliation(s)
- Chitmandeep Josan
- Department of Pediatrics and the Graduate Program in Medical Sciences, McMaster University, Hamilton, ON, Canada
| | - Sachin Kakar
- Department of Pediatrics, McMaster University, Hamilton, ON, Canada
| | - Sandeep Raha
- Department of Pediatrics and the Graduate Program in Medical Sciences, McMaster University, Hamilton, ON, Canada
- Department of Pediatrics, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
40
|
Paoletti C, Chiono V. Bioengineering Methods in MicroRNA-Mediated Direct Reprogramming of Fibroblasts Into Cardiomyocytes. Front Cardiovasc Med 2021; 8:750438. [PMID: 34760946 PMCID: PMC8573325 DOI: 10.3389/fcvm.2021.750438] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2021] [Accepted: 09/13/2021] [Indexed: 12/29/2022] Open
Abstract
Ischemic heart disease is the major cause of mortality worldwide. Despite the most recent pharmacological progresses, cardiac regeneration is yet not possible, and heart transplantation is the only therapeutic option for end-stage heart failure. Traditional cardiac regenerative medicine approaches, such as cell therapies and tissue engineering, have failed in the obtainment of human functional cardiac tissue, mainly due to unavailability of high quantities of autologous functional cardiomyocytes (CMs), low grafting efficiency, and/or arrhythmic events. Direct reprogramming (DR) of fibroblasts into induced CMs (iCMs) has emerged as a new promising approach for myocardial regeneration by in situ transdifferentiation or providing additional CM source for cell therapy. Among available DR methods, non-viral transfection with microRNAs (miRcombo: miR-1, miR-133, miR-208, and miR-499) appears promising for future clinical translation. MiRcombo transfection of fibroblasts could be significantly improved by the development of safe nanocarriers, efficiently delivering their cargo to target cells at the required stoichiometric ratio and overall dose in due times. Newly designed in vitro 3D culture microenvironments, providing biomimetic biophysical and biochemical stimuli to miRcombo-transfected cells, significantly increase the yield of fibroblast transdifferentiation into iCMs, enhancing CM gene expression. Epigenetic regulation of gene expression programs, critical to cell lineage commitment, can also be promoted by the administration of specific anti-inflammatory and anti-fibrotic soluble factors, helping in suppressing fibroblast signature. The aim of this mini-review is to introduce the readers to a relatively unknown field of cardiac research integrating bioengineering tools as relevant for the progress of miRNA-mediated cardiac DR.
Collapse
Affiliation(s)
- Camilla Paoletti
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| | - Valeria Chiono
- Department of Mechanical and Aerospace Engineering, Politecnico di Torino, Turin, Italy
| |
Collapse
|
41
|
Kort-Mascort J, Bao G, Elkashty O, Flores-Torres S, Munguia-Lopez JG, Jiang T, Ehrlicher AJ, Mongeau L, Tran SD, Kinsella JM. Decellularized Extracellular Matrix Composite Hydrogel Bioinks for the Development of 3D Bioprinted Head and Neck in Vitro Tumor Models. ACS Biomater Sci Eng 2021; 7:5288-5300. [PMID: 34661396 DOI: 10.1021/acsbiomaterials.1c00812] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023]
Abstract
Reinforced extracellular matrix (ECM)-based hydrogels recapitulate several mechanical and biochemical features found in the tumor microenvironment (TME) in vivo. While these gels retain several critical structural and bioactive molecules that promote cell-matrix interactivity, their mechanical properties tend toward the viscous regime limiting their ability to retain ordered structural characteristics when considered as architectured scaffolds. To overcome this limitation characteristic of pure ECM hydrogels, we present a composite material containing alginate, a seaweed-derived polysaccharide, and gelatin, denatured collagen, as rheological modifiers which impart mechanical integrity to the biologically active decellularized ECM (dECM). After an optimization process, the reinforced gel proposed is mechanically stable and bioprintable and has a stiffness within the expected physiological values. Our hydrogel's elastic modulus has no significant difference when compared to tumors induced in preclinical xenograft head and neck squamous cell carcinoma (HNSCC) mouse models. The bioprinted cell-laden model is highly reproducible and allows proliferation and reorganization of HNSCC cells while maintaining cell viability above 90% for periods of nearly 3 weeks. Cells encapsulated in our bioink produce spheroids of at least 3000 μm2 of cross-sectional area by day 15 of culture and are positive for cytokeratin in immunofluorescence quantification, a common marker of HNSCC model validation in 2D and 3D models. We use this in vitro model system to evaluate the standard-of-care small molecule therapeutics used to treat HNSCC clinically and report a 4-fold increase in the IC50 of cisplatin and an 80-fold increase for 5-fluorouracil compared to monolayer cultures. Our work suggests that fabricating in vitro models using reinforced dECM provides a physiologically relevant system to evaluate malignant neoplastic phenomena in vitro due to the physical and biological features replicated from the source tissue microenvironment.
