1
|
Shi M, Fischer P, Melde K. Acoustic holographic assembly of cell-dense tissue constructs. Biofabrication 2025; 17:035009. [PMID: 40328276 DOI: 10.1088/1758-5090/add49e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2025] [Accepted: 05/06/2025] [Indexed: 05/08/2025]
Abstract
Tissue engineering aims to develop tissue constructs as models or substitutes for native tissues. For organ-level biological studies and regenerative medicine applications, it is essential to fabricate tissue constructs with physiologically relevant cell densities (on the order of 10 million to 1 billion cells·ml-1), large size (centimeter scale and larger), and a controllable geometry to guide tissue maturation. State-of-the-art biofabrication methods, however, struggle to simultaneously meet all of these demands. The recently proposed acoustic holographic assembly (AHA) method shows promise, as it is compatible with culture media and enables the contactless, label-free, and volumetric assembly of biological cells in a predefined geometry within few minutes. Here we present an AHA biofabrication scheme designated for fabricating cell-dense, centimeter-scale, and arbitrarily-shaped tissue constructs using a compact benchtop instrument compatible with a biolab environment. We demonstrate the assembly of C2C12 myoblasts in gelatin methacryloyl (GelMA) into large and asymmetric branch-shaped constructs, which are rapidly formed with an average cell density of 40 million cells·ml-1and a local density of up to 260 million cells·ml-1. Featuring a high viability of 90.5 ± 4.3%, the assembled cell constructs are observed to grow within the GelMA hydrogel under perfusion over five days. Further, we show how AHA can-in a single step-assemble cells into layered and three-dimensional geometries inside standard cell culture labware. It can therefore help obtain engineered tissue constructs with structural and functional characteristics seen in more complex native tissues.
Collapse
Affiliation(s)
- Minghui Shi
- Max Planck Institute for Medical Research, Jahnstr. 29, 69120 Heidelberg, Germany
- Institute for Molecular Systems Engineering and Advanced Materials, Heidelberg University, Im Neuenheimer Feld 225, 69120 Heidelberg, Germany
| | - Peer Fischer
- Max Planck Institute for Medical Research, Jahnstr. 29, 69120 Heidelberg, Germany
- Institute for Molecular Systems Engineering and Advanced Materials, Heidelberg University, Im Neuenheimer Feld 225, 69120 Heidelberg, Germany
- Institute for Basic Science (IBS), Center for Nanomedicine, Seoul 03722, Republic of Korea
- Department of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea
| | - Kai Melde
- Max Planck Institute for Medical Research, Jahnstr. 29, 69120 Heidelberg, Germany
- Institute for Molecular Systems Engineering and Advanced Materials, Heidelberg University, Im Neuenheimer Feld 225, 69120 Heidelberg, Germany
| |
Collapse
|
2
|
Chen Y, Yu K, Jiang Z, Yang G. CRISPR-based genetically modified scaffold-free biomaterials for tissue engineering and regenerative medicine. Biomater Sci 2025. [PMID: 40326747 DOI: 10.1039/d5bm00194c] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
CRISPR-based genetically modified scaffold-free biomaterials, including extracellular vehicles, cell sheets, cell aggregates, organoids and organs, have attracted significant attention in the fields of regenerative medicine and tissue engineering in recent years. With a wide range of applications in gene therapy, modeling disease, tissue regeneration, organ xenotransplantation, modeling organogenesis as well as gene and drug screening, they are at a critical juncture from clinical trials to therapeutic applications. Xenografts have already been tested on non-human primates and humans. However, we have to admit that a series of obstacles still need to be addressed, such as immune response, viral infection, off-target effects, difficulty in mass production, and ethical issues. Therefore, future research should pay more attention to improving their safety, accuracy of gene editing, flexibility of production, and ethical rationality. This review summarizes various types of CRISPR-based genetically modified scaffold-free biomaterials, including their preparation procedures, applications, and possible improvements.
Collapse
Affiliation(s)
- Yunxuan Chen
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| | - Ke Yu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| | - Zhiwei Jiang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| | - Guoli Yang
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou 310000, China.
| |
Collapse
|
3
|
Abdollahi S, Zarin B, Vatani M, Vajhadin F, Hassani M, Jalali P, Kim K, Sanati-Nezhad A. Biomimetic culture substrates for modelling homeostatic intestinal epithelium in vitro. Nat Commun 2025; 16:4120. [PMID: 40316543 PMCID: PMC12048609 DOI: 10.1038/s41467-025-59459-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 04/22/2025] [Indexed: 05/04/2025] Open
Abstract
The increasing interest in utilizing three-dimensional (3D) in vitro models with innovative biomaterials to engineer functional tissues arises from the limitations of conventional cell culture methods in accurately reproducing the complex physiological conditions of living organisms. This study presents a strategy for replicating the intricate microenvironment of the intestine by cultivating intestinal cells within bioinspired 3D interfaces that recapitulate the villus-crypt architecture and 3D tissue arrangement of the intestine. Intestinal cells cultured on these biomimetic substrates exhibited phenotypes and differentiation characteristics resembling intestinal-specific cell types, effectively replicating intestinal tissue. Notably, tissue proliferation and differentiation were achieved within 72-120 h-significantly faster than the several weeks required by conventional bioengineered materials, which often pose risks of tissue necrosis or cross-contamination. Additionally, the differentiated cells on these villi-crypts mimicking bio-interfaces exhibit higher production of natural antimicrobial peptides, resulting in reduced pathogenic infection compared to control samples. Furthermore, our method stands out for simplicity in fabrication, eliminating the need for cleanroom procedures and complex microfabrication techniques.
Collapse
Affiliation(s)
- Sorosh Abdollahi
- Department of Biomedical Engineering, University of Calgary, Calgary, Alberta, T2N 1N4, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, T2N 2T8, Canada
| | - Bahareh Zarin
- Department of Biomedical Engineering, University of Calgary, Calgary, Alberta, T2N 1N4, Canada
| | - Maryam Vatani
- Department of Biomedical Engineering, University of Calgary, Calgary, Alberta, T2N 1N4, Canada
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, T2N 2T8, Canada
| | - Fereshteh Vajhadin
- Department of Biomedical Engineering, University of Calgary, Calgary, Alberta, T2N 1N4, Canada
| | - Mohsen Hassani
- Department of Mechanical & Manufacturing Engineering, University of Calgary, Calgary, Alberta, T2N 1N4, Canada
| | - Pezhman Jalali
- Department of Biomedical Engineering, University of Calgary, Calgary, Alberta, T2N 1N4, Canada
| | - Keekyoung Kim
- Department of Biomedical Engineering, University of Calgary, Calgary, Alberta, T2N 1N4, Canada
- Department of Mechanical & Manufacturing Engineering, University of Calgary, Calgary, Alberta, T2N 1N4, Canada
| | - Amir Sanati-Nezhad
- Department of Biomedical Engineering, University of Calgary, Calgary, Alberta, T2N 1N4, Canada.
- Snyder Institute for Chronic Diseases, University of Calgary, Calgary, Alberta, T2N 2T8, Canada.
- Department of Mechanical & Manufacturing Engineering, University of Calgary, Calgary, Alberta, T2N 1N4, Canada.
- Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, T2N 2T8, Canada.
| |
Collapse
|
4
|
Lu D, Xia B, Feng T, Qi G, Ma Z. The Role of Cancer Organoids in Ferroptosis, Pyroptosis, and Necroptosis: Functions and Clinical Implications. Biomolecules 2025; 15:659. [PMID: 40427552 PMCID: PMC12108908 DOI: 10.3390/biom15050659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Revised: 04/28/2025] [Accepted: 05/01/2025] [Indexed: 05/29/2025] Open
Abstract
The enduring prevalence of cancer worldwide constitutes a significant public health challenge, thereby emphasizing the imperative for the development of therapeutic models capable of accounting for the heterogeneity inherent in tumors. In this context, cancer organoids have emerged as powerful tools for studying tumor biology, providing valuable insights into the complex interactions within the tumor microenvironment. Concurrently, research is increasingly focused on non-apoptotic forms of regulated cell death (RCD)-including ferroptosis, pyroptosis, and necroptosis-which exert pivotal influences on cancer development and progression. Cancer organoids not only recapitulate the genetic and phenotypic heterogeneity of the original tumors but also enable more precise investigations into the roles of non-apoptotic RCDs within oncology. This review explores the utility of cancer organoids in delineating the molecular mechanisms underlying RCDs and their implications for cancer biology and treatment responses. By synthesizing recent research findings, it highlights the essential role of organoid models in uncovering the intricate details of non-apoptotic RCDs. Furthermore, it emphasizes promising directions for future research that aim to deepen our understanding of these pathways and their therapeutic potential. The integration of organoid models into investigations of ferroptosis, pyroptosis, and necroptosis provides novel insights into oncogenic mechanisms and facilitates the development of targeted therapeutic strategies. By bridging cancer organoids with human pathophysiology, this approach not only provides a transformative framework for dissecting oncogenic pathways but also enables the design of precision therapeutics that selectively target the molecular machinery underlying non-apoptotic RCDs.
Collapse
Affiliation(s)
- Dingci Lu
- The First Affiliated Hospital of Yangtze University, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China; (D.L.); (B.X.); (T.F.)
- School of Basic Medicine, Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China
- Department of Clinical Laboratory, The Second Hospital of Jingzhou, Jingzhou 434000, China
| | - Bingqian Xia
- The First Affiliated Hospital of Yangtze University, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China; (D.L.); (B.X.); (T.F.)
- School of Basic Medicine, Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China
| | - Tianquan Feng
- The First Affiliated Hospital of Yangtze University, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China; (D.L.); (B.X.); (T.F.)
- School of Basic Medicine, Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China
| | - Gui Qi
- The First Affiliated Hospital of Yangtze University, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China; (D.L.); (B.X.); (T.F.)
- School of Basic Medicine, Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China
| | - Zhaowu Ma
- The First Affiliated Hospital of Yangtze University, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China; (D.L.); (B.X.); (T.F.)
- School of Basic Medicine, Health Science Center, Yangtze University, Nanhuan Road 1, Jingzhou 434023, China
| |
Collapse
|
5
|
Revokatova D, Bikmulina P, Heydari Z, Solovieva A, Vosough M, Shpichka A, Timashev P. Getting Blood out of a Stone: Vascularization via Spheroids and Organoids in 3D Bioprinting. Cells 2025; 14:665. [PMID: 40358189 PMCID: PMC12071597 DOI: 10.3390/cells14090665] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2025] [Revised: 04/25/2025] [Accepted: 04/28/2025] [Indexed: 05/15/2025] Open
Abstract
Current developments in bioequivalent technology have led to the creation of excellent models that mimic the structure and function of human organs. These models are based on the original tissues and organs of the human body, but they lack the complex interaction with the extensive network of vasculature, and this is a major challenge for these models. A functional vasculature is essential for oxygen, nutrient, and waste exchange. It is also responsible for inductive biochemical exchange, and provides a structural pattern for organ growth. In vitro systems, containing no perfusable vessels, suffer from the quick formation of a necrotic core of organoids, and further development does not occur due to increased metabolic demands. Another key limitation of 3D-based techniques is the absence of accurate architectural structures and large-scale tissue sizes. Recently, new 3D bioprinting methods have been developed for organoids and spheroids as living building blocks. These methods aim to address some of the challenges associated with 3D technologies. In this review, we discuss recent strategies for vascularization via organoids and spheroids, which are used as structural units in bioprinting to recreate natural organs and tissues with ever-increasing accuracy in structure and function.
Collapse
Affiliation(s)
- Daria Revokatova
- Institute for Regenerative Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Polina Bikmulina
- Institute for Regenerative Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Zahra Heydari
- Institute for Regenerative Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Anna Solovieva
- Semenov Institute of Chemical Physics, 119991 Moscow, Russia
| | - Massoud Vosough
- Regenerative Medicine Department, Royan Institute for Stem Cell Science, Tehran 16635148, Iran
| | - Anastasia Shpichka
- Institute for Regenerative Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Peter Timashev
- Institute for Regenerative Medicine, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| |
Collapse
|
6
|
Xie D, Chen B, Wang W, Guo W, Sun Z, Wang L, Shi B, Song Y, Su M. Nanocomposite Hydrogels and Micro/Nanostructures for Printing Organoids. ACS NANO 2025; 19:12458-12466. [PMID: 40162603 DOI: 10.1021/acsnano.4c17587] [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: 04/02/2025]
Abstract
Organoids are 3D artificial miniature organs composed of a cluster of self-renewing and self-organizing cells in vitro, which mimic the functions of real organs. Nanotechnologies, including the preparation of nanomaterials and the fabrication of micro/nanostructures, have been proven to promote cell proliferation, guide cell differentiation, and regulate cell self-organization, showing great promise in engineering organoids. In this Perspective, different types of nanocomposite hydrogels for organoid culture are summarized, the effects of micro/nanostructures on organoid growth and development are discussed, and 3D bioprinting technologies for constructing organoid models are introduced.
