1
|
Cong R, Lu C, Li X, Xu Z, Wang Y, Sun S. Tumor organoids in cancer medicine: from model systems to natural compound screening. PHARMACEUTICAL BIOLOGY 2025; 63:89-109. [PMID: 39893515 PMCID: PMC11789228 DOI: 10.1080/13880209.2025.2458149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/04/2024] [Accepted: 01/17/2025] [Indexed: 02/04/2025]
Abstract
CONTEXT The advent of tissue engineering and biomedical techniques has significantly advanced the development of three-dimensional (3D) cell culture systems, particularly tumor organoids. These self-assembled 3D cell clusters closely replicate the histopathological, genetic, and phenotypic characteristics of primary tissues, making them invaluable tools in cancer research and drug screening. OBJECTIVE This review addresses the challenges in developing in vitro models that accurately reflect tumor heterogeneity and explores the application of tumor organoids in cancer research, with a specific focus on the screening of natural products for antitumor therapies. METHODS This review synthesizes information from major databases, including Chemical Abstracts, Medicinal and Aromatic Plants Abstracts, ScienceDirect, Google Scholar, Scopus, PubMed and Springer Link. Publications were selected without date restrictions, using terms such as 'organoid', 'natural product', 'pharmacological', 'extract', 'nanomaterial' and 'traditional uses'. Articles related to agriculture, ecology, synthetic work or published in languages other than English were excluded. RESULTS AND CONCLUSIONS The review identifies key challenges related to the efficiency and variability of organoid generation and discusses ongoing efforts to enhance their predictive capabilities in drug screening and personalized medicine. Recent studies utilizing patient-derived organoid models for natural compound screening are highlighted, demonstrating the potential of these models in developing new classes of anticancer agents. The integration of natural products with patient-derived organoid models presents a promising approach for discovering novel anticancer compounds and elucidating their mechanisms of action.
Collapse
Affiliation(s)
- Rong Cong
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Can Lu
- Department of Pathology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Xinying Li
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Zhijie Xu
- Department of Pathology, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yaqin Wang
- Clinical Research Center for Reproduction and Genetics in Hunan Province, Reproductive and Genetic Hospital of CITIC-Xiangya, Changsha, China
| | - Shusen Sun
- College of Pharmacy and Health Sciences, Western New England University, Springfield, MA, USA
| |
Collapse
|
2
|
Williams E, Hocking JS, Fairley CK, Chen MY, Williamson DA, McCarthy JS, Jamrozik E. Rationale and Ethical Assessment of an Oropharyngeal Gonorrhea Controlled Human Infection Model. J Infect Dis 2025; 231:841-848. [PMID: 39825788 DOI: 10.1093/infdis/jiaf029] [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: 10/30/2024] [Revised: 12/28/2024] [Accepted: 01/16/2025] [Indexed: 01/20/2025] Open
Abstract
Infection with Neisseria gonorrhoeae, the causative agent of gonorrhea, causes significant morbidity worldwide and can have long-term impacts on reproductive health. The greatest global burden of gonorrhea occurs in low- and middle-income settings. Global public health significance is increasing due to rising antimicrobial resistance, which threatens future gonorrhea management. The oropharynx is an important asymptomatic reservoir for gonorrhea transmission and a high-risk site for development of antimicrobial resistance and treatment failure. Controlled human infection model (CHIM) studies using N gonorrhoeae may provide a means to accelerate the development of urgently needed therapeutics, vaccines, and other biomedical prevention strategies. A gonorrhea urethritis CHIM has been used since the 1980s with no reported serious adverse events. Here, we describe the rationale for an oropharyngeal gonorrhea CHIM, including analysis of potential ethical issues that should inform the development of this novel study design.
Collapse
Affiliation(s)
- Eloise Williams
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity
- Victorian Infectious Diseases Reference Laboratory, Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity
| | - Jane S Hocking
- Melbourne School of Population and Global Health, The University of Melbourne
| | - Christopher K Fairley
- Melbourne Sexual Health Centre, Alfred Health
- School of Translational Medicine, Monash University, Melbourne, Australia
| | - Marcus Y Chen
- Melbourne Sexual Health Centre, Alfred Health
- School of Translational Medicine, Monash University, Melbourne, Australia
| | - Deborah A Williamson
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity
- Department of Medicine, University of St Andrews
- School of Infection and Immunity, Medical Research Council-University of Glasgow Centre for Virus Research, United Kingdom
| | - James S McCarthy
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity
- Division of Infection and Global Health, The Walter and Eliza Hall Institute of Medical Research
- Victorian Infectious Diseases Service, The Royal Melbourne Hospital at the Peter Doherty Institute for Infection and Immunity, Melbourne, Australia
| | - Euzebiusz Jamrozik
- Department of Infectious Diseases, The University of Melbourne at the Peter Doherty Institute for Infection and Immunity
- Nuffield Department of Population Health, The Ethox Centre and Wellcome Centre for Ethics and the Humanities, University of Oxford, United Kingdom
- Monash Bioethics Centre, Monash University, Clayton, Australia
| |
Collapse
|
3
|
Zhou L, Huang J, Li C, Gu Q, Li G, Li ZA, Xu J, Zhou J, Tuan RS. Organoids and organs-on-chips: Recent advances, applications in drug development, and regulatory challenges. MED 2025; 6:100667. [PMID: 40220744 DOI: 10.1016/j.medj.2025.100667] [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: 03/21/2024] [Revised: 08/30/2024] [Accepted: 03/12/2025] [Indexed: 04/14/2025]
Abstract
Organoids and organs-on-chips (OoCs) are rapidly evolving technologies for creating miniature human tissue models. They can mimic complex physiological functions and pathological conditions, offering more realistic platforms for disease modeling, drug screening, precision medicine, and regenerative therapies. The passing of the FDA Modernization Act 2.0 has reduced animal testing requirements for drug trials, marking a significant milestone in using advanced in vitro models such as organoids and OoCs for therapeutic discovery. Apart from technical and ethical challenges, regulatory issues persist in ensuring the reliability, scientificity, and applicability of these models in drug development. This perspective explores the concept, advancements, pros and cons, and applications of organoids and OoCs, particularly in drug research and development. It also examines global regulatory agencies' policies and actions on using these models in drug evaluation, aiming to guide industry standard setting and advance regulatory science.
Collapse
Affiliation(s)
- Liangbin Zhou
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China; Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science and Technology Park, Hong Kong SAR, China; Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Innovative Orthopaedic Biomaterials and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Jingjing Huang
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Cun Li
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Qi Gu
- Key Laboratory of Organ Regeneration and Reconstruction, Beijing Institute for Stem Cell and Regenerative Medicine, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Gang Li
- Department of Otolaryngology-Head and Neck Surgery, Nanfang Hospital of the Southern Medical University, Guangzhou, China
| | - Zhong Alan Li
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China; Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science and Technology Park, Hong Kong SAR, China; Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Jiankun Xu
- Musculoskeletal Research Laboratory, Department of Orthopaedics and Traumatology, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; Innovative Orthopaedic Biomaterials and Drug Translational Research Laboratory, Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Jie Zhou
- Department of Microbiology, School of Clinical Medicine, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China.
| | - Rocky S Tuan
- Department of Biomedical Engineering, Faculty of Engineering, The Chinese University of Hong Kong, Hong Kong SAR, China; Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science and Technology Park, Hong Kong SAR, China; Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China; School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
4
|
Miedel MT, Varmazyad M, Xia M, Brooks MM, Gavlock DC, Reese C, Behari J, Soto-Gutierrez A, Gough A, Taylor DL, Schurdak ME. Validation of microphysiological systems for interpreting patient heterogeneity requires robust reproducibility analytics and experimental metadata. CELL REPORTS METHODS 2025:101028. [PMID: 40233763 DOI: 10.1016/j.crmeth.2025.101028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Revised: 12/24/2024] [Accepted: 03/20/2025] [Indexed: 04/17/2025]
Abstract
Multi-cell-type, 3D microphysiological systems (MPS) that recapitulate normal organ/organ system functions and the progression of diseases are being applied in drug discovery and development programs to enable precision medicine. A critical step for this application is to demonstrate the reproducibility of the MPS and its ability to identify biologic/clinical heterogeneity from experimental variability, which requires capturing detailed metadata associated with MPS studies as well as a strong analytical approach for assessing reproducibility. Detailed metadata ensure that identical study parameters are being compared when evaluating reproducibility. We have developed the Pittsburgh reproducibility protocol (PReP), which uses a set of common statistical metrics, the coefficient of variation (CV), ANOVA, and intraclass correlation coefficient (ICC), in a pipeline as a standard approach to evaluate the intra- and interstudy reproducibility of MPS performance. The PReP can be employed to identify biological/clinical heterogeneity relevant to precision medicine.
Collapse
Affiliation(s)
- Mark T Miedel
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA; Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Mahboubeh Varmazyad
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Mengying Xia
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Maria Mori Brooks
- Department of Epidemiology, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA; Department of Biostatistics, University of Pittsburgh School of Public Health, Pittsburgh, PA, USA
| | - Dillon C Gavlock
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Celeste Reese
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Jaideep Behari
- Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA 15261, USA; Division of Gastroenterology, Hepatology and Nutrition, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - Alejandro Soto-Gutierrez
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261, USA; Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA 15261, USA; Center for Transcriptional Medicine, Pittsburgh, PA 15261, USA
| | - Albert Gough
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA; Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | - D Lansing Taylor
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA; Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Computational and System Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Mark E Schurdak
- Drug Discovery Institute, University of Pittsburgh, Pittsburgh, PA 15261, USA; Pittsburgh Liver Research Center, University of Pittsburgh, Pittsburgh, PA 15261, USA; Department of Computational and System Biology, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15260, USA.
| |
Collapse
|
5
|
Korbmacher F, Bernabeu M. Induced pluripotent stem cell-based tissue models to study malaria: a new player in the research game. Curr Opin Microbiol 2025; 84:102585. [PMID: 40010012 DOI: 10.1016/j.mib.2025.102585] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 01/27/2025] [Accepted: 02/07/2025] [Indexed: 02/28/2025]
Abstract
Most in vitro studies on parasite development and pathogenesis in the human host have been conducted using traditional primary or immortalized cells, despite their inherent limitations. Breakthroughs in the field of induced pluripotent stem cells (iPSCs) are revolutionizing disease modeling, offering alternatives to traditional in vivo and in vitro infection models. Human iPSCs differentiate into all cell types, proliferate indefinitely, and offer experimental advantages, like genome editing and donor control. iPSCs can be engineered into complex 3D tissue models that closely mimic morphology and function of their in vivo counterparts and allow for precise experimental manipulation. The physiological complexity of iPSC-based tissue models has improved rapidly. Given Plasmodium's systemic impact across multiple organs, these models provide an invaluable resource for studying parasite-tissue interactions. This opinion article focuses on recent developments of iPSC-based models for Plasmodium research. We describe the main highlights and potential use of these systems while acknowledging current limitations.
Collapse
Affiliation(s)
- François Korbmacher
- European Molecular Biology Laboratory (EMBL) Barcelona, Carrer del Doctor Aiguader 88, 08003 Barcelona, Spain
| | - Maria Bernabeu
- European Molecular Biology Laboratory (EMBL) Barcelona, Carrer del Doctor Aiguader 88, 08003 Barcelona, Spain.
| |
Collapse
|
6
|
Kainz MP, Polz M, Ziesel D, Nowakowska M, Üçal M, Kienesberger S, Hasiba-Pappas S, Winter R, Tabrizi-Wizsy NG, Kager S, Rienmüller T, Fuchs J, Terzano M, Baumgartner C, Holzapfel GA. Biointegration of soft tissue-inspired hydrogels on the chorioallantoic membrane: An experimental characterization. Mater Today Bio 2025; 31:101508. [PMID: 39990742 PMCID: PMC11846936 DOI: 10.1016/j.mtbio.2025.101508] [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: 10/21/2024] [Revised: 01/08/2025] [Accepted: 01/19/2025] [Indexed: 02/25/2025] Open
Abstract
Soft scaffold materials for cell cultures grafted onto the chorioallantoic membrane (CAM) provide innovative solutions for creating physiologically relevant environments by mimicking the host tissue. Biocompatible hydrogels represent an ideal medium for such applications, but the relationship between scaffold mechanical properties and reactions at the biological interface remains poorly understood. This study examines the attachment and integration of soft hydrogels on the CAM using an accessible ex ovo system. Composite hydrogels of polyvinyl alcohol and Phytagel were fabricated by sterile freeze-thawing. CAM assays, as an alternative to traditional in vivo models, enabled the evaluation of the compatibility, attachment, and biointegration of hydrogels with three distinct compositions. The mechanomimetic properties of the hydrogels were assessed through cyclic compression-tension tests, with nominal peak stresses ranging from 0 . 26 to 2 . 82 kPa in tension and - 0 . 33 to - 2 . 92 kPa in compression. Mechanical attachment to the CAM was measured by pull-off tests after five days of incubation. On the first day, the interface strength was similar for all hydrogel compositions. On day 5 , softer hydrogels showed the greatest increase ( p = 0 . 008 ), followed by intermediate hydrogels ( p = 0 . 020 ), while the denser hydrogels showed negligible changes ( p = 0 . 073 ). Histological analyses revealed cell infiltration in 100 % of soft, 75 % of intermediate, and 13 % of dense hydrogels, suggesting that softer hydrogels integrate better into the CAM by facilitating cell migration and enhancing interface strength. Chicken embryo survival rates and cytotoxicity assays confirmed the biocompatibility of the hydrogels and supported their potential for use in soft, hydrated three-dimensional scaffolds that mimic tissue environments in dynamic biological systems. Statement of significance Current research on soft scaffold materials for cell cultures often overlooks the critical relationship between mechanical properties and biological integration of these materials with host tissues. Although hydrogels, as soft porous materials, hold promise for creating physiologically relevant environments, the mechanisms driving their attachment and biointegration, especially on the chorioallantoic membrane (CAM), remain largely unexplored. This study addresses this gap by investigating the interaction between soft hydrogels and the CAM, providing valuable insights into how material properties and microstructure influence cellular responses. Our findings emphasize the importance of understanding these dynamics to develop biocompatible scaffolds that better mimic tissue environments, advancing applications in three-dimensional cell cultures on CAM assays and other biological systems.
Collapse
Affiliation(s)
- Manuel P. Kainz
- Institute of Biomechanics, Graz University of Technology, Austria
| | - Mathias Polz
- Institute of Health Care Engineering with European Testing Center of Medical Devices, Graz University of Technology, Austria
| | - Daniel Ziesel
- Institute of Health Care Engineering with European Testing Center of Medical Devices, Graz University of Technology, Austria
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Division of Medical Physics and Biophysics, Medical University of Graz, Austria
| | - Marta Nowakowska
- Department of Neurosurgery, Medical University of Graz, Austria
- BioTechMed-Graz, Austria
| | - Muammer Üçal
- Department of Neurosurgery, Medical University of Graz, Austria
- BioTechMed-Graz, Austria
- Department of Neurology, Medical University of Graz, Austria
| | - Sabine Kienesberger
- BioTechMed-Graz, Austria
- Institute of Molecular Biosciences, University of Graz, Austria
| | - Sophie Hasiba-Pappas
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, Austria
| | - Raimund Winter
- Research Unit for Tissue Regeneration, Repair and Reconstruction, Division of Plastic, Aesthetic and Reconstructive Surgery, Department of Surgery, Medical University of Graz, Austria
| | | | - Sarah Kager
- Institute of Health Care Engineering with European Testing Center of Medical Devices, Graz University of Technology, Austria
- Division of Immunology, Research Unit CAM Lab, Otto Loewi Research Center, Medical University of Graz, Austria
| | - Theresa Rienmüller
- Institute of Health Care Engineering with European Testing Center of Medical Devices, Graz University of Technology, Austria
- BioTechMed-Graz, Austria
| | - Julia Fuchs
- Institute of Health Care Engineering with European Testing Center of Medical Devices, Graz University of Technology, Austria
- Division of Cell Biology, Histology and Embryology, Gottfried Schatz Research Center, Medical University of Graz, Austria
| | - Michele Terzano
- Institute of Biomechanics, Graz University of Technology, Austria
| | - Christian Baumgartner
- Institute of Health Care Engineering with European Testing Center of Medical Devices, Graz University of Technology, Austria
| | - Gerhard A. Holzapfel
- Institute of Biomechanics, Graz University of Technology, Austria
- Department of Structural Engineering, NTNU, Trondheim, Norway
| |
Collapse
|
7
|
Xia X, Hu M, Zhou W, Jin Y, Yao X. Engineering cardiology with miniature hearts. Mater Today Bio 2025; 31:101505. [PMID: 39911371 PMCID: PMC11795835 DOI: 10.1016/j.mtbio.2025.101505] [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: 11/07/2024] [Revised: 12/28/2024] [Accepted: 01/18/2025] [Indexed: 02/07/2025] Open
Abstract
Cardiac organoids offer sophisticated 3D structures that emulate key aspects of human heart development and function. This review traces the evolution of cardiac organoid technology, from early stem cell differentiation protocols to advanced bioengineering approaches. We discuss the methodologies for creating cardiac organoids, including self-organization techniques, biomaterial-based scaffolds, 3D bioprinting, and organ-on-chip platforms, which have significantly enhanced the structural complexity and physiological relevance of in vitro cardiac models. We examine their applications in fundamental research and medical innovations, highlighting their potential to transform our understanding of cardiac biology and pathology. The integration of multiple cell types, vascularization strategies, and maturation protocols has led to more faithful representations of the adult human heart. However, challenges remain in achieving full functional maturity and scalability. We critically assess the current limitations and outline future directions for advancing cardiac organoid technology. By providing a comprehensive analysis of the field, this review aims to catalyze further innovation in cardiac tissue engineering and facilitate its translation to clinical applications.
Collapse
Affiliation(s)
- Xiaojun Xia
- Department of Cardiology, Center of Regenerative and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Miner Hu
- Department of Cardiology, Children's Hospital, Zhejiang University School of Medicine, National Clinical Research Center for Child Health, Hangzhou, 310000, China
| | - Wenyan Zhou
- School of Medicine, Taizhou University, Taizhou, Zhejiang, 318000, China
| | - Yunpeng Jin
- Department of Cardiology, Center of Regenerative and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
| | - Xudong Yao
- Department of Cardiology, Center of Regenerative and Aging Medicine, The Fourth Affiliated Hospital of School of Medicine, and International School of Medicine, International Institutes of Medicine, Zhejiang University, Yiwu, 322000, China
- Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Zhejiang University School of Medicine, Hangzhou, 310058, China
| |
Collapse
|
8
|
Nguyen NTB, Gevers S, Kok RNU, Burgering LM, Neikes H, Akkerman N, Betjes MA, Ludikhuize MC, Gulersonmez C, Stigter ECA, Vercoulen Y, Drost J, Clevers H, Vermeulen M, van Zon JS, Tans SJ, Burgering BMT, Rodríguez Colman MJ. Lactate controls cancer stemness and plasticity through epigenetic regulation. Cell Metab 2025; 37:903-919.e10. [PMID: 39933514 DOI: 10.1016/j.cmet.2025.01.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 11/04/2024] [Accepted: 01/03/2025] [Indexed: 02/13/2025]
Abstract
Tumors arise from uncontrolled cell proliferation driven by mutations in genes that regulate stem cell renewal and differentiation. Intestinal tumors, however, retain some hierarchical organization, maintaining both cancer stem cells (CSCs) and cancer differentiated cells (CDCs). This heterogeneity, coupled with cellular plasticity enabling CDCs to revert to CSCs, contributes to therapy resistance and relapse. Using genetically encoded fluorescent reporters in human tumor organoids, combined with our machine-learning-based cell tracker, CellPhenTracker, we simultaneously traced cell-type specification, metabolic changes, and reconstructed cell lineage trajectories during tumor organoid development. Our findings reveal distinctive metabolic phenotypes in CSCs and CDCs. We find that lactate regulates tumor dynamics, suppressing CSC differentiation and inducing dedifferentiation into a proliferative CSC state. Mechanistically, lactate increases histone acetylation, epigenetically activating MYC. Given that lactate's regulation of MYC depends on the bromodomain-containing protein 4 (BRD4), targeting cancer metabolism and BRD4 inhibitors emerge as a promising strategy to prevent tumor relapse.
Collapse
Affiliation(s)
- Nguyen T B Nguyen
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CG Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Sira Gevers
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CG Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Rutger N U Kok
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CG Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Lotte M Burgering
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CG Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Hannah Neikes
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, Nijmegen 6525 GA, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Ninouk Akkerman
- Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, University Medical Center Utrecht, Utrecht, the Netherlands
| | | | - Marlies C Ludikhuize
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CG Utrecht, the Netherlands
| | - Can Gulersonmez
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CG Utrecht, the Netherlands
| | - Edwin C A Stigter
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CG Utrecht, the Netherlands
| | - Yvonne Vercoulen
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CG Utrecht, the Netherlands
| | - Jarno Drost
- Oncode Institute, Utrecht, the Netherlands; Princess Máxima Center for Pediatric Oncology, Heidelberglaan 25, 3584 CS Utrecht, the Netherlands
| | - Hans Clevers
- Oncode Institute, Utrecht, the Netherlands; Hubrecht Institute, Royal Netherlands Academy of Arts and Sciences, University Medical Center Utrecht, Utrecht, the Netherlands
| | - Michiel Vermeulen
- Department of Molecular Biology, Faculty of Science, Radboud Institute for Molecular Life Sciences, Oncode Institute, Radboud University Nijmegen, Nijmegen 6525 GA, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | | | - Sander J Tans
- AMOLF, Amsterdam, the Netherlands; Bionanoscience Department, Kavli Institute of Nanoscience Delft, Delft University of Technology, Delft, the Netherlands
| | - Boudewijn M T Burgering
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CG Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands
| | - Maria J Rodríguez Colman
- Molecular Cancer Research, Center for Molecular Medicine, University Medical Center Utrecht, Heidelberglaan 100, 3584 CG Utrecht, the Netherlands; Oncode Institute, Utrecht, the Netherlands.
| |
Collapse
|
9
|
Mukhare R, Gandhi KA, Kadam A, Raja A, Singh A, Madhav M, Chaubal R, Pandey S, Gupta S. Integration of Organoids With CRISPR Screens: A Narrative Review. Biol Cell 2025; 117:e70006. [PMID: 40223602 PMCID: PMC11995251 DOI: 10.1111/boc.70006] [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: 03/05/2025] [Revised: 03/05/2025] [Accepted: 03/18/2025] [Indexed: 04/15/2025]
Abstract
Organoids represent a significant advancement in disease modeling, demonstrated by their capacity to mimic the physiological/pathological structure and functional characteristics of the native tissue. Recently CRISPR/Cas9 technology has emerged as a powerful tool in combination with organoids for the development of novel therapies in preclinical settings. This review explores the current literature on applications of pooled CRISPR screening in organoids and the emerging role of these models in understanding cancer. We highlight the evolution of genome-wide CRISPR gRNA library screens in organoids, noting their increasing adoption in the field over the past decade. Noteworthy studies utilizing these screens to investigate oncogenic vulnerabilities and developmental pathways in various organoid systems are discussed. Despite the promise organoids hold, challenges such as standardization, reproducibility, and the complexity of data interpretation remain. The review also addresses the ideas of assessing tumor organoids (tumoroids) against established cancer hallmarks and the potential of studying intercellular cooperation within these models. Ultimately, we propose that organoids, particularly when personalized for patient-specific applications, could revolutionize drug screening and therapeutic approaches, minimizing the reliance on traditional animal models and enhancing the precision of clinical interventions.
Collapse
Affiliation(s)
- Rushikesh Mukhare
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in CancerTata Memorial CentreNavi MumbaiMaharashtraIndia
- Training School ComplexHomi Bhabha National InstituteMumbaiMaharashtraIndia
- Department of Medical OncologyTata Memorial Hospital, Tata Memorial CentreMumbaiMaharashtraIndia
| | - Khushboo A. Gandhi
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in CancerTata Memorial CentreNavi MumbaiMaharashtraIndia
| | - Anushree Kadam
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in CancerTata Memorial CentreNavi MumbaiMaharashtraIndia
| | - Aishwarya Raja
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in CancerTata Memorial CentreNavi MumbaiMaharashtraIndia
- Training School ComplexHomi Bhabha National InstituteMumbaiMaharashtraIndia
- Department of Medical OncologyTata Memorial Hospital, Tata Memorial CentreMumbaiMaharashtraIndia
| | - Ankita Singh
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in CancerTata Memorial CentreNavi MumbaiMaharashtraIndia
| | - Mrudula Madhav
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in CancerTata Memorial CentreNavi MumbaiMaharashtraIndia
| | - Rohan Chaubal
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in CancerTata Memorial CentreNavi MumbaiMaharashtraIndia
- Training School ComplexHomi Bhabha National InstituteMumbaiMaharashtraIndia
- Department of Surgical OncologyTata Memorial Hospital, Tata Memorial CentreMumbaiMaharashtraIndia
| | - Shwetali Pandey
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in CancerTata Memorial CentreNavi MumbaiMaharashtraIndia
| | - Sudeep Gupta
- Clinical Genomics and Hypoxia Lab (Clinician Scientist Laboratory), Advanced Centre for Treatment, Research, and Education in CancerTata Memorial CentreNavi MumbaiMaharashtraIndia
- Department of Medical OncologyTata Memorial Hospital, Tata Memorial CentreMumbaiMaharashtraIndia
| |
Collapse
|
10
|
Li J, Isaakidou A, van Zanten LJ, Tas RP, Mirzaali MJ, Fratila-Apachitei LE, Zadpoor AA. Multi-scale additive manufacturing of 3D porous networks integrated with hydrogel for sustained in vitro tissue growth. Acta Biomater 2025; 196:198-212. [PMID: 40049309 DOI: 10.1016/j.actbio.2025.03.005] [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: 10/31/2024] [Revised: 02/06/2025] [Accepted: 03/03/2025] [Indexed: 03/15/2025]
Abstract
The development of high-fidelity three-dimensional (3D) tissue models can minimize the need for animal models in clinical medicine and drug development. However, physical limitations regarding the distances within which diffusion processes are effective impose limitations on the size of such constructs. That is because larger-size constructs experience necrosis, especially in their centers, due to the cells residing deep inside such constructs not receiving enough oxygen and nutrients. This hampers the sustained in vitro growth of the tissues which is required for achieving functional microtissues. To address this challenge, we used three types of 3D printing technologies to create perfusable networks at different length scales and integrate them into such constructs. Toward this aim, networks incorporating porous conduits with increasingly complex configurations were designed and fabricated using fused deposition modeling, stereolithography, and two-photon polymerization while optimizing the printing conditions for each of these technologies. Furthermore, following network embedding in hydrogels, contrast agent-enhanced micro-computed tomography and confocal fluorescence microscopy were employed to characterize one of the essential network functionalities, namely the diffusion function. The investigations revealed the effects of various design parameters on the diffusion behavior of the porous conduits over 24 h. We found that the number of pores exerts the most significant influence on the diffusion behavior of the contrast agent, followed by variations in the pore size and hydrogel concentration. The analytical approach and the findings of this study establish a solid base for a new technological platform to fabricate perfusable multiscale 3D porous networks with complex designs while enabling the customization of diffusion characteristics to meet specific requirements for sustained in vitro tissue growth. STATEMENT OF SIGNIFICANCE: This study addresses an essential limitation of current 3D tissue engineering, namely, sustaining tissue viability in larger constructs through optimized nutrient and oxygen delivery. By utilizing advanced 3D printing techniques this research proposes the fabrication of perfusable, multiscale and customizable networks that enhance diffusion and enable cell access to essential nutrients throughout the construct. The findings highlighted the role of network characteristics on the diffusion of a model compound within a hydrogel matrix. This work represents a promising technological platform for creating advanced in vitro 3D tissue models that can reduce the use of animal models in research involving tissue regeneration, disease models and drug development.
Collapse
Affiliation(s)
- J Li
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft 2628 CD, The Netherlands.
| | - A Isaakidou
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft 2628 CD, The Netherlands
| | - L J van Zanten
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft 2628 CD, The Netherlands
| | - R P Tas
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft 2628 CD, The Netherlands
| | - M J Mirzaali
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft 2628 CD, The Netherlands
| | - L E Fratila-Apachitei
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft 2628 CD, The Netherlands.
| | - A A Zadpoor
- Department of Biomechanical Engineering, Faculty of Mechanical Engineering, Delft University of Technology (TU Delft), Mekelweg 2, Delft 2628 CD, The Netherlands
| |
Collapse
|
11
|
Deltourbe LG, Durand M, Costas A, Ingersoll MA. A bladder blueprint to build better models for understanding homeostasis and disease. Nat Rev Urol 2025:10.1038/s41585-025-01013-x. [PMID: 40140722 DOI: 10.1038/s41585-025-01013-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/24/2025] [Indexed: 03/28/2025]
Abstract
The bladder is a complex organ that can be affected by various pathologies, such as cancer or infection. It has a specific tissue structure composed of many different cell types and layers, including urothelial and endothelial cells but also a muscle layer controlling stretch and contraction to void urine. The bladder has constitutive and induced immune responses to infection or damage and harbours a microbiome. Each of these features can be influenced by factors including age and biological sex, which makes modelling homeostasis and disease in the bladder complex and challenging. To model diseases that affect the bladder, mouse models are an invaluable tool to understand the bladder in situ. However, stark differences exist between mice and humans, and so mouse models of human disease have limitations. Thus, models that more closely approximate human physiology would be expected to contribute to improved understanding of bladder biology. As technology advances, improvements in model development and creation of 3D bladder structures are enabling scientists to recapitulate essential aspects of human bladder physiology to gain increased understanding of bladder homeostasis and diseases.
Collapse
Affiliation(s)
- Léa G Deltourbe
- Mucosal Inflammation and Immunity Team, Université Paris Cité, CNRS, Inserm, Institut Cochin and Department of Immunology, Institut Pasteur, Paris, France
| | - Méline Durand
- Mucosal Inflammation and Immunity Team, Université Paris Cité, CNRS, Inserm, Institut Cochin and Department of Immunology, Institut Pasteur, Paris, France
| | - Ariana Costas
- Mucosal Inflammation and Immunity Team, Université Paris Cité, CNRS, Inserm, Institut Cochin and Department of Immunology, Institut Pasteur, Paris, France
- Australian Institute for Microbiology and Infection, University of Technology Sydney, ULTIMO, Sydney, Australia
| | - Molly A Ingersoll
- Mucosal Inflammation and Immunity Team, Université Paris Cité, CNRS, Inserm, Institut Cochin and Department of Immunology, Institut Pasteur, Paris, France.
| |
Collapse
|
12
|
Lin Z, Wang W, Liu R, Li Q, Lee J, Hirschler C, Liu J. Cyborg organoids integrated with stretchable nanoelectronics can be functionally mapped during development. Nat Protoc 2025:10.1038/s41596-025-01147-7. [PMID: 40140634 DOI: 10.1038/s41596-025-01147-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2024] [Accepted: 12/31/2024] [Indexed: 03/28/2025]
Abstract
Organoids are in vitro miniaturized cellular models of organs that offer opportunities for studying organ development, disease mechanisms and drug screening. Understanding the complex processes governing organoid development and function requires methods suitable for the continuous, long-term monitoring of cell activities (for example, electrophysiological and mechanical activity) at single-cell resolution throughout the entire three-dimensional (3D) structure. Cyborg organoid technology addresses this need by seamlessly integrating stretchable mesh nanoelectronics with tissue-like properties, such as tissue-level flexibility, subcellular feature size and mesh-like networks, into 3D organoids through a 2D-to-3D tissue reconfiguration process during organogenesis. This approach enables longitudinal, tissue-wide, single-cell functional mapping, thereby overcoming the limitations of existing techniques including recording duration, spatial coverage, and the ability to maintain stable contact with the tissue during organoid development. This protocol describes the fabrication and characterization of stretchable mesh nanoelectronics, their electrical performance, their integration with organoids and the acquisition of long-term functional organoid activity requiring multimodal data analysis techniques. Cyborg organoid technology represents a transformative tool for investigating organoid development and function, with potential for improving in vitro disease models, drug screening and personalized medicine. The procedure is suitable for users within a multidisciplinary team with expertise in stem cell biology, tissue engineering, nanoelectronics fabrication, electrophysiology and data science.
Collapse
Affiliation(s)
- Zuwan Lin
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Wenbo Wang
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Ren Liu
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
| | - Qiang Li
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
| | - Jaeyong Lee
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
| | - Charles Hirschler
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA
| | - Jia Liu
- John A. Paulson School of Engineering and Applied Sciences, Harvard University, Boston, MA, USA.
| |
Collapse
|
13
|
Alnasser SM. From gut to liver: organoids as platforms for next-generation toxicology assessment vehicles for xenobiotics. Stem Cell Res Ther 2025; 16:150. [PMID: 40140938 PMCID: PMC11948905 DOI: 10.1186/s13287-025-04264-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2024] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
Traditional toxicological assessment relied heavily on 2D cell cultures and animal models of study, which were inadequate for the precise prediction of human response to chemicals. Researchers have now shifted focus on organoids for toxicological assessment. Organoids are 3D structures produced from stem cells that mimic the shape and functionality of human organs and have a number of advantages compared to traditional models of study. They have the capacity to replicate the intricate cellular microenvironment and in vivo interactions. They offer a physiologically pertinent platform that is useful for the researchers to monitor cellular responses in a more realistic manner and evaluate drug toxicity. Additionally, organoids can be created from cells unique to a patient, allowing for individualized toxicological research and providing understanding of the inter-individual heterogeneity in drug responses. Recent developments in the use of gut and liver organoids for assessment of the xenobiotics (environmental toxins and drugs) is reviewed in this article. Gut organoids can reveal potential damage to the digestive system and how xenobiotics affect nutrient absorption and barrier function. Liver is the primary site of detoxification and metabolism of xenobiotics, usually routed from the gut. Hence, these are linked and crucial for evaluating chemical or pollutant induced organ toxicity, forecasting their metabolism and pharmacokinetics. When incorporated into the drug development process, organoid models have the potential to improve the accuracy and efficiency of drug safety assessments, leading to safer and more effective treatments. We also discuss the limitations of using organoid-based toxicological assays, and future prospects, including the need for standardized protocols for overcoming reproducibility issues.
Collapse
Affiliation(s)
- Sulaiman Mohammed Alnasser
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, 51452, Buraydah, Qassim, Saudi Arabia.
| |
Collapse
|
14
|
Zhu M, Zhang H, Zhou Q, Sheng S, Gao Q, Geng Z, Chen X, Lai Y, Jing Y, Xu K, Bai L, Wang G, Wang J, Jiang Y, Su J. Dynamic GelMA/DNA Dual-Network Hydrogels Promote Woven Bone Organoid Formation and Enhance Bone Regeneration. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025:e2501254. [PMID: 40123197 DOI: 10.1002/adma.202501254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/28/2025] [Indexed: 03/25/2025]
Abstract
Bone organoids, in vitro models mimicking native bone structure and function, rely on 3D stem cell culture for self-organization, differentiation, ECM secretion, and biomineralization, ultimately forming mineralized collagen hierarchies. However, their development is often limited by the lack of suitable matrices with optimal mechanical properties for sustained cell growth and differentiation. To address this, a dynamic DNA/Gelatin methacryloyl (GelMA) hydrogel (CGDE) is developed to recapitulate key biochemical and mechanical features of the bone ECM, providing a supportive microenvironment for bone organoid formation. This dual-network hydrogel is engineered through hydrogen bonding between DNA and GelMA, combined with GelMA network crosslinking, resulting in appropriate mechanical strength and enhanced viscoelasticity. During a 21-day 3D culture, the CGDE hydrogel facilitates cellular migration and self-organization, promoting woven bone organoid (WBO) formation via intramembranous ossification. These WBOs exhibit spatiotemporal architectures supporting dynamic mineralization and tissue remodeling. In vivo studies demonstrate that CGDE-derived WBOs exhibit self-adaptive properties, enabling rapid osseointegration within 4 weeks. This work highlights the CGDE hydrogel as a robust and scalable platform for bone organoid development, offering new insights into bone biology and innovative strategies for bone tissue regeneration.
Collapse
Affiliation(s)
- Mengru Zhu
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, P. R. China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Hao Zhang
- Department of Orthopedics, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
| | - Qirong Zhou
- Department of Orthopedics, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
| | - Shihao Sheng
- Department of Orthopedics, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
| | - Qianmin Gao
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, P. R. China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Zhen Geng
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, P. R. China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Xiao Chen
- Department of Orthopedics, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
| | - Yuxiao Lai
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, P. R. China
| | - Yingying Jing
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, P. R. China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Ke Xu
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, P. R. China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Long Bai
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, P. R. China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Guangchao Wang
- Department of Orthopedics, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
| | - Jianhua Wang
- Department of Orthopedics, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
| | - Yingying Jiang
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, P. R. China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Jiacan Su
- Organoid Research Center, Institute of Translational Medicine, Shanghai University, Shanghai, 200444, P. R. China
- National Center for Translational Medicine (Shanghai) SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
- Department of Orthopedics, Xinhua Hospital affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
| |
Collapse
|
15
|
Kirsche L, He J, Müller A, Leary P. MARMOT: A multifaceted R pipeline for analysing spectral flow cytometry data from subcutaneously growing murine gastric organoids. J Immunol Methods 2025; 540:113854. [PMID: 40122453 DOI: 10.1016/j.jim.2025.113854] [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: 12/17/2024] [Revised: 03/10/2025] [Accepted: 03/20/2025] [Indexed: 03/25/2025]
Abstract
The analysis of murine immune cell types is a critical component of immunological research, necessitating precise and reproducible methodologies. Here, we present a comprehensive protocol and pipeline designed to streamline the process from murine gastric organoid transplant sample preparation to figure generation. This pipeline includes a detailed staining panel tailored for murine immune cells, ensuring accurate and comprehensive identification of various cell types. Additionally, it integrates an R-based analysis script (MARMOT Pipeline), encompassing data processing and visualisation. A key feature of this pipeline is its ability to produce publication-quality figures with minimal direct R coding, thus making advanced data analysis accessible to researchers with limited programming experience. Additionally, figures can be customised using a provided Shiny application. This approach both enhances the efficiency of data analysis and enables the reproducibility required for high-quality scientific research.
Collapse
Affiliation(s)
- Lydia Kirsche
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Jiazhuo He
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland
| | - Anne Müller
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland; Comprehensive Cancer Center Zürich, Zürich, Switzerland
| | - Peter Leary
- Institute of Molecular Cancer Research, University of Zürich, Zürich, Switzerland; Functional Genomics Center Zürich, University of Zürich/ETHZ, Zürich, Switzerland.
| |
Collapse
|
16
|
Nityanandam A, Patton MH, Bayazitov IT, Newman KD, Thomas KT, Zakharenko SS. Protocol for generating human assembloids to investigate thalamocortical and corticothalamic synaptic transmission and plasticity. STAR Protoc 2025; 6:103630. [PMID: 39921865 PMCID: PMC11850219 DOI: 10.1016/j.xpro.2025.103630] [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: 10/28/2024] [Revised: 12/05/2024] [Accepted: 01/15/2025] [Indexed: 02/10/2025] Open
Abstract
Human induced pluripotent stem cells (hiPSCs) can be used to generate assembloids that recreate thalamocortical circuitry displaying short-term and long-term synaptic plasticity. Here, we describe a protocol for differentiating hiPSCs into thalamic and cortical organoids and then fusing them to generate thalamocortical assembloids. We detail the steps for using whole-cell patch-clamp electrophysiology to investigate the properties of synaptic transmission and synaptic plasticity in this model system. For complete details on the use and execution of this protocol, please refer to Patton et al.1.
Collapse
Affiliation(s)
- Anjana Nityanandam
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| | - Mary H Patton
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Ildar T Bayazitov
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Kyle D Newman
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Kristen T Thomas
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA
| | - Stanislav S Zakharenko
- Department of Developmental Neurobiology, St. Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
17
|
Taglieri M, Di Gregorio L, Matis S, Uras CRM, Ardy M, Casati S, Marchese M, Poggi A, Raffaghello L, Benelli R. Colorectal Organoids: Models, Imaging, Omics, Therapy, Immunology, and Ethics. Cells 2025; 14:457. [PMID: 40136707 PMCID: PMC11941511 DOI: 10.3390/cells14060457] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/06/2025] [Accepted: 03/12/2025] [Indexed: 03/27/2025] Open
Abstract
Colorectal epithelium was the first long-term 3D organoid culture established in vitro. Identification of the key components essential for the long-term survival of the stem cell niche allowed an indefinite propagation of these cultures and the modulation of their differentiation into various lineages of mature intestinal epithelial cells. While these methods were eventually adapted to establish organoids from different organs, colorectal organoids remain a pioneering model for the development of new applications in health and disease. Several basic and applicative aspects of organoid culture, modeling, monitoring and testing are analyzed in this review. We also tackle the ethical problems of biobanking and distribution of these precious research tools, frequently confined in the laboratory of origin or condemned to destruction at the end of the project.
Collapse
Affiliation(s)
- Martina Taglieri
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.T.); (L.D.G.); (S.M.); (C.R.M.U.); (M.A.); (M.M.); (A.P.); (L.R.)
| | - Linda Di Gregorio
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.T.); (L.D.G.); (S.M.); (C.R.M.U.); (M.A.); (M.M.); (A.P.); (L.R.)
| | - Serena Matis
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.T.); (L.D.G.); (S.M.); (C.R.M.U.); (M.A.); (M.M.); (A.P.); (L.R.)
| | - Chiara Rosa Maria Uras
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.T.); (L.D.G.); (S.M.); (C.R.M.U.); (M.A.); (M.M.); (A.P.); (L.R.)
| | - Massimo Ardy
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.T.); (L.D.G.); (S.M.); (C.R.M.U.); (M.A.); (M.M.); (A.P.); (L.R.)
| | - Sara Casati
- Istituto per l’Endocrinologia e l’Oncologia Sperimentale “Gaetano Salvatore” CNR, 80131 Naples, Italy;
- Common Service ELSI, BBMRI.it (UNIMIB National Node Headquarter), 20126 Milan, Italy
| | - Monica Marchese
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.T.); (L.D.G.); (S.M.); (C.R.M.U.); (M.A.); (M.M.); (A.P.); (L.R.)
| | - Alessandro Poggi
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.T.); (L.D.G.); (S.M.); (C.R.M.U.); (M.A.); (M.M.); (A.P.); (L.R.)
| | - Lizzia Raffaghello
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.T.); (L.D.G.); (S.M.); (C.R.M.U.); (M.A.); (M.M.); (A.P.); (L.R.)
| | - Roberto Benelli
- IRCCS Ospedale Policlinico San Martino, 16132 Genoa, Italy; (M.T.); (L.D.G.); (S.M.); (C.R.M.U.); (M.A.); (M.M.); (A.P.); (L.R.)
| |
Collapse
|
18
|
Abueva CD, Yoon SR, Carpena NT, Ahn SC, Chang SY, Choi JE, Lee MY, Jung JY. Development of NIR photocleavable nanoparticles with BDNF for vestibular neuron regeneration. J Nanobiotechnology 2025; 23:209. [PMID: 40075449 PMCID: PMC11905548 DOI: 10.1186/s12951-025-03298-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: 12/10/2024] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
Among nanoparticle platforms, light or photoresponsive nanoparticles have emerged as a promising drug delivery strategy with spatiotemporal control while minimizing off-target effects. The characteristic absorption spectrum of the photoresponsive moiety dictates the wavelength of light needed to activate bond cleavage. However, the low tissue penetration depth limit and short-wavelength ultraviolet (UV) cellular toxicity are considered disadvantageous. This study developed a vestibular ganglion neuron organoid as a model for vestibulopathy. UV and near-infrared (NIR) radiation targeted the inner ear and neural cells, followed by toxicity evaluation. A significantly smaller toxicity of NIR light was confirmed. The photocleavage release of brain-derived neurotrophic factor (BDNF) was used by applying NIR wavelength. The results indicate that polyethylene glycol octamethylene diamine derivative conjugated with leucomethylene blue with an ethanolamine linker nanoparticle can be effectively disassembled and release BDNF when using the 808 nm laser as a trigger. The findings of the cytotoxicity assay suggest that photocleavable nanoparticles (PCNs) and laser irradiation are safe and biocompatible for human-derived and neural progenitor types of cells. Phototriggered BDNF release by NIR laser supported the growth and differentiation of human neural progenitor cells in culture. In addition, the vestibulopathy organoid exhibited a significant regenerative effect. This study harnesses the full potential of NIR laser PCNs to treat vestibular neuropathies.
Collapse
Affiliation(s)
- Celine Dg Abueva
- Beckman Laser Institute Korea, College of Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Medical Laser Research Center, College of Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Sung Ryeong Yoon
- Department of Medical Science, Graduate School of Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Nathaniel T Carpena
- Department of Medical Science, Graduate School of Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Seung Cheol Ahn
- Department of Physiology, College of Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - So-Young Chang
- Beckman Laser Institute Korea, College of Medicine, Dankook University, Cheonan, 31116, Republic of Korea
- Medical Laser Research Center, College of Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Ji Eun Choi
- Department of Otolaryngology-Head & Neck Surgery, College of Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Min Young Lee
- Department of Otolaryngology-Head & Neck Surgery, College of Medicine, Dankook University, Cheonan, 31116, Republic of Korea.
| | - Jae Yun Jung
- Department of Otolaryngology-Head & Neck Surgery, College of Medicine, Dankook University, Cheonan, 31116, Republic of Korea.
| |
Collapse
|
19
|
Acosta-Virgen K, González-Conchillos HD, Vallejo-Flores G, Salazar-Villatoro LI, Guerrero-Sánchez E, Martínez-Palomo A, Espinosa-Cantellano M. Digital PCR characterizes epithelial cell populations in murine duodenal organoids. PLoS One 2025; 20:e0319701. [PMID: 40080477 PMCID: PMC11906084 DOI: 10.1371/journal.pone.0319701] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Accepted: 02/06/2025] [Indexed: 03/15/2025] Open
Abstract
Three-dimensional cultures are powerful tools to recapitulate animal and human tissues. Under the influence of specific growth factors, adult stem cells differentiate and organize into 3D cultures named organoids. The molecular phenotyping of these structures is an essential step for validating an organoid model. However, the limited number of organoids generated in culture yields very low amounts of genetic material, making phenotyping difficult. Recently, digital PCR (dPCR) techniques have become available for the highly sensitive detection of genetic material at low concentrations. The aim of this work was to apply dPCR to the identification of the various cell populations expected to be present in murine duodenal organoids. Results show the potential use of dPCR as a genetic characterization tool for organoids.
Collapse
Affiliation(s)
- Karla Acosta-Virgen
- Departamento de Infectómica y Patogénesis Molecular, Center for Research and Advanced Studies, Mexico City, Mexico
| | | | - Gabriela Vallejo-Flores
- Department of Molecular Biology, Max Planck Institute for Infection Biology, Berlin, Germany
| | | | | | - Adolfo Martínez-Palomo
- Departamento de Infectómica y Patogénesis Molecular, Center for Research and Advanced Studies, Mexico City, Mexico
| | - Martha Espinosa-Cantellano
- Departamento de Infectómica y Patogénesis Molecular, Center for Research and Advanced Studies, Mexico City, Mexico
| |
Collapse
|
20
|
Kogler S, Skottvoll F, Hrušková H, Rise F, Aizenshtadt A, Krauss S, Ro̷berg-Larsen H, Hansen FA, Wilson SR. Electromembrane Extraction Provides Unprecedented Selectivity for Drugs in Cell Culture Media Used in Organoid and Organ-on-Chip Systems. Anal Chem 2025; 97:4923-4931. [PMID: 39995235 PMCID: PMC11912128 DOI: 10.1021/acs.analchem.4c04994] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Revised: 01/06/2025] [Accepted: 02/18/2025] [Indexed: 02/26/2025]
Abstract
The use of organoids and organ-on-chip technologies as nonanimal methodologies in drug discovery and personalized medicine is rapidly expanding. However, the complexity and small volumes of organoid culture samples present significant analytical challenges, e.g., in drug analysis using liquid chromatography-mass spectrometry (LC-MS). Essentially an electrophoresis across an oil membrane, electromembrane extraction (EME) offers a promising approach for measuring drugs, as it is, for example, compatible with small samples such as organoid and organ-on-chip formats. Given the potential of the technology, there is a need to assess the extraction purity of EME extracts to ensure EME's compatibility with high-throughput, downstream analysis. This study evaluates the effectiveness of EME for sample cleanup in various common cell culture media used for organoids and organs-on-chips. The media were spiked with 90 small-molecule drugs. Using gel electrophoresis (sodium dodecyl sulfate polyacrylamide gel electrophoresis), high-resolution nuclear magnetic resonance spectroscopy, and LC-MS, we demonstrate that EME provides exhaustive removal of unwanted medium components (proteins, polar molecules, and apolar/neutral molecules) while selectively extracting the spiked small-molecule drugs. The approach was demonstrated with human stem-cell-derived liver organoids, allowing simple detection and monitoring of telltale cytochrome P450 metabolism. Taken together, our observations highlight an unprecedented ability of EME to provide sample cleanup for drug analysis in matrixes compatible with organoids and organ-on-chip technology.
Collapse
Affiliation(s)
- Stian Kogler
- Section for
Chemical Life Sciences, Department of Chemistry, Faculty of Mathematics
and Natural Sciences, University of Oslo, Oslo 0371, Norway
- Hybrid Technology
Hub - Centre of Excellence, Institute of Basic Medical Sciences, Faculty
of Medicine, University of Oslo, Oslo 0315, Norway
| | | | - Helena Hrušková
- Section for
Chemical Life Sciences, Department of Chemistry, Faculty of Mathematics
and Natural Sciences, University of Oslo, Oslo 0371, Norway
- Hybrid Technology
Hub - Centre of Excellence, Institute of Basic Medical Sciences, Faculty
of Medicine, University of Oslo, Oslo 0315, Norway
| | - Frode Rise
- Section for
Catalysis and Organic Chemistry, Department of Chemistry, Faculty
of Mathematics and Natural Sciences, University
of Oslo, Oslo 0371, Norway
| | - Aleksandra Aizenshtadt
- Hybrid Technology
Hub - Centre of Excellence, Institute of Basic Medical Sciences, Faculty
of Medicine, University of Oslo, Oslo 0315, Norway
| | - Stefan Krauss
- Hybrid Technology
Hub - Centre of Excellence, Institute of Basic Medical Sciences, Faculty
of Medicine, University of Oslo, Oslo 0315, Norway
| | - Hanne Ro̷berg-Larsen
- Section for
Chemical Life Sciences, Department of Chemistry, Faculty of Mathematics
and Natural Sciences, University of Oslo, Oslo 0371, Norway
- Hybrid Technology
Hub - Centre of Excellence, Institute of Basic Medical Sciences, Faculty
of Medicine, University of Oslo, Oslo 0315, Norway
| | - Frederik André Hansen
- Department
of Pharmacy, Faculty of Mathematics and Natural Sciences, University of Oslo, Oslo 0371, Norway
| | - Steven Ray Wilson
- Section for
Chemical Life Sciences, Department of Chemistry, Faculty of Mathematics
and Natural Sciences, University of Oslo, Oslo 0371, Norway
- Hybrid Technology
Hub - Centre of Excellence, Institute of Basic Medical Sciences, Faculty
of Medicine, University of Oslo, Oslo 0315, Norway
| |
Collapse
|
21
|
Chang W, Cai Y, Sun J, Deng Y, Li Y, Xing L, Simayijiang Z, Gu Z, Xie Z. Surface Double Dendritic Magnetic Microfibrils for Rapid Isolation and Proteomic Profiling of Extracellular Vesicles from Microliters of Biofluids. ACS NANO 2025; 19:9196-9206. [PMID: 39996590 DOI: 10.1021/acsnano.4c18711] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/26/2025]
Abstract
The extracellular vesicles (EVs) are crucial for intercellular communication, and their proteomic analysis offers significant insights into their functions, although rapid and efficient analysis in trace biofluids is challenging due to their low abundance and potential protein loss. This study developed functionalized double dendritic magnetic microfibrils (fDDMMs) for efficient isolation and proteomic analysis of EVs from microliter biofluids. The fDDMMs possess dendritic mesoporous silica shell and magnetic Fe3O4 core, with bifunctional groups, Ti ions and R8 cell-penetrating peptide, grafted on the surface by dendritic molecules for enhanced EV capture. The multifunctional properties, including dynamic magnetic mixing and accelerated protein digestion, streamline the proteomic sample preparation process. The results demonstrated that fDDMMs enabled the rapid batch separation and proteomic sample preparation of EVs from 1 μL of plasma samples and 100 μL of tumor organoid culture medium. The rapid EV isolation and proteomic profiling approach holds great potential for liquid biopsy and personalized medicine with tiny clinic biofluids.
Collapse
Affiliation(s)
- Wenya Chang
- Institute for Molecular Medical Technology, State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, P. R. China
| | - Yuhan Cai
- Institute for Molecular Medical Technology, State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, P. R. China
| | - Jie Sun
- Institute for Molecular Medical Technology, State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, P. R. China
| | - Yuanyuan Deng
- Institute for Molecular Medical Technology, State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, P. R. China
| | - Yu Li
- Institute for Molecular Medical Technology, State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, P. R. China
| | - Likai Xing
- Institute for Molecular Medical Technology, State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, P. R. China
| | - Zulihabire Simayijiang
- Institute for Molecular Medical Technology, State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, P. R. China
| | - Zhongze Gu
- Institute of Microphysiological Systems, State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, P. R. China
| | - Zhuoying Xie
- Institute for Molecular Medical Technology, State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, P. R. China
- Institute of Microphysiological Systems, State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing, Jiangsu 210096, P. R. China
| |
Collapse
|
22
|
Krasner M, Barbiro-Michaely E, Abu-Shach UB, Onn A, Broday L, Gerber D. OncoFlow: A multiplexed microfluidic platform for personalized drug sensitivity assessment. N Biotechnol 2025; 87:105-111. [PMID: 40074170 DOI: 10.1016/j.nbt.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/16/2024] [Accepted: 03/07/2025] [Indexed: 03/14/2025]
Abstract
While biomarker-guided treatments and NGS-based approaches are refining precision medicine, they are not universally applicable. The gap between the genomic characterization of tumors and their functional behavior is becoming increasingly evident. There is an escalating demand for functional assays that can customize cancer treatments for individual patients and bridge this gap. We have developed OncoFlow, an integrated microfluidic platform that automates viability assays. This platform customizes treatment options by assessing the functional responses of a patient's tumor cells to a specific drug panel. This study specifically addressed non-small cell lung adenocarcinoma (NSCLC) in patients presenting pleural effusion. We used the NCI-H2228 adenocarcinoma cell line, which harbors the EML4-ALK fusion oncogene, to develop and fine-tune the viability assay. Cells cultivated in microfluidic chambers were treated with various concentrations of the tyrosine kinase inhibitors alectinib and crizotinib, and the cytotoxic effects were measured. The results were consistent with those from conventional cell culture methods, thereby validating the assay's reliability. Next, pleural effusion samples from six NSCLC patients, four of them harboring the EML4-ALK rearrangement were tested with alectinib and crizotinib using the OncoFlow system. Monitoring and analysis of cell viability showed varied sensitivities to crizotinib, while all samples exhibited resistance to alectinib. These findings underscore OncoFlow's potential to enhance physician decision-making and customize treatment plans, ultimately improving patient outcomes.
Collapse
Affiliation(s)
- Matan Krasner
- The Mina & Everard Goodman Faculty of Life Sciences and the Institute for Nanotechnology and Advanced Materials, Bar Ilan University, Ramat-Gan, Israel
| | - Efrat Barbiro-Michaely
- The Mina & Everard Goodman Faculty of Life Sciences and the Institute for Nanotechnology and Advanced Materials, Bar Ilan University, Ramat-Gan, Israel
| | - Ulrike Bening Abu-Shach
- Department of Cell and Developmental Biology, School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv 69978, Israel
| | - Amir Onn
- Thoracic Cancer Unit, Cancer Center, Sheba Medical Center, Tel HaShomer, Ramat Gan 52621, Israel
| | - Limor Broday
- Department of Cell and Developmental Biology, School of Medicine, Faculty of Medical and Health Sciences, Tel Aviv University, Tel Aviv 69978, Israel.
| | - Doron Gerber
- The Mina & Everard Goodman Faculty of Life Sciences and the Institute for Nanotechnology and Advanced Materials, Bar Ilan University, Ramat-Gan, Israel.
| |
Collapse
|
23
|
Vanhoeijen R, Okkelman IA, Rogier N, Sedlačík T, Stöbener DD, Devriendt B, Dmitriev RI, Hoogenboom R. Poly(2-alkyl-2-oxazoline) Hydrogels as Synthetic Matrices for Multicellular Spheroid and Intestinal Organoid Cultures. Biomacromolecules 2025; 26:1860-1872. [PMID: 39898884 DOI: 10.1021/acs.biomac.4c01627] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
The extracellular matrix (ECM) plays a crucial role in organoid cultures by supporting cell proliferation and differentiation. A key feature of the ECM is its mechanical influence on the surrounding cells, directly affecting their behavior. Matrigel, the most commonly used ECM, is limited by its animal-derived origin, batch variability, and uncontrollable mechanical properties, restricting its use in 3D cell-model-based mechanobiological studies. Poly(2-alkyl-2-oxazoline) (PAOx) synthetic hydrogels represent an appealing alternative because of their reproducibility and versatile chemistry, enabling tuning of hydrogel stiffness and functionalization. Here, we studied PAOx hydrogels with differing compressive moduli for their potential to support 3D cell growth. PAOx hydrogels support spheroid and organoid growth over several days without the addition of ECM components. Furthermore, we discovered intestinal organoid epithelial polarity reversion in PAOx hydrogels and demonstrate how the tunable mechanical properties of PAOx can be used to study effects on the morphology and oxygenation of live multicellular spheroids.
Collapse
Affiliation(s)
- Robin Vanhoeijen
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Krijgslaan 281-S4, Ghent 9000, Belgium
- Tissue Engineering and Biomaterials Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, C. Heymanslaan 10, Ghent 9000, Belgium
| | - Irina A Okkelman
- Tissue Engineering and Biomaterials Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, C. Heymanslaan 10, Ghent 9000, Belgium
| | - Nette Rogier
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Krijgslaan 281-S4, Ghent 9000, Belgium
- Tissue Engineering and Biomaterials Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, C. Heymanslaan 10, Ghent 9000, Belgium
| | - Tomáš Sedlačík
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Krijgslaan 281-S4, Ghent 9000, Belgium
- Hydrogel Lab, Department of Polymers, Faculty of Chemical Technology, University of Chemistry and Technology, Technicka 1903/5, Prague 6 166 28, Czech Republic
| | - Daniel D Stöbener
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Krijgslaan 281-S4, Ghent 9000, Belgium
| | - Bert Devriendt
- Laboratory of Immunology, Department of Translational Physiology, Infectiology and Public Health, Faculty of Veterinary Medicine, Ghent University, Merelbeke 9820, Belgium
| | - Ruslan I Dmitriev
- Tissue Engineering and Biomaterials Group, Department of Human Structure and Repair, Faculty of Medicine and Health Sciences, Ghent University, C. Heymanslaan 10, Ghent 9000, Belgium
| | - Richard Hoogenboom
- Supramolecular Chemistry Group, Centre of Macromolecular Chemistry (CMaC), Department of Organic and Macromolecular Chemistry, Faculty of Sciences, Ghent University, Krijgslaan 281-S4, Ghent 9000, Belgium
| |
Collapse
|
24
|
Rahman OM, Tarantino R, Waldman SD, Hwang DK. Single-Step Fabrication of V-Shaped Polymeric Microwells to Enhance Cancer Spheroid Formation. ACS Biomater Sci Eng 2025; 11:1857-1868. [PMID: 39967243 DOI: 10.1021/acsbiomaterials.4c02359] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/20/2025]
Abstract
Traditional cancer research has long relied on two-dimensional (2D) cell cultures, which inadequately mimic the complex three-dimensional (3D) microenvironments of in vivo tumors. Recent advancements in 3D cell cultures, particularly cancer spheroids, have highlighted their superior physiological relevance. However, existing methods for spheroid generation often require complex, multistep fabrication processes that limit scalability and reproducibility. In this study, we present a novel single-step photolithographic technique to fabricate high-aspect-ratio V-slanted hydrogel microwells. By employing polyethylene glycol (PEG)-based hydrogels, we create biocompatible, extracellular matrix (ECM)-like scaffolds that enhance gas and nutrient exchange while promoting uniform spheroid formation. The hydrogel microwells allow precise control of spheroid size, achieving a physiologically relevant diameter of 425 μm within 12-24 h, and the resulting spheroids exhibiting high viability over 3 weeks. Moreover, the method facilitates the creation of scalable multiwell arrays for high-throughput applications, making it suitable for both small-scale and large-scale experimental needs. This platform addresses the limitations of traditional microwell fabrication, offering a robust, efficient, and reproducible system for generating physiologically relevant 3D models with valuable applications in cancer research, drug testing, and tissue engineering.
Collapse
Affiliation(s)
- Omar M Rahman
- Department of Electrical, Computer, and Biomedical Engineering, Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada
- Li Ka Shing Knowledge Institute, Unity Health Toronto (St. Michael's Hospital), Toronto, Ontario M5B 1W8, Canada
- Institute of Biomedical Engineering, Science and Technology (iBEST), Unity Health Toronto and Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada
| | - Roberto Tarantino
- Department of Chemical Engineering, Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada
- Li Ka Shing Knowledge Institute, Unity Health Toronto (St. Michael's Hospital), Toronto, Ontario M5B 1W8, Canada
- Institute of Biomedical Engineering, Science and Technology (iBEST), Unity Health Toronto and Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada
| | - Stephen D Waldman
- Department of Chemical Engineering, Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada
- Li Ka Shing Knowledge Institute, Unity Health Toronto (St. Michael's Hospital), Toronto, Ontario M5B 1W8, Canada
- Institute of Biomedical Engineering, Science and Technology (iBEST), Unity Health Toronto and Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada
| | - Dae Kun Hwang
- Department of Chemical Engineering, Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada
- Li Ka Shing Knowledge Institute, Unity Health Toronto (St. Michael's Hospital), Toronto, Ontario M5B 1W8, Canada
- Institute of Biomedical Engineering, Science and Technology (iBEST), Unity Health Toronto and Toronto Metropolitan University, Toronto, Ontario M5B 2K3, Canada
| |
Collapse
|
25
|
Derobertmasure A, Toh LS, Wotring VE, Williams PM, Morbidelli L, Stingl JC, Vinken M, Ramadan R, Chhun S, Boutouyrie P. Pharmacological countermeasures for long-duration space missions: addressing cardiovascular challenges and advancing space-adapted healthcare. Eur J Pharm Sci 2025; 209:107063. [PMID: 40064402 DOI: 10.1016/j.ejps.2025.107063] [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: 11/29/2024] [Revised: 02/10/2025] [Accepted: 03/07/2025] [Indexed: 03/17/2025]
Abstract
Future long-duration crewed space missions beyond Low Earth Orbit (LEO) will bring new healthcare challenges for astronauts for which pharmacological countermeasures (pharmacological countermeasures) are crucial. This paper highlights current pharmacological countermeasures challenges described in the ESA SciSpacE Roadmap, with a focus on the cardiovascular system as a model to demonstrate the potential implication of the challenges and recommendations. New pharmacological approaches and procedures need to be adapted to spaceflight (spaceflight) conditions, including ethical and reglementary considerations. Potential strategies include combining pharmacological biomarkers such as pharmacogenomics with therapeutic drug monitoring, advancing microsampling techniques, and implementing a pharmacovigilance system to gain deep insights into pharmacokinetics/pharmacodynamics (PK/PD) spaceflight alteration on drug exposure. Emerging therapeutic approaches (such as long-term regimens) or manufacturing drugs in the space environment, can address specific issues related to drug storage and stability. The integration of biobanks and innovative technologies like organoids and organ-on-a-chip, artificial intelligence (AI), including machine learning will further enhance PK modelling leading to personalized treatments. These innovative pharmaceutical tools will also enable reciprocal game-changing healthcare developments to be made on Earth as well as in space and are essential to ensure space explorers receive safe effective pharmaceutical care.
Collapse
Affiliation(s)
- Audrey Derobertmasure
- Faculty of Medicine, Paris Cité University, INSERM PARCC, Service de Pharmacologie Clinique, Hôpital Européen Georges Pompidou Hospital (AP-HP), Paris, France
| | - Li Shean Toh
- School of Pharmacy, Faculty of Science, University of Nottingham, University Park, Nottingham NG7 2RD, United Kingdom
| | - Virginia E Wotring
- International Space University, 1 rue Jean-Dominique Cassini, Parc d'Innovation, 6700 Illkirch-Graffenstaden, France
| | - Philip M Williams
- School of Pharmacy, Faculty of Science, University of Nottingham, University Park, Nottingham NG7 2RD, United Kingdom
| | - Lucia Morbidelli
- Department of Life Sciences, University of Siena, Via A. Moro 2, 53100 Siena, Italy
| | - Julia C Stingl
- Institute of Clinical Pharmacology, University Hospital of RWTH Aachen, Wendlingweg 2, 52064, Aachen, Germany
| | - Mathieu Vinken
- Department of Pharmaceutical and Pharmacological Sciences, Vrije Universiteit Brussel, Brussels, Belgium
| | - Raghda Ramadan
- Interdisciplinary Biosciences Group, Radiobiology Unit, Belgian Nuclear Research Centre (SCK CEN), Mol, Belgium
| | - Stephanie Chhun
- Faculty of Medicine, Paris Cité University, Paris, France; Institut Necker-Enfants Malades (INEM), INSERM U1151-CNRS UMR 8253; AP-HP, Laboratory of Immunology, Necker-Enfants Malades Hospital, Paris, France
| | - Pierre Boutouyrie
- Faculty of Medicine, Paris Cité University, INSERM PARCC, Service de Pharmacologie Clinique, Hôpital Européen Georges Pompidou Hospital (AP-HP), Paris, France.
| |
Collapse
|
26
|
Zhou R, Brislinger D, Fuchs J, Lyons A, Langthaler S, Hauser CAE, Baumgartner C. Vascularised organoids: Recent advances and applications in cancer research. Clin Transl Med 2025; 15:e70258. [PMID: 40045486 PMCID: PMC11882480 DOI: 10.1002/ctm2.70258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 03/09/2025] Open
Abstract
Organoids are three-dimensional (3D) cellular models designed to replicate human tissues and organs while preserving their physiological complexity and functionality. Among these, vascularised organoids represent a groundbreaking advancement in 3D tissue engineering, incorporating vascular networks into engineered tissues to more accurately mimic the in vivo tumour microenvironment. These models offer significantly improved physiological relevance compared to conventional two-dimensional cultures or animal models, positioning them as invaluable tools in cancer research. Despite their potential, the rapid proliferation of techniques and materials for developing vascularised organoids presents challenges for researchers navigating this dynamic field. This systematic review provides a comprehensive examination of methodologies for fabricating vascularised organoids, with a focus on strategies that enhance vascularisation and support organoid growth. It critically evaluates the materials used, emphasising those that effectively mimic the extracellular matrix and facilitate vascular network formation. Key advancements in engineered organoids models are highlighted, emphasising their potential for studying interactions between vasculature and cancer cells, conducting drug screening, and understanding cytokine regulation. In summary, this review provides an in-depth overview of the current landscape of vascularised organoid fabrication and functionality, addressing challenges and opportunities within the field. A detailed understanding of the scope and future trajectories is essential for advancing organoid development and expanding their applications in both basic cancer research and clinical practice. KEY POINTS: Comparative analysis: Evaluation of organoids, animal models, and 2D models, highlighting their respective strengths and limitations in replicating physiological conditions and studying disease processes. Vascularisation techniques: Comparative evaluation of vascularised organoid fabrication methods, emphasising their efficiency, scalability and ability to replicate physiological vascular networks. Material selection: Thorough evaluation of materials for vascularised organoid culture system, focusing on those that effectively mimic the extracellular matrix and support vascular network formation. Applications: Overview of organoid applications in basic cancer research and clinical settings, with an emphasis on their potential in drug discovery, disease modelling and exploring complex biological processes.
Collapse
Affiliation(s)
- Rui Zhou
- Institute of Health Care Engineering with European Testing Center of Medical DevicesGraz University of TechnologyGrazAustria
| | - Dagmar Brislinger
- Department of Cell BiologyHistology and EmbryologyGottfried Schatz Research CenterMedical University of GrazGrazAustria
| | - Julia Fuchs
- Institute of Health Care Engineering with European Testing Center of Medical DevicesGraz University of TechnologyGrazAustria
- Department of Cell BiologyHistology and EmbryologyGottfried Schatz Research CenterMedical University of GrazGrazAustria
| | - Alicia Lyons
- Institute of Health Care Engineering with European Testing Center of Medical DevicesGraz University of TechnologyGrazAustria
| | - Sonja Langthaler
- Institute of Health Care Engineering with European Testing Center of Medical DevicesGraz University of TechnologyGrazAustria
| | - Charlotte A. E. Hauser
- Institute of Health Care Engineering with European Testing Center of Medical DevicesGraz University of TechnologyGrazAustria
| | - Christian Baumgartner
- Institute of Health Care Engineering with European Testing Center of Medical DevicesGraz University of TechnologyGrazAustria
| |
Collapse
|
27
|
Guha Ray P, Rajasekaran R, Pratihar B, De S, Dhara S, Fussenegger M. Skin-Integrated Electrogenetic Regulation of Vasculature for Accelerated Wound Healing. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412257. [PMID: 39792704 PMCID: PMC11884547 DOI: 10.1002/advs.202412257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/16/2024] [Indexed: 01/12/2025]
Abstract
Neo-vascularization plays a key role in achieving long-term viability of engineered cells contained in medical implants used in precision medicine. Moreover, strategies to promote neo-vascularization around medical implants may also be useful to promote the healing of deep wounds. In this context, a biocompatible, electroconductive borophene-poly(ε-caprolactone) (PCL) 3D platform is developed, which is called VOLT, to support designer cells engineered with a direct-current (DC) voltage-controlled gene circuit that drives secretion of vascular endothelial growth factor A (VEGFA). The VOLT platform consists of a 3D-printed borophene-PCL honeycomb-shaped matrix decorated with borophene-PCL nanofibers by electrospinning. The honeycomb structure provides mechanical stability, while the nanofibers facilitate the adhesion, migration, and proliferation of the engineered cells. The cells incorporate a DC-powered reactive oxygen species (ROS)-sensing gene circuit wired to an engineered synthetic promoter that triggers secretion of VEGFA to promote vascularization in the adjacent extracellular matrix. Cells engineered with this gene circuit and enclosed in the VOLT matrix, termed the VOLTVEGFA system, can be simply triggered using off-the-shelf AA batteries, utilizing the established ability of a brief DC bias to generate non-cytotoxic levels of ROS. For proof-of-concept, a subcutaneous wound-healing model in rats is chosen. Electrostimulation of a VOLTVEGFA implant (5 V, 20 s per day) induced secretion of VEGFA, and significantly accelerated neovascularization and granulation tissue formation, resulting in faster wound closure compared with non-stimulated controls. Complete re-epithelialization and dermal regeneration are observed within 15 days of application.
Collapse
Affiliation(s)
- Preetam Guha Ray
- ETH ZurichDepartment of Biosystems Science and EngineeringKlingelbergstrasse 48BaselCH‐4056Switzerland
- Biomaterials and Tissue Engineering LaboratorySchool of Medical Science and Technology (SMST)Indian Institute of Technology KharagpurKharagpur721302India
| | - Ragavi Rajasekaran
- Biomaterials and Tissue Engineering LaboratorySchool of Medical Science and Technology (SMST)Indian Institute of Technology KharagpurKharagpur721302India
| | - Bitan Pratihar
- Department of Chemical EngineeringIndian Institute of Technology KharagpurKharagpur721302India
| | - Sirshendu De
- Department of Chemical EngineeringIndian Institute of Technology KharagpurKharagpur721302India
| | - Santanu Dhara
- Biomaterials and Tissue Engineering LaboratorySchool of Medical Science and Technology (SMST)Indian Institute of Technology KharagpurKharagpur721302India
| | - Martin Fussenegger
- ETH ZurichDepartment of Biosystems Science and EngineeringKlingelbergstrasse 48BaselCH‐4056Switzerland
- Faculty of ScienceUniversity of BaselKlingelbergstrasse 48BaselCH‐4056Switzerland
| |
Collapse
|
28
|
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
|
29
|
Ma L, Zhang Z, Mu Y, Liu B, Zhou H, Wang DA. The Application of Biomaterial-Based Spinal Cord Tissue Engineering. Macromol Biosci 2025; 25:e2400444. [PMID: 39472074 DOI: 10.1002/mabi.202400444] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 10/11/2024] [Indexed: 03/14/2025]
Abstract
Advancements in biomaterial-based spinal cord tissue engineering technology have profoundly influenced regenerative medicine, providing innovative solutions for both spinal cord organoid development and engineered spinal cord injury (SCI) repair. In spinal cord organoids, biomaterials offer a supportive microenvironment that mimics the natural extracellular matrix, facilitating cell differentiation and organization and advancing the understanding of spinal cord development and pathophysiology. Furthermore, biomaterials are essential in constructing engineered spinal cords for SCI repair. The incorporation of biomaterials with growth factors, fabrication of ordered scaffold structures, and artificial spinal cord assemblies are critical insights for SCI to ensure structural integrity, enhance cell viability, and promote neural regeneration in transplantation. In summary, this review summarizes the contribution of biomaterials to the spinal cord organoids progression and discusses strategies for biomaterial-based spinal cord engineering in SCI therapy. These achievements underscore the transformative potential of biomaterials to improve treatment options for SCI and accelerate future clinical applications.
Collapse
Affiliation(s)
- Liang Ma
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, 999077, China
| | - Zhen Zhang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, 999077, China
| | - Yulei Mu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, 999077, China
| | - Bangheng Liu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, 999077, China
| | - Huiqun Zhou
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, 999077, China
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, 999077, China
- Centre for Neuromusculoskeletal Restorative Medicine, InnoHK HKSTP, Sha Tin, Hong Kong, 999077, China
- Tung Biomedical Sciences Centre, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong, 999077, China
| |
Collapse
|
30
|
Kesharwani A, Tani S, Nishikawa M, Sakai Y, Okada H, Ohba S, Chung UI, Hojo H. Modeling vascular dynamics at the initial stage of endochondral ossification on a microfluidic chip using a human embryonic-stem-cell-derived organoid. Regen Ther 2025; 28:90-100. [PMID: 39703814 PMCID: PMC11655692 DOI: 10.1016/j.reth.2024.11.018] [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: 11/13/2024] [Revised: 11/20/2024] [Accepted: 11/21/2024] [Indexed: 12/21/2024] Open
Abstract
Vascular interactions play a crucial role in embryogenesis, including skeletal development. During endochondral ossification, vascular networks are formed as mesenchymal cells condense and later invade skeletal elements to form the bone marrow. We and other groups developed a model of endochondral ossification by implanting human embryonic stem cell (hESC)-derived sclerotome into immunodeficient mice. However, in vitro models of endochondral ossification, particularly vascular interaction with mesenchymal cells at its initial stage, are yet to be established. Therefore, we developed a method to model the initial stage of endochondral ossification using a microfluidic chip-based platform, with a particular focus on the vascular interaction. On the chip, we found that the fibrin gel helped align mCherry-expressing human umbilical vein endothelial cells (HUVECs) better than the collagen-I gel, suggesting that the fibrin gel is more suitable for the formation of a vascular-like network. The perfusability of the vascular-like networks was partially confirmed using fluorescein isothiocyanate (FITC)-dextran and fluorescent microbeads. We then mixed hESC-derived sclerotome with enhanced green fluorescent protein (EGFP)-expressing HUVECs and applied this mixture on the chip. We named this mixture of cells SH organoids. The SH organoids showed superior abilities to maintain the vascular-like network, which was formed by the mCherry-expressing HUVECs, compared with the sclerotome spheroids on the chip. The EGFP-expressing HUVECs migrated from the SH organoid, formed a vascular-like networks, and partially interacted with the mCherry-expressing vascular-like networks on the chip. Histological analysis showed that SRY-box transcription factor 9 (SOX9) and type I collagen were expressed mutually exclusively in the condensed mesenchymal cells and perichondrial-like cells, respectively. This study demonstrates that our SH organoid-on-a-chip method reproduces vascular networks that are formed at the initial stage of endochondral ossification. This model may provide insights into human endochondral ossification and has potential applications in bone disease modeling and drug screening.
Collapse
Affiliation(s)
- Abhiraj Kesharwani
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8654, Japan
| | - Shoichiro Tani
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
- Department of Orthopaedic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Masaki Nishikawa
- Department of Chemical Systems Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8654, Japan
| | - Yasuyuki Sakai
- Department of Chemical Systems Engineering, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8654, Japan
| | - Hiroyuki Okada
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
- Department of Orthopaedic Surgery, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Shinsuke Ohba
- Department of Tissue and Developmental Biology, Graduate School of Dentistry, Osaka University, Osaka 565-0871, Japan
| | - Ung-il Chung
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8654, Japan
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| | - Hironori Hojo
- Department of Bioengineering, Graduate School of Engineering, The University of Tokyo, Tokyo 113-8654, Japan
- Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, The University of Tokyo, Tokyo 113-8655, Japan
| |
Collapse
|
31
|
Li R, Wang J, Lin Q, Yin Z, Zhou F, Chen X, Tan H, Su J. Mechano-Responsive Biomaterials for Bone Organoid Construction. Adv Healthc Mater 2025; 14:e2404345. [PMID: 39740101 DOI: 10.1002/adhm.202404345] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 12/08/2024] [Indexed: 01/02/2025]
Abstract
Mechanical force is essential for bone development, bone homeostasis, and bone fracture healing. In the past few decades, various biomaterials have been developed to provide mechanical signals that mimic the natural bone microenvironment, thereby promoting bone regeneration. Bone organoids, emerging as a novel research approach, are 3D micro-bone tissues that possess the ability to self-renew and self-organize, exhibiting biomimetic spatial characteristics. Incorporating mechano-responsive biomaterials in the construction of bone organoids presents a promising avenue for simulating the mechanical bone microenvironment. Therefore, this review commences by elucidating the impact of mechanical force on bone health, encompassing both cellular interactions and alterations in bone structure. Furthermore, the most recent applications of mechano-responsive biomaterials within the realm of bone tissue engineering are highlighted. Three different types of mechano-responsive biomaterials are introduced with a focus on their responsive mechanisms, construction strategies, and efficacy in facilitating bone regeneration. Based on a comprehensive overview, the prospective utilization and future challenges of mechano-responsive biomaterials in the construction of bone organoids are discussed. As bone organoid technology advances, these biomaterials are poised to become powerful tools in bone regeneration.
Collapse
Affiliation(s)
- Ruiyang Li
- Department of Orthopedics, Trauma Orthopedics Center, Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
- Institute of Translational Medicine, National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Jian Wang
- Department of Orthopedics, Trauma Orthopedics Center, Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
- Institute of Translational Medicine, National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| | - Qiushui Lin
- Department of Spine Surgery, First Affiliated Hospital of Naval Medical University, Shanghai, 200433, P. R. China
| | - Zhifeng Yin
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 200941, P. R. China
| | - Fengjin Zhou
- Department of Orthopedics, Honghui Hospital, Xi'an Jiao Tong University, Xi'an, 710000, P. R. China
| | - Xiao Chen
- Department of Orthopedics, Trauma Orthopedics Center, Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
| | - Hongbo Tan
- Department of Orthopedics, The 920th Hospital of Joint Logistics Support Force, Yunnan, 650020, P. R. China
| | - Jiacan Su
- Department of Orthopedics, Trauma Orthopedics Center, Institute of Musculoskeletal Injury and Translational Medicine of Organoids, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200092, P. R. China
- Institute of Translational Medicine, National Center for Translational Medicine SHU Branch, Shanghai University, Shanghai, 200444, P. R. China
| |
Collapse
|
32
|
Hu Y, Peng Z, Qiu M, Xue L, Ren H, Wu X, Zhu X, Ding Y. Developing biotechnologies in organoids for liver cancer. BIOMEDICAL TECHNOLOGY 2025; 9:100067. [DOI: 10.1016/j.bmt.2024.100067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
|
33
|
Rembotte L, Beneyton T, Buisson L, Badon A, Boyreau A, Douillet C, Hermant L, Jana A, Nassoy P, Baret J. Pheno-Morphological Screening and Acoustic Sorting of 3D Multicellular Aggregates Using Drop Millifluidics. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2410677. [PMID: 39792815 PMCID: PMC11884609 DOI: 10.1002/advs.202410677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 12/20/2024] [Indexed: 01/12/2025]
Abstract
Three-dimensional multicellular aggregates (MCAs) like organoids and spheroids have become essential tools to study the biological mechanisms involved in the progression of diseases. In cancer research, they are now widely used as in vitro models for drug testing. However, their analysis still relies on tedious manual procedures, which hinders their routine use in large-scale biological assays. Here, a novel drop millifluidic approach is introduced to screen and sort large populations containing over one thousand MCAs: ImOCAS (Image-based Organoid Cytometry and Acoustic Sorting). This system utilizes real-time image processing to detect pheno-morphological traits in MCAs. They are then encapsulated in millimetric drops, actuated on-demand using the acoustic radiation force. The performance of ImOCAS is demonstrated by sorting spheroids with uniform sizes from a heterogeneous population, and by isolating organoids from spheroids with different phenotypes. This approach lays the groundwork for high-throughput screening and high-content analysis of MCAs with controlled morphological and phenotypical properties, which promises accelerated progress in biomedical research.
Collapse
Affiliation(s)
- Leon Rembotte
- CNRSUniv. BordeauxCRPPUMR 5031PessacF‐33600France
- LP2NUniv. BordeauxTalenceF‐33400France
| | | | | | - Amaury Badon
- LP2NUniv. BordeauxTalenceF‐33400France
- IOGSCNRSUMR5298TalenceF‐33400France
| | | | - Camille Douillet
- LP2NUniv. BordeauxTalenceF‐33400France
- IOGSCNRSUMR5298TalenceF‐33400France
| | - Loic Hermant
- LP2NUniv. BordeauxTalenceF‐33400France
- IOGSCNRSUMR5298TalenceF‐33400France
| | - Anirban Jana
- LP2NUniv. BordeauxTalenceF‐33400France
- TreeFrog TherapeuticsPessacF‐33600France
| | - Pierre Nassoy
- LP2NUniv. BordeauxTalenceF‐33400France
- IOGSCNRSUMR5298TalenceF‐33400France
| | | |
Collapse
|
34
|
Kim J, Ślęczkowska M, Nobre B, Wieringa P. Study Models for Chlamydia trachomatis Infection of the Female Reproductive Tract. Microorganisms 2025; 13:553. [PMID: 40142446 PMCID: PMC11945960 DOI: 10.3390/microorganisms13030553] [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: 12/23/2024] [Revised: 02/13/2025] [Accepted: 02/14/2025] [Indexed: 03/28/2025] Open
Abstract
Chlamydia trachomatis (Ct) is a leading cause of sexually transmitted infections globally, often resulting in inflammatory disorders, ectopic pregnancies, and infertility. Studying Ct's pathogenesis remains challenging due to its unique life cycle and host-specific interactions, which require diverse experimental models. Animal studies using mouse, guinea pig, pig, and non-human primate models provide valuable insights into immune responses, hormonal influences, and disease progression. However, they face limitations in terms of translational relevance due to physiological differences, as well as ethical concerns. Complementing these, in vitro systems, ranging from simple monolayer to advanced three-dimensional models, exhibit improved physiological relevance by replicating the human tissue architecture. This includes the detailed investigation of epithelial barrier disruptions, epithelium-stroma interactions, and immune responses at a cellular level. Nonetheless, in vitro models fall short in mimicking the intricate tissue structures found in vivo and, therefore, cannot faithfully replicate the host-pathogen interactions or infection dynamics observed in living organisms. This review presents a comprehensive overview of the in vivo and in vitro models employed over the past few decades to investigate Ct and its pathogenesis, addressing their strengths and limitations. Furthermore, we explore emerging technologies, including organ-on-chip and in silico models, as promising tools to overcome the existing challenges and refine our understanding of Ct infections.
Collapse
Affiliation(s)
| | | | | | - Paul Wieringa
- Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands; (J.K.); (M.Ś.); (B.N.)
| |
Collapse
|
35
|
Bokhout L, Campeiro JD, Dalm SU. Exploring the landscape of current in vitro and in vivo models and their relevance for targeted radionuclide theranostics. Eur J Nucl Med Mol Imaging 2025:10.1007/s00259-025-07123-3. [PMID: 40016527 DOI: 10.1007/s00259-025-07123-3] [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: 10/07/2024] [Accepted: 01/28/2025] [Indexed: 03/01/2025]
Abstract
Cancer remains a leading cause of mortality globally, driving ongoing research into innovative treatment strategies. Preclinical research forms the base for developing these novel treatments, using both in vitro and in vivo model systems that are, ideally, as clinically representative as possible. Emerging as a promising approach for cancer management, targeted radionuclide theranostics (TRT) uses radiotracers to deliver (cytotoxic) radionuclides specifically to cancer cells. Since the field is relatively new, more advanced preclinical models are not yet regularly applied in TRT research. This narrative review examines the currently applied in vitro, ex vivo and in vivo models for oncological research, discusses if and how these models are now applied for TRT studies, and whether not yet applied models can be of benefit for the field. A selection of different models is discussed, ranging from in vitro two-dimensional (2D) and three-dimensional (3D) cell models, including spheroids, organoids and tissue slice cultures, to in vivo mouse cancer models, such as cellline-derived models, patient-derived xenograft models and humanized models. Each of the models has advantages and limitations for studying human cancer biology, radiopharmaceutical assessment and treatment efficacy. Overall, there is a need to apply more advanced models in TRT research that better address specific TRT phenomena, such as crossfire and abscopal effects, to enhance the clinical relevance and effectiveness of preclinical TRT evaluations.
Collapse
Affiliation(s)
- Lisa Bokhout
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Joana D Campeiro
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands
| | - Simone U Dalm
- Department of Radiology and Nuclear Medicine, Erasmus MC, Rotterdam, The Netherlands.
| |
Collapse
|
36
|
Finch SC, Harwood DT. Past, Current and Future Techniques for Monitoring Paralytic Shellfish Toxins in Bivalve Molluscs. Toxins (Basel) 2025; 17:105. [PMID: 40137878 PMCID: PMC11945588 DOI: 10.3390/toxins17030105] [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: 01/28/2025] [Revised: 02/19/2025] [Accepted: 02/21/2025] [Indexed: 03/29/2025] Open
Abstract
Paralytic shellfish poisoning is a threat to human health caused by the consumption of shellfish contaminated with toxins of the saxitoxin class. Human health is protected by the setting of regulatory limits and the analysis of shellfish prior to sale. Both robust toxicity data, generated from experiments fitting into the ethical 3R framework, and appropriate analysis methods are required to ensure the success of this approach. A literature review of in vivo animal bioassays and in vitro and analytical methods showed that in vitro methods are the best option to screen shellfish for non-regulatory purposes. However, since neither the receptor nor antibody binding of paralytic shellfish toxin analogues correlate with toxicity, these assays cannot accurately quantify toxicity in shellfish nor be used to calculate toxicity equivalence factors. Fully replacing animals in testing is rightfully the ultimate goal, but this cannot be at a cost to human health. More modern technology, such as organ-on-a-chip, represent an exciting development, but animal bioassays cannot currently be replaced in the determination of toxicity. Analytical methods that employ toxicity equivalence factors calculated using oral animal toxicity data result in an accurate assessment of the food safety risk posed by paralytic shellfish toxin contamination in bivalve molluscs.
Collapse
Affiliation(s)
- Sarah C. Finch
- AgResearch Ltd., Ruakura Research Centre, Private Bag 3123, Hamilton 3240, New Zealand
| | - D. Tim Harwood
- Cawthron Institute, Private Bag 2, Nelson 7042, New Zealand;
- New Zealand Food Safety Science and Research Centre, Massey University, Private Bag 11 222, Palmerston North 4442, New Zealand
| |
Collapse
|
37
|
Borrelli MJ, Kolendowski B, DiMattia GE, Shepherd TG. Spatiotemporal analysis of ratiometric biosensors in live multicellular spheroids using SPoRTS. CELL REPORTS METHODS 2025; 5:100987. [PMID: 39965566 PMCID: PMC11955269 DOI: 10.1016/j.crmeth.2025.100987] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 11/06/2024] [Accepted: 01/27/2025] [Indexed: 02/20/2025]
Abstract
Here, we describe SPoRTS, an open-source workflow for high-throughput spatiotemporal image analysis of fluorescence-based ratiometric biosensors in living spheroids. To achieve this, we have implemented a fully automated algorithm for the acquisition of line intensity profile data, ultimately enabling semi-quantitative measurement of biosensor activity as a function of distance from the center of the spheroid. We demonstrate the functionality of SPoRTS via spatial analysis of live spheroids expressing a ratiometric biosensor based on the fluorescent, ubiquitin-based cell-cycle indicator (FUCCI) system, which identifies mitotic cells. We compare this FUCCI-based SPoRTS analysis with spatially quantified immunostaining for proliferation markers, finding that the results are strongly correlated.
Collapse
Affiliation(s)
- Matthew J Borrelli
- The Mary and John Knight Translational Ovarian Cancer Research Unit, Verspeeten Family Cancer Centre, London, ON N6A 5W9, Canada; Department of Anatomy and Cell Biology, Western University, London, ON N6A 5C1, Canada
| | - Bart Kolendowski
- The Mary and John Knight Translational Ovarian Cancer Research Unit, Verspeeten Family Cancer Centre, London, ON N6A 5W9, Canada
| | - Gabriel E DiMattia
- The Mary and John Knight Translational Ovarian Cancer Research Unit, Verspeeten Family Cancer Centre, London, ON N6A 5W9, Canada; Department of Oncology, Western University, London, ON N6A 5W9, Canada; Department of Biochemistry, Western University, London, ON N6A 5C1, Canada
| | - Trevor G Shepherd
- The Mary and John Knight Translational Ovarian Cancer Research Unit, Verspeeten Family Cancer Centre, London, ON N6A 5W9, Canada; Department of Anatomy and Cell Biology, Western University, London, ON N6A 5C1, Canada; Department of Oncology, Western University, London, ON N6A 5W9, Canada; Department of Obstetrics and Gynecology, Western University, London, ON N6A 5W9, Canada.
| |
Collapse
|
38
|
Artegiani B, Hendriks D. Organoids from pluripotent stem cells and human tissues: When two cultures meet each other. Dev Cell 2025; 60:493-511. [PMID: 39999776 DOI: 10.1016/j.devcel.2025.01.005] [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: 03/11/2024] [Revised: 06/13/2024] [Accepted: 01/10/2025] [Indexed: 02/27/2025]
Abstract
Human organoids are a widely used tool in cell biology to study homeostatic processes, disease, and development. The term organoids covers a plethora of model systems from different cellular origins that each have unique features and applications but bring their own challenges. This review discusses the basic principles underlying organoids generated from pluripotent stem cells (PSCs) as well as those derived from tissue stem cells (TSCs). We consider how well PSC- and TSC-organoids mimic the different intended organs in terms of cellular complexity, maturity, functionality, and the ongoing efforts to constitute predictive complex models of in vivo situations. We discuss the advantages and limitations associated with each system to answer different biological questions including in the field of cancer and developmental biology, and with respect to implementing emerging advanced technologies, such as (spatial) -omics analyses, CRISPR screens, and high-content imaging screens. We postulate how the two fields may move forward together, integrating advantages of one to the other.
Collapse
Affiliation(s)
| | - Delilah Hendriks
- Princess Máxima Center for Pediatric Oncology, Utrecht, the Netherlands.
| |
Collapse
|
39
|
Malak M, Qian C, James J, Nair S, Grantham J, Ericson MB. Insights into metabolic changes during epidermal differentiation as revealed by multiphoton microscopy with fluorescence lifetime imaging. Sci Rep 2025; 15:6377. [PMID: 39984626 PMCID: PMC11845624 DOI: 10.1038/s41598-025-90101-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2024] [Accepted: 02/10/2025] [Indexed: 02/23/2025] Open
Abstract
Rapid developments in the field of organotypic cultures have generated a growing need for effective and non-invasive methods for quality control during tissue development. In this study, we correlate metabolic changes with epidermal differentiation and demonstrate that multiphoton microscopy with fluorescence lifetime imaging (MPM-FLIM) can be applied to monitor epidermal differentiation of keratinocytes with respect to proliferative and differentiated states. In a 2D keratinocyte tissue culture model, increased expression of differentiation markers keratin-1 and keratin-10 was induced with calcium supplementation. An accompanying shift from glycolysis to mitochondrial respiration was detected in metabolic flux assays. Analysis of MPM-FLIM images acquired at 750 nm and 900 nm excitation revealed a decreased relative fraction of intracellular NADH and FAD after high calcium treatment, consistent with increased oxidative phosphorylation. Epidermal differentiation could be monitored over a 96 h period. Discrimination analysis based on k-means clustering generated clusters that correlated well with the duration of high Ca2+ treatment, suggesting that MPM-FLIM can provide useful parameters for monitoring keratinocyte differentiation.
Collapse
Affiliation(s)
- Monika Malak
- Department of Chemistry and Molecular Biology, Faculty of Science, University of Gothenburg, Gothenburg, 412 96, Sweden
| | - Chen Qian
- Department of Chemistry and Molecular Biology, Faculty of Science, University of Gothenburg, Gothenburg, 412 96, Sweden.
| | - Jeemol James
- Department of Chemistry and Molecular Biology, Faculty of Science, University of Gothenburg, Gothenburg, 412 96, Sweden
| | - Syam Nair
- Institute of Neuroscience and Physiology, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, 413 90, Sweden
- Institute of Clinical Sciences, The Sahlgrenska Academy, University of Gothenburg, Gothenburg, 416 85, Sweden
| | - Julie Grantham
- Department of Chemistry and Molecular Biology, Faculty of Science, University of Gothenburg, Gothenburg, 412 96, Sweden
| | - Marica B Ericson
- Department of Chemistry and Molecular Biology, Faculty of Science, University of Gothenburg, Gothenburg, 412 96, Sweden.
| |
Collapse
|
40
|
Ali M, Benfante V, Basirinia G, Alongi P, Sperandeo A, Quattrocchi A, Giannone AG, Cabibi D, Yezzi A, Di Raimondo D, Tuttolomondo A, Comelli A. Applications of Artificial Intelligence, Deep Learning, and Machine Learning to Support the Analysis of Microscopic Images of Cells and Tissues. J Imaging 2025; 11:59. [PMID: 39997561 PMCID: PMC11856378 DOI: 10.3390/jimaging11020059] [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: 12/31/2024] [Revised: 02/08/2025] [Accepted: 02/12/2025] [Indexed: 02/26/2025] Open
Abstract
Artificial intelligence (AI) transforms image data analysis across many biomedical fields, such as cell biology, radiology, pathology, cancer biology, and immunology, with object detection, image feature extraction, classification, and segmentation applications. Advancements in deep learning (DL) research have been a critical factor in advancing computer techniques for biomedical image analysis and data mining. A significant improvement in the accuracy of cell detection and segmentation algorithms has been achieved as a result of the emergence of open-source software and innovative deep neural network architectures. Automated cell segmentation now enables the extraction of quantifiable cellular and spatial features from microscope images of cells and tissues, providing critical insights into cellular organization in various diseases. This review aims to examine the latest AI and DL techniques for cell analysis and data mining in microscopy images, aid the biologists who have less background knowledge in AI and machine learning (ML), and incorporate the ML models into microscopy focus images.
Collapse
Affiliation(s)
- Muhammad Ali
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy; (M.A.); (G.B.)
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
| | - Viviana Benfante
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
- Advanced Diagnostic Imaging—INNOVA Project, Department of Radiological Sciences, A.R.N.A.S. Civico, Di Cristina e Benfratelli Hospitals, P.zza N. Leotta 4, 90127 Palermo, Italy;
- Pharmaceutical Factory, La Maddalena S.P.A., Via San Lorenzo Colli, 312/d, 90146 Palermo, Italy;
| | - Ghazal Basirinia
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy; (M.A.); (G.B.)
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
| | - Pierpaolo Alongi
- Advanced Diagnostic Imaging—INNOVA Project, Department of Radiological Sciences, A.R.N.A.S. Civico, Di Cristina e Benfratelli Hospitals, P.zza N. Leotta 4, 90127 Palermo, Italy;
- Department of Biomedicine, Neuroscience and Advanced Diagnostics (BiND), University of Palermo, 90127 Palermo, Italy
| | - Alessandro Sperandeo
- Pharmaceutical Factory, La Maddalena S.P.A., Via San Lorenzo Colli, 312/d, 90146 Palermo, Italy;
| | - Alberto Quattrocchi
- Pathologic Anatomy Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy; (A.Q.); (A.G.G.); (D.C.)
| | - Antonino Giulio Giannone
- Pathologic Anatomy Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy; (A.Q.); (A.G.G.); (D.C.)
| | - Daniela Cabibi
- Pathologic Anatomy Unit, Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, University of Palermo, 90127 Palermo, Italy; (A.Q.); (A.G.G.); (D.C.)
| | - Anthony Yezzi
- Department of Electrical and Computer Engineering, Georgia Institute of Technology, Atlanta, GA 30332, USA;
| | - Domenico Di Raimondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
| | - Antonino Tuttolomondo
- Department of Health Promotion, Mother and Child Care, Internal Medicine and Medical Specialties, Molecular and Clinical Medicine, University of Palermo, 90127 Palermo, Italy; (D.D.R.); (A.T.)
| | - Albert Comelli
- Ri.MED Foundation, Via Bandiera 11, 90133 Palermo, Italy; (M.A.); (G.B.)
| |
Collapse
|
41
|
Cabriel C, Córdova-Castro RM, Berenschot E, Dávila-Lezama A, Pondman K, Le Gac S, Tas N, Susarrey-Arce A, Izeddin I. 3D Single-Molecule Super-Resolution Imaging of Microfabricated Multiscale Fractal Substrates for Calibration and Cell Imaging. ACS APPLIED MATERIALS & INTERFACES 2025; 17:9019-9034. [PMID: 39901441 DOI: 10.1021/acsami.4c19431] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2025]
Abstract
Microstructures arrayed over a substrate have shown increasing interest due to their ability to provide advanced 3D cellular models, which open up new possibilities for cell culture, proliferation, and differentiation. Still, the mechanisms by which physical cues impact the cell phenotype are not fully understood, hence the necessity to interrogate cell behavior at the highest resolution. However, cell 3D high-resolution optical imaging on such microstructured substrates remains challenging due to their complexity as well as axial calibration issues. In this work, we address this issue by leveraging the geometrical characteristics of fractal-like structures, which serve as axial calibration tools and modulate cell growth. To this end, we use multiscale 3D SiO2 substrates consisting of spatially arrayed octahedral features of a few micrometers to hundreds of nanometers. Through optimizations of both the structures and optical imaging conditions, we demonstrate the potential of these 3D multiscale structures as an alternative to electron microscopy for material imaging but also as calibration tools for 3D super-resolution microscopy. We used their multiscale and known geometry to perform lateral and axial calibrations in 3D single-molecule localization microscopy (SMLM) and assess imaging resolutions. We then utilized these substrates as a platform for high-resolution bioimaging. As a proof of concept, we cultivate human mesenchymal stem cells on these substrates, revealing very different growth patterns compared to flat glass. Specifically, the spatial distribution of cytoskeleton proteins is vastly modified, as we demonstrate with a 3D SMLM assessment.
Collapse
Affiliation(s)
- Clément Cabriel
- Institut Langevin, ESPCI Paris, CNRS, Université PSL, Paris 75005, France
| | - R Margoth Córdova-Castro
- Institut Langevin, ESPCI Paris, CNRS, Université PSL, Paris 75005, France
- Department of Physics, University of Ottawa, 25 Templeton Street, Ottawa, ON K1N 6N5, Canada
| | - Erwin Berenschot
- Mesoscale Chemical Systems, MESA+ Institute, University of Twente, PO. Box 217, Enschede 7500 AE, The Netherlands
| | - Amanda Dávila-Lezama
- Mesoscale Chemical Systems, MESA+ Institute, University of Twente, PO. Box 217, Enschede 7500 AE, The Netherlands
- Facultad de Ciencias de la Salud, Universidad Autónoma de Baja California, Blvd. Universitario número 1000, Valle de las Palmas 22260, México
| | - Kirsten Pondman
- Applied Microfluidics for BioEngineering Research, MESA+ Institute for Nanotechnology & TechMed Centre, and Organ-on-Chip Centre, University of Twente, Enschede 7500 AE, The Netherlands
| | - Séverine Le Gac
- Applied Microfluidics for BioEngineering Research, MESA+ Institute for Nanotechnology & TechMed Centre, and Organ-on-Chip Centre, University of Twente, Enschede 7500 AE, The Netherlands
| | - Niels Tas
- Mesoscale Chemical Systems, MESA+ Institute, University of Twente, PO. Box 217, Enschede 7500 AE, The Netherlands
| | - Arturo Susarrey-Arce
- Mesoscale Chemical Systems, MESA+ Institute, University of Twente, PO. Box 217, Enschede 7500 AE, The Netherlands
| | - Ignacio Izeddin
- Institut Langevin, ESPCI Paris, CNRS, Université PSL, Paris 75005, France
| |
Collapse
|
42
|
Sun Y, Zhang H, Huang F, Gao Q, Li P, Li D, Luo G. Deliod a lightweight detection model for intestinal organoids based on deep learning. Sci Rep 2025; 15:5040. [PMID: 39934224 DOI: 10.1038/s41598-025-89409-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 02/05/2025] [Indexed: 02/13/2025] Open
Abstract
Intestinal organoids are indispensable tools for exploring intestinal disorders. Deep learning methodologies are often employed in morphological analysis to evaluate the condition of these organoids. Nonetheless, prevailing analytical techniques face obstacles such as many organisational overlaps and tiny targets lead to a high incidence of errors and limited applicability. This paper presents Deliod, a streamlined intestinal organoid detection model founded on YOLOv8 and designed to automate the identification of organoid morphology. Deliod performed excellently compared to leading detection models when applied to an intestinal organoid dataset, attaining an mAP50 of 87.5%. Ablation experiments verified the module's efficacy in improving detection performance. Furthermore, Deliod features a modest parameter count of 5.41 M and a computational load of 16.6 GFLOPs, facilitating the broader application of the detection model in the realm of intestinal organoid image recognition. This streamlined model not only enables efficient and accurate recognition of organoid morphology but also minimizes hardware deployment requirements, broadening its range of potential applications.
Collapse
Affiliation(s)
- Yu Sun
- College of Electrical and Automation Engineering, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Hanwen Zhang
- Engineering Laboratory of Advanced In Vitro Diagnostic Technology, Suzhou Institute of Biomedical Engineering and Technology Chinese Academy of Sciences, Chinese Academy of Sciences, Suzhou, 215163, P. R. China
| | - Fengliang Huang
- College of Electrical and Automation Engineering, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Qin Gao
- College of Electrical and Automation Engineering, Nanjing Normal University, Nanjing, 210023, P. R. China
| | - Peng Li
- Engineering Laboratory of Advanced In Vitro Diagnostic Technology, Suzhou Institute of Biomedical Engineering and Technology Chinese Academy of Sciences, Chinese Academy of Sciences, Suzhou, 215163, P. R. China
| | - Dong Li
- Engineering Laboratory of Advanced In Vitro Diagnostic Technology, Suzhou Institute of Biomedical Engineering and Technology Chinese Academy of Sciences, Chinese Academy of Sciences, Suzhou, 215163, P. R. China.
| | - Gangyin Luo
- Engineering Laboratory of Advanced In Vitro Diagnostic Technology, Suzhou Institute of Biomedical Engineering and Technology Chinese Academy of Sciences, Chinese Academy of Sciences, Suzhou, 215163, P. R. China.
| |
Collapse
|
43
|
Zabiegalov O, Berger A, Kamdar D, Adamou K, Tian C, Mbefo M, Quinodoz M, Udry F, Rivolta C, Kostic C, Arsenijevic Y. Generation of a Double Reporter mES Cell Line to Simultaneously Trace the Generation of Retinal Progenitors and Photoreceptors. Cells 2025; 14:252. [PMID: 39996725 PMCID: PMC11854395 DOI: 10.3390/cells14040252] [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: 08/06/2024] [Revised: 01/25/2025] [Accepted: 01/29/2025] [Indexed: 02/26/2025] Open
Abstract
Three-dimensional retinal culture systems help to understand eye development and the pathology of disorders. There is a need for reporter stem cell lines to allow in vitro studies on retinal progenitors and photoreceptors and their developmental dynamics or properties and to test therapeutic approaches. The isolation of pure progenitor populations or photoreceptor precursors may serve for drug, gene, and cell therapy development. Here, we generated a dual-reporter mouse embryonic stem cell line Crx-GFP;Rax-mCherry enabling the visualization or isolation of photoreceptors and retinal progenitors from retinal organoid settings. From day 4 organoids, we isolated mCherry-positive cells to assess their early retinal progenitor identity with proliferation tests as well as transcriptomic and proteomic profiling. The timing of eye field transcription factor expression at the transcriptomic and protein levels is in accordance with mouse retinogenesis. This new line will be helpful for rapidly investigating biological questions or testing therapeutics before using human induced pluripotent stem cells (iPSCs), which require a much longer time for retinal organoid formation.
Collapse
Affiliation(s)
- Oleksandr Zabiegalov
- Unit of Retinal Degeneration and Regeneration, Department of Ophthalmology, University of Lausanne, 1004 Lausanne, Switzerland; (C.T.); (M.M.); (F.U.)
| | - Adeline Berger
- Unit of Epigenetics of Ocular Diseases, Department of Ophthalmology, University of Lausanne, 1004 Lausanne, Switzerland;
| | - Dhryata Kamdar
- Ophthalmic Genetics Group, Institute of Molecular and Clinical Ophthalmology, 4031 Basel, Switzerland; (D.K.); (M.Q.); (C.R.)
- Department of Ophthalmology, University of Basel, 4031 Basel, Switzerland
| | - Kabirou Adamou
- Group for Retinal Disorders Research, Department of Ophthalmology, University of Lausanne, 1004 Lausanne, Switzerland; (K.A.); (C.K.)
| | - Chuanxi Tian
- Unit of Retinal Degeneration and Regeneration, Department of Ophthalmology, University of Lausanne, 1004 Lausanne, Switzerland; (C.T.); (M.M.); (F.U.)
| | - Martial Mbefo
- Unit of Retinal Degeneration and Regeneration, Department of Ophthalmology, University of Lausanne, 1004 Lausanne, Switzerland; (C.T.); (M.M.); (F.U.)
| | - Mathieu Quinodoz
- Ophthalmic Genetics Group, Institute of Molecular and Clinical Ophthalmology, 4031 Basel, Switzerland; (D.K.); (M.Q.); (C.R.)
- Department of Ophthalmology, University of Basel, 4031 Basel, Switzerland
| | - Florian Udry
- Unit of Retinal Degeneration and Regeneration, Department of Ophthalmology, University of Lausanne, 1004 Lausanne, Switzerland; (C.T.); (M.M.); (F.U.)
| | - Carlo Rivolta
- Ophthalmic Genetics Group, Institute of Molecular and Clinical Ophthalmology, 4031 Basel, Switzerland; (D.K.); (M.Q.); (C.R.)
- Department of Ophthalmology, University of Basel, 4031 Basel, Switzerland
| | - Corinne Kostic
- Group for Retinal Disorders Research, Department of Ophthalmology, University of Lausanne, 1004 Lausanne, Switzerland; (K.A.); (C.K.)
| | - Yvan Arsenijevic
- Unit of Retinal Degeneration and Regeneration, Department of Ophthalmology, University of Lausanne, 1004 Lausanne, Switzerland; (C.T.); (M.M.); (F.U.)
| |
Collapse
|
44
|
Wu Z, Chen X, Han F, Leeansyah E. MAIT cell homing in intestinal homeostasis and inflammation. SCIENCE ADVANCES 2025; 11:eadu4172. [PMID: 39919191 PMCID: PMC11804934 DOI: 10.1126/sciadv.adu4172] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Accepted: 01/08/2025] [Indexed: 02/09/2025]
Abstract
Mucosa-associated invariant T (MAIT) cells are a large population of unconventional T cells widely distributed in the human gastrointestinal tract. Their homing to the gut is central to maintaining mucosal homeostasis and immunity. This review discusses the potential mechanisms that guide MAIT cells to the intestinal mucosa during homeostasis and inflammation, emphasizing the roles of chemokines, chemokine receptors, and tissue adhesion molecules. The potential influence of the gut microbiota on MAIT cell homing to different regions of the human gut is also discussed. Last, we introduce how organoid technology offers a potentially valuable approach to advance our understanding of MAIT cell tissue homing by providing a more physiologically relevant model that mimics the human gut tissue. These models may enable a detailed investigation of the gut-specific homing mechanisms of MAIT cells. By understanding the regulation of MAIT cell homing to the human gut, potential avenues for therapeutic interventions targeting gut inflammatory conditions such as inflammatory bowel diseases (IBD) may emerge.
Collapse
Affiliation(s)
- Zhengyu Wu
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Xingchi Chen
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Fei Han
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| | - Edwin Leeansyah
- Institute of Biopharmaceutical and Health Engineering, Tsinghua Shenzhen International Graduate School, Tsinghua University, Shenzhen 518055, China
| |
Collapse
|
45
|
Ekanger CT, Dinesh Kumar N, Koutstaal RW, Zhou F, Beukema M, Waldock J, Jochems SP, Mulder N, van Els CACM, Engelhardt OG, Mantel N, Buno KP, Brokstad KA, Engelsen AST, Cox RJ, Melgert BN, Huckriede ALW, van Kasteren PB. Comparison of air-liquid interface transwell and airway organoid models for human respiratory virus infection studies. Front Immunol 2025; 16:1532144. [PMID: 39981254 PMCID: PMC11839712 DOI: 10.3389/fimmu.2025.1532144] [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: 11/21/2024] [Accepted: 01/21/2025] [Indexed: 02/22/2025] Open
Abstract
Introduction Complex in vitro respiratory models, including air-liquid interface (ALI) transwell cultures and airway organoids, have emerged as promising tools for studying human respiratory virus infections. These models address several limitations of conventional two-dimensional cell line and animal models. However, the lack of standardized protocols for the application of these models in infection studies limits the possibilities for comparing results across different research groups. Therefore, we applied a collaborative approach to harmonize several aspects of experimental methodology between different research laboratories, aiming to assess the comparability of different models of human airway epithelium in the context of respiratory viral infections. Methods In this study, we compared three different models of human respiratory epithelium: a primary human bronchial epithelial cell-derived ALI transwell model, and two airway organoid models established from human airway- and lung-derived adult stem cells. We first assessed the presence of various differentiated cell types using immunofluorescence microscopy. Using a shared stock of influenza A virus, we then assessed viral growth kinetics, epithelial cytokine responses, and serum-mediated inhibition of infection. Results The presence of club, goblet, and ciliated cells was confirmed in all models. We observed similar viral replication kinetics with a >4-log increase in virus titre across all models using a TCID50 assay. Following infection, a reproducible antiviral cytokine response, including a consistent increase in CXCL10, IL-6, IFN-λ1, IFN-λ2/3, and IFN-β, was detected across all models. Finally, neutralization was assessed by pre-incubation of virus with human serum. Reduced viral replication was observed across all models, resulting in a 3- to 6-log decrease in virus titres as quantified by TCID50. Discussion In conclusion, all three models produced consistent results regardless of the varying cell sources, culturing approaches, and infection methods. Our collaborative efforts to harmonize infection experiments and compare ALI transwell and airway organoid models described here aid in advancing our understanding and improving the standardization of these complex in vitro respiratory models for future studies.
Collapse
Affiliation(s)
- Camilla T. Ekanger
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Medicine, University of Bergen, Bergen, Norway
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Nilima Dinesh Kumar
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Rosanne W. Koutstaal
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| | - Fan Zhou
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Martin Beukema
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Joanna Waldock
- Influenza Resource Centre, Vaccines, Science Research & Innovation, Medicines and Healthcare Products Regulatory Agency (MHRA), Potters Bar, United Kingdom
| | - Simon P. Jochems
- Leiden University Center for Infectious Diseases (LU-CID), Leiden University Medical Center, Leiden, Netherlands
| | - Noa Mulder
- Groningen Research Institute for Pharmacy, Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
| | - Cécile A. C. M. van Els
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
- Section Immunology, Department Biomolecular Health Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Othmar G. Engelhardt
- Influenza Resource Centre, Vaccines, Science Research & Innovation, Medicines and Healthcare Products Regulatory Agency (MHRA), Potters Bar, United Kingdom
| | | | | | - Karl Albert Brokstad
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Safety, Chemistry and Biomedical Laboratory Sciences, Western Norway University of Applied Sciences, Bergen, Norway
| | - Agnete S. T. Engelsen
- Department of Biomedicine, Faculty of Medicine, University of Bergen, Bergen, Norway
- Centre for Cancer Biomarkers (CCBIO), Department of Clinical Medicine, University of Bergen, Bergen, Norway
| | - Rebecca J. Cox
- Influenza Centre, Department of Clinical Science, University of Bergen, Bergen, Norway
- Department of Microbiology, Haukeland University Hospital, Bergen, Norway
| | - Barbro N. Melgert
- Groningen Research Institute for Pharmacy, Department of Molecular Pharmacology, University of Groningen, Groningen, Netherlands
- Groningen Research Institute for Asthma and COPD, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Anke L. W. Huckriede
- Department of Medical Microbiology and Infection Prevention, University Medical Center Groningen, University of Groningen, Groningen, Netherlands
| | - Puck B. van Kasteren
- Center for Infectious Disease Control, National Institute for Public Health and the Environment (RIVM), Bilthoven, Netherlands
| |
Collapse
|
46
|
Naghipour S, Corben LA, Hulme AJ, Dottori M, Delatycki MB, Lees JG, Lim SY. Omaveloxolone for the Treatment of Friedreich Ataxia: Efficacy, Safety, and Future Perspectives. Mov Disord 2025; 40:226-230. [PMID: 39559924 DOI: 10.1002/mds.30070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2024] [Revised: 11/06/2024] [Accepted: 11/08/2024] [Indexed: 11/20/2024] Open
Affiliation(s)
- Saba Naghipour
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
| | - Louise A Corben
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, Melbourne University, Parkville, Victoria, Australia
| | - Amy J Hulme
- School of Medical, Indigenous and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, New South Wales, Australia
| | - Mirella Dottori
- School of Medical, Indigenous and Health Sciences, Molecular Horizons, University of Wollongong, Wollongong, New South Wales, Australia
| | - Martin B Delatycki
- Bruce Lefroy Centre for Genetic Health Research, Murdoch Children's Research Institute, Parkville, Victoria, Australia
- Department of Paediatrics, Melbourne University, Parkville, Victoria, Australia
- Victorian Clinical Genetics Services, Parkville, Victoria, Australia
| | - Jarmon G Lees
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
- Department of Medicine and Surgery, University of Melbourne, Parkville, Victoria, Australia
- Drug Discovery Biology, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Clayton, Victoria, Australia
| | - Shiang Y Lim
- O'Brien Institute Department, St Vincent's Institute of Medical Research, Fitzroy, Victoria, Australia
- Department of Medicine and Surgery, University of Melbourne, Parkville, Victoria, Australia
- Drug Discovery Biology, Faculty of Pharmacy and Pharmaceutical Sciences, Monash University, Clayton, Victoria, Australia
- National Heart Research Institute, National Heart Center, Singapore
| |
Collapse
|
47
|
Li K, Gu L, Cai H, Lu HC, Mackie K, Guo F. Human brain organoids for understanding substance use disorders. Drug Metab Pharmacokinet 2025; 60:101036. [PMID: 39567282 PMCID: PMC11825288 DOI: 10.1016/j.dmpk.2024.101036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2024]
Abstract
Substance use disorders (SUDs) are complex mental health conditions involving a problematic pattern of substance use. Challenges remain in understanding its neural mechanisms, which are likely to lead to improved SUD treatments. Human brain organoids, brain-like 3D in vitro cultures derived from human stem cells, show unique potential in recapitulating the response of a developing human brain to substances. Here, we review the recent progress in understanding SUD using human brain organoid models focusing on neurodevelopmental perspectives. We first summarize the background of SUD in humans. Moreover, we introduce the development of various human brain organoid models and then discuss current progress and findings underlying the abuse of substances like nicotine, alcohol, and other addictive drugs using organoid models. Furthermore, we review efforts to develop organ chips and microphysiological systems to engineer better human brain organoids for advancing SUD studies. Lastly, we conclude by elaborating on the current challenges and future directions of SUD studies using human brain organoids.
Collapse
Affiliation(s)
- Kangle Li
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, USA
| | - Longjun Gu
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, USA
| | - Hongwei Cai
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, USA
| | - Hui-Chen Lu
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Indiana University Bloomington, IN, 47405, USA
| | - Ken Mackie
- Gill Center for Biomolecular Science, Department of Psychological and Brain Sciences, Indiana University Bloomington, IN, 47405, USA
| | - Feng Guo
- Department of Intelligent Systems Engineering, Indiana University Bloomington, IN, 47405, USA.
| |
Collapse
|
48
|
Brauer J, Tumani M, Frey N, Lehmann LH. The cardio-oncologic burden of breast cancer: molecular mechanisms and importance of preclinical models. Basic Res Cardiol 2025; 120:91-112. [PMID: 39621070 PMCID: PMC11790711 DOI: 10.1007/s00395-024-01090-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 10/22/2024] [Accepted: 10/22/2024] [Indexed: 02/04/2025]
Abstract
Breast cancer, the most prevalent cancer affecting women worldwide, poses a significant cardio-oncological burden. Despite advancements in novel therapeutic strategies, anthracyclines, HER2 antagonists, and radiation remain the cornerstones of oncological treatment. However, each carries a risk of cardiotoxicity, though the molecular mechanisms underlying these adverse effects differ. Common mechanisms include DNA damage response, increased reactive oxygen species, and mitochondrial dysfunction, which are key areas of ongoing research for potential cardioprotective strategies. Since these mechanisms are also essential for effective tumor cytotoxicity, we explore tumor-specific effects, particularly in hereditary breast cancer linked to BRCA1 and BRCA2 mutations. These genetic variants impair DNA repair mechanisms, increase the risk of tumorigenesis and possibly for cardiotoxicity from treatments such as anthracyclines and HER2 antagonists. Novel therapies, including immune checkpoint inhibitors, are used in the clinic for triple-negative breast cancer and improve the oncological outcomes of breast cancer patients. This review discusses the molecular mechanisms underlying BRCA dysfunction and the associated pathological pathways. It gives an overview of preclinical models of breast cancer, such as genetically engineered mouse models, syngeneic murine models, humanized mouse models, and various in vitro and ex vivo systems and models to study cardiovascular side effects of breast cancer therapies. Understanding the underlying mechanism of cardiotoxicity and developing cardioprotective strategies in preclinical models are essential for improving treatment outcomes and reducing long-term cardiovascular risks in breast cancer patients.
Collapse
Affiliation(s)
- J Brauer
- Department of Cardiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- German Center of Cardiovascular Research (DZHK), Partnersite Heidelberg, Mannheim, Germany
| | - M Tumani
- Department of Cardiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- German Center of Cardiovascular Research (DZHK), Partnersite Heidelberg, Mannheim, Germany
| | - N Frey
- Department of Cardiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany
- German Center of Cardiovascular Research (DZHK), Partnersite Heidelberg, Mannheim, Germany
| | - L H Lehmann
- Department of Cardiology, University Hospital Heidelberg, Im Neuenheimer Feld 410, 69120, Heidelberg, Germany.
- German Center of Cardiovascular Research (DZHK), Partnersite Heidelberg, Mannheim, Germany.
- German Cancer Research Center (DKFZ), Heidelberg, Germany.
| |
Collapse
|
49
|
Shao Y, Wang J, Jin A, Jiang S, Lei L, Liu L. Biomaterial-assisted organoid technology for disease modeling and drug screening. Mater Today Bio 2025; 30:101438. [PMID: 39866785 PMCID: PMC11757232 DOI: 10.1016/j.mtbio.2024.101438] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 12/10/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025] Open
Abstract
Developing disease models and screening for effective drugs are key areas of modern medical research. Traditional methodologies frequently fall short in precisely replicating the intricate architecture of bodily tissues and organs. Nevertheless, recent advancements in biomaterial-assisted organoid technology have ushered in a paradigm shift in biomedical research. This innovative approach enables the cultivation of three-dimensional cellular structures in vitro that closely emulate the structural and functional attributes of organs, offering physiologically superior models compared to conventional techniques. The evolution of biomaterials plays a pivotal role in supporting the culture and development of organ tissues, thereby facilitating more accurate disease state modeling and the rigorous evaluation of drug efficacy and safety profiles. In this review, we will explore the roles that various biomaterials play in organoid development, examine the fundamental principles and advantages of utilizing these technologies in constructing disease models, and highlight recent advances and practical applications in drug screening using disease-specific organoid models. Additionally, the challenges and future directions of organoid technology are discussed. Through continued research and innovation, we aim to make remarkable strides in disease treatment and drug development, ultimately enhancing patient quality of life.
Collapse
Affiliation(s)
- Yunyuan Shao
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, China
| | - Juncheng Wang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Anqi Jin
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, China
| | - Shicui Jiang
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| | - Lanjie Lei
- Key Laboratory of Artificial Organs and Computational Medicine in Zhejiang Province, Institute of Translational Medicine, Zhejiang Shuren University, Hangzhou, 310015, China
| | - Liangle Liu
- The Third Affiliated Hospital of Wenzhou Medical University, Wenzhou, 325200, China
| |
Collapse
|
50
|
Kalla J, Pfneissl J, Mair T, Tran L, Egger G. A systematic review on the culture methods and applications of 3D tumoroids for cancer research and personalized medicine. Cell Oncol (Dordr) 2025; 48:1-26. [PMID: 38806997 PMCID: PMC11850459 DOI: 10.1007/s13402-024-00960-8] [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] [Accepted: 05/11/2024] [Indexed: 05/30/2024] Open
Abstract
Cancer is a highly heterogeneous disease, and thus treatment responses vary greatly between patients. To improve therapy efficacy and outcome for cancer patients, more representative and patient-specific preclinical models are needed. Organoids and tumoroids are 3D cell culture models that typically retain the genetic and epigenetic characteristics, as well as the morphology, of their tissue of origin. Thus, they can be used to understand the underlying mechanisms of cancer initiation, progression, and metastasis in a more physiological setting. Additionally, co-culture methods of tumoroids and cancer-associated cells can help to understand the interplay between a tumor and its tumor microenvironment. In recent years, tumoroids have already helped to refine treatments and to identify new targets for cancer therapy. Advanced culturing systems such as chip-based fluidic devices and bioprinting methods in combination with tumoroids have been used for high-throughput applications for personalized medicine. Even though organoid and tumoroid models are complex in vitro systems, validation of results in vivo is still the common practice. Here, we describe how both animal- and human-derived tumoroids have helped to identify novel vulnerabilities for cancer treatment in recent years, and how they are currently used for precision medicine.
Collapse
Affiliation(s)
- Jessica Kalla
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Janette Pfneissl
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Theresia Mair
- Department of Pathology, Medical University of Vienna, Vienna, Austria
| | - Loan Tran
- Department of Pathology, Medical University of Vienna, Vienna, Austria
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria
| | - Gerda Egger
- Department of Pathology, Medical University of Vienna, Vienna, Austria.
- Ludwig Boltzmann Institute Applied Diagnostics, Vienna, Austria.
- Comprehensive Cancer Center, Medical University of Vienna, Vienna, Austria.
| |
Collapse
|