Collapse
Affiliation(s)
- Jacqueline Kort-Mascort
- Department of Bioengineering, McGill University, McConnell Engineering Building, 3480 University, Room 350, Montreal, Quebec H3A 0E9, Canada
| | - Guangyu Bao
- Department of Mechanical Engineering, McGill University, Macdonald Engineering Building, Room 270, 817 Sherbrooke Street West, Montreal, Quebec H3A 0C3, Canada
| | - Osama Elkashty
- Faculty of Dentistry, McGill University, 3640 rue University, Montreal, Quebec H3A 0C7, Canada.,Oral Pathology Department, Faculty of Dentistry, Mansoura University, Mansoura 29R6+Q3F, Egypt
| | - Salvador Flores-Torres
- Department of Bioengineering, McGill University, McConnell Engineering Building, 3480 University, Room 350, Montreal, Quebec H3A 0E9, Canada
| | - Jose G Munguia-Lopez
- Department of Bioengineering, McGill University, McConnell Engineering Building, 3480 University, Room 350, Montreal, Quebec H3A 0E9, Canada.,Faculty of Dentistry, McGill University, 3640 rue University, Montreal, Quebec H3A 0C7, Canada
| | - Tao Jiang
- Department of Intelligent Machinery and Instrument, College of Intelligence Science and Technology, National University of Defense Technology Changsha, No. 109 Deya Road, Kaifu District, Changsha, Hunan 410073, China
| | - Allen J Ehrlicher
- Department of Bioengineering, McGill University, McConnell Engineering Building, 3480 University, Room 350, Montreal, Quebec H3A 0E9, Canada.,Department of Mechanical Engineering, McGill University, Macdonald Engineering Building, Room 270, 817 Sherbrooke Street West, Montreal, Quebec H3A 0C3, Canada
| | - Luc Mongeau
- Department of Mechanical Engineering, McGill University, Macdonald Engineering Building, Room 270, 817 Sherbrooke Street West, Montreal, Quebec H3A 0C3, Canada
| | - Simon D Tran
- Faculty of Dentistry, McGill University, 3640 rue University, Montreal, Quebec H3A 0C7, Canada
| | - Joseph M Kinsella
- Department of Bioengineering, McGill University, McConnell Engineering Building, 3480 University, Room 350, Montreal, Quebec H3A 0E9, Canada
| |
Collapse
|
42
|
Wu C, Chin CSM, Huang Q, Chan HY, Yu X, Roy VAL, Li WJ. Rapid nanomolding of nanotopography on flexible substrates to control muscle cell growth with enhanced maturation. MICROSYSTEMS & NANOENGINEERING 2021; 7:89. [PMID: 34754504 PMCID: PMC8571286 DOI: 10.1038/s41378-021-00316-4] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/23/2021] [Revised: 08/20/2021] [Accepted: 09/13/2021] [Indexed: 05/11/2023]
Abstract
In vivo, multiple biophysical cues provided by highly ordered connective tissues of the extracellular matrix regulate skeletal muscle cells to align in parallel with one another. However, in routine in vitro cell culture environments, these key factors are often missing, which leads to changes in cell behavior. Here, we present a simple strategy for using optical media discs with nanogrooves and other polymer-based substrates nanomolded from the discs to directly culture muscle cells to study their response to the effect of biophysical cues such as nanotopography and substrate stiffness. We extend the range of study of biophysical cues for myoblasts by showing that they can sense ripple sizes as small as a 100 nm width and a 20 nm depth for myotube alignment, which has not been reported previously. The results revealed that nanotopography and substrate stiffness regulated myoblast proliferation and morphology independently, with nanotopographical cues showing a higher effect. These biophysical cues also worked synergistically, and their individual effects on cells were additive; i.e., by comparing cells grown on different polymer-based substrates (with and without nanogrooves), the cell proliferation rate could be reduced by as much as ~29%, and the elongation rate could be increased as much as ~116%. Moreover, during myogenesis, muscle cells actively responded to nanotopography and consistently showed increases in fusion and maturation indices of ~28% and ~21%, respectively. Finally, under electrical stimulation, the contraction amplitude of well-aligned myotubes was found to be almost 3 times greater than that for the cells on a smooth surface, regardless of the substrate stiffness.