Collapse
Affiliation(s)
- Daixi Xie
- Key Laboratory of Green Printing, Institute of Chemistry, Chinese Academy of Sciences, Beijing Engineering Research Center of Nanomaterials for Green Printing Technology, Beijing National Laboratory for Molecular Sciences (BNLMS), Zhongguancun North First Street 2, 100190 Beijing, P. R. China
- University of Chinese Academy of Sciences, Yuquan Road No. 19A, 100049 Beijing, P. R. China
| | - Bingda Chen
- Key Laboratory of Green Printing, Institute of Chemistry, Chinese Academy of Sciences, Beijing Engineering Research Center of Nanomaterials for Green Printing Technology, Beijing National Laboratory for Molecular Sciences (BNLMS), Zhongguancun North First Street 2, 100190 Beijing, P. R. China
| | - Wenqing Wang
- Key Laboratory of Green Printing, Institute of Chemistry, Chinese Academy of Sciences, Beijing Engineering Research Center of Nanomaterials for Green Printing Technology, Beijing National Laboratory for Molecular Sciences (BNLMS), Zhongguancun North First Street 2, 100190 Beijing, P. R. China
- University of Chinese Academy of Sciences, Yuquan Road No. 19A, 100049 Beijing, P. R. China
| | - Wenjing Guo
- University of Chinese Academy of Sciences, Yuquan Road No. 19A, 100049 Beijing, P. R. China
| | - Zhiyuan Sun
- Macao Institute of Materials Science and Engineering, Faculty of Innovation Engineering, Macau University of Science and Technology, Taipa, Macao 999078, China
| | - Long Wang
- Department of Orthopedics, the Fourth Medical Center of Chinese PLA General Hospital, Beijing 100048, China
| | - Bin Shi
- Department of Organ Transplantation, the Third Medical Center of PLA General Hospital, Beijing 100039, China
| | - Yanlin Song
- Key Laboratory of Green Printing, Institute of Chemistry, Chinese Academy of Sciences, Beijing Engineering Research Center of Nanomaterials for Green Printing Technology, Beijing National Laboratory for Molecular Sciences (BNLMS), Zhongguancun North First Street 2, 100190 Beijing, P. R. China
- University of Chinese Academy of Sciences, Yuquan Road No. 19A, 100049 Beijing, P. R. China
| | - Meng Su
- Key Laboratory of Green Printing, Institute of Chemistry, Chinese Academy of Sciences, Beijing Engineering Research Center of Nanomaterials for Green Printing Technology, Beijing National Laboratory for Molecular Sciences (BNLMS), Zhongguancun North First Street 2, 100190 Beijing, P. R. China
- University of Chinese Academy of Sciences, Yuquan Road No. 19A, 100049 Beijing, P. R. China
| |
Collapse
|
7
|
Yang J, Li W, Zhang Z, Xu Z, Zhu W, Wang J, Wang W. Development and Applications of Organoids in Gynecological Diseases. Stem Cell Rev Rep 2025; 21:629-644. [PMID: 39666266 PMCID: PMC11965162 DOI: 10.1007/s12015-024-10833-0] [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] [Accepted: 12/04/2024] [Indexed: 12/13/2024]
Abstract
Organoids are rapidly self-organizing 3D in vitro cultures derived from pluripotent stem cells (PSCs) or adult stem cells (ASCs) that possess disease-like characteristics with high success rates. Due to their ability to retain tissue structure, biological phenotypes, and genetic information, they have been utilized as a novel in vitro model for disease research. In recent years, scientists have established self-organizing 3D organoids for human endometrium, fallopian tubes, ovaries, and cervix by culturing stem cells with cytokines in 3D scaffolds. The integration of organoids with animal models, organ-on-a-chip systems, and 3D printing technologies offers a novel preclinical model for exploring disease mechanisms and developing treatments. This review elaborate on the recent research progress of stem cells-formed organoids in the field of gynecology from the aspects of constructing gynecological disease organoids, drug screening and new drug development, simulation modeling, allogeneic transplantation, regenerative medicine and personalized treatment."
Collapse
Affiliation(s)
- Jian Yang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Wenwen Li
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zihan Zhang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Zhonglei Xu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Wenjing Zhu
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Jing Wang
- Department of Obstetrics and Gynecology, Anhui Women and Children's Medical Center, Hefei, Anhui, China
| | - Wenyan Wang
- Department of Obstetrics and Gynecology, Second Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
8
|
Kim JE, Jeong GJ, Yoo YM, Bhang SH, Kim JH, Shin YM, Yoo KH, Lee BC, Baek W, Heo DN, Mongrain R, Lee JB, Yoon JK. 3D bioprinting technology for modeling vascular diseases and its application. Biofabrication 2025; 17:022014. [PMID: 40081017 DOI: 10.1088/1758-5090/adc03a] [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: 08/25/2024] [Accepted: 03/13/2025] [Indexed: 03/15/2025]
Abstract
In vitromodeling of vascular diseases provides a useful platform for drug screening and mechanistic studies, by recapitulating the essential structures and physiological characteristics of the native tissue. Bioprinting is an emerging technique that offers high-resolution 3D capabilities, which have recently been employed in the modeling of various tissues and associated diseases. Blood vessels are composed of multiple layers of distinct cell types, and experience different mechanical conditions depending on the vessel type. The intimal layer, in particular, is directly exposed to such hemodynamic conditions inducing shear stress, which in turn influence vascular physiology. 3D bioprinting techniques have addressed the structural limitations of the previous vascular models, by incorporating supporting cells such as smooth muscle cells, geometrical properties such as dilation, curvature, or branching, or mechanical stimulation such as shear stress and pulsatile pressure. This paper presents a review of the physiology of blood vessels along with the pathophysiology of the target diseases including atherosclerosis, thrombosis, aneurysms, and tumor angiogenesis. Additionally, it discusses recent advances in fabricatingin vitro3D vascular disease models utilizing bioprinting techniques, while addressing the current challenges and future perspectives for the potential clinical translation into therapeutic interventions.
Collapse
Affiliation(s)
- Ju-El Kim
- Department of Systems Biotechnology, Chung-Ang University, Anseong-Si, Gyeonggi-Do 17546, Republic of Korea
| | - Gun-Jae Jeong
- Institute of Cell and Tissue Engineering, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Young Min Yoo
- Department of Biological Science, Research Institute of Women's Health, Brain Korea 21 Project, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jae Hoon Kim
- Department of Systems Biotechnology, Chung-Ang University, Anseong-Si, Gyeonggi-Do 17546, Republic of Korea
| | - Young Min Shin
- Department of Biological Science, Research Institute of Women's Health, Brain Korea 21 Project, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Kyung Hyun Yoo
- Department of Biological Science, Research Institute of Women's Health, Brain Korea 21 Project, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Byung-Chul Lee
- Department of Biological Science, Research Institute of Women's Health, Brain Korea 21 Project, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Wooyeol Baek
- Department of Plastic and Reconstructive Surgery, Institute for Human Tissue Restoration, Severance Hospital, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Dong Nyoung Heo
- Department of Dental Materials, School of Dentistry, Kyung Hee University, Seoul 02447, Republic of Korea
- Biofriends Inc., Seoul 02447, Republic of Korea
| | - Rosaire Mongrain
- Mechanical Engineering Department, McGill University, H3A 0C3 Montréal, Canada
| | - Jung Bok Lee
- Department of Biological Science, Research Institute of Women's Health, Brain Korea 21 Project, Sookmyung Women's University, Seoul 04310, Republic of Korea
| | - Jeong-Kee Yoon
- Department of Systems Biotechnology, Chung-Ang University, Anseong-Si, Gyeonggi-Do 17546, Republic of Korea
| |
Collapse
|
9
|
O'Laughlin R, Cheng F, Song H, Ming GL. Bioengineering tools for next-generation neural organoids. Curr Opin Neurobiol 2025; 92:103011. [PMID: 40132519 DOI: 10.1016/j.conb.2025.103011] [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: 10/15/2024] [Revised: 02/14/2025] [Accepted: 02/25/2025] [Indexed: 03/27/2025]
Abstract
Human stem cell-derived neural organoids were recently introduced as powerful in vitro 3D experimental model systems that innately undergo critical steps of organogenesis in culture and exhibit molecular, cellular, and structural features similar to the fetal human nervous system. These organoids have yielded new insights into human neurodevelopment and associated disorders. However, neural organoids have some crucial limitations that arise from the loosely controlled conditions for their development, an inability to maintain their spatial orientation in culture and a lack of technologies for taking long-term measurements on their morphology and electrical activity. Here, we review recent progress in using bioengineering methods to improve neural organoid formation and analysis by leveraging microfabrication, biomaterials, 3D printing, and flexible electrodes. We discuss how the applications of each technique can help to address critical limitations with standard neural organoid models. We conclude with a perspective on future applications of bioengineered next-generation neural organoids.
Collapse
Affiliation(s)
- Richard O'Laughlin
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Fangyi Cheng
- Graduate Program in Bioengineering, School of Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Hongjun Song
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Graduate Program in Bioengineering, School of Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Neurosurgery, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; The Epigenetics Institute, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Guo-Li Ming
- Department of Neuroscience and Mahoney Institute for Neurosciences, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Graduate Program in Bioengineering, School of Engineering, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Cell and Developmental Biology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Institute for Regenerative Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Department of Psychiatry, Perelman School for Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
10
|
Wang K, Wang Y, Han J, Liang Z, Zhang W, Li X, Chen J, Wang L. Biofabrication and simulation techniques for gut-on-a-chip. Biofabrication 2025; 17:022011. [PMID: 39965538 DOI: 10.1088/1758-5090/adb7c1] [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: 08/23/2024] [Accepted: 02/18/2025] [Indexed: 02/20/2025]
Abstract
Biomimetic gut models show promise for enhancing our understanding of intestinal disorder pathogenesis and accelerating therapeutic strategy development. Currentin vitromodels predominantly comprise traditional static cell culture and animal models. Static cell culture lacks the precise control of the complex microenvironment governing human intestinal function. Animal models provide greater microenvironment complexity but fail to accurately replicate human physiological conditions due to interspecies differences. As the available models do not accurately reflect the microphysiological environment and functions of the human intestine, their applications are limited. An optimal approach to intestinal modeling is yet to be developed, but the field will probably benefit from advances in biofabrication techniques. This review highlights biofabrication strategies for constructing biomimetic intestinal models and research approaches for simulating key intestinal physiological features. We also discuss potential biomedical applications of these models and provide an outlook on multi-scale intestinal modeling.
Collapse
Affiliation(s)
- Ke Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, People's Republic of China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, People's Republic of China
| | - Yushen Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, People's Republic of China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, People's Republic of China
| | - Junlei Han
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, People's Republic of China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, People's Republic of China
| | - Zhixiang Liang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, People's Republic of China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, People's Republic of China
| | - Wenhong Zhang
- College of Mechanical Engineering, Donghua University, Shanghai 201620, People's Republic of China
| | - Xinyu Li
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong 250021, People's Republic of China
| | - Jun Chen
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, People's Republic of China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, People's Republic of China
| | - Li Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, People's Republic of China
- Shandong Institute of Mechanical Design and Research, Jinan 250353, People's Republic of China
| |
Collapse
|
11
|
Taglieri M, Di Gregorio L, Matis S, Uras CRM, Ardy M, Casati S, Marchese M, Poggi A, Raffaghello L, Benelli R. Colorectal Organoids: Models, Imaging, Omics, Therapy, Immunology, and Ethics. Cells 2025; 14:457. [PMID: 40136707 PMCID: PMC11941511 DOI: 10.3390/cells14060457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/06/2025] [Accepted: 03/12/2025] [Indexed: 03/27/2025] Open
Abstract
Colorectal epithelium was the first long-term 3D organoid culture established in vitro. Identification of the key components essential for the long-term survival of the stem cell niche allowed an indefinite propagation of these cultures and the modulation of their differentiation into various lineages of mature intestinal epithelial cells. While these methods were eventually adapted to establish organoids from different organs, colorectal organoids remain a pioneering model for the development of new applications in health and disease. Several basic and applicative aspects of organoid culture, modeling, monitoring and testing are analyzed in this review. We also tackle the ethical problems of biobanking and distribution of these precious research tools, frequently confined in the laboratory of origin or condemned to destruction at the end of the project.
Collapse
Affiliation(s)
- Martina Taglieri
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.T.); (L.D.G.); (S.M.); (C.R.M.U.); (M.A.); (M.M.); (A.P.); (L.R.)
| | - Linda Di Gregorio
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.T.); (L.D.G.); (S.M.); (C.R.M.U.); (M.A.); (M.M.); (A.P.); (L.R.)
| | - Serena Matis
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.T.); (L.D.G.); (S.M.); (C.R.M.U.); (M.A.); (M.M.); (A.P.); (L.R.)
| | - Chiara Rosa Maria Uras
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.T.); (L.D.G.); (S.M.); (C.R.M.U.); (M.A.); (M.M.); (A.P.); (L.R.)
| | - Massimo Ardy
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.T.); (L.D.G.); (S.M.); (C.R.M.U.); (M.A.); (M.M.); (A.P.); (L.R.)
| | - Sara Casati
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “Gaetano Salvatore” CNR, 80131 Naples, Italy;
- Common Service ELSI, BBMRI.it (UNIMIB National Node Headquarter), 20126 Milan, Italy
| | - Monica Marchese
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.T.); (L.D.G.); (S.M.); (C.R.M.U.); (M.A.); (M.M.); (A.P.); (L.R.)
| | - Alessandro Poggi
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.T.); (L.D.G.); (S.M.); (C.R.M.U.); (M.A.); (M.M.); (A.P.); (L.R.)
| | - Lizzia Raffaghello
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.T.); (L.D.G.); (S.M.); (C.R.M.U.); (M.A.); (M.M.); (A.P.); (L.R.)
| | - Roberto Benelli
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.T.); (L.D.G.); (S.M.); (C.R.M.U.); (M.A.); (M.M.); (A.P.); (L.R.)
| |
Collapse
|
12
|
Bakirci E, Asghari Adib A, Ashraf SF, Feinberg AW. Advancing extrusion-based embedded 3D bioprinting via scientific, engineering, and process innovations. Biofabrication 2025; 17:023002. [PMID: 39965539 DOI: 10.1088/1758-5090/adb7c3] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 02/18/2025] [Indexed: 02/20/2025]
Abstract
Extrusion-based embedded 3D bioprinting, where bioinks and biomaterials are extruded within a support bath, has greatly expanded the achievable tissue architectures and complexity of biologic constructs that can be fabricated. However, significant scientific, engineering, and process-related challenges remain to recreate the full anatomic structure and physiologic function required for many therapeutic applications. This perspective explores the future advances in extrusion-based embedded 3D bioprinting that could address these challenges, paving the way for clinical translation of bioprinted tissues.
Collapse
Affiliation(s)
- Ezgi Bakirci
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, United States of America
| | - Ali Asghari Adib
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, United States of America
| | - Syed Faaz Ashraf
- Department of Cardiothoracic Surgery, University of Pittsburgh, Pittsburgh, PA 15213, United States of America
| | - Adam W Feinberg
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, United States of America
- Department of Materials Science & Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, United States of America
| |
Collapse
|
13
|
Sugihara HY, Okamoto R, Mizutani T. Intestinal organoids: The path towards clinical application. Eur J Cell Biol 2025; 104:151474. [PMID: 39740324 DOI: 10.1016/j.ejcb.2024.151474] [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: 08/30/2024] [Revised: 11/14/2024] [Accepted: 11/17/2024] [Indexed: 01/02/2025] Open
Abstract
Organoids have revolutionized the whole field of biology with their ability to model complex three-dimensional human organs in vitro. Intestinal organoids were especially consequential as the first successful long-term culture of intestinal stem cells, which raised hopes for translational medical applications. Despite significant contributions to basic research, challenges remain to develop intestinal organoids into clinical tools for diagnosis, prognosis, and therapy. In this review, we outline the current state of translational research involving adult stem cell and pluripotent stem cell derived intestinal organoids, highlighting the advances and limitations in disease modeling, drug-screening, personalized medicine, and stem cell therapy. Preclinical studies have demonstrated a remarkable functional recapitulation of infectious and genetic diseases, and there is mounting evidence for the reliability of intestinal organoids as a patient-specific avatar. Breakthroughs now allow the generation of structurally and cellularly complex intestinal models to better capture a wider range of intestinal pathophysiology. As the field develops and evolves, there is a need for standardized frameworks for generation, culture, storage, and analysis of intestinal organoids to ensure reproducibility, comparability, and interpretability of these preclinical and clinical studies to ultimately enable clinical translation.
Collapse
Affiliation(s)
- Hady Yuki Sugihara
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Ryuichi Okamoto
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Tomohiro Mizutani
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan.
| |
Collapse
|
14
|
Truong H, Abaci A, Gharacheh H, Guvendiren M. Embedded bioprinting of dense cellular constructs in bone allograft-enhanced hydrogel matrices for bone tissue engineering. Biomater Sci 2025. [PMID: 40018866 DOI: 10.1039/d4bm01616e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
Bone tissue engineering aims to address critical-sized defects by developing biomimetic scaffolds that promote repair and regeneration. This study introduces a material extrusion-based embedded bioprinting approach to fabricate dense cellular constructs within methacrylated hyaluronic acid (MeHA) hydrogels enhanced with bioactive microparticles. Composite matrices containing human bone allograft or tricalcium phosphate (TCP) particles were evaluated for their rheological, mechanical, and osteoinductive properties. High cell viability (>95%) and uniform strand dimensions were achieved across all bioprinting conditions, demonstrating the method's ability to preserve cellular integrity and structural fidelity. The inclusion of bone or TCP particles did not significantly alter the viscosity, crosslinking kinetics, or compressive modulus of the MeHA hydrogels, ensuring robust mechanical stability and shape retention. However, bone allograft particles significantly enhanced osteogenic differentiation of human mesenchymal stem cells (hMSCs), as evidenced by increased alkaline phosphatase (ALP) activity and calcium deposition. Notably, osteogenesis was observed even in basal media, with a dose-dependent response to bone particle concentration, highlighting the intrinsic bioactivity of allograft particles. This study demonstrates the potential of combining embedded bioprinting with bioactive matrices to create dense, osteoinductive cellular constructs. The ability to induce osteogenesis without external growth factors positions this platform as a scalable and clinically relevant solution for bone repair and regeneration.
Collapse
Affiliation(s)
- Hang Truong
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA.
| | - Alperen Abaci
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA.
| | - Hadis Gharacheh
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA.
| | - Murat Guvendiren
- Otto H. York Department of Chemical and Materials Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA.
- Department of Biomedical Engineering, New Jersey Institute of Technology, Newark, NJ 07102, USA
| |
Collapse
|
15
|
Mao R, Zhang J, Qin H, Liu Y, Xing Y, Zeng W. Application progress of bio-manufacturing technology in kidney organoids. Biofabrication 2025; 17:022007. [PMID: 39933190 DOI: 10.1088/1758-5090/adb4a1] [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: 08/04/2024] [Accepted: 02/11/2025] [Indexed: 02/13/2025]
Abstract
Kidney transplantation remains a pivotal treatment modality for kidney disease, yet its progress is significantly hindered by the scarcity of donor kidneys and ethical dilemmas surrounding their procurement. As organoid technology evolves and matures, the creation of bionic human kidney organoids offers profound potential for advancing kidney disease research, drug nephrotoxicity screening, and regenerative medicine. Nevertheless, current kidney organoid models grapple with limitations such as constrained cellular differentiation, underdeveloped functional structures, and a crucial absence of vascularization. This deficiency in vascularization, in particular, stunts organoid development, restricts their size, diminishes filtration capabilities, and may trigger immune inflammatory reactions through the resulting ischemic microenvironment. Hence, the achievement of vascularization within kidney organoids and the successful establishment of functional microvascular networks constitutes a paramount goal for their future progression. In this review, we provide an overview of recent advancements in biotechnology domains, encompassing organ-on-a-chip technology, biomimetic matrices, and bioprinting, with the aim of catalyzing technological breakthroughs that can enhance the vascularization of kidney organoids and broaden their applicability. These technologies hold the key to unlocking the full potential of kidney organoids as a transformative therapeutic option for kidney disease.
Collapse
Affiliation(s)
- Runqi Mao
- Department of Cell Biology, Third Military Medical University, Chongqing, People's Republic of China
| | - Junming Zhang
- Department of Cell Biology, Third Military Medical University, Chongqing, People's Republic of China
| | - Haoxiang Qin
- Department of Cell Biology, Third Military Medical University, Chongqing, People's Republic of China
| | - Yuanyuan Liu
- Department of Cell Biology, Third Military Medical University, Chongqing, People's Republic of China
| | - Yuxin Xing
- Department of Cell Biology, Third Military Medical University, Chongqing, People's Republic of China
| | - Wen Zeng
- Department of Cell Biology, Third Military Medical University, Chongqing, People's Republic of China
- State Key Laboratory of Trauma, Burn and Combined Injury, Chongqing, People's Republic of China
- Jinfeng Laboratory, Chongqing 401329, People's Republic of China
| |
Collapse
|
16
|
Wu Z, Liu R, Shao N, Zhao Y. Developing 3D bioprinting for organs-on-chips. LAB ON A CHIP 2025; 25:1081-1096. [PMID: 39775492 DOI: 10.1039/d4lc00769g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
Abstract
Organs-on-chips (OoCs) have significantly advanced biomedical research by precisely reconstructing human microphysiological systems with biomimetic functions. However, achieving greater structural complexity of cell cultures on-chip for enhanced biological mimicry remains a challenge. To overcome these challenges, 3D bioprinting techniques can be used in directly building complex 3D cultures on chips, facilitating the in vitro engineering of organ-level models. Herein, we review the distinctive features of OoCs, along with the technical and biological challenges associated with replicating complex organ structures. We discuss recent bioprinting innovations that simplify the fabrication of OoCs while increasing their architectural complexity, leading to breakthroughs in the field and enabling the investigation of previously inaccessible biological problems. We highlight the challenges for the development of 3D bioprinted OoCs, concluding with a perspective on future directions aimed at facilitating their clinical translation.
Collapse
Affiliation(s)
- Zhuhao Wu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Rui Liu
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Ning Shao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
| | - Yuanjin Zhao
- Department of Rheumatology and Immunology, Nanjing Drum Tower Hospital, School of Biological Science and Medical Engineering, Southeast University, Nanjing 210096, China.
- Shenzhen Research Institute, Southeast University, Shenzhen 518071, China
- Institute of Organoids on Chips Translational Research, Henan Academy of Sciences, Zhengzhou 450009, China
| |
Collapse
|
17
|
Yamada T, Trentesaux C, Brunger JM, Xiao Y, Stevens AJ, Martyn I, Kasparek P, Shroff NP, Aguilar A, Bruneau BG, Boffelli D, Klein OD, Lim WA. Synthetic organizer cells guide development via spatial and biochemical instructions. Cell 2025; 188:778-795.e18. [PMID: 39706189 PMCID: PMC12027307 DOI: 10.1016/j.cell.2024.11.017] [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: 09/26/2023] [Revised: 07/10/2024] [Accepted: 11/08/2024] [Indexed: 12/23/2024]
Abstract
In vitro development relies primarily on treating progenitor cells with media-borne morphogens and thus lacks native-like spatial information. Here, we engineer morphogen-secreting organizer cells programmed to self-assemble, via cell adhesion, around mouse embryonic stem (ES) cells in defined architectures. By inducing the morphogen WNT3A and its antagonist DKK1 from organizer cells, we generated diverse morphogen gradients, varying in range and steepness. These gradients were strongly correlated with morphogenetic outcomes: the range of minimum-maximum WNT activity determined the resulting range of anterior-to-posterior (A-P) axis cell lineages. Strikingly, shallow WNT activity gradients, despite showing truncated A-P lineages, yielded higher-resolution tissue morphologies, such as a beating, chambered cardiac-like structure associated with an endothelial network. Thus, synthetic organizer cells, which integrate spatial, temporal, and biochemical information, provide a powerful way to systematically and flexibly direct the development of ES or other progenitor cells in different directions within the morphogenetic landscape.
Collapse
Affiliation(s)
- Toshimichi Yamada
- Cell Design Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Coralie Trentesaux
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Jonathan M Brunger
- Cell Design Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Yini Xiao
- Cell Design Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Adam J Stevens
- Cell Design Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Iain Martyn
- Cell Design Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Petr Kasparek
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Neha P Shroff
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Angelica Aguilar
- Cell Design Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Benoit G Bruneau
- Gladstone Institutes, San Francisco, CA 94158, USA; Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA; Cardiovascular Research Institute, University of California, San Francisco, San Francisco, CA 94158, USA
| | - Dario Boffelli
- Department of Pediatrics, Cedars-Sinai Guerin Children's, Los Angeles, CA 90048, USA
| | - Ophir D Klein
- Department of Orofacial Sciences and Program in Craniofacial Biology, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Pediatrics, University of California, San Francisco, San Francisco, CA 94143, USA; Department of Pediatrics, Cedars-Sinai Guerin Children's, Los Angeles, CA 90048, USA.
| | - Wendell A Lim
- Cell Design Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA 94158, USA.
| |
Collapse
|
18
|
Verstegen MMA, Coppes RP, Beghin A, De Coppi P, Gerli MFM, de Graeff N, Pan Q, Saito Y, Shi S, Zadpoor AA, van der Laan LJW. Clinical applications of human organoids. Nat Med 2025; 31:409-421. [PMID: 39901045 DOI: 10.1038/s41591-024-03489-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Accepted: 12/17/2024] [Indexed: 02/05/2025]
Abstract
Organoids are innovative three-dimensional and self-organizing cell cultures of various lineages that can be used to study diverse tissues and organs. Human organoids have dramatically increased our understanding of developmental and disease biology. They provide a patient-specific model to study known diseases, with advantages over animal models, and can also provide insights into emerging and future health threats related to climate change, zoonotic infections, environmental pollutants or even microgravity during space exploration. Furthermore, organoids show potential for regenerative cell therapies and organ transplantation. Still, several challenges for broad clinical application remain, including inefficiencies in initiation and expansion, increasing model complexity and difficulties with upscaling clinical-grade cultures and developing more organ-specific human tissue microenvironments. To achieve the full potential of organoid technology, interdisciplinary efforts are needed, integrating advances from biology, bioengineering, computational science, ethics and clinical research. In this Review, we showcase pivotal achievements in epithelial organoid research and technologies and provide an outlook for the future of organoids in advancing human health and medicine.
Collapse
Affiliation(s)
- Monique M A Verstegen
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands.
| | - Rob P Coppes
- Departments of Biomedical Sciences and Radiation Oncology, University Medical Center Groningen, University of Groningen, Groningen, the Netherlands
| | - Anne Beghin
- Mechanobiology Institute, National University of Singapore, Singapore, Singapore
- Department of Microbiology and Immunology, Immunology Translational Research Program, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Centre for Research and Engineering in Space Technology, Universite Libre de Bruxelles, Bruxelles, Belgium
| | - Paolo De Coppi
- Stem Cell and Regenerative Medicine Section, Zayed Centre for Research into Rare Disease in Children, Great Ormond Street Institute of Child Health, University College London, London, UK
| | - Mattia F M Gerli
- Division of Surgery and Interventional Science, Department of Surgical Biotechnology, University College London, London, UK
| | - Nienke de Graeff
- Department of Medical Ethics and Health Law, Leiden University Medical Center, Leiden University, Leiden, the Netherlands
- The Novo Nordisk Foundation Center for Stem Cell Medicine (reNEW), Leiden Node, Leiden, the Netherlands
| | - Qiuwei Pan
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, the Netherlands
| | - Yoshimasa Saito
- Division of Pharmacotherapeutics, Keio University Faculty of Pharmacy, Tokyo, Japan
| | - Shaojun Shi
- Department of Organ Transplantation, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Amir A Zadpoor
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Delft, the Netherlands
| | - Luc J W van der Laan
- Department of Surgery, Erasmus MC Transplant Institute, University Medical Center Rotterdam, Rotterdam, the Netherlands
- Department of Biomechanical Engineering, Faculty of Mechanical, Maritime, and Materials Engineering, Delft University of Technology (TU Delft), Delft, the Netherlands
| |
Collapse
|
19
|
Kim Y, Kang M, Mamo MG, Adisasmita M, Huch M, Choi D. Liver organoids: Current advances and future applications for hepatology. Clin Mol Hepatol 2025; 31:S327-S348. [PMID: 39722609 PMCID: PMC11925438 DOI: 10.3350/cmh.2024.1040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 12/13/2024] [Accepted: 12/24/2024] [Indexed: 12/28/2024] Open
Abstract
The creation of self-organizing liver organoids represents a significant, although modest, step toward addressing the ongoing organ shortage crisis in allogeneic liver transplantation. However, researchers have recognized that achieving a fully functional whole liver remains a distant goal, and the original ambition of organoid-based liver generation has been temporarily put on hold. Instead, liver organoids have revolutionized the field of hepatology, extending their influence into various domains of precision and molecular medicine. These 3D cultures, capable of replicating key features of human liver function and pathology, have opened new avenues for human-relevant disease modeling, CRISPR gene editing, and high-throughput drug screening that animal models cannot accomplish. Moreover, advancements in creating more complex systems have led to the development of multicellular assembloids, dynamic organoid-on-chip systems, and 3D bioprinting technologies. These innovations enable detailed modeling of liver microenvironments and complex tissue interactions. Progress in regenerative medicine and transplantation applications continues to evolve and strives to overcome the obstacles of biocompatibility and tumorigenecity. In this review, we examine the current state of liver organoid research by offering insights into where the field currently stands, and the pivotal developments that are shaping its future.
Collapse
Affiliation(s)
- Yohan Kim
- Department of MetaBioHealth, Sungkyunkwan University, Suwon, Korea
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon, Korea
- Biomedical Institute for Convergence at SKKU, Sungkyunkwan University, Suwon, Korea
| | - Minseok Kang
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
| | - Michael Girma Mamo
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
- Research Institute of Regenerative Medicine and Stem Cells, Hanyang University, Seoul, Korea
| | - Michael Adisasmita
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
- Research Institute of Regenerative Medicine and Stem Cells, Hanyang University, Seoul, Korea
| | - Meritxell Huch
- Max Planck Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Dongho Choi
- Department of Surgery, Hanyang University College of Medicine, Seoul, Korea
- Research Institute of Regenerative Medicine and Stem Cells, Hanyang University, Seoul, Korea
- Hanyang Institute of Bioscience and Biotechnology, Hanyang University, Seoul, Korea
- Department of HY-KIST Bio-convergence, Hanyang University, Seoul, Korea
| |
Collapse
|
20
|
Moss SP, Bakirci E, Feinberg AW. Engineering the 3D structure of organoids. Stem Cell Reports 2025; 20:102379. [PMID: 39706178 PMCID: PMC11784486 DOI: 10.1016/j.stemcr.2024.11.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 12/23/2024] Open
Abstract
Organoids form through the sel f-organizing capabilities of stem cells to produce a variety of differentiated cell and tissue types. Most organoid models, however, are limited in terms of the structure and function of the tissues that form, in part because it is difficult to regulate the cell type, arrangement, and cell-cell/cell-matrix interactions within these systems. In this article, we will discuss the engineering approaches to generate more complex organoids with improved function and translational relevance, as well as their advantages and disadvantages. Additionally, we will explore how biofabrication strategies can manipulate the cell composition, 3D organization, and scale-up of organoids, thus improving their utility for disease modeling, drug screening, and regenerative medicine applications.
Collapse
Affiliation(s)
- Samuel P Moss
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Ezgi Bakirci
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA
| | - Adam W Feinberg
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA, USA; Department of Materials Science and Engineering, Carnegie Mellon University, Pittsburgh, PA, USA.
| |
Collapse
|
21
|
Zhu Z, Cheng Y, Liu X, Ding W, Liu J, Ling Z, Wu L. Advances in the Development and Application of Human Organoids: Techniques, Applications, and Future Perspectives. Cell Transplant 2025; 34:9636897241303271. [PMID: 39874083 PMCID: PMC11775963 DOI: 10.1177/09636897241303271] [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] [Revised: 10/10/2024] [Accepted: 11/11/2024] [Indexed: 01/30/2025] Open
Abstract
Organoids are three-dimensional (3D) cell cultures derived from human pluripotent stem cells or adult stem cells that recapitulate the cellular heterogeneity, structure, and function of human organs. These microstructures are invaluable for biomedical research due to their ability to closely mimic the complexity of native tissues while retaining human genetic material. This fidelity to native organ systems positions organoids as a powerful tool for advancing our understanding of human biology and for enhancing preclinical drug testing. Recent advancements have led to the successful development of a variety of organoid types, reflecting a broad range of human organs and tissues. This progress has expanded their application across several domains, including regenerative medicine, where organoids offer potential for tissue replacement and repair; disease modeling, which allows for the study of disease mechanisms and progression in a controlled environment; drug discovery and evaluation, where organoids provide a more accurate platform for testing drug efficacy and safety; and microecological research, where they contribute to understanding the interactions between microbes and host tissues. This review provides a comprehensive overview of the historical development of organoid technology, highlights the key achievements and ongoing challenges in the field, and discusses the current and emerging applications of organoids in both laboratory research and clinical practice.
Collapse
Affiliation(s)
- Zhangcheng Zhu
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Yiwen Cheng
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Xia Liu
- Department of Intensive Care Unit, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Wenwen Ding
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, China
| | - Jiaming Liu
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, China
| | - Zongxin Ling
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, China
| | - Lingbin Wu
- Department of Laboratory Medicine, Lishui Second People’s Hospital, Lishui, China
| |
Collapse
|
22
|
Hu Y, Zhu T, Cui H, Cui H. Integrating 3D Bioprinting and Organoids to Better Recapitulate the Complexity of Cellular Microenvironments for Tissue Engineering. Adv Healthc Mater 2025; 14:e2403762. [PMID: 39648636 DOI: 10.1002/adhm.202403762] [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: 09/30/2024] [Revised: 11/16/2024] [Indexed: 12/10/2024]
Abstract
Organoids, with their capacity to mimic the structures and functions of human organs, have gained significant attention for simulating human pathophysiology and have been extensively investigated in the recent past. Additionally, 3D bioprinting, as an emerging bio-additive manufacturing technology, offers the potential for constructing heterogeneous cellular microenvironments, thereby promoting advancements in organoid research. In this review, the latest developments in 3D bioprinting technologies aimed at enhancing organoid engineering are introduced. The commonly used bioprinting methods and materials for organoids, with a particular emphasis on the potential advantages of combining 3D bioprinting with organoids are summarized. These advantages include achieving high cell concentrations to form large cellular aggregates, precise deposition of building blocks to create organoids with complex structures and functions, and automation and high throughput to ensure reproducibility and standardization in organoid culture. Furthermore, this review provides an overview of relevant studies from recent years and discusses the current limitations and prospects for future development.
Collapse
Affiliation(s)
- Yan Hu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Tong Zhu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Haitao Cui
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Haijun Cui
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| |
Collapse
|
23
|
Lavrador P, Moura BS, Almeida-Pinto J, Gaspar VM, Mano JF. Engineered nascent living human tissues with unit programmability. NATURE MATERIALS 2025; 24:143-154. [PMID: 39117911 DOI: 10.1038/s41563-024-01958-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Accepted: 06/25/2024] [Indexed: 08/10/2024]
Abstract
Leveraging human cells as materials precursors is a promising approach for fabricating living materials with tissue-like functionalities and cellular programmability. Here we describe a set of cellular units with metabolically engineered glycoproteins that allow cells to tether together to function as macrotissue building blocks and bioeffectors. The generated human living materials, termed as Cellgels, can be rapidly assembled in a wide variety of programmable three-dimensional configurations with physiologically relevant cell densities (up to 108 cells per cm3), tunable mechanical properties and handleability. Cellgels inherit the ability of living cells to sense and respond to their environment, showing autonomous tissue-integrative behaviour, mechanical maturation, biological self-healing, biospecific adhesion and capacity to promote wound healing. These living features also enable the modular bottom-up assembly of multiscale constructs, which are reminiscent of human tissue interfaces with heterogeneous composition. This technology can potentially be extended to any human cell type, unlocking the possibility for fabricating living materials that harness the intrinsic biofunctionalities of biological systems.
Collapse
Affiliation(s)
- Pedro Lavrador
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Beatriz S Moura
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - José Almeida-Pinto
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Vítor M Gaspar
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Aveiro, Portugal.
| | - João F Mano
- CICECO - Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Aveiro, Portugal.
| |
Collapse
|
24
|
Derman ID, Moses JC, Rivera T, Ozbolat IT. Understanding the cellular dynamics, engineering perspectives and translation prospects in bioprinting epithelial tissues. Bioact Mater 2025; 43:195-224. [PMID: 39386221 PMCID: PMC11462153 DOI: 10.1016/j.bioactmat.2024.09.025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/04/2024] [Accepted: 09/18/2024] [Indexed: 10/12/2024] Open
Abstract
The epithelium is one of the important tissues in the body as it plays a crucial barrier role serving as a gateway into and out of the body. Most organs in the body contain an epithelial tissue component, where the tightly connected, organ-specific epithelial cells organize into cysts, invaginations, or tubules, thereby performing distinct to endocrine or exocrine secretory functions. Despite the significance of epithelium, engineering functional epithelium in vitro has remained a challenge due to it is special architecture, heterotypic composition of epithelial tissues, and most importantly, difficulty in attaining the apico-basal and planar polarity of epithelial cells. Bioprinting has brought a paradigm shift in fabricating such apico-basal polarized tissues. In this review, we provide an overview of epithelial tissues and provide insights on recapitulating their cellular arrangement and polarization to achieve epithelial function. We describe the different bioprinting techniques that have been successful in engineering polarized epithelium, which can serve as in vitro models for understanding homeostasis and studying diseased conditions. We also discuss the different attempts that have been investigated to study these 3D bioprinted engineered epithelium for preclinical use. Finally, we highlight the challenges and the opportunities that need to be addressed for translation of 3D bioprinted epithelial tissues towards paving way for personalized healthcare in the future.
Collapse
Affiliation(s)
- Irem Deniz Derman
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
| | - Joseph Christakiran Moses
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
| | - Taino Rivera
- Biomedical Engineering Department, Penn State University, University Park, PA, 16802, USA
| | - Ibrahim T. Ozbolat
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, 16802, USA
- Biomedical Engineering Department, Penn State University, University Park, PA, 16802, USA
- Materials Research Institute, Penn State University, University Park, PA, 16802, USA
- Cancer Institute, Penn State University, University Park, PA, 16802, USA
- Neurosurgery Department, Penn State University, University Park, PA, 16802, USA
- Department of Medical Oncology, Cukurova University, Adana, 01330, Turkey
| |
Collapse
|
25
|
Takahashi J, Sugihara HY, Kato S, Kawasaki S, Nagata S, Okamoto R, Mizutani T. Controlled aggregative assembly to form self-organizing macroscopic human intestine from induced pluripotent stem cells. CELL REPORTS METHODS 2024; 4:100930. [PMID: 39662475 PMCID: PMC11704612 DOI: 10.1016/j.crmeth.2024.100930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/26/2024] [Revised: 10/11/2024] [Accepted: 11/14/2024] [Indexed: 12/13/2024]
Abstract
Human intestinal organoids (HIOs) derived from human pluripotent stem cells (hPSCs) are promising resources for intestinal regenerative therapy as they recapitulate both endodermal and mesodermal components of the intestine. However, due to their hPSC-line-dependent mesenchymal development and spherical morphology, HIOs have limited applicability beyond basic research and development. Here, we demonstrate the incorporation of separately differentiated mesodermal and mid/hindgut cells into assembled spheroids to stabilize mesenchymal growth in HIOs. In parallel, we generate tubular intestinal constructs (assembled human intestinal tubules [a-HITs]) by leveraging the high aggregative property of assembled spheroids. Through rotational culture in a bioreactor, a-HITs self-organize to develop epithelium and supportive mesenchyme. Upon mesenteric transplantation, a-HITs mature into centimeter-scale tubular intestinal tissue with complex architectures. Our aggregation- and suspension-based approach offers basic technology for engineering tubular intestinal tissue from hPSCs, which could be ultimately applied to the generation of the human intestine for clinical application.
Collapse
Affiliation(s)
- Junichi Takahashi
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Hady Yuki Sugihara
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Shu Kato
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Sho Kawasaki
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Sayaka Nagata
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Ryuichi Okamoto
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Tomohiro Mizutani
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan.
| |
Collapse
|
26
|
Moro LG, Guarnier LP, Azevedo MF, Fracasso JAR, Lucio MA, de Castro MV, Dias ML, Lívero FADR, Ribeiro-Paes JT. A Brief History of Cell Culture: From Harrison to Organs-on-a-Chip. Cells 2024; 13:2068. [PMID: 39768159 PMCID: PMC11674496 DOI: 10.3390/cells13242068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2024] [Revised: 10/11/2024] [Accepted: 10/20/2024] [Indexed: 01/11/2025] Open
Abstract
This comprehensive overview of the historical milestones in cell culture underscores key breakthroughs that have shaped the field over time. It begins with Wilhelm Roux's seminal experiments in the 1880s, followed by the pioneering efforts of Ross Granville Harrison, who initiated groundbreaking experiments that fundamentally shaped the landscape of cell culture in the early 20th century. Carrel's influential contributions, notably the immortalization of chicken heart cells, have marked a significant advancement in cell culture techniques. Subsequently, Johannes Holtfreter, Aron Moscona, and Joseph Leighton introduced methodological innovations in three-dimensional (3D) cell culture, initiated by Alexis Carrel, laying the groundwork for future consolidation and expansion of the use of 3D cell culture in different areas of biomedical sciences. The advent of induced pluripotent stem cells by Takahashi and Yamanaka in 2006 was revolutionary, enabling the reprogramming of differentiated cells into a pluripotent state. Since then, recent innovations have included spheroids, organoids, and organ-on-a-chip technologies, aiming to mimic the structure and function of tissues and organs in vitro, pushing the boundaries of biological modeling and disease understanding. In this review, we overview the history of cell culture shedding light on the main discoveries, pitfalls and hurdles that were overcome during the transition from 2D to 3D cell culture techniques. Finally, we discussed the future directions for cell culture research that may accelerate the development of more effective and personalized treatments.
Collapse
Affiliation(s)
- Lincoln Gozzi Moro
- Human Genome and Stem Cell Research Center, Institute of Biosciences, University of São Paulo—USP, São Paulo 01246-904, Brazil; (L.G.M.); (M.V.d.C.)
| | - Lucas Pires Guarnier
- Department of Genetic, Ribeirão Preto Medical School, University of São Paulo—USP, Ribeirão Preto 14040-904, Brazil;
| | | | | | - Marco Aurélio Lucio
- Graduate Program in Environment and Regional Development, University of Western São Paulo, Presidente Prudente 19050-920, Brazil;
| | - Mateus Vidigal de Castro
- Human Genome and Stem Cell Research Center, Institute of Biosciences, University of São Paulo—USP, São Paulo 01246-904, Brazil; (L.G.M.); (M.V.d.C.)
| | - Marlon Lemos Dias
- Precision Medicine Research Center, Carlos Chagas Filho Biophysics Institute, Federal University of Rio de Janeiro—UFRJ, Rio de Janeiro 21941-630, Brazil;
| | | | - João Tadeu Ribeiro-Paes
- Department of Genetic, Ribeirão Preto Medical School, University of São Paulo—USP, Ribeirão Preto 14040-904, Brazil;
- Laboratory of Genetics and Cell Therapy (GenTe Cel), Department of Biotechnology, São Paulo State University—UNESP, Assis 19806-900, Brazil
| |
Collapse
|
27
|
Liu H, Mei H, Jiang H, Jiang L, Lin K, Jiang M, Ding N, Li X, Gao Z, Liu B, Lin W, Li J, Zhou J. Bioprinted Symbiotic Dressings: A Lichen-Inspired Approach to Diabetic Wound Healing with Enhanced Bioactivity and Structural Integrity. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2407105. [PMID: 39663708 DOI: 10.1002/smll.202407105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Revised: 11/13/2024] [Indexed: 12/13/2024]
Abstract
Providing oxygen and preventing infection at wound sites are effective ways to heal diabetic chronic wounds. Inspired by natural lichens, a bioprinted biogenic hydrogel (BBH) containing microalgae and probiotics is developed for diabetic chronic wound therapeutics, which offers prolonged biogenetic oxygen supply by microalgae and infection inhibition by probiotics. The rational design of symbiotic BBH with customizable structure and microorganism composition enhances wound resilience against elevated glucose levels and hypoxia, leading to the increased migration ability of fibroblasts and the angiogenic potential of human umbilical vein endothelial cells. Notably, BBH-treated diabetic wounds exhibit dense vascular distribution, reduced hypoxia levels and inflammatory responses, and enhanced epithelial differentiation and keratinization abilities. Consequently, the BBH achieves rapid tissue repairing within 3 d and restores approximately 90% of the whole skin structure within 12 d. This work presents an engineered platform for regulating biological microenvironment of diabetic wounds and provides insights for developing bioprinted hybrid microorganism systems.
Collapse
Affiliation(s)
- Hai Liu
- College of Biomass Science and Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
| | - Hongxiang Mei
- College of Biomass Science and Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Hejin Jiang
- College of Biomass Science and Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
- Key Laboratory of Leather Chemistry and Engineering of Ministry of Education, National Engineering Laboratory for Clean Technology of Leather Manufacture, Sichuan University, Chengdu, 610065, China
| | - Linli Jiang
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Kaifeng Lin
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Minwen Jiang
- College of Biomass Science and Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
| | - Ning Ding
- College of Biomass Science and Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
| | - Xiaojie Li
- College of Biomass Science and Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
| | - Ziqi Gao
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Bin Liu
- Shenzhen Key Laboratory of Marine Microbiome Engineering, Shenzhen Key Laboratory of Food Nutrition and Health, Institute for Advanced Study, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, 518060, China
| | - Wei Lin
- College of Biomass Science and Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
- Key Laboratory of Leather Chemistry and Engineering of Ministry of Education, National Engineering Laboratory for Clean Technology of Leather Manufacture, Sichuan University, Chengdu, 610065, China
| | - Juan Li
- College of Biomass Science and Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
- State Key Laboratory of Oral Diseases & National Center for Stomatology & National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University, Chengdu, 610041, China
| | - Jiajing Zhou
- College of Biomass Science and Engineering, West China Hospital of Stomatology, Sichuan University, Chengdu, 610065, China
- Key Laboratory of Leather Chemistry and Engineering of Ministry of Education, National Engineering Laboratory for Clean Technology of Leather Manufacture, Sichuan University, Chengdu, 610065, China
| |
Collapse
|
28
|
Zhang Y, Qi F, Chen P, Liu BF, Li Y. Spatially defined microenvironment for engineering organoids. BIOPHYSICS REVIEWS 2024; 5:041302. [PMID: 39679203 PMCID: PMC11646138 DOI: 10.1063/5.0198848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/19/2024] [Accepted: 10/01/2024] [Indexed: 12/17/2024]
Abstract
In the intricately defined spatial microenvironment, a single fertilized egg remarkably develops into a conserved and well-organized multicellular organism. This observation leads us to hypothesize that stem cells or other seed cell types have the potential to construct fully structured and functional tissues or organs, provided the spatial cues are appropriately configured. Current organoid technology, however, largely depends on spontaneous growth and self-organization, lacking systematic guided intervention. As a result, the structures replicated in vitro often emerge in a disordered and sparse manner during growth phases. Although existing organoids have made significant contributions in many aspects, such as advancing our understanding of development and pathogenesis, aiding personalized drug selection, as well as expediting drug development, their potential in creating large-scale implantable tissue or organ constructs, and constructing multicomponent microphysiological systems, together with functioning at metabolic levels remains underutilized. Recent discoveries have demonstrated that the spatial definition of growth factors not only induces directional growth and migration of organoids but also leads to the formation of assembloids with multiple regional identities. This opens new avenues for the innovative engineering of higher-order organoids. Concurrently, the spatial organization of other microenvironmental cues, such as physical stresses, mechanical loads, and material composition, has been minimally explored. This review delves into the burgeoning field of organoid engineering with a focus on potential spatial microenvironmental control. It offers insight into the molecular principles, expected outcomes, and potential applications, envisioning a future perspective in this domain.
Collapse
Affiliation(s)
- Yilan Zhang
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Fukang Qi
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Peng Chen
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Bi-Feng Liu
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yiwei Li
- The Key Laboratory for Biomedical Photonics of MOE at Wuhan National Laboratory for Optoelectronics-Hubei Bioinformatics and Molecular Imaging Key Laboratory, Department of Biomedical Engineering, College of Life Science and Technology, Huazhong University of Science and Technology, Wuhan 430074, China
| |
Collapse
|
29
|
Lee SJ, Jeong W, Atala A. 3D Bioprinting for Engineered Tissue Constructs and Patient-Specific Models: Current Progress and Prospects in Clinical Applications. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2408032. [PMID: 39420757 PMCID: PMC11875024 DOI: 10.1002/adma.202408032] [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: 06/05/2024] [Revised: 09/24/2024] [Indexed: 10/19/2024]
Abstract
Advancements in bioprinting technology are driving the creation of complex, functional tissue constructs for use in tissue engineering and regenerative medicine. Various methods, including extrusion, jetting, and light-based bioprinting, have their unique advantages and drawbacks. Over the years, researchers and industry leaders have made significant progress in enhancing bioprinting techniques and materials, resulting in the production of increasingly sophisticated tissue constructs. Despite this progress, challenges still need to be addressed in achieving clinically relevant, human-scale tissue constructs, presenting a hurdle to widespread clinical translation. However, with ongoing interdisciplinary research and collaboration, the field is rapidly evolving and holds promise for personalized medical interventions. Continued development and refinement of bioprinting technologies have the potential to address complex medical needs, enabling the development of functional, transplantable tissues and organs, as well as advanced in vitro tissue models.
Collapse
Affiliation(s)
| | | | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27157, United States
| |
Collapse
|
30
|
Kadotani A, Hayase G, Yoshino D. Geometrically engineered organoid units and their assembly for pre-construction of organ structures. APL Bioeng 2024; 8:046112. [PMID: 39606711 PMCID: PMC11602216 DOI: 10.1063/5.0222866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 11/11/2024] [Indexed: 11/29/2024] Open
Abstract
Regenerative medicine is moving from the nascent to the transitional stage as researchers are actively engaged in creating mini-organs from pluripotent stem cells to construct artificial models of physiological and pathological conditions. Currently, mini-organs can express higher-order functions, but their size is limited to the order of a few millimeters. Therefore, one of the ultimate goals of regenerative medicine, "organ replication and transplantation with organoid," remains a major obstacle. Three-dimensional (3D) bioprinting technology is expected to be an innovative breakthrough in this field, but various issues have been raised, such as cell damage, versatility of bioink, and printing time. In this study, we established a method for fabricating, connecting, and assembling organoid units of various shapes independent of cell type, extracellular matrix, and adhesive composition (unit construction method). We also fabricated kidney tissue-like structures using three types of parenchymal and interstitial cells that compose the human kidney and obtained findings suggesting the possibility of crosstalk between the units. This study mainly focuses on methods for reproducing the structure of organs, and there are still issues to be addressed in terms of the expression of their higher-order functions. We anticipate that engineering innovation based on this technique will bring us closer to the realization of highly efficient and rapid fabrication of full-scale organoids that can withstand organ transplantation.
Collapse
Affiliation(s)
- Ayaka Kadotani
- Department of Biomedical Engineering, Graduate School of Engineering, Tokyo University of Agriculture and Technology, 2-24-16 Naka-cho, Koganei, Tokyo 184-8588, Japan
| | - Gen Hayase
- Research Center for Electronic and Optical Materials, National Institute for Materials Science, 1-1 Namiki, Tsukuba, Ibaraki 305-0044, Japan
| | | |
Collapse
|
31
|
Deng X, Qi C, Meng S, Dong H, Wang T, Liu Z, Kong T. All-Aqueous Embedded 3D Printing for Freeform Fabrication of Biomimetic 3D Constructs. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2406825. [PMID: 39520386 DOI: 10.1002/adma.202406825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 10/19/2024] [Indexed: 11/16/2024]
Abstract
All-aqueous embedded 3D printing, which involves extruding inks in an aqueous bath, has emerged as a transformative platform for the freeform fabrication of 3D constructs with precise control. The use of a supporting bath not only enables the printing of arbitrarily designed 3D constructs but also broadens ink selection for various soft matters, advancing the wide application of this technology. This review focuses on recent progress in the freeform preparation of 3D constructs using all-aqueous embedded 3D printing. It begins by discussing the significance of ultralow interfacial tension in all-liquid embedded printing and highlights the fundamental concepts and properties of all-aqueous system. The review then introduces recent advances in all-aqueous embedded 3D printing and clarifies the key factors affecting printing stability and shape fidelity, aiming to guide expansion and assessment of emerging printing systems used for various representative applications. Furthermore, it proposes the potential scope and applications of this technology, including in vitro models, cytomimetic microreactors, and soft ionic electronics. Finally, the review discusses the challenges facing current all-aqueous embedded 3D printing and offers future perspectives on possible improvements and developments.
Collapse
Affiliation(s)
- Xiaokang Deng
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, Guangdong, 518000, 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
| | - Cheng Qi
- College of Mechatronics and Control Engineering, Shenzhen University, Shenzhen, Guangdong, 518000, China
| | - Si Meng
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, Guangdong, 518000, China
| | - Haifeng Dong
- Huizhou Institute of Green Energy and Advanced Materials, Huizhou, Guangdong, 516081, China
| | - Tianfu Wang
- 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
| | - Zhou Liu
- College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, Guangdong, 518000, China
| | - Tiantian Kong
- 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
| |
Collapse
|
32
|
McCormack A, Porcza LM, Leslie NR, Melchels FPW. Gellan gum-based granular gels as suspension media for biofabrication. PLoS One 2024; 19:e0312726. [PMID: 39602414 PMCID: PMC11602023 DOI: 10.1371/journal.pone.0312726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 10/13/2024] [Indexed: 11/29/2024] Open
Abstract
Engineering 3D tissue-like constructs for applications such as regenerative medicine remains a major challenge in biomedical research. Recently, self-healing, viscoplastic fluids have been introduced as suspension media to allow lower viscosity, water-rich bioinks to be printed within them for the fabrication of more biomimetic structures. Here, we present gellan gum granular gels produced through the application of shear during gelation, as a candidate suspension medium. We demonstrate that these granular gels exhibit viscoplasticity over a wide range of temperatures, permitting their use for 3D bioprinting of filaments and droplets at low (4°C) as well as physiological temperatures. These granular gels exhibit very low yield stresses (down to 0.4 Pa) which facilitated printing at print speeds up to 60 mm.s-1. Furthermore, we demonstrate the printing of cell-laden droplets maintained over 7 days to show the potential for multiple days of cell culture, as well as the fabrication of hydrogel features within a crosslinkable version of the suspension medium containing granular gellan gum and gelatine-methacryloyl. The combination of ease of preparation, high printing speed, wide temperature tolerance, and crosslinkability makes this gellan gum sheared through cooling-induced gelation an attractive candidate for suspended biofabrication.
Collapse
Affiliation(s)
- Andrew McCormack
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, United Kingdom
| | - Laura M. Porcza
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, United Kingdom
| | - Nicholas R. Leslie
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, United Kingdom
| | - Ferry P. W. Melchels
- Institute of Biological Chemistry, Biophysics and Bioengineering, School of Engineering and Physical Sciences, Heriot-Watt University, Edinburgh, United Kingdom
- Future Industries Institute, University of South Australia Mawson Lakes, Adelaide, Australia
| |
Collapse
|
33
|
Al Monla R, Daien V, Michon F. Advanced bioengineering strategies broaden the therapeutic landscape for corneal failure. Front Bioeng Biotechnol 2024; 12:1480772. [PMID: 39605752 PMCID: PMC11598527 DOI: 10.3389/fbioe.2024.1480772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Accepted: 10/30/2024] [Indexed: 11/29/2024] Open
Abstract
The cornea acts as the eye foremost protective layer and is essential for its focusing power. Corneal blindness may arise from physical trauma or conditions like dystrophies, keratitis, keratoconus, or ulceration. While conventional treatments involve medical therapies and donor allografts-sometimes supplemented with keratoprostheses-these options are not suitable for all corneal defects. Consequently, the development of bioartificial corneal tissue has emerged as a critical research area, aiming to address the global shortage of human cornea donors. Bioengineered corneas hold considerable promise as substitutes, with the potential to replace either specific layers or the entire thickness of damaged corneas. This review first delves into the structural anatomy of the human cornea, identifying key attributes necessary for successful corneal tissue bioengineering. It then examines various corneal pathologies, current treatments, and their limitations. Finally, the review outlines the primary approaches in corneal tissue engineering, exploring cell-free, cell-based, and scaffold-based options as three emerging strategies to address corneal failure.
Collapse
Affiliation(s)
- Reem Al Monla
- Institute for Neurosciences of Montpellier, INSERM, University of Montpellier, Montpellier, France
| | - Vincent Daien
- Department of Ophthalmology, Gui de Chauliac Hospital, Montpellier, France
- Sydney Medical School, The Save Sight Institute, The University of Sydney, Sydney, NSW, Australia
| | - Frederic Michon
- Institute for Neurosciences of Montpellier, INSERM, University of Montpellier, Montpellier, France
- Department of Ophthalmology, Gui de Chauliac Hospital, Montpellier, France
| |
Collapse
|
34
|
Shuai Y, Zheng M, Kundu SC, Mao C, Yang M. Bioengineered Silk Protein-Based 3D In Vitro Models for Tissue Engineering and Drug Development: From Silk Matrix Properties to Biomedical Applications. Adv Healthc Mater 2024; 13:e2401458. [PMID: 39009465 DOI: 10.1002/adhm.202401458] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2024] [Revised: 06/22/2024] [Indexed: 07/17/2024]
Abstract
3D in vitro model has emerged as a valuable tool for studying tissue development, drug screening, and disease modeling. 3D systems can accurately replicate tissue microstructures and physiological features, mirroring the in vivo microenvironment departing from conventional 2D cell cultures. Various 3D in vitro models utilizing biomacromolecules like collagen and synthetic polymers have been developed to meet diverse research needs and address the complex challenges of contemporary research. Silk proteins, bearing structural and functional similarities to collagen, have been increasingly employed to construct advanced 3D in vitro systems, surpassing the limitations of 2D cultures. This review examines silk proteins' composition, structure, properties, and functions, elucidating their role in 3D in vitro models. Furthermore, recent advances in biomedical applications involving silk-based organoid models are discussed. In particular, the unique physiological attributes of silk matrix constituents in in vitro tissue constructs are highlighted, providing a meticulous evaluation of their importance. Additionally, it outlines the current research hurdles and complexities while contemplating future avenues, thereby paving the way for developing complex and biomimetic silk protein-based microtissues.
Collapse
Affiliation(s)
- Yajun Shuai
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Meidan Zheng
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Hangzhou, 310058, P. R. China
| | - Subhas C Kundu
- 3B's Research Group, I3Bs - Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Zona Industrial da Gandra, Barco, Guimarães, 4805-017, Portugal
| | - Chuanbin Mao
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Sha Tin, Hong Kong, SAR, P. R. China
| | - Mingying Yang
- Key Laboratory of Silkworm and Bee Resource Utilization and Innovation of Zhejiang Province, Institute of Applied Bioresource Research, College of Animal Science, Zhejiang University, Hangzhou, 310058, P. R. China
| |
Collapse
|
35
|
Murphy JF, Lavelle M, Asciak L, Burdis R, Levis HJ, Ligorio C, McGuire J, Polleres M, Smith PO, Tullie L, Uribe-Gomez J, Chen B, Dawson JI, Gautrot JE, Hooper NM, Kelly DJ, Li VSW, Mata A, Pandit A, Phillips JB, Shu W, Stevens MM, Williams RL, Armstrong JPK, Huang YYS. Biofabrication and biomanufacturing in Ireland and the UK. Biodes Manuf 2024; 7:825-856. [PMID: 39650072 PMCID: PMC11618173 DOI: 10.1007/s42242-024-00316-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/11/2024] [Indexed: 12/11/2024]
Abstract
As we navigate the transition from the Fourth to the Fifth Industrial Revolution, the emerging fields of biomanufacturing and biofabrication are transforming life sciences and healthcare. These sectors are benefiting from a synergy of synthetic and engineering biology, sustainable manufacturing, and integrated design principles. Advanced techniques such as 3D bioprinting, tissue engineering, directed assembly, and self-assembly are instrumental in creating biomimetic scaffolds, tissues, organoids, medical devices, and biohybrid systems. The field of biofabrication in the United Kingdom and Ireland is emerging as a pivotal force in bioscience and healthcare, propelled by cutting-edge research and development. Concentrating on the production of biologically functional products for use in drug delivery, in vitro models, and tissue engineering, research institutions across these regions are dedicated to innovating healthcare solutions that adhere to ethical standards while prioritising sustainability, affordability, and healthcare system benefits. Graphic abstract
Collapse
Affiliation(s)
- Jack F. Murphy
- Department of Engineering, University of Cambridge, Cambridge, CB2 1PZ UK
| | - Martha Lavelle
- Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS1 3NY UK
| | - Lisa Asciak
- Department of Biomedical Engineering, University of Strathclyde, Glasgow, G4 0NW UK
| | - Ross Burdis
- Department of Materials, Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ UK
- Department of Bioengineering, Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ UK
| | - Hannah J. Levis
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX UK
| | - Cosimo Ligorio
- Biodiscovery Institute, University of Nottingham, Nottingham, NG7 2RD UK
- Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham, NG7 2RD UK
| | - Jamie McGuire
- Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, S016 6YD UK
| | - Marlene Polleres
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT UK
| | - Poppy O. Smith
- UCL Centre for Nerve Engineering, Department of Pharmacology, UCL School of Pharmacy, University College London, London, WC1N 1AX UK
| | - Lucinda Tullie
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, NW1 1AT UK
| | - Juan Uribe-Gomez
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, H91 W2TY Ireland
| | - Biqiong Chen
- School of Mechanical and Aerospace Engineering, Queen’s University Belfast, Belfast, BT9 5AH UK
| | - Jonathan I. Dawson
- Centre for Human Development, Stem Cells and Regeneration, Human Development and Health, Faculty of Medicine, University of Southampton, Southampton, S016 6YD UK
| | - Julien E. Gautrot
- School of Engineering and Materials Science, Queen Mary University of London, London, E1 4NS UK
| | - Nigel M. Hooper
- Division of Neuroscience, School of Biological Sciences, Faculty of Biology, Medicine and Health, University of Manchester, Manchester, M13 9PT UK
- Geoffrey Jefferson Brain Research Centre, Manchester Academic Health Science Centre, Northern Care Alliance and University of Manchester, Manchester, M13 9PL UK
| | - Daniel J. Kelly
- Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland
- Department of Mechanical, Manufacturing and Biomedical Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland
- Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin 2, Ireland
- Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, Dublin, D02 H903 Ireland
| | - Vivian S. W. Li
- Stem Cell and Cancer Biology Laboratory, The Francis Crick Institute, London, NW1 1AT UK
| | - Alvaro Mata
- Biodiscovery Institute, University of Nottingham, Nottingham, NG7 2RD UK
- Department of Chemical and Environmental Engineering, University of Nottingham, Nottingham, NG7 2RD UK
- School of Pharmacy, University of Nottingham, Nottingham, NG7 2RD UK
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, H91 W2TY Ireland
| | - James B. Phillips
- UCL Centre for Nerve Engineering, Department of Pharmacology, UCL School of Pharmacy, University College London, London, WC1N 1AX UK
| | - Wenmiao Shu
- Department of Biomedical Engineering, University of Strathclyde, Glasgow, G4 0NW UK
| | - Molly M. Stevens
- Department of Materials, Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ UK
- Department of Bioengineering, Institute of Biomedical Engineering, Imperial College London, London, SW7 2AZ UK
- Department of Physiology, Anatomy and Genetics, Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, OX1 3QU UK
- Department of Engineering Science, Kavli Institute for Nanoscience Discovery, University of Oxford, Oxford, OX1 3QU UK
| | - Rachel L. Williams
- Department of Eye and Vision Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX UK
| | - James P. K. Armstrong
- Department of Translational Health Sciences, Bristol Medical School, University of Bristol, Bristol, BS1 3NY UK
| | | |
Collapse
|
36
|
Luo Y, Xu R, Hu Z, Ni R, Zhu T, Zhang H, Zhu Y. Gel-Based Suspension Medium Used in 3D Bioprinting for Constructing Tissue/Organ Analogs. Gels 2024; 10:644. [PMID: 39451297 PMCID: PMC11507232 DOI: 10.3390/gels10100644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 10/05/2024] [Accepted: 10/07/2024] [Indexed: 10/26/2024] Open
Abstract
Constructing tissue/organ analogs with natural structures and cell types in vitro offers a valuable strategy for the in situ repair of damaged tissues/organs. Three-dimensional (3D) bioprinting is a flexible method for fabricating these analogs. However, extrusion-based 3D bioprinting faces the challenge of balancing the use of soft bioinks with the need for high-fidelity geometric shapes. To address these challenges, recent advancements have introduced various suspension mediums based on gelatin, agarose, and gellan gum microgels. The emergence of these gel-based suspension mediums has significantly advanced the fabrication of tissue/organ constructs using 3D bioprinting. They effectively stabilize and support soft bioinks, enabling the formation of complex spatial geometries. Moreover, they provide a stable, cell-friendly environment that maximizes cell viability during the printing process. This minireview will summarize the properties, preparation methods, and potential applications of gel-based suspension mediums in constructing tissue/organ analogs, while also addressing current challenges and providing an outlook on the future of 3D bioprinting.
Collapse
Affiliation(s)
- Yang Luo
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Rong Xu
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Zeming Hu
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Renhao Ni
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Tong Zhu
- Health Science Center, Ningbo University, Ningbo 315211, China
| | - Hua Zhang
- Health Science Center, Ningbo University, Ningbo 315211, China
- Research Institute of Smart Medicine and Biological Engineering, Ningbo University, Ningbo 315211, China
| | - Yabin Zhu
- Health Science Center, Ningbo University, Ningbo 315211, China
| |
Collapse
|
37
|
Graham AJ, Khoo MW, Srivastava V, Viragova S, Kim H, Parekh K, Hennick KM, Bird M, Goldhammer N, Yu JZ, Morley CD, Lebel P, Kumar S, Rosenbluth JM, Nowakowski TJ, Klein O, Gómez-Sjöberg R, Gartner ZJ. MAGIC matrices: freeform bioprinting materials to support complex and reproducible organoid morphogenesis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.02.01.578324. [PMID: 38370663 PMCID: PMC10871257 DOI: 10.1101/2024.02.01.578324] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/20/2024]
Abstract
Organoids are powerful models of tissue physiology, yet their applications remain limited due to their relatively simple morphology and high organoid-to-organoid structural variability. To address these limitations we developed a soft, composite yield-stress extracellular matrix that supports optimal organoid morphogenesis following freeform 3D bioprinting of cell slurries at tissue-like densities. The material is designed with two temperature regimes: at 4 °C it exhibits reversible yield-stress behavior to support long printing times without compromising cell viability. When transferred to cell culture at 37 °C, the material cross-links and exhibits similar viscoelasticity and plasticity to basement membrane extracts such as Matrigel. We first characterize the rheological properties of MAGIC matrices that optimize organoid morphogenesis, including low stiffness and high stress relaxation. Next, we combine this material with a custom piezoelectric printhead that allows more reproducible and robust self-organization from uniform and spatially organized tissue "seeds." We apply MAGIC matrix bioprinting for high-throughput generation of intestinal, mammary, vascular, salivary gland, and brain organoid arrays that are structurally similar to those grown in pure Matrigel, but exhibit dramatically improved homogeneity in organoid size, shape, maturation time, and efficiency of morphogenesis. The flexibility of this method and material enabled fabrication of fully 3D microphysiological systems, including perfusable organoid tubes that experience cyclic 3D strain in response to pressurization. Furthermore, the reproducibility of organoid structure increased the statistical power of a drug response assay by up to 8 orders-of-magnitude for a given number of comparisons. Combined, these advances lay the foundation for the efficient fabrication of complex tissue morphologies by canalizing their self-organization in both space and time.
Collapse
Affiliation(s)
- Austin J. Graham
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
- Chan Zuckerberg Biohub SF, San Francisco, CA
| | | | - Vasudha Srivastava
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
| | - Sara Viragova
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA
| | - Honesty Kim
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
- Chan Zuckerberg Biohub SF, San Francisco, CA
| | - Kavita Parekh
- Department of Bioengineering, University of California Berkeley, Berkeley, CA
| | - Kelsey M. Hennick
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Malia Bird
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
| | - Nadine Goldhammer
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Jie Zeng Yu
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Cameron D. Morley
- Department of Bioengineering, University of California Berkeley, Berkeley, CA
| | - Paul Lebel
- Chan Zuckerberg Biohub SF, San Francisco, CA
| | - Sanjay Kumar
- Department of Bioengineering, University of California Berkeley, Berkeley, CA
- Department of Chemical and Biomolecular Engineering, University of California Berkeley, Berkeley, CA
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA
| | - Jennifer M. Rosenbluth
- Chan Zuckerberg Biohub SF, San Francisco, CA
- Department of Medicine, University of California San Francisco, San Francisco, CA
| | - Tomasz J. Nowakowski
- Weill Institute for Neurosciences, University of California San Francisco, San Francisco, CA
| | - Ophir Klein
- Department of Orofacial Sciences, University of California San Francisco, San Francisco, CA
- Department of Pediatrics, Cedars-Sinai Guerin Children’s, Los Angeles, CA
| | | | - Zev J. Gartner
- Department of Pharmaceutical Chemistry, University of California San Francisco, San Francisco, CA
- Chan Zuckerberg Biohub SF, San Francisco, CA
- Department of Bioengineering and Therapeutic Sciences, University of California San Francisco, San Francisco, CA
- Center for Cellular Construction, University of California San Francisco, San Francisco, CA
| |
Collapse
|
38
|
Miklosic G, Ferguson SJ, D'Este M. Engineering complex tissue-like microenvironments with biomaterials and biofabrication. Trends Biotechnol 2024; 42:1241-1257. [PMID: 38658198 DOI: 10.1016/j.tibtech.2024.03.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Revised: 03/19/2024] [Accepted: 03/20/2024] [Indexed: 04/26/2024]
Abstract
Advances in tissue engineering for both system modeling and organ regeneration depend on embracing and recapitulating the target tissue's functional and structural complexity. Microenvironmental features such as anisotropy, heterogeneity, and other biochemical and mechanical spatiotemporal cues are essential in regulating tissue development and function. Novel biofabrication strategies and innovative biomaterial design have emerged as promising tools to better reproduce such features. These facilitate a transition towards high-fidelity biomimetic structures, offering opportunities for a deeper understanding of tissue function and the development of superior therapies. In this review, we explore some of the key structural and compositional aspects of tissues, lay out how to achieve similar outcomes with current fabrication strategies, and identify the main challenges and promising avenues for future research.
Collapse
Affiliation(s)
- Gregor Miklosic
- AO Research Institute Davos, Davos, Switzerland; Institute for Biomechanics, ETH Zurich, Zurich, Switzerland
| | | | | |
Collapse
|
39
|
Soliman BG, Nguyen AK, Gooding JJ, Kilian KA. Advancing Synthetic Hydrogels through Nature-Inspired Materials Chemistry. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2404235. [PMID: 38896849 PMCID: PMC11486603 DOI: 10.1002/adma.202404235] [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: 03/23/2024] [Revised: 05/25/2024] [Indexed: 06/21/2024]
Abstract
Synthetic extracellular matrix (ECM) mimics that can recapitulate the complex biochemical and mechanical nature of native tissues are needed for advanced models of development and disease. Biomedical research has heavily relied on the use of animal-derived biomaterials, which is now impeding their translational potential and convoluting the biological insights gleaned from in vitro tissue models. Natural hydrogels have long served as a convenient and effective cell culture tool, but advances in materials chemistry and fabrication techniques now present promising new avenues for creating xenogenic-free ECM substitutes appropriate for organotypic models and microphysiological systems. However, significant challenges remain in creating synthetic matrices that can approximate the structural sophistication, biochemical complexity, and dynamic functionality of native tissues. This review summarizes key properties of the native ECM, and discusses recent approaches used to systematically decouple and tune these properties in synthetic matrices. The importance of dynamic ECM mechanics, such as viscoelasticity and matrix plasticity, is also discussed, particularly within the context of organoid and engineered tissue matrices. Emerging design strategies to mimic these dynamic mechanical properties are reviewed, such as multi-network hydrogels, supramolecular chemistry, and hydrogels assembled from biological monomers.
Collapse
Affiliation(s)
- Bram G Soliman
- School of Chemistry, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales Sydney, Sydney, NSW, 2052, Australia
| | - Ashley K Nguyen
- School of Chemistry, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales Sydney, Sydney, NSW, 2052, Australia
| | - J Justin Gooding
- School of Chemistry, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales Sydney, Sydney, NSW, 2052, Australia
| | - Kristopher A Kilian
- School of Chemistry, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Centre for NanoMedicine, University of New South Wales Sydney, Sydney, NSW, 2052, Australia
- School of Materials Science and Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| |
Collapse
|
40
|
Liu S, Chen Y, Wang Z, Liu M, Zhao Y, Tan Y, Qu Z, Du L, Wu C. The cutting-edge progress in bioprinting for biomedicine: principles, applications, and future perspectives. MedComm (Beijing) 2024; 5:e753. [PMID: 39314888 PMCID: PMC11417428 DOI: 10.1002/mco2.753] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 09/03/2024] [Accepted: 09/03/2024] [Indexed: 09/25/2024] Open
Abstract
Bioprinting is a highly promising application area of additive manufacturing technology that has been widely used in various fields, including tissue engineering, drug screening, organ regeneration, and biosensing. Its primary goal is to produce biomedical products such as artificial implant scaffolds, tissues and organs, and medical assistive devices through software-layered discrete and numerical control molding. Despite its immense potential, bioprinting technology still faces several challenges. It requires concerted efforts from researchers, engineers, regulatory bodies, and industry stakeholders are principal to overcome these challenges and unlock the full potential of bioprinting. This review systematically discusses bioprinting principles, applications, and future perspectives while also providing a topical overview of research progress in bioprinting over the past two decades. The most recent advancements in bioprinting are comprehensively reviewed here. First, printing techniques and methods are summarized along with advancements related to bioinks and supporting structures. Second, interesting and representative cases regarding the applications of bioprinting in tissue engineering, drug screening, organ regeneration, and biosensing are introduced in detail. Finally, the remaining challenges and suggestions for future directions of bioprinting technology are proposed and discussed. Bioprinting is one of the most promising application areas of additive manufacturing technology that has been widely used in various fields. It aims to produce biomedical products such as artificial implant scaffolds, tissues and organs, and medical assistive devices. This review systematically discusses bioprinting principles, applications, and future perspectives, which provides a topical description of the research progress of bioprinting.
Collapse
Affiliation(s)
- Shuge Liu
- Department of BiophysicsInstitute of Medical EngineeringSchool of Basic Medical SciencesHealth Science CenterXi'an Jiaotong UniversityXi'anShaanxiChina
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University)Ministry of Education of ChinaXi'anShaanxiChina
| | - Yating Chen
- Department of BiophysicsInstitute of Medical EngineeringSchool of Basic Medical SciencesHealth Science CenterXi'an Jiaotong UniversityXi'anShaanxiChina
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University)Ministry of Education of ChinaXi'anShaanxiChina
| | - Zhiyao Wang
- Department of BiophysicsInstitute of Medical EngineeringSchool of Basic Medical SciencesHealth Science CenterXi'an Jiaotong UniversityXi'anShaanxiChina
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University)Ministry of Education of ChinaXi'anShaanxiChina
| | - Minggao Liu
- Department of BiophysicsInstitute of Medical EngineeringSchool of Basic Medical SciencesHealth Science CenterXi'an Jiaotong UniversityXi'anShaanxiChina
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University)Ministry of Education of ChinaXi'anShaanxiChina
| | - Yundi Zhao
- Department of BiophysicsInstitute of Medical EngineeringSchool of Basic Medical SciencesHealth Science CenterXi'an Jiaotong UniversityXi'anShaanxiChina
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University)Ministry of Education of ChinaXi'anShaanxiChina
| | - Yushuo Tan
- Department of BiophysicsInstitute of Medical EngineeringSchool of Basic Medical SciencesHealth Science CenterXi'an Jiaotong UniversityXi'anShaanxiChina
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University)Ministry of Education of ChinaXi'anShaanxiChina
| | - Zhan Qu
- Department of BiophysicsInstitute of Medical EngineeringSchool of Basic Medical SciencesHealth Science CenterXi'an Jiaotong UniversityXi'anShaanxiChina
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University)Ministry of Education of ChinaXi'anShaanxiChina
| | - Liping Du
- Department of BiophysicsInstitute of Medical EngineeringSchool of Basic Medical SciencesHealth Science CenterXi'an Jiaotong UniversityXi'anShaanxiChina
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University)Ministry of Education of ChinaXi'anShaanxiChina
| | - Chunsheng Wu
- Department of BiophysicsInstitute of Medical EngineeringSchool of Basic Medical SciencesHealth Science CenterXi'an Jiaotong UniversityXi'anShaanxiChina
- Key Laboratory of Environment and Genes Related to Diseases (Xi'an Jiaotong University)Ministry of Education of ChinaXi'anShaanxiChina
| |
Collapse
|
41
|
Zhu T, Hu Y, Cui H, Cui H. 3D Multispheroid Assembly Strategies towards Tissue Engineering and Disease Modeling. Adv Healthc Mater 2024; 13:e2400957. [PMID: 38924326 DOI: 10.1002/adhm.202400957] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 06/21/2024] [Indexed: 06/28/2024]
Abstract
Cell spheroids (esp. organoids) as 3D culture platforms are popular models for representing cell-cell and cell-extracellular matrix (ECM) interactions, bridging the gap between 2D cell cultures and natural tissues. 3D cell models with spatially organized multiple cell types are preferred for gaining comprehensive insights into tissue pathophysiology and constructing in vitro tissues and disease models because of the complexities of natural tissues. In recent years, an assembly strategy using cell spheroids (or organoids) as living building blocks has been developed to construct complex 3D tissue models with spatial organization. Here, a comprehensive overview of recent advances in multispheroid assembly studies is provided. The different mechanisms of the multispheroid assembly techniques, i.e., automated directed assembly, noncontact remote assembly, and programmed self-assembly, are introduced. The processing steps, advantages, and technical limitations of the existing methodologies are summarized. Applications of the multispheroid assembly strategies in disease modeling, drug screening, tissue engineering, and organogenesis are reviewed. Finally, this review concludes by emphasizing persistent issues and future perspectives, encouraging researchers to adopt multispheroid assembly techniques for generating advanced 3D cell models that better resemble real tissues.
Collapse
Affiliation(s)
- Tong Zhu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Yan Hu
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Haitao Cui
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| | - Haijun Cui
- Key Laboratory of Biorheological Science and Technology, Ministry of Education, College of Bioengineering, Chongqing University, Chongqing, 400044, China
| |
Collapse
|
42
|
Hall GN, Fan Y, Viellerobe B, Iazzolino A, Dimopoulos A, Poiron C, Clapies A, Luyten FP, Guillemot F, Papantoniou I. Laser-assisted bioprinting of targeted cartilaginous spheroids for high density bottom-up tissue engineering. Biofabrication 2024; 16:045029. [PMID: 39136309 DOI: 10.1088/1758-5090/ad6e1a] [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: 04/03/2024] [Accepted: 08/12/2024] [Indexed: 08/23/2024]
Abstract
Multicellular spheroids such as microtissues and organoids have demonstrated great potential for tissue engineering applications in recent years as these 3D cellular units enable improved cell-cell and cell-matrix interactions. Current bioprinting processes that use multicellular spheroids as building blocks have demonstrated limited control on post printing distribution of cell spheroids or moderate throughput and printing efficiency. In this work, we presented a laser-assisted bioprinting approach able to transfer multicellular spheroids as building blocks for larger tissue structures. Cartilaginous multicellular spheroids formed by human periosteum derived cells (hPDCs) were successfully bioprinted possessing high viability and the capacity to undergo chondrogenic differentiation post printing. Smaller hPDC spheroids with diameters ranging from ∼100 to 150µm were successfully bioprinted through the use of laser-induced forward transfer method (LIFT) however larger spheroids constituted a challenge. For this reason a novel alternative approach was developed termed as laser induced propulsion of mesoscopic objects (LIPMO) whereby we were able to bioprint spheroids of up to 300µm. Moreover, we combined the bioprinting process with computer aided image analysis demonstrating the capacity to 'target and shoot', through automated selection, multiple large spheroids in a single sequence. By taking advantage of target and shoot system, multilayered constructs containing high density cell spheroids were fabricated.
Collapse
Affiliation(s)
- Gabriella Nilsson Hall
- Prometheus Division of Skeletal Tissue Engineering, KU Leuven, O&N1, Herestraat 49, PB 813, 3000 Leuven, Belgium
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, O&N1, Herestraat 49, PB 813, 3000 Leuven, Belgium
| | - Yuchao Fan
- Prometheus Division of Skeletal Tissue Engineering, KU Leuven, O&N1, Herestraat 49, PB 813, 3000 Leuven, Belgium
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, O&N1, Herestraat 49, PB 813, 3000 Leuven, Belgium
| | - Bertrand Viellerobe
- POIETIS, Bioparc Bordeaux Métropole, 27 allée Charles Darwin, Pessac 33600, France
| | - Antonio Iazzolino
- POIETIS, Bioparc Bordeaux Métropole, 27 allée Charles Darwin, Pessac 33600, France
| | - Andreas Dimopoulos
- Prometheus Division of Skeletal Tissue Engineering, KU Leuven, O&N1, Herestraat 49, PB 813, 3000 Leuven, Belgium
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, O&N1, Herestraat 49, PB 813, 3000 Leuven, Belgium
| | - Claire Poiron
- POIETIS, Bioparc Bordeaux Métropole, 27 allée Charles Darwin, Pessac 33600, France
| | - Aude Clapies
- POIETIS, Bioparc Bordeaux Métropole, 27 allée Charles Darwin, Pessac 33600, France
| | - Frank P Luyten
- Prometheus Division of Skeletal Tissue Engineering, KU Leuven, O&N1, Herestraat 49, PB 813, 3000 Leuven, Belgium
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, O&N1, Herestraat 49, PB 813, 3000 Leuven, Belgium
| | - Fabien Guillemot
- POIETIS, Bioparc Bordeaux Métropole, 27 allée Charles Darwin, Pessac 33600, France
| | - Ioannis Papantoniou
- Prometheus Division of Skeletal Tissue Engineering, KU Leuven, O&N1, Herestraat 49, PB 813, 3000 Leuven, Belgium
- Department of Development and Regeneration, Skeletal Biology and Engineering Research Center, KU Leuven, O&N1, Herestraat 49, PB 813, 3000 Leuven, Belgium
- Institute of Chemical Engineering Sciences, Foundation for Research and Technology-Hellas, Stadiou 26504, Platani, Patras, Greece
| |
Collapse
|
43
|
Zhao X, Li N, Zhang Z, Hong J, Zhang X, Hao Y, Wang J, Xie Q, Zhang Y, Li H, Liu M, Zhang P, Ren X, Wang X. Beyond hype: unveiling the Real challenges in clinical translation of 3D printed bone scaffolds and the fresh prospects of bioprinted organoids. J Nanobiotechnology 2024; 22:500. [PMID: 39169401 PMCID: PMC11337604 DOI: 10.1186/s12951-024-02759-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2024] [Accepted: 08/06/2024] [Indexed: 08/23/2024] Open
Abstract
Bone defects pose significant challenges in healthcare, with over 2 million bone repair surgeries performed globally each year. As a burgeoning force in the field of bone tissue engineering, 3D printing offers novel solutions to traditional bone transplantation procedures. However, current 3D-printed bone scaffolds still face three critical challenges in material selection, printing methods, cellular self-organization and co-culture, significantly impeding their clinical application. In this comprehensive review, we delve into the performance criteria that ideal bone scaffolds should possess, with a particular focus on the three core challenges faced by 3D printing technology during clinical translation. We summarize the latest advancements in non-traditional materials and advanced printing techniques, emphasizing the importance of integrating organ-like technologies with bioprinting. This combined approach enables more precise simulation of natural tissue structure and function. Our aim in writing this review is to propose effective strategies to address these challenges and promote the clinical translation of 3D-printed scaffolds for bone defect treatment.
Collapse
Affiliation(s)
- Xiangyu Zhao
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Na Li
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Ziqi Zhang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Jinjia Hong
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Xiaoxuan Zhang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Yujia Hao
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Jia Wang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Qingpeng Xie
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Yuan Zhang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Huifei Li
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Meixian Liu
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Pengfei Zhang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China
| | - Xiuyun Ren
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China.
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China.
| | - Xing Wang
- Shanxi Medical University School and Hospital of Stomatology, Taiyuan, 030001, China.
- Shanxi Province Key Laboratory of Oral Diseases Prevention and New Materials, Taiyuan, 030001, China.
| |
Collapse
|
44
|
Zheng L, Zhan Y, Wang C, Fan Q, Sun D, Li Y, Xiong Y. Technological advances and challenges in constructing complex gut organoid systems. Front Cell Dev Biol 2024; 12:1432744. [PMID: 39206092 PMCID: PMC11349554 DOI: 10.3389/fcell.2024.1432744] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 08/05/2024] [Indexed: 09/04/2024] Open
Abstract
Recent advancements in organoid technology have heralded a transformative era in biomedical research, characterized by the emergence of gut organoids that replicate the structural and functional complexity of the human intestines. These stem cell-derived structures provide a dynamic platform for investigating intestinal physiology, disease pathogenesis, and therapeutic interventions. This model outperforms traditional two-dimensional cell cultures in replicating cell interactions and tissue dynamics. Gut organoids represent a significant leap towards personalized medicine. They provide a predictive model for human drug responses, thereby minimizing reliance on animal models and paving the path for more ethical and relevant research approaches. However, the transition from basic organoid models to more sophisticated, biomimetic systems that encapsulate the gut's multifaceted environment-including its interactions with microbial communities, immune cells, and neural networks-presents significant scientific challenges. This review concentrates on recent technological strides in overcoming these barriers, emphasizing innovative engineering approaches for integrating diverse cell types to replicate the gut's immune and neural components. It also explores the application of advanced fabrication techniques, such as 3D bioprinting and microfluidics, to construct organoids that more accurately replicate human tissue architecture. They provide insights into the intricate workings of the human gut, fostering the development of targeted, effective treatments. These advancements hold promise in revolutionizing disease modeling and drug discovery. Future research directions aim at refining these models further, making them more accessible and scalable for wider applications in scientific inquiry and clinical practice, thus heralding a new era of personalized and predictive medicine.
Collapse
Affiliation(s)
- Longjin Zheng
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, China
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, China
| | - Yang Zhan
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, China
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, China
| | - Chenxuan Wang
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, China
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, China
| | - Qigui Fan
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, China
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, China
| | - Denglong Sun
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, China
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, China
| | - Yingmeng Li
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, China
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, China
| | - Yanxia Xiong
- State Key Laboratory for the Modernization of Classical and Famous Prescriptions of Chinese Medicine, Nanchang, China
- Research and Development Department, Jiangzhong Pharmaceutical Co., Ltd., Nanchang, China
| |
Collapse
|
45
|
Polak R, Zhang ET, Kuo CJ. Cancer organoids 2.0: modelling the complexity of the tumour immune microenvironment. Nat Rev Cancer 2024; 24:523-539. [PMID: 38977835 DOI: 10.1038/s41568-024-00706-6] [Citation(s) in RCA: 48] [Impact Index Per Article: 48.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 05/09/2024] [Indexed: 07/10/2024]
Abstract
The development of neoplasia involves a complex and continuous interplay between malignantly transformed cells and the tumour microenvironment (TME). Cancer immunotherapies targeting the immune TME have been increasingly validated in clinical trials but response rates vary substantially between tumour histologies and are often transient, idiosyncratic and confounded by resistance. Faithful experimental models of the patient-specific tumour immune microenvironment, capable of recapitulating tumour biology and immunotherapy effects, would greatly improve patient selection, target identification and definition of resistance mechanisms for immuno-oncology therapeutics. In this Review, we discuss currently available and rapidly evolving 3D tumour organoid models that capture important immune features of the TME. We highlight diverse opportunities for organoid-based investigations of tumour immunity, drug development and precision medicine.
Collapse
Affiliation(s)
- Roel Polak
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
- Princess Máxima Center for Pediatric Oncology, Utrecht, Netherlands
| | - Elisa T Zhang
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
| | - Calvin J Kuo
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA.
| |
Collapse
|
46
|
Chen A, Wang W, Mao Z, He Y, Chen S, Liu G, Su J, Feng P, Shi Y, Yan C, Lu J. Multimaterial 3D and 4D Bioprinting of Heterogenous Constructs for Tissue Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307686. [PMID: 37737521 DOI: 10.1002/adma.202307686] [Citation(s) in RCA: 27] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 09/06/2023] [Indexed: 09/23/2023]
Abstract
Additive manufacturing (AM), which is based on the principle of layer-by-layer shaping and stacking of discrete materials, has shown significant benefits in the fabrication of complicated implants for tissue engineering (TE). However, many native tissues exhibit anisotropic heterogenous constructs with diverse components and functions. Consequently, the replication of complicated biomimetic constructs using conventional AM processes based on a single material is challenging. Multimaterial 3D and 4D bioprinting (with time as the fourth dimension) has emerged as a promising solution for constructing multifunctional implants with heterogenous constructs that can mimic the host microenvironment better than single-material alternatives. Notably, 4D-printed multimaterial implants with biomimetic heterogenous architectures can provide a time-dependent programmable dynamic microenvironment that can promote cell activity and tissue regeneration in response to external stimuli. This paper first presents the typical design strategies of biomimetic heterogenous constructs in TE applications. Subsequently, the latest processes in the multimaterial 3D and 4D bioprinting of heterogenous tissue constructs are discussed, along with their advantages and challenges. In particular, the potential of multimaterial 4D bioprinting of smart multifunctional tissue constructs is highlighted. Furthermore, this review provides insights into how multimaterial 3D and 4D bioprinting can facilitate the realization of next-generation TE applications.
Collapse
Affiliation(s)
- Annan Chen
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China
| | - Wanying Wang
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
| | - Zhengyi Mao
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
| | - Yunhu He
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
| | - Shiting Chen
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
| | - Guo Liu
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
| | - Jin Su
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China
| | - Pei Feng
- State Key Laboratory of High-Performance Complex Manufacturing, College of Mechanical and Electrical Engineering, Central South University, Changsha, 410083, China
| | - Yusheng Shi
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China
| | - Chunze Yan
- State Key Laboratory of Materials Processing and Die & Mould Technology, School of Materials Science and Engineering, Huazhong University of Science and Technology, Wuhan, 430074, China
- Engineering Research Center of Ceramic Materials for Additive Manufacturing, Ministry of Education, Wuhan, 430074, China
| | - Jian Lu
- Centre for Advanced Structural Materials, Department of Mechanical Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Centre for Advanced Structural Materials, City University of Hong Kong Shenzhen Research Institute, Greater Bay Joint Division, Shenyang National Laboratory for Materials Science, Shenzhen, 518057, China
- CityU-Shenzhen Futian Research Institute, Shenzhen, 518045, China
- Department of Materials Science and Engineering, City University of Hong Kong, Kowloon, Hong Kong, 999077, China
- Hong Kong Branch of National Precious Metals Material Engineering Research, Center (NPMM), City University of Hong Kong, Kowloon, Hong Kong, 999077, China
| |
Collapse
|
47
|
Na DH, Cui S, Fang X, Lee H, Eum SH, Shin YJ, Lim SW, Yang CW, Chung BH. Advancements in Research on Genetic Kidney Diseases Using Human-Induced Pluripotent Stem Cell-Derived Kidney Organoids. Cells 2024; 13:1190. [PMID: 39056771 PMCID: PMC11274677 DOI: 10.3390/cells13141190] [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: 05/14/2024] [Revised: 07/09/2024] [Accepted: 07/11/2024] [Indexed: 07/28/2024] Open
Abstract
Genetic or hereditary kidney disease stands as a pivotal cause of chronic kidney disease (CKD). The proliferation and widespread utilization of DNA testing in clinical settings have notably eased the diagnosis of genetic kidney diseases, which were once elusive but are now increasingly identified in cases previously deemed CKD of unknown etiology. However, despite these diagnostic strides, research into disease pathogenesis and novel drug development faces significant hurdles, chiefly due to the dearth of appropriate animal models and the challenges posed by limited patient cohorts in clinical studies. Conversely, the advent and utilization of human-induced pluripotent stem cells (hiPSCs) offer a promising avenue for genetic kidney disease research. Particularly, the development of hiPSC-derived kidney organoid systems presents a novel platform for investigating various forms of genetic kidney diseases. Moreover, the integration of the CRISPR/Cas9 technique into this system holds immense potential for efficient research on genetic kidney diseases. This review aims to explore the applications of in vitro kidney organoids generated from hiPSCs in the study of diverse genetic kidney diseases. Additionally, it will delve into the limitations of this research platform and outline future perspectives for advancing research in this crucial area.
Collapse
Affiliation(s)
- Do Hyun Na
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sheng Cui
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
| | - Xianying Fang
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
| | - Hanbi Lee
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Sang Hun Eum
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
- Division of Nephrology, Department of Internal Medicine, Incheon St. Mary’s Hospital, The College of Medicine, The Catholic University of Korea, Incheon 21431, Republic of Korea
| | - Yoo Jin Shin
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
| | - Sun Woo Lim
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
| | - Chul Woo Yang
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| | - Byung Ha Chung
- Transplantation Research Center, College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea; (D.H.N.); (S.C.); (X.F.); (H.L.); (S.H.E.); (Y.J.S.); (S.W.L.); (C.W.Y.)
- Division of Nephrology, Department of Internal Medicine, Seoul St. Mary’s Hospital, The College of Medicine, The Catholic University of Korea, Seoul 06591, Republic of Korea
| |
Collapse
|
48
|
Li Y, Cheng S, Shi H, Yuan R, Gao C, Wang Y, Zhang Z, Deng Z, Huang J. 3D embedded bioprinting of large-scale intestine with complex structural organization and blood capillaries. Biofabrication 2024; 16:045001. [PMID: 38914075 DOI: 10.1088/1758-5090/ad5b1b] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2024] [Accepted: 06/24/2024] [Indexed: 06/26/2024]
Abstract
Accurate reproduction of human intestinal structure and functionin vitrois of great significance for understanding the development and disease occurrence of the gut. However, mostin vitrostudies are often confined to 2D models, 2.5D organ chips or 3D organoids, which cannot fully recapitulate the tissue architecture, microenvironment and cell compartmentalization foundin vivo. Herein, a centimeter-scale intestine tissue that contains intestinal features, such as hollow tubular structure, capillaries and tightly connected epithelium with invivo-likering folds, crypt-villi, and microvilli is constructed by 3D embedding bioprinting. In our strategy, a novel photocurable bioink composed of methacrylated gelatin, methacrylated sodium alginate and poly (ethylene glycol) diacrylate is developed for the fabrication of intestinal model. The Caco-2 cells implanted in the lumen are induced by the topological structures of the model to derive microvilli, crypt-villi, and tight junctions, simulating the intestinal epithelial barrier. The human umbilical vein endothelial cells encapsulated within the model gradually form microvessels, mimicking the dense capillary network in the intestine. This intestine-like tissue, which closely resembles the structure and cell arrangement of the human gut, can act as a platform to predict the therapeutic and toxic side effects of new drugs on the intestine.
Collapse
Affiliation(s)
- Yuxuan Li
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, People's Republic of China
- Organoid Innovation Center, CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Shengnan Cheng
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, People's Republic of China
- Organoid Innovation Center, CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Haihua Shi
- Department of Gastrointestinal surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou 215001, People's Republic of China
| | - Renshun Yuan
- Department of Gastrointestinal surgery, Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Suzhou 215001, People's Republic of China
| | - Chen Gao
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, People's Republic of China
- Organoid Innovation Center, CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Yuhan Wang
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, People's Republic of China
- Organoid Innovation Center, CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Zhijun Zhang
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, People's Republic of China
- Organoid Innovation Center, CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Zongwu Deng
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, People's Republic of China
- Organoid Innovation Center, CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| | - Jie Huang
- School of Nano-Tech and Nano-Bionics, University of Science and Technology of China, Hefei 230026, People's Republic of China
- Organoid Innovation Center, CAS Key Laboratory of Nano-Bio Interface, Division of Nanobiomedicine, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, Suzhou 215123, People's Republic of China
| |
Collapse
|
49
|
Almeida-Pinto J, Moura BS, Gaspar VM, Mano JF. Advances in Cell-Rich Inks for Biofabricating Living Architectures. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2313776. [PMID: 38639337 DOI: 10.1002/adma.202313776] [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: 12/17/2023] [Revised: 04/15/2024] [Indexed: 04/20/2024]
Abstract
Advancing biofabrication toward manufacturing living constructs with well-defined architectures and increasingly biologically relevant cell densities is highly desired to mimic the biofunctionality of native human tissues. The formulation of tissue-like, cell-dense inks for biofabrication remains, however, challenging at various levels of the bioprinting process. Promising advances have been made toward this goal, achieving relatively high cell densities that surpass those found in conventional platforms, pushing the current boundaries closer to achieving tissue-like cell densities. On this focus, herein the overarching challenges in the bioprocessing of cell-rich living inks into clinically grade engineered tissues are discussed, as well as the most recent advances in cell-rich living ink formulations and their processing technologies are highlighted. Additionally, an overview of the foreseen developments in the field is provided and critically discussed.
Collapse
Affiliation(s)
- José Almeida-Pinto
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - Beatriz S Moura
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - Vítor M Gaspar
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| | - João F Mano
- Department of Chemistry, CICECO - Aveiro Institute of Materials, University of Aveiro, Campus Universitário de Santiago, Aveiro, 3810-193, Portugal
| |
Collapse
|
50
|
Hermans F, Hasevoets S, Vankelecom H, Bronckaers A, Lambrichts I. From Pluripotent Stem Cells to Organoids and Bioprinting: Recent Advances in Dental Epithelium and Ameloblast Models to Study Tooth Biology and Regeneration. Stem Cell Rev Rep 2024; 20:1184-1199. [PMID: 38498295 PMCID: PMC11222197 DOI: 10.1007/s12015-024-10702-w] [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] [Accepted: 02/20/2024] [Indexed: 03/20/2024]
Abstract
Ameloblasts are the specialized dental epithelial cell type responsible for enamel formation. Following completion of enamel development in humans, ameloblasts are lost and biological repair or regeneration of enamel is not possible. In the past, in vitro models to study dental epithelium and ameloblast biology were limited to freshly isolated primary cells or immortalized cell lines, both with limited translational potential. In recent years, large strides have been made with the development of induced pluripotent stem cell and organoid models of this essential dental lineage - both enabling modeling of human dental epithelium. Upon induction with several different signaling factors (such as transforming growth factor and bone morphogenetic proteins) these models display elevated expression of ameloblast markers and enamel matrix proteins. The advent of 3D bioprinting, and its potential combination with these advanced cellular tools, is poised to revolutionize the field - and its potential for tissue engineering, regenerative and personalized medicine. As the advancements in these technologies are rapidly evolving, we evaluate the current state-of-the-art regarding in vitro cell culture models of dental epithelium and ameloblast lineage with a particular focus toward their applicability for translational tissue engineering and regenerative/personalized medicine.
Collapse
Affiliation(s)
- Florian Hermans
- Department of Cardiology and Organ Systems (COS), Biomedical Research Institute (BIOMED), Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, 3590, Belgium.
| | - Steffie Hasevoets
- Department of Cardiology and Organ Systems (COS), Biomedical Research Institute (BIOMED), Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, 3590, Belgium
| | - Hugo Vankelecom
- Laboratory of Tissue Plasticity in Health and Disease, Cluster of Stem Cell and Developmental Biology, Department of Development and Regeneration, KU Leuven, Leuven, 3000, Belgium
| | - Annelies Bronckaers
- Department of Cardiology and Organ Systems (COS), Biomedical Research Institute (BIOMED), Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, 3590, Belgium
| | - Ivo Lambrichts
- Department of Cardiology and Organ Systems (COS), Biomedical Research Institute (BIOMED), Faculty of Medicine and Life Sciences, Hasselt University, Diepenbeek, 3590, Belgium.
| |
Collapse
|