Sign in to use this feature.

Years

Between: -

Subjects

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Article Types

Countries / Regions

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline

Search Results (347)

Search Parameters:
Keywords = 3D-organoid cell culture

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
22 pages, 2846 KB  
Review
From Cells to Organoids: Approaches, Regulatory Mechanisms, Applications, and Challenges of Organoids
by Lihong Wang, Anqi Chen, Dong Zhang and Zuping He
Cells 2025, 14(23), 1898; https://doi.org/10.3390/cells14231898 - 29 Nov 2025
Viewed by 86
Abstract
Organoids refer to three-dimensional (3D) multicellular tissues derived from stem cells or single cells through their self-assembly capacity, and significantly, they mimic structural and functional characteristics of the organ from which they are derived. Organoids can maintain the gene expression profiles and mutational [...] Read more.
Organoids refer to three-dimensional (3D) multicellular tissues derived from stem cells or single cells through their self-assembly capacity, and significantly, they mimic structural and functional characteristics of the organ from which they are derived. Organoids can maintain the gene expression profiles and mutational features of parental cells during long-term culture. This makes organoids more relevant to the human bodies than gene knockout or overexpression animal models. Consequently, organoids have been widely used in various kinds of fields, including studies on organ developmental mechanisms, regenerative medicine, organ repair, the construction of disease models, high-throughput drug screening, and personalized medicine. Notably, significant progress has recently been made in organoid construction methodologies and regulatory mechanisms. These include the selections of starting cell sources, optimizing matrix materials, and the related cell signaling pathways. The rapid development of organoid technologies has provided new opportunities for their applications in organ transplantation, drug and toxicity screening, and molecular mechanisms for cell and tissue development. In this review, we discuss organoid construction methods involving the starting cell selection and spatiotemporal mediation, regulatory mechanisms with signaling molecules and pathways, and their applications in unveiling organogenesis mechanisms and disease etiology, drug screening, toxicity testing, personalized medicine, regenerative medicine, and alternatives to animal experiments. We also address the perspectives and challenges in this field with an aim to promote the development of organoids in basic research and translational medicine. Full article
(This article belongs to the Special Issue Organ Regeneration: Cells, Organoids and Organs)
Show Figures

Figure 1

18 pages, 2566 KB  
Article
Feasibility of Patient-Derived 3D Gastrointestinal Stromal Tumour Models as Alternatives for In Vivo Mouse Models
by Dina Mönch, Julia Thiel, Meng Dong, Annika Maaß, Eileen Wegner, Anna Binner, Annette M. Staiger, Katrin S. Kurz, German Ott, Philipp Renner, Tobias Leibold, Christian Schmees, Thomas E. Mürdter, Matthias Schwab, Marc-H. Dahlke and Jana Koch
Int. J. Mol. Sci. 2025, 26(23), 11456; https://doi.org/10.3390/ijms262311456 - 26 Nov 2025
Viewed by 51
Abstract
Gastrointestinal stromal tumours (GISTs) are the most common mesenchymal tumours of the gastrointestinal tract and a key example for targeted therapy with tyrosine kinase inhibitors (TKIs), which have significantly improved survival rates. However, no effective treatments exist for TKI-resistant or mutation-negative tumours. Until [...] Read more.
Gastrointestinal stromal tumours (GISTs) are the most common mesenchymal tumours of the gastrointestinal tract and a key example for targeted therapy with tyrosine kinase inhibitors (TKIs), which have significantly improved survival rates. However, no effective treatments exist for TKI-resistant or mutation-negative tumours. Until now, research on the effects of TKIs has mainly used 2D cultures or mouse models, lacking patient-specific 3D GIST models. We investigated various 3D GIST models, including spheroids, organoids, patient-derived microtumours (PDMs), and precision-cut tumour slices (PCTSs), to assess their feasibility as alternatives for 2D cell culture or in vivo mouse models. Moreover, 2D monolayer and 3D spheroid GIST cell lines showed mutation-dependent responses to TKI treatment, but differences between 2D and 3D cultures were minimal. Thus, patient-derived 3D models, incorporating tumour microenvironment cells, were developed for more accurate in vivo representation. PDMs and PCTSs were successfully isolated from primary tumours and cultivated for up to two weeks. Three-dimensional models were immunohistochemically characterised, and the response to TKI therapies was tested and compared with expected clinical outcomes. In addition to already established 2D cell cultures and mouse models, PDMs and PCTSs are novel patient-derived 3D models that can be used to study tumour cell interactions within the microenvironment. Moreover, they could be used to investigate TKI resistance, and novel treatment options such as immunotherapies and combination therapies. Full article
(This article belongs to the Special Issue Recent Advances in 3D Tumor Models for Cancer Research)
Show Figures

