1
|
Di Stefano J, Di Marco F, Cicalini I, FitzGerald U, Pieragostino D, Verhoye M, Ponsaerts P, Van Breedam E. Generation, interrogation, and future applications of microglia-containing brain organoids. Neural Regen Res 2025; 20:3448-3460. [PMID: 39665813 PMCID: PMC11974650 DOI: 10.4103/nrr.nrr-d-24-00921] [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: 08/13/2024] [Revised: 10/29/2024] [Accepted: 11/05/2024] [Indexed: 12/13/2024] Open
Abstract
Brain organoids encompass a large collection of in vitro stem cell-derived 3D culture systems that aim to recapitulate multiple aspects of in vivo brain development and function. First, this review provides a brief introduction to the current state-of-the-art for neuro-ectoderm brain organoid development, emphasizing their biggest advantages in comparison with classical two-dimensional cell cultures and animal models. However, despite their usefulness for developmental studies, a major limitation for most brain organoid models is the absence of contributing cell types from endodermal and mesodermal origin. As such, current research is highly investing towards the incorporation of a functional vasculature and the microglial immune component. In this review, we will specifically focus on the development of immune-competent brain organoids. By summarizing the different approaches applied to incorporate microglia, it is highlighted that immune-competent brain organoids are not only important for studying neuronal network formation, but also offer a clear future as a new tool to study inflammatory responses in vitro in 3D in a brain-like environment. Therefore, our main focus here is to provide a comprehensive overview of assays to measure microglial phenotype and function within brain organoids, with an outlook on how these findings could better understand neuronal network development or restoration, as well as the influence of physical stress on microglia-containing brain organoids. Finally, we would like to stress that even though the development of immune-competent brain organoids has largely evolved over the past decade, their full potential as a pre-clinical tool to study novel therapeutic approaches to halt or reduce inflammation-mediated neurodegeneration still needs to be explored and validated.
Collapse
Affiliation(s)
- Julia Di Stefano
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
- Bio-Imaging Lab, University of Antwerp, Wilrijk, Belgium
| | - Federica Di Marco
- Center for Advanced Studies and Technology (CAST), G. d’Annunzio University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
| | - Ilaria Cicalini
- Center for Advanced Studies and Technology (CAST), G. d’Annunzio University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
| | - Una FitzGerald
- CÚRAM, Center for Research in Medical Devices, Biomedical Engineering, University of Galway, Ireland
- Galway Neuroscience Center, University of Galway, Ireland
| | - Damiana Pieragostino
- Center for Advanced Studies and Technology (CAST), G. d’Annunzio University of Chieti-Pescara, Chieti, Italy
- Department of Innovative Technologies in Medicine and Dentistry, University “G. d’Annunzio” of Chieti-Pescara, Chieti, Italy
| | - Marleen Verhoye
- Bio-Imaging Lab, University of Antwerp, Wilrijk, Belgium
- μNEURO Research Center of Excellence, University of Antwerp, Wilrijk, Belgium
| | - Peter Ponsaerts
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| | - Elise Van Breedam
- Laboratory of Experimental Hematology, Vaccine and Infectious Disease Institute (Vaxinfectio), University of Antwerp, Wilrijk, Belgium
| |
Collapse
|
2
|
Han HJ, Han J, Choi Y, Hwang GJ, Kim S, Ryoo IJ, Kim BY, Soung NK. A novel tubulin inhibitor, No.07, shows anti-cancer and anti-metastatic effects in colon cancer and tumoroids. Life Sci 2025; 372:123644. [PMID: 40252756 DOI: 10.1016/j.lfs.2025.123644] [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: 01/02/2025] [Revised: 04/08/2025] [Accepted: 04/12/2025] [Indexed: 04/21/2025]
Abstract
Colorectal cancer is a highly metastatic disease and the second leading cause of cancer-related death worldwide. Despite the use of various treatment strategies, including chemotherapy and targeted therapy, challenges such as toxicity, drug resistance, and poor response indicate the critical need for new therapeutic agents. Microtubule target agents are one of the major treatment options for chemotherapy in various cancer patients. However, most of these agents are substrates of the MDR1 protein, which leads to the development of multidrug resistance, significantly limiting their effectiveness. Therefore, the development of new drugs is being actively pursued. In this study, we synthesized a novel compound, No.07, which demonstrates significant anti-cancer activity in 3D spheroid models, patient-derived colon cancer organoid models, and mice xenograft models. No.07 directly binds to tubulin dimers, interfering with microtubule polymerization and thereby disrupting tubulin dynamics, ultimately inducing mitotic arrest. Furthermore, No.07 increases mitochondria reactive oxygen species level, leading to the inactivation of the RAF-MEK-ERK signaling cascade, which consequently inhibits metastasis. Notably, Swiss ADME predictions suggest that No.07 is not a substrate of MDR1 and can cross the blood-brain barrier, unlike other microtubule target agents that are limited by MDR1-mediated drug resistance and poor brain penetration. Additionally, experiments using multidrug-resistant cell lines confirmed that No.07 effectively overcomes multidrug resistance, providing a significant improvement over traditionally used chemotherapy agents. In conclusion, No.07 has the potential to address the limitations of existing treatments as a novel therapeutic option.
Collapse
Affiliation(s)
- Ho Jin Han
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Junyeol Han
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; Department of Biomolecular Science, Korea National University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Yerim Choi
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; Department of Biomolecular Science, Korea National University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Gwi-Ja Hwang
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Sumin Kim
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; Department of Biomolecular Science, Korea National University of Science and Technology, Daejeon 34113, Republic of Korea
| | - In-Ja Ryoo
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea
| | - Bo Yeon Kim
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; Department of Biomolecular Science, Korea National University of Science and Technology, Daejeon 34113, Republic of Korea
| | - Nak-Kyun Soung
- Chemical Biology Research Center, Korea Research Institute of Bioscience and Biotechnology, Cheongju 28116, Republic of Korea; Department of Biomolecular Science, Korea National University of Science and Technology, Daejeon 34113, Republic of Korea.
| |
Collapse
|
3
|
Sharin T, Williams KL, Mueller RW, Crump D, O'Brien JM. Avian-Specific Evidence for an Estrogen Receptor Agonism Adverse Outcome Pathway Based on Chicken Embryos and LMH 3D Spheroids Exposed to Ethinylestradiol and Bisphenol A. ENVIRONMENTAL SCIENCE & TECHNOLOGY 2025. [PMID: 40383999 DOI: 10.1021/acs.est.4c10887] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Several adverse outcome pathways (AOPs) describe the effects of endocrine disrupting compounds on estrogen signaling. Substantial data support an AOP related to estrogen receptor (ER) antagonism, leading to decreased fecundity in fish. In this study, data were generated for an ER agonism AOP leading to reduced fecundity in avian species (AOP537). Chicken embryos and the chicken leghorn male hepatoma cell line, LMH, were used to elucidate key events associated with estrogen signaling following exposure to 17α-ethinylestradiol (EE2) and bisphenol A (BPA). Embryos were exposed via egg injection. Viability and hepatic estrogen-responsive gene expression data were collected at midincubation (embryonic day [ED] 11). Changes in plasma vitellogenin (VTG), gonad morphology and growth were evaluated prior to pipping (ED20). Both chemicals dysregulated estrogen-responsive genes in hepatic tissue and increased plasma VTG concentrations. In LMH spheroids, EE2 and BPA altered estrogen-responsive genes and VTG concentrations at 24 and 48 h, respectively. Gonadal histology revealed oocyte-type cells and loss of testicular cords in male embryos exposed to EE2 and BPA. Overall, EE2 and BPA upregulated VTG mRNA expression, increased plasma VTG, and caused impairments in gonadal development. These results contribute avian-specific evidence to support an endocrine disruption AOP describing the relationship between disrupted VTG synthesis and impaired reproduction.
Collapse
Affiliation(s)
- Tasnia Sharin
- National Wildlife Research Centre, Environment and Climate Change Canada, Ottawa K1S 5R2, Canada
| | - Kim L Williams
- National Wildlife Research Centre, Environment and Climate Change Canada, Ottawa K1S 5R2, Canada
| | - Rudolf W Mueller
- Department of Pathology and Laboratory Medicine, University of Ottawa, Ottawa K1H 8M5, Canada
| | - Doug Crump
- National Wildlife Research Centre, Environment and Climate Change Canada, Ottawa K1S 5R2, Canada
| | - Jason M O'Brien
- National Wildlife Research Centre, Environment and Climate Change Canada, Ottawa K1S 5R2, Canada
| |
Collapse
|
4
|
Hans A, Salil, Sawant P, Ajgaonkar B, Jain R, Dandekar P. Cryopreservation of human lung adenocarcinoma spheroids using MMC based cryomixtures. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2025:1-22. [PMID: 40372794 DOI: 10.1080/09205063.2025.2502096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 04/04/2025] [Indexed: 05/17/2025]
Abstract
Cryopreservation remains crucial bottleneck for storing and transporting bioengineered 3D cell models, vital for preclinical drug development and cancer research. Conventional cryoprotectants like fetal bovine serum (FBS) and dimethyl sulfoxide (DMSO) present cytotoxicity challenges and lack efficacy in maintaining structural integrity and viability in complex 3D culture models. This study investigates the efficacy of two carbohydrate-based macromolecular crowders (MMCs), polydextrose III (PD) and resistant maltodextrin (rMD), in cryopreserving human lung adenocarcinoma spheroids as alternatives to FBS. Spheroids were cryopreserved at -80 and -196 °C using MMC-based cryomixtures, with subsequent evaluation of cell viability, structural integrity, and proliferation markers post-thaw. Results indicate that MMC-based cryomixtures, particularly PD, provide superior cryoprotection, preserving the structural and functional integrity of A549 spheroids over a 60-day storage period at -196 °C. Immunocytochemistry of vimentin and Ki67 biomarkers demonstrated that PD-cryopreserved spheroids exhibited consistent structural stability and retained proliferative capacity, contrasting with those stored in conventional FBS-based cryomixtures, which showed marked deterioration in cellular morphology and viability. Apoptosis profiling revealed a lower incidence of cell death in MMC-preserved spheroids, with live cell percentages stabilizing around 50% at -80 °C and approximately 54% at -196 °C over the extended storage period. Further characterization revealed protection of the necrotic core and cellular junctions PD-cryopreserved spheroids. These findings suggest that MMC-based cryomixtures, especially PD, are effective alternatives for cryopreservation of tumor spheroids. The increased cellular viability and structural preservation provided by MMCs could advance their application in 3D culture preservation, addressing limitations of conventional cryopreservation in drug testing, regenerative medicine, and cancer research.
