1
|
Xu R, Ooi HS, Bian L, Ouyang L, Sun W. Dynamic hydrogels for biofabrication: A review. Biomaterials 2025; 320:123266. [PMID: 40120174 DOI: 10.1016/j.biomaterials.2025.123266] [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: 12/09/2024] [Revised: 03/06/2025] [Accepted: 03/17/2025] [Indexed: 03/25/2025]
Abstract
Reversibly crosslinked dynamic hydrogels have emerged as a significant material platform for biomedical applications owing to their distinctive time-dependent characteristics, including shear-thinning, self-healing, stress relaxation, and creep. These physical properties permit the use of dynamic hydrogels as injectable carriers or three-dimensional printable bioinks. It is noteworthy that matrix dynamics can serve as physical cues that stimulate cellular processes. Therefore, dynamic hydrogels are preferred for tissue engineering and biofabrication, which seek to create functional tissue constructs that require regulation of cellular processes. This review summarizes the critical biophysical properties of dynamic hydrogels, various cellular processes and related mechanisms triggered by hydrogel dynamics, particularly in three-dimensional culture scenarios. Subsequently, we present an overview of advanced biofabrication techniques, particularly 3D bioprinting, of dynamic hydrogels for the large-scale production of tissue and organ engineering models. This review presents an overview of the strategies that can be used to expand the range of applications of dynamic hydrogels in biofabrication, while also addressing the challenges and opportunities that arise in the field. This review highlights the importance of matrix dynamics in regulating cellular processes and elucidates strategies for leveraging them in the context of biofabrication.
Collapse
Affiliation(s)
- Runze Xu
- Biomanufacturing and Engineering Living Systems Innovation International Talents Base (111 Base), Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Hon Son Ooi
- Biomanufacturing and Engineering Living Systems Innovation International Talents Base (111 Base), Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China
| | - Liming Bian
- School of Biomedical Sciences and Engineering, Guangzhou International Campus, South China University of Technology, Guangzhou, 511442, China
| | - Liliang Ouyang
- Biomanufacturing and Engineering Living Systems Innovation International Talents Base (111 Base), Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China; State Key Laboratory of Tribology in Advanced Equipment, Tsinghua University, Beijing, 100084, China.
| | - Wei Sun
- Biomanufacturing and Engineering Living Systems Innovation International Talents Base (111 Base), Department of Mechanical Engineering, Tsinghua University, Beijing, 100084, China; Department of Mechanical Engineering and Mechanics, Drexel University, Philadelphia, PA, 19104, USA
| |
Collapse
|
2
|
Li K, He Y, Jin X, Jin K, Qian J. Reproducible extracellular matrices for tumor organoid culture: challenges and opportunities. J Transl Med 2025; 23:497. [PMID: 40312683 PMCID: PMC12044958 DOI: 10.1186/s12967-025-06349-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Accepted: 03/03/2025] [Indexed: 05/03/2025] Open
Abstract
Tumor organoid models have emerged as valuable 3D in vitro systems to study cancer behavior in a physiologically relevant environment. The composition and architecture of the extracellular matrix (ECM) play critical roles in tumor organoid culture by influencing the tumor microenvironment and tumor behavior. Traditional matrices such as Matrigel and collagen, have been widely used, but their batch-to-batch variability and limited tunability hinder their reproducibility and broader applications. To address these challenges, researchers have turned to synthetic/engineered matrices and biopolymer-based matrices, which offer precise tunability, reproducibility, and chemically defined compositions. However, these matrices also present challenges of their own. In this review, we explore the significance of ECMs in tumor organoid culture, discuss the limitations of commonly used matrices, and highlight recent advancements in engineered/synthetic matrices for improved tumor organoid modeling.
Collapse
Affiliation(s)
- Kan Li
- School of Basic Medical Sciences, Health Science Center, Ningbo University, Ningbo, 315211, China
| | - Yibo He
- Department of Surgical Oncology, Hangzhou Cancer Hospital, Hangzhou, Zhejiang, 310006, China
- Department of Breast Surgery, Affiliated Hangzhou First People'S Hospital, School of Medicine, Westlake University, Hangzhou, Zhejiang, 310006, China
| | - Xue Jin
- Center for Clinical Pharmacy, Cancer Center, Department of Pharmacy, Zhejiang Provincial People'S Hospital (Affiliated People'S Hospital, Hangzhou Medical College), Hangzhou, Zhejiang, 310014, China
| | - Ketao Jin
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Zhejiang Chinese Medical University (Zhejiang Provincial Hospital of Chinese Medicine), Hangzhou, Zhejiang, 310003, China.
| | - Jun Qian
- Department of Colorectal Surgery, Xinchang People'S Hospital, Affiliated Xinchang Hosptial, Wenzhou Medical University, Xinchang, Zhejiang, 312500, China.
| |
Collapse
|
3
|
Wu Y, Song Y, Soto J, Hoffman T, Lin X, Zhang A, Chen S, Massad RN, Han X, Qi D, Yeh KW, Fang Z, Eoh J, Gu L, Rowat AC, Gu Z, Li S. Viscoelastic extracellular matrix enhances epigenetic remodeling and cellular plasticity. Nat Commun 2025; 16:4054. [PMID: 40307238 PMCID: PMC12043949 DOI: 10.1038/s41467-025-59190-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Accepted: 04/14/2025] [Indexed: 05/02/2025] Open
Abstract
Extracellular matrices of living tissues exhibit viscoelastic properties, yet how these properties regulate chromatin and the epigenome remains unclear. Here, we show that viscoelastic substrates induce changes in nuclear architecture and epigenome, with more pronounced effects on softer surfaces. Fibroblasts on viscoelastic substrates display larger nuclei, lower chromatin compaction, and differential expression of distinct sets of genes related to the cytoskeleton and nuclear function, compared to those on elastic surfaces. Slow-relaxing viscoelastic substrates reduce lamin A/C expression and enhance nuclear remodeling. These structural changes are accompanied by a global increase in euchromatin marks and local increase in chromatin accessibility at cis-regulatory elements associated with neuronal and pluripotent genes. Consequently, viscoelastic substrates improve the reprogramming efficiency from fibroblasts into neurons and induced pluripotent stem cells. Collectively, our findings unravel the roles of matrix viscoelasticity in epigenetic regulation and cell reprogramming, with implications for designing smart materials for cell fate engineering.
Collapse
Affiliation(s)
- Yifan Wu
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Yang Song
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Institute of Biomedical Engineering, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, 610041, Chengdu, China
| | - Jennifer Soto
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Tyler Hoffman
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Xiao Lin
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Aaron Zhang
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Siyu Chen
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Ramzi N Massad
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Xiao Han
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Dongping Qi
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Kun-Wei Yeh
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Zhiwei Fang
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Joon Eoh
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Luo Gu
- Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
- Institute for NanoBioTechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
- Translational Tissue Engineering Center, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Amy C Rowat
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Department of Integrative Biology and Physiology, University of California Los Angeles, Los Angeles, CA, 90095, USA
- California NanoSystems Institute, University of California Los Angeles, Los Angeles, CA, 90095, USA
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA
| | - Zhen Gu
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA
- National Key Laboratory of Advanced Drug Delivery and Release Systems, College of Pharmaceutical Sciences, Zhejiang University, 310058, Hangzhou, China
- Key Laboratory of Advanced Drug Delivery Systems of Zhejiang Province, College of Pharmaceutical Sciences, Zhejiang University, 310058, Hangzhou, China
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, 310027, Hangzhou, China
| | - Song Li
- Department of Bioengineering, University of California Los Angeles, Los Angeles, CA, 90095, USA.
- Jonsson Comprehensive Cancer Center, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA.
- Department of Medicine, University of California Los Angeles, Los Angeles, CA, 90095, USA.
- Broad Stem Cell Research Center, University of California Los Angeles, Los Angeles, CA, 90095, USA.
| |
Collapse
|
4
|
Kuhn MR, Wolcott EA, Langer EM. Developments in gastrointestinal organoid cultures to recapitulate tissue environments. Front Bioeng Biotechnol 2025; 13:1521044. [PMID: 40313639 PMCID: PMC12043594 DOI: 10.3389/fbioe.2025.1521044] [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: 11/01/2024] [Accepted: 03/21/2025] [Indexed: 05/03/2025] Open
Abstract
Culture platforms that closely mimic the spatial architecture, cellular diversity, and extracellular matrix composition of native tissues can serve as invaluable tools for a range of scientific discovery and biomedical applications. Organoids have emerged as a promising alternative to both traditional 2D cell culture and animal models, offering a physiologically relevant 3D culture system for studying human cell biology. Organoids provide a manipulable platform to investigate organ development and function as well as to model patient-specific phenotypes. This mini review examines various methods used for culturing organoids to model normal and disease conditions in gastrointestinal tissues. We focus on how the matrix composition and media formulations can impact cell signaling, altering the baseline cellular phenotypes as well as response to perturbations. We discuss future directions for optimizing organoid culture conditions to improve basic and translational potential.
Collapse
Affiliation(s)
- Madeline R. Kuhn
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
- Division of Oncological Sciences, Oregon Health and Science University, Portland, OR, United States
| | - Emma A. Wolcott
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
- Division of Oncological Sciences, Oregon Health and Science University, Portland, OR, United States
| | - Ellen M. Langer
- Cancer Early Detection Advanced Research Center, Knight Cancer Institute, Oregon Health and Science University, Portland, OR, United States
- Division of Oncological Sciences, Oregon Health and Science University, Portland, OR, United States
- Brenden-Colson Center for Pancreatic Care, Oregon Health and Science University, Portland, OR, United States
| |
Collapse
|
5
|
Wu Y, Song Y, Soto J, Hoffman T, Lin X, Zhang A, Chen S, Massad RN, Han X, Qi D, Yeh KW, Fang Z, Eoh J, Gu L, Rowat AC, Gu Z, Li S. Viscoelastic Extracellular Matrix Enhances Epigenetic Remodeling and Cellular Plasticity. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.04.14.589442. [PMID: 38659850 PMCID: PMC11042188 DOI: 10.1101/2024.04.14.589442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Extracellular matrices of living tissues exhibit viscoelastic properties, yet how these properties regulate chromatin and the epigenome remains unclear. Here, we show that viscoelastic substrates induce changes in nuclear architecture and epigenome, with more pronounced effects on softer surfaces. Fibroblasts on viscoelastic substrates display larger nuclei, lower chromatin compaction, and differential expression of distinct sets of genes related to the cytoskeleton and nuclear function compared to those on purely elastic surfaces. Slow-relaxing viscoelastic substrates reduce lamin A/C expression and enhance nuclear remodeling. These structural changes are accompanied by a global increase in euchromatin marks and local increase in chromatin accessibility at cis-regulatory elements associated with neuronal and pluripotent genes. Consequently, viscoelastic substrates improve the reprogramming efficiency from fibroblasts into neurons and induced pluripotent stem cells. Collectively, our findings unravel the roles of matrix viscoelasticity in epigenetic regulation and cell reprogramming, with implications for designing smart materials for cell fate engineering.
Collapse
|
6
|
Wang Y, Zhang R, Qiao Z, Dou B, Xu H, Meng F, Huang J. Polyacrylamide-Based Hydrogel with Biocompatibility and Tunable Stiffness for Three-Dimensional Cell Culture. ACS APPLIED BIO MATERIALS 2025; 8:2356-2364. [PMID: 39949138 DOI: 10.1021/acsabm.4c01846] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/18/2025]
Abstract
Three-dimensional (3D) culture of cells has gained increasing popularity because of its enhanced physiological relevance and more accurate representation of in vivo tissues. Matrigel, alginate, hyaluronic acid, and collagen are biocompatible 3D culture platforms with cell biofunctions, while it is difficult to decouple the biofunctions with mechanical properties. Polyacrylamide (PAAm) is a biocompatible but biologically nonfunctional platform heavily used in 2D culture. However, the cytotoxicity of acrylamide (AAm) prevents the application of PAAm as a platform for the 3D culture. Here, through RAFT copolymerization of AAm with a primary amine-bearing functional monomer, followed by postpolymerization modification, we synthesized nontoxic, linear PAAm featuring either multithiol or multinorbornene groups, available in various chain lengths. PAAm networks were fabricated by photoinduced thiol-norbornene coupling. The resulting PAAm hydrogel was biocompatible and structurally homogeneous with highly tunable and reproducible mechanical properties. PAAm hydrogels supported the 3D culture of human umbilical vein endothelial cells (HUVECs), where a higher adhesive ligand density promoted the viability of HUVECs. Furthermore, in combination with Matrigel, the PAAm hydrogel was used in the 3D culture of intestinal organoids, demonstrating that a lower mechanical strength was favorable. In summary, this report paves the way for the use of PAAm hydrogels in 3D culture, which is especially appealing for the decoupling of biological functions and mechanical properties.