Figure 1

28 pages, 1795 KB  
Review
Transcription Factor-Based Differentiation of Pluripotent Stem Cells: Overcoming the Traps of Random Neuronal Fate
by Georgie McDaid, Jaime Vanek, Brett Cromer and Huseyin Sumer
Biomedicines 2025, 13(11), 2783; https://doi.org/10.3390/biomedicines13112783 - 14 Nov 2025
Viewed by 1110
Abstract
Developing robust methods to differentiate pluripotent stem cells (PSCs) into specific neuronal subtypes is crucial for advancing neuroscience research, including disease modelling and regenerative medicine. Research in this area has primarily focused on generating and studying excitatory neurons, often in co-culture with primary [...] Read more.
Developing robust methods to differentiate pluripotent stem cells (PSCs) into specific neuronal subtypes is crucial for advancing neuroscience research, including disease modelling and regenerative medicine. Research in this area has primarily focused on generating and studying excitatory neurons, often in co-culture with primary astrocytes to support maturation. Due to the shared ectodermal lineage of these cell types, any mesoderm derived cells, such as microglia, are absent using traditional methods of culture. To more accurately model the intricate complexity of the brain and its normal neuronal physiology, it is important to incorporate other critical neural subtypes, such as inhibitory interneurons and various glial cells. This review highlights recent progress in using transcription factor-based in vitro differentiation strategies to generate these diverse neural populations. A major advantage of this approach is the ability to rapidly produce highly specific cell types in a controlled manner, allowing for the precise seeding of cells at defined anatomical and physiological ratios. This controlled methodology enables the creation of more accurate and reproducible in vitro models, including two-dimensional (2D) and three-dimensional (3D) cultures and organoids, thereby moving beyond the limitations of random differentiation from neuronal progenitor cells. Despite these advances, key challenges remain, including reproducibility between pluripotent stem cell lines, off-target transcriptional effects of exogenous factors, and incomplete phenotypic maturation of derived cells. Addressing these constraints is essential for translating transcription factor-based approaches into robust and clinically relevant neural models. Full article
(This article belongs to the Special Issue Stem Cell Therapy: Traps and Tricks)
Show Figures

Figure 1

23 pages, 1957 KB  
Review
Three-Dimensional Models of the Dental Pulp: Bridging Fundamental Biology and Regenerative Therapy
by Rana Smaida, Guoqiang Hua, Nadia Benkirane-Jessel and Florence Fioretti
Int. J. Mol. Sci. 2025, 26(22), 10960; https://doi.org/10.3390/ijms262210960 - 12 Nov 2025
Viewed by 519
Abstract
The dental pulp is a dynamic connective tissue essential for tooth vitality, sensory function, immune defense, and reparative dentinogenesis. Conventional endodontic procedures, while effective in eradicating infection, often result in a non-functional, devitalized tooth, highlighting the need for biologically based regenerative approaches. The [...] Read more.
The dental pulp is a dynamic connective tissue essential for tooth vitality, sensory function, immune defense, and reparative dentinogenesis. Conventional endodontic procedures, while effective in eradicating infection, often result in a non-functional, devitalized tooth, highlighting the need for biologically based regenerative approaches. The emergence of three-dimensional (3D) culture systems has transformed pulp biology and endodontic research by providing physiologically relevant microenvironments that better reproduce the dentino-pulp interface, vascular and neural networks, and immune interactions. This review synthesizes current advances in 3D dental pulp modeling, from scaffold-based and hydrogel systems to spheroids, organoids, bioprinted constructs, and microfluidic “tooth-on-a-chip” platforms. Each system’s composition, biological relevance, and translational potential are critically examined with respect to odontogenic differentiation, angiogenesis, neurogenesis, and inflammatory response. Applications in disease modeling, biomaterial screening, and regenerative endodontics are highlighted, showing how these models bridge fundamental biology and therapeutic innovation. Finally, we discuss key challenges including vascularization, innervation, standardization, and clinical translation, and propose integrative strategies combining bioprinting, stem-cell engineering, and organ-on-chip technologies to achieve functional pulp regeneration. Overall, 3D pulp models represent a paradigm shift from reductionist cultures to bioinstructive, patient-relevant platforms that accelerate the development of next-generation endodontic therapies. Full article
(This article belongs to the Special Issue Application of Biotechnology to Dental Treatment)
Show Figures