Collapse
Affiliation(s)
- Cong Wu
- Department of Mechanical Engineering, City University of Hong Kong, Hong Kong, China
| | - Chriss S. M. Chin
- Department of Mechanical Engineering, City University of Hong Kong, Hong Kong, China
| | - Qingyun Huang
- Department of Mechanical Engineering, City University of Hong Kong, Hong Kong, China
| | - Ho-Yin Chan
- Department of Mechanical Engineering, City University of Hong Kong, Hong Kong, China
| | - Xinge Yu
- Department of Biomedical Engineering, City University of Hong Kong, Hong Kong, China
| | | | - Wen J. Li
- Department of Mechanical Engineering, City University of Hong Kong, Hong Kong, China
| |
Collapse
|
43
|
Bae M, Hwang DW, Ko MK, Jin Y, Shin WJ, Park W, Chae S, Lee HJ, Jang J, Yi HG, Lee DS, Cho DW. Neural stem cell delivery using brain-derived tissue-specific bioink for recovering from traumatic brain injury. Biofabrication 2021; 13. [PMID: 34551404 DOI: 10.1088/1758-5090/ac293f] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2021] [Accepted: 09/22/2021] [Indexed: 01/02/2023]
Abstract
Traumatic brain injury is one of the leading causes of accidental death and disability. The loss of parts in a severely injured brain induces edema, neuronal apoptosis, and neuroinflammation. Recently, stem cell transplantation demonstrated regenerative efficacy in an injured brain. However, the efficacy of current stem cell therapy needs improvement to resolve issues such as low survival of implanted stem cells and low efficacy of differentiation into respective cells. We developed brain-derived decellularized extracellular matrix (BdECM) bioink that is printable and has native brain-like stiffness. This study aimed to fabricate injured cavity-fit scaffold with BdECM bioink and assessed the utility of BdECM bioink for stem cell delivery to a traumatically injured brain. Our BdECM bioink had shear thinning property for three-dimensional (3D)-cell-printing and physical properties and fiber structures comparable to those of the native brain, which is important for tissue integration after implantation. The human neural stem cells (NSCs) (F3 cells) laden with BdECM bioink were found to be fully differentiated to neurons; the levels of markers for mature differentiated neurons were higher than those observed with collagen bioinkin vitro. Moreover, the BdECM bioink demonstrated potential in defect-fit carrier fabrication with 3D cell-printing, based on the rheological properties and shape fidelity of the material. As F3 cell-laden BdECM bioink was transplanted into the motor cortex of a rat brain, high efficacy of differentiation into mature neurons was observed in the transplanted NSCs; notably increased level of MAP2, a marker of neuronal differentiation, was observed. Furthermore, the transplanted-cell bioink suppressed reactive astrogliosis and microglial activation that may impede regeneration of the injured brain. The brain-specific material reported here is favorable for NSC differentiation and suppression of neuroinflammation and is expected to successfully support regeneration of a traumatically injured brain.
