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
remove_circle_outline
remove_circle_outline

Journals

remove_circle_outline
remove_circle_outline
remove_circle_outline
remove_circle_outline
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 (621)

Search Parameters:
Keywords = 3D organoids

Order results
Result details
Results per page
Select all
Export citation of selected articles as:
16 pages, 4363 KB  
Article
Lactoferrin and Osteopontin Cooperatively Promote Intestinal Epithelial Maturation in Neonatal Mice by Activating the Brg1/Notch1/Hes1 Pathway
by Wen Zhang, Chuangang Li, Ran Bi, Yao Lu, Yiran Zhang, Chenhong Shi, Ziyu Qiao, Yanan Sun, Juan Chen, Pengjie Wang, Ran Wang, Fazheng Ren and Yixuan Li
Nutrients 2025, 17(19), 3176; https://doi.org/10.3390/nu17193176 - 8 Oct 2025
Abstract
Background/Objectives: Early life is crucial for infant gut development and intestinal homeostasis. Lactoferrin (LF) and osteopontin (OPN) are bioactive breast milk proteins that are supplemented into infant formula to promote gut development. However, the combined effect of LF and OPN (LOP) on in [...] Read more.
Background/Objectives: Early life is crucial for infant gut development and intestinal homeostasis. Lactoferrin (LF) and osteopontin (OPN) are bioactive breast milk proteins that are supplemented into infant formula to promote gut development. However, the combined effect of LF and OPN (LOP) on in vivo gut maturation has not been fully elucidated. This study investigated the effects of LF, OPN, and LOP on intestinal epithelium maturation in C57BL/6N mice from postnatal days 7 to 21. Methods: 3-day-old pups were assigned to four groups: Control group, LF group: 300 mg/kg LF; OPN group: 300 mg/kg OPN, LOP group: 300 mg/kg of a 1:5 (w/w) mixture of LF and OPN. Results: Compared to controls, LOP reduced plasma Diamine Oxidase (DAO) activity by 1.54-fold and D-lactate levels by 1.41-fold, demonstrating greater efficacy than LF or OPN alone in reducing intestinal permeability. LOP also significantly increased intestinal absorptive cells versus controls or single proteins. Mechanistically, LOP promoted directional intestinal stem cell differentiation, increasing jejunal transit-amplifying cells by 1.40-fold in 21-day-old mice. LOP upregulated expression of the Notch pathway target Hes1 by 1.70-fold. Further investigations revealed LOP activated Notch signaling via the transcription factor Brg1. Validation using intestinal organoids and IEC-6 cells confirmed intact OPN within LOP mediates increased Brg1 expression, activating the Notch pathway to direct intestinal stem cell differentiation into absorptive cells. Conclusions: Collectively, these findings in neonatal mice suggest that LOP cooperatively enhances intestinal barrier maturation and directs stem cell differentiation via Brg1-Notch signaling, offering potential insights for future research on bioactive protein supplementation in infant nutrition. Full article
Show Figures

Figure 1

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
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

0 pages, 1439 KB  
Review
Unlocking the Secrets of the Endometrium: Stem Cells, Niches and Modern Methodologies
by Lijun Huang, Miaoxian Ou, Dunjin Chen and Shuang Zhang
Biomedicines 2025, 13(10), 2435; https://doi.org/10.3390/biomedicines13102435 - 6 Oct 2025
Viewed by 187
Abstract
The endometrium is a highly dynamic tissue central to female reproductive function, undergoing nearly 500 cycles of proliferation, differentiation, shedding, and regeneration throughout a woman’s reproductive life. This remarkable regenerative capacity is driven by a reservoir of endometrial stem/progenitor cells (ESCs), which are [...] Read more.
The endometrium is a highly dynamic tissue central to female reproductive function, undergoing nearly 500 cycles of proliferation, differentiation, shedding, and regeneration throughout a woman’s reproductive life. This remarkable regenerative capacity is driven by a reservoir of endometrial stem/progenitor cells (ESCs), which are crucial for maintaining tissue homeostasis. Dysregulation of these cells is linked to a variety of clinical disorders, including menstrual abnormalities, infertility, recurrent pregnancy loss, and serious gynecological conditions such as endometriosis and endometrial cancer. Recent advancements in organoid technology and lineage-tracing models have provided insights into the complex cellular hierarchy that underlies endometrial regeneration and differentiation. This review highlights the latest breakthroughs in endometrial stem cell biology, focusing particularly on 3D in vitro platforms that replicate endometrial physiology and disease states. By integrating these cutting-edge approaches, we aim to offer new perspectives on the pathogenesis of endometrial disorders and establish a comprehensive framework for developing precision regenerative therapies. Full article
Show Figures