Figure 1

35 pages, 22753 KB  
Review
Integrating 3D Bioprinting with Organoid Technology-Based Breast Cancer Models for Drug Evaluation
by Arvind Kumar Shukla, Sandhya Shukla, Raj Kumar Mongre, Adarsha Mahendra Upadhyay, Govindhan Thiruppathi, Chandra Dhar Shukla, Shuktika Mishra and Sayan Deb Dutta
Organoids 2025, 4(4), 26; https://doi.org/10.3390/organoids4040026 - 5 Nov 2025
Viewed by 763
Abstract
Breast cancer remains one of the leading causes of cancer morbidity and mortality among women worldwide. Conventional two-dimensional (2D) cell culture models and animal studies often fail to accurately recapitulate the complex tumor microenvironment and heterogeneous nature of breast cancer. Recent advancements in [...] Read more.
Breast cancer remains one of the leading causes of cancer morbidity and mortality among women worldwide. Conventional two-dimensional (2D) cell culture models and animal studies often fail to accurately recapitulate the complex tumor microenvironment and heterogeneous nature of breast cancer. Recent advancements in tissue engineering have enabled the development of more physiologically relevant models using three-dimensional (3D) bioprinting and organoid technology. This study focuses on integrating 3D bioprinting with patient-derived organoid models to replicate breast cancer tissue architecture, cellular heterogeneity, and tumor-stroma interactions. Utilizing biomimetic bioinks and customized bioprinting protocols, we successfully fabricated breast cancer tissue constructs embedded with stromal and immune components. These engineered models demonstrated high fidelity in mimicking in vivo tumor pathophysiology, including angiogenesis, epithelial–mesenchymal transition, and extracellular matrix remodeling. Furthermore, the platform allowed for high-throughput drug screening and evaluation of therapeutic responses, revealing differential sensitivities to chemotherapeutics and targeted therapies. Our findings highlight the potential of bioprinted organoid models as powerful tools for personalized medicine, enabling more predictive and reliable cancer research and drug development. Full article
Show Figures

Figure 1

38 pages, 5411 KB  
Review
3D Bioprinting Functional Engineered Heart Tissues
by Man Chi Leung and Zachary Laksman
Int. J. Mol. Sci. 2025, 26(21), 10707; https://doi.org/10.3390/ijms262110707 - 3 Nov 2025
Viewed by 1184
Abstract
Three-dimensional (3D) bioprinting is increasingly explored as a strategy for myocardial repair and regenerative medicine. Conventional 3D casting often yields heterogeneous cellularization, slow electromechanical maturation, and inadequate vascularization; by contrast, bioprinting places cells and biomaterials in predefined architectures to program alignment, stiffness, vascular [...] Read more.
Three-dimensional (3D) bioprinting is increasingly explored as a strategy for myocardial repair and regenerative medicine. Conventional 3D casting often yields heterogeneous cellularization, slow electromechanical maturation, and inadequate vascularization; by contrast, bioprinting places cells and biomaterials in predefined architectures to program alignment, stiffness, vascular pathways, and electrical coupling that better recapitulate native myocardium. This review focuses on cardiac-specific advances in 3D bioprinting. We compare major platforms (jetting, light-based, extrusion, and volumetric) and their trade-offs for cardiac applications; distill bioink design principles trending toward natural–synthetic hybrids, including conductive and shape-morphing components; and outline practical characterization readouts spanning rheology, print fidelity, swelling/degradation, and cardiac function. We also summarize cell sources and co-culture strategies. Applications surveyed include cardiac patches, engineered tissues, chambered constructs, and organoids. Finally, we discuss current limitations and potential future directions for 3D bioprinting cardiac tissues. Collectively, recent advances position 3D bioprinting to accelerate the realization of in vivo-like engineered heart tissues. Full article
(This article belongs to the Section Biochemistry)
Show Figures

Figure 1

27 pages, 1591 KB  
Review
Human-Induced Pluripotent Stem Cell Models for Amyloid Cardiomyopathy: From Mechanistic Insights to Therapeutic Discovery
by Yufeng Liu and Muhammad Riaz
J. Cardiovasc. Dev. Dis. 2025, 12(11), 434; https://doi.org/10.3390/jcdd12110434 - 2 Nov 2025
Viewed by 659
Abstract
Amyloid cardiomyopathy (ACM), driven by transthyretin (TTR) and immunoglobulin light chain (LC) amyloid fibrils, remains a major clinical challenge due to limited mechanistic understanding and insufficient preclinical models. Human-induced pluripotent stem cells (iPSCs) have emerged as a transformative platform to model ACM, offering [...] Read more.
Amyloid cardiomyopathy (ACM), driven by transthyretin (TTR) and immunoglobulin light chain (LC) amyloid fibrils, remains a major clinical challenge due to limited mechanistic understanding and insufficient preclinical models. Human-induced pluripotent stem cells (iPSCs) have emerged as a transformative platform to model ACM, offering patient-specific and genetically controlled systems. In this review, we summarize recent advances in the use of iPSC-derived cardiomyocytes (iPSC-CMs) in both two-dimensional (2D) monolayer cultures and three-dimensional (3D) constructs—including spheroids, organoids, cardiac microtissues, and engineered heart tissues (EHTs)—for disease modeling, mechanistic research, and drug discovery. While 2D culture of iPSC-CMs reproduces hallmark proteotoxic phenotypes such as sarcomeric disorganization, oxidative stress, and apoptosis in ACM, 3D models provide enhanced physiological relevance through incorporating multicellularity, extracellular matrix interactions, and mechanical load-related features. Genome editing with Clustered Regularly Interspaced Short Palindromic Repeats (CRISPR)-Cas9 further broadens the scope of iPSC-based models, enabling isogenic comparisons and the dissection of mutation-specific effects, particularly in transthyretin-related amyloidosis (ATTR). Despite limitations such as cellular immaturity and challenges in recapitulating aging-associated phenotypes, ongoing refinements in differentiation, maturation, and dynamic training of iPSC-cardiac models hold great promise for overcoming these barriers. Together, these advances position iPSC-based systems as powerful human-relevant platforms for modeling and elucidating disease mechanisms and accelerating therapeutic development to prevent ACM. Full article
(This article belongs to the Section Acquired Cardiovascular Disease)
Show Figures

Graphical abstract

19 pages, 8252 KB  
Article
A Thymus-Independent Artificial Organoid System Supports Complete Thymopoiesis from Rhesus Macaque-Derived Hematopoietic Stem and Progenitor Cells
by Callie Wilde, Saleem Anwar, Yu-Tim Yau, Sunil Badve, Yesim Gökmen-Polar, John D. Roback, Rama Rao Amara, R. Paul Johnson and Sheikh Abdul Rahman
Biomedicines 2025, 13(11), 2692; https://doi.org/10.3390/biomedicines13112692 - 1 Nov 2025
Viewed by 900
Abstract
Background: T cell regeneration in the thymus is intrinsically linked to the T cell-biased lineage differentiation of hematopoietic stem and progenitor cells (HSPCs). Although nonhuman primates (NHPs) serve as indispensable models for studying thymic output under physiological and pathological conditions, a non-animal technology [...] Read more.
Background: T cell regeneration in the thymus is intrinsically linked to the T cell-biased lineage differentiation of hematopoietic stem and progenitor cells (HSPCs). Although nonhuman primates (NHPs) serve as indispensable models for studying thymic output under physiological and pathological conditions, a non-animal technology facilitating efficient TCR-selected T cell development and evaluating T cell output from NHP-derived HSPCs has been lacking. To address this gap, we established a rhesus macaque-specific artificial thymic organoid (RhATO) modeling primary thymus-tissue-free thymopoiesis. Methods: The RhATO was developed by expressing Rhesus macaque (RM) Delta-like Notch ligand 1 in mouse bone marrow stromal cell line (MS5-RhDLL1). The bone marrow-derived HSPCs were aggregated with MS5-RhDLL1 and cultured forming 3D artificial thymic organoids. These organoids were maintained under defined cytokine conditions to support complete T cell developmental ontogeny. T cell developmental progression was assessed by flow cytometry, and TCR-selected subsets were analyzed for phenotypic and functional properties. Results: RhATOs recapitulated the complete spectrum of thymopoietic events, including emergence of thymus-seeding progenitors, CD4+CD3 immature single-positive and CD4+CD8+ double-positive early thymocytes, and mature CD4+ or CD8+ single-positive subsets. These subsets expressed CD38, consistent with the recent thymic emigrant phenotype, and closely mirrored canonical T cell ontogeny described in humans. RhATO-derived T cells were TCR-selected and demonstrated cytokine expression upon stimulation. Conclusions: This study provides the first demonstration of an NHP-specific artificial thymic technology that faithfully models thymopoiesis. RhATO represents a versatile ex vivo platform for studying T cell development, immunopathogenesis, and generating TCR selected T cells. Full article
Show Figures

Figure 1

33 pages, 2286 KB  
Review
Antigenic Dark Matter: Unexplored Post-Translational Modifications of Tumor-Associated and Tumor-Specific Antigens in Pancreatic Cancer
by Amin Safa, Idris Vruzhaj, Marta Gambirasi and Giuseppe Toffoli
Cancers 2025, 17(21), 3506; https://doi.org/10.3390/cancers17213506 - 30 Oct 2025
Viewed by 910
Abstract
Background: Pancreatic ductal adenocarcinoma (PDAC) exhibits marked resistance to immunotherapy. Beyond its characteristically low tumor mutational burden, post-translational modifications (PTMs) remodel the immunopeptidome and promote immune escape through reversible, enzyme-driven programs. Subject Matter: We synthesize evidence that aberrant glycosylation, O-GlcNAcylation, phosphorylation, and citrullination [...] Read more.
Background: Pancreatic ductal adenocarcinoma (PDAC) exhibits marked resistance to immunotherapy. Beyond its characteristically low tumor mutational burden, post-translational modifications (PTMs) remodel the immunopeptidome and promote immune escape through reversible, enzyme-driven programs. Subject Matter: We synthesize evidence that aberrant glycosylation, O-GlcNAcylation, phosphorylation, and citrullination constitute core determinants of antigen visibility operating within spatially discrete tumor niches and a desmoplastic stroma. In hypoxic regions, HIF-linked hexosamine metabolism and OGT activity stabilize immune checkpoints and attenuate antigen processing; at tumor margins, sialylated mucins engage inhibitory Siglec receptors on innate and adaptive lymphocytes; within the stroma, PAD4-dependent NET formation enforces T cell exclusion. We also delineate technical barriers to discovering PTM antigens labile chemistry, low stoichiometry, and method-embedded biases and outline practical solutions: ETD/EThcD/AI-ETD fragmentation, PTM-aware database searching and machine-learning models, and autologous validation in patient-derived organoid–T cell co-cultures. Finally, we highlight therapeutic strategies that either immunize against PTM neoepitopes or inhibit PTM machinery (e.g., PAD4, OGT, ST6GAL1), with stromal remodeling as an enabling adjunct. Conclusions: PTM biology, spatial omics, and patient sample models can uncover targetable niches and speed up PDAC vaccination, TCR, and enzyme-directed treatment development. Full article
Show Figures

Figure 1

16 pages, 356 KB  
Review
Mycotoxins and the Intestinal Epithelium: From Barrier Injury to Stem Cell Dysfunction
by Wenying Huo, Yingying Qiao, Xiangru He, Cailing Wang, Ruiqing Li, Long Che and Enkai Li
Toxins 2025, 17(11), 534; https://doi.org/10.3390/toxins17110534 - 30 Oct 2025
Viewed by 783
Abstract
Mycotoxins are toxic secondary metabolites produced by filamentous fungi that contaminate agricultural commodities, posing risks to food safety, animal productivity, and human health. The gastrointestinal tract is the first and most critical site of exposure, where the intestinal epithelium functions as both a [...] Read more.
Mycotoxins are toxic secondary metabolites produced by filamentous fungi that contaminate agricultural commodities, posing risks to food safety, animal productivity, and human health. The gastrointestinal tract is the first and most critical site of exposure, where the intestinal epithelium functions as both a physical and immunological barrier against luminal toxins and pathogens. While extensive research has demonstrated that mycotoxins disrupt epithelial integrity through tight junction impairment, oxidative stress, apoptosis, and inflammation, their effects on the intestinal stem cell (ISC) compartment and epithelial regeneration remain insufficiently understood. This review integrates recent findings from in vivo, cell culture, and advanced 3D intestinal organoid and gut-on-chip models to elucidate how mycotoxins such as deoxynivalenol and zearalenone impair ISC proliferation, alter Wnt/Notch signaling, and compromise mucosal repair. We also discuss dose relevance, species differences, and the modulatory roles of the microbiome and short-chain fatty acids, as well as emerging evidence of additive or synergistic toxicity under co-exposure conditions. By bridging well-established mechanisms of barrier disruption with the emerging concept of ISC-driven regenerative failure, this review identifies a critical knowledge gap in mycotoxin toxicology and highlights the need for integrative models that link epithelial damage to impaired regeneration. Collectively, these insights advance understanding of mycotoxin-induced intestinal dysfunction and provide a foundation for developing nutritional, microbial, and pharmacological strategies to preserve gut integrity and repair. Full article
19 pages, 1106 KB  
Article
Effects of n-3 Long-Chain Polyunsaturated Fatty Acid and Vitamin D Supplementation on Transcriptional Profiles of Human Lung Organoids
by Mina Ali, Martin Steen Mortensen, Ole Bæk, Nicklas Brustad, Tingting Wang, Liang Chen, Min Kim, Casper-Emil Tingskov Pedersen, Trevor D. Lawley, Athanasios Pasias, Jakub Sedzinski, Jakob Stokholm, Klaus Bønnelykke and Bo Chawes
Metabolites 2025, 15(10), 670; https://doi.org/10.3390/metabo15100670 - 14 Oct 2025
Viewed by 595
Abstract
Background/Objectives: Randomized clinical trials (RCTs) suggest that n-3 long-chain polyunsaturated fatty acids (n-3 LCPUFA) and high-dose vitamin D supplementation during pregnancy may protect against childhood asthma. However, the underlying mechanisms remain unclear. Methods: To explore the transcriptional effects of various concentrations of n-3 [...] Read more.
Background/Objectives: Randomized clinical trials (RCTs) suggest that n-3 long-chain polyunsaturated fatty acids (n-3 LCPUFA) and high-dose vitamin D supplementation during pregnancy may protect against childhood asthma. However, the underlying mechanisms remain unclear. Methods: To explore the transcriptional effects of various concentrations of n-3 LCPUFA and vitamin D supplementation on in utero lung development, we cultured human lung organoids derived from BILX and SEHP human-induced pluripotent stem cell lines at the Sanger Institute (Cambridge, UK). The organoids were treated with either no supplementation, or low (0.01 µL/mL) or high (0.1 µL/mL) concentrations of n-3 LCPUFA, as well as no supplementation, or low (5 pM) or high (50 pM) concentrations of vitamin D. Organoids were matured for 50 days, with foregut spheroids embedded in Matrigel and later re-embedded individually to ensure robust growth. We then assessed the impact of these supplementations using RNA sequencing. Results: RNA sequencing of four replicates per condition (36 total samples) revealed that n-3 LCPUFA supplementation had a more substantial impact on gene regulation than vitamin D (differentially expressed genes, n = 907 vs. n = 23). CPT1A and ANGPTL4 genes were highly expressed in media cultured with a high concentration of n-3 LCPUFA, while CYP24A1 was among the highly expressed genes in media cultured with a high concentration of vitamin D. Enrichment analysis showed activation of PPAR pathways, suggesting that n-3 LCPUFA supplementation may protect against asthma by regulating lipid metabolism and inflammation. Conclusions: We identified several genes and pathways that may provide insights into the biological effects of n-3 LCPUFA and vitamin D supplementation on asthma pathophysiology. Full article
Show Figures

Graphical abstract

41 pages, 7020 KB  
Review
Recent Insights into Organoid-Derived Extracellular Vesicles and Their Biomedical Applications
by Ahmed Abdal Dayem, Yeonjoo Kwak, Hyemin Jeun and Ssang-Goo Cho
J. Pers. Med. 2025, 15(10), 492; https://doi.org/10.3390/jpm15100492 - 14 Oct 2025
Viewed by 1771
Abstract
Extracellular vesicles (EVs) play a crucial role in cell-to-cell communication by transporting functionally active molecules, including proteins, lipids, and nucleic acids. While extensive research has focused on EVs generated from traditional two-dimensional (2D) monolayer cultures (2D-EVs), the emergence of three-dimensional (3D) organoid systems [...] Read more.
Extracellular vesicles (EVs) play a crucial role in cell-to-cell communication by transporting functionally active molecules, including proteins, lipids, and nucleic acids. While extensive research has focused on EVs generated from traditional two-dimensional (2D) monolayer cultures (2D-EVs), the emergence of three-dimensional (3D) organoid systems has led to the development of organoid-derived EVs (OEVs), which more closely mimic the physiological conditions of native tissues. In contrast to 2D cultures, 3D systems offer improved EV yield and cargo specificity, enhancing their translational potential. This review discusses the distinctive features of OEVs, including their enhanced tissue relevance, diverse molecular composition, and promising therapeutic applications in areas like disease modeling, regenerative therapies, and targeted drug delivery. We also present an overview of the current organoid-based platforms used to produce OEVs, recent innovations in EV modification and bioengineering, and the practical barriers to their clinical adoption. By comparing the strengths and limitations of OEVs with those of 2D-EVs, we provide a comprehensive perspective on their future role in precision healthcare, biomarker identification, and advanced therapeutic strategies. Full article
(This article belongs to the Section Disease Biomarkers)
Show Figures

Figure 1

36 pages, 1531 KB  
Review
From AI-Assisted In Silico Computational Design to Preclinical In Vivo Models: A Multi-Platform Approach to Small Molecule Anti-IBD Drug Discovery
by Joya Datta Ripa, Sarfaraz Ali, Matt Field, John Smithson and Phurpa Wangchuk
Pharmaceuticals 2025, 18(10), 1536; https://doi.org/10.3390/ph18101536 - 13 Oct 2025
Viewed by 2284
Abstract
Background: Inflammatory Bowel Disease (IBD), including Ulcerative Colitis and Crohn’s Disease, is a multifactorial inflammatory condition of the intestinal tract driven by a complex interplay of genetic factors, immune system dysfunction, and gut microbiota alterations. This review aims to synthesize current advancements [...] Read more.
Background: Inflammatory Bowel Disease (IBD), including Ulcerative Colitis and Crohn’s Disease, is a multifactorial inflammatory condition of the intestinal tract driven by a complex interplay of genetic factors, immune system dysfunction, and gut microbiota alterations. This review aims to synthesize current advancements in modern drug development strategies for IBD. It emphasizes the integration of computational modelling, cell-based experiments, and animal model studies to enhance translational outcomes. Methods: To compile this review, an extensive literature search was performed utilizing PubMed, Scopus, and Google Scholar databases for English-language research and review articles published between 2000 and 2025 using keywords such as “IBD,” “molecular docking,” “bioinformatics,” “organoids,” “animal models,” and “network pharmacology,” among others. A total of 199 peer-reviewed studies were identified for inclusion based on relevance, transparency, and methodological robustness. Results: The review outlines a range of cutting-edge approaches to IBD drug discovery. These include computer modelling, molecular docking, and network analysis to accelerate early-stage target prediction and drug screening. The review further highlights the critical importance of utilizing 2D and 3D cell culture systems in parallel with advanced animal models. It emphasizes the critical integration of computational predictions with biologically relevant in vitro and in vivo validations to improve the reliability and efficiency of drug development. Conclusions: The integration of computer modelling, cell culture systems, and animal studies provides a revolutionary paradigm for accelerating drug discovery to IBD and other diseases enabling personalized and more effective treatment approaches. Full article
(This article belongs to the Collection Feature Review Collection in Medicinal Chemistry)
Show Figures

Graphical abstract

41 pages, 2919 KB  
Review
Organoids as Next-Generation Models for Tumor Heterogeneity, Personalized Therapy, and Cancer Research: Advancements, Applications, and Future Directions
by Ayush Madan, Ramandeep Saini, Nainci Dhiman, Shu-Hui Juan and Mantosh Kumar Satapathy
Organoids 2025, 4(4), 23; https://doi.org/10.3390/organoids4040023 - 8 Oct 2025
Cited by 2 | Viewed by 3027
Abstract
Organoid technology has emerged as a revolutionary tool in cancer research, offering physiologically accurate, three-dimensional models that preserve the histoarchitecture, genetic stability, and phenotypic complexity of primary tumors. These self-organizing structures, derived from adult stem cells, induced pluripotent stem cells, or patient tumor [...] Read more.
Organoid technology has emerged as a revolutionary tool in cancer research, offering physiologically accurate, three-dimensional models that preserve the histoarchitecture, genetic stability, and phenotypic complexity of primary tumors. These self-organizing structures, derived from adult stem cells, induced pluripotent stem cells, or patient tumor biopsies, recapitulate critical aspects of tumor heterogeneity, clonal evolution, and microenvironmental interactions. Organoids serve as powerful systems for modeling tumor progression, assessing drug sensitivity and resistance, and guiding precision oncology strategies. Recent innovations have extended organoid capabilities beyond static culture systems. Integration with microfluidic organoid-on-chip platforms, high-throughput CRISPR-based functional genomics, and AI-driven phenotypic analytics has enhanced mechanistic insight and translational relevance. Co-culture systems incorporating immune, stromal, and endothelial components now permit dynamic modeling of tumor–host interactions, immunotherapeutic responses, and metastatic behavior. Comparative analyses with conventional platforms, 2D monolayers, spheroids, and patient-derived xenografts emphasize the superior fidelity and clinical potential of organoids. Despite these advances, several challenges remain, such as protocol variability, incomplete recapitulation of systemic physiology, and limitations in scalability, standardization, and regulatory alignment. Addressing these gaps with unified workflows, synthetic matrices, vascularized and innervated co-cultures, and GMP-compliant manufacturing will be crucial for clinical integration. Proactive engagement with regulatory frameworks and ethical guidelines will be pivotal to ensuring safe, responsible, and equitable clinical translation. With the convergence of bioengineering, multi-omics, and computational modeling, organoids are poised to become indispensable tools in next-generation oncology, driving mechanistic discovery, predictive diagnostics, and personalized therapy optimization. Full article
Show Figures

Figure 1

20 pages, 4013 KB  
Review
Bioengineering 3D Pancreatic Cancer Models with Fibrotic Stroma for In Vitro Cancer Modeling
by Xingrun Lan, Keke Chen and Xiaoyun Wei
Micromachines 2025, 16(10), 1140; https://doi.org/10.3390/mi16101140 - 2 Oct 2025
Viewed by 1163
Abstract
Pancreatic ductal adenocarcinoma (PDAC) remains highly lethal due to late diagnosis, high malignancy, and profound resistance to therapy. Traditional two-dimensional (2D) cell cultures fail to recapitulate the complex tumor microenvironment (TME), especially the fibrotic stroma, which is crucial for the progression of PDAC [...] Read more.
Pancreatic ductal adenocarcinoma (PDAC) remains highly lethal due to late diagnosis, high malignancy, and profound resistance to therapy. Traditional two-dimensional (2D) cell cultures fail to recapitulate the complex tumor microenvironment (TME), especially the fibrotic stroma, which is crucial for the progression of PDAC and drug response. In vitro three-dimensional (3D) models, which provide more physiologically relevant features such as tight cell–cell and cell-extracellular matrix (ECM) interactions, as well as 3D architecture, have been regarded as highly promising models in PDAC research. This review summarizes some representative in vitro PDAC models, including 3D spheroids, tumor-on-a-chip, bioprinted constructs, and patient-derived organoids (PDOs), particularly focused on the advances in bioengineering strategies for the integration of the key stomal components for microenvironment recapitulation and their applications. Additionally, we discuss the current challenges facing 3D models and propose potential strategies for constructing in vitro models that more accurately simulate the pathophysiology of the fibrotic stroma, aiming for their application in clinical settings. Full article
(This article belongs to the Special Issue 3D Tissue Engineering Techniques and Their Applications)
Show Figures

Figure 1

Back to TopTop