Collapse
Affiliation(s)
- Mihyeon Bae
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeonsangbuk-do 37673, Republic of Korea
| | - Do Won Hwang
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,THERABEST, Co. Ltd, Seocho-daero 40-gil, Seoul 06657, Republic of Korea
| | - Min Kyung Ko
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,THERABEST, Co. Ltd, Seocho-daero 40-gil, Seoul 06657, Republic of Korea
| | - Yeona Jin
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea
| | - Woo Jung Shin
- THERABEST, Co. Ltd, Seocho-daero 40-gil, Seoul 06657, Republic of Korea
| | - Wonbin Park
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeonsangbuk-do 37673, Republic of Korea
| | - Suhun Chae
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeonsangbuk-do 37673, Republic of Korea
| | - Hong Jun Lee
- College of Medicine and Medical Research Institute, Chungbuk National University, Cheongju, Chungbuk, Republic of Korea.,Research Institute eBiogen Inc., Seoul, Republic of Korea
| | - Jinah Jang
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeonsangbuk-do 37673, Republic of Korea.,Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeonsangbuk-do 37673, Republic of Korea.,Institute for Convergence Research and Education in Advanced Technology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| | - Hee-Gyeong Yi
- Department of Rural and Biosystems Engineering, College of Agriculture and Life Sciences, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Dong Soo Lee
- Department of Nuclear Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul 03080, Republic of Korea.,Department of Molecular Medicine and Biopharmaceutical Sciences, Graduate School of Convergence Science and Technology, and College of Medicine or College of Pharmacy, Seoul National University, Seoul, Republic of Korea
| | - Dong-Woo Cho
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, Gyeonsangbuk-do 37673, Republic of Korea.,Institute for Convergence Research and Education in Advanced Technology, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea
| |
Collapse
|
44
|
Lee S, Kim MS, Patel KD, Choi H, Thangam R, Yoon J, Koo TM, Jung HJ, Min S, Bae G, Kim Y, Han SB, Kang N, Kim M, Li N, Fu HE, Jeon YS, Song JJ, Kim DH, Park S, Choi JW, Paulmurugan R, Kang YC, Lee H, Wei Q, Dravid VP, Lee KB, Kim YK, Kang H. Magnetic Control and Real-Time Monitoring of Stem Cell Differentiation by the Ligand Nanoassembly. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2021; 17:e2102892. [PMID: 34515417 DOI: 10.1002/smll.202102892] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Revised: 07/04/2021] [Indexed: 06/13/2023]
Abstract
Native extracellular matrix (ECM) exhibits dynamic change in the ligand position. Herein, the ECM-emulating control and real-time monitoring of stem cell differentiation are demonstrated by ligand nanoassembly. The density of gold nanoassembly presenting cell-adhesive Arg-Gly-Asp (RGD) ligand on Fe3 O4 (magnetite) nanoparticle in nanostructures flexibly grafted to material is changed while keeping macroscale ligand density invariant. The ligand nanoassembly on the Fe3 O4 can be magnetically attracted to mediate rising and falling ligand movements via linker stretching and compression, respectively. High ligand nanoassembly density stimulates integrin ligation to activate the mechanosensing-assisted stem cell differentiation, which is monitored via in situ real-time electrochemical sensing. Magnetic control of rising and falling ligand movements hinders and promotes the adhesion-mediated mechanotransduction and differentiation of stem cells, respectively. These rising and falling ligand states yield the difference in the farthest distance (≈34.6 nm) of the RGD from material surface, thereby dynamically mimicking static long and short flexible linkers, which hinder and promote cell adhesion, respectively. Design of cytocompatible ligand nanoassemblies can be made with combinations of dimensions, shapes, and biomimetic ligands for remotely regulating stem cells for offering novel methodologies to advance regenerative therapies.