Figure 1

15 pages, 993 KB  
Review
Antioxidants in Cardiovascular Health: Implications for Disease Modeling Using Cardiac Organoids
by Gracious R. Ross and Ivor J. Benjamin
Antioxidants 2025, 14(10), 1202; https://doi.org/10.3390/antiox14101202 - 3 Oct 2025
Viewed by 350
Abstract
Cardiovascular disease remains the leading cause of mortality worldwide, and at its molecular core lies a silent disruptor: oxidative stress. This imbalance between reactive oxygen species (ROS) and antioxidant defenses not only damages cellular components but also orchestrates a cascade of pathological events [...] Read more.
Cardiovascular disease remains the leading cause of mortality worldwide, and at its molecular core lies a silent disruptor: oxidative stress. This imbalance between reactive oxygen species (ROS) and antioxidant defenses not only damages cellular components but also orchestrates a cascade of pathological events across diverse cardiac cell types. In cardiomyocytes, ROS overload impairs contractility and survival, contributing to heart failure and infarction. Cardiac fibroblasts respond by promoting fibrosis through excessive collagen deposition. Macrophages intensify inflammatory responses, such as atherosclerosis, via ROS-mediated lipid oxidation—acting both as mediators of damage and targets for antioxidant intervention. This review examines how oxidative stress affects cardiac cell types and evaluates antioxidant-based therapeutic strategies. Therapeutic approaches include natural antioxidants (e.g., polyphenols and vitamins) and synthetic agents (e.g., enzyme modulators), which show promise in experimental models by improving myocardial remodeling. However, clinical trials reveal inconsistent outcomes, underscoring translational challenges (e.g., clinical biomarkers). Emerging strategies—such as targeted antioxidant delivery, activation of endogenous pathways, and disease modeling using 3D organoids—aim to enhance efficacy. In conclusion, we spotlight innovative technologies—like lab-grown heart tissue models—that help scientists better understand how oxidative stress affects heart health. These tools are bridging the gap between early-stage research and personalized medicine, opening new possibilities for diagnosing and treating heart disease more effectively. 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 266
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

15 pages, 10820 KB  
Article
Wnt/β-Catenin Pathway Activation Confers Fumonisin B1 Tolerance in Chicken Intestinal Organoid Monolayers by Enhancing Intestinal Stem Cell Function
by Shuai Zhang, Yanan Cao, Yiyi Shan, Xueli Zhang, Liangxing Xia, Haifei Wang, Shenglong Wu and Wenbin Bao
Animals 2025, 15(19), 2850; https://doi.org/10.3390/ani15192850 - 29 Sep 2025
Viewed by 266
Abstract
Fumonisin B1 (FB1) is a prevalent mycotoxin in moldy grains and feeds, highly toxic to livestock and compromising product quality while threatening food safety. Poultry exhibit low susceptibility to FB1, but the underlying tolerance mechanisms remain unclear. Traditional 3D chicken intestinal organoid models [...] Read more.
Fumonisin B1 (FB1) is a prevalent mycotoxin in moldy grains and feeds, highly toxic to livestock and compromising product quality while threatening food safety. Poultry exhibit low susceptibility to FB1, but the underlying tolerance mechanisms remain unclear. Traditional 3D chicken intestinal organoid models cannot simulate direct interaction between the epithelial monolayer and FB1, limiting the study of FB1–chicken intestinal crosstalk. Here, we established a 2D chicken intestinal organoid monolayer model, derived from intestinal crypts of 18-day-old specific pathogen-free chicken embryos, to systematically explore poultry’s resistance mechanisms against FB1. Using this model, we compared FB1-induced effects with those in a porcine intestinal epithelial cell model. Results showed that FB1 exposure did not reduce transepithelial electrical resistance, induce abnormal expression of tight junction genes, or cause significant fluctuations in inflammatory factor levels in chicken intestinal organoid monolayers. Mechanistically, FB1 enhances chicken intestinal stem cell function by activating the Wnt/β-catenin pathway, thereby promoting epithelial regeneration and renewal to increase FB1 resistance and decrease toxin sensitivity in chickens. This study reveals a strategy for enhancing FB1 tolerance in poultry by promoting intestinal stem cell function, providing a new perspective for developing mycotoxin prevention and control strategies. Full article
(This article belongs to the Section Poultry)
Show Figures

Figure 1

69 pages, 3282 KB  
Review
Formulation Strategies for Immunomodulatory Natural Products in 3D Tumor Spheroids and Organoids: Current Challenges and Emerging Solutions
by Chang-Eui Hong and Su-Yun Lyu
Pharmaceutics 2025, 17(10), 1258; https://doi.org/10.3390/pharmaceutics17101258 - 25 Sep 2025
Viewed by 692
Abstract
Background/Objectives: Natural products exhibit significant immunomodulatory potential but face severe efficacy loss in three-dimensional (3D) tumor models. This review comprehensively examines the penetration–activity trade-off and proposes integrated strategies for developing effective natural product-based cancer immunotherapies. Methods: We analyzed formulation strategies across three natural [...] Read more.
Background/Objectives: Natural products exhibit significant immunomodulatory potential but face severe efficacy loss in three-dimensional (3D) tumor models. This review comprehensively examines the penetration–activity trade-off and proposes integrated strategies for developing effective natural product-based cancer immunotherapies. Methods: We analyzed formulation strategies across three natural product categories (hydrophobic, macromolecular, stability-sensitive), evaluating penetration enhancement versus activity preservation in spheroids, organoids, and advanced 3D platforms. Results: Tumor spheroids present formidable barriers: dense extracellular matrix (33-fold increased fibronectin), pH gradients (7.4 → 6.5), and extreme cell density (6 × 107 cells/cm3). While nanoparticles, liposomes, and cyclodextrins achieve 3–20-fold penetration improvements, biological activity frequently declines through conformational changes, incomplete release (10–75%), and surface modification interference. Critically, immune cells remain peripheral (30–50 μm), questioning deep penetration pursuit. Patient-derived organoids display 68% predictive accuracy, while emerging vascularized models unveil additional complexity. Food and Drug Administration (FDA) Modernization Act 2.0 enables regulatory acceptance of these advanced models. Conclusions: Effective therapeutic outcomes depend on maintaining immunomodulatory activity in peripherally-located immune cell populations rather than achieving maximum tissue penetration depth. Our five-stage evaluation framework and standardization protocols guide development. Future priorities include artificial intelligence-driven optimization, personalized formulation strategies, and integration of multi-organ platforms to bridge the critical gap between enhanced delivery and therapeutic efficacy. Full article
Show Figures

Figure 1

27 pages, 5128 KB  
Article
Lepidium meyenii Walpers Promotes the Regeneration of Salivary Gland and Prevents Xerostomia After Irradiation Injury
by Yi-Ting Tsai, Yuan-Chuan Lin, Ming-Jen Cheng, Chun-Ming Shih, Chien-Sung Tsai, Ze-Hao Lai, Ching-Yi Wu, Chen-Wei Liu, Feng-Yen Lin and Yi-Wen Lin
Nutrients 2025, 17(19), 3033; https://doi.org/10.3390/nu17193033 - 23 Sep 2025
Viewed by 475
Abstract
Objectives: Lepidium meyenii Walpers (LMW), a high-altitude plant, is known to stimulate hormone release, counteract neurodegeneration, and protect against oxidative stress. Saliva is vital for oral health, and reduced production leads to xerostomia, often caused by aging, radiation, or Sjögren’s syndrome. Key pathological [...] Read more.
Objectives: Lepidium meyenii Walpers (LMW), a high-altitude plant, is known to stimulate hormone release, counteract neurodegeneration, and protect against oxidative stress. Saliva is vital for oral health, and reduced production leads to xerostomia, often caused by aging, radiation, or Sjögren’s syndrome. Key pathological features include mesenchymal fibrosis and acinar atrophy, largely regulated by the TGF-β1 pathway. Current treatments are limited, with many patients relying on artificial saliva. Developing therapies to restore salivary function could offer significant benefits. Methods: In this study, we assessed the protective effects of LMW extract (LMWE) in irradiated C57BL/6J mice and TGF-β1-treated rat parotid acinar cells (Par-C10) using histological, molecular, bioenergetic, and 3D organoid analyses to evaluate salivary gland regeneration and lineage-specific differentiation. Results: LMWE significantly restored gland weight, shortened secretion lag time, and increased amylase activity in irradiated mice. Histological and molecular analyses showed reduced acinar atrophy and fibrosis, preservation of epithelial polarity, and upregulation of Mist1, AQP5, and amylase. In vitro, LMWE protected Par-C10 cells from TGF-β1-induced senescence, preserved mitochondrial membrane potential, and improved epithelial barrier function. In 3D organoid cultures of Par-C10 cells embedded in matrix, (1E,4Z)-1-(2,4-dihydroxyphenyl)-5-(3,4-dihydroxyphenyl) penta-1,4-dien-3-one (DHPPD) and (Z)-N-phenyldodec-2-enamide (E4Z-PD)-selectively enhanced acinar and ductal lineage differentiation, respectively. Conclusions: These results suggest that LMWE promotes salivary gland regeneration through antioxidative and lineage-specific mechanisms and may represent a safe and effective therapeutic strategy for xerostomia. Full article
(This article belongs to the Special Issue Diet and Oral Health)
Show Figures

Figure 1

23 pages, 4959 KB  
Review
Advanced 3D In Vitro Liver Fibrosis Models: Spheroids, Organoids, and Liver-on-Chips
by Jae Eun Lee, Yu-Jeong Lee and Jeong-Kee Yoon
Biomimetics 2025, 10(10), 639; https://doi.org/10.3390/biomimetics10100639 - 23 Sep 2025
Viewed by 634
Abstract
Liver fibrosis (LF) is a progressive and increasingly prevalent condition, yet current therapeutic options remain limited. This underscores the growing demand for advanced three-dimensional (3D) preclinical models that better recapitulate the complex pathophysiology of human LF and overcome the limitations of conventional systems. [...] Read more.
Liver fibrosis (LF) is a progressive and increasingly prevalent condition, yet current therapeutic options remain limited. This underscores the growing demand for advanced three-dimensional (3D) preclinical models that better recapitulate the complex pathophysiology of human LF and overcome the limitations of conventional systems. Although a number of in vitro models have been proposed in recent years, many still rely on two-dimensional (2D) hepatocyte cultures, which fail to represent the multicellular interactions and spatial architecture of the fibrotic liver. In contrast, 3D in vitro models, including spheroids, organoids, and liver-on-a-chip (LoC) platforms, offer more physiologically relevant microenvironments, enabling improved disease modeling and patient-specific drug testing. In this review, we summarize current bioengineering strategies for constructing 3D LF models and highlight their advantages, limitations, and future directions for clinical translation. Full article
(This article belongs to the Special Issue Organ-on-a-Chip Platforms for Drug Delivery and Tissue Engineering)
Show Figures

Figure 1

13 pages, 8472 KB  
Article
Radiation-Induced EMT of Adipose-Derived Stem Cells in 3D Organotypic Culture via Notch Signaling Pathway
by Seon Jeong Choi, Meesun Kim, Kyung Tae Chung and Tae Gen Son
Biology 2025, 14(9), 1306; https://doi.org/10.3390/biology14091306 - 22 Sep 2025
Viewed by 301
Abstract
In our previous study, adipose-derived stem cells (ASCs) cultured in a three-dimensional (3D) organotypic system exhibited mesenchymal-to-epithelial transition (MET) features, including cobblestone morphology and increased expression of E-cadherin and CK18. In this study, we investigated whether ionizing radiation could reverse this phenotype via [...] Read more.
In our previous study, adipose-derived stem cells (ASCs) cultured in a three-dimensional (3D) organotypic system exhibited mesenchymal-to-epithelial transition (MET) features, including cobblestone morphology and increased expression of E-cadherin and CK18. In this study, we investigated whether ionizing radiation could reverse this phenotype via epithelial–mesenchymal transition (EMT) and examined the involvement of Notch signaling. Mouse ASCs were cultured in Matrigel-based 3D organotypic conditions and exposed to 8 Gy of γ-radiation, and EMT- and Notch-related gene and protein expression were assessed 96 h post-irradiation using ATP viability assays, RT-qPCR, and Western blotting. Exposure to 8 Gy significantly reduced cell viability in 2D ASCs to 49.50 ± 6.50% compared with 61.02 ± 5.77% in 3D organoids (p < 0.0001). Irradiated 3D organoids showed EMT-like changes, including an increase of ~2.5-fold in fibronectin and an increase of ~2.0-fold in Twist1 expression, while epithelial CK18 was modestly elevated. Notch signaling was concurrently activated, with Notch1 and Jagged1 increasing by more than twofold and Fra-1 being significantly upregulated. Pretreatment with 20 μM of the γ-secretase inhibitor (GSI) kept cell viability above 90% and suppressed radiation-induced fibronectin, Twist1, Notch1, and Jagged1 expression. These findings indicate that ionizing radiation promotes EMT in 3D-cultured ASCs and reverses prior epithelialization, with Notch signaling playing a key regulatory role. The 3D ASC organoid model may thus provide a physiologically relevant platform for investigating radiation-induced plasticity and potential antifibrotic interventions. Full article
Show Figures

Figure 1

46 pages, 4099 KB  
Review
Hypoxia and Multilineage Communication in 3D Organoids for Human Disease Modeling
by Seif Ehab, Ola A. Gaser and Ahmed Abdal Dayem
Biomimetics 2025, 10(9), 624; https://doi.org/10.3390/biomimetics10090624 - 16 Sep 2025
Viewed by 1030
Abstract
Organoids, self-organizing, three-dimensional (3D) multicellular structures derived from tissues or stem cells, offer physiologically relevant models for studying human development and disease. Compared to conventional two-dimensional (2D) cell cultures and animal models, organoids more accurately recapitulate the architecture and function of human organs. [...] Read more.
Organoids, self-organizing, three-dimensional (3D) multicellular structures derived from tissues or stem cells, offer physiologically relevant models for studying human development and disease. Compared to conventional two-dimensional (2D) cell cultures and animal models, organoids more accurately recapitulate the architecture and function of human organs. Among the critical microenvironmental cues influencing organoid behavior, hypoxia and multilineage communication are particularly important for guiding cell fate, tissue organization, and pathological modeling. Hypoxia, primarily regulated by hypoxia-inducible factors (HIFs), modulates cellular proliferation, differentiation, metabolism, and gene expression, making it a key component in disease modeling. Similarly, multilineage communication, facilitated by intercellular interactions and extracellular matrix (ECM) remodeling, enhances organoid complexity and immunological relevance. This review explores the dynamic interplay between hypoxia and multilineage signaling in 3D organoid-based disease models, emphasizing recent advances in engineering hypoxic niches and co-culture systems to improve preclinical research fidelity. We also discuss their translational implications for drug screening, regenerative medicine, and precision therapies, while highlighting current challenges and future opportunities. By integrating biophysical, biochemical, and computational approaches, next-generation organoid models may be further optimized for translational research and therapeutic innovation. Full article
Show Figures

Figure 1

13 pages, 9662 KB  
Article
Modeling Synucleinopathy Using hESC-Derived Cerebral Organoids
by So Jin Kim, Won Hee Jung, Mu Seog Choe, Ye Seong Jeon and Min Young Lee
Cells 2025, 14(18), 1436; https://doi.org/10.3390/cells14181436 - 15 Sep 2025
Viewed by 556
Abstract
Animal and cellular models harboring SNCA gene mutations have been instrumental in synucleinopathy, but faithful human brain models remain limited. Here, we report the development of a human cerebral organoid (CO) model of synucleinopathy carrying the Ala53Thr mutation in SNCA (SNCAA53T [...] Read more.
Animal and cellular models harboring SNCA gene mutations have been instrumental in synucleinopathy, but faithful human brain models remain limited. Here, we report the development of a human cerebral organoid (CO) model of synucleinopathy carrying the Ala53Thr mutation in SNCA (SNCAA53T). Using a human embryonic stem cell (hESC) line overexpressing SNCAA53T (A53T hESC line), we generated COs (A53T COs) that recapitulate hallmark features of synucleinopathy. These A53T COs exhibited elevated α-synuclein (α-Syn) expression, the increased phosphorylation of α-Syn, and Lewy body-like aggregations. Notably, we also observed the increased expression of phosphorylated tau and neurofibrillary tangle-like silver deposits, although amyloid β expression and accumulation remained unchanged. To evaluate the utility of this model in drug screening, we treated A53T COs with synuclean D (SynD), an inhibitor of α-Syn aggregation, which significantly reduced both α-Syn and tau phosphorylation without affecting total α-Syn levels. Together, our findings establish a robust hESC-derived synucleinopathy CO model harboring the SNCAA53T mutation, demonstrating its potential as a valuable tool for therapeutic drug screening. Full article
(This article belongs to the Special Issue Brain Organoids for Disease Modeling)
Show Figures

Figure 1

30 pages, 7327 KB  
Article
Interaction Between Human Skeletal and Mesenchymal Stem Cells Under Physioxia Enhances Cartilage Organoid Formation: A Phenotypic, Molecular, and Functional Characterization
by Cristian Mera Azain, Astrid Natalia Santamaría Durán, Tatiana Camila Castañeda, Luis Fernando Useche, Efraín Leal Garcia, Jaime Mariño Valero, Rodrigo Jaramillo Quintero, Luis Fernando Jaramillo, Jorge Andrés Franco, Rubiela Castañeda Salazar, Juan Carlos Ulloa, Ivonne Gutiérrez Rojas, Rodrigo Somoza Palacios, Claudia Cuervo Patiño and Viviana Marcela Rodríguez-Pardo
Cells 2025, 14(18), 1423; https://doi.org/10.3390/cells14181423 - 11 Sep 2025
Viewed by 588
Abstract
Articular cartilage regeneration remains a major challenge due to its limited self-repair capacity. Bone marrow-derived skeletal stem cells (SSCs) and mesenchymal stem cells (MSCs) are promising candidates for cartilage engineering, although they differ in their chondrogenic potential. This study explored whether co-culturing SSCs [...] Read more.
Articular cartilage regeneration remains a major challenge due to its limited self-repair capacity. Bone marrow-derived skeletal stem cells (SSCs) and mesenchymal stem cells (MSCs) are promising candidates for cartilage engineering, although they differ in their chondrogenic potential. This study explored whether co-culturing SSCs and MSCs in three-dimensional (3D) organoid systems under cartilage physioxia (5% O2) and chondrogenic induction could improve cartilage tissue formation. SSCs, MSCs, and SSC–MSC co-cultures were characterized for morphology, phenotype, and differentiation capacity. Organoids were generated and cultured for 10 days, followed by analysis of morphology, viability, gene expression (SOX9, RUNX2, ACAN, COL2A1, COL10A1, PRG4, and PDPN), chondrocyte-associated antigens (CD44, CD105, CD146, and PDPN), and cartilage ECM proteins (aggrecan, collagen types I, II, and X, and PRG4). SSCs showed robust chondrogenic and osteogenic potential, while MSCs exhibited a balanced multipotency. Co-culture-derived organoids enhanced chondrogenesis and reduced adipogenesis, with higher expression of cartilage-specific ECM and lower hypertrophic marker levels. These findings highlight the functional synergy between SSCs and MSCs in co-culture, promoting the formation of stable, cartilage-like structures under physioxia. The approach offers a promising strategy for generating preclinical models and advancing regenerative therapies for hyaline cartilage repair. Full article
(This article belongs to the Special Issue Organoids and Models from Stem Cells)
Show Figures

Figure 1

43 pages, 4033 KB  
Review
Updated Applications of Stem Cells in Hypoplastic Left Heart Syndrome
by Rui Xiao, Haleema Darr, Zarif Khan and Qingzhong Xiao
Cells 2025, 14(17), 1396; https://doi.org/10.3390/cells14171396 - 6 Sep 2025
Viewed by 2223
Abstract
Hypoplastic left heart syndrome (HLHS) is a severe congenital heart disease affecting 2–3 neonates every 10,000 live births. While prior research has highlighted associations of HLHS with specific chromosomal abnormalities and genetic mutations, the precise pathophysiology remains elusive. Despite early surgical intervention potentially [...] Read more.
Hypoplastic left heart syndrome (HLHS) is a severe congenital heart disease affecting 2–3 neonates every 10,000 live births. While prior research has highlighted associations of HLHS with specific chromosomal abnormalities and genetic mutations, the precise pathophysiology remains elusive. Despite early surgical intervention potentially allowing most HLHS patients to survive their critical heart disease with a single-ventricle physiology, patients frequently experience complications of arrhythmias and right ventricular heart failure, culminating in the need for an eventual heart transplant. Scarcity of suitable donors combined with limited understanding of mechanisms of development highlights the need for furthering our understanding of HLHS and alternative treatment options. Over the past decades, stem cell research has significantly advanced our understanding of cardiac conditions, repair, development, and therapy, opening the door for a new exciting field of regenerative medicine in cardiology with significant implications for HLHS. This review serves to provide a comprehensive overview of a much focused-on area related to HLHS. Specifically, we will first discuss the key pathophysiological basis and signalling molecules of HLHS. We then outline the emerging role of stem cell-based therapy, with a focus on adult stem cells and pluripotent stem cells (PSCs) in uncovering the pathophysiology of HLHS and optimising future treatment directions. Finally, we will also explore the latest and possible future directions of stem cell-derived techniques such as cardiac organoids and bioengineering cardiac tissues and their utility for investigating disease mechanisms, drug screening, and novel therapy for HLHF. Full article
Show Figures

Figure 1

42 pages, 1890 KB  
Review
Toxicity of Magnetic Nanoparticles in Medicine: Contributing Factors and Modern Assessment Methods
by Julia Nowak-Jary and Beata Machnicka
Int. J. Mol. Sci. 2025, 26(17), 8586; https://doi.org/10.3390/ijms26178586 - 3 Sep 2025
Viewed by 1002
Abstract
With the rapid evolution of nanotechnology, magnetic iron oxide nanoparticles (MNPs)—primarily Fe3O4 and γ-Fe2O3—have gained prominence in biomedicine. Their extensive specific surface area, tunable surface functionalities, and intrinsic magnetic characteristics render them highly versatile for diverse [...] Read more.
With the rapid evolution of nanotechnology, magnetic iron oxide nanoparticles (MNPs)—primarily Fe3O4 and γ-Fe2O3—have gained prominence in biomedicine. Their extensive specific surface area, tunable surface functionalities, and intrinsic magnetic characteristics render them highly versatile for diverse clinical applications, including tumor visualization through Magnetic Resonance Imaging (MRI), radiolabeling, targeted radiotherapy, hyperthermia, gene transfer, drug delivery, Magnetic Particle Imaging (MPI), magnetic blood filtration and theranostic strategies. Nevertheless, ensuring the biocompatibility and non-toxicity of these nanostructures remains a fundamental prerequisite for their medical implementation. Hence, it is essential to continuously refine our understanding of MNP-related toxicity and pursue comprehensive research on this front. This article consolidates up-to-date insights into the evaluation of MNPs’ toxicological profiles, emphasizing the influence of physicochemical properties such as morphology, surface modifications, and electrostatic characteristics, along with operational factors like dosage and administration routes. Traditional toxicity testing strategies, including in vitro assays as first-line screening tools, together with standard ex vivo and in vivo models, are discussed. Special attention is given to the emerging role of New Approach Methodologies (NAMs), such as organoid formation, 3D bioprinting, in ovo chicken embryo assays, and image cytometry. These techniques offer ethical, human-relevant, and informative alternatives to animal testing, supporting more predictive and translationally relevant toxicity assessment of MNPs. Taken together, the integration of conventional assays with innovative NAMs, alongside careful consideration of physicochemical and operational factors, is essential to translate the laboratory promise of MNPs into safe and clinically effective nanomedicines. Full article
(This article belongs to the Special Issue Toxicity of Heavy Metal Compounds)
Show Figures

Figure 1

Back to TopTop