Collapse
Affiliation(s)
- Aakarsh Hans
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| | - Salil
- Department of Biological Sciences and Biotechnology, Institute of Chemical Technology, Mumbai, India
| | - Pooja Sawant
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| | - Bhargavi Ajgaonkar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| | - Ratnesh Jain
- Department of Biological Sciences and Biotechnology, Institute of Chemical Technology, Mumbai, India
| | - Prajakta Dandekar
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Mumbai, India
| |
Collapse
|
5
|
Boix-Montesinos P, Carrascosa-Marco P, Armiñán A, Vicent MJ. Identification of functional biomarkers for personalized nanomedicine in advanced breast cancer in vitro models. J Control Release 2025; 381:113584. [PMID: 40086758 DOI: 10.1016/j.jconrel.2025.113584] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2024] [Revised: 01/30/2025] [Accepted: 02/26/2025] [Indexed: 03/16/2025]
Abstract
Nanomedicines represent promising advanced therapeutics for the enhanced treatment of breast cancer, the primary cause of cancer-related deaths in women; however, the clinical translation of nanomedicines remains challenging. Advanced in vitro models of breast cancer may improve preclinical evaluations and the identification of biomarkers that aid the stratification of patients who would benefit from a given nanomedicine. In this study, we first developed a matrix-embedded breast cancer cell spheroid model representing the extracellular matrix and confirmed the faithful recapitulation of disease aggressiveness in vitro. We then characterized factors influencing nanomedicine drug release (i.e., cathepsin B levels/activity, reactive oxygen species levels, glutathione levels, and cytoplasmic pH values) and evaluated nanomedicine internalization and cytotoxicity evaluation in our spheroid model. We confirmed the reduced-to-oxidized glutathione ratio as a functional biomarker of disulfide linker-containing polypeptide-drug conjugate effectiveness. We then established a biobank of patient-derived breast cancer organoids that recapitulate clinical intra-tumor and inter-tumor heterogeneity as a more advanced model. Analysis in organoids revealed that patient-specific responses to a polypeptide-based nanomedicine correlated with cathepsin B levels, supporting the potential of the functional biomarker for patient-tailored nanomedicine selection. Our findings highlight that exhaustively characterized advanced in vitro models support the evaluation of nanomedicines and the identification of functional biomarkers.
Collapse
Affiliation(s)
- Paz Boix-Montesinos
- Polymer Therapeutics Lab, Centro de Investigación Príncipe Felipe, Av. Eduardo Primo Yúfera 3, Valencia 46012, Spain
| | - Paula Carrascosa-Marco
- Polymer Therapeutics Lab, Centro de Investigación Príncipe Felipe, Av. Eduardo Primo Yúfera 3, Valencia 46012, Spain
| | - Ana Armiñán
- Polymer Therapeutics Lab, Centro de Investigación Príncipe Felipe, Av. Eduardo Primo Yúfera 3, Valencia 46012, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), ISCCIII, Madrid, Spain.
| | - María J Vicent
- Polymer Therapeutics Lab, Centro de Investigación Príncipe Felipe, Av. Eduardo Primo Yúfera 3, Valencia 46012, Spain; Centro de Investigación Biomédica en Red de Cáncer (CIBERONC), ISCCIII, Madrid, Spain.
| |
Collapse
|
6
|
Roy S, Parveen M, Bala A, Sur D. The 3C (Cell Culture, Computer Simulation, Clinical Trial) Solution for Optimizing the 3R (Replace, Reduction, Refine) Framework during Preclinical Research Involving Laboratory Animals. ACS Pharmacol Transl Sci 2025; 8:1188-1204. [PMID: 40370984 PMCID: PMC12070318 DOI: 10.1021/acsptsci.4c00661] [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: 11/11/2024] [Revised: 03/20/2025] [Accepted: 04/01/2025] [Indexed: 05/16/2025]
Abstract
Preclinical research has traditionally utilized laboratory animals to elucidate the safety, tolerability, pharmacokinetics, and pharmacodynamics of new chemical entities prior to human trials. The use of animal models has been pivotal in advancing scientific knowledge and medical breakthroughs, contributing significantly to our understanding of the complex biological processes and human diseases. However, many promising treatments that have demonstrated efficacy in animal studies have failed to translate to human subjects during clinical trials. Consequently, animal testing faces ethical concerns and criticism regarding its predictive reliability for human responses. This has led to the development of 3R principles (Replacement, Reduction, Refinement), introduced in 1959, advocating for alternative methods and improved animal welfare in research. Furthermore, regulatory frameworks and recent legislation, such as the 2022 FDA Modernisation Act, emphasize modern scientific alternatives to traditional animal testing. Emerging approaches, known as the 3Cs-cell culture, computer simulation, and phase 0 clinical trials-offer promising nonanimal solutions that could accelerate drug development and address ethical concerns, potentially rendering preclinical research more humane and efficient.
Collapse
Affiliation(s)
- Susmita Roy
- Haldia
Institute of Pharmacy, ICARE Complex, Haldia Purba Medinipur 721657, India
| | - Mehnaz Parveen
- Division
of Pharmacology, Guru Nanak Institute of
Pharmaceutical Science and Technology, 157/F Nilgunj Road, Panihati, Kolkata 700114, India
| | - Asis Bala
- Pharmacology
and Drug Discovery Research Laboratory, Division of Life Sciences, Institute of Advanced Study in Science and Technology,
an Autonomous Institute under the Department of Science and Technology
(Govt. of India), Vigyan Path, Guwahati, Assam 781035, India
| | - Debjeet Sur
- Division
of Pharmacology, Guru Nanak Institute of
Pharmaceutical Science and Technology, 157/F Nilgunj Road, Panihati, Kolkata 700114, India
| |
Collapse
|
7
|
Alavi R, Chancy O, Trudel B, Dewit L, Luthold C, Piquet L, Akbarzadeh A, Desjardins M, Landreville S, Bordeleau F. Quantitative polarization microscopy as a potential tool for quantification of mechanical stresses within 3D matrices. Acta Biomater 2025:S1742-7061(25)00306-X. [PMID: 40348695 DOI: 10.1016/j.actbio.2025.04.052] [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: 04/02/2024] [Revised: 04/21/2025] [Accepted: 04/28/2025] [Indexed: 05/14/2025]
Abstract
3D mechanical stresses within tissues/extracellular matrices (ECMs) play a significant role in pathological and physiological processes, making their quantification a necessary step to understand the mechanobiological phenomena. Unfortunately, it is rather challenging to quantify these 3D mechanical stresses due to the highly nonlinear and heterogeneous nature of the fibrous matrix. A number of techniques have been developed to address this challenge, including 3D traction force microscopy (TFM), micropillar devices or microparticle-based force sensors; yet, these techniques come with certain drawbacks. Here, we are presenting quantitative polarization microscopy (QPOL) as a non-invasive and label-free technique to quantify mechanical stresses in 3D matrix without a necessity to assume a matrix material model. Taking collagen as a birefringent material, we demonstrated the correlation between the retardance signals obtained by QPOL and the mechanical parameters associated with the 3D collagen hydrogel, i.e. applied external forces and maximum shear stresses. Using cantilever-collagen systems wherein cantilevers applied external loads on the collagen hydrogel, we showed that the retardance signal within loaded collagen positively correlated with the applied load. Also, the retardance signal values within the collagen hydrogel correlated with the maximum shear stress values derived from computational finite element (FE) models. Finally, we obtained the retardance signals around the spheroids of different contractility levels embedded in collagen hydrogel, and the retardance distribution around the spheroids reflected the stress distribution and applied force. This study provides the framework to use QPOL as a tool for quantification of mechanical stresses within 3D ECM. STATEMENT OF SIGNIFICANCE: Mechanical stresses within the 3D extracellular matrix play an important role during physiological and pathological processes. Quantification of such 3D forces is paramount to our understanding of such phenomena and potentially developing therapeutic interventions based on mechanobiological status of the disease. The existing approaches to quantify these 3D mechanical stresses face certain drawbacks such as high computational cost or introduce discontinuities and alteration within the natural 3D microenvironment of the cells. Here, we provide the framework to use quantitative polarization microscopy (QPOL) as an optical-based, non-invasive and computationally efficient technique to quantify the 3D mechanical stresses within the 3D matrix.
Collapse
Affiliation(s)
- Reza Alavi
- Centre de recherche sur le cancer, Université Laval, Québec, QC, Canada; Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Université Laval, Québec, QC, Canada; Oncology division, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Olivier Chancy
- Centre de recherche sur le cancer, Université Laval, Québec, QC, Canada; Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Université Laval, Québec, QC, Canada; Oncology division, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Regenerative Medicine division, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Benjamin Trudel
- Centre de recherche sur le cancer, Université Laval, Québec, QC, Canada; Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Université Laval, Québec, QC, Canada; Oncology division, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Louise Dewit
- Oncology division, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Carole Luthold
- Oncology division, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Léo Piquet
- Centre de recherche sur le cancer, Université Laval, Québec, QC, Canada; Oncology division, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Regenerative Medicine division, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada
| | - Abdolhamid Akbarzadeh
- AM(3)L Laboratory, Department of Bioresource Engineering, McGill University, Island of Montreal, QC, Canada; Department of Mechanical Engineering, McGill University, Montreal, QC, Canada
| | - Michèle Desjardins
- Oncology division, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Department of Physics, Physics Engineering and Optics, Faculty of Sciences and Engineering, Université Laval, Québec, QC, Canada
| | - Solange Landreville
- Centre de recherche sur le cancer, Université Laval, Québec, QC, Canada; Oncology division, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Regenerative Medicine division, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Department of Ophthalmology and Otorhinolaryngology-Cervico-Facial Surgery, Faculty of Medicine, Université Laval, Québec, QC, Canada
| | - François Bordeleau
- Centre de recherche sur le cancer, Université Laval, Québec, QC, Canada; Centre de recherche en organogénèse expérimentale de l'Université Laval/LOEX, Université Laval, Québec, QC, Canada; Oncology division, Centre de recherche du CHU de Québec-Université Laval, Québec, QC, Canada; Department of Molecular Biology, Medical Biochemistry and Pathology, Faculty of Medicine, Université Laval, Québec, QC, Canada.
| |
Collapse
|
8
|
Rahman MS, Qi G, Li C, Li Y, Wang W, Atala A, Sun XS. Differential Effects of Wheat Bran Antioxidants on the Growth Dynamics of Human Cancer Cells. Foods 2025; 14:1633. [PMID: 40361715 PMCID: PMC12071416 DOI: 10.3390/foods14091633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2025] [Revised: 04/10/2025] [Accepted: 04/27/2025] [Indexed: 05/15/2025] Open
Abstract
Wheat bran, rich in phenolic compounds like ferulic acid, possesses notable antioxidant properties that may contribute to cancer treatment strategies. This study examined the effects of hydrolyzed arabinoxylan oligomers (HAOs) linked with ferulic acid from hard wheat bran on three human cancer cell lines: colon cancer (SW480), liver cancer (HepG2), and cervical cancer (HeLa). Cells were cultured in a three-dimensional (3D) 0.5% PGS matrix and exposed to varying concentrations (100, 500, and 1000 μg/mL) of wheat bran antioxidants (WBA) extracts. Results show that WBA inhibited growth of SW480 cells, significantly reducing spheroid expansion and promoting dehydration. In contrast, HepG2 cells exhibited increased growth under WBA treatment, suggesting a non-toxic, growth-enhancing effect. No significant changes were observed in HeLa cell growth, with cell viability remaining high across all treatments. These findings highlight the selective influence of WBA on cancer cell behavior, underscoring its potential for targeted, personalized cancer therapies. This study provides valuable insights into the application of antioxidant-rich compounds for modulating specific cancer cell dynamics, paving the way for novel therapeutic approaches.
Collapse
Affiliation(s)
- Md Sharifur Rahman
- Department of Biological and Agricultural Engineering, Kansas State University, Manhattan, KS 66506, USA;
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA; (G.Q.); (A.A.)
| | - Guangyan Qi
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA; (G.Q.); (A.A.)
| | - Cheng Li
- Department of Grain Science and Industry, Kansas State University, Manhattan, KS 66506, USA; (C.L.); (Y.L.)
| | - Yonghui Li
- Department of Grain Science and Industry, Kansas State University, Manhattan, KS 66506, USA; (C.L.); (Y.L.)
| | - Weiqun Wang
- Department of Food, Nutrition, Dietetics and Health, Kansas State University, Manhattan, KS 66506, USA;
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA; (G.Q.); (A.A.)
| | - Xiuzhi Susan Sun
- Department of Biological and Agricultural Engineering, Kansas State University, Manhattan, KS 66506, USA;
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC 27101, USA; (G.Q.); (A.A.)
| |
Collapse
|
9
|
Johnston EK, Fang Z, Soto-Gutierrez A, Taner CB, Cook KE, Yang L, Abbott RD. Engineering a three-dimensional liver steatosis model. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167888. [PMID: 40328412 DOI: 10.1016/j.bbadis.2025.167888] [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: 07/25/2024] [Revised: 04/28/2025] [Accepted: 05/02/2025] [Indexed: 05/08/2025]
Abstract
Liver transplantation is the key treatment for liver failure, yet organ scarcity, exacerbated by high discard rates of steatotic livers, leads to high waitlist mortality. Preclinical models of steatosis are necessary to understand the pathophysiology of the disease and to develop pharmacological interventions to decrease disease burden and liver discard rate. In this paper, we develop an expedited 3D steatotic organoid model containing primary human hepatocytes and non-parenchymal cells. We present our iterative approach as we transition from 2D to 3D models and from immortalized to primary cells to optimize conditions for the development of a 3D human steatosis model. Both primary cell aggregation and steatosis induction time were reduced from the standard, 5-7 days, to 2 days. Our 3D model incorporates human primary hepatocytes from discarded liver tissues, which have not been used in organoids previously due to their rapid loss of phenotype in culture. After optimizing our steatosis induction media there was a mix of macro- and micro-steatosis in these primary hepatocytes which is consistent with the human pathology. Our approach achieves a model reflective of the liver pathology, preserving cellular phenotypes and viability while exhibiting markers of oxidative stress, a key factor contributing to complications in the transplantation of steatotic livers.
Collapse
Affiliation(s)
- Elizabeth K Johnston
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Zhou Fang
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | | | - C Burcin Taner
- Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Keith E Cook
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Liu Yang
- Department of Transplantation, Mayo Clinic, Jacksonville, FL 32224, USA
| | - Rosalyn D Abbott
- Department of Biomedical Engineering, Carnegie Mellon University, Pittsburgh, PA 15213, USA.
| |
Collapse
|
10
|
Hockaden N, Leriger G, Wang J, Ray H, Chakrabarti S, Downing N, Desmond J, Williams D, Hollenhorst PC, Longmore G, Carpenter RL. Amyloidogenesis promotes HSF1 activity enhancing cell survival during breast cancer metastatic colonization. Cell Stress Chaperones 2025; 30:143-159. [PMID: 40147541 PMCID: PMC12002613 DOI: 10.1016/j.cstres.2025.03.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 02/10/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025] Open
Abstract
Breast cancer is the most commonly diagnosed cancer among women and the second leading cause of cancer deaths in women. A majority of these breast cancer deaths are due to metastasis, which occurs when primary tumor cells invade into the blood stream to travel and colonize at distant organ sites. Metastatic colonization is the rate-limiting step of metastasis. Heat shock factor 1 (HSF1) is a transcription factor that has been shown to be involved in promoting malignancy with a function in metastatic dissemination due to its contribution to promoting epithelial-to-mesenchymal transition. The role of HSF1 in colonization is unclear. In this study, we observed that HSF1 was essential for metastatic colonization. Consistent with these findings, we also observed that HSF1 was more active in human metastatic tumors compared to primary tumors. HSF1 was also seen to be activated during in vitro colony formation, which was accompanied by increases in amyloid beta (Aβ) fibrils, which was also observed in human metastatic tumors. Aβ fibrils led to HSF1 activation and depletion or inhibition of HSF1 led to increases in Aβ fibrils. HSF1 inhibition with small molecule inhibitors suppressed in vitro colony formation and mammosphere growth of metastatic breast cancer cells. These results suggest that colonization increases Aβ fibril formation that subsequently activates HSF1 as a cell survival mechanism that is essential for metastatic initiation and outgrowth.
Collapse
Affiliation(s)
| | - Gabi Leriger
- Medical Sciences, Indiana University, Bloomington, IN 47405
| | - John Wang
- Medical Sciences, Indiana University, Bloomington, IN 47405
| | - Haimanti Ray
- Medical Sciences, Indiana University, Bloomington, IN 47405
| | | | | | - Jacob Desmond
- Department of Chemistry, Indiana University, Bloomington, IN 47405
| | - David Williams
- Department of Chemistry, Indiana University, Bloomington, IN 47405
| | - Peter C Hollenhorst
- Medical Sciences, Indiana University, Bloomington, IN 47405; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202; Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202
| | - Gregory Longmore
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110
| | - Richard L Carpenter
- Medical Sciences, Indiana University, Bloomington, IN 47405; Department of Biochemistry and Molecular Biology, Indiana University School of Medicine, Indianapolis, IN 46202; Simon Comprehensive Cancer Center, Indiana University School of Medicine, Indianapolis, IN 46202.
| |
Collapse
|
11
|
Joshi T, Chan YO, Qiao Z, Kheirandish-Gozal L, Gozal D, Khalyfa A. Circulating exosomes in pediatric obstructive sleep apnea with or without neurocognitive deficits and their effects on a 3D-blood-brain barrier spheroid model. Exp Neurol 2025; 387:115188. [PMID: 39986553 DOI: 10.1016/j.expneurol.2025.115188] [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: 10/19/2024] [Revised: 01/31/2025] [Accepted: 02/16/2025] [Indexed: 02/24/2025]
Abstract
Obstructive sleep apnea (OSA) in children is linked to cognitive impairments, potentially due to blood-brain barrier (BBB) dysfunction. Exosomes, small vesicles released by most cells, reflect cellular changes. This study examined the effects of exosomes from children with OSA, with or without cognitive deficits, on neurovascular unit (NVU) models. Twenty-six children were categorized into three groups: healthy controls (Cont, n = 6), OSA without cognitive deficits (OSA-NG, n = 10), and OSA with neurocognitive deficits (OSA-POS, n = 10). Plasma exosomes were characterized and applied to human 3D NVU spheroids for 24 h. Barrier integrity, permeability, and angiogenesis were assessed using trans-endothelial electrical resistance (TEER), tight junction integrity, and tube formation assays. Single-nucleus RNA sequencing (snRNA-seq) and bioinformatics, including CellChat analysis, identified intercellular signaling pathways. Results showed that exosomes from OSA-POS children disrupted TEER, increased permeability, and impaired ZO1 staining in spheroids, compared to the other groups. Both OSA-POS and OSA-NG exosomes increased permeability in NVU cells in monolayer and microfluidic BBB models. snRNA-seq analysis further revealed distinct cell clusters and pathways associated with the different groups. This 3D NVU spheroid model provides a robust platform to study BBB properties and the role of exosomes in OSA. These findings suggest that integrating snRNA-seq with exosome studies can uncover mechanisms underlying neurocognitive dysfunction in pediatric OSA, potentially leading to personalized therapeutic approaches.
Collapse
Affiliation(s)
- Trupti Joshi
- MU Institute for Data Science and Informatics, University of Missouri, Columbia, MO, USA; Christophers S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.
| | - Yen On Chan
- MU Institute for Data Science and Informatics, University of Missouri, Columbia, MO, USA; Christophers S. Bond Life Sciences Center, University of Missouri, Columbia, MO, USA.
| | - Zhuanhong Qiao
- Department of Neurology, School of Medicine, University of Missouri, Columbia, MO, USA
| | | | - David Gozal
- Department of Pediatrics, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, USA
| | - Abdelnaby Khalyfa
- Department of Biomedical Sciences, Joan C. Edwards School of Medicine, Marshall University, Huntington, West Virginia, United States of America.
| |
Collapse
|
12
|
Liu K, Wang H, Wang L, Ma W, Yang J, Li C, Liu J, Bao W, Li L, Du Y, Gao H. Benzeneboronic acid-modified hyaluronic acid hydrogel enhances the differentiation of dorsal root ganglion stem cells in a three-dimensional environment. Int J Biol Macromol 2025; 309:142786. [PMID: 40185459 DOI: 10.1016/j.ijbiomac.2025.142786] [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: 01/15/2025] [Revised: 03/29/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Peripheral nerve injuries (PNI) remain challenging to treat due to limited regeneration capacity and the lack of effective therapeutic scaffolds to support nerve repair. This study aims to develop and evaluate a 3-aminophenylboronic acid-modified hyaluronic acid (HAB) hydrogel as a 3D scaffold to enhance Dorsal root ganglion-derived stem cells (DRGSCs) attachment, migration, and neuronal differentiation for peripheral nerve regeneration. The HAB hydrogel was synthesized through an amidation reaction and characterized using Fourier transform infrared spectroscopy (FTIR) and 1H nuclear magnetic resonance (1H NMR). DRGSCs were cultured in HAB hydrogel, and neuronal differentiation was assessed through immunofluorescence staining, PCR, and multi-electrode array (MEA) recordings. Cytotoxicity, proliferation, and in vivo biocompatibility were evaluated through live/dead staining, CCK-8 assays, and subcutaneous implantation in rats. Transcriptomic analysis was performed to explore gene expression profiles. Our results shown that DRGSCs cultured in HAB hydrogel exhibited significantly improved attachment (78.5 % ± 3.2 % vs. 45.3 % ± 2.8 %, p < 0.05) and migration speeds (21.4 μm/h vs. 12.9 μm/h, p < 0.05) compared to 2D cultures. Neuronal differentiation efficiency, as indicated by Tuj1-positive cells, was also higher (72.6 % ± 4.1 % vs. 42.8 % ± 3.9 %, p < 0.01). RNA sequencing identified 990 differentially expressed genes (627 upregulated, 363 downregulated), with pathways involved in synaptic vesicle cycling, glutamatergic and GABAergic synapses significantly enriched (p < 0.05). Validation revealed that the expression trends of Gnao1 and Grm7 in the plastic petri dish and HAB hydrogel groups were consistent with the RNA sequencing results. In vivo, the hydrogel showed excellent biocompatibility, with reduced TNF-α and IL-1β expression over a 28-day degradation cycle (p < 0.01). The HAB hydrogel provides a supportive 3D microenvironment that enhances DRGSCs differentiation and electrophysiological activity, highlighting its potential as a promising scaffold for peripheral nerve regeneration and neuroregenerative medicine.