Collapse
Affiliation(s)
- Sungkyu Lee
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Myeong Soo Kim
- Institute for High Technology Materials and Devices, Korea University, Seoul, 02841, Republic of Korea
| | - Kapil D Patel
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Hyojun Choi
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Ramar Thangam
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
- Institute for High Technology Materials and Devices, Korea University, Seoul, 02841, Republic of Korea
| | - Jinho Yoon
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ, 08854, USA
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, 04107, Republic of Korea
| | - Thomas Myeongseok Koo
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Hee Joon Jung
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, 60208, USA
- International Institute for Nanotechnology, Evanston, IL, 60208, USA
- NUANCE Center, Northwestern University, Evanston, IL, 60208, USA
| | - Sunhong Min
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Gunhyu Bae
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Yuri Kim
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Seong-Beom Han
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Nayeon Kang
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Minjin Kim
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Na Li
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, 08308, Republic of Korea
| | - Hong En Fu
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Yoo Sang Jeon
- Institute of Engineering Research, Korea University, Seoul, 02841, Republic of Korea
| | - Jae-Jun Song
- Department of Otorhinolaryngology-Head and Neck Surgery, Korea University College of Medicine, Seoul, 08308, Republic of Korea
| | - Dong-Hwee Kim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Steve Park
- Department of Materials Science and Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, 34141, Republic of Korea
| | - Jeong-Woo Choi
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul, 04107, Republic of Korea
| | - Ramasamy Paulmurugan
- Department of Radiology, Molecular Imaging Program at Stanford, Stanford University School of Medicine, Stanford University, Palo Alto, CA, 94304, USA
- Department of Radiology, Canary Center at Stanford for Cancer Early Detection, Stanford University School of Medicine, Stanford University, Palo Alto, CA, 94304, USA
| | - Yun Chan Kang
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Heon Lee
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Qiang Wei
- College of Polymer Science and Engineering, State Key Laboratory of Polymer Materials and Engineering, Sichuan University, Chengdu, 610065, China
| | - Vinayak P Dravid
- Department of Materials Science and Engineering, Northwestern University, Evanston, IL, 60208, USA
- International Institute for Nanotechnology, Evanston, IL, 60208, USA
- NUANCE Center, Northwestern University, Evanston, IL, 60208, USA
| | - Ki-Bum Lee
- Department of Chemistry and Chemical Biology, Rutgers University, Piscataway, NJ, 08854, USA
| | - Young Keun Kim
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
| | - Heemin Kang
- Department of Materials Science and Engineering, Korea University, Seoul, 02841, Republic of Korea
- Department of Biomicrosystem Technology, Korea University, Seoul, 02841, Republic of Korea
| |
Collapse
|
45
|
Bang S, Hwang KS, Jeong S, Cho IJ, Choi N, Kim J, Kim HN. Engineered neural circuits for modeling brain physiology and neuropathology. Acta Biomater 2021; 132:379-400. [PMID: 34157452 DOI: 10.1016/j.actbio.2021.06.024] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Revised: 05/16/2021] [Accepted: 06/14/2021] [Indexed: 12/14/2022]
Abstract
The neural circuits of the central nervous system are the regulatory pathways for feeling, motion control, learning, and memory, and their dysfunction is closely related to various neurodegenerative diseases. Despite the growing demand for the unraveling of the physiology and functional connectivity of the neural circuits, their fundamental investigation is hampered because of the inability to access the components of neural circuits and the complex microenvironment. As an alternative approach, in vitro human neural circuits show principles of in vivo human neuronal circuit function. They allow access to the cellular compartment and permit real-time monitoring of neural circuits. In this review, we summarize recent advances in reconstituted in vitro neural circuits using engineering techniques. To this end, we provide an overview of the fabrication techniques and methods for stimulation and measurement of in vitro neural circuits. Subsequently, representative examples of in vitro neural circuits are reviewed with a particular focus on the recapitulation of structures and functions observed in vivo, and we summarize their application in the study of various brain diseases. We believe that the in vitro neural circuits can help neuroscience and the neuropharmacology. STATEMENT OF SIGNIFICANCE: Despite the growing demand to unravel the physiology and functional connectivity of the neural circuits, the studies on the in vivo neural circuits are frequently limited due to the poor accessibility. Furthermore, single neuron-based analysis has an inherent limitation in that it does not reflect the full spectrum of the neural circuit physiology. As an alternative approach, in vitro engineered neural circuit models have arisen because they can recapitulate the structural and functional characteristics of in vivo neural circuits. These in vitro neural circuits allow the mimicking of dysregulation of the neural circuits, including neurodegenerative diseases and traumatic brain injury. Emerging in vitro engineered neural circuits will provide a better understanding of the (patho-)physiology of neural circuits.
Collapse
Affiliation(s)
- Seokyoung Bang
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea
| | - Kyeong Seob Hwang
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; School of Mechanical Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Sohyeon Jeong
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea
| | - Il-Joo Cho
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea; School of Electrical and Electronics Engineering, Yonsei University, Seoul 03722, Republic of Korea; Yonsei-KIST Convergence Research Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Nakwon Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea; KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul 02841, Republic of Korea.
| | - Jongbaeg Kim
- School of Mechanical Engineering, Yonsei University, Seoul 03722, Republic of Korea.
| | - Hong Nam Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Republic of Korea; Division of Bio-Medical Science & Technology, KIST School, Korea University of Science and Technology (UST), Seoul 02792, Republic of Korea.
| |
Collapse
|
46
|
Kim SK, Kim YH, Park S, Cho SW. Organoid engineering with microfluidics and biomaterials for liver, lung disease, and cancer modeling. Acta Biomater 2021; 132:37-51. [PMID: 33711526 DOI: 10.1016/j.actbio.2021.03.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Revised: 02/14/2021] [Accepted: 03/01/2021] [Indexed: 02/08/2023]
Abstract
As life expectancy improves and the number of people suffering from various diseases increases, the need for developing effective personalized disease models is rapidly rising. The development of organoid technology has led to better recapitulation of the in vivo environment of organs, and can overcome the constraints of existing disease models. However, for more precise disease modeling, engineering approaches such as microfluidics and biomaterials, that aid in mimicking human physiology, need to be integrated with the organoid models. In this review, we introduce key elements for disease modeling and recent engineering advances using both liver and lung organoids. Due to the importance of personalized medicine, we also emphasize patient-derived cancer organoid models and their engineering approaches. These organoid-based disease models combined with microfluidics, biomaterials, and co-culture systems will provide a powerful research platform for understanding disease mechanisms and developing precision medicine; enabling preclinical drug screening and drug development. STATEMENT OF SIGNIFICANCE: The development of organoid technology has led to better recapitulation of the in vivo environment of organs, and can overcome the constraints of existing disease models. However, for more precise disease modeling, engineering approaches such as microfluidics and biomaterials, that aid in mimicking human physiology, need to be integrated with the organoid models. In this review, we introduce liver, lung, and cancer organoids integrated with various engineering approaches as a novel platform for personalized disease modeling. These engineered organoid-based disease models will provide a powerful research platform for understanding disease mechanisms and developing precision medicine.
Collapse
|
47
|
Jin Y, Cho SW. Bioengineering platforms for cell therapeutics derived from pluripotent and direct reprogramming. APL Bioeng 2021; 5:031501. [PMID: 34258498 PMCID: PMC8263070 DOI: 10.1063/5.0040621] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2020] [Accepted: 06/01/2021] [Indexed: 12/13/2022] Open
Abstract
Pluripotent and direct reprogramming technologies hold great potential for tissue repair and restoration of tissue and organ function. The implementation of induced pluripotent stem cells and directly reprogrammed cells in biomedical research has resulted in a significant leap forward in the highly promising area of regenerative medicine. While these therapeutic strategies are promising, there are several obstacles to overcome prior to the introduction of these therapies into clinical settings. Bioengineering technologies, such as biomaterials, bioprinting, microfluidic devices, and biostimulatory systems, can enhance cell viability, differentiation, and function, in turn the efficacy of cell therapeutics generated via pluripotent and direct reprogramming. Therefore, cellular reprogramming technologies, in combination with tissue-engineering platforms, are poised to overcome current bottlenecks associated with cell-based therapies and create new ways of producing engineered tissue substitutes.
Collapse
Affiliation(s)
- Yoonhee Jin
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | | |
Collapse
|
48
|
Cho AN, Jin Y, An Y, Kim J, Choi YS, Lee JS, Kim J, Choi WY, Koo DJ, Yu W, Chang GE, Kim DY, Jo SH, Kim J, Kim SY, Kim YG, Kim JY, Choi N, Cheong E, Kim YJ, Je HS, Kang HC, Cho SW. Microfluidic device with brain extracellular matrix promotes structural and functional maturation of human brain organoids. Nat Commun 2021; 12:4730. [PMID: 34354063 PMCID: PMC8342542 DOI: 10.1038/s41467-021-24775-5] [Citation(s) in RCA: 193] [Impact Index Per Article: 48.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 07/06/2021] [Indexed: 11/10/2022] Open
Abstract
Brain organoids derived from human pluripotent stem cells provide a highly valuable in vitro model to recapitulate human brain development and neurological diseases. However, the current systems for brain organoid culture require further improvement for the reliable production of high-quality organoids. Here, we demonstrate two engineering elements to improve human brain organoid culture, (1) a human brain extracellular matrix to provide brain-specific cues and (2) a microfluidic device with periodic flow to improve the survival and reduce the variability of organoids. A three-dimensional culture modified with brain extracellular matrix significantly enhanced neurogenesis in developing brain organoids from human induced pluripotent stem cells. Cortical layer development, volumetric augmentation, and electrophysiological function of human brain organoids were further improved in a reproducible manner by dynamic culture in microfluidic chamber devices. Our engineering concept of reconstituting brain-mimetic microenvironments facilitates the development of a reliable culture platform for brain organoids, enabling effective modeling and drug development for human brain diseases.
Collapse
Affiliation(s)
- Ann-Na Cho
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Yoonhee Jin
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Yeonjoo An
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Jin Kim
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Yi Sun Choi
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Jung Seung Lee
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Junghoon Kim
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Won-Young Choi
- Department of Biochemistry, Yonsei University, Seoul, Republic of Korea
| | - Dong-Jun Koo
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
| | - Weonjin Yu
- Signature Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Gyeong-Eon Chang
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Dong-Yoon Kim
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
| | - Sung-Hyun Jo
- Department of Chemical Engineering, Soongsil University, Seoul, Republic of Korea
| | - Jihun Kim
- Division of Pediatric Neurology, Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Sung-Yon Kim
- Institute of Molecular Biology and Genetics, Seoul National University, Seoul, Republic of Korea
- Department of Chemistry, Seoul National University, Seoul, Republic of Korea
| | - Yun-Gon Kim
- Department of Chemical Engineering, Soongsil University, Seoul, Republic of Korea
| | - Ju Young Kim
- Department of Advanced Materials Engineering, Kangwon National University, Samcheok, Republic of Korea
| | - Nakwon Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Eunji Cheong
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Young-Joon Kim
- Department of Biochemistry, Yonsei University, Seoul, Republic of Korea
| | - Hyunsoo Shawn Je
- Signature Program in Neuroscience and Behavioral Disorders, Duke-NUS Medical School, Singapore, Singapore
| | - Hoon-Chul Kang
- Division of Pediatric Neurology, Department of Pediatrics, Severance Children's Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea.
- Center for Nanomedicine, Institute for Basic science (IBS), Seoul, Republic of Korea.
- Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
49
|
Kim J, Lee KT, Lee JS, Shin J, Cui B, Yang K, Choi YS, Choi N, Lee SH, Lee JH, Bahn YS, Cho SW. Fungal brain infection modelled in a human-neurovascular-unit-on-a-chip with a functional blood-brain barrier. Nat Biomed Eng 2021; 5:830-846. [PMID: 34127820 DOI: 10.1038/s41551-021-00743-8] [Citation(s) in RCA: 94] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2019] [Accepted: 04/30/2021] [Indexed: 02/05/2023]
Abstract
The neurovascular unit, which consists of vascular cells surrounded by astrocytic end-feet and neurons, controls cerebral blood flow and the permeability of the blood-brain barrier (BBB) to maintain homeostasis in the neuronal milieu. Studying how some pathogens and drugs can penetrate the human BBB and disrupt neuronal homeostasis requires in vitro microphysiological models of the neurovascular unit. Here we show that the neurotropism of Cryptococcus neoformans-the most common pathogen causing fungal meningitis-and its ability to penetrate the BBB can be modelled by the co-culture of human neural stem cells, brain microvascular endothelial cells and brain vascular pericytes in a human-neurovascular-unit-on-a-chip maintained by a stepwise gravity-driven unidirectional flow and recapitulating the structural and functional features of the BBB. We found that the pathogen forms clusters of cells that penetrate the BBB without altering tight junctions, suggesting a transcytosis-mediated mechanism. The neurovascular-unit-on-a-chip may facilitate the study of the mechanisms of brain infection by pathogens, and the development of drugs for a range of brain diseases.
Collapse
Affiliation(s)
- Jin Kim
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Kyung-Tae Lee
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Jong Seung Lee
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Jisoo Shin
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Baofang Cui
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Kisuk Yang
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Yi Sun Choi
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea
| | - Nakwon Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea.,KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, Republic of Korea.,Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Republic of Korea
| | - Soo Hyun Lee
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Jae-Hyun Lee
- Institute for Basic Science (IBS), Center for Nanomedicine, Seoul, Republic of Korea.,Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea
| | - Yong-Sun Bahn
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea.
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul, Republic of Korea. .,Institute for Basic Science (IBS), Center for Nanomedicine, Seoul, Republic of Korea. .,Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul, Republic of Korea.
| |
Collapse
|
50
|
Mattiassi S, Rizwan M, Grigsby CL, Zaw AM, Leong KW, Yim EKF. Enhanced efficiency of nonviral direct neuronal reprogramming on topographical patterns. Biomater Sci 2021; 9:5175-5191. [PMID: 34128504 DOI: 10.1039/d1bm00400j] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Nonviral direct neuronal reprogramming holds significant potential in the fields of tissue engineering and regenerative medicine. However, the issue of low reprogramming efficiency poses a major barrier to its application. We propose that topographical cues, which have been applied successfully to enhance lineage-directed differentiation and multipotent stem cell transdifferentiation, could improve nonviral direct neuronal reprogramming efficiency. To investigate, we used a polymer-BAM (Brn2, Ascl1, Myt1l) factor transfection polypex to reprogram primary mouse embryonic fibroblasts. Using a multiarchitecture chip, we screened for patterns that may improve transfection and/or subsequent induced neuron reprogramming efficiency. Selected patterns were then investigated further by analyzing β-tubulin III (TUJ1) and microtubule-associated protein 2 (MAP2) protein expression, cell morphology and electrophysiological function of induced neurons. Certain hierarchical topographies, with nanopatterns imprinted on micropatterns, significantly improved the percentage of TUJ1+ and MAP2+ cells. It is postulated that the microscale base pattern enhances initial BAM expression while the nanoscale sub-pattern promotes subsequent maturation. This is because the base pattern alone increased expression of TUJ1 and MAP2, while the nanoscale pattern was the only pattern yielding induced neurons capable of firing multiple action potentials. Nanoscale patterns also produced the highest fraction of cells showing spontaneous synaptic activity. Overall, reprogramming efficiency with one dose of polyplex on hierarchical patterns was comparable to that of five doses without topography. Thus, topography can enhance nonviral direct reprogramming of fibroblasts into induced neurons.
Collapse
Affiliation(s)
- Sabrina Mattiassi
- Department of Chemical Engineering, University of Waterloo, 200 University Ave. W, Waterloo, Ontario N2L 3G1, Canada.
| | | | | | | | | | | |
Collapse
|