1
|
Toyofuku T, Ishikawa T, Kumanogoh A. Deletion of the plexin-D1 ectodomain leads to anoikis by suppressing integrin inside-out signaling. Mol Biol Cell 2025; 36:ar71. [PMID: 40266804 DOI: 10.1091/mbc.e25-02-0075] [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: 04/25/2025] Open
Abstract
Plexin-D1, mainly expressed in endothelial and cancer cells, regulates diverse effects, suppresses endothelial cell growth, and induces cancer cell migration and proliferation. Here, we demonstrated that plexin-D1 was cleaved by proteinase on cancer cells. To examine the role of cleaved plexin-D1 in cells, Madin-Darby canine kidney (MDCK) cells overexpressing truncated plexin-D1 were cultured in Matrigel. MDCK cells expressing plexin-D1 lacking the ectodomain (plexin-D1 ΔEC) underwent apoptosis. An adhesion assay for extracellular matrix (ECM) molecules showed that plexin-D1 ΔEC-expressing MDCK cells lost their affinity for the ECM. These results suggest that plexin-D1 ΔEC blocks integrin inside-out signaling, leading to detachment from the ECM and apoptosis, so-called anoikis. By contrast, MDCK cells expressing full-length plexin-D1 or plexin-D1 lacking the cytoplasmic domain (plexin-D1 ΔIC) developed multicellular branching tubular structures in Matrigel. This morphological change was blocked in plexin-D1-expressing MDCK cells by the hepatocyte growth factor receptor (Met) loss of function or by Met inhibitors. These results suggest that plexin-D1 associates with Met through the plexin-D1 extracellular domain, and this activates Met cytoplasmic kinase activity. We therefore conclude that plexin-D1 contains distinct domains that determine the fate of cancer cells.
Collapse
Affiliation(s)
- Toshihiko Toyofuku
- Department of Immunology and Molecular Medicine, Graduate School of Medicine, The Center of Medical Innovation and Translational Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Takako Ishikawa
- Department of Immunology and Molecular Medicine, Graduate School of Medicine, The Center of Medical Innovation and Translational Research, Osaka University, Suita, Osaka 565-0871, Japan
| | - Atsushi Kumanogoh
- Department of Respiratory Medicine and Clinical Immunology, Graduate School of Medicine, Osaka University, Suita, Osaka 565-0871, Japan
- Laboratory of Immunopathology, WPI Immunology Frontier Research Center, Osaka University, Suita, Osaka 565-0871, Japan
- Integrated Frontier Research for Medical Science Division, Institute for Open and Transdisciplinary Research Initiatives, Osaka University, Suita, Osaka 565-0871, Japan
| |
Collapse
|
2
|
Tonini L, Ahn C. Latest Advanced Techniques for Improving Intestinal Organoids Limitations. Stem Cell Rev Rep 2025:10.1007/s12015-025-10894-9. [PMID: 40388043 DOI: 10.1007/s12015-025-10894-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 05/06/2025] [Indexed: 05/20/2025]
Abstract
Intestinal organoids are valuable tools across different disciplines, from a clinical aspect to the biomedical research, providing a unique perspective on the complexity of the gastrointestinal system. They are alternatives to common cell lines as they can offer insights into architectural functionality and reduce the use of animal models. A deeper understanding of their organoid characteristics is required to harness their full potential. Despite their beneficial uses and multiple advantages, organoids have limitations that remain unaddressed. This review aims to elucidate the principal limitations of intestinal organoids, investigate structural defects such as the deficiency in a vascularized and lymphatic system, and absence of the microbiome, restrictions in mimicking the physiological gut model, including the lack of an acid-neutralizing system or a shortage of digestive enzymes, and the difficulties in their long-term maintenance and polarity accessibility. Development of innovative techniques to address these limitations will lead to improve in vivo recapitulation and pioneering further advancements in this field.
Collapse
Affiliation(s)
- Lisa Tonini
- Laboratory of Veterinary Physiology, College of Veterinary Medicine, Jeju National University, Jeju, 63243, Republic of Korea
| | - Changhwan Ahn
- Laboratory of Veterinary Physiology, College of Veterinary Medicine, Jeju National University, Jeju, 63243, Republic of Korea.
- Veterinary Medical Research Institute, Jeju National University, Jeju, 63243, Republic of Korea.
| |
Collapse
|
3
|
Kumar D, Gupta S, Gupta V, Tanwar R, Chandel A. Engineering the Future of Regenerative Medicines in Gut Health with Stem Cell-Derived Intestinal Organoids. Stem Cell Rev Rep 2025:10.1007/s12015-025-10893-w. [PMID: 40380985 DOI: 10.1007/s12015-025-10893-w] [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: 05/05/2025] [Indexed: 05/19/2025]
Abstract
The advent of intestinal organoids, three-dimensional structures derived from stem cells, has significantly advanced the field of biology by providing robust in vitro models that closely mimic the architecture and functionality of the human intestine. These organoids, generated from induced pluripotent stem cells (iPSCs), embryonic stem cells (ESCs), or adult stem cells, possess remarkable capabilities for self-renewal, differentiation into diverse intestinal cell types, and functional recapitulation of physiological processes, including nutrient absorption, epithelial barrier integrity, and host-microbe interactions. The utility of intestinal organoids has been extensively demonstrated in disease modeling, drug screening, and personalized medicine. Notable examples include iPSC-derived organoids, which have been effectively employed to model enteric infections, and ESC-derived organoids, which have provided critical insights into fetal intestinal development. Patient-derived organoids have emerged as powerful tools for investigating personalized therapeutics and regenerative interventions for conditions such as inflammatory bowel disease (IBD), cystic fibrosis, and colorectal cancer. Preclinical studies involving transplantation of human intestinal organoids into murine models have shown promising outcomes, including functional integration, epithelial restoration, and immune system interactions. Despite these advancements, several challenges persist, particularly in achieving reproducibility, scalability, and maturation of organoids, which hinder their widespread clinical translation. Addressing these limitations requires the establishment of standardized protocols for organoid generation, culture, storage, and analysis to ensure reproducibility and comparability of findings across studies. Nevertheless, intestinal organoids hold immense promise for transforming our understanding of gastrointestinal pathophysiology, enhancing drug development pipelines, and advancing personalized medicine. By bridging the gap between preclinical research and clinical applications, these organoids represent a paradigm shift in the exploration of novel therapeutic strategies and the investigation of gut-associated diseases.
Collapse
Affiliation(s)
- Dinesh Kumar
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab, India.
| | - Sonia Gupta
- Swami Devi Dyal Group of Professional Institute, Panchkula, India
| | - Vrinda Gupta
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab, India
| | - Rajni Tanwar
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab, India
| | - Anchal Chandel
- School of Pharmacy, Desh Bhagat University, Mandi Gobindgarh, Punjab, India
| |
Collapse
|
4
|
Song Y, Seitz M, Kowalczewski A, Mai NY, Jain E, Yang H, Ma Z. Mechanically and Chemically Defined PEG Hydrogels Improve Reproducibility in Human Cardioid Development. Adv Healthc Mater 2025:e2403997. [PMID: 40376871 DOI: 10.1002/adhm.202403997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2024] [Revised: 03/22/2025] [Indexed: 05/18/2025]
Abstract
Cardioids are 3D self-organized heart organoids directly derived from induced pluripotent stem cells (hiPSCs) aggregates. The growth and culture of cardioids is either conducted in suspension culture or heavily relies on Matrigel encapsulation. Despite the significant advancements in cardioid technology, reproducibility remains a major challenge, limiting their widespread use in both basic research and translational applications. Here, for the first time, we employed synthetic, matrix metalloproteinase (MMP)-degradable polyethylene glycol (PEG)-based hydrogels to define the effect of mechanical and biochemical cues on cardioid development. Successful cardiac differentiation is demonstrated in all the hydrogel conditions, while cardioid cultured in optimized PEG hydrogel (3 wt.% PEG-2mM RGD) underwent similar morphological development and comparable tissue functions to those cultured in Matrigel. Matrix stiffness and cell adhesion motif play a critical role in cardioid development, nascent chamber formation, contractile physiology, and endothelial cell gene enrichment. More importantly, synthetic hydrogel improved the reproducibility in cardioid properties compared to traditional suspension culture and Matrigel encapsulation. Therefore, PEG-based hydrogel has the potential to be used as an alternative to Matrigel for human cardioid culture in a variety of clinical applications including cell therapy and tissue engineering.
Collapse
Affiliation(s)
- Yuanhui Song
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, 13244, USA
| | - Michael Seitz
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, 13244, USA
| | - Andrew Kowalczewski
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, 13244, USA
| | - Nhu Y Mai
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, 13244, USA
| | - Era Jain
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, 13244, USA
| | - Huaxiao Yang
- Department of Biomedical Engineering, University of North Texas, Denton, TX, 76205, USA
| | - Zhen Ma
- Department of Biomedical & Chemical Engineering, Syracuse University, Syracuse, NY, 13244, USA
- BioInspired Institute for Material and Living Systems, Syracuse University, Syracuse, NY, 13244, USA
| |
Collapse
|
5
|
Wang Q, Yuan F, Zuo X, Li M. Breakthroughs and challenges of organoid models for assessing cancer immunotherapy: a cutting-edge tool for advancing personalised treatments. Cell Death Discov 2025; 11:222. [PMID: 40335487 PMCID: PMC12059183 DOI: 10.1038/s41420-025-02505-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 04/16/2025] [Accepted: 04/23/2025] [Indexed: 05/09/2025] Open
Abstract
Organoid models are powerful tools for evaluating cancer immunotherapy that provide a more accurate representation of the tumour microenvironment (TME) and immune responses than traditional models. This review focuses on the latest advancements in organoid technologies, including immune cell co-culture, 3D bioprinting, and microfluidic systems, which enhance the modelling of TME and facilitate the assessment of immune therapies such as immune checkpoint inhibitors (ICIs), CAR-T therapies, and oncolytic viruses. Although these models have great potential in personalised cancer treatment, challenges persist in immune cell diversity, long-term culture stability, and reproducibility. Future developments integrating artificial intelligence (AI), multi-omics, and high-throughput platforms are expected to improve the predictive power of organoid models and accelerate the clinical translation of immunotherapy.
Collapse
Affiliation(s)
- Qian Wang
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210009, Jiangsu, PR China
- The Fourth Clinical College of Nanjing Medical University, Nanjing, 210009, Jiangsu, PR China
| | - Fangwei Yuan
- Department of Thoracic Surgery, Lian Shui County People's Hospital, Huaian, 223400, Jiangsu, PR China
| | - Xianglin Zuo
- Biobank of Jiangsu Cancer Hospital (Jiangsu Institute of Cancer Research & The Affiliated Cancer Hospital of Nanjing Medical University), Nanjing, 210000, Jiangsu, PR China.
| | - Ming Li
- Department of Thoracic Surgery, The Affiliated Cancer Hospital of Nanjing Medical University & Jiangsu Cancer Hospital & Jiangsu Institute of Cancer Research, Jiangsu Key Laboratory of Molecular and Translational Cancer Research, Collaborative Innovation Center for Cancer Personalized Medicine, Nanjing, 210009, Jiangsu, PR China.
- The Fourth Clinical College of Nanjing Medical University, Nanjing, 210009, Jiangsu, PR China.
| |
Collapse
|
6
|
Ramírez-González GA, Consumi-Tubito C, Vargas-Méndez E, Centeno-Cerdas C. Advancing Organ-on-a-Chip Systems: The Role of Scaffold Materials and Coatings in Engineering Cell Microenvironment. Polymers (Basel) 2025; 17:1263. [PMID: 40363048 PMCID: PMC12074455 DOI: 10.3390/polym17091263] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/03/2025] [Accepted: 04/14/2025] [Indexed: 05/15/2025] Open
Abstract
For organ-on-a-chip (OoC) engineering, the use of biocompatible coatings and materials is not only recommended but essential. Extracellular matrix (ECM) components are commonly used as coatings due to their effects on cell orientation, protein expression, differentiation, and adhesion. Among the most frequently used coatings are collagen, fibronectin, and Matrigel, according to the specific cell type and intended OoC application. Additionally, materials such as polydimethylsiloxane (PDMS), thermoplastics, chitosan, and alginate serve as scaffolding components due to their biomechanical properties and biocompatibility. Here, we discuss some of the most employed coating techniques, including SAMs, dip coating, spin coating, microcontact printing, and 3D bioprinting, each offering advantages and drawbacks. Current challenges comprise enhancing biocompatibility, exploring novel materials, and improving scalability and reproducibility.
Collapse
Affiliation(s)
- Guido Andrés Ramírez-González
- Master’s Program in Medical Device Engineering, School of Materials Science and Engineering, Costa Rica Institute of Technology, Cartago 30109, Costa Rica;
- Biotechnology Research Center, Costa Rica Institute of Technology, Cartago 30109, Costa Rica;
| | - Chiara Consumi-Tubito
- Biotechnology Research Center, Costa Rica Institute of Technology, Cartago 30109, Costa Rica;
| | - Ernesto Vargas-Méndez
- Department of Biochemistry, School of Medicine, University of Costa Rica, San Jose 11501-2060, Costa Rica;
| | - Carolina Centeno-Cerdas
- Biotechnology Research Center, Costa Rica Institute of Technology, Cartago 30109, Costa Rica;
| |
Collapse
|
7
|
Hao LT, Lee S, Hwang DS, Jeon H, Park J, Kim HJ, Oh DX. Self-Healing Scaffolding Technology with Strong, Reversible Interactions under Physiological Conditions for Engineering Marbled Cultured Meat. ACS APPLIED MATERIALS & INTERFACES 2025. [PMID: 40317268 DOI: 10.1021/acsami.5c03479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
Cultured meat offers a sustainable alternative to animal farming, with the potential to reduce environmental impacts and improve food security. However, recapitulating natural meat marbling remains a significant challenge. This study presents a straightforward technology for achieving precise marbling patterns in large-scale cultured meat using self-healing hydrogels containing boronic acid-conjugated chitosan. Unlike conventional hydrogels, which require nonphysiological conditions for strong, reversible bonding, our system achieves robust reversible bonding at neutral pH through a unique mechanism: the nucleophilic groups of chitosan facilitate boronic acid-diol bond formation, exhibiting half the strength of a typical covalent bond, as demonstrated by nanomechanics analysis. The hydrogels form dual reversible networks of boronic acid-diol and hydrogen bonds, enabling self-healing and tunable stiffness. Biocompatibility studies confirm that they support the growth of mouse-derived cells and bovine-derived primary muscle cells. Each hydrogel variant optimizes mechanotransduction for the distinct requirements of fat or muscle cell culture and differentiation. This self-healing scaffolding technology enables the seamless assembly of muscle and fat monocultures into centimeter-thick meat with micrometer-scale marbling patterns, tailoring organoleptic properties and nutritional profiles without the need for meat glues or processing equipment.
Collapse
Affiliation(s)
- Lam Tan Hao
- Research Center for Bio-based Chemistry, Korea Research Institute of Chemical Technology (KRICT), Ulsan 44429, Republic of Korea
| | - Seunghyeon Lee
- Division of Environmental Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Dong Soo Hwang
- Division of Environmental Science and Engineering, Pohang University of Science and Technology (POSTECH), Pohang 37673, Republic of Korea
| | - Hyeonyeol Jeon
- Research Center for Bio-based Chemistry, Korea Research Institute of Chemical Technology (KRICT), Ulsan 44429, Republic of Korea
- Advanced Materials and Chemical Engineering, Korea National University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Jeyoung Park
- Department of Chemical and Biomolecular Engineering, Sogang University, Seoul 04107, Republic of Korea
| | - Hyo Jeong Kim
- Research Center for Bio-based Chemistry, Korea Research Institute of Chemical Technology (KRICT), Ulsan 44429, Republic of Korea
- Advanced Materials and Chemical Engineering, Korea National University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Dongyeop X Oh
- Department of Polymer Science and Engineering and Program in Environmental and Polymer Engineering, Inha University, Incheon 22212, Republic of Korea
| |
Collapse
|
8
|
Vetter J, Palagi I, Waisman A, Blaeser A. Recent advances in blood-brain barrier-on-a-chip models. Acta Biomater 2025; 197:1-28. [PMID: 40127880 DOI: 10.1016/j.actbio.2025.03.041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2024] [Revised: 03/19/2025] [Accepted: 03/21/2025] [Indexed: 03/26/2025]
Abstract
The blood-brain barrier is a physiological barrier between the vascular system and the nervous system. Under healthy conditions, it restricts the passage of most biomolecules into the brain, making drug development exceedingly challenging. Conventional cell-based in vitro models provide valuable insights into certain features of the BBB. Nevertheless, these models often lack the three-dimensional structure and dynamic interactions of the surrounding microenvironment, which greatly influence cell functionality. Consequently, considerable efforts have been made to enhance in vitro models for drug development and disease research. Recently, microfluidic organ-on-a-chip systems have emerged as promising candidates to better mimic the dynamic nature of the BBB. This review provides a comprehensive overview of recent BBB-on-chip devices. The typical building blocks, chip designs, the perfusion infrastructure, and readouts used to characterize and evaluate BBB formation are presented, analyzed, and discussed in detail. STATEMENT OF SIGNIFICANCE: The blood-brain barrier (BBB) is a highly selective barrier that controls what can enter the brain. While it protects the brain from harmful substances, it also hinders the delivery of treatments for neurological diseases such as Alzheimer's and Parkinson's. Due to its complexity, studying the BBB in living organisms remains difficult. However, recent advances in "organ-on-a-chip" technology have allowed scientists to create small, engineered models that replicate the BBB. These models provide a powerful platform to study diseases and test potential drugs with greater accuracy than traditional methods. Organ-on-a-chip devices are designed to mimic the behavior of organs or tissues in the human body, offering a more realistic and controlled environment for research. This review highlights recent breakthroughs in BBB-on-a-chip technology, showing how these models enhance current research and have the potential to transform the way we study brain diseases and develop new drugs. By integrating biology and engineering, BBB-on-a-chip technology has the potential to transform neuroscience research, improve drug development, and enhance our understanding of brain disorders.
Collapse
Affiliation(s)
- Johanna Vetter
- Institute for BioMedical Printing Technology, Technical University of Darmstadt, Darmstadt, Germany
| | - Ilaria Palagi
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Ari Waisman
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany; Research Center for Immunotherapy (FZI), University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Andreas Blaeser
- Institute for BioMedical Printing Technology, Technical University of Darmstadt, Darmstadt, Germany; Centre for Synthetic Biology, Technical University of Darmstadt, Darmstadt, Germany.
| |
Collapse
|
9
|
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
|
10
|
Busa D, Herudkova Z, Hyl J, Vlazny J, Sokol F, Matulova K, Folta A, Hynst J, Vojtova L, Kren L, Repko M, Racil Z, Mayer J, Culen M. Robust acute myeloid leukemia engraftment in humanized scaffolds using injectable biomaterials and intravenous xenotransplantation. Mol Oncol 2025; 19:1371-1385. [PMID: 39840700 PMCID: PMC12077274 DOI: 10.1002/1878-0261.13790] [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: 05/27/2024] [Revised: 10/11/2024] [Accepted: 12/11/2024] [Indexed: 01/23/2025] Open
Abstract
Patient-derived xenografts (PDXs) can be improved by implantation of a humanized niche. Nevertheless, the overall complexity of the current protocols, as well as the use of specific biomaterials and procedures, limits the wider adoption of this approach. Here, we identify the essential minimum steps required to create the humanized scaffolds and achieve successful acute myeloid leukemia (AML) engraftment. We compared seven biomaterials, which included both published and custom-designed materials. The highest level of bone marrow niche was achieved with extracellular matrix gels and custom collagen fiber, both of which allowed for a simple non-surgical implantation. The biomaterial selection did not influence the following AML infiltration. Regarding xenotransplantation, standard intravenous administration produced the most robust engraftment, even for two out of four otherwise non-engrafting AML samples. In contrast, direct intra-scaffold xenotransplantation did not offer any advantage. In summary, we demonstrate that the combination of an injectable biomaterial for scaffold creation plus an intravenous route for AML xenotransplantation provide the most convenient and robust approach to produce AML PDX using a humanized niche.
Collapse
Affiliation(s)
- Daniel Busa
- Department of Internal Medicine, Hematology and Oncology, Faculty of MedicineMasaryk UniversityBrnoCzech Republic
| | - Zdenka Herudkova
- Department of Internal Medicine, Hematology and Oncology, Faculty of MedicineMasaryk UniversityBrnoCzech Republic
| | - Jan Hyl
- Department of Internal Medicine, Hematology and Oncology, Faculty of MedicineMasaryk UniversityBrnoCzech Republic
| | - Jakub Vlazny
- Department of PathologyUniversity Hospital BrnoCzech Republic
- Department of Pathology, Faculty of MedicineMasaryk UniversityBrnoCzech Republic
| | - Filip Sokol
- Department of PathologyUniversity Hospital BrnoCzech Republic
| | - Kvetoslava Matulova
- Department of PathologyUniversity Hospital BrnoCzech Republic
- Department of Pathology, Faculty of MedicineMasaryk UniversityBrnoCzech Republic
| | - Adam Folta
- Department of Internal Medicine, Hematology and OncologyUniversity Hospital BrnoCzech Republic
| | - Jakub Hynst
- Central European Institute of TechnologyMasaryk UniversityBrnoCzech Republic
| | - Lucy Vojtova
- Central European Institute of TechnologyBrno Institute of TechnologyCzech Republic
| | - Leos Kren
- Department of PathologyUniversity Hospital BrnoCzech Republic
- Department of Pathology, Faculty of MedicineMasaryk UniversityBrnoCzech Republic
| | - Martin Repko
- Orthopedic ClinicUniversity Hospital BrnoCzech Republic
- Department of Orthopedic Surgery, Faculty of MedicineMasaryk UniversityBrnoCzech Republic
| | - Zdenek Racil
- Department of Physiology, Faculty of MedicineMasaryk UniversityBrnoCzech Republic
| | - Jiri Mayer
- Department of Internal Medicine, Hematology and Oncology, Faculty of MedicineMasaryk UniversityBrnoCzech Republic
- Department of Internal Medicine, Hematology and OncologyUniversity Hospital BrnoCzech Republic
- Central European Institute of TechnologyMasaryk UniversityBrnoCzech Republic
| | - Martin Culen
- Department of Internal Medicine, Hematology and Oncology, Faculty of MedicineMasaryk UniversityBrnoCzech Republic
- Department of Internal Medicine, Hematology and OncologyUniversity Hospital BrnoCzech Republic
| |
Collapse
|
11
|
Ozdil B, Avci CB, Calik-Kocaturk D, Gorgulu V, Uysal A, Güler G, Karabay Yavaşoğlu NÜ, Aktug H. Modulating Cancer Stem Cell Characteristics in CD133+ Melanoma Cells through Hif1α, KLF4, and SHH Silencing. ACS OMEGA 2025; 10:16804-16814. [PMID: 40321496 PMCID: PMC12044452 DOI: 10.1021/acsomega.5c00799] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 03/19/2025] [Accepted: 04/04/2025] [Indexed: 05/08/2025]
Abstract
Malignant melanoma is a highly aggressive form of skin cancer, partly driven by a subset of cancer stem cells (CSCs) with remarkable capacities for self-renewal, differentiation, and resistance to therapy. In this study, we examined how silencing three key genes-Hif1α, KLF4, and SHH-affects CSC characteristics. Using small interfering RNA (siRNA)-based approaches, we observed significant changes at both the gene and protein levels, shedding light on how these pathways influence melanoma progression. Our results demonstrated that silencing these genes reduces the stem-like features of CSCs. Notably, Hif1α silencing triggered a marked decrease in hypoxia-related gene expression, while targeting SHH led to a reduction in Gli1, a downstream effector of SHH signaling, highlighting its potential as a therapeutic target. We also observed changes in epigenetic markers such as HDAC9 and EP300, which play crucial roles in maintaining stemness and regulating gene expression. Interestingly, these interventions appeared to reprogram CSCs, pushing them toward a phenotype distinct from both traditional CSCs and non-stem cancer cells (NCSCs). Our findings emphasize the importance of targeting key signaling pathways in melanoma CSCs and underscore the value of mimicking the tumor microenvironment in experimental models. By revealing the dynamic plasticity of melanoma CSCs, this study offers fresh insights into potential therapeutic strategies, particularly using siRNA to modulate pathways associated with tumor progression and stem cell behavior.
Collapse
Affiliation(s)
- Berrin Ozdil
- Department
of Histology and Embryology, Faculty of Medicine, Suleyman Demirel University, Isparta 32260, Turkey
- Department
of Histology and Embryology, Faculty of Medicine, Ege University, Izmir 35100, Turkey
- Department
of Physics, Biophysics Laboratory, Izmir
Institute of Technology, Izmir 35430, Turkey
| | - Cigir Biray Avci
- Department
of Medical Biology, Faculty of Medicine, Ege University, Izmir 35100, Turkey
| | | | - Volkan Gorgulu
- Department
of Histology and Embryology, Faculty of Medicine, Ege University, Izmir 35100, Turkey
| | - Aysegul Uysal
- Department
of Histology and Embryology, Faculty of Medicine, Ege University, Izmir 35100, Turkey
| | - Günnur Güler
- Department
of Physics, Biophysics Laboratory, Izmir
Institute of Technology, Izmir 35430, Turkey
| | | | - Huseyin Aktug
- Department
of Histology and Embryology, Faculty of Medicine, Ege University, Izmir 35100, Turkey
| |
Collapse
|
12
|
Liu J, Liu Q, Guo M, Jiang C, Chen J, Wang T, Sung TC, Chou SJ, Chiou SH, Fan G, Higuchi A. Differentiation of human induced pluripotent stem cells into retinal pigment epithelium cells during culture on peptide-grafted hydrogels. Regen Biomater 2025; 12:rbaf035. [PMID: 40416648 PMCID: PMC12098265 DOI: 10.1093/rb/rbaf035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 04/19/2025] [Accepted: 04/23/2025] [Indexed: 05/27/2025] Open
Abstract
A variety of novel peptide-grafted hydrogels, of which peptides were derived from vitronectin (PQVTRGDVFTMP) or the laminin β4 chain (PMQKMRGDVFSP), were prepared in this study. The peptide-grafted hydrogels promoted the adhesion, proliferation and colony formation of hiPSCs and maintained their pluripotency up to passage 5 under xeno-free conditions. We successfully generated RPE cells from hiPSCs using one of the most suitable xeno-free peptide-grafted hydrogels, KVN2CK (KGCGGKGG-PQVTRGDVFTMP), which was derived from vitronectin, and confirmed the effect of these hiPSC-derived RPE cells in a rat retinal degeneration model (Royal College of Surgeons (RCS) rats) via subretinal transplantation, when we investigated functional improvements in vision in RCS rats after the transplantation of hiPSC-derived RPE cells. Our novel peptide-grafted hydrogels provided a safe and robust platform for generating single-layer hiPSC-derived RPE cells under xeno-free conditions, which indicates the potential of these hydrogels for stem cell therapy for retinal degenerative diseases in the future.
Collapse
Affiliation(s)
- Jun Liu
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Qian Liu
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Minmei Guo
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Chengyu Jiang
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Jianyang Chen
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Ting Wang
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Tzu-Cheng Sung
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Shih-Jie Chou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan, China
- Institute of Pharmacology, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, China
| | - Shih-Hwa Chiou
- Department of Medical Research, Taipei Veterans General Hospital, Taipei 112201, Taiwan, China
- Institute of Pharmacology, School of Medicine, National Yang Ming Chiao Tung University, Taipei 112304, Taiwan, China
- Department of Ophthalmology, Taipei Veterans General Hospital, Taipei 11217, Taiwan, China
| | - Guoping Fan
- Department of Human Genetics, David Geffen School of Medicine, UCLA, Los Angeles, CA 90095, USA
| | - Akon Higuchi
- State Key Laboratory of Eye Health, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
- Department of Chemical and Materials Engineering, National Central University, Taoyuan 32001, Taiwan, China
- R&D Center for Membrane Technology, Chung Yuan Christian University, Taoyuan 320, Taiwan, China
| |
Collapse
|
13
|
Zhao N, Pessell AF, Chung TD, Searson PC. Brain vascular basement membrane: comparison of human and mouse brain at the transcriptomic and proteomic levels. Matrix Biol 2025:S0945-053X(25)00036-8. [PMID: 40294830 DOI: 10.1016/j.matbio.2025.04.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Revised: 04/23/2025] [Accepted: 04/24/2025] [Indexed: 04/30/2025]
Abstract
The cerebrovascular basement membrane (BM) is a key component of the blood-brain barrier (BBB). The BM provides structural support for brain microvascular endothelial cells and the supporting cells of the neurovascular unit, and facilitates cell signaling through adhesion receptors, regulates the concentration of soluble factors, and serves as an additional barrier for transport. However, our understanding of the composition of BM remains incomplete. Here we analyze recent proteomic and genomic data to assess the composition of BM in human and mouse brain, and in tissue-engineered BBB models. All data sets confirm that the main components of brain BM are collagen IV a1/2, laminin, along with agrin, perlecan, and nidogen. Transcriptomic data from human BMECs suggests that the main laminin isoform is Laminin 321, while transcriptomic data from mice and proteomic data from mice and humans suggest that Laminin 521 is the predominant isoform. Transcriptomic data from iBMECs suggest that Laminin 511 is the predominant isoform. The supporting molecules agrin, perlecan, and nidogen were detected at significant levels in all studies, although only nidogen 1 was detected in the human transcriptomic data sets. No significant differences in human BM composition were observed in BMECs along the arterio-venous axis, or in comparison of healthy and AD brains.
Collapse
Affiliation(s)
- Nan Zhao
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Alexander F Pessell
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Tracy D Chung
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA
| | - Peter C Searson
- Institute for Nanobiotechnology, Johns Hopkins University, Baltimore, MD, 21218, USA; Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA; Department of Materials Science and Engineering, Johns Hopkins University, Baltimore, MD, 21218, USA.
| |
Collapse
|
14
|
Wang J, Tao X, Zhu J, Dai Z, Du Y, Xie Y, Chu X, Fu G, Lei Z. Tumor organoid-immune co-culture models: exploring a new perspective of tumor immunity. Cell Death Discov 2025; 11:195. [PMID: 40268893 PMCID: PMC12019369 DOI: 10.1038/s41420-025-02407-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Revised: 02/20/2025] [Accepted: 03/18/2025] [Indexed: 04/25/2025] Open
Abstract
Recent advancements in technology have significantly expanded the scope of tumor research, progressing from the study of individual cells to more intricate tissue and organ-level analyses. Tumor organoids have emerged as a highly realistic platform for investigating tumor growth, development, and their interactions with the surrounding microenvironment. However, a notable limitation of these organoids is their lack of the diverse cellular composition typically observed in actual tumors, which hinders their ability to fully replicate the complexity of the tumor microenvironment. Immune cells play a pivotal role, and tumor immunology has become a major research hotspot. Research in tumor immunology aims to elucidate how the immune system recognizes and attacks tumor cells, as well as how tumor cells evade immune surveillance. In recent years, there has been growing interest in co-culturing immune cells with tumor organoids, an approach that has yielded valuable insights into the intricate interactions between tumors and the immune system. The aim of this paper is to review and discuss the progress achieved in co-culturing tumor organoids with immune cells. By doing so, we hope to offer a new perspective and enhance our understanding of the complexity and diversity inherent in the tumor microenvironment.
Collapse
Affiliation(s)
- Jing Wang
- Department of Oncology, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, China
| | - Xiaoyue Tao
- Department of Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jialong Zhu
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Zhe Dai
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yuanyang Du
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China
| | - Yiyang Xie
- Department of Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China
| | - Xiaoyuan Chu
- Department of Oncology, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, China.
- Department of Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China.
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- Department of Oncology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, China.
| | - Gongbo Fu
- Department of Oncology, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, China.
- Department of Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China.
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- Department of Oncology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, China.
| | - Zengjie Lei
- Department of Oncology, Jinling Clinical Medical College, Nanjing Medical University, Nanjing, China.
- Department of Oncology, Jinling Clinical Medical College, Nanjing University of Chinese Medicine, Nanjing, China.
- Department of Oncology, Nanjing Jinling Hospital, Affiliated Hospital of Medical School, Nanjing University, Nanjing, China.
- Department of Oncology, Jinling Hospital, The First School of Clinical Medicine, Southern Medical University, Nanjing, China.
| |
Collapse
|
15
|
Amel A, Brown R, Rabeling A, Goolam M. Matrigel inhibits elongation and drives endoderm differentiation in aggregates of mouse embryonic stem cells. FEBS Open Bio 2025. [PMID: 40251891 DOI: 10.1002/2211-5463.70044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 04/05/2025] [Accepted: 04/11/2025] [Indexed: 04/21/2025] Open
Abstract
Modelling peri-implantation mammalian development using the self-organising properties of stem cells is a rapidly growing field that has advanced our understanding of cell fate decisions occurring in the early embryo. Matrigel, a basement membrane matrix, is a critical substrate used in various protocols for its efficacy in promoting stem cell growth and self-organisation. However, its role in driving stem cell lineage commitment, and whether this effect is driven by biochemical or physical cues, is not currently clear. Here, we grow embryoid bodies in suspension, Matrigel and agarose, an inert polysaccharide, to attempt to decouple the physical and biochemical roles of Matrigel and better understand how it drives stem cell differentiation. We use a combination of light microscopy, quantitative PCR and immunostaining to investigate gene and protein changes in our different culture conditions. We show that stem cell aggregates in Matrigel are hindered in their ability to elongate compared with those grown in agarose or in suspension, indicating that prohibitive role in self-organisation. Aggregates in Matrigel are also driven to differentiate into endoderm, with ectoderm differentiation inhibited. Furthermore, these effects are not due to the physical presence of Matrigel, as the same effects are not witnessed in aggregates grown in agarose. Our results thus indicate that Matrigel has a significant and complex effect on the differentiation and morphology of embryoid bodies.
Collapse
Affiliation(s)
- Atoosa Amel
- Department of Human Biology, University of Cape Town, South Africa
- UCT Neuroscience Institute, Cape Town, South Africa
| | - Rachel Brown
- Department of Human Biology, University of Cape Town, South Africa
- UCT Neuroscience Institute, Cape Town, South Africa
| | - Alexa Rabeling
- Department of Human Biology, University of Cape Town, South Africa
- UCT Neuroscience Institute, Cape Town, South Africa
| | - Mubeen Goolam
- Department of Human Biology, University of Cape Town, South Africa
- UCT Neuroscience Institute, Cape Town, South Africa
| |
Collapse
|
16
|
Kary AD, Noelle H, Magin CM. Tissue-Informed Biomaterial Innovations Advance Pulmonary Regenerative Engineering. ACS Macro Lett 2025; 14:434-447. [PMID: 40102038 DOI: 10.1021/acsmacrolett.5c00075] [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: 03/20/2025]
Abstract
Irreversible progressive pulmonary diseases drastically reduce the patient quality of life, while transplantation remains the only definitive cure. Research into lung regeneration pathways holds significant potential to expand and promote the discovery of new treatment options. Polymeric biomaterials designed to replicate key tissue characteristics (i.e., biochemical composition and mechanical cues) show promise for creating environments in which to study chronic lung diseases and initiate lung tissue regeneration. In this Viewpoint, we explore how naturally derived materials can be employed alone or combined with engineered polymer systems to create advanced tissue culture platforms. Pulmonary tissue models have historically leveraged natural materials, including basement membrane extracts and a decellularized extracellular matrix, as platforms for lung regeneration studies. Here, we provide an overview of the progression of pulmonary regenerative engineering, exploring how innovations in the growing field of tissue-informed biomaterials have the potential to advance lung regeneration research by bridging the gap between biological relevance and mechanical precision.
Collapse
Affiliation(s)
- Anton D Kary
- Department of Bioengineering, University of Colorado, Denver | Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Haley Noelle
- Department of Bioengineering, University of Colorado, Denver | Anschutz Medical Campus, Aurora, Colorado 80045, United States
| | - Chelsea M Magin
- Department of Bioengineering, University of Colorado, Denver | Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Department of Pediatrics, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045, United States
- Division of Pulmonary Sciences and Critical Care Medicine, Department of Medicine, University of Colorado, Anschutz Medical Campus, Aurora, Colorado 80045, United States
| |
Collapse
|
17
|
Xu S, Yin SY, Bie ZX, Li YM, Qi J, Ma YD, Wang Z, Xi JJ, Li XG. Personalized drug screening of patient-derived tumor-like cell clusters based on specimens obtained from percutaneous transthoracic needle biopsy in patients with lung malignancy: a real-world study. BMC Cancer 2025; 25:649. [PMID: 40205326 PMCID: PMC11983848 DOI: 10.1186/s12885-025-14069-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025] Open
Abstract
BACKGROUND Patient-derived xenografts and organoids were the most common patient-derived tumor models in vitro that were utilized in personalized drug screening, and the establishment rate and duration required to be improved. Patient-derived tumor-like cell clusters (PTCs) could be established within ten days for drug screening, with high establishment rate and accuracy in predicting clinical outcomes. This study aims to explore the accuracy of PTCs based on specimens obtained from percutaneous transthoracic needle biopsy (PTNB) in lung malignancy (LM) patients, and to investigate the predictors for the success of PTC culture. MATERIALS AND METHODS This retrospective cohort study included LM patients who underwent image-guided PTNB, and the specimens were used for PTC culture, which was followed by personalized drug screening of chemotherapy and molecular targeted therapy, and the accuracy was validated by previous or further treatments. The predictors of the success of PTC culture were identified by univariable and multivariable analyses. RESULTS A total of 68 LM patients were enrolled, consisting of 57, 7, and 4 patients with non-small cell lung cancer, small cell lung cancer, and lung metastases, respectively. Pneumothorax was the predominant adverse event for PTNB, with an incidence rate of 20.6% (14/68). PTC models based on PTNB specimens were established successfully for 56 patients in 3.8 ± 2.3 days, with an 82.4% success rate. Five patients had not received treatments before or after PTC culture. PTC drug screening reveals 88.2% (45/51) overall consistency in predicting clinical outcomes. Necrotic area over half of the tumor (hazard ratio, 0.121; 95% confidence interval, 0.025-0.598; P = 0.010) was identified as the negative predictor for the success of PTC culture. CONCLUSIONS PTC culture based on PTNB specimens could be established in 82.4% of LM patients, with a high accuracy in predicting clinical outcomes. Excessive necrosis in the tumor may predict the failure of PTC culture. Image-guided PTNB targeting enhanced or fluorodeoxyglucose avid regions on images might contribute to improving the success rate of PTC culture.
Collapse
Affiliation(s)
- Sheng Xu
- Department of Minimally Invasive Tumor Therapies Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Shen-Yi Yin
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, Beijing, 100871, China
| | - Zhi-Xin Bie
- Department of Minimally Invasive Tumor Therapies Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yuan-Ming Li
- Department of Minimally Invasive Tumor Therapies Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Jing Qi
- Department of Neurology, Beijing Chao-Yang Hospital, Capital Medical University, Beijing, 100020, China
| | - Yi-Dan Ma
- Department of Pathology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Zheng Wang
- Department of Pathology, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Jianzhong Jeff Xi
- State Key Laboratory of Natural and Biomimetic Drugs, Department of Biomedical Engineering, College of Future Technology, Peking University, No.5 Yiheyuan Road, Haidian District, Beijing, 100871, China.
| | - Xiao-Guang Li
- Department of Minimally Invasive Tumor Therapies Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, 100730, China.
- Department of Minimally Invasive Tumor Therapies Center, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, No.1 Da Hua Road, Dong Dan, Beijing, 100730, China.
- Graduate School of Peking Union Medical College, Chinese Academy of Medical Sciences, 9 Dongdansantiao Street, Dongcheng District, Beijing, 100730, China.
| |
Collapse
|
18
|
Amitrano MJ, Cho M, Coughlin EM, Palecek SP, Murphy WL. Synthetic hydrogels support robust and reproducible cardiomyocyte differentiation. Biomater Sci 2025; 13:2142-2151. [PMID: 40091790 DOI: 10.1039/d4bm01636j] [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: 03/19/2025]
Abstract
Cardiomyocyte manufacturing from human pluripotent stem cells is limited by the variability of differentiation efficiencies, partly attributed to the widespread use of the tumor-derived substrate Matrigel. Here, we describe a screening approach to identify fully-defined synthetic PEG hydrogels that support iPSC-derived cardiac progenitor cell (iPSC-CPC) adhesion, survival, and differentiation into iPSC-derived cardiomyocytes (iPSC-CMs). Our PEG hydrogels supported superior iPSC-CM differentiation efficiency, with a 24% increase in cTnT expression, and greater reproducibility when compared to cells cultured on Matrigel. By combining our 5-level, 3-variable full factorial screening approach with multi-variate analysis, we showed that all substrate variables manipulated here (adhesion ligand type/concentration, stiffness) had a significant influence on iPSC-CPC confluency and that iPSC-CM differentiation was significantly influenced by adhesion ligands. These results highlight the benefit of synthetic, tunable cell culture substrates and multi-variate screening studies to identify substrate formulations for a targeted cell behavior.
Collapse
Affiliation(s)
- Margot J Amitrano
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1111 Highland Avenue Room 5405, 53705, Madison, WI, USA.
| | - Mina Cho
- Department of Materials Science and Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - Eva M Coughlin
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1111 Highland Avenue Room 5405, 53705, Madison, WI, USA.
| | - Sean P Palecek
- Department of Chemical and Biological Engineering, University of Wisconsin-Madison, Madison, WI, USA
| | - William L Murphy
- Department of Biomedical Engineering, University of Wisconsin-Madison, 1111 Highland Avenue Room 5405, 53705, Madison, WI, USA.
- Department of Orthopedics and Rehabilitation, University of Wisconsin-Madison, Madison, WI, USA
| |
Collapse
|
19
|
Ma Z, Liu Y, Chen R, Fan H, Kong L, Cao X. A novel perspective on bone tumors: advances in organoid research. Front Pharmacol 2025; 16:1550163. [PMID: 40271075 PMCID: PMC12015983 DOI: 10.3389/fphar.2025.1550163] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2024] [Accepted: 03/27/2025] [Indexed: 04/25/2025] Open
Abstract
Bone tumor organoids are three-dimensional cell culture models derived from patient tissues or cells, capable of highly replicating the growth patterns and cell interactions of bone tumors in vitro. Current treatments for bone tumors are hindered by challenges such as drug resistance, recurrence, and metastasis. Organoids enhance the physiological relevance of bone tumor models, thereby improving treatment precision and overcoming the limitations of current therapeutic approaches. Organoid technology has made preliminary applications in bone tumor research, including primary bone tumors, metastatic bone tumors, and bone marrow-derived bone tumors. This review will explore the establishment of bone tumor organoids, summarize their applications and prospects in various bone tumor diseases, and discuss their integration with emerging technologies. Additionally, the limitations and future directions of bone tumor organoid research will be discussed. In the future, bone tumor organoids are expected to promote the further development of precision medicine.
Collapse
Affiliation(s)
- Zebing Ma
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Yibing Liu
- Hunan University of Chinese Medicine, Changsha, Hunan, China
| | - Rui Chen
- Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, Henan, China
| | - Huayu Fan
- Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, Henan, China
| | - Liang Kong
- Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, Henan, China
| | - Xiangyang Cao
- Hunan University of Chinese Medicine, Changsha, Hunan, China
- Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, Henan, China
- Institute of Intelligent Medical and Bioengineering Henan Academy of Traditional Chinese Medicine Sciences, Zhengzhou, Henan, China
- Henan Province Artificial Intelligence Engineering Research Center for Bone Injury Rehabilitation, Luoyang Orthopedic Hospital of Henan Province (Orthopedic Hospital of Henan Province), Zhengzhou, Henan, China
- Henan University of Chinese Medicine, Zhengzhou, Henan, China
| |
Collapse
|
20
|
Courbot O, Elosegui-Artola A. The role of extracellular matrix viscoelasticity in development and disease. NPJ BIOLOGICAL PHYSICS AND MECHANICS 2025; 2:10. [PMID: 40191103 PMCID: PMC11968406 DOI: 10.1038/s44341-025-00014-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 02/14/2025] [Indexed: 04/09/2025]
Abstract
For several decades, research has studied the influence of the extracellular matrix (ECM) mechanical properties in cell response, primarily emphasising its elasticity as the main determinant of cell and tissue behaviour. However, the ECM is not purely elastic; it is viscoelastic. ECM viscoelasticity has now emerged as a major regulator of collective cell dynamics. This review highlights recent findings on the role of ECM viscoelasticity in development and pathology.
Collapse
Affiliation(s)
- Olivia Courbot
- Cell and Tissue Mechanobiology Laboratory, The Francis Crick Institute, London, UK
- Department of Physics, King’s College London, London, UK
| | - Alberto Elosegui-Artola
- Cell and Tissue Mechanobiology Laboratory, The Francis Crick Institute, London, UK
- Department of Physics, King’s College London, London, UK
| |
Collapse
|
21
|
Zhang W, Ding Y, He H, Chen K, Zeng Q, Cao X, Xiang Y, Zeng H. Prospects and challenges of ovarian cancer organoids in chemotherapy research (Review). Oncol Lett 2025; 29:198. [PMID: 40052067 PMCID: PMC11883337 DOI: 10.3892/ol.2025.14944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2024] [Accepted: 01/20/2025] [Indexed: 03/09/2025] Open
Abstract
Ovarian cancer (OC), a prevalent and severe malignancy of the female reproductive system, often presents with mild early symptoms and is therefore diagnosed at advanced stages, leading to a poor prognosis. Current chemotherapeutic treatment relies on platinum-based combinational therapy and there have been no recent breakthroughs in the development of new drugs. Advances in organoid technology offer a novel approach to study OC by simulating tumors and their microenvironment, enhancing drug screening effectiveness and accuracy, and providing a foundation for personalized therapy. In recent years, researchers have made notable advancements, successfully developing a diverse array of OC organoid models, with biobanks serving a pivotal role in enhancing their success rates and overall efficiency. The present review summarizes the advantages of organoids over other models, such as two-dimensional cell models, three-dimensional spheres and patient-derived xenograft models, as well as the application of organoids. In particular, the current review emphasizes the application of organoids in chemotherapeutic drug screening, testing and personalized treatment. The limitations and prospects of organoid technology are also discussed. The present study aimed to reveal the unique advantages of OC organoids in chemotherapeutic applications, so as to provide insights into screening and testing new drugs for OC.
Collapse
Affiliation(s)
- Weijia Zhang
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Yuqing Ding
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Hui He
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Keming Chen
- Department of Gynecology and Obstetrics, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Qingsong Zeng
- Department of Gynecology and Obstetrics, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Xiaoming Cao
- Department of Gynecology and Obstetrics, First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Ying Xiang
- Department of Cell Biology and Medical Genetics, School of Basic Medicine, Health Science Center, Yangtze University, Jingzhou, Hubei 434023, P.R. China
| | - Hai Zeng
- Department of Oncology, The First Affiliated Hospital of Yangtze University, Jingzhou, Hubei 434023, P.R. China
| |
Collapse
|
22
|
Breideband L, Wächtershäuser KN, Sarkar R, Puspathasan M, Stelzer EH, Pampaloni F. Gravitational forces and matrix stiffness modulate the invasiveness of breast cancer cells in bioprinted spheroids. Mater Today Bio 2025; 31:101640. [PMID: 40124331 PMCID: PMC11930500 DOI: 10.1016/j.mtbio.2025.101640] [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: 10/13/2024] [Revised: 01/29/2025] [Accepted: 03/03/2025] [Indexed: 03/25/2025] Open
Abstract
The progression of breast cancer is influenced by the stiffness of the extracellular matrix (ECM), which becomes stiffer as cancer advances due to increased collagen IV and laminin secretion by cancer-associated fibroblasts. Intriguingly, breast cancer cells cultivated in two-dimensions exhibit a less aggressive behavior when exposed to weightlessness, or microgravity conditions. This study aims to elucidate the interplay between matrix stiffness and microgravity on breast cancer progression. For this purpose, three-dimensional spheroids of breast cancer cell lines (MCF-7 and MDA-MB-231) were formed. These spheroids were subsequently bioprinted in hydrogels of varying stiffness, obtained by the mixing of gelatin methacrylate and poly(ethylene) glycol diacrylate mixed at different ratios. The constructs were printed with a custom stereolithography (SLA) bioprinter converted from a low-cost, commercially available 3D printer. These bioprinted structures, encapsulating breast cancer spheroids, were then placed in a clinostat (microgravity simulation device) for a duration of seven days. Comparative analyses were conducted between objects cultured under microgravity and standard earth gravity conditions. Protein expression was characterized through fluorescent microscopy, while gene expression of MCF-7 constructs was analyzed via RNA sequencing. Remarkably, the influence of a stiffer ECM on the protein and gene expression levels of breast cancer cells could be modulated and sometimes even reversed in microgravity conditions. The study's findings hold implications for refining therapeutic strategies for advanced breast cancer stages - an array of genes involved in reversing aggressive or even metastatic behavior might lead to the discovery of new compounds that could be used in a clinical setting.
Collapse
Affiliation(s)
- Louise Breideband
- Biological Sciences (IZN), Buchman Institute for Molecular Life Sciences (BMLS), Goethe-Universität Frankfurt am Main, DE-Frankfurt am Main, Germany
| | - Kaja Nicole Wächtershäuser
- Biological Sciences (IZN), Buchman Institute for Molecular Life Sciences (BMLS), Goethe-Universität Frankfurt am Main, DE-Frankfurt am Main, Germany
| | - Ryan Sarkar
- Biological Sciences (IZN), Buchman Institute for Molecular Life Sciences (BMLS), Goethe-Universität Frankfurt am Main, DE-Frankfurt am Main, Germany
| | - Melosha Puspathasan
- Biological Sciences (IZN), Buchman Institute for Molecular Life Sciences (BMLS), Goethe-Universität Frankfurt am Main, DE-Frankfurt am Main, Germany
| | - Ernst H.K. Stelzer
- Biological Sciences (IZN), Buchman Institute for Molecular Life Sciences (BMLS), Goethe-Universität Frankfurt am Main, DE-Frankfurt am Main, Germany
| | - Francesco Pampaloni
- Biological Sciences (IZN), Buchman Institute for Molecular Life Sciences (BMLS), Goethe-Universität Frankfurt am Main, DE-Frankfurt am Main, Germany
| |
Collapse
|
23
|
Soulat A, Mohsenpour T, Roshangar L, Moaddab SY, Soulat F. Innovative Therapeutic Approach Targeting Colon Cancer Stem Cells: Transitional Cold Atmospheric Plasma. ACS OMEGA 2025; 10:12109-12121. [DOI: https:/doi.org/10.1021/acsomega.4c10378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/12/2025]
Affiliation(s)
- Abolfazl Soulat
- Department of Atomic and Molecular Physics, Faculty of Sciences
- University of Mazandaran
| | - Taghi Mohsenpour
- Department of Atomic and Molecular Physics, Faculty of Sciences
- University of Mazandaran
| | - Leila Roshangar
- Department of Histology, Faculty of Medicine
- Tabriz University of Medical Sciences
| | | | - Fatemeh Soulat
- Applied Chemistry laboratory, Department of Chemistry, Faculty of Basic Science
- Azarbaijan Shahid Madani University (ASMU)
| |
Collapse
|
24
|
Guan Y, Jia Z, Xiong X, He R, Ouyang Y, Liu H, Liang L, Meng X, Zhang R, Guan C, Wang S, Li D, Cui Y, Bai J, Zhao J, Meng H, Peng J, Wang Y. Tissue-specific extracellular matrix for the larger-scaled expansion of spinal cord organoids. Mater Today Bio 2025; 31:101561. [PMID: 40083838 PMCID: PMC11904521 DOI: 10.1016/j.mtbio.2025.101561] [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: 10/31/2024] [Revised: 02/02/2025] [Accepted: 02/07/2025] [Indexed: 03/16/2025] Open
Abstract
Spinal cord organoids (SCOs) are in vitro models that faithfully recapitulate the basic tissue architecture and cell types of the spinal cord and play a crucial role in developmental studies, disease modeling, and drug screening. Physiological cues are required for proliferation and differentiation during SCO culture. However, commonly used basement membrane matrix products, such as Matrigel®, lack tissue-specific biophysical signals. The current study utilizes decellularization process to fabricate tissue-derived hydrogel from porcine spinal cord tissue that retain intrinsic matrix components. This gel system supported an expanded neuroepithelial scale and enhanced ventral recognition patterns during SCO cultivation. Based on the characteristics of the enlarged aggregate size, a technical system for SCO cutting and subculture are proposed to improve the economic feasibility. Finally, the advantage of S-gel in maintaining neurite outgrowth are also found, which suggests its potential application in neural-related microphysiological systems.
Collapse
Affiliation(s)
- Yanjun Guan
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Co-innovation Center of Neuroregeneration, Nantong University Nantong, Jiangsu Province 226007, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Zhibo Jia
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Xing Xiong
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Ruichao He
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- School of Medicine, Nankai University, Tianjin 300071, PR China
| | - Yiben Ouyang
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- School of Medicine, Nankai University, Tianjin 300071, PR China
| | - Haolin Liu
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Lijing Liang
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Xiaoran Meng
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Ranran Zhang
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Congcong Guan
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Sice Wang
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Dongdong Li
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Yuhui Cui
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Graduate School of Chinese PLA General Hospital, No. 28 Fuxing Road, Beijing, 100853, PR China
| | - Jun Bai
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Jinjuan Zhao
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Haoye Meng
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
| | - Jiang Peng
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Co-innovation Center of Neuroregeneration, Nantong University Nantong, Jiangsu Province 226007, PR China
| | - Yu Wang
- Institute of Orthopedics, The Fourth Medical Center of Chinese PLA General Hospital, Beijing Key Lab of Regenerative Medicine in Orthopedics, Key Laboratory of Musculoskeletal Trauma & War Injuries PLA, No. 51 Fucheng Road, Beijing, 100048, PR China
- Co-innovation Center of Neuroregeneration, Nantong University Nantong, Jiangsu Province 226007, PR China
| |
Collapse
|
25
|
Soulat A, Mohsenpour T, Roshangar L, Moaddab SY, Soulat F. Innovative Therapeutic Approach Targeting Colon Cancer Stem Cells: Transitional Cold Atmospheric Plasma. ACS OMEGA 2025; 10:12109-12121. [PMID: 40191350 PMCID: PMC11966581 DOI: 10.1021/acsomega.4c10378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 02/22/2025] [Accepted: 03/04/2025] [Indexed: 04/09/2025]
Abstract
Transitional cold atmospheric plasma (TCAP) represents a novel technique for generating plasma remotely from a primary source. It consists of a partially nonthermal ionized gas mixture containing charged and neutral particles, photons, and free radicals. In recent years, TCAP has attracted considerable attention in biomedical applications. In order to evaluate colon cancer stem cells' (CCSCs) proliferation, apoptotic induction, inflammatory response, and survival, TCAP was utilized both directly and indirectly in this study. Using argon and helium gases, TCAP was continuously delivered in two stages during the experiment. For direct state, TCAP was irradiated onto CCSCs for 3 and 5 min. In the indirect technique, Matrigel was treated with TCAP for 5 min before the introduction of cells. In vitro assays demonstrated that TCAP exposure significantly reduced the viability of CCSCs; helium gas and direct application had greater impacts than argon. Numerous investigations confirmed the induction of apoptosis, showing that the treated groups had more apoptotic cells and altered cellular structures than controls (****p < 0.0001). A substantial increase in the Bax/Bcl-2 ratio was found by analyzing the expression of the Bax and Bcl-2 genes, indicating increased susceptibility to apoptosis (*p = 0.0177 and ***p = 0.0004). The higher efficacy of the direct helium mode was further highlighted by inflammatory marker analysis, which showed a significant reduction in interleukin-6 and interleukin-8 expression in cells directly treated with TCAP-helium compared to TCAP-argon (**p = 0.0015 and ***p = 0.0007). Lastly, the proliferation test, which relies on K i-67 expression, demonstrated a noteworthy decline in all TCAP-treated groups, with the direct helium group exhibiting the most robust impact (**p = 0.0014). Overall, the findings highlight the potential of TCAP, particularly with helium, as a promising approach for selectively targeting CCSCs and providing insights into its therapeutic mechanisms for cancer treatment. TCAP, therefore, emerges as a unique therapeutic strategy with potential applications in cancer stem cell-targeted therapies.
Collapse
Affiliation(s)
- Abolfazl Soulat
- Department of Atomic and Molecular Physics, Faculty of Sciences, University of Mazandaran, 4741613534 Babolsar, Iran
| | - Taghi Mohsenpour
- Department of Atomic and Molecular Physics, Faculty of Sciences, University of Mazandaran, 4741613534 Babolsar, Iran
| | - Leila Roshangar
- Department of Histology, Faculty of Medicine, Tabriz University of Medical Sciences, 5166614766 Tabriz, Iran
| | - Seyyed Yaghoub Moaddab
- Liver and Gastrointestinal Disease Research Center, Tabriz University of Medical Sciences, 5166614766 Tabriz, Iran
| | - Fatemeh Soulat
- Applied Chemistry laboratory, Department of Chemistry, Faculty of Basic Science, Azarbaijan Shahid Madani University (ASMU), 5375171379 Tabriz, Iran
| |
Collapse
|
26
|
Okuda K, Takagi A, Shimizu R, Nishi K, Hayano N, Takashima I, Konishi M. Total Synthesis of Antiausterity Agent Callistrilone O Reveals Promising Antitumor Activity in a Melanoma Homograft Mouse Model. ChemMedChem 2025; 20:e202400818. [PMID: 39812162 DOI: 10.1002/cmdc.202400818] [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: 10/17/2024] [Revised: 12/16/2024] [Accepted: 01/13/2025] [Indexed: 01/16/2025]
Abstract
The antiausterity strategy in anticancer drug discovery has attracted much attention as a way to exterminate cancer cells under nutrient deprived conditions which are commonly found in solid tumors. These tumors under low nutrient stress are known to be malignant and often resist conventional drug therapy. As a potential drug candidate, we focused on the meroterpenoid natural product callistrilone O which has demonstrated extremely potent antiausterity properties toward PANC-1 pancreatic carcinoma in vitro. Here, we report for the first time the total synthesis of callistrilone O in seven steps from phloroglucinol. A Friedel-Crafts-type Michael addition and an oxidative [3+2] cycloaddition with Fetizon's reagent were used to construct the molecular skeleton. The preferential cytotoxicity of callistrilone O was also evaluated with multiple starvation-resistant cancer cell lines under low nutrient conditions. Furthermore, callistrilone O was found to strongly suppress B16 melanoma tumor growth without critical toxicity in vivo. Overall, this study presents a novel anticancer agent candidate from natural products with a concise synthetic route which can be readily applied to the synthesis of derivatives.
Collapse
Affiliation(s)
- Kensuke Okuda
- Laboratory of Bioorganic & Natural Products Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita, Higashinada, Kobe, Hyogo, 658-8558, Japan
| | - Akira Takagi
- Laboratory of Bioorganic & Natural Products Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita, Higashinada, Kobe, Hyogo, 658-8558, Japan
| | - Ryohei Shimizu
- Laboratory of Microbial Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita, Higashinada, Kobe, Hyogo, 658-8558, Japan
- Department of Molecular Pharmaceutics, Hoshi University, 2-4-41 Ebara, Shinagawa, Tokyo, 142-8501, Japan
| | - Kensuke Nishi
- Laboratory of Bioorganic & Natural Products Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita, Higashinada, Kobe, Hyogo, 658-8558, Japan
| | - Narumi Hayano
- Laboratory of Microbial Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita, Higashinada, Kobe, Hyogo, 658-8558, Japan
| | - Ippei Takashima
- Laboratory of Bioorganic & Natural Products Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita, Higashinada, Kobe, Hyogo, 658-8558, Japan
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, 2-1-1 Katahira, Aoba, Sendai, Miyagi, 980-8577, Japan
| | - Morichika Konishi
- Laboratory of Microbial Chemistry, Kobe Pharmaceutical University, 4-19-1 Motoyamakita, Higashinada, Kobe, Hyogo, 658-8558, Japan
| |
Collapse
|
27
|
Mulero-Russe A, Mora-Boza A, Marquez EN, Ziegelski M, Helmrath M, García AJ. Synthetic hydrogel substrate for human induced pluripotent stem cell definitive endoderm differentiation. Biomaterials 2025; 315:122920. [PMID: 39504708 PMCID: PMC11625597 DOI: 10.1016/j.biomaterials.2024.122920] [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: 06/27/2024] [Revised: 10/18/2024] [Accepted: 10/23/2024] [Indexed: 11/08/2024]
Abstract
Human induced pluripotent stem cells (hiPSCs) can give rise to multiple lineages derived from three germ layers, endoderm, mesoderm and ectoderm. Definitive endoderm (DE) cell types and tissues have great potential for regenerative medicine applications. Current hiPSC differentiation protocols focus on the addition of soluble factors; however, extracellular matrix properties are known to also play a role in dictating cell fate. Matrigel™ is the gold standard for DE differentiation, but this xenogeneic, poorly defined basement membrane extract limits the clinical translatability of DE-derived tissues. Here we present a fully defined PEG-based hydrogel substrate to support hiPSC-derived DE differentiation. We screened hydrogel formulations presenting different adhesive peptides and matrix stiffness. Our results demonstrate that presenting a short peptide, cyclic RGD, on the engineered PEG hydrogel supports the transition from undifferentiated hiPSCs to DE using a serum-free, commercially available kit. We show that increasing substrate stiffness (G' = 1.0-4.0 kPa) results in an increased linear response in DE differentiation efficiency. We also include a temporal analysis of the expression of integrin and syndecan receptors as the hiPSCs undergo specification towards DE lineage. Finally, we show that focal adhesion kinase activity regulates hiPSC growth and DE differentiation efficiency. Overall, we present a fully defined matrix as a synthetic alternative for Matrigel™ supporting DE differentiation.
Collapse
Affiliation(s)
- Adriana Mulero-Russe
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Ana Mora-Boza
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Elijah N Marquez
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Morgan Ziegelski
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Michael Helmrath
- Division of Pediatric Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Andrés J García
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
28
|
Fukunaga I, Takebe T. In vitro liver models for toxicological research. Drug Metab Pharmacokinet 2025; 62:101478. [PMID: 40203632 DOI: 10.1016/j.dmpk.2025.101478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 02/25/2025] [Accepted: 03/04/2025] [Indexed: 04/11/2025]
Abstract
Drug-induced liver injury (DILI) presents a major challenge not only in new drug development but also in post-marketing withdrawals and the safety of food, cosmetics, and chemicals. Experimental model organisms such as the rodents have been widely used for preclinical toxicological testing. However, the tension exists associated with the ethical and sustainable use of animals in part because animals do not necessarily inform the human-specific ADME (adsorption, dynamics, metabolism and elimination) profiling. To establish alternative models in humans, in vitro hepatic tissue models have been proposed, ranging from primary hepatocytes, immortal hepatocytes, to the development of new cell resources such as stem cell-derived hepatocytes. Given the evolving number of novel alternative methods, understanding possible combinations of cell sources and culture methods will be crucial to develop the context-of-use assays. This review primarily focuses on 3D liver organoid models for conducting. We will review the relevant cell sources, bioengineering methods, selection of training compounds, and biomarkers towards the rationale design of in vitro toxicology testing.
Collapse
Affiliation(s)
- Ichiro Fukunaga
- Center for Genomic and Regenerative Medicine, Juntendo University Graduate School of Medicine, 2-1-1, Hongo, Bunkyo-ku, Tokyo, 113-8431, Japan.
| | - Takanori Takebe
- Human Biology Research Unit, Institute of Integrated Research, Institute of Science Tokyo, 1-5-45, Yushima, Bunkyo-ku, Tokyo, 113-8510, Japan; Department of Genome Biology, Graduate School of Medicine, Osaka University, Suita, Osaka, 565-0871, Japan; Divisions of Gastroenterology, Hepatology & Nutrition, Developmental Biology and Biomedical Informatics, Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229-3039, USA; Department of Pediatrics, University of Cincinnati College of Medicine, 3333 Burnet Avenue, Cincinnati, OH, 45229-3039, USA; Center for Stem Cell and Organoid Medicine (CuSTOM), Cincinnati Children's Hospital Medical Center, 3333 Burnet Avenue, Cincinnati, OH, 45229-3039, USA; Premium Research Institute for Human Metaverse Medicine (WPI-PRIMe), Osaka University, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
29
|
Kozalak G, Koşar A. Bone-on-a-Chip Systems for Hematological Cancers. BIOSENSORS 2025; 15:176. [PMID: 40136973 PMCID: PMC11940066 DOI: 10.3390/bios15030176] [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] [Subscribe] [Scholar Register] [Received: 02/04/2025] [Revised: 02/28/2025] [Accepted: 03/06/2025] [Indexed: 03/27/2025]
Abstract
Hematological malignancies originating from blood, bone marrow, and lymph nodes include leukemia, lymphoma, and myeloma, which necessitate the use of a distinct chemotherapeutic approach. Drug resistance frequently complicates their treatment, highlighting the need for predictive tools to guide therapeutic decisions. Conventional 2D/3D cell cultures do not fully encompass in vivo criteria, and translating disease models from mice to humans proves challenging. Organ-on-a-chip technology presents an avenue to surmount genetic disparities between species, offering precise design, concurrent manipulation of various cell types, and extrapolation of data to human physiology. The development of bone-on-a-chip (BoC) systems is crucial for accurately representing the in vivo bone microenvironment, predicting drug responses for hematological cancers, mitigating drug resistance, and facilitating personalized therapeutic interventions. BoC systems for modeling hematological cancers and drug research can encompass intricate designs and integrated platforms for analyzing drug response data to simulate disease scenarios. This review provides a comprehensive examination of BoC systems applicable to modeling hematological cancers and visualizing drug responses within the intricate context of bone. It thoroughly discusses the materials pertinent to BoC systems, suitable in vitro techniques, the predictive capabilities of BoC systems in clinical settings, and their potential for commercialization.
Collapse
Affiliation(s)
- Gül Kozalak
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul 34956, Turkey;
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabancı University, Istanbul 34956, Turkey
| | - Ali Koşar
- Faculty of Engineering and Natural Sciences, Sabancı University, Istanbul 34956, Turkey;
- Center of Excellence for Functional Surfaces and Interfaces for Nano Diagnostics (EFSUN), Sabancı University, Istanbul 34956, Turkey
- Turkish Academy of Sciences (TÜBA), Çankaya, Ankara 06700, Turkey
| |
Collapse
|
30
|
Wu P, Chen D, Wang F, Lu K, Sigurdsson EM, Jin C. Formaldehyde induces and promotes Alzheimer's disease pathologies in a 3D human neural cell culture system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.27.640690. [PMID: 40093146 PMCID: PMC11908216 DOI: 10.1101/2025.02.27.640690] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/19/2025]
Abstract
Alzheimer's disease (AD) arises from complex multilevel interactions between genetic, epigenetic, and environmental factors. Recent studies suggest that exposure to the environmental and occupational toxicant formaldehyde (FA) may play a significant role in AD development. However, the effects of FA exposure on Aβ and tau pathologies in human neural cell 3D culture systems remain unexplored. To investigate FA's role in AD initiation, we differentiated 3D-cultured immortalized human neural progenitor ReN cells (ReNcell VM) into neurons and glial cells, followed by FA treatment. FA exposure for 12 weeks resulted in a dose-dependent increase in Aβ40, Aβ42, and phosphorylated tau levels. To further examine FA's role in AD progression, we established a 3D human neural cell culture AD model by transfecting ReN cells with AD-related mutant genes, including mutant APP and PSEN1, which recapitulate key AD pathological events. Our findings demonstrate that FA exposure significantly elevated Aβ40, Aβ42, and phosphorylated tau levels in this 3D-cultured AD model. These results suggest that FA exposure contributes to the initiation and progression of AD pathology in 3D-cultured human neural cells.
Collapse
|
31
|
Martinet A, Miebach L, Weltmann K, Emmert S, Bekeschus S. Biomimetic Hydrogels - Tools for Regenerative Medicine, Oncology, and Understanding Medical Gas Plasma Therapy. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2403856. [PMID: 39905967 PMCID: PMC11878268 DOI: 10.1002/smll.202403856] [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: 05/13/2024] [Revised: 01/23/2025] [Indexed: 02/06/2025]
Abstract
Biomimetic hydrogels enable biochemical, cell biology, and tissue-like studies in the third dimension. Smart hydrogels are also frequently used in tissue engineering and as drug carriers for intra- or extracutaneous regenerative medicine. They have also been studied in bio-sensor development, 3D cell culture, and organoid growth optimization. Yet, many hydrogel types, adjuvant components, and cross-linking methods have emerged over decades, diversifying and complexifying such studies. Here, an evaluative overview is provided, mapping potential applications to the corresponding hydrogel tuning. Strikingly, hydrogels are ideal for studying locoregional therapy modalities, such as cold medical gas plasma technology. These partially ionized gases produce various reactive oxygen species (ROS) types along with other physico-chemical components such as ions and electric fields, and the spatio-temporal effects of these components delivered to diseased tissues remain largely elusive to date. Hence, this work outlines the promising applications of hydrogels in biomedical research in general and cold plasma science in particular and underlines the great potential of these smart scaffolds for current and future research and therapy.
Collapse
Affiliation(s)
- Alice Martinet
- Department of Dermatology and VenerologyRostock University Medical CenterStrempelstr. 1318057RostockGermany
- ZIK plasmatisLeibniz Institute for Plasma Science and Technology (INP)Felix‐Hausdorff‐Str. 217489GreifswaldGermany
| | - Lea Miebach
- ZIK plasmatisLeibniz Institute for Plasma Science and Technology (INP)Felix‐Hausdorff‐Str. 217489GreifswaldGermany
| | - Klaus‐Dieter Weltmann
- Department of Dermatology and VenerologyRostock University Medical CenterStrempelstr. 1318057RostockGermany
| | - Steffen Emmert
- Department of Dermatology and VenerologyRostock University Medical CenterStrempelstr. 1318057RostockGermany
| | - Sander Bekeschus
- Department of Dermatology and VenerologyRostock University Medical CenterStrempelstr. 1318057RostockGermany
- ZIK plasmatisLeibniz Institute for Plasma Science and Technology (INP)Felix‐Hausdorff‐Str. 217489GreifswaldGermany
| |
Collapse
|
32
|
Zhao J, Zhi Y, Ren H, Wang J, Zhao Y. Emerging biotechnologies for engineering liver organoids. Bioact Mater 2025; 45:1-18. [PMID: 39588483 PMCID: PMC11585797 DOI: 10.1016/j.bioactmat.2024.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 11/02/2024] [Accepted: 11/02/2024] [Indexed: 11/27/2024] Open
Abstract
The engineering construction of the liver has attracted enormous attention. Organoids, as emerging miniature three-dimensional cultivation units, hold significant potential in the biomimetic simulation of liver structure and function. Despite notable successes, organoids still face limitations such as high variability and low maturity. To overcome these challenges, engineering strategies have been established to maintain organoid stability and enhance their efficacy, laying the groundwork for the development of advanced liver organoids. The present review comprehensively summarizes the construction of engineered liver organoids and their prospective applications in biomedicine. Initially, we briefly present the latest research progress on matrix materials that maintain the three-dimensional morphology of organoids. Next, we discuss the manipulative role of engineering technologies in organoid assembly. Additionally, we outline the impact of gene-level regulation on organoid growth and development. Further, we introduce the applications of liver organoids in disease modeling, drug screening and regenerative medicine. Lastly, we overview the current obstacles and forward-looking perspectives on the future of engineered liver organoids. We anticipate that ongoing innovations in engineered liver organoids will lead to significant advancements in medical applications.
Collapse
Affiliation(s)
- Junqi Zhao
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
| | - Yue Zhi
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Haozhen Ren
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Jinglin Wang
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
| | - Yuanjin Zhao
- Department of Hepatobiliary Surgery, Hepatobiliary Institute, Nanjing Drum Tower Hospital, Medical School, Nanjing University, Nanjing, 210008, China
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
- Shenzhen Research Institute, Southeast University, Shenzhen, 518038, China
| |
Collapse
|
33
|
Sugihara HY, Okamoto R, Mizutani T. Intestinal organoids: The path towards clinical application. Eur J Cell Biol 2025; 104:151474. [PMID: 39740324 DOI: 10.1016/j.ejcb.2024.151474] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Revised: 11/14/2024] [Accepted: 11/17/2024] [Indexed: 01/02/2025] Open
Abstract
Organoids have revolutionized the whole field of biology with their ability to model complex three-dimensional human organs in vitro. Intestinal organoids were especially consequential as the first successful long-term culture of intestinal stem cells, which raised hopes for translational medical applications. Despite significant contributions to basic research, challenges remain to develop intestinal organoids into clinical tools for diagnosis, prognosis, and therapy. In this review, we outline the current state of translational research involving adult stem cell and pluripotent stem cell derived intestinal organoids, highlighting the advances and limitations in disease modeling, drug-screening, personalized medicine, and stem cell therapy. Preclinical studies have demonstrated a remarkable functional recapitulation of infectious and genetic diseases, and there is mounting evidence for the reliability of intestinal organoids as a patient-specific avatar. Breakthroughs now allow the generation of structurally and cellularly complex intestinal models to better capture a wider range of intestinal pathophysiology. As the field develops and evolves, there is a need for standardized frameworks for generation, culture, storage, and analysis of intestinal organoids to ensure reproducibility, comparability, and interpretability of these preclinical and clinical studies to ultimately enable clinical translation.
Collapse
Affiliation(s)
- Hady Yuki Sugihara
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Ryuichi Okamoto
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan
| | - Tomohiro Mizutani
- Department of Gastroenterology and Hepatology, Institute of Science Tokyo, 1-5-45, Yushima, Bunkyo-ku, Tokyo 113-8510, Japan.
| |
Collapse
|
34
|
Sisakht MM, Gholizadeh F, Hekmatirad S, Mahmoudi T, Montazeri S, Sharifi L, Daemi H, Romal S, Yazdi MH, Faramarzi MA, Shahverdi AR, Hamidieh AA. Cost-reduction strategy to culture patient derived bladder tumor organoids. Sci Rep 2025; 15:4223. [PMID: 39905065 PMCID: PMC11794879 DOI: 10.1038/s41598-025-87509-3] [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: 08/12/2024] [Accepted: 01/20/2025] [Indexed: 02/06/2025] Open
Abstract
Organoids as self-organized structure derived from stem cells can recapitulate the function of an organ in miniature form which have developed great potential for clinical translation, drug screening and personalized medicine. Nevertheless, the majority of patient-derived organoids (PDOs) are currently being cultured in the basement membrane matrices (BMMs), which are constrained by xenogeneic origin, batch-to-batch variability, cost, and complexity. Besides, organoid culture relies on biochemical signals provided by various growth factors in the composition of medium. We propose sodium alginate hydrogel scaffold in addition to the fibroblast conditioned medium (FCM)-enriched culture medium that is inexpensive and easily amenable to clinical applications for the culture of bladder cancer PDOs. PDOs grown in sodium alginate and FCM based medium have proliferation potential, growth rate, and gene expression that are similar to PDOs cultured in BME. According to the results, sodium alginate has substantial mechanical properties and reduces variance in early passage bladder tumor organoid cultures collected from patients. Furthermore, using FCM based medium as an alternative solution to eliminate some essential growth factors can be considered, especially for low-resource situation and develop cost effective tumor organoids.
Collapse
Affiliation(s)
- Mahsa Mollapour Sisakht
- Biotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
- Stem Cell and Regenerative Medicine innovation center, Tehran University of Medical Sciences, Tehran, Iran.
| | - Fatemeh Gholizadeh
- Stem Cell and Regenerative Medicine innovation center, Tehran University of Medical Sciences, Tehran, Iran
| | - Shirin Hekmatirad
- Stem Cell and Regenerative Medicine innovation center, Tehran University of Medical Sciences, Tehran, Iran
| | - Tokameh Mahmoudi
- Department of Urology, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Saeed Montazeri
- Uro-oncology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Laleh Sharifi
- Uro-oncology Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Hamed Daemi
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran
| | - Shahla Romal
- Department of Urology, Erasmus MC Cancer Institute, Erasmus University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Mohammad Hosein Yazdi
- Biotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Ali Faramarzi
- Biotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Ahmad Reza Shahverdi
- Biotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Recombinant Vaccine Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Amir Ali Hamidieh
- Biotechnology Research Center, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
- Pediatric Cell and Gene Therapy Research Center, Gene, Cell & Tissue Research Institute, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
35
|
Dutta A, Chowdhury N, Chandra S, Guha P, Saha V, GuhaSarkar D. Gallbladder cholangiocyte organoids. Biol Cell 2025; 117:e2400132. [PMID: 39945546 PMCID: PMC11823593 DOI: 10.1111/boc.202400132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/23/2024] [Accepted: 01/30/2025] [Indexed: 02/16/2025]
Abstract
Organoids are miniature three-dimensional (3D) organ-like structures developed from primary cells that closely mimic the key histological, functional, and molecular characteristics of their parent organs. These structures self-organize through cell-cell and cell-matrix interaction in culture. In the last decade, organoids and allied 3D culture technologies have catalyzed studies involving developmental biology, disease biology, high-throughput drug screening, personalized medicine, biomarker discovery, tissue engineering, and regenerative medicine. Many organoid systems have been generated from the gastrointestinal system, for example, intestine, stomach, liver, pancreas, or colon. Gallbladder cancer (GBC) is the most common and highly aggressive form of biliary tract cancer. GBC is rare in the west but has a high incidence in South America and India. Prolonged chronic inflammation is implicated in the pathogenesis of GBC but the driving molecular pathways leading to neoplasia are not well understood. Gallbladder cholangiocyte organoids (GCO) will facilitate the understanding of the evolution of the disease and novel therapeutic strategies. In this review, we have discussed alternative methodologies and culture conditions developed to generate GCO models, applications that these models have been subjected to and the current limitations for the use of GCOs in addressing the challenges in GBC research.
Collapse
Affiliation(s)
- Ankita Dutta
- SOLi3D LaboratoryTata Translational Cancer Research CentreKolkataIndia
- School of Medical Science and TechnologyIndian Institute of Technology KharagpurKharagpurIndia
| | - Nandita Chowdhury
- SOLi3D LaboratoryTata Translational Cancer Research CentreKolkataIndia
| | - Shinjini Chandra
- SOLi3D LaboratoryTata Translational Cancer Research CentreKolkataIndia
| | - Payel Guha
- SOLi3D LaboratoryTata Translational Cancer Research CentreKolkataIndia
| | - Vaskar Saha
- SOLi3D LaboratoryTata Translational Cancer Research CentreKolkataIndia
- Department of Paediatric Haematology and Oncology Tata Medical CenterKolkataIndia
- Division of Cancer SciencesFaculty of BiologyMedicine and HealthSchool of Medical SciencesUniversity of ManchesterManchesterUK
| | - Dwijit GuhaSarkar
- SOLi3D LaboratoryTata Translational Cancer Research CentreKolkataIndia
| |
Collapse
|
36
|
Antonacci M, Maqoud F, Di Turi A, Miciaccia M, Perrone MG, Scilimati A, Tricarico D. KATP Channel Inhibitors Reduce Cell Proliferation Through Upregulation of H3K27ac in Diffuse Intrinsic Pontine Glioma: A Functional Expression Investigation. Cancers (Basel) 2025; 17:358. [PMID: 39941728 PMCID: PMC11816144 DOI: 10.3390/cancers17030358] [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: 12/20/2024] [Revised: 01/04/2025] [Accepted: 01/14/2025] [Indexed: 02/16/2025] Open
Abstract
BACKGROUND Diffuse intrinsic pontine glioma [DIPG] is a fatal pediatric disease characterized by a post-translational modification, a replacement of lysine by methionine in position 27 of the N-terminal [H3K27M] tail of histone 3 isoform-1 [H3.1] or histone 3 isoform-3 [H3.3], respectively, expressed in the DIPG-36 and DIPG-50 cells. We investigated the role of cation channels in DIPG cells for the first time and the effects of ATP-sensitive K+[KATP] and TRPV1 channel modulators. METHODS Experiments were performed using "in vitro" cytotoxic assays combined with the patch clamp technique, RT-PCR, Western blot, and flow cytometry assays. RESULTS The most effective anti-proliferative drugs were repaglinide and glibenclamide after short and long-term incubation [6-96 h]. These drugs reduced macroscopic currents of the DIPG cells recorded in whole-cell patch clamp. Repaglinide concentration dependently enhanced the target protein H3K27ac in Western blotting after 48 h of incubation. This drug reduced cell diameter and enhanced cleaved caspase-3 in DIPG cells; total AKT/mTOR levels and phospho-mTOR were downregulated in DIPG-36. CONCLUSIONS KATP and TRPV1 channels are functionally expressed, and sulphonylureas are effective antiproliferative upregulating H3K27ac with apoptosis in DIPG cells and the sub-micromolar concentrations in DIPG-50.
Collapse
Affiliation(s)
- Marina Antonacci
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (M.A.); (F.M.); (A.D.T.); (M.M.); (M.G.P.)
| | - Fatima Maqoud
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (M.A.); (F.M.); (A.D.T.); (M.M.); (M.G.P.)
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology Saverio de Bellis, I.R.C.C.S. Research Hospital, 70013 Castellana Grotte, Italy
| | - Annamaria Di Turi
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (M.A.); (F.M.); (A.D.T.); (M.M.); (M.G.P.)
| | - Morena Miciaccia
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (M.A.); (F.M.); (A.D.T.); (M.M.); (M.G.P.)
| | - Maria Grazia Perrone
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (M.A.); (F.M.); (A.D.T.); (M.M.); (M.G.P.)
| | - Antonio Scilimati
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (M.A.); (F.M.); (A.D.T.); (M.M.); (M.G.P.)
| | - Domenico Tricarico
- Department of Pharmacy-Pharmaceutical Sciences, University of Bari “Aldo Moro”, 70125 Bari, Italy; (M.A.); (F.M.); (A.D.T.); (M.M.); (M.G.P.)
| |
Collapse
|
37
|
Dohi K, Manabe Y, Fujii NL, Furuichi Y. Achieving myoblast engraftment into intact skeletal muscle via extracellular matrix. Front Cell Dev Biol 2025; 12:1502332. [PMID: 39877158 PMCID: PMC11772487 DOI: 10.3389/fcell.2024.1502332] [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: 09/26/2024] [Accepted: 11/29/2024] [Indexed: 01/31/2025] Open
Abstract
Cell therapy of skeletal muscles is a promising approach for the prevention of muscular diseases and age-related muscle atrophy. However, cell transplantation to treat muscle atrophy that does not involve disease, such as sarcopenia, is considered impossible because externally injected cells rarely engraft into non-injured muscle tissue. Additionally, skeletal muscle-specific somatic stem cells, called satellite cells, lose their ability to adhere to tissue after being cultured in vitro and transforming into myoblasts. To overcome these hurdles, we explored using extracellular matrix (ECM) components to create a niche environment conducive for myoblasts during transplantation. We demonstrated that myoblasts mixed with ECM components can be engrafted into intact skeletal muscle and significantly increase muscle mass in a mouse model. These findings implicate cell transplantation therapy as a viable option for the treatment of sarcopenia. The findings will inform advancements in regenerative medicine for skeletal muscles.
Collapse
Affiliation(s)
| | | | | | - Yasuro Furuichi
- Department of Health Promotion Sciences, Graduated School of Human Health Sciences, Tokyo Metropolitan University, Hachioji, Japan
| |
Collapse
|
38
|
Eiken MK, Childs CJ, Brastrom LK, Frum T, Plaster EM, Ahmed DW, Spencer RC, Shachaf O, Pfeiffer S, Levine JE, Alysandratos KD, Kotton DN, Spence JR, Loebel C. Nascent matrix deposition supports alveolar organoid formation from aggregates in synthetic hydrogels. Stem Cell Reports 2025; 20:102376. [PMID: 39672155 PMCID: PMC11784465 DOI: 10.1016/j.stemcr.2024.11.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 11/12/2024] [Accepted: 11/14/2024] [Indexed: 12/15/2024] Open
Abstract
Human induced pluripotent stem cell (iPSC)-derived alveolar organoids have emerged as a system to model the alveolar epithelium in homeostasis and disease. However, alveolar organoids are typically grown in Matrigel, a mouse sarcoma-derived basement membrane matrix that offers poor control over matrix properties, prompting the development of synthetic hydrogels as a Matrigel alternative. Here, we develop a two-step culture method that involves pre-aggregation of organoids in hydrogel-based microwells followed by embedding in a synthetic hydrogel that supports alveolar organoid growth, while also offering considerable control over organoid and hydrogel properties. We find that the aggregated organoids secrete their own nascent extracellular matrix (ECM) both in the microwells and upon embedding in synthetic hydrogels, which supports their growth. Thus, the synthetic hydrogels described here allow us to de-couple exogenous and nascent ECM to interrogate the role of ECM in organoid formation.
Collapse
Affiliation(s)
- Madeline K Eiken
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Charlie J Childs
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Lindy K Brastrom
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Tristan Frum
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Eleanor M Plaster
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Donia W Ahmed
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Ryan C Spencer
- Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Orren Shachaf
- Department of Biomedical Engineering, University of Texas, Austin, TX, USA
| | - Suzanne Pfeiffer
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Justin E Levine
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA
| | - Konstantinos-Dionysios Alysandratos
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Darrell N Kotton
- Center for Regenerative Medicine, Boston University and Boston Medical Center, Boston, MA 02118, USA; The Pulmonary Center and Department of Medicine, Boston University Chobanian & Avedisian School of Medicine, Boston, MA 02118, USA
| | - Jason R Spence
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA; Department of Internal Medicine, University of Michigan Medical School, Ann Arbor, MI, USA; Department of Cell and Developmental Biology, University of Michigan Medical School, Ann Arbor, MI, USA; Program in Cell and Molecular Biology, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Claudia Loebel
- Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, USA; Department of Materials Science and Engineering, University of Michigan, Ann Arbor, MI, USA.
| |
Collapse
|
39
|
Wijnakker JJ, van Son GJ, Krueger D, van de Wetering WJ, Lopez-Iglesias C, Schreurs R, van Rijt F, Lim S, Lin L, Peters PJ, Isberg RR, Janda CY, de Lau W, Clevers H. Integrin-activating Yersinia protein Invasin sustains long-term expansion of primary epithelial cells as 2D organoid sheets. Proc Natl Acad Sci U S A 2025; 122:e2420595121. [PMID: 39793062 PMCID: PMC11725944 DOI: 10.1073/pnas.2420595121] [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: 10/07/2024] [Accepted: 11/14/2024] [Indexed: 01/12/2025] Open
Abstract
Matrigel®/BME®, a basement membrane-like preparation, supports long-term growth of epithelial 3D organoids from adult stem cells [T. Sato et al., Nature 459, 262-265 (2009); T. Sato et al., Gastroenterology 141, 1762-1772 (2011)]. Here, we show that interaction between Matrigel's major component laminin-111 with epithelial α6β1-integrin is crucial for this process. The outer membrane protein Invasin of Yersinia is known to activate multiple integrin-β1 complexes, including integrin α6β1. A C-terminal integrin-binding fragment of Invasin, coated on culture plates, mediated gut epithelial cell adhesion. Addition of organoid growth factors allowed multipassage expansion in 2D. Polarization, junction formation, and generation of enterocytes, goblet cells, Paneth cells, and enteroendocrine cells were stable over time. Sustained expansion of other human, mouse, and even snake epithelia was accomplished under comparable conditions. The 2D "organoid sheet" format holds advantages over the 3D "in gel" format in terms of imaging, accessibility of basal and apical domains, and automation for high-throughput screening. Invasin represents a fully defined, affordable, versatile, and animal-free complement to Matrigel®/BME®.
Collapse
Affiliation(s)
- Joost J.A.P.M. Wijnakker
- Oncode Institute, Hubrecht Institute-Royal Netherlands Academy of Arts and Science, Utrecht3584 CT, The Netherlands
- University Medical Centre, Utrecht3584 CT, The Netherlands
| | - Gijs J.F. van Son
- Princess Maxima Center of Pediatric Oncology, Utrecht3584 CS, The Netherlands
| | - Daniel Krueger
- Oncode Institute, Hubrecht Institute-Royal Netherlands Academy of Arts and Science, Utrecht3584 CT, The Netherlands
- University Medical Centre, Utrecht3584 CT, The Netherlands
| | | | - Carmen Lopez-Iglesias
- The Maastricht Multimodal Imaging Institute, Maastricht University, Maastricht6229 ER, The Netherlands
| | - Robin Schreurs
- Oncode Institute, Hubrecht Institute-Royal Netherlands Academy of Arts and Science, Utrecht3584 CT, The Netherlands
- University Medical Centre, Utrecht3584 CT, The Netherlands
| | - Fenna van Rijt
- Princess Maxima Center of Pediatric Oncology, Utrecht3584 CS, The Netherlands
| | - Sangho Lim
- Oncode Institute, Hubrecht Institute-Royal Netherlands Academy of Arts and Science, Utrecht3584 CT, The Netherlands
- University Medical Centre, Utrecht3584 CT, The Netherlands
| | - Lin Lin
- Oncode Institute, Hubrecht Institute-Royal Netherlands Academy of Arts and Science, Utrecht3584 CT, The Netherlands
- University Medical Centre, Utrecht3584 CT, The Netherlands
- Princess Maxima Center of Pediatric Oncology, Utrecht3584 CS, The Netherlands
| | - Peter J. Peters
- The Maastricht Multimodal Imaging Institute, Maastricht University, Maastricht6229 ER, The Netherlands
| | - Ralph R. Isberg
- Department of Molecular Biology and Microbiology, School of Medicine, Tufts University, Boston, MA02111
| | - Claudia Y. Janda
- Princess Maxima Center of Pediatric Oncology, Utrecht3584 CS, The Netherlands
| | - Wim de Lau
- Oncode Institute, Hubrecht Institute-Royal Netherlands Academy of Arts and Science, Utrecht3584 CT, The Netherlands
- University Medical Centre, Utrecht3584 CT, The Netherlands
| | - Hans Clevers
- Oncode Institute, Hubrecht Institute-Royal Netherlands Academy of Arts and Science, Utrecht3584 CT, The Netherlands
- University Medical Centre, Utrecht3584 CT, The Netherlands
- Princess Maxima Center of Pediatric Oncology, Utrecht3584 CS, The Netherlands
| |
Collapse
|
40
|
Larrañaga E, Marin-Riera M, Abad-Lázaro A, Bartolomé-Català D, Otero A, Fernández-Majada V, Batlle E, Sharpe J, Ojosnegros S, Comelles J, Martinez E. Long-range organization of intestinal 2D-crypts using exogenous Wnt3a micropatterning. Nat Commun 2025; 16:382. [PMID: 39753580 PMCID: PMC11698991 DOI: 10.1038/s41467-024-55651-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2023] [Accepted: 12/19/2024] [Indexed: 01/06/2025] Open
Abstract
Intestinal epithelial cells are segregated into proliferative crypts and differentiated regions. This organization relies on specific signals, including Wnt3a, which regulates cell proliferation within crypts, and Eph/Ephrin, which dictates cell positioning along the crypt-villus axis. However, studying how the spatial distributions of these signals influences crypt-villus organization is challenging both in vitro and in vivo. Here we show that micropatterns of Wnt3a can govern the size, shape and long-range organization of crypts in vitro. By adjusting the spacing between Wnt3a ligand patterns at the microscale over large surfaces, we override endogenous Wnt3a to precisely control the distribution and long-range order of crypt-like regions in primary epithelial monolayers. Additionally, an agent-based model integrating Wnt3a/BMP feedback and Eph/Ephrin repulsion effectively replicates experimental tissue compartmentalization, crypt size, shape, and organization. This combined experimental and computational approach offers a framework to study how signaling pathways help organize intestinal epithelial tissue.
Collapse
Affiliation(s)
- Enara Larrañaga
- Biomimetic Systems for Cell Engineering Laboratory, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | | | - Aina Abad-Lázaro
- Biomimetic Systems for Cell Engineering Laboratory, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - David Bartolomé-Català
- Biomimetic Systems for Cell Engineering Laboratory, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Aitor Otero
- Biomimetic Systems for Cell Engineering Laboratory, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Vanesa Fernández-Majada
- Biomimetic Systems for Cell Engineering Laboratory, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Eduard Batlle
- Institute for Research in Biomedicine (IRB), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
- Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - James Sharpe
- European Molecular Biology Laboratory (EMBL), Barcelona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
| | - Samuel Ojosnegros
- Bioengineering in Reproductive Health, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain
| | - Jordi Comelles
- Biomimetic Systems for Cell Engineering Laboratory, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Department of Electronics and Biomedical Engineering, University of Barcelona (UB), Barcelona, Spain.
| | - Elena Martinez
- Biomimetic Systems for Cell Engineering Laboratory, Institute for Bioengineering of Catalonia (IBEC), The Barcelona Institute of Science and Technology (BIST), Barcelona, Spain.
- Department of Electronics and Biomedical Engineering, University of Barcelona (UB), Barcelona, Spain.
- Centro de Investigación Biomédica en Red - Bioingeniería, Biomateriales y Nanomedicina (CIBER-BBN), Madrid, Spain.
| |
Collapse
|
41
|
Sajjad MW, Muzamil F, Sabir M, Ashfaq UA. Regenerative Medicine and Nanotechnology Approaches against Cardiovascular Diseases: Recent Advances and Future Prospective. Curr Stem Cell Res Ther 2025; 20:50-71. [PMID: 38343052 DOI: 10.2174/011574888x263530230921074827] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2023] [Revised: 07/07/2023] [Accepted: 07/14/2023] [Indexed: 01/31/2025]
Abstract
Regenerative medicine refers to medical research focusing on repairing, replacing, or regenerating damaged or diseased tissues or organs. Cardiovascular disease (CVDs) is a significant health issue globally and is the leading cause of death in many countries. According to the Centers for Disease Control and Prevention (CDC), one person dies every 34 seconds in the United States from cardiovascular diseases, and according to a World Health Organization (WHO) report, cardiovascular diseases are the leading cause of death globally, taking an estimated 17.9 million lives each year. Many conventional treatments are available using different drugs for cardiovascular diseases, but these treatments are inadequate. Stem cells and nanotechnology are promising research areas for regenerative medicine treating CVDs. Regenerative medicines are a revolutionary strategy for advancing and successfully treating various diseases, intending to control cardiovascular disorders. This review is a comprehensive study of different treatment methods for cardiovascular diseases using different types of biomaterials as regenerative medicines, the importance of different stem cells in therapeutics, the expanded role of nanotechnology in treatment, the administration of several types of stem cells, their tracking, imaging, and the final observation of clinical trials on many different levels as well as it aims to keep readers up to pace on emerging therapeutic applications of some specific organs and disorders that may improve from regenerative medicine shortly.
Collapse
Affiliation(s)
- Muhammad Waseem Sajjad
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Fatima Muzamil
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Maida Sabir
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| | - Usman Ali Ashfaq
- Department of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
| |
Collapse
|
42
|
Olvera-Valencia M, Garcia-Castillo V, Ramos-Payan R, Aguilar-Medina M, Trujano-Camacho S, López-Saavedra A, Marchat LA, López-Camarillo C, Sumagin R, Pérez-Yepez E, Pérez-Plasencia C. Development of a reliable method for human triple-negative breast cancer organotypic culture: Improving imaging and genomic studies in 3D cultures. J Tissue Eng 2025; 16:20417314251326668. [PMID: 40342587 PMCID: PMC12059422 DOI: 10.1177/20417314251326668] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 02/25/2025] [Indexed: 05/11/2025] Open
Abstract
Triple-negative breast cancer (TNBC) is highly aggressive and lacks targeted therapies, posing a major challenge in oncology. Traditional two-dimensional (2D) cell cultures fail to capture the tumor microenvironment's complexity, whereas three-dimensional (3D) cultures provide a more accurate model of tumor biology. We developed an advanced 3D culture system for TNBC cell lines BT-20 and MDA-MB-231, enhancing the hanging-drop method with Matrigel to restore essential extracellular matrix interactions. Confocal imaging showed MDA-MB-231 cells forming clusters typical of aggressive carcinoma, while BT-20 cells organized into duct-like structures. Molecular analysis of PI3K and β-catenin target genes revealed distinct expression patterns, with PI3K overexpressed and β-catenin downregulated in 3D cultures. Moreover, β-catenin distribution in the 3D cell culture closely resembles its pattern in tissue. These findings underscore the value of 3D models in understanding TNBC progression and in supporting the exploration of novel therapeutic strategies.
Collapse
Affiliation(s)
- Mercedes Olvera-Valencia
- Programa Institucional de Biomedicina Molecular, Escuela Nacional de Medicina y Homeopatía del Instituto Politécnico Nacional, Ticoman, CDMX, Mexico
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Tlalpan, CDMX, Mexico
| | - Verónica Garcia-Castillo
- Laboratorio de Genómica Funcional, Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla Estado de México, Mexico
| | - Rosalío Ramos-Payan
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacan, Sinaloa, Mexico
| | - Maribel Aguilar-Medina
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacan, Sinaloa, Mexico
| | - Samuel Trujano-Camacho
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Tlalpan, CDMX, Mexico
- Experimental Biology PhD Program, DCBS, Universidad Autónoma Metropolitana- Iztapalapa, Iztapalapa, Mexico
| | - Alejandro López-Saavedra
- Advanced Microscopy Applications Unit (ADMIRA)-Instituto Nacional de Cancerología, Tlalpan, CDMX, Mexico
- Tecnológico de Monterrey, Escuela de Medicina y Ciencias de la Salud, Mexico City, Mexico
| | - Laurence A. Marchat
- Programa Institucional de Biomedicina Molecular, Escuela Nacional de Medicina y Homeopatía del Instituto Politécnico Nacional, Ticoman, CDMX, Mexico
| | - Cesar López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de México, Benito Juarez, CDMX, Mexico
| | - Ronen Sumagin
- Department of Pathology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Eloy Pérez-Yepez
- Laboratorio de Genómica, Instituto Nacional de Cancerología, Tlalpan, CDMX, Mexico
| | - Carlos Pérez-Plasencia
- Laboratorio de Genómica Funcional, Unidad de Biomedicina, Facultad de Estudios Superiores Iztacala, UNAM, Tlalnepantla Estado de México, Mexico
| |
Collapse
|
43
|
Wongpakham T, Chunfong T, Jeamsaksiri W, Chessadangkul K, Bhanpattanakul S, Kallayanathum W, Tharasanit T, Pimpin A. Development of Pyramidal Microwells for Enhanced Cell Spheroid Formation in a Cell-on-Chip Microfluidic System for Cardiac Differentiation of Mouse Embryonic Stem Cells. Cells 2024; 13:2132. [PMID: 39768221 PMCID: PMC11674798 DOI: 10.3390/cells13242132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
Three-dimensional (3D) tissue culture models provide in vivo-like conditions for studying cell physiology. This study aimed to examine the efficiency of pyramidal microwell geometries in microfluidic devices on spheroid formation, cell growth, viability, and differentiation in mouse embryonic stem cells (mESCs). The static culture using the hanging drop (HD) method served as a control. The microfluidic chips were fabricated to have varying pyramidal tip angles, including 66°, 90°, and 106°. From flow simulations, when the tip angle increased, streamline distortion decreased, resulting in more uniform flow and a lower velocity gradient near the spheroids. These findings demonstrate the significant influence of microwell geometry on fluid dynamics. The 90° microwells provide optimal conditions, including uniform flow and reduced shear stress, while maintaining the ability for waste removal, resulting in superior spheroid growth compared to the HD method and other microwell designs. From the experiments, by Day 3, spheroids in the 90° microwells reached approximately 400 µm in diameter which was significantly larger than those in the 66° microwells, 106° microwells, and HD cultures. Brachyury gene expression in the 90° microwells was four times higher than the HD method, indicating enhanced mesodermal differentiation essential for cardiac differentiation. Immunofluorescence staining confirmed cardiomyocyte differentiation. In conclusion, microwell geometry significantly influences 3D cell culture outcomes. The pyramidal microwells with a 90° tip angle proved most effective in promoting spheroid growth and cardiac differentiation of mESC differentiation, providing insights for optimizing microfluidic systems in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Tepparit Wongpakham
- Department of Mechanical Engineering, Faculty of Engineering, Chulalongkorn University, Bangkok 10330, Thailand; (T.W.); (T.C.); (K.C.)
| | - Thanapat Chunfong
- Department of Mechanical Engineering, Faculty of Engineering, Chulalongkorn University, Bangkok 10330, Thailand; (T.W.); (T.C.); (K.C.)
| | - Wutthinan Jeamsaksiri
- Thai Microelectronics Center, National Electronics and Computer Technology Center, Chachoengsao 24000, Thailand;
| | - Kriengkai Chessadangkul
- Department of Mechanical Engineering, Faculty of Engineering, Chulalongkorn University, Bangkok 10330, Thailand; (T.W.); (T.C.); (K.C.)
| | - Sudchaya Bhanpattanakul
- Department of Pathology, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand;
- Center of Excellence for Veterinary Clinical Stem Cells and Bioengineering, Chulalongkorn University, Bangkok 10330, Thailand
| | - Wirakan Kallayanathum
- Department of Obstetrics, Gynaecology and Reproduction, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Theerawat Tharasanit
- Center of Excellence for Veterinary Clinical Stem Cells and Bioengineering, Chulalongkorn University, Bangkok 10330, Thailand
- Department of Obstetrics, Gynaecology and Reproduction, Faculty of Veterinary Science, Chulalongkorn University, Bangkok 10330, Thailand;
| | - Alongkorn Pimpin
- Department of Mechanical Engineering, Faculty of Engineering, Chulalongkorn University, Bangkok 10330, Thailand; (T.W.); (T.C.); (K.C.)
- Micro/Nano Electromechanical Integrated Device Research Unit, Faculty of Engineering, Chulalongkorn University, Bangkok 10330, Thailand
| |
Collapse
|
44
|
Sonnentag SJ, Ibrahim NSM, Orian-Rousseau V. CD44: a stemness driver, regulator, and marker-all in one? Stem Cells 2024; 42:1031-1039. [PMID: 39364735 DOI: 10.1093/stmcls/sxae060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 08/23/2024] [Indexed: 10/05/2024]
Abstract
Although the concept of cancer stem cells is still controversial, previous studies have shown that blood cancers, as well as specific types of solid cancers such as colorectal cancer, rely on stem cells during the onset of tumor growth and further tumor development. Moreover, resistance to therapeutic treatment in leukemias such as acute myeloid leukemia and in colorectal cancer can be attributed to a small population of cells with stemness properties known as minimal residual disease. In this review, we look back on the discovery of cancer stem cells and the contribution of the findings in blood cancer to a parallel discovery in solid cancers. We focus on CD44 as a stem cell marker, both in blood cancers and in several types of solid cancers, particularly of the gastrointestinal tract. This review highlights newly discovered molecular mechanisms of action of CD44 which indicate that CD44 has indeed a function in stemness, stem cell maintenance, and drug resistance. We attempt here to make the link between the functions of CD44 isoforms in stemness and their involvement in specific steps of tumor growth and metastasis.
Collapse
Affiliation(s)
- Steffen J Sonnentag
- Karlsruhe Institute of Technology, Institute of Biological and Chemical Systems-Functional Molecular Systems, Kaiserstraße 12, 76131 Karlsruhe, Germany
| | - Nagwa S M Ibrahim
- Karlsruhe Institute of Technology, Institute of Biological and Chemical Systems-Functional Molecular Systems, Kaiserstraße 12, 76131 Karlsruhe, Germany
| | - Veronique Orian-Rousseau
- Karlsruhe Institute of Technology, Institute of Biological and Chemical Systems-Functional Molecular Systems, Kaiserstraße 12, 76131 Karlsruhe, Germany
| |
Collapse
|
45
|
Nitaramorn N, Kobpornchai P, Tongkrajang N, Chaisri U, Imwong M, Kulkeaw K. Human liver organoids are susceptible to Plasmodium vivax infection. Malar J 2024; 23:368. [PMID: 39639330 PMCID: PMC11622667 DOI: 10.1186/s12936-024-05202-8] [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: 05/15/2024] [Accepted: 11/28/2024] [Indexed: 12/07/2024] Open
Abstract
BACKGROUND The eradication of Plasmodium vivax malaria is complicated due to the presence of hypnozoites, the hidden dormant form of the parasite that is present in the liver. Currently available drug regimens are effective at killing hypnozoites but cause side effects and are difficult to administer. Studies testing drugs for liver-stage malaria remain rare and mainly rely on the use of cancerous or immortalized hepatic cells and primary hepatocytes. METHODS Organoids were used as platform to model liver-stage vivax malaria. Hepatic endoderm cells, endothelial progenitor cells and mesenchymal cells were generated from human induced pluripotent stem cells and self-assembled into liver organoids on top of Matrigel layer. Liver characteristic and maturity were examined through genes and proteins expression of liver markers, and liver functional tests before infected with Plasmodium vivax sporozoites. The infection was then verified by the detection of parasitophorous vacuole membrane proteins, Upregulated in Infectious Sporozoite 4 (UIS4), and blood-stage infection following co-culture with human reticulocytes. RESULTS Generated liver organoids showed upregulation of liver specific transcripts including hepatic nuclear factor 4A (HNF4A), alpha-fetoprotein (AFP), and albumin (ALB) which also confirmed by the protein expression. Furthermore, those organoids resembled mature hepatocytes in terms of albumin secretion, fat and glycogen storage and cytochrome activity. Following invasion of P. vivax sporozoites, PvUIS4 was detected and the hepatic merozoites could develop into ring-stage and early trophozoites in human reticulocytes. Moreover, differential expression patterns of genes involved in lipid and cholesterol synthesis were also detected. CONCLUSIONS Stem cell-derived liver organoids resemble mature liver cells in terms of liver functions and are susceptible to infection with P. vivax sporozoites, paving the way for studies on the mechanism of hypnozoite formation and testing of possible hypnozoitocidal drugs.
Collapse
Affiliation(s)
- Norapat Nitaramorn
- Graduate Program in Biodesign in Medicine, Department of Parasitology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Porntida Kobpornchai
- Siriraj Integrative Center for Neglected Parasitic Diseases, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
- Siriraj-Long Read Laboratory, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Nongnat Tongkrajang
- Siriraj Integrative Center for Neglected Parasitic Diseases, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand
| | - Urai Chaisri
- Department of Tropical Pathology, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Mallika Imwong
- Department of Molecular Tropical Medicine and Genetics, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Kasem Kulkeaw
- Siriraj Integrative Center for Neglected Parasitic Diseases, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
- Siriraj-Long Read Laboratory, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, 10700, Thailand.
| |
Collapse
|
46
|
Ireland J, Kilian KA. The importance of matrix in cardiomyogenesis: Defined substrates for maturation and chamber specificity. Matrix Biol Plus 2024; 24:100160. [PMID: 39291079 PMCID: PMC11403269 DOI: 10.1016/j.mbplus.2024.100160] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 08/15/2024] [Accepted: 08/16/2024] [Indexed: 09/19/2024] Open
Abstract
Human embryonic stem cell-derived cardiomyocytes (hESC-CM) are a promising source of cardiac cells for disease modelling and regenerative medicine. However, current protocols invariably lead to mixed population of cardiac cell types and often generate cells that resemble embryonic phenotypes. Here we developed a combinatorial approach to assess the importance of extracellular matrix proteins (ECMP) in directing the differentiation of cardiomyocytes from human embryonic stem cells (hESC). We did this by focusing on combinations of ECMP commonly found in the developing heart with a broad goal of identifying combinations that promote maturation and influence chamber specific differentiation. We formulated 63 unique ECMP combinations fabricated from collagen 1, collagen 3, collagen 4, fibronectin, laminin, and vitronectin, presented alone and in combinations, leading to the identification of specific ECMP combinations that promote hESC proliferation, pluripotency, and germ layer specification. When hESC were subjected to a differentiation protocol on the ECMP combinations, it revealed precise protein combinations that enhance differentiation as determined by the expression of cardiac progenitor markers kinase insert domain receptor (KDR) and mesoderm posterior transcription factor 1 (MESP1). High expression of cardiac troponin (cTnT) and the relative expression of myosin light chain isoforms (MLC2a and MLC2v) led to the identification of three surfaces that promote a mature cardiomyocyte phenotype. Action potential morphology was used to assess chamber specificity, which led to the identification of matrices that promote chamber-specific cardiomyocytes. This study provides a matrix-based approach to improve control over cardiomyocyte phenotypes during differentiation, with the scope for translation to cardiac laboratory models and for the generation of functional chamber specific cardiomyocytes for regenerative therapies.
Collapse
Affiliation(s)
- Jake Ireland
- School of Chemistry, UNSW Sydney, Sydney, New South Wales, Australia
| | - Kristopher A Kilian
- School of Chemistry, UNSW Sydney, Sydney, New South Wales, Australia
- School of Materials Science and Engineering, UNSW Sydney, Sydney, New South Wales, Australia
- Australian Centre for NanoMedicine, UNSW Sydney, Sydney, New South Wales, Australia
| |
Collapse
|
47
|
Du C, Liu J, Liu S, Xiao P, Chen Z, Chen H, Huang W, Lei Y. Bone and Joint-on-Chip Platforms: Construction Strategies and Applications. SMALL METHODS 2024; 8:e2400436. [PMID: 38763918 DOI: 10.1002/smtd.202400436] [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: 03/25/2024] [Revised: 04/28/2024] [Indexed: 05/21/2024]
Abstract
Organ-on-a-chip, also known as "tissue chip," is an advanced platform based on microfluidic systems for constructing miniature organ models in vitro. They can replicate the complex physiological and pathological responses of human organs. In recent years, the development of bone and joint-on-chip platforms aims to simulate the complex physiological and pathological processes occurring in human bones and joints, including cell-cell interactions, the interplay of various biochemical factors, the effects of mechanical stimuli, and the intricate connections between multiple organs. In the future, bone and joint-on-chip platforms will integrate the advantages of multiple disciplines, bringing more possibilities for exploring disease mechanisms, drug screening, and personalized medicine. This review explores the construction and application of Organ-on-a-chip technology in bone and joint disease research, proposes a modular construction concept, and discusses the new opportunities and future challenges in the construction and application of bone and joint-on-chip platforms.
Collapse
Affiliation(s)
- Chengcheng Du
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Jiacheng Liu
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Senrui Liu
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Pengcheng Xiao
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Zhuolin Chen
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Hong Chen
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Wei Huang
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| | - Yiting Lei
- Department of Orthopedics, Orthopedic Laboratory of Chongqing Medical University, The First Affiliated Hospital of Chongqing Medical University, Chongqing, 400016, China
| |
Collapse
|
48
|
Walker M, Morton JP. Hydrogel models of pancreatic adenocarcinoma to study cell mechanosensing. Biophys Rev 2024; 16:851-870. [PMID: 39830124 PMCID: PMC11735828 DOI: 10.1007/s12551-024-01265-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/06/2024] [Indexed: 01/22/2025] Open
Abstract
Pancreatic adenocarcinoma (PDAC) is the predominant form of pancreatic cancer and one of the leading causes of cancer-related death worldwide, with an extremely poor prognosis after diagnosis. High mortality from PDAC arises partly due to late diagnosis resulting from a lack of early-stage biomarkers and due to chemotherapeutic drug resistance, which arises from a highly fibrotic stromal response known as desmoplasia. Desmoplasia alters tissue mechanics, which triggers changes in cell mechanosensing and leads to dysregulated transcriptional activity and disease phenotypes. Hydrogels are effective in vitro models to mimic mechanical changes in tissue mechanics during PDAC progression and to study the influence of these changes on mechanosensitive cell responses. Despite the complex biophysical changes that occur within the PDAC microenvironment, carefully designed hydrogels can very closely recapitulate these properties during PDAC progression. Hydrogels are relatively inexpensive, highly reproducible and can be designed in a humanised manner to increase their relevance for human PDAC studies. In vivo models have some limitations, including species-species differences, high variability, expense and legal/ethical considerations, which make hydrogel models a promising alternative. Here, we comprehensively review recent advancements in hydrogel bioengineering for developing our fundamental understanding of mechanobiology in PDAC, which is critical for informing advanced therapeutics.
Collapse
Affiliation(s)
- M Walker
- Centre for the Cellular Microenvironment, Advanced Research Centre, 11 Chapel Lane, James Watt School of Engineering, University of Glasgow, Glasgow, G11 6EW UK
| | - JP Morton
- Cancer Research UK Scotland Institute, Garscube Estate, Switchback Rd, Glasgow, G61 1BD UK
- School of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Rd, Glasgow, G61 1QH UK
| |
Collapse
|
49
|
Pfeifer LM, Sensbach J, Pipp F, Werkmann D, Hewitt P. Increasing sustainability and reproducibility of in vitro toxicology applications: serum-free cultivation of HepG2 cells. FRONTIERS IN TOXICOLOGY 2024; 6:1439031. [PMID: 39650261 PMCID: PMC11621109 DOI: 10.3389/ftox.2024.1439031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 10/30/2024] [Indexed: 12/11/2024] Open
Abstract
Fetal Bovine Serum (FBS) is an important ingredient in cell culture media and the current standard for most cells in vitro. However, the use of FBS is controversial for several reasons, including ethical concerns, political, and societal pressure, as well as scientific problems due to the undefined and variable nature of FBS. Nevertheless, scientists hesitate to change the paradigm without solid data de-risking the switch of their assays to alternatives. In this study, HepG2 cells, a human hepatoblastoma cell line commonly used to study drug hepatotoxicity, were adapted to serum-free conditions by using different commercially available media and FBS replacements. After transition to these new culture conditions, the success of adaptation was determined based on cell morphology and growth characteristics. Long-term culturing capacity for each medium was defined as the number of passages HepG2 cells could be cultured without any alterations in morphology or growth behavior. Two media (Advanced DMEM/F12 from ThermoFisher and TCM® Serum Replacement from MP Biomedicals) showed a long-term cultivation capacity comparable to media containing FBS and were selected for further analysis. Both media can be characterized as serum-free, however still contain animal-derived components: bovine serum albumin (both media) and bovine transferrin (only TCM® serum replacement). To assess the functionality of the cells cultivated in either of the two media, HepG2 cells were treated with reference compounds, specifically selected for their known hepatotoxicity characteristics in man. Different toxicological assays focusing on viability, mitochondrial toxicity, oxidative stress, and intracellular drug response were performed. Throughout the different assays, response to reference compounds was comparable, with a slightly higher sensitivity of serum-free cultivated HepG2 cells when assessing viability/cell death and a lower sensitivity towards oxidative stress. Taken together, the two selected media were shown to support growth, morphology, and function of serum-free cultivated HepG2 cells in the early preclinical safety space. Therefore, these results can serve as a starting point to further optimize culture conditions with the goal to remove any remaining animal-derived components.
Collapse
Affiliation(s)
| | - Janike Sensbach
- Early Investigative Toxicology, Merck Healthcare KGaA, Darmstadt, Germany
| | - Frederic Pipp
- Corporate Animal Affairs, Merck KGaA, Darmstadt, Germany
| | - Daniela Werkmann
- Cell Design Lab, Molecular Biology, Merck KGaA, Darmstadt, Germany
| | - Philip Hewitt
- Early Investigative Toxicology, Merck Healthcare KGaA, Darmstadt, Germany
| |
Collapse
|
50
|
Varlı HS, Akkurt Yıldırım M, Kızılbey K, Türkoğlu N. Gene Delivery via Octadecylamine-Based Nanoparticles for iPSC Generation from CCD1072-SK Fibroblast Cells. Curr Issues Mol Biol 2024; 46:12588-12607. [PMID: 39590341 PMCID: PMC11593313 DOI: 10.3390/cimb46110747] [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: 11/01/2024] [Accepted: 11/03/2024] [Indexed: 11/28/2024] Open
Abstract
This study presents a novel biotechnological approach using octadecylamine-based solid lipid nanoparticles (OCTNPs) for the first-time reprogramming of human CCD1072-SK fibroblast cells into induced pluripotent stem cells (iPSCs). OCTNPs, with an average size of 178.9 nm and a positive zeta potential of 22.8 mV, were synthesized, thoroughly characterized, and utilized as a non-viral vector to efficiently deliver reprogramming factors, achieving a remarkable transfection efficiency of 82.0%. iPSCs were characterized through immunofluorescence, flow cytometry, and RT-qPCR, confirming the expression of key pluripotency markers such as OCT4, SOX2, and KLF4, with alkaline phosphatase activity further validating their pluripotent state. Following this comprehensive characterization, the iPSCs were successfully differentiated into cardiomyocyte-like cells using 5-azacytidine. Our research highlights the innovative application of OCTNPs as a safe and effective alternative to viral vectors, addressing key limitations of iPSC reprogramming. The novel application of OCTNPs for efficient gene delivery demonstrates a powerful tool for advancing stem cell technologies, minimizing risks associated with viral vectors. These findings pave the way for further innovations in biotechnological applications, particularly in tissue engineering and personalized medicine.
Collapse
Affiliation(s)
- Hanife Sevgi Varlı
- Department of Molecular Biology and Genetics, Institute of Science and Technology, Yildiz Technical University, 34220 Istanbul, Türkiye; (H.S.V.); (M.A.Y.)
- Central Research Laboratory, Yildiz Technical University, 34220 Istanbul, Türkiye
| | - Meryem Akkurt Yıldırım
- Department of Molecular Biology and Genetics, Institute of Science and Technology, Yildiz Technical University, 34220 Istanbul, Türkiye; (H.S.V.); (M.A.Y.)
| | - Kadriye Kızılbey
- Basic Sciences, Faculty of Engineering and Natural Sciences, Acıbadem Mehmet Ali Aydınlar University, 34752 Istanbul, Türkiye
| | - Nelisa Türkoğlu
- Department of Molecular Biology and Genetics, Institute of Science and Technology, Yildiz Technical University, 34220 Istanbul, Türkiye; (H.S.V.); (M.A.Y.)
| |
Collapse
|