Collapse
Affiliation(s)
- Yi Wang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
- SINOPEC Research Institute of Petroleum Engineering Co. Ltd., Beijing 102206, China
| | - Rui Zhang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| | - Ziwen Qiao
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| | - Bohan Dou
- Department of Mechanical Engineering, Tsinghua University, Beijing 100084, China
| | - Hongwei Xu
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| | - Fanlu Meng
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| | - Jianyong Huang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| |
Collapse
|
7
|
Vanhoeijen R, Okkelman IA, Rogier N, Sedlačík T, Stöbener DD, Devriendt B, Dmitriev RI, Hoogenboom R. Poly(2-alkyl-2-oxazoline) Hydrogels as Synthetic Matrices for Multicellular Spheroid and Intestinal Organoid Cultures. Biomacromolecules 2025; 26:1860-1872. [PMID: 39898884 DOI: 10.1021/acs.biomac.4c01627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
The extracellular matrix (ECM) plays a crucial role in organoid cultures by supporting cell proliferation and differentiation. A key feature of the ECM is its mechanical influence on the surrounding cells, directly affecting their behavior. Matrigel, the most commonly used ECM, is limited by its animal-derived origin, batch variability, and uncontrollable mechanical properties, restricting its use in 3D cell-model-based mechanobiological studies. Poly(2-alkyl-2-oxazoline) (PAOx) synthetic hydrogels represent an appealing alternative because of their reproducibility and versatile chemistry, enabling tuning of hydrogel stiffness and functionalization. Here, we studied PAOx hydrogels with differing compressive moduli for their potential to support 3D cell growth. PAOx hydrogels support spheroid and organoid growth over several days without the addition of ECM components. Furthermore, we discovered intestinal organoid epithelial polarity reversion in PAOx hydrogels and demonstrate how the tunable mechanical properties of PAOx can be used to study effects on the morphology and oxygenation of live multicellular spheroids.
Collapse
Affiliation(s)
- Robin Vanhoeijen
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Krijgslaan 281-S4, Ghent 9000, Belgium
- Tissue Engineering and Biomaterials Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, C. Heymanslaan 10, Ghent 9000, Belgium
| | - Irina A Okkelman
- Tissue Engineering and Biomaterials Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, C. Heymanslaan 10, Ghent 9000, Belgium
| | - Nette Rogier
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Krijgslaan 281-S4, Ghent 9000, Belgium
- Tissue Engineering and Biomaterials Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, C. Heymanslaan 10, Ghent 9000, Belgium
| | - Tomáš Sedlačík
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Krijgslaan 281-S4, Ghent 9000, Belgium
- Hydrogel Lab, Department of Polymers, Faculty of Chemical Technology, University of Chemistry and Technology, Technicka 1903/5, Prague 6 166 28, Czech Republic
| | - Daniel D Stöbener
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Krijgslaan 281-S4, Ghent 9000, Belgium
| | - Bert Devriendt
- Laboratory of Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke 9820, Belgium
| | - Ruslan I Dmitriev
- Tissue Engineering and Biomaterials Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, C. Heymanslaan 10, Ghent 9000, Belgium
| | - Richard Hoogenboom
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Krijgslaan 281-S4, Ghent 9000, Belgium
| |
Collapse
|
8
|
Shao Y, Wang J, Jin A, Jiang S, Lei L, Liu L. Biomaterial-assisted organoid technology for disease modeling and drug screening. Mater Today Bio 2025; 30:101438. [PMID: 39866785 PMCID: PMC11757232 DOI: 10.1016/j.mtbio.2024.101438] [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: 10/17/2024] [Revised: 12/10/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025] Open
Abstract
Developing disease models and screening for effective drugs are key areas of modern medical research. Traditional methodologies frequently fall short in precisely replicating the intricate architecture of bodily tissues and organs. Nevertheless, recent advancements in biomaterial-assisted organoid technology have ushered in a paradigm shift in biomedical research. This innovative approach enables the cultivation of three-dimensional cellular structures in vitro that closely emulate the structural and functional attributes of organs, offering physiologically superior models compared to conventional techniques. The evolution of biomaterials plays a pivotal role in supporting the culture and development of organ tissues, thereby facilitating more accurate disease state modeling and the rigorous evaluation of drug efficacy and safety profiles. In this review, we will explore the roles that various biomaterials play in organoid development, examine the fundamental principles and advantages of utilizing these technologies in constructing disease models, and highlight recent advances and practical applications in drug screening using disease-specific organoid models. Additionally, the challenges and future directions of organoid technology are discussed. Through continued research and innovation, we aim to make remarkable strides in disease treatment and drug development, ultimately enhancing patient quality of life.
Collapse
Affiliation(s)
- Yunyuan Shao
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, China
| | - Juncheng Wang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Anqi Jin
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, China
| | - Shicui Jiang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Lanjie Lei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, China
| | - Liangle Liu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| |
Collapse
|
9
|
Deng Y, Yuan X, Lu X, Wu J, Luo C, Zhang T, Liu Q, Tang S, Li Z, Mu X, Hu Y, Du Q, Xu J, Xie R. The Use of Gut Organoids: To Study the Physiology and Disease of the Gut Microbiota. J Cell Mol Med 2025; 29:e70330. [PMID: 39968926 PMCID: PMC11836903 DOI: 10.1111/jcmm.70330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 11/02/2024] [Accepted: 12/16/2024] [Indexed: 02/20/2025] Open
Abstract
The intestinal flora has attracted much attention in recent years. An imbalance in the intestinal flora can cause not only intestinal diseases but also cause a variety of parenteral diseases, such as endocrine diseases, nervous system diseases and cardiovascular diseases. Research on the mechanism of disease is likely to be hampered by sample accessibility, ethical issues, and differences between cellular animal and physiological studies. However, advances in stem cell culture have made it possible to reproduce 3D human tissues in vitro that mimic the cellular, anatomical and functional characteristics of real organs. Recent studies have shown that organoids can be used to simulate the development and disease of the gut and intestinal flora and have a wide range of applications in intestinal flora physiology and disease. Intestinal organoids provide a preeminent in vitro model system for cultivating microbiota that influence GI physiology, as well as for understanding how they encounter intestinal epithelial cells and cause disease. The mechanistic details obtained from such modelling may provide new avenues for the prevention and treatment of many gastrointestinal (GI) disorders. Researchers are now starting to take inspiration from other fields, such as bioengineering, and the rise of interdisciplinary approaches, including organoid chip technology and microfluidics, has greatly accelerated the development of organoids to generate intestinal organoids that are more physiologically relevant and suitable for gut microbiota studies. Here, we describe the development of organoid models of gut biology and the application of organoids to study the pathophysiology of diseases caused by intestinal dysbiosis.
Collapse
Affiliation(s)
- Ya Deng
- Department of Endoscopy and Digestive SystemGuizhou Provincial People's HospitalGuiyangGuizhouChina
- Zunyi Medical UniversityZunyiGuizhouChina
| | - Xiaolu Yuan
- The Second Affiliated Hospital of Zunyi Medical UniversityZunyiGuizhouChina
| | - XianMin Lu
- Zunyi Medical UniversityZunyiGuizhouChina
| | - Jiangbo Wu
- Zunyi Medical UniversityZunyiGuizhouChina
| | - Chen Luo
- Zunyi Medical UniversityZunyiGuizhouChina
| | - Ting Zhang
- Zunyi Medical UniversityZunyiGuizhouChina
| | - Qi Liu
- Zunyi Medical UniversityZunyiGuizhouChina
| | - Siqi Tang
- Zunyi Medical UniversityZunyiGuizhouChina
| | - Zhuo Li
- Zunyi Medical UniversityZunyiGuizhouChina
| | - Xingyi Mu
- Zunyi Medical UniversityZunyiGuizhouChina
| | - Yanxia Hu
- Zunyi Medical UniversityZunyiGuizhouChina
| | - Qian Du
- Department of Endoscopy and Digestive SystemGuizhou Provincial People's HospitalGuiyangGuizhouChina
| | - Jingyu Xu
- Guizhou Medical UniversityGuiyangGuizhouChina
| | - Rui Xie
- Department of Endoscopy and Digestive SystemGuizhou Provincial People's HospitalGuiyangGuizhouChina
| |
Collapse
|
10
|
Park S, Kwon O, Lee H, Cho Y, Yeun J, Yoon SH, Sun SY, Huh Y, Yu WD, Park S, Son N, Jeon S, Lee S, Kim DS, Lee SY, Son JG, Lee KJ, Kim YI, Lim JH, Yoo J, Lee TG, Son MY, Im SG. Xenogeneic-free culture of human intestinal stem cells on functional polymer-coated substrates for scalable, clinical-grade stem cell therapy. Nat Commun 2024; 15:10492. [PMID: 39622824 PMCID: PMC11612142 DOI: 10.1038/s41467-024-54653-9] [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: 03/06/2024] [Accepted: 11/18/2024] [Indexed: 12/06/2024] Open
Abstract
The need for basement membrane extract (BME) with undefined constituents, such as Matrigel, for intestinal stem cell (ISC) culture in traditional systems poses a significant barrier that must be overcome for the development of clinical-grade, scalable, ready-to-use ISCs. Here, we propose a functional polymer-based xenogeneic-free dish for the culture of intestinal stem cells (XF-DISC), ensuring substantially prolonged maintenance of ISCs derived from 3-dimensional human intestinal organoids (ISCs3D-hIO). XF-DISC enables remarkable expandability, exhibiting a 24-fold increase in cell numbers within 30 days, with long-term maintenance of ISCs3D-hIO for more than 30 consecutive passages (>210 days). In addition, XF-DISC is fully compatible with a cell banking system. Notably, human pluripotent stem cell-derived ISCs3D-hIO cultured on XF-DISC are successfully transplanted into intestinal injury and inflammation mouse models, leading to engraftment and regeneration of damaged mouse intestinal epithelium. As a reliable and scalable xenogeneic-free ISC3D-hIO culture method, XF-DISC is highly promising for the development of regenerative ISC therapy for human intestinal diseases.
Collapse
Affiliation(s)
- Seonghyeon Park
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Ohman Kwon
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea
- KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Hana Lee
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Younghak Cho
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Jemin Yeun
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Sung Hyun Yoon
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Sang Yu Sun
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Yubin Huh
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea
- KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Won Dong Yu
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea
- KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Sohee Park
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea
- KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Naeun Son
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea
- KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Sojeong Jeon
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea
- KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
| | - Sugi Lee
- Digital Biotech Innovation Center, KRIBB, Daejeon, Republic of Korea
| | - Dae-Soo Kim
- KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea
- Digital Biotech Innovation Center, KRIBB, Daejeon, Republic of Korea
| | - Sun Young Lee
- Nanobio Measurement Group, Division of Biomedical Metrology, Korea Research Institute of Standards and Science (KRISS), 267 Gajeong-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Jin Gyeong Son
- Nanobio Measurement Group, Division of Biomedical Metrology, Korea Research Institute of Standards and Science (KRISS), 267 Gajeong-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Kyung Jin Lee
- R&D Institute, ORGANOIDSCIENCES Ltd., Seongnam, Republic of Korea
| | - Yong Il Kim
- R&D Institute, ORGANOIDSCIENCES Ltd., Seongnam, Republic of Korea
| | - Jin Hong Lim
- Department of Surgery, Gangnam Severance Hospital, Pancreatobiliary Cancer Clinic, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jongman Yoo
- R&D Institute, ORGANOIDSCIENCES Ltd., Seongnam, Republic of Korea
- Organoid Standards Initiative (OSI), Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea
- CHA Organoid Research Center, CHA University, Seongnam, Republic of Korea
| | - Tae Geol Lee
- Nanobio Measurement Group, Division of Biomedical Metrology, Korea Research Institute of Standards and Science (KRISS), 267 Gajeong-ro, Yuseong-gu, Daejeon, Republic of Korea
| | - Mi-Young Son
- Stem Cell Convergence Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), 125 Gwahak-ro, Yuseong-gu, Daejeon, Republic of Korea.
- KRIBB School of Bioscience, Korea University of Science and Technology (UST), Daejeon, Republic of Korea.
- Organoid Standards Initiative (OSI), Department of Biophysics, Institute of Quantum Biophysics, Sungkyunkwan University, Suwon, Republic of Korea.
- School of Medicine, Sungkyunkwan University, Suwon, Republic of Korea.
| | - Sung Gap Im
- Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), 291 Daehak-ro, Yuseong-gu, Daejeon, Republic of Korea.
- KAIST Stem Cell Center, Department of Chemical and Biomolecular Engineering, Graduate School of Stem Cell & Regenerative Biology, KAIST, Daejeon, Republic of Korea.
| |
Collapse
|
11
|
Lin YH, Lou J, Xia Y, Chaudhuri O. Cross-Linker Architectures Impact Viscoelasticity in Dynamic Covalent Hydrogels. Adv Healthc Mater 2024; 13:e2402059. [PMID: 39407436 PMCID: PMC11617263 DOI: 10.1002/adhm.202402059] [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: 06/04/2024] [Revised: 08/30/2024] [Indexed: 10/23/2024]
Abstract
Dynamic covalent cross-linked (DCC) hydrogels represent a significant advance in biomaterials for regenerative medicine and mechanobiology, offering viscoelasticity, and self-healing properties that more closely mimic in vivo tissue mechanics than traditional, predominantly elastic, covalent hydrogels. However, the effects of varying cross-linker architecture on DCC hydrogel viscoelasticity have not been thoroughly investigated. This study introduces hydrazone-based alginate hydrogels to explore how cross-linker architectures impact stiffness and viscoelasticity. In hydrogels with side-chain cross-linker (SCX), higher cross-linker concentrations enhance stiffness and decelerate stress relaxation, while an off-stoichiometric hydrazine-to-aldehyde ratio reduces stiffness and shortens relaxation time. In hydrogels with telechelic cross-linking, maximal stiffness and relaxation time occurs at intermediate cross-linker mixing ratio for both linear cross-linker (LX) and star cross-linker (SX), with higher cross-linker valency further enhancing these properties. Further, the ranges of stiffness and viscoelasticity accessible with the different cross-linker architectures are found to be distinct, with SCX hydrogels leading to slower stress relaxation relative to the other architectures, and SX hydrogels providing increased stiffness and slower stress relaxation versus LX hydrogels. This research underscores the pivotal role of cross-linker architecture in defining hydrogel stiffness and viscoelasticity, providing insights for designing DCC hydrogels with tailored mechanical properties for specific biomedical applications.
Collapse
Affiliation(s)
- Yung-Hao Lin
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Junzhe Lou
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, MA, 02215, USA
| | - Yan Xia
- Department of Chemistry, Stanford University, Stanford, CA, 94305, USA
| | - Ovijit Chaudhuri
- Department of Mechanical Engineering, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
12
|
Liu Y, Gilchrist AE, Heilshorn SC. Engineered Protein Hydrogels as Biomimetic Cellular Scaffolds. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407794. [PMID: 39233559 PMCID: PMC11573243 DOI: 10.1002/adma.202407794] [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: 05/31/2024] [Revised: 08/01/2024] [Indexed: 09/06/2024]
Abstract
The biochemical and biophysical properties of the extracellular matrix (ECM) play a pivotal role in regulating cellular behaviors such as proliferation, migration, and differentiation. Engineered protein-based hydrogels, with highly tunable multifunctional properties, have the potential to replicate key features of the native ECM. Formed by self-assembly or crosslinking, engineered protein-based hydrogels can induce a range of cell behaviors through bioactive and functional domains incorporated into the polymer backbone. Using recombinant techniques, the amino acid sequence of the protein backbone can be designed with precise control over the chain-length, folded structure, and cell-interaction sites. In this review, the modular design of engineered protein-based hydrogels from both a molecular- and network-level perspective are discussed, and summarize recent progress and case studies to highlight the diverse strategies used to construct biomimetic scaffolds. This review focuses on amino acid sequences that form structural blocks, bioactive blocks, and stimuli-responsive blocks designed into the protein backbone for highly precise and tunable control of scaffold properties. Both physical and chemical methods to stabilize dynamic protein networks with defined structure and bioactivity for cell culture applications are discussed. Finally, a discussion of future directions of engineered protein-based hydrogels as biomimetic cellular scaffolds is concluded.
Collapse
Affiliation(s)
- Yueming Liu
- Department of Materials Science & Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Aidan E Gilchrist
- Department of Biomedical Engineering, University of California, Davis 451 Health Sciences Dr, GBSF 3315, Davis, CA, 95616, USA
| | - Sarah C Heilshorn
- Department of Materials Science & Engineering, 476 Lomita Mall, McCullough Room 246, Stanford, CA, 94305, USA
| |
Collapse
|
13
|
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
|
14
|
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
|
15
|
Giraldo-Castaño MC, Littlejohn KA, Avecilla ARC, Barrera-Villamizar N, Quiroz FG. Programmability and biomedical utility of intrinsically-disordered protein polymers. Adv Drug Deliv Rev 2024; 212:115418. [PMID: 39094909 PMCID: PMC11389844 DOI: 10.1016/j.addr.2024.115418] [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: 04/20/2024] [Revised: 07/03/2024] [Accepted: 07/29/2024] [Indexed: 08/04/2024]
Abstract
Intrinsically disordered proteins (IDPs) exhibit molecular-level conformational dynamics that are functionally harnessed across a wide range of fascinating biological phenomena. The low sequence complexity of IDPs has led to the design and development of intrinsically-disordered protein polymers (IDPPs), a class of engineered repeat IDPs with stimuli-responsive properties. The perfect repetitive architecture of IDPPs allows for repeat-level encoding of tunable protein functionality. Designer IDPPs can be modeled on endogenous IDPs or engineered de novo as protein polymers with dual biophysical and biological functionality. Their properties can be rationally tailored to access enigmatic IDP biology and to create programmable smart biomaterials. With the goal of inspiring the bioengineering of multifunctional IDP-based materials, here we synthesize recent multidisciplinary progress in programming and exploiting the bio-functionality of IDPPs and IDPP-containing proteins. Collectively, expanding beyond the traditional sequence space of extracellular IDPs, emergent sequence-level control of IDPP functionality is fueling the bioengineering of self-assembling biomaterials, advanced drug delivery systems, tissue scaffolds, and biomolecular condensates -genetically encoded organelle-like structures. Looking forward, we emphasize open challenges and emerging opportunities, arguing that the intracellular behaviors of IDPPs represent a rich space for biomedical discovery and innovation. Combined with the intense focus on IDP biology, the growing landscape of IDPPs and their biomedical applications set the stage for the accelerated engineering of high-value biotechnologies and biomaterials.
Collapse
Affiliation(s)
- Maria Camila Giraldo-Castaño
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Kai A Littlejohn
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Alexa Regina Chua Avecilla
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Natalia Barrera-Villamizar
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA
| | - Felipe Garcia Quiroz
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology and Emory University, Atlanta, GA, USA.
| |
Collapse
|
16
|
LeSavage BL, Zhang D, Huerta-López C, Gilchrist AE, Krajina BA, Karlsson K, Smith AR, Karagyozova K, Klett KC, Huang MS, Long C, Kaber G, Madl CM, Bollyky PL, Curtis C, Kuo CJ, Heilshorn SC. Engineered matrices reveal stiffness-mediated chemoresistance in patient-derived pancreatic cancer organoids. NATURE MATERIALS 2024; 23:1138-1149. [PMID: 38965405 DOI: 10.1038/s41563-024-01908-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/25/2022] [Accepted: 04/30/2024] [Indexed: 07/06/2024]
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is characterized by its fibrotic and stiff extracellular matrix. However, how the altered cell/extracellular-matrix signalling contributes to the PDAC tumour phenotype has been difficult to dissect. Here we design and engineer matrices that recapitulate the key hallmarks of the PDAC tumour extracellular matrix to address this knowledge gap. We show that patient-derived PDAC organoids from three patients develop resistance to several clinically relevant chemotherapies when cultured within high-stiffness matrices mechanically matched to in vivo tumours. Using genetic barcoding, we find that while matrix-specific clonal selection occurs, cellular heterogeneity is not the main driver of chemoresistance. Instead, matrix-induced chemoresistance occurs within a stiff environment due to the increased expression of drug efflux transporters mediated by CD44 receptor interactions with hyaluronan. Moreover, PDAC chemoresistance is reversible following transfer from high- to low-stiffness matrices, suggesting that targeting the fibrotic extracellular matrix may sensitize chemoresistant tumours. Overall, our findings support the potential of engineered matrices and patient-derived organoids for elucidating extracellular matrix contributions to human disease pathophysiology.
Collapse
Affiliation(s)
- Bauer L LeSavage
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Daiyao Zhang
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Carla Huerta-López
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Aidan E Gilchrist
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Brad A Krajina
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Kasper Karlsson
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Amber R Smith
- Department of Medicine, Division of Hematology, Stanford University School of Medicine, Stanford, CA, USA
| | - Kremena Karagyozova
- Stanford Cancer Institute, Stanford University School of Medicine, Stanford, CA, USA
| | - Katarina C Klett
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Michelle S Huang
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Christopher Long
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Gernot Kaber
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Christopher M Madl
- Department of Bioengineering, Stanford University, Stanford, CA, USA
- Department of Materials Science and Engineering, University of Pennsylvania, Philadelphia, PA, USA
| | - Paul L Bollyky
- Division of Infectious Diseases and Geographic Medicine, Department of Medicine, Stanford University School of Medicine, Stanford, CA, USA
| | - Christina Curtis
- Department of Medicine, Division of Oncology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, USA
- Stanford Cancer Institute, 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
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|
17
|
Cai PC, Braunreuther M, Shih A, Spakowitz AJ, Fuller GG, Heilshorn SC. Air-liquid intestinal cell culture allows in situ rheological characterization of intestinal mucus. APL Bioeng 2024; 8:026112. [PMID: 38721267 PMCID: PMC11078553 DOI: 10.1063/5.0187974] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 03/25/2024] [Indexed: 01/06/2025] Open
Abstract
Intestinal health heavily depends on establishing a mucus layer within the gut with physical properties that strike a balance between being sufficiently elastic to keep out harmful pathogens yet viscous enough to flow and turnover the contents being digested. Studies investigating dysfunction of the mucus layer in the intestines are largely confined to animal models, which require invasive procedures to collect the mucus fluid. In this work, we develop a nondestructive method to study intestinal mucus. We use an air-liquid interface culture of primary human intestinal epithelial cells that exposes their apical surface to allow in situ analysis of the mucus layer. Mucus collection is not only invasive but also disrupts the mucus microstructure, which plays a crucial role in the interaction between mucus and the gut microbiome. Therefore, we leverage a noninvasive rheology technique that probes the mechanical properties of the mucus without removal from the culture. Finally, to demonstrate biomedical uses for this cell culture system, we characterize the biochemical and biophysical properties of intestinal mucus due to addition of the cytokine IL-13 to recapitulate the gut environment of Nippostrongylus brasiliensis infection.
Collapse
Affiliation(s)
- Pamela C. Cai
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, USA
| | - Margaret Braunreuther
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, USA
| | - Audrey Shih
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, USA
| | | | - Gerald G. Fuller
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, USA
| | - Sarah C. Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA
| |
Collapse
|
18
|
Guan Y, Peltz G. Hepatic organoids move from adolescence to maturity. Liver Int 2024; 44:1290-1297. [PMID: 38451053 DOI: 10.1111/liv.15893] [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: 08/17/2023] [Revised: 02/08/2024] [Accepted: 02/28/2024] [Indexed: 03/08/2024]
Abstract
Since organoids were developed 15 years ago, they are now in their adolescence as a research tool. The ability to generate 'tissue in a dish' has created enormous opportunities for biomedical research. We examine the contributions that hepatic organoids have made to three areas of liver research: as a source of cells and tissue for basic research, for drug discovery and drug safety testing, and for understanding disease pathobiology. We discuss the features that enable hepatic organoids to provide useful models for human liver diseases and identify four types of advances that will enable them to become a mature (i.e., adult) research tool over the next 5 years. During this period, advances in single-cell RNA sequencing and CRISPR technologies coupled with improved hepatic organoid methodology, which enables them to have a wider range of cell types that are present in liver and to be grown in microwells, will generate discoveries that will dramatically advance our understanding of liver development and the pathogenesis of liver diseases. It will generate also new approaches for treating liver fibrosis, which remains a major public health problem with few treatment options.
Collapse
Affiliation(s)
- Yuan Guan
- Department of Anesthesia, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, California, USA
| | - Gary Peltz
- Department of Anesthesia, Pain and Perioperative Medicine, Stanford University School of Medicine, Stanford, California, USA
| |
Collapse
|
19
|
Park S, Cho SW. Bioengineering toolkits for potentiating organoid therapeutics. Adv Drug Deliv Rev 2024; 208:115238. [PMID: 38447933 DOI: 10.1016/j.addr.2024.115238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 01/28/2024] [Accepted: 02/27/2024] [Indexed: 03/08/2024]
Abstract
Organoids are three-dimensional, multicellular constructs that recapitulate the structural and functional features of specific organs. Because of these characteristics, organoids have been widely applied in biomedical research in recent decades. Remarkable advancements in organoid technology have positioned them as promising candidates for regenerative medicine. However, current organoids still have limitations, such as the absence of internal vasculature, limited functionality, and a small size that is not commensurate with that of actual organs. These limitations hinder their survival and regenerative effects after transplantation. Another significant concern is the reliance on mouse tumor-derived matrix in organoid culture, which is unsuitable for clinical translation due to its tumor origin and safety issues. Therefore, our aim is to describe engineering strategies and alternative biocompatible materials that can facilitate the practical applications of organoids in regenerative medicine. Furthermore, we highlight meaningful progress in organoid transplantation, with a particular emphasis on the functional restoration of various organs.
Collapse
Affiliation(s)
- Sewon Park
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Seung-Woo Cho
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; Center for Nanomedicine, Institute for Basic Science (IBS), Seoul 03722, Republic of Korea; Graduate Program of Nano Biomedical Engineering (NanoBME), Advanced Science Institute, Yonsei University, Seoul 03722, Republic of Korea.
| |
Collapse
|
20
|
Puertas-Bartolomé M, Venegas-Bustos D, Acosta S, Rodríguez-Cabello JC. Contribution of the ELRs to the development of advanced in vitro models. Front Bioeng Biotechnol 2024; 12:1363865. [PMID: 38650751 PMCID: PMC11033926 DOI: 10.3389/fbioe.2024.1363865] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2023] [Accepted: 03/18/2024] [Indexed: 04/25/2024] Open
Abstract
Developing in vitro models that accurately mimic the microenvironment of biological structures or processes holds substantial promise for gaining insights into specific biological functions. In the field of tissue engineering and regenerative medicine, in vitro models able to capture the precise structural, topographical, and functional complexity of living tissues, prove to be valuable tools for comprehending disease mechanisms, assessing drug responses, and serving as alternatives or complements to animal testing. The choice of the right biomaterial and fabrication technique for the development of these in vitro models plays an important role in their functionality. In this sense, elastin-like recombinamers (ELRs) have emerged as an important tool for the fabrication of in vitro models overcoming the challenges encountered in natural and synthetic materials due to their intrinsic properties, such as phase transition behavior, tunable biological properties, viscoelasticity, and easy processability. In this review article, we will delve into the use of ELRs for molecular models of intrinsically disordered proteins (IDPs), as well as for the development of in vitro 3D models for regenerative medicine. The easy processability of the ELRs and their rational design has allowed their use for the development of spheroids and organoids, or bioinks for 3D bioprinting. Thus, incorporating ELRs into the toolkit of biomaterials used for the fabrication of in vitro models, represents a transformative step forward in improving the accuracy, efficiency, and functionality of these models, and opening up a wide range of possibilities in combination with advanced biofabrication techniques that remains to be explored.
Collapse
Affiliation(s)
- María Puertas-Bartolomé
- Technical Proteins Nanobiotechnology, S.L. (TPNBT), Valladolid, Spain
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER's Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| | - Desiré Venegas-Bustos
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER's Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| | - Sergio Acosta
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER's Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| | - José Carlos Rodríguez-Cabello
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER's Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| |
Collapse
|
21
|
DeFrates KG, Tong E, Cheng J, Heber‐Katz E, Messersmith PB. A Pro-Regenerative Supramolecular Prodrug Protects Against and Repairs Colon Damage in Experimental Colitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2304716. [PMID: 38247203 PMCID: PMC10987129 DOI: 10.1002/advs.202304716] [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: 07/12/2023] [Revised: 10/13/2023] [Indexed: 01/23/2024]
Abstract
Structural repair of the intestinal epithelium is strongly correlated with disease remission in inflammatory bowel disease (IBD); however, ulcer healing is not addressed by existing therapies. To address this need, this study reports the use of a small molecule prolyl hydroxylase (PHD) inhibitor (DPCA) to upregulate hypoxia-inducible factor one-alpha (HIF-1α) and induce mammalian regeneration. Sustained delivery of DPCA is achieved through subcutaneous injections of a supramolecular hydrogel, formed through the self-assembly of PEG-DPCA conjugates. Pre-treatment of mice with PEG-DPCA is shown to protect mice from epithelial erosion and symptoms of dextran sodium sulfate (DSS)-induced colitis. Surprisingly, a single subcutaneous dose of PEG-DPCA, administered after disease onset, leads to accelerated weight gain and complete restoration of healthy tissue architecture in colitic mice. Rapid DPCA-induced restoration of the intestinal barrier is likely orchestrated by increased expression of HIF-1α and associated targets leading to an epithelial-to-mesenchymal transition. Further investigation of DPCA as a potential adjunctive or stand-alone restorative treatment to combat active IBD is warranted.
Collapse
Affiliation(s)
- Kelsey G. DeFrates
- Department of BioengineeringUniversity of California, BerkeleyBerkeleyCA94720USA
| | - Elaine Tong
- Department of BioengineeringUniversity of California, BerkeleyBerkeleyCA94720USA
| | - Jing Cheng
- Department of BioengineeringUniversity of California, BerkeleyBerkeleyCA94720USA
| | | | - Phillip B. Messersmith
- Department of BioengineeringUniversity of California, BerkeleyBerkeleyCA94720USA
- Department of Materials Science and EngineeringUniversity of California, BerkeleyBerkeleyCA94720USA
- Materials Sciences DivisionLawrence Berkeley National LaboratoryBerkeleyCA94720USA
| |
Collapse
|
22
|
Yang H, Li J, Wang Z, Khutsishvili D, Tang J, Zhu Y, Cai Y, Dai X, Ma S. Bridging the organoid translational gap: integrating standardization and micropatterning for drug screening in clinical and pharmaceutical medicine. LIFE MEDICINE 2024; 3:lnae016. [PMID: 39872665 PMCID: PMC11748978 DOI: 10.1093/lifemedi/lnae016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/29/2023] [Accepted: 04/12/2024] [Indexed: 01/30/2025]
Abstract
Synthetic organ models such as organoids and organ-on-a-chip have been receiving recognition from administrative agencies. Despite the proven success of organoids in predicting drug efficacy on laboratory scales, their translational advances have not fully satisfied the expectations for both clinical implementation and commercial applications. The transition from laboratory settings to clinical applications continues to encounter challenges. Employing engineering methodologies to facilitate the bridging of this gap for organoids represents one of the key directions for future advancement. The main measures to bridge the gap include environmental and phenotypic recapitulation, 3D patterning, matrix engineering, and multi-modality information acquisition and processing. Pilot whole-process clinical/pharmaceutical applications with fast and standardized organoid models will continuously offer convincing frontline optimization clues and driving forces to the organoid community, which is a promising path to translational organoid technologies.
Collapse
Affiliation(s)
- Haowei Yang
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
- Tsinghua-Berkeley Shenzhen Institute, Shenzhen 518055, China
| | - Jiawei Li
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
- Tsinghua-Berkeley Shenzhen Institute, Shenzhen 518055, China
| | - Zitian Wang
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
| | - Davit Khutsishvili
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
| | - Jiyuan Tang
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
| | - Yu Zhu
- Guangdong Research Center of Organoid Engineering and Technology, Guangzhou 510530, China
| | - Yongde Cai
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
| | - Xiaoyong Dai
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
| | - Shaohua Ma
- Tsinghua Shenzhen International Graduate School (SIGS), Tsinghua University, Shenzhen 518055, China
- Tsinghua-Berkeley Shenzhen Institute, Shenzhen 518055, China
- Key Laboratory of Industrial Biocatalysis (Ministry of Education), Tsinghua University, Beijing 100084, China
| |
Collapse
|
23
|
Guo L, Li C, Gong W. Toward reproducible tumor organoid culture: focusing on primary liver cancer. Front Immunol 2024; 15:1290504. [PMID: 38571961 PMCID: PMC10987700 DOI: 10.3389/fimmu.2024.1290504] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2023] [Accepted: 02/29/2024] [Indexed: 04/05/2024] Open
Abstract
Organoids present substantial potential for pushing forward preclinical research and personalized medicine by accurately recapitulating tissue and tumor heterogeneity in vitro. However, the lack of standardized protocols for cancer organoid culture has hindered reproducibility. This paper comprehensively reviews the current challenges associated with cancer organoid culture and highlights recent multidisciplinary advancements in the field with a specific focus on standardizing liver cancer organoid culture. We discuss the non-standardized aspects, including tissue sources, processing techniques, medium formulations, and matrix materials, that contribute to technical variability. Furthermore, we emphasize the need to establish reproducible platforms that accurately preserve the genetic, proteomic, morphological, and pharmacotypic features of the parent tumor. At the end of each section, our focus shifts to organoid culture standardization in primary liver cancer. By addressing these challenges, we can enhance the reproducibility and clinical translation of cancer organoid systems, enabling their potential applications in precision medicine, drug screening, and preclinical research.
Collapse
Affiliation(s)
| | | | - Weiqiang Gong
- Department of Hepatobiliary and Pancreatic Surgery, Weifang People’s Hospital, Weifang, Shandong, China
| |
Collapse
|
24
|
Mai S, Inkielewicz-Stepniak I. Graphene Oxide Nanoparticles and Organoids: A Prospective Advanced Model for Pancreatic Cancer Research. Int J Mol Sci 2024; 25:1066. [PMID: 38256139 PMCID: PMC10817028 DOI: 10.3390/ijms25021066] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2023] [Revised: 01/02/2024] [Accepted: 01/04/2024] [Indexed: 01/24/2024] Open
Abstract
Pancreatic cancer, notorious for its grim 10% five-year survival rate, poses significant clinical challenges, largely due to late-stage diagnosis and limited therapeutic options. This review delves into the generation of organoids, including those derived from resected tissues, biopsies, pluripotent stem cells, and adult stem cells, as well as the advancements in 3D printing. It explores the complexities of the tumor microenvironment, emphasizing culture media, the integration of non-neoplastic cells, and angiogenesis. Additionally, the review examines the multifaceted properties of graphene oxide (GO), such as its mechanical, thermal, electrical, chemical, and optical attributes, and their implications in cancer diagnostics and therapeutics. GO's unique properties facilitate its interaction with tumors, allowing targeted drug delivery and enhanced imaging for early detection and treatment. The integration of GO with 3D cultured organoid systems, particularly in pancreatic cancer research, is critically analyzed, highlighting current limitations and future potential. This innovative approach has the promise to transform personalized medicine, improve drug screening efficiency, and aid biomarker discovery in this aggressive disease. Through this review, we offer a balanced perspective on the advancements and future prospects in pancreatic cancer research, harnessing the potential of organoids and GO.
Collapse
Affiliation(s)
| | - Iwona Inkielewicz-Stepniak
- Department of Pharmaceutical Pathophysiology, Faculty of Pharmacy, Medical University of Gdańsk, 80-210 Gdańsk, Poland;
| |
Collapse
|
25
|
Li J, Liu J, Xia W, Yang H, Sha W, Chen H. Deciphering the Tumor Microenvironment of Colorectal Cancer and Guiding Clinical Treatment With Patient-Derived Organoid Technology: Progress and Challenges. Technol Cancer Res Treat 2024; 23:15330338231221856. [PMID: 38225190 PMCID: PMC10793199 DOI: 10.1177/15330338231221856] [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: 08/30/2023] [Revised: 11/10/2023] [Accepted: 11/30/2023] [Indexed: 01/17/2024] Open
Abstract
Colorectal cancer (CRC) is one of the most prevalent malignant tumors of the digestive tract worldwide. Despite notable advancements in CRC treatment, there is an urgent requirement for preclinical model systems capable of accurately predicting drug efficacy in CRC patients, to identify more effective therapeutic options. In recent years, substantial strides have been made in the field of organoid technology, patient-derived organoid models can phenotypically replicate the original intra-tumor and inter-tumor heterogeneity of CRC, reflecting cellular interactions of the tumor microenvironment. Patient-derived organoid models have become an indispensable tool for investigating the pathogenesis of CRC and facilitating translational research. This review focuses on the application of organoid technology in CRC modeling, tumor microenvironment, and guiding clinical treatment, particularly in drug screening and personalized medicine. It also examines the existing challenges encountered in clinical organoid research and provides a prospective outlook on the future development directions of clinical organoid research, encompassing the standardization of organoid culture technology and the application of tissue engineering technology.
Collapse
Affiliation(s)
- Jingwei Li
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Jianhua Liu
- The Second School of Clinical Medicine, Southern Medical University, Guangzhou, China
| | - Wuzheng Xia
- Department of Organ Transplantation, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Hongwei Yang
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, China
| | - Weihong Sha
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Hao Chen
- Department of Gastroenterology, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
26
|
Suhar RA, Huang MS, Navarro RS, Aviles Rodriguez G, Heilshorn SC. A Library of Elastin-like Proteins with Tunable Matrix Ligands for In Vitro 3D Neural Cell Culture. Biomacromolecules 2023; 24:5926-5939. [PMID: 37988588 DOI: 10.1021/acs.biomac.3c00941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2023]
Abstract
Hydrogels with encapsulated cells have widespread biomedical applications, both as tissue-mimetic 3D cultures in vitro and as tissue-engineered therapies in vivo. Within these hydrogels, the presentation of cell-instructive extracellular matrix (ECM)-derived ligands and matrix stiffness are critical factors known to influence numerous cell behaviors. While individual ECM biopolymers can be blended together to alter the presentation of cell-instructive ligands, this typically results in hydrogels with a range of mechanical properties. Synthetic systems that allow for the facile incorporation and modulation of multiple ligands without modification of matrix mechanics are highly desirable. In the present work, we leverage protein engineering to design a family of xeno-free hydrogels (i.e., devoid of animal-derived components) consisting of recombinant hyaluronan and recombinant elastin-like proteins (ELPs), cross-linked together with dynamic covalent bonds. The ELP components incorporate cell-instructive peptide ligands derived from ECM proteins, including fibronectin (RGD), laminin (IKVAV and YIGSR), collagen (DGEA), and tenascin-C (PLAEIDGIELTY and VFDNFVL). By carefully designing the protein primary sequence, we form 3D hydrogels with defined and tunable concentrations of cell-instructive ligands that have similar matrix mechanics. Utilizing this system, we demonstrate that neurite outgrowth from encapsulated embryonic dorsal root ganglion (DRG) cultures is significantly modified by cell-instructive ligand content. Thus, this library of protein-engineered hydrogels is a cell-compatible system to systematically study cell responses to matrix-derived ligands.
Collapse
Affiliation(s)
- Riley A Suhar
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| | - Michelle S Huang
- Department of Chemical Engineering, Stanford University, Stanford, California 94305, United States
- The Institute for Chemistry, Stanford University, Engineering & Medicine for Human Health (Sarafan ChEM-H), Stanford, California 94305, United States
| | - Renato S Navarro
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| | - Giselle Aviles Rodriguez
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, United States
| |
Collapse
|
27
|
Guo Y, Liu S, Jing D, Liu N, Luo X. The construction of elastin-like polypeptides and their applications in drug delivery system and tissue repair. J Nanobiotechnology 2023; 21:418. [PMID: 37951928 PMCID: PMC10638729 DOI: 10.1186/s12951-023-02184-8] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2023] [Accepted: 11/02/2023] [Indexed: 11/14/2023] Open
Abstract
Elastin-like polypeptides (ELPs) are thermally responsive biopolymers derived from natural elastin. These peptides have a low critical solution temperature phase behavior and can be used to prepare stimuli-responsive biomaterials. Through genetic engineering, biomaterials prepared from ELPs can have unique and customizable properties. By adjusting the amino acid sequence and length of ELPs, nanostructures, such as micelles and nanofibers, can be formed. Correspondingly, ELPs have been used for improving the stability and prolonging drug-release time. Furthermore, ELPs have widespread use in tissue repair due to their biocompatibility and biodegradability. Here, this review summarizes the basic property composition of ELPs and the methods for modulating their phase transition properties, discusses the application of drug delivery system and tissue repair and clarifies the current challenges and future directions of ELPs in applications.
Collapse
Affiliation(s)
- Yingshu Guo
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China.
| | - Shiwei Liu
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Dan Jing
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Nianzu Liu
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Xiliang Luo
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China.
| |
Collapse
|
28
|
Hefferon ME, Huang MS, Liu Y, Navarro RS, de Paiva Narciso N, Zhang D, Aviles-Rodriguez G, Heilshorn SC. Cell Microencapsulation Within Engineered Hyaluronan Elastin-Like Protein (HELP) Hydrogels. Curr Protoc 2023; 3:e917. [PMID: 37929691 PMCID: PMC10629846 DOI: 10.1002/cpz1.917] [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] [Indexed: 11/07/2023]
Abstract
Three-dimensional cell encapsulation has rendered itself a staple in the tissue engineering field. Using recombinantly engineered, biopolymer-based hydrogels to encapsulate cells is especially promising due to the enhanced control and tunability it affords. Here, we describe in detail the synthesis of our hyaluronan (i.e., hyaluronic acid) and elastin-like protein (HELP) hydrogel system. In addition to validating the efficacy of our synthetic process, we also demonstrate the modularity of the HELP system. Finally, we show that cells can be encapsulated within HELP gels over a range of stiffnesses, exhibit strong viability, and respond to stiffness cues. © 2023 Wiley Periodicals LLC. Basic Protocol 1: Elastin-like protein modification with hydrazine Basic Protocol 2: Nuclear magnetic resonance quantification of elastin-like protein modification with hydrazine Basic Protocol 3: Hyaluronic acid-benzaldehyde synthesis Basic Protocol 4: Nuclear magnetic resonance quantification of hyaluronic acid-benzaldehyde Basic Protocol 5: 3D cell encapsulation in hyaluronan elastin-like protein gels.
Collapse
Affiliation(s)
- Meghan E. Hefferon
- Department of Materials Science and Engineering, Stanford University, 496 Lomita Mall, Stanford, California 94305, 605-724-6784
| | - Michelle S. Huang
- Department of Chemical Engineering, Stanford University, 443 Via Ortega, Stanford, California 94305, 650-723-4906, 605-724-6784
| | - Yueming Liu
- Department of Materials Science and Engineering, Stanford University, 496 Lomita Mall, Stanford, California 94305, 605-724-6784
| | - Renato S. Navarro
- Department of Materials Science and Engineering, Stanford University, 496 Lomita Mall, Stanford, California 94305, 605-724-6784
| | - Narelli de Paiva Narciso
- Department of Materials Science and Engineering, Stanford University, 496 Lomita Mall, Stanford, California 94305, 605-724-6784
| | - Daiyao Zhang
- Department of Chemical Engineering, Stanford University, 443 Via Ortega, Stanford, California 94305, 650-723-4906, 605-724-6784
| | - Giselle Aviles-Rodriguez
- Department of Materials Science and Engineering, Stanford University, 496 Lomita Mall, Stanford, California 94305, 605-724-6784
| | - Sarah C. Heilshorn
- Department of Materials Science and Engineering, Stanford University, 496 Lomita Mall, Stanford, California 94305, 605-724-6784
| |
Collapse
|
29
|
Nguyen AK, Molley TG, Kardia E, Ganda S, Chakraborty S, Wong SL, Ruan J, Yee BE, Mata J, Vijayan A, Kumar N, Tilley RD, Waters SA, Kilian KA. Hierarchical assembly of tryptophan zipper peptides into stress-relaxing bioactive hydrogels. Nat Commun 2023; 14:6604. [PMID: 37872151 PMCID: PMC10593748 DOI: 10.1038/s41467-023-41907-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2023] [Accepted: 09/22/2023] [Indexed: 10/25/2023] Open
Abstract
Soft materials in nature are formed through reversible supramolecular assembly of biological polymers into dynamic hierarchical networks. Rational design has led to self-assembling peptides with structural similarities to natural materials. However, recreating the dynamic functional properties inherent to natural systems remains challenging. Here we report the discovery of a short peptide based on the tryptophan zipper (trpzip) motif, that shows multiscale hierarchical ordering that leads to emergent dynamic properties. Trpzip hydrogels are antimicrobial and self-healing, with tunable viscoelasticity and unique yield-stress properties that allow immediate harvest of embedded cells through a flick of the wrist. This characteristic makes Trpzip hydrogels amenable to syringe extrusion, which we demonstrate with examples of cell delivery and bioprinting. Trpzip hydrogels display innate bioactivity, allowing propagation of human intestinal organoids with apical-basal polarization. Considering these extensive attributes, we anticipate the Trpzip motif will prove a versatile building block for supramolecular assembly of soft materials for biotechnology and medicine.
Collapse
Affiliation(s)
- Ashley K Nguyen
- School of Chemistry, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Center for Nanomedicine, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Thomas G Molley
- School of Chemistry, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Center for Nanomedicine, University of New South Wales, Sydney, NSW, 2052, Australia
- School of Materials Science and Engineering, University of New South Wales Sydney, Sydney, NSW, 2052, Australia
| | - Egi Kardia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, 2052, Australia
- Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales, Sydney, NSW, 2052, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Sylvia Ganda
- School of Chemistry, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Center for Nanomedicine, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Sudip Chakraborty
- School of Chemistry, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Sharon L Wong
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, 2052, Australia
- Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales, Sydney, NSW, 2052, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Juanfang Ruan
- Electron Microscopy Unit, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Bethany E Yee
- School of Chemistry, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Center for Nanomedicine, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Jitendra Mata
- School of Chemistry, University of New South Wales, Sydney, NSW, 2052, Australia
- Australian Centre for Neutron Scattering, Australian Nuclear Science and Technology Organization, Lucas Heights, NSW, 2234, Australia
| | - Abhishek Vijayan
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, 2052, Australia
- Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales, Sydney, NSW, 2052, Australia
- School of Biotechnology and Biomolecular Sciences, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Naresh Kumar
- School of Chemistry, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Richard D Tilley
- School of Chemistry, University of New South Wales, Sydney, NSW, 2052, Australia
- Electron Microscopy Unit, Mark Wainwright Analytical Centre, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Shafagh A Waters
- Australian Center for Nanomedicine, University of New South Wales, Sydney, NSW, 2052, Australia
- School of Biomedical Sciences, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, 2052, Australia
- Molecular and Integrative Cystic Fibrosis Research Centre (miCF_RC), University of New South Wales, Sydney, NSW, 2052, Australia
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, 2052, Australia
- Department of Respiratory Medicine, Sydney Children's Hospital, Randwick, NSW, 2031, Australia
| | - Kristopher A Kilian
- School of Chemistry, University of New South Wales, Sydney, NSW, 2052, Australia.
- Australian Center for Nanomedicine, University of New South Wales, Sydney, NSW, 2052, Australia.
- School of Materials Science and Engineering, University of New South Wales Sydney, Sydney, NSW, 2052, Australia.
- School of Clinical Medicine, Faculty of Medicine and Health, University of New South Wales, Sydney, NSW, 2052, Australia.
| |
Collapse
|
30
|
Roth JG, Huang MS, Navarro RS, Akram JT, LeSavage BL, Heilshorn SC. Tunable hydrogel viscoelasticity modulates human neural maturation. SCIENCE ADVANCES 2023; 9:eadh8313. [PMID: 37862423 PMCID: PMC10588948 DOI: 10.1126/sciadv.adh8313] [Citation(s) in RCA: 34] [Impact Index Per Article: 17.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 09/15/2023] [Indexed: 10/22/2023]
Abstract
Human-induced pluripotent stem cells (hiPSCs) have emerged as a promising in vitro model system for studying neurodevelopment. However, current models remain limited in their ability to incorporate tunable biomechanical signaling cues imparted by the extracellular matrix (ECM). The native brain ECM is viscoelastic and stress-relaxing, exhibiting a time-dependent response to an applied force. To recapitulate the remodelability of the neural ECM, we developed a family of protein-engineered hydrogels that exhibit tunable stress relaxation rates. hiPSC-derived neural progenitor cells (NPCs) encapsulated within these gels underwent relaxation rate-dependent maturation. Specifically, NPCs within hydrogels with faster stress relaxation rates extended longer, more complex neuritic projections, exhibited decreased metabolic activity, and expressed higher levels of genes associated with neural maturation. By inhibiting actin polymerization, we observed decreased neuritic projections and a concomitant decrease in neural maturation gene expression. Together, these results suggest that microenvironmental viscoelasticity is sufficient to bias human NPC maturation.
Collapse
Affiliation(s)
- Julien G. Roth
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, USA
- Complex in Vitro Systems, Safety Assessment, Genentech Inc., South San Francisco, CA, USA
| | - Michelle S. Huang
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Renato S. Navarro
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Jason T. Akram
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Bauer L. LeSavage
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Sarah C. Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| |
Collapse
|
31
|
de Paiva Narciso N, Navarro RS, Gilchrist A, Trigo MLM, Rodriguez GA, Heilshorn SC. Design Parameters for Injectable Biopolymeric Hydrogels with Dynamic Covalent Chemistry Crosslinks. Adv Healthc Mater 2023; 12:e2301265. [PMID: 37389811 PMCID: PMC10638947 DOI: 10.1002/adhm.202301265] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2023] [Revised: 06/28/2023] [Accepted: 06/29/2023] [Indexed: 07/01/2023]
Abstract
Dynamic covalent chemistry (DCC) crosslinks can form hydrogels with tunable mechanical properties permissive to injectability and self-healing. However, not all hydrogels with transient crosslinks are easily extrudable. For this reason, two additional design parameters must be considered when formulating DCC-crosslinked hydrogels: 1) degree of functionalization (DoF) and 2) polymer molecular weight (MW). To investigate these parameters, hydrogels comprised of two recombinant biopolymers: 1) a hyaluronic acid (HA) modified with benzaldehyde and 2) an elastin-like protein (ELP) modified with hydrazine (ELP-HYD), are formulated. Several hydrogel families are synthesized with distinct HA MW and DoF while keeping the ELP-HYD component constant. The resulting hydrogels have a range of stiffnesses, G' ≈ 10-1000 Pa, and extrudability, which is attributed to the combined effects of DCC crosslinks and polymer entanglements. In general, lower MW formulations require lower forces for injectability, regardless of stiffness. Higher DoF formulations exhibit more rapid self-healing. Gel extrusion through a cannula (2 m length, 0.25 mm diameter) demonstrates the potential for minimally invasive delivery for future biomedical applications. In summary, this work highlights additional parameters that influence the injectability and network formation of DCC-crosslinked hydrogels and aims to guide future design of injectable hydrogels.
Collapse
Affiliation(s)
| | - Renato S. Navarro
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| | - Aidan Gilchrist
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| | - Miriam L. M. Trigo
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| | | | - Sarah C. Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA 94305, USA
| |
Collapse
|
32
|
Shayan M, Huang MS, Navarro R, Chiang G, Hu C, Oropeza BP, Johansson PK, Suhar RA, Foster AA, LeSavage BL, Zamani M, Enejder A, Roth JG, Heilshorn SC, Huang NF. Elastin-like protein hydrogels with controllable stress relaxation rate and stiffness modulate endothelial cell function. J Biomed Mater Res A 2023; 111:896-909. [PMID: 36861665 PMCID: PMC10159914 DOI: 10.1002/jbm.a.37520] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 01/26/2023] [Accepted: 02/15/2023] [Indexed: 03/03/2023]
Abstract
Mechanical cues from the extracellular matrix (ECM) regulate vascular endothelial cell (EC) morphology and function. Since naturally derived ECMs are viscoelastic, cells respond to viscoelastic matrices that exhibit stress relaxation, in which a cell-applied force results in matrix remodeling. To decouple the effects of stress relaxation rate from substrate stiffness on EC behavior, we engineered elastin-like protein (ELP) hydrogels in which dynamic covalent chemistry (DCC) was used to crosslink hydrazine-modified ELP (ELP-HYD) and aldehyde/benzaldehyde-modified polyethylene glycol (PEG-ALD/PEG-BZA). The reversible DCC crosslinks in ELP-PEG hydrogels create a matrix with independently tunable stiffness and stress relaxation rate. By formulating fast-relaxing or slow-relaxing hydrogels with a range of stiffness (500-3300 Pa), we examined the effect of these mechanical properties on EC spreading, proliferation, vascular sprouting, and vascularization. The results show that both stress relaxation rate and stiffness modulate endothelial spreading on two-dimensional substrates, on which ECs exhibited greater cell spreading on fast-relaxing hydrogels up through 3 days, compared with slow-relaxing hydrogels at the same stiffness. In three-dimensional hydrogels encapsulating ECs and fibroblasts in coculture, the fast-relaxing, low-stiffness hydrogels produced the widest vascular sprouts, a measure of vessel maturity. This finding was validated in a murine subcutaneous implantation model, in which the fast-relaxing, low-stiffness hydrogel produced significantly more vascularization compared with the slow-relaxing, low-stiffness hydrogel. Together, these results suggest that both stress relaxation rate and stiffness modulate endothelial behavior, and that the fast-relaxing, low-stiffness hydrogels supported the highest capillary density in vivo.
Collapse
Affiliation(s)
- Mahdis Shayan
- Department of Cardiothoracic Surgery, Stanford University, Palo Alto, CA, USA
- The Stanford Cardiovascular Institute, Stanford University, Palo Alto, CA, USA
| | - Michelle S. Huang
- Department of Chemical Engineering, Stanford University, Palo Alto, CA, USA
| | - Renato Navarro
- Department of Materials Science & Engineering, Stanford University, Palo Alto, CA, USA
| | - Gladys Chiang
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Caroline Hu
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Beu P. Oropeza
- Department of Cardiothoracic Surgery, Stanford University, Palo Alto, CA, USA
- The Stanford Cardiovascular Institute, Stanford University, Palo Alto, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| | - Patrik K. Johansson
- Geballe Laboratory for Advanced Materials, Stanford University, Palo Alto, CA, USA
| | - Riley A. Suhar
- Department of Materials Science & Engineering, Stanford University, Palo Alto, CA, USA
| | - Abbygail A. Foster
- Department of Materials Science & Engineering, Stanford University, Palo Alto, CA, USA
| | - Bauer L. LeSavage
- Department of Bioengineering, Stanford University, Palo Alto, CA, USA
| | - Maedeh Zamani
- Department of Cardiothoracic Surgery, Stanford University, Palo Alto, CA, USA
- The Stanford Cardiovascular Institute, Stanford University, Palo Alto, CA, USA
| | - Annika Enejder
- Geballe Laboratory for Advanced Materials, Stanford University, Palo Alto, CA, USA
| | - Julien G. Roth
- Institute for Stem Cell Biology & Regenerative Medicine, Stanford University School of Medicine, Palo Alto, CA, USA
| | - Sarah C. Heilshorn
- The Stanford Cardiovascular Institute, Stanford University, Palo Alto, CA, USA
- Department of Chemical Engineering, Stanford University, Palo Alto, CA, USA
- Department of Materials Science & Engineering, Stanford University, Palo Alto, CA, USA
- Geballe Laboratory for Advanced Materials, Stanford University, Palo Alto, CA, USA
| | - Ngan F. Huang
- Department of Cardiothoracic Surgery, Stanford University, Palo Alto, CA, USA
- The Stanford Cardiovascular Institute, Stanford University, Palo Alto, CA, USA
- Department of Chemical Engineering, Stanford University, Palo Alto, CA, USA
- Veterans Affairs Palo Alto Health Care System, Palo Alto, CA, USA
| |
Collapse
|
33
|
Madl CM. Accelerating aging with dynamic biomaterials: Recapitulating aged tissue phenotypes in engineered platforms. iScience 2023; 26:106825. [PMID: 37250776 PMCID: PMC10213044 DOI: 10.1016/j.isci.2023.106825] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2023] Open
Abstract
Aging is characterized by progressive decline in tissue function and represents the greatest risk factor for many diseases. Nevertheless, many fundamental mechanisms driving human aging remain poorly understood. Aging studies using model organisms are often limited in their applicability to humans. Mechanistic studies of human aging rely on relatively simple cell culture models that fail to replicate mature tissue function, making them poor surrogates for aged tissues. These culture systems generally lack well-controlled cellular microenvironments that capture the changes in tissue mechanics and microstructure that occur during aging. Biomaterial platforms presenting dynamic, physiologically relevant mechanical, structural, and biochemical cues can capture the complex changes in the cellular microenvironment in a well-defined manner, accelerating the process of cellular aging in model laboratory systems. By enabling selective tuning of relevant microenvironmental parameters, these biomaterials systems may enable identification of new therapeutic approaches to slow or reverse the detrimental effects of aging.
Collapse
Affiliation(s)
- Christopher M. Madl
- Department of Materials Science and Engineering, School of Engineering and Applied Sciences, University of Pennsylvania, Philadelphia, PA 19104, USA
| |
Collapse
|
34
|
Lee JC, Brien HJ, Walton BL, Eidman ZM, Toda S, Lim WA, Brunger JM. Instructional materials that control cellular activity through synthetic Notch receptors. Biomaterials 2023; 297:122099. [PMID: 37023529 PMCID: PMC10320837 DOI: 10.1016/j.biomaterials.2023.122099] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2022] [Revised: 03/13/2023] [Accepted: 03/22/2023] [Indexed: 03/31/2023]
Abstract
The field of regenerative engineering relies primarily on the dual technical platforms of cell selection/conditioning and biomaterial fabrication to support directed cell differentiation. As the field has matured, an appreciation for the influence of biomaterials on cell behaviors has resulted in engineered matrices that meet biomechanical and biochemical demands of target pathologies. Yet, despite advances in methods to produce designer matrices, regenerative engineers remain unable to reliably orchestrate behaviors of therapeutic cells in situ. Here, we present a platform named MATRIX whereby cellular responses to biomaterials can be custom defined by combining engineered materials with cells expressing cognate synthetic biology control modules. Such privileged channels of material-to-cell communication can activate synthetic Notch receptors and govern activities as diverse as transcriptome engineering, inflammation attenuation, and pluripotent stem cell differentiation, all in response to materials decorated with otherwise bioinert ligands. Further, we show that engineered cellular behaviors are confined to programmed biomaterial surfaces, highlighting the potential to use this platform to spatially organize cellular responses to bulk, soluble factors. This integrated approach of co-engineering cells and biomaterials for orthogonal interactions opens new avenues for reproducible control of cell-based therapies and tissue replacements.
Collapse
Affiliation(s)
- Joanne C Lee
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - Hannah J Brien
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - Bonnie L Walton
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - Zachary M Eidman
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA
| | - Satoshi Toda
- WPI Nano Life Science Institute (NanoLSI), Kanazawa University, Kanazawa, Ishikawa, Japan
| | - Wendell A Lim
- Cell Design Institute and Department of Cellular and Molecular Pharmacology, University of California, San Francisco, San Francisco, CA, 94158, USA.
| | - Jonathan M Brunger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37212, USA; Center for Stem Cell Biology, Vanderbilt University, Nashville, TN, 37212, USA.
| |
Collapse
|
35
|
Yang R, Yu Y. Patient-derived organoids in translational oncology and drug screening. Cancer Lett 2023; 562:216180. [PMID: 37061121 DOI: 10.1016/j.canlet.2023.216180] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/04/2023] [Accepted: 04/07/2023] [Indexed: 04/17/2023]
Abstract
Patient-derived organoids (PDO) are a new biomedical research model that can reconstruct phenotypic and genetic characteristics of the original tissue and are useful for research on pathogenesis and drug screening. To introduce the progression in this field, we review the key factors of constructing organoids derived from epithelial tissues and cancers, covering culture medium and matrix, morphological characteristics, genetic profiles, high-throughput drug screening, and application potential. We also discuss the co-culture system of cancer organoids with tumor microenvironment (TME) associated cells. The co-culture system is widely used in evaluating crosstalk of cancer cells with TME components, such as fibroblasts, endothelial cells, immune cells, and microorganisms. The article provides a prospective for standardized cultivation mode, automatic morphological evaluation, and drug sensitivity screening using high-throughput methods.
Collapse
Affiliation(s)
- Ruixin Yang
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China
| | - Yingyan Yu
- Department of General Surgery of Ruijin Hospital, Shanghai Institute of Digestive Surgery, and Shanghai Key Laboratory for Gastric Neoplasms, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| |
Collapse
|
36
|
Hull SM, Lou J, Lindsay CD, Navarro RS, Cai B, Brunel LG, Westerfield AD, Xia Y, Heilshorn SC. 3D bioprinting of dynamic hydrogel bioinks enabled by small molecule modulators. SCIENCE ADVANCES 2023; 9:eade7880. [PMID: 37000873 PMCID: PMC10065439 DOI: 10.1126/sciadv.ade7880] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 02/24/2023] [Indexed: 06/19/2023]
Abstract
Three-dimensional bioprinting has emerged as a promising tool for spatially patterning cells to fabricate models of human tissue. Here, we present an engineered bioink material designed to have viscoelastic mechanical behavior, similar to that of living tissue. This viscoelastic bioink is cross-linked through dynamic covalent bonds, a reversible bond type that allows for cellular remodeling over time. Viscoelastic materials are challenging to use as inks, as one must tune the kinetics of the dynamic cross-links to allow for both extrudability and long-term stability. We overcome this challenge through the use of small molecule catalysts and competitors that temporarily modulate the cross-linking kinetics and degree of network formation. These inks were then used to print a model of breast cancer cell invasion, where the inclusion of dynamic cross-links was found to be required for the formation of invasive protrusions. Together, we demonstrate the power of engineered, dynamic bioinks to recapitulate the native cellular microenvironment for disease modeling.
Collapse
Affiliation(s)
- Sarah M. Hull
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | - Junzhe Lou
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | | | - Renato S. Navarro
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Betty Cai
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Lucia G. Brunel
- Department of Chemical Engineering, Stanford University, Stanford, CA, USA
| | | | - Yan Xia
- Department of Chemistry, Stanford University, Stanford, CA, USA
| | - Sarah C. Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| |
Collapse
|
37
|
Blatchley MR, Anseth KS. Middle-out methods for spatiotemporal tissue engineering of organoids. NATURE REVIEWS BIOENGINEERING 2023; 1:329-345. [PMID: 37168734 PMCID: PMC10010248 DOI: 10.1038/s44222-023-00039-3] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 02/03/2023] [Indexed: 05/13/2023]
Abstract
Organoids recapitulate many aspects of the complex three-dimensional (3D) organization found within native tissues and even display tissue and organ-level functionality. Traditional approaches to organoid culture have largely employed a top-down tissue engineering strategy, whereby cells are encapsulated in a 3D matrix, such as Matrigel, alongside well-defined biochemical cues that direct morphogenesis. However, the lack of spatiotemporal control over niche properties renders cellular processes largely stochastic. Therefore, bottom-up tissue engineering approaches have evolved to address some of these limitations and focus on strategies to assemble tissue building blocks with defined multi-scale spatial organization. However, bottom-up design reduces the capacity for self-organization that underpins organoid morphogenesis. Here, we introduce an emerging framework, which we term middle-out strategies, that relies on existing design principles and combines top-down design of defined synthetic matrices that support proliferation and self-organization with bottom-up modular engineered intervention to limit the degrees of freedom in the dynamic process of organoid morphogenesis. We posit that this strategy will provide key advances to guide the growth of organoids with precise geometries, structures and function, thereby facilitating an unprecedented level of biomimicry to accelerate the utility of organoids to more translationally relevant applications.
Collapse
Affiliation(s)
- Michael R. Blatchley
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO USA
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO USA
| | - Kristi S. Anseth
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO USA
- The BioFrontiers Institute, University of Colorado Boulder, Boulder, CO USA
| |
Collapse
|
38
|
Tumor decellularization reveals proteomic and mechanical characteristics of the extracellular matrix of primary liver cancer. BIOMATERIALS ADVANCES 2023; 146:213289. [PMID: 36724550 DOI: 10.1016/j.bioadv.2023.213289] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 01/09/2023] [Accepted: 01/10/2023] [Indexed: 01/19/2023]
Abstract
Tumor initiation and progression are critically dependent on interaction of cancer cells with their cellular and extracellular microenvironment. Alterations in the composition, integrity, and mechanical properties of the extracellular matrix (ECM) dictate tumor processes including cell proliferation, migration, and invasion. Also in primary liver cancer, consisting of hepatocellular carcinoma (HCC) and cholangiocarcinoma (CCA), the dysregulation of the extracellular environment by liver fibrosis and tumor desmoplasia is pertinent. Yet, the exact changes occurring in liver cancer ECM remain uncharacterized and underlying tumor-promoting mechanisms remain largely unknown. Herein, an integrative molecular and mechanical approach is used to extensively characterize the ECM of HCC and CCA tumors by utilizing an optimized decellularization technique. We identified a myriad of proteins in both tumor and adjacent liver tissue, uncovering distinct malignancy-related ECM signatures. The resolution of this approach unveiled additional ECM-related proteins compared to large liver cancer transcriptomic datasets. The differences in ECM protein composition resulted in divergent mechanical properties on a macro- and micro-scale that are tumor-type specific. Furthermore, the decellularized tumor ECM was employed to create a tumor-specific hydrogel that supports patient-derived tumor organoids, which provides a new avenue for personalized medicine applications. Taken together, this study contributes to a better understanding of alterations to composition, stiffness, and collagen alignment of the tumor ECM that occur during liver cancer development.
Collapse
|
39
|
Yavitt FM, Kirkpatrick BE, Blatchley MR, Speckl KF, Mohagheghian E, Moldovan R, Wang N, Dempsey PJ, Anseth KS. In situ modulation of intestinal organoid epithelial curvature through photoinduced viscoelasticity directs crypt morphogenesis. SCIENCE ADVANCES 2023; 9:eadd5668. [PMID: 36662859 PMCID: PMC9858500 DOI: 10.1126/sciadv.add5668] [Citation(s) in RCA: 27] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Accepted: 12/15/2022] [Indexed: 06/17/2023]
Abstract
Spatiotemporally coordinated transformations in epithelial curvature are necessary to generate crypt-villus structures during intestinal development. However, the temporal regulation of mechanotransduction pathways that drive crypt morphogenesis remains understudied. Intestinal organoids have proven useful to study crypt morphogenesis in vitro, yet the reliance on static culture scaffolds limits the ability to assess the temporal effects of changing curvature. Here, a photoinduced hydrogel cross-link exchange reaction is used to spatiotemporally alter epithelial curvature and study how dynamic changes in curvature influence mechanotransduction pathways to instruct crypt morphogenesis. Photopatterned curvature increased membrane tension and depolarization, which was required for subsequent nuclear localization of yes-associated protein 1 (YAP) observed 24 hours following curvature change. Curvature-directed crypt morphogenesis only occurred following a delay in the induction of differentiation that coincided with the delay in spatially restricted YAP localization, indicating that dynamic changes in curvature initiate epithelial curvature-dependent mechanotransduction pathways that temporally regulate crypt morphogenesis.
Collapse
Affiliation(s)
- F. Max Yavitt
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80309, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Bruce E. Kirkpatrick
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80309, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80309, USA
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Michael R. Blatchley
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80309, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Kelly F. Speckl
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80309, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80309, USA
| | - Erfan Mohagheghian
- Department of Mechanical Science and Engineering, The Grainger College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Radu Moldovan
- Advanced Light Microscopy Core Facility, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, USA
| | - Ning Wang
- Department of Mechanical Science and Engineering, The Grainger College of Engineering, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Peter J. Dempsey
- Section of Developmental Biology, Department of Pediatrics, University of Colorado, Denver, CO 80204, USA
| | - Kristi S. Anseth
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO 80309, USA
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO 80309, USA
| |
Collapse
|
40
|
Londoño-Berrio M, Castro C, Cañas A, Ortiz I, Osorio M. Advances in Tumor Organoids for the Evaluation of Drugs: A Bibliographic Review. Pharmaceutics 2022; 14:pharmaceutics14122709. [PMID: 36559203 PMCID: PMC9784359 DOI: 10.3390/pharmaceutics14122709] [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: 11/01/2022] [Revised: 11/25/2022] [Accepted: 11/27/2022] [Indexed: 12/11/2022] Open
Abstract
Tumor organoids are defined as self-organized three-dimensional assemblies of heterogeneous cell types derived from patient samples that mimic the key histopathological, genetic, and phenotypic characteristics of the original tumor. This technology is proposed as an ideal candidate for the evaluation of possible therapies against cancer, presenting advantages over other models which are currently used. However, there are no reports in the literature that relate the techniques and material development of tumor organoids or that emphasize in the physicochemical and biological properties of materials that intent to biomimicry the tumor extracellular matrix. There is also little information regarding the tools to identify the correspondence of native tumors and tumoral organoids (tumoroids). Moreover, this paper relates the advantages of organoids compared to other models for drug evaluation. A growing interest in tumoral organoids has arisen from 2009 to the present, aimed at standardizing the process of obtaining organoids, which more accurately resemble patient-derived tumor tissue. Likewise, it was found that the characteristics to consider for the development of organoids, and therapeutic responses of them, are cell morphology, physiology, the interaction between cells, the composition of the cellular matrix, and the genetic, phenotypic, and epigenetic characteristics. Currently, organoids have been used for the evaluation of drugs for brain, lung, and colon tumors, among others. In the future, tumor organoids will become closer to being considered a better model for studying cancer in clinical practice, as they can accurately mimic the characteristics of tumors, in turn ensuring that the therapeutic response aligns with the clinical response of patients.
Collapse
Affiliation(s)
- Maritza Londoño-Berrio
- Systems Biology Research Group, Pontifical Bolivarian University (Universidad Pontificia Bolivariana), Carrera 78B No. 72a-109, Medellin 050034, Colombia
| | - Cristina Castro
- New Materials Research Group, School of Engineering, Pontifical Bolivarian University, Circular 1 No. 70-01, Medellin 050031, Colombia
| | - Ana Cañas
- Corporation for Biological Research, Medical, and Experimental Research Group, Carrera 72A # 78b-141, Medellin 050034, Colombia
| | - Isabel Ortiz
- Systems Biology Research Group, Pontifical Bolivarian University (Universidad Pontificia Bolivariana), Carrera 78B No. 72a-109, Medellin 050034, Colombia
| | - Marlon Osorio
- Systems Biology Research Group, Pontifical Bolivarian University (Universidad Pontificia Bolivariana), Carrera 78B No. 72a-109, Medellin 050034, Colombia
- New Materials Research Group, School of Engineering, Pontifical Bolivarian University, Circular 1 No. 70-01, Medellin 050031, Colombia
- Correspondence:
| |
Collapse
|
41
|
Günther C, Winner B, Neurath MF, Stappenbeck TS. Organoids in gastrointestinal diseases: from experimental models to clinical translation. Gut 2022; 71:1892-1908. [PMID: 35636923 PMCID: PMC9380493 DOI: 10.1136/gutjnl-2021-326560] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Accepted: 05/13/2022] [Indexed: 12/12/2022]
Abstract
We are entering an era of medicine where increasingly sophisticated data will be obtained from patients to determine proper diagnosis, predict outcomes and direct therapies. We predict that the most valuable data will be produced by systems that are highly dynamic in both time and space. Three-dimensional (3D) organoids are poised to be such a highly valuable system for a variety of gastrointestinal (GI) diseases. In the lab, organoids have emerged as powerful systems to model molecular and cellular processes orchestrating natural and pathophysiological human tissue formation in remarkable detail. Preclinical studies have impressively demonstrated that these organs-in-a-dish can be used to model immunological, neoplastic, metabolic or infectious GI disorders by taking advantage of patient-derived material. Technological breakthroughs now allow to study cellular communication and molecular mechanisms of interorgan cross-talk in health and disease including communication along for example, the gut-brain axis or gut-liver axis. Despite considerable success in culturing classical 3D organoids from various parts of the GI tract, some challenges remain to develop these systems to best help patients. Novel platforms such as organ-on-a-chip, engineered biomimetic systems including engineered organoids, micromanufacturing, bioprinting and enhanced rigour and reproducibility will open improved avenues for tissue engineering, as well as regenerative and personalised medicine. This review will highlight some of the established methods and also some exciting novel perspectives on organoids in the fields of gastroenterology. At present, this field is poised to move forward and impact many currently intractable GI diseases in the form of novel diagnostics and therapeutics.
Collapse
Affiliation(s)
- Claudia Günther
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Beate Winner
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Department of Stem Cell Biology, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
- Center of Rare Diseases Erlangen (ZSEER), University Hospital Erlangen, Friedrich-Alexander University of Erlangen-Nürnberg (FAU), Erlangen, Germany
| | - Markus F Neurath
- Department of Medicine 1, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
- Deutsches Zentrum Immuntherapie, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Thaddeus S Stappenbeck
- Department of Inflammation and Immunity, Lerner Research Institute, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
42
|
Lou J, Mooney DJ. Chemical strategies to engineer hydrogels for cell culture. Nat Rev Chem 2022; 6:726-744. [PMID: 37117490 DOI: 10.1038/s41570-022-00420-7] [Citation(s) in RCA: 146] [Impact Index Per Article: 48.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/27/2022] [Indexed: 12/12/2022]
Abstract
Two-dimensional and three-dimensional cell culture systems are widely used for biological studies, and are the basis of the organoid, tissue engineering and organ-on-chip research fields in applications such as disease modelling and drug screening. The natural extracellular matrix of tissues, a complex scaffold with varying chemical and mechanical properties, has a critical role in regulating important cellular functions such as spreading, migration, proliferation and differentiation, as well as tissue morphogenesis. Hydrogels are biomaterials that are used in cell culture systems to imitate critical features of a natural extracellular matrix. Chemical strategies to synthesize and tailor the properties of these hydrogels in a controlled manner, and manipulate their biological functions in situ, have been developed. In this Review, we provide the rational design criteria for predictably engineering hydrogels to mimic the properties of the natural extracellular matrix. We highlight the advances in using biocompatible strategies to engineer hydrogels for cell culture along with recent developments to dynamically control the cellular environment by exploiting stimuli-responsive chemistries. Finally, future opportunities to engineer hydrogels are discussed, in which the development of novel chemical methods will probably have an important role.
Collapse
|
43
|
Navarro RS, Huang MS, Roth JG, Hubka KM, Long CM, Enejder A, Heilshorn SC. Tuning Polymer Hydrophilicity to Regulate Gel Mechanics and Encapsulated Cell Morphology. Adv Healthc Mater 2022; 11:e2200011. [PMID: 35373510 PMCID: PMC9262823 DOI: 10.1002/adhm.202200011] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Revised: 03/20/2022] [Indexed: 12/20/2022]
Abstract
Mechanically tunable hydrogels are attractive platforms for 3D cell culture, as hydrogel stiffness plays an important role in cell behavior. Traditionally, hydrogel stiffness has been controlled through altering either the polymer concentration or the stoichiometry between crosslinker reactive groups. Here, an alternative strategy based upon tuning the hydrophilicity of an elastin-like protein (ELP) is presented. ELPs undergo a phase transition that leads to protein aggregation at increasing temperatures. It is hypothesized that increasing this transition temperature through bioconjugation with azide-containing molecules of increasing hydrophilicity will allow direct control of the resulting gel stiffness by making the crosslinking groups more accessible. These azide-modified ELPs are crosslinked into hydrogels with bicyclononyne-modified hyaluronic acid (HA-BCN) using bioorthogonal, click chemistry, resulting in hydrogels with tunable storage moduli (100-1000 Pa). Human mesenchymal stromal cells (hMSCs), human umbilical vein endothelial cells (HUVECs), and human neural progenitor cells (hNPCs) are all observed to alter their cell morphology when encapsulated within hydrogels of varying stiffness. Taken together, the use of protein hydrophilicity as a lever to tune hydrogel mechanical properties is demonstrated. These hydrogels have tunable moduli over a stiffness range relevant to soft tissues, support the viability of encapsulated cells, and modify cell spreading as a consequence of gel stiffness.
Collapse
Affiliation(s)
- Renato S Navarro
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Michelle S Huang
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Julien G Roth
- Institute for Stem Cell Biology and Regenerative Medicine, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Kelsea M Hubka
- Maternal and Child Health Research Institute, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | - Chris M Long
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Annika Enejder
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, 94305, USA
| |
Collapse
|
44
|
Advances of Engineered Hydrogel Organoids within the Stem Cell Field: A Systematic Review. Gels 2022; 8:gels8060379. [PMID: 35735722 PMCID: PMC9222364 DOI: 10.3390/gels8060379] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/10/2022] [Accepted: 06/13/2022] [Indexed: 02/06/2023] Open
Abstract
Organoids are novel in vitro cell culture models that enable stem cells (including pluripotent stem cells and adult stem cells) to grow and undergo self-organization within a three-dimensional microenvironment during the process of differentiation into target tissues. Such miniature structures not only recapitulate the histological and genetic characteristics of organs in vivo, but also form tissues with the capacity for self-renewal and further differentiation. Recent advances in biomaterial technology, particularly hydrogels, have provided opportunities to improve organoid cultures; by closely integrating the mechanical and chemical properties of the extracellular matrix microenvironment, with novel synthetic materials and stem cell biology. This systematic review critically examines recent advances in various strategies and techniques utilized for stem-cell-derived organoid culture, with particular emphasis on the application potential of hydrogel technology in organoid culture. We hope this will give a better understanding of organoid cultures for modelling diseases and tissue engineering applications.
Collapse
|
45
|
Suhar RA, Doulames VM, Liu Y, Hefferon ME, Figueroa O, Buabbas H, Heilshorn SC. Hyaluronan and elastin-like protein (HELP) gels significantly improve microsphere retention in the myocardium. Biomater Sci 2022; 10:2590-2608. [PMID: 35411353 PMCID: PMC9123900 DOI: 10.1039/d1bm01890f] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2023]
Abstract
Heart disease is the leading cause of death globally, and delivery of therapeutic cargo (e.g., particles loaded with proteins, drugs, or genes and cells) through direct injection into the myocardium is a promising clinical intervention. However, retention of deliverables to the contracting myocardium is low, with as much as 60-90% of payload being lost within 24 hr. Commercially-available injectable hydrogels, including Matrigel, have been hypothesized to increase payload retention but have not yielded significant improvements in quantified analyses. Here, we assess a recombinant hydrogel composed of chemically modified hyaluronan and elastin-like protein (HELP) as an alternative injectable carrier to increase cargo retention. HELP is crosslinked using dynamic covalent bonds, and tuning the hyaluronan chemistry significantly alters hydrogel mechanical properties including stiffness, stress relaxation rate, and ease of injectability through a needle or catheter. These materials can be injected even after complete crosslinking, extending the time window for surgical delivery. We show that HELP gels significantly improve in vivo retention of microsphere cargo compared to Matrigel, both 1 day and 7 days post-injection directly into the rat myocardium. These data suggest that HELP gels may assist with the clinical translation of therapeutic cargo designed for delivery into the contracting myocardium by preventing acute cargo loss.
Collapse
Affiliation(s)
- Riley A Suhar
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA.
| | - Vanessa M Doulames
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA.
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California 94305, USA
| | - Yueming Liu
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA.
| | - Meghan E Hefferon
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA.
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, California 94305, USA
| | | | - Hana Buabbas
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA.
- Department of Biology, Stanford University, Stanford, California, 94305, USA
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305, USA.
| |
Collapse
|
46
|
Yavitt FM, Kirkpatrick BE, Blatchley MR, Anseth KS. 4D Materials with Photoadaptable Properties Instruct and Enhance Intestinal Organoid Development. ACS Biomater Sci Eng 2022; 8:4634-4638. [PMID: 35298149 DOI: 10.1021/acsbiomaterials.1c01450] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Intestinal organoids are self-organized tissue constructs, grown in vitro, that resemble the structure and function of the intestine and are often considered promising as a prospective platform for drug testing and disease modeling. Organoid development in vitro is typically instructed by exogenous cues delivered from the media, but cellular responses also depend on properties of the surrounding microenvironmental niche, such as mechanical stiffness and extracellular matrix (ECM) ligands. In recent years, synthetic hydrogel platforms have been engineered to resemble the in vivo niche, with the goal of generating physiologically relevant environments that can promote mature and reproducible organoid development. However, a few of these approaches consider the importance of intestinal organoid morphology or how morphology changes during development, as cues that may dictate organoid functionality. For example, intestinal organoids grown in vitro often lack the physical boundary conditions found in vivo that are responsible for shaping a collection of cells into developmentally relevant morphologies, resulting in organoids that often differ in structure and cellular organization from the parent organ. This disconnect relates, in part, to a lack of appropriate adaptable and programmable materials for cell culture, especially those that enable control over colony growth and differentiation in space and time (i.e., 4D materials). We posit that the future of organoid culture platforms may benefit from advances in photoadaptable chemistries and integration into biomaterials scaffolds, thereby allowing greater user-directed control over both the macro- and microscale material properties. In this way, synthetic materials can begin to better replicate changes in the ECM during development or regeneration in vivo. Recapitulation of cellular and tissue morphological changes, along with an appreciation for the appropriate developmental time scales, should help instruct the next generation of organoid models to facilitate predictable outcomes.
Collapse
Affiliation(s)
| | - Bruce E Kirkpatrick
- Medical Scientist Training Program, University of Colorado Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | | | | |
Collapse
|
47
|
Barhouse PS, Andrade MJ, Smith Q. Home Away From Home: Bioengineering Advancements to Mimic the Developmental and Adult Stem Cell Niche. FRONTIERS IN CHEMICAL ENGINEERING 2022. [DOI: 10.3389/fceng.2022.832754] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
The inherent self-organizing capacity of pluripotent and adult stem cell populations has advanced our fundamental understanding of processes that drive human development, homeostasis, regeneration, and disease progression. Translating these principles into in vitro model systems has been achieved with the advent of organoid technology, driving innovation to harness patient-specific, cell-laden regenerative constructs that can be engineered to augment or replace diseased tissue. While developmental organization and regenerative adult stem cell niches are tightly regulated in vivo, in vitro analogs lack defined architecture and presentation of physicochemical cues, leading to the unhindered arrangement of mini-tissues that lack complete physiological mimicry. This review aims to highlight the recent integrative engineering approaches that elicit spatio-temporal control of the extracellular niche to direct the structural and functional maturation of pluripotent and adult stem cell derivatives. While the advances presented here leverage multi-pronged strategies ranging from synthetic biology to microfabrication technologies, the methods converge on recreating the biochemical and biophysical milieu of the native tissue to be modeled or regenerated.
Collapse
|
48
|
LeSavage BL, Suhar RA, Broguiere N, Lutolf MP, Heilshorn SC. Next-generation cancer organoids. NATURE MATERIALS 2022; 21:143-159. [PMID: 34385685 DOI: 10.1038/s41563-021-01057-5] [Citation(s) in RCA: 216] [Impact Index Per Article: 72.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2021] [Accepted: 06/21/2021] [Indexed: 05/13/2023]
Abstract
Organotypic models of patient-specific tumours are revolutionizing our understanding of cancer heterogeneity and its implications for personalized medicine. These advancements are, in part, attributed to the ability of organoid models to stably preserve genetic, proteomic, morphological and pharmacotypic features of the parent tumour in vitro, while also offering unprecedented genomic and environmental manipulation. Despite recent innovations in organoid protocols, current techniques for cancer organoid culture are inherently uncontrolled and irreproducible, owing to several non-standardized facets including cancer tissue sources and subsequent processing, medium formulations, and animal-derived three-dimensional matrices. Given the potential for cancer organoids to accurately recapitulate the intra- and intertumoral biological heterogeneity associated with patient-specific cancers, eliminating the undesirable technical variability accompanying cancer organoid culture is necessary to establish reproducible platforms that accelerate translatable insights into patient care. Here we describe the current challenges and recent multidisciplinary advancements and opportunities for standardizing next-generation cancer organoid systems.
Collapse
Affiliation(s)
- Bauer L LeSavage
- Department of Bioengineering, Stanford University, Stanford, CA, USA
| | - Riley A Suhar
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA
| | - Nicolas Broguiere
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Chemical Sciences and Engineering, School of Basic Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Matthias P Lutolf
- Laboratory of Stem Cell Bioengineering, Institute of Bioengineering, School of Life Sciences and School of Engineering, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- Institute of Chemical Sciences and Engineering, School of Basic Science, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Sarah C Heilshorn
- Department of Materials Science and Engineering, Stanford University, Stanford, CA, USA.
| |
Collapse
|
49
|
Häfliger J, Morsy Y, Scharl M, Wawrzyniak M. From Patient Material to New Discoveries: a Methodological Review and Guide for Intestinal Stem Cell Researchers. Stem Cell Rev Rep 2022; 18:1309-1321. [PMID: 35038103 DOI: 10.1007/s12015-021-10307-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/21/2021] [Indexed: 10/19/2022]
Abstract
Intestinal stem cells (ISC) are characterized by their ability to continuously self-renew and differentiate into various functionally distinct intestinal epithelial cell types. Impaired stem cell proliferation and differentiation can cause severe dysfunction of the gastrointestinal tract and lead to the development of several clinical disorders. Animal mouse models provide a valuable platform to study ISC function, disease mechanisms, and the intestinal epithelium's regenerative capacity upon tissue damage. However, advanced in vitro systems that are more relevant to human physiology are needed to understand better the diverse disease-triggering factors and the heterogeneity in clinical manifestations. Intestinal biopsies from patients might serve as potent starting material for such "gut-in-a-dish" approaches. While many promising tools for intestinal tissue processing, in vitro expansion, and downstream analysis have been developed in recent years, a comprehensive guide with recommendations to successfully launch or improve intestinal stem cell culture is missing. In this review, we present a selection of currently established methods, highlight recent publications and discuss the potential and limitations of those methodological approaches to facilitate and support the future design of novel and more personalized therapeutic options.
Collapse
Affiliation(s)
- Janine Häfliger
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Sternwartstrasse 14, 8091, Zurich, Switzerland
| | - Yasser Morsy
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Sternwartstrasse 14, 8091, Zurich, Switzerland
| | - Michael Scharl
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Sternwartstrasse 14, 8091, Zurich, Switzerland
| | - Marcin Wawrzyniak
- Department of Gastroenterology and Hepatology, University Hospital Zurich, University of Zurich, Sternwartstrasse 14, 8091, Zurich, Switzerland.
| |
Collapse
|
50
|
Poudel H, Sanford K, Szwedo PK, Pathak R, Ghosh A. Synthetic Matrices for Intestinal Organoid Culture: Implications for Better Performance. ACS OMEGA 2022; 7:38-47. [PMID: 35036676 PMCID: PMC8756583 DOI: 10.1021/acsomega.1c05136] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Accepted: 12/10/2021] [Indexed: 05/04/2023]
Abstract
Organoids are three-dimensional (3D) self-renewing and self-organizing clusters of cells that imitate an organ's structure and function, making them an important tool in various fields ranging from regenerative medicine to drug discovery. Organoids can be developed ex vivo by isolating adult stem cells from an organ-specific tissue (e.g., intestine, brain, and lung) and allowing the stem cells to grow and differentiate in an appropriate growth media with some structural support elements. A 3D extracellular matrix (ECM) hydrogel, a network of highly hydrophilic cross-linked polymer chains, provides essential support and cues for ex vivo organoid growth. Commercially available hydrogel matrices (for example, Matrigel and collagen) are primarily derived from animal tissues. Notably, these animal-derived hydrogel matrices are not suitable for controlled modifications and pose risks of immunogen and pathogen transfer, thus diminishing their clinical application. These limitations of animal-derived hydrogel matrices can, however, be overcome using synthetic hydrogel matrices based on polymers such as polyethylene glycol, nanocellulose, alginate, hyaluronic acid, and polylactic-co-glycolic acid. This review highlights some of the current approaches and advantages of developing synthetic ECM-mimic hydrogels, focusing primarily on intestinal organoid culture.
Collapse
Affiliation(s)
- Humendra Poudel
- Department
of Chemistry, University of Arkansas at
Little Rock, 2801 South University Avenue, Little Rock, Arkansas 72204, United States
| | - Karie Sanford
- Department
of Chemistry, University of Arkansas at
Little Rock, 2801 South University Avenue, Little Rock, Arkansas 72204, United States
| | - Peter K. Szwedo
- Department
of Chemistry, University of Arkansas at
Little Rock, 2801 South University Avenue, Little Rock, Arkansas 72204, United States
| | - Rupak Pathak
- Department
of Pharmaceutical Sciences, University of
Arkansas for Medical Sciences, 4301 West Markham Street, Little Rock, Arkansas 72205, United States
- . Phone: 501-603-1472. Fax: 501-686-6057
| | - Anindya Ghosh
- Department
of Chemistry, University of Arkansas at
Little Rock, 2801 South University Avenue, Little Rock, Arkansas 72204, United States
- . Phone: 501-916-6542. Fax: 501-916-3838
| |
Collapse
|