Collapse
Affiliation(s)
- Kuangpin Liu
- College of Rehabilitation, Kunming Medical University, Kunming 650500, China
| | - Hailei Wang
- Hepatic Surgery, Affiliated Calmette Hospital of Kunming Medical University, Kunming 650500, China
| | - Le Wang
- Department of Clinical Laboratory, Qingdao Central Hospital, University of Health and Rehabilitation Sciences, Qingdao 266000, China
| | - Wei Ma
- Institute of Neuroscience, Kunming Medical University, Kunming 650500, China
| | - Jinwei Yang
- Second Department of General Surgery, First People's Hospital of Yunnan Province, Kunming 650032, China
| | - Chunyan Li
- Neurology Department, The Second Affiliated Hospital of Kunming Medical University, Kunming 650032, China
| | - Jinhua Liu
- College of Rehabilitation, Kunming Medical University, Kunming 650500, China
| | - Wenli Bao
- College of Rehabilitation, Kunming Medical University, Kunming 650500, China
| | - Liyan Li
- Institute of Neuroscience, Kunming Medical University, Kunming 650500, China.
| | - Yan Du
- College of Information Engineering and Automation, Kunming University of Science and Technology, Kunming 650032, China.
| | - Hongqiang Gao
- Hepatic Surgery, Affiliated Calmette Hospital of Kunming Medical University, Kunming 650500, China.
| |
Collapse
|
13
|
Yilmaz EG, Hacıosmanoğlu N, Jordi SBU, Yilmaz B, Inci F. Revolutionizing IBD research with on-chip models of disease modeling and drug screening. Trends Biotechnol 2025; 43:1062-1078. [PMID: 39523166 DOI: 10.1016/j.tibtech.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Revised: 08/30/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024]
Abstract
Inflammatory bowel disease (IBD) comprises chronic inflammatory conditions with complex mechanisms and diverse manifestations, posing significant clinical challenges. Traditional animal models and ethical concerns in human studies necessitate innovative approaches. This review provides an overview of human intestinal architecture in health and inflammation, emphasizing the role of microfluidics and on-chip technologies in IBD research. These technologies allow precise manipulation of cellular and microbial interactions in a physiologically relevant context, simulating the intestinal ecosystem microscopically. By integrating cellular components and replicating 3D tissue architecture, they offer promising models for studying host-microbe interactions, wound healing, and therapeutic approaches. Continuous refinement of these technologies promises to advance IBD understanding and therapy development, inspiring further innovation and cross-disciplinary collaboration.
Collapse
Affiliation(s)
- Eylul Gulsen Yilmaz
- UNAM-National Nanotechnology Research Center, Bilkent University, 06800 Ankara, Turkey; Institute of Materials Science and Nanotechnology, Bilkent University, 06800 Ankara, Turkey
| | - Nedim Hacıosmanoğlu
- UNAM-National Nanotechnology Research Center, Bilkent University, 06800 Ankara, Turkey; Institute of Materials Science and Nanotechnology, Bilkent University, 06800 Ankara, Turkey
| | - Sebastian Bruno Ulrich Jordi
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, 3010, Bern, Switzerland; Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, 3008, Bern, Switzerland
| | - Bahtiyar Yilmaz
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, 3010, Bern, Switzerland; Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, 3008, Bern, Switzerland.
| | - Fatih Inci
- UNAM-National Nanotechnology Research Center, Bilkent University, 06800 Ankara, Turkey; Institute of Materials Science and Nanotechnology, Bilkent University, 06800 Ankara, Turkey.
| |
Collapse
|
14
|
Hanitrarimalala V, Prgomet Z, Hedhammar M, Tassidis H, Wingren AG. In vitro 3D modeling of colorectal cancer: the pivotal role of the extracellular matrix, stroma and immune modulation. Front Genet 2025; 16:1545017. [PMID: 40376304 PMCID: PMC12078225 DOI: 10.3389/fgene.2025.1545017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Accepted: 04/23/2025] [Indexed: 05/18/2025] Open
Abstract
Colorectal cancer (CRC) is a leading global cancer with high mortality, especially in metastatic cases, with limited therapeutic options. The tumor microenvironment (TME), a network comprising various immune cells, stromal cells and extracellular (ECM) components plays a crucial role in influencing tumor progression and therapy outcome. The genetic heterogeneity of CRC and the complex TME complicates the development of effective, personalized treatment strategies. The prognosis has slowly improved during the past decades, but metastatic CRC (mCRC) is common among patients and is still associated with low survival. The therapeutic options for CRC differ from those for mCRC and include surgery (mostly for CRC), chemotherapy, growth factor receptor signaling pathway targeting, as well as immunotherapy. Malignant CRC cells are established in the TME, which varies depending on the primary or metastatic site. Herein, we review the role and interactions of several ECM components in 3D models of CRC and mCRC tumor cells, with an emphasis on how the TME affects tumor growth and treatment. This comprehensive summary provides support for the development of 3D models that mimic the interactions within the TME, which will be essential for the development of novel anticancer therapies.
Collapse
Affiliation(s)
- Veroniaina Hanitrarimalala
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden
- Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden
| | - Zdenka Prgomet
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden
- Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden
| | - My Hedhammar
- KTH Royal Institute of Technology, Division of Protein Technology, Stockholm, Sweden
| | - Helena Tassidis
- Department of Bioanalysis, Faculty of Natural Sciences, Kristianstad University, Kristianstad, Sweden
| | - Anette Gjörloff Wingren
- Department of Biomedical Sciences, Faculty of Health and Society, Malmö University, Malmö, Sweden
- Biofilms-Research Center for Biointerfaces, Malmö University, Malmö, Sweden
- Department of Bioanalysis, Faculty of Natural Sciences, Kristianstad University, Kristianstad, Sweden
| |
Collapse
|
15
|
Sleep SL, Ranjit E, Gunter J, Hryciw DH, Arany P, George R. Mitochondrial Oxygen Consumption and Immunocytochemistry of Human Dental Pulp Stem Cell Following 808 nm PBM Therapy: A 3D Cell Culture Study. JOURNAL OF BIOPHOTONICS 2025:e70051. [PMID: 40294940 DOI: 10.1002/jbio.70051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 03/27/2025] [Accepted: 04/14/2025] [Indexed: 04/30/2025]
Abstract
This study investigated the impact of 808 nm laser photobiomodulation (PBM) on mitochondrial respiration and osteogenic protein expression (OCN, OPN, ALP, RUNX2, COL-1, BMP-2) in human dental pulp stem cells (hDPSCs) within a 3D hydrogel model. hDPSCs were isolated from third molars and maintained under hypoxic conditions. Cells received PBM at 5 and 15 J/cm2 using an 808 nm diode laser. The study showed that 808 nm PBM can alter mitochondrial respiration, with 5 J/cm2 enhancing osteogenic protein expression (OCN, ALP, OPN, RUNX2) but failing to sustain BMP-2 at 24 h. In contrast, 15 J/cm2 induced stronger upregulation and prolonged BMP-2 expression, suggesting an optimal dose for sustained osteogenic activity. BMP-2 was later downregulated, and COL-1 remained unchanged post-PBM. Importantly, this study indicates the dose-specific PBM modulation of mitochondrial respiration and protein expression, but further research is required to optimize treatment protocols.
Collapse
Affiliation(s)
- Simone L Sleep
- School of Medicine and Dentistry, Griffith University, Southport, Australia
| | - Eliza Ranjit
- School of Medicine and Dentistry, Griffith University, Southport, Australia
| | - Jennifer Gunter
- Australian Prostate Cancer Research Centre, QLD, Centre for Genomics and Personalised Health, School of Biomedical Science, Queensland University of Technology, Translational Research Institute, Brisbane, Australia
| | - Deanne H Hryciw
- School of Environment and Science, Griffith University, Nathan, Australia
- Institute for Biomedicine and Glycomics, Griffith University, Nathan, Australia
| | - Praveen Arany
- Oral Biology, Surgery, and Biomedical Engineering, University at Buffalo, Buffalo, New York, USA
| | - Roy George
- School of Medicine and Dentistry, Griffith University, Southport, Australia
| |
Collapse
|
16
|
Diao XJ, Soto C, Wang F, Wang Y, Wu YC, Mukherjee A. The potential of brain organoids in addressing the heterogeneity of synucleinopathies. Cell Mol Life Sci 2025; 82:188. [PMID: 40293500 PMCID: PMC12037466 DOI: 10.1007/s00018-025-05686-w] [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: 09/16/2024] [Revised: 03/26/2025] [Accepted: 03/30/2025] [Indexed: 04/30/2025]
Abstract
Synucleinopathies are a group of diseases characterized by neuronal and glial accumulation of α-synuclein (aSyn) linked with different clinical presentations, including Parkinson's disease (PD), Parkinson's disease with dementia (PDD), Dementia with Lewy Bodies (DLB) and Multiple system atrophy (MSA). Interestingly, the structure of the aSyn aggregates can vary across different synucleinopathies. Currently, it is unclear how the aSyn protein can aggregate into diverse structures and affect distinct cell types and various brain regions, leading to different clinical symptoms. Recent advances in induced pluripotent stem cells (iPSCs)-based brain organoids (BOs) technology provide an unprecedented opportunity to define the etiology of synucleinopathies in human brain cells within their three-dimensional (3D) context. In this review, we will summarize current advances in investigating the mechanisms of synucleinopathies using BOs and discuss the scope of this platform to define mechanisms underlining the selective vulnerability of cell types and brain regions in synucleinopathies.
Collapse
Affiliation(s)
- Xiao-Jun Diao
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Claudio Soto
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Fei Wang
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA
| | - Yu Wang
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yun-Cheng Wu
- Department of Neurology, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.
| | - Abhisek Mukherjee
- Mitchell Center for Alzheimer's Disease and Related Brain Disorders, Department of Neurology, McGovern Medical School at the University of Texas Health Science Center at Houston, Houston, TX, USA.
| |
Collapse
|
17
|
Hsieh HC, Han Q, Brenes D, Bishop KW, Wang R, Wang Y, Poudel C, Glaser AK, Freedman BS, Vaughan JC, Allbritton NL, Liu JTC. Imaging 3D cell cultures with optical microscopy. Nat Methods 2025:10.1038/s41592-025-02647-w. [PMID: 40247123 DOI: 10.1038/s41592-025-02647-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2024] [Accepted: 01/16/2025] [Indexed: 04/19/2025]
Abstract
Three-dimensional (3D) cell cultures have gained popularity in recent years due to their ability to represent complex tissues or organs more faithfully than conventional two-dimensional (2D) cell culture. This article reviews the application of both 2D and 3D microscopy approaches for monitoring and studying 3D cell cultures. We first summarize the most popular optical microscopy methods that have been used with 3D cell cultures. We then discuss the general advantages and disadvantages of various microscopy techniques for several broad categories of investigation involving 3D cell cultures. Finally, we provide perspectives on key areas of technical need in which there are clear opportunities for innovation. Our goal is to guide microscope engineers and biomedical end users toward optimal imaging methods for specific investigational scenarios and to identify use cases in which additional innovations in high-resolution imaging could be helpful.
Collapse
Affiliation(s)
- Huai-Ching Hsieh
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Qinghua Han
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - David Brenes
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Kevin W Bishop
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Rui Wang
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA
| | - Yuli Wang
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Chetan Poudel
- Department of Chemistry, University of Washington, Seattle, WA, USA
| | - Adam K Glaser
- Allen Institute for Neural Dynamics, Seattle, WA, USA
| | - Benjamin S Freedman
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Medicine, Division of Nephrology, Kidney Research Institute and Institute for Stem Cell and Regenerative Medicine, Seattle, WA, USA
- Plurexa LLC, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Joshua C Vaughan
- Department of Chemistry, University of Washington, Seattle, WA, USA
- Department of Physiology and Biophysics, University of Washington, Seattle, WA, USA
| | - Nancy L Allbritton
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Jonathan T C Liu
- Department of Bioengineering, University of Washington, Seattle, WA, USA.
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA.
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA.
| |
Collapse
|
18
|
Zhu X, Zhao Y, Bai X, Dong Q, Tian C, Sun R, Yan C, Ruan J, Liu Z, Gao J. Small molecules direct the generation of ameloblast-like cells from human embryonic stem cells. Stem Cell Res Ther 2025; 16:173. [PMID: 40221796 PMCID: PMC11993985 DOI: 10.1186/s13287-025-04294-6] [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: 11/06/2024] [Accepted: 03/25/2025] [Indexed: 04/14/2025] Open
Abstract
BACKGROUND Ameloblasts present a promising avenue for the investigation of enamel and tooth regeneration. Previous protocols for directing the differentiation of human embryonic stem cells (hESCs) into dental epithelial (DE) cells involving the need for additional cells, conditional medium, and the use of costly cytokines. Importantly, ameloblasts have not been generated from hESCs in previous studies. Hence, we aimed to identify defined differentiation conditions that would solely utilize small molecules to achieve the production of ameloblasts. METHODS We developed a three-step strategy entailing the progression of hESCs through non-neural ectoderm (NNE) and DE to generate functional ameloblasts in vitro. Initially, the NNE fate was induced from hESCs using a 6-day differentiation protocol with 1 µmol/L Retinoic acid (RA). Subsequently, the NNE lineage was differentiated into DE by employing a combination of 1 µmol/L LDN193189 (a BMP signaling inhibitor) and 1 µmol/L XAV939 (a WNT signaling inhibitor). In the final phase, 3 µmol/L CHIR99021 (a WNT signaling activator) and 2 µmol/L DAPT (a NOTCH signaling inhibitor) were utilized to achieve the fate of ameloblasts from DE cells. Three-dimensional cultures were investigated to enhance the ameloblast differentiation ability of the induced DE cells. Quantitative reverse transcription polymerase chain reaction (qRT-PCR) and immunofluorescence were conducted to assess the expression of lineage-specific markers. Alizarin Red S (ARS) staining was performed to evaluate the formation of mineralization nodules. RESULTS The application of RA facilitated the efficient generation of NNE within a six-day period. Subsequently, upon stimulation with LDN193189 and XAV939, a notable emergence of DE cells was observed on the eighth days. By the tenth day, ameloblast-like cells derived from hESCs were generated. Upon cultivation in spheroids, these cells exhibited elevated levels of ameloblast markers AMBN and AMELX expression, suggesting that spheroid culture augments the differentiation of ameloblasts. CONCLUSION We established an efficient small molecule-based method to differentiate hESCs into ameloblast-like cells through the concerted modulation of RA, BMP, WNT, and NOTCH signaling pathways, potentially advancing research in enamel and tooth regeneration.
Collapse
Affiliation(s)
- Ximei Zhu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
- Center of Oral Public Health, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - YiMeng Zhao
- Department of Pediatric Dentistry, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Xiaofan Bai
- Department of Pediatric Dentistry, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Qiannan Dong
- Center of Oral Public Health, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Chunli Tian
- Center of Oral Public Health, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Ruilin Sun
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
- Center of Oral Public Health, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Congjuan Yan
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
- Center of Oral Public Health, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Jianping Ruan
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
- Center of Oral Public Health, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Zhongbo Liu
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
- Laboratory Center of Stomatology, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China
| | - Jianghong Gao
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China.
- Center of Oral Public Health, College of Stomatology, Xi'an Jiaotong University, Xi'an, Shaanxi, 710004, China.
| |
Collapse
|
19
|
Wang P, Zhang XP, Liu F, Wang W. Progressive Deactivation of Hydroxylases Controls Hypoxia-Inducible Factor-1α-Coordinated Cellular Adaptation to Graded Hypoxia. RESEARCH (WASHINGTON, D.C.) 2025; 8:0651. [PMID: 40171017 PMCID: PMC11960303 DOI: 10.34133/research.0651] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/20/2024] [Revised: 02/26/2025] [Accepted: 03/08/2025] [Indexed: 04/03/2025]
Abstract
Graded hypoxia is a common microenvironment in malignant solid tumors. As a central regulator in the hypoxic response, hypoxia-inducible factor-1 (HIF-1) can induce multiple cellular processes including glycolysis, angiogenesis, and necroptosis. How cells exploit the HIF-1 pathway to coordinate different processes to survive hypoxia remains unclear. We developed an integrated model of the HIF-1α network to elucidate the mechanism of cellular adaptation to hypoxia. By numerical simulations and bifurcation analysis, we found that HIF-1α is progressively activated with worsening hypoxia due to the sequential deactivation of the hydroxylases prolyl hydroxylase domain enzymes and factor inhibiting HIF (FIH). Bistable switches control the activation and deactivation processes. As a result, glycolysis, immunosuppression, angiogenesis, and necroptosis are orderly elicited in aggravating hypoxia. To avoid the excessive accumulation of lactic acid during glycolysis, HIF-1α induces monocarboxylate transporter and carbonic anhydrase 9 sequentially to export intracellular hydrogen ions, facilitating tumor cell survival. HIF-1α-induced miR-182 facilitates vascular endothelial growth factor production to promote angiogenesis under moderate hypoxia. The imbalance between accumulation and removal of lactic acid in severe hypoxia may result in acidosis and induce cell necroptosis. In addition, the deactivation of FIH results in the destabilization of HIF-1α in anoxia. Collectively, HIF-1α orchestrates the adaptation of tumor cells to hypoxia by selectively inducing its targets according to the severity of hypoxia. Our work may provide clues for tumor therapy by targeting the HIF-1 pathway.
Collapse
Affiliation(s)
- Ping Wang
- Kuang Yaming Honors School,
Nanjing University, Nanjing 210023, China
- Key Laboratory of High Performance Scientific Computation, School of Science,
Xihua University, Chengdu 610039, China
| | - Xiao-Peng Zhang
- Kuang Yaming Honors School,
Nanjing University, Nanjing 210023, China
- Institute of Brain Sciences,
Nanjing University, Nanjing 210093, China
| | - Feng Liu
- Institute of Brain Sciences,
Nanjing University, Nanjing 210093, China
- National Laboratory of Solid State Microstructures and Department of Physics,
Nanjing University, Nanjing 210093, China
| | - Wei Wang
- Institute of Brain Sciences,
Nanjing University, Nanjing 210093, China
- National Laboratory of Solid State Microstructures and Department of Physics,
Nanjing University, Nanjing 210093, China
| |
Collapse
|
20
|
Abdurakhmanova МM, Leonteva AA, Vasilieva NS, Kuligina EV, Nushtaeva AA. 3D cell culture models: how to obtain and characterize the main models. Vavilovskii Zhurnal Genet Selektsii 2025; 29:175-188. [PMID: 40264808 PMCID: PMC12011624 DOI: 10.18699/vjgb-25-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 11/28/2024] [Accepted: 11/28/2024] [Indexed: 04/24/2025] Open
Abstract
For many years, the gold standard in the study of malignant tumors has been the in vitro culture of tumor cells, in vivo xenografts or genetically modified animal models. Meanwhile, three-dimensional cell models (3D cultures) have been added to the arsenal of modern biomedical research. 3D cultures reproduce tissue-specific features of tissue topology. This makes them relevant tissue models in terms of cell differentiation, metabolism and the development of drug resistance. Such models are already being used by many research groups for both basic and translational research, and may substantially reduce the number of animal studies, for example in the field of oncological research. In the current literature, 3D cultures are classified according to the technique of their formation (with or without a scaffold), cultivation conditions (static or dynamic), as well as their cellular organization and function. In terms of cellular organization, 3D cultures are divided into "spheroid models", "organoids", "organs-on-a-chip" and "microtissues". Each of these models has its own unique features, which should be taken into account when using a particular model in an experiment. The simplest 3D cultures are spheroid models which are floating spherical cell aggregates. An organoid is a more complex 3D model, in which a self-organizing 3D structure is formed from stem cells (SCs) capable of self-renewal and differentiation within the model. Organ-on-a-chip models are chips of microfluidic systems that simulate dynamic physical and biological processes found in organs and tissues in vitro. By combining different cell types into a single structure, spheroids and organoids can act as a basis for the formation of a microtissue - a hybrid 3D model imitating a specific tissue phenotype and containing tissue-specific extracellular matrix (ECM) components. This review presents a brief history of 3D cell culture. It describes the main characteristics and perspectives of the use of "spheroid models", "organoids", "organ-on-a-chip" models and "microtissues" in immune oncology research of solid tumors.
Collapse
Affiliation(s)
- М M Abdurakhmanova
- Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - A A Leonteva
- Sirius University of Science and Technology, Sirius Federal Territory, Krasnodar Region, Russia Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - N S Vasilieva
- Sirius University of Science and Technology, Sirius Federal Territory, Krasnodar Region, Russia Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - E V Kuligina
- Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - A A Nushtaeva
- Sirius University of Science and Technology, Sirius Federal Territory, Krasnodar Region, Russia Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
21
|
Bhuker S, Sinha AK, Arora A, Tuli HS, Datta S, Saini AK, Saini RV, Ramniwas S. Genes and proteins expression profile of 2D vs 3D cancer models: a comparative analysis for better tumor insights. Cytotechnology 2025; 77:51. [PMID: 39867829 PMCID: PMC11759753 DOI: 10.1007/s10616-025-00714-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2024] [Accepted: 01/08/2025] [Indexed: 01/28/2025] Open
Abstract
When juxtaposed with 2D cell culture models, multicellular tumor spheroids demonstrate a capacity to faithfully replicate certain features inherent to solid tumors. These include spatial architecture, physiological responses, the release of soluble mediators, patterns of gene expression, and mechanisms of drug resistance. The morphological and behavioural similarities between 3D-cultured cells and cells within tumor masses highlight the potential of these models in studying cancer biology and drug responses. The liquid overlay method, hanging drop technique, and ultra-low adhesion plates are among the various methods for generating tumor spheroids, each with its advantages and applications. Gene expression studies, employing advanced methods such as microarrays, suppression subtractive hybridization, qRT-PCR, and mass-spectrometry-based proteomics revealed distinct expression patterns in 3D spheroids compared to 2D cultures, uncovering upregulation and downregulation of genes associated with tumor development, metastasis, and drug resistance. Protein expression studies identified alterations in key signaling pathways, metabolic characteristics, and phosphorylation levels, highlighting the impact of 3D culture on cellular responses. This study explores genes and proteins expression variations in various cancer cell lines cultivated in 3D spheroids, shedding light on the complexity of interactions in a more tumor-mimicking environment. The fusion of these analytical approaches not only advances scientific understanding but also holds promise for the development of more effective cancer treatments.
Collapse
Affiliation(s)
- Sunaina Bhuker
- Department of Bio-Sciences & Technology, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana, 133207 India
| | - Abhinav Kumar Sinha
- Department of Bio-Sciences & Technology, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana, 133207 India
| | - Anuksha Arora
- Department of Bio-Sciences & Technology, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana, 133207 India
| | - Hardeep Singh Tuli
- Department of Bio-Sciences & Technology, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana, 133207 India
| | - Sonal Datta
- Department of Bio-Sciences & Technology, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana, 133207 India
| | - Adesh K. Saini
- Department of Bio-Sciences & Technology, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana, 133207 India
- Central Research Laboratory, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana-Ambala, Haryana 133207 India
| | - Reena V. Saini
- Department of Bio-Sciences & Technology, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana, 133207 India
- Central Research Laboratory, MMEC, Maharishi Markandeshwar (Deemed to Be University), Mullana-Ambala, Haryana 133207 India
| | - Seema Ramniwas
- University Centre for Research and Development, University Institute of Pharmaceutical Sciences, Chandigarh University, Gharuan, Mohali, 140413 India
| |
Collapse
|
22
|
Zheng J, He L, Shi Q, Wang M, Ma Y, Yu W, Liu L, Yu G, Liu X, Wang B, Zhong J. In vivo and In vitro assessment of the retinal toxicity of polystyrene nanoplastics. ENVIRONMENT INTERNATIONAL 2025; 198:109420. [PMID: 40158453 DOI: 10.1016/j.envint.2025.109420] [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: 09/21/2024] [Revised: 01/28/2025] [Accepted: 03/26/2025] [Indexed: 04/02/2025]
Abstract
Plastic pollution has emerged as a critical global environmental challenge, yet the effects of the ingested plastic particles on ocular health remain largely unexplored. In this study, we investigated the impact of orally ingested polystyrene nanoplastics (PS-NPs) on the mouse retina. The in vivo experimental results showed that PS-NPs could penetrate the mouse retina within 2 h after gavage. Their levels increased at 4 h and remained detectable up to 24 h post-gavage. Prolonged exposure (28 days) to PS-NPs might disrupt the tight junctions of the inner blood-retinal barrier (iBRB). Moreover, PS-NPs induced oxidative stress in the retina by downregulating the expression of Nrf2 and HO-1, and potentially promoted apoptosis via the upregulation of Cleaved caspase 3. Additionally, we used human retinal microvascular endothelial cells (HRMECs) to model the iBRB and employed a human retinal pigment epithelial cell line (ARPE-19) to assess the potential toxicity of PS-NPs on the human retina. Our results indicated that PS-NPs penetrated and disrupted the simulated iBRB, inducing oxidative stress and promoting apoptosis in ARPE-19 cells. This study provides critical insights into the potential risks of ingested PS-NPs to retinal health and offers novel perspectives on the broader implications of plastic pollution for humans.
Collapse
Affiliation(s)
- Jiang Zheng
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China
| | - Lun He
- The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou Medical University, Guangzhou 510260, China
| | - Qi Shi
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China
| | - Meilan Wang
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China
| | - Yu Ma
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China
| | - Wenjuan Yu
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China
| | - Lian Liu
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China
| | - Guocheng Yu
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China
| | - Xiaoting Liu
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China; Institute for Environmental and Climate Research, Jinan University, Guangzhou 511443, China
| | - Boguang Wang
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China; Institute for Environmental and Climate Research, Jinan University, Guangzhou 511443, China.
| | - Jingxiang Zhong
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou 510630, China; The Sixth Affiliated Hospital of Jinan University, Jinan University, Dongguan 523129, China.
| |
Collapse
|
23
|
Kim H, Kang S, Cho B, An S, Kim Y, Kim J. Parkinson's Disease Modeling Using Directly Converted 3D Induced Dopaminergic Neuron Organoids and Assembloids. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2412548. [PMID: 39965129 PMCID: PMC11984911 DOI: 10.1002/advs.202412548] [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: 10/08/2024] [Revised: 02/03/2025] [Indexed: 02/20/2025]
Abstract
Parkinson's disease (PD) is characterized by the progressive loss of dopaminergic neurons and the accumulation of α-synuclein aggregates, yet current models inadequately mimic the complex human brain environment. Recent advances in brain organoid models offer a more physiologically relevant platform for studying PD, however, iPSC-derived brain organoids require long maturation times and may not accurately represent the aged brain's epigenetics and cellular contexts, limiting their applicability for modeling late-onset diseases like PD. In this study, a novel approach for generating 3D-induced dopaminergic (iDA) neuron organoids directly from human fibroblasts is presented. It is confirmed that these 3D iDA organoids more closely resemble the aged human brain and accurately replicate PD pathologies. Furthermore, this model is extended by incorporating astrocytes to create 3D iDA neuron-astrocyte assembloids, recognizing the critical role of glial cells in neurodegenerative processes. It is identified that PD assembloids incorporating control astrocytes with A53T mutant iDAs demonstrated the neuroprotective effects of healthy astrocytes. In contrast, A53T mutant astrocytes progressively contributed to neuronal degeneration and synucleinopathy in 3D-iDA assembloids. These findings suggest that directly converted 3D-iDA organoids and assembloids provide a robust and physiologically relevant model for studying PD pathogenesis and evaluating therapeutic interventions.
Collapse
Affiliation(s)
- Hongwon Kim
- Department of ChemistryDongguk UniversityPudong 1‐gil 30, Jung‐guSeoul04620Republic of Korea
- Department of Chemistry and Chemical BiologyRutgersThe State University of New JerseyPiscatawayNJ08854USA
| | - Soi Kang
- Department of ChemistryDongguk UniversityPudong 1‐gil 30, Jung‐guSeoul04620Republic of Korea
| | - Byounggook Cho
- Department of ChemistryDongguk UniversityPudong 1‐gil 30, Jung‐guSeoul04620Republic of Korea
| | - Saemin An
- Department of ChemistryDongguk UniversityPudong 1‐gil 30, Jung‐guSeoul04620Republic of Korea
| | - Yunkyung Kim
- Department of ChemistryDongguk UniversityPudong 1‐gil 30, Jung‐guSeoul04620Republic of Korea
| | - Jongpil Kim
- Department of ChemistryDongguk UniversityPudong 1‐gil 30, Jung‐guSeoul04620Republic of Korea
| |
Collapse
|
24
|
Luanpitpong S, Janan M, Poohadsuan J, Rodboon N, Samart P, Rungarunlert S, Issaragrisil S. A High-Throughput, Three-Dimensional Multiple Myeloma Model Recapitulating Tumor-Stroma Interactions for CAR-Immune Cell-Mediated Cytotoxicity Assay. Immunotargets Ther 2025; 14:321-338. [PMID: 40182067 PMCID: PMC11967349 DOI: 10.2147/itt.s503984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Accepted: 03/18/2025] [Indexed: 04/05/2025] Open
Abstract
Background Multiple myeloma (MM) is characterized by an excessive proliferation of clonal plasma cells in the bone marrow (BM). Components in BM niche contribute to the immunosuppressive tumor microenvironment (TME), but three-dimensional (3D) MM models that recreate the complex TME and enable high-throughput cytotoxicity assay of chimeric antigen receptor (CAR)-engineered immune cells are still lacking. Methods Stable, luciferase (Luc)-labeled target MM cells were generated using Luc/RFP dual reporter system to track MM growth. 3D spheroids were formed in a 96-well plate in the presence or absence of cancer-associated fibroblast (CAF)-like stromal cells activated by MM-derived conditioned medium and the cytotoxicity of CAR-immune cells, which were represented by third-generation anti-CD138 CAR-NK-92 cells, was evaluated by luciferase assay using a multimode microplate reader. Immune cell infiltration was visualized under a fluorescence microscope by using multiple fluorescent dyes. Results We first showed that luciferase assay provides a relatively simple and robust means to specifically monitor Luc-labeled tumor cell growth in a coculture system, allowing the high-throughput assessment of CAR-immune cytotoxicity. Through this assay, we demonstrated that CAF-like stromal cells impaired NK cell effector function in 2D culture and 3D spheroids, likely via paracrine signaling and physical barrier function. Importantly, we showed that 3D spheroids consisting of MM cells and CAF-like stromal cells provide a more comprehensive, physiologically relevant immuno-oncology model. Our established model could also be used to investigate the trafficking and infiltration of immune cells into the core of spheroids. Herein, we showed that CAR incorporation did improve the ability of NK cells to infiltrate 3D spheroids. Conclusion Our established 3D spheroid model, which partially recapitulates the complex TME with immunosuppressive environment, is suitable for high-throughput screening of CAR-immune cytotoxicity and could be important in accelerating immuno-oncology drug discovery for MM since there is a pressing need to establish innovative CAR-immune cells.
Collapse
Affiliation(s)
- Sudjit Luanpitpong
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Cell Factory for Cancer Immunotherapy, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Blood Products and Cellular Immunotherapy Research Group, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Montira Janan
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Siriraj Cell Factory for Cancer Immunotherapy, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Blood Products and Cellular Immunotherapy Research Group, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Jirarat Poohadsuan
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Napachai Rodboon
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Parinya Samart
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Sasitorn Rungarunlert
- Department of Preclinic and Applied Animal Science, Faculty of Veterinary Science, Mahidol University, Nakhon Pathom, Thailand
| | - Surapol Issaragrisil
- Siriraj Center of Excellence for Stem Cell Research, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
- BDMS Center of Excellence for Hematology, Wattanosoth Cancer Hospital, Bangkok, Thailand
| |
Collapse
|
25
|
Petrauskas V, Damaseviciute R, Gulla A. Pancreatic 3D Organoids and Microfluidic Systems-Applicability and Utilization in Surgery: A Literature Review. MEDICINA (KAUNAS, LITHUANIA) 2025; 61:623. [PMID: 40282914 PMCID: PMC12028617 DOI: 10.3390/medicina61040623] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/05/2025] [Accepted: 03/26/2025] [Indexed: 04/29/2025]
Abstract
Background: Pancreatic organoids are a rapidly advancing field of research with new discoveries being made every day. A literature review was performed to answer the question of how relevant 3D pancreatic organoids are for surgery. Materials and Methods: We started our investigation by identifying articles in PubMed within the last 5 years using the keywords (("pancreatic organoid", OR "organ-on-a-chip", OR "pancreatic chip" OR "3D culture methods") AND pancreatic surgery). Only English articles were included in this literature review. This literature review was performed in a non-systematic way; articles were chosen without a predetermined protocol of inclusion and were based on the aim of the review. Results and Conclusions: There are many promising innovations in the field of 3D cultures. Drug sensitivity testing in particular holds great potential for surgical application. For locally advanced PDAC, EUS-FNB obtained cancer tissue can be cultured as organoids, and after 4 weeks, neoadjuvant treatment could be adjusted for each patient individually. Utilizing this approach could increase the number of R0 resections and possibly cure the disease. Furthermore, microfluidic devices, as a platform for pancreatic islet pre-transplant evaluation or cultivation of beta cells derived from HiPSC in vitro, promise broad application of islet transplantation to T1DM patients in the near future.
Collapse
Affiliation(s)
- Vidas Petrauskas
- Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, LT-01131 Vilnius, Lithuania
| | - Ryte Damaseviciute
- Center of Visceral Medicine and Translational Research, Faculty of Medicine, Vilnius University, LT-01131 Vilnius, Lithuania
| | - Aiste Gulla
- Institute of Clinical Medicine, Faculty of Medicine, Vilnius University, LT-01131 Vilnius, Lithuania
- Center of Visceral Medicine and Translational Research, Faculty of Medicine, Vilnius University, LT-01131 Vilnius, Lithuania
- Department of Surgery, George Washington University, Washington, DC 20052, USA
| |
Collapse
|
26
|
Peng Z, Lv X, Sun H, Zhao L, Huang S. 3D tumor cultures for drug resistance and screening development in clinical applications. Mol Cancer 2025; 24:93. [PMID: 40119343 PMCID: PMC11927140 DOI: 10.1186/s12943-025-02281-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2025] [Accepted: 02/24/2025] [Indexed: 03/24/2025] Open
Abstract
Tumor drug resistance presents a growing challenge in medical practice, particularly during anti-cancer therapies, where the emergence of drug-resistant cancer cells significantly complicates clinical treatment. In recent years, three-dimensional (3D) tumor culture technology, which more effectively simulates the in vivo physiological environment, has gained increasing attention in tumor drug resistance research and clinical applications. By mimicking the in vivo cellular microenvironment, 3D tumor culture technology not only recapitulates cell-cell interactions but also more faithfully reproduces the biological effects of therapeutic agents. Consequently, 3D tumor culture technology is emerging as a crucial tool in biomedical and clinical research. We summarize the benefits of 3D culture models and organoid technology, explore their application in the realm of drug resistance, drug screening, and personalized therapy, and discuss their potential application prospects and challenges in clinical transformation, with the aim of providing insights for optimizing cancer treatment strategies and advancing precision therapy.
Collapse
Affiliation(s)
- Zheng Peng
- Department of Clinical Laboratory, Liuzhou Traditional Chinese Medical Hospital, Liuzhou, Guangxi, China
| | - Xiaolan Lv
- Department of Clinical Laboratory, Liuzhou Maternity and Child Healthcare Hospital, Liuzhou, Guangxi, China
| | - Hao Sun
- Faculty of Science, Autonomous University of Madrid, Spainish National Research Council -Consejo Superior de Investigaciones Científicas,(UAM-CSIC), Madrid, 28049, Spain
| | - Lina Zhao
- State Key Laboratory of Holistic Integrative Management of Gastrointestinal Cancers, Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| | - Shigao Huang
- Department of Radiation Oncology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
27
|
Dondi C, Tsikritsis D, Vorng JL, Greenidge G, Kepiro IE, Belsey NA, McMahon G, Gilmore IS, Ryadnov MG, Shaw M. Multiparametric physicochemical analysis of a type 1 collagen 3D cell culture model using light and electron microscopy and mass spectrometry imaging. Sci Rep 2025; 15:9578. [PMID: 40113888 PMCID: PMC11926111 DOI: 10.1038/s41598-025-93700-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
Three-dimensional cell culture systems underpin cell-based technologies ranging from tissue scaffolds for regenerative medicine to tumor models and organoids for drug screening. However, to realise the full potential of these technologies requires analytical methods able to capture the diverse information needed to characterize constituent cells, scaffold components and the extracellular milieu. Here we describe a multimodal imaging workflow which combines fluorescence, vibrational and second harmonic generation microscopy with secondary ion mass spectrometry imaging and transmission electron microscopy to analyse the morphological, chemical and ultrastructural properties of cell-seeded scaffolds. Using cell nuclei as landmarks we register fluorescence with label-free optical microscopy images and high mass resolution with high spatial resolution secondary ion mass spectrometry images, with an accuracy comparable to the intrinsic spatial resolution of the techniques. We apply these methods to investigate relationships between cell distribution, cytoskeletal morphology, scaffold fiber organisation and biomolecular composition in type I collagen scaffolds seeded with human dermal fibroblasts.
Collapse
Affiliation(s)
- Camilla Dondi
- National Physical Laboratory, Hampton Road, Teddington, TW11 0LW, UK
| | | | - Jean-Luc Vorng
- National Physical Laboratory, Hampton Road, Teddington, TW11 0LW, UK
| | - Gina Greenidge
- National Physical Laboratory, Hampton Road, Teddington, TW11 0LW, UK
| | - Ibolya E Kepiro
- National Physical Laboratory, Hampton Road, Teddington, TW11 0LW, UK
| | - Natalie A Belsey
- National Physical Laboratory, Hampton Road, Teddington, TW11 0LW, UK
- School of Chemistry and Chemical Engineering, University of Surrey, Guildford, GU2 7XH, UK
| | - Greg McMahon
- National Physical Laboratory, Hampton Road, Teddington, TW11 0LW, UK
| | - Ian S Gilmore
- National Physical Laboratory, Hampton Road, Teddington, TW11 0LW, UK
| | - Maxim G Ryadnov
- National Physical Laboratory, Hampton Road, Teddington, TW11 0LW, UK
| | - Michael Shaw
- National Physical Laboratory, Hampton Road, Teddington, TW11 0LW, UK.
- UCL Hawkes Institute and Department of Computer Science, University College London, London, UK.
| |
Collapse
|
28
|
Kumar A, Shahvej SK, Yadav P, Modi U, Yadav AK, Solanki R, Bhatia D. Clinical Applications of Targeted Nanomaterials. Pharmaceutics 2025; 17:379. [PMID: 40143042 PMCID: PMC11944548 DOI: 10.3390/pharmaceutics17030379] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2025] [Revised: 03/07/2025] [Accepted: 03/12/2025] [Indexed: 03/28/2025] Open
Abstract
Targeted nanomaterials are at the forefront of advancements in nanomedicine due to their unique and versatile properties. These include nanoscale size, shape, surface chemistry, mechanical flexibility, fluorescence, optical behavior, magnetic and electronic characteristics, as well as biocompatibility and biodegradability. These attributes enable their application across diverse fields, including drug delivery. This review explores the fundamental characteristics of nanomaterials and emphasizes their importance in clinical applications. It further delves into methodologies for nanoparticle programming alongside discussions on clinical trials and case studies. We discussed some of the promising nanomaterials, such as polymeric nanoparticles, carbon-based nanoparticles, and metallic nanoparticles, and their role in biomedical applications. This review underscores significant advancements in translating nanomaterials into clinical applications and highlights the potential of these innovative approaches in revolutionizing the medical field.
Collapse
Affiliation(s)
- Ankesh Kumar
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar 382355, Gujarat, India
| | - SK Shahvej
- Amrita School of Biotechnology, Amrita Vishwa Vidyapeetham, Kollam 690525, Kerala, India
| | - Pankaj Yadav
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar 382355, Gujarat, India
| | - Unnati Modi
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar 382355, Gujarat, India
| | - Amit K. Yadav
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar 382355, Gujarat, India
| | - Raghu Solanki
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar 382355, Gujarat, India
| | - Dhiraj Bhatia
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar, Palaj, Gandhinagar 382355, Gujarat, India
| |
Collapse
|
29
|
Zhou D, Li X, Liu W, Zhang M, Cheng Y, Xu Z, Gao J, Wang Y. A novel approach for engineering DHCM/GelMA microgels: application in hepatocellular carcinoma cell encapsulation and chemoresistance research. Front Bioeng Biotechnol 2025; 13:1564543. [PMID: 40161518 PMCID: PMC11949893 DOI: 10.3389/fbioe.2025.1564543] [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: 01/21/2025] [Accepted: 02/28/2025] [Indexed: 04/02/2025] Open
Abstract
Liver cancer, a highly aggressive malignancy, continues to present significant challenges in therapeutic management due to its pronounced chemoresistance. This resistance, which undermines the efficacy of conventional chemotherapy and targeted therapies, is driven by multifaceted mechanisms, with increasing emphasis placed on the protective role of the tumor microenvironment (TME). The hepatocellular carcinoma extracellular matrix (ECM), a primary non-cellular component of the TME, has emerged as a critical regulator in cancer progression and drug resistance, particularly in hepatocellular carcinoma cell (HCC). In this study, a hybrid biomimetic hydrogel was engineered by integrating decellularized hepatocellular carcinoma matrix (DHCM) with gelatin methacrylate (GelMA) precursors. This composite DHCM/GelMA hydrogel was designed to replicate the physicochemical and functional properties of the hepatocellular carcinoma ECM, thereby offering a biomimetic platform to explore the interactions between HCCs and their microenvironment. Leveraging a custom-designed microfluidic 3D printing platform, we achieved high-throughput fabrication of HCC-encapsulated DHCM/GelMA microgels, characterized by enhanced uniformity, biocompatibility, and scalability. These microgels facilitated the construction of hepatocellular carcinoma microtissues, which were subsequently employed for chemoresistance studies. Our findings revealed that DHCM/GelMA microgels closely mimic the hepatocellular carcinoma tumor microenvironment, effectively recapitulating key features of ECM-mediated drug resistance. Mechanistic studies further demonstrated that DHCM significantly upregulates the expression of Aquaporin 3 (AQP3) in the encapsulated HCCs. This upregulation potentially activates mTOR signaling-associated autophagy pathways, thereby enhancing chemoresistance in HCCs. These biomimetic models provide a robust and versatile platform for studying the underlying mechanisms of drug resistance and evaluating therapeutic interventions. This innovative approach highlights the potential of DHCM/GelMA microgels as a transformative tool in cancer-associated tissue engineering and anticancer drug screening. By enabling detailed investigations into the role of ECM in chemoresistance, this study contributes to advancing therapeutic research and offers promising strategies to overcome drug resistance, ultimately improving clinical outcomes in liver cancer treatment.
Collapse
Affiliation(s)
- Dandan Zhou
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Department of Geriatric Medicine, Jiulongpo People’s Hospital of Chongqing, Chongqing, China
| | - Xiaoxiao Li
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Tissue Repairing and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Wencun Liu
- Department of Radiology, Jiulongpo People’s Hospital of Chongqing, Chongqing, China
| | - Mingjun Zhang
- Department of Clinical Laboratory, Jiulongpo People’s Hospital of Chongqing, Chongqing, China
| | - Ying Cheng
- Department of Clinical Laboratory, Jiulongpo People’s Hospital of Chongqing, Chongqing, China
| | - Zhousong Xu
- Department of Clinical Laboratory, Jiulongpo People’s Hospital of Chongqing, Chongqing, China
| | - Jian Gao
- Department of Gastroenterology, The Second Affiliated Hospital of Chongqing Medical University, Chongqing, China
| | - Yiyang Wang
- Department of Orthopedics, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
- Tissue Repairing and Biotechnology Research Center, The Third Affiliated Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
30
|
Mahadeo CO, Shahin-Shamsabadi A, Khodamoradi M, Fahnestock M, Selvaganapathy PR. The Effects of Electrical Stimulation on a 3D Osteoblast Cell Model. Cells 2025; 14:396. [PMID: 40136645 PMCID: PMC11941504 DOI: 10.3390/cells14060396] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Revised: 01/31/2025] [Accepted: 03/04/2025] [Indexed: 03/27/2025] Open
Abstract
Electrical stimulation has been used with tissue engineering-based models to develop three-dimensional (3D), dynamic, research models that are more physiologically relevant than static two-dimensional (2D) cultures. For bone tissue, the effect of electrical stimulation has focused on promoting healing and regeneration of tissue to prevent bone loss. However, electrical stimulation can also potentially affect mature bone parenchymal cells such as osteoblasts to guide bone formation and the secretion of paracrine or endocrine factors. Due to a lack of physiologically relevant models, these phenomena have not been studied in detail. In vitro electrical stimulation models can be useful for gaining an understanding of bone physiology and its effects on paracrine tissues under different physiological and pathological conditions. Here, we use a 3D, dynamic, in vitro model of bone to study the effects of electrical stimulation conditions on protein and gene expression of SaOS-2 human osteosarcoma osteoblast-like cells. We show that different stimulation regimens, including different frequencies, exposure times, and stimulation patterns, can have different effects on the expression and secretion of the osteoblastic markers alkaline phosphatase and osteocalcin. These results reveal that electrical stimulation can potentially be used to guide osteoblast gene and protein expression.
Collapse
Affiliation(s)
- Crystal O. Mahadeo
- Neuroscience Graduate Program, McMaster University, Hamilton, ON L8S 4K1, Canada;
| | - Alireza Shahin-Shamsabadi
- School of Biomedical Engineering, McMaster University, Hamilton, ON L8S 4L8, Canada; (A.S.-S.); (M.K.)
- Department of Mechanical Engineering, McMaster University, Hamilton, ON L8S 4L8, Canada
| | - Maedeh Khodamoradi
- School of Biomedical Engineering, McMaster University, Hamilton, ON L8S 4L8, Canada; (A.S.-S.); (M.K.)
| | - Margaret Fahnestock
- Department of Psychiatry and Behavioural Neurosciences, McMaster University, Hamilton, ON L8S 4K1, Canada
| | - Ponnambalam Ravi Selvaganapathy
- School of Biomedical Engineering, McMaster University, Hamilton, ON L8S 4L8, Canada; (A.S.-S.); (M.K.)
- Department of Mechanical Engineering, McMaster University, Hamilton, ON L8S 4L8, Canada
| |
Collapse
|
31
|
Potter MJ, Heywood JD, Coeyman SJ, Richardson WJ. Heart Scar-In-A-Dish: Tissue Culture Platform to Study Myocardial Infarct Healing In Vitro. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.28.640625. [PMID: 40060569 PMCID: PMC11888419 DOI: 10.1101/2025.02.28.640625] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/15/2025]
Abstract
Myocardial Infarction (MI) is a major contributor to morbidity and mortality, wherein blood flow is blocked to a portion of the left ventricle and leads to myocardial necrosis and scar formation. Cardiac remodeling in response to MI is a major determinant of patient prognosis, so many therapies are under development to improve infarct healing. Part of this development involves in vitro therapy screening which can be accelerated by engineered heart tissues (EHTs). Unfortunately, EHTs often over-simplify the infarcted tissue microarchitecture by neglecting spatial variation found in infarcted ventricles. MI results in a spatially heterogeneous environment with an infarct zone composed mostly of extracellular matrix (ECM) and cardiac fibroblasts, contrasted with a remote (non-infarct) zone composed mostly of cardiomyocytes, and a border zone transitioning in between. The heterogeneous structure is accompanied by heterogeneous mechanics where the passive infarct zone is cyclically stretched under tension as the remote zone cyclically contracts with every heartbeat. We present an in vitro 3-dimensional tissue culture platform focused on mimicking the heterogeneous mechanical environment of post-infarct myocardium. Herein, EHTs were subjected to a cryowound injury to induce localized cell death in a central portion of beating tissues composed of neonatal rat cardiomyocytes and cardiac fibroblasts. After injury, the remote zone continued to contract (i.e., negative strains) while the wounded zone was cyclically stretched (i.e., positive tensile strains) with intermediate strains in the border zone. We also observed increased tissue stiffnesses in the wounded zone and border zone following injury, while the remote zone did not show the same stiffening. Collectively, this work establishes a novel in vitro platform for characterizing myocardial wound remodeling with both spatial and temporal resolution, contributing to a deeper understanding of MI and offering insights for potential therapeutic approaches.
Collapse
Affiliation(s)
- M J Potter
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - J D Heywood
- Bioengineering Department, Clemson University, Clemson, SC, USA
| | - S J Coeyman
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, Fayetteville, AR, USA
| | - W J Richardson
- Ralph E. Martin Department of Chemical Engineering, University of Arkansas, Fayetteville, AR, USA
| |
Collapse
|
32
|
Braccini S, Pecorini G, Biagini S, Tacchini C, Battisti A, Puppi D. Chitosan/alginate polyelectrolyte complex hydrogels by additive manufacturing for in vitro 3D ovarian cancer modeling. Int J Biol Macromol 2025; 296:139795. [PMID: 39805455 DOI: 10.1016/j.ijbiomac.2025.139795] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 01/04/2025] [Accepted: 01/10/2025] [Indexed: 01/16/2025]
Abstract
Polyelectrolyte complexes (PECs) are self-assembled systems formed from oppositely charged polymers, used to create hydrogels for cell culture. This work was aimed at additive manufacturing 3D hydrogels made of a PEC between chitosan (Cs) and alginate, as well as their investigation for in vitro 3D ovarian cancer modeling. PEC hydrogels stability in cell culture medium demonstrated their suitability for long-term cell culture applications. Higher in vitro viability of two human ovarian cancer cell lines was detected at different time points on PEC hydrogels than on Cs hydrogels, used as a control. In addition, during the 63-day culture experiment, cells effectively colonized the scaffolds while retaining their aggressive tumor characteristics. A significantly lower sensitivity to cisplatin and eugenol, also when combined, was observed in the developed 3D ovarian cancer models, in comparison to what was achieved in relevant 2D cell cultures. The obtained results demonstrated therefore the suitability of the developed scaffolds for in vitro investigation of tumor modeling.
Collapse
Affiliation(s)
- Simona Braccini
- BIOLab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM - Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| | - Gianni Pecorini
- BIOLab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM - Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| | - Serena Biagini
- BIOLab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM - Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| | - Chiara Tacchini
- BIOLab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM - Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy
| | - Antonella Battisti
- NEST, Istituto Nanoscienze-CNR and Scuola Normale Superiore, p.zza San Silvestro 12, 56127 Pisa, Italy
| | - Dario Puppi
- BIOLab Research Group, Department of Chemistry and Industrial Chemistry, University of Pisa, UdR INSTM - Pisa, Via G. Moruzzi 13, 56124 Pisa, Italy.
| |
Collapse
|
33
|
Verdugo-Avello F, Wychowaniec JK, Villacis-Aguirre CA, D'Este M, Toledo JR. Bone microphysiological models for biomedical research. LAB ON A CHIP 2025; 25:806-836. [PMID: 39906932 DOI: 10.1039/d4lc00762j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2025]
Abstract
Bone related disorders are highly prevalent, and many of these pathologies still do not have curative and definitive treatment methods. This is due to a complex interplay of multiple factors, such as the crosstalk between different tissues and cellular components, all of which are affected by microenvironmental factors. Moreover, these bone pathologies are specific, and current treatment results vary from patient to patient owing to their intrinsic biological variability. Current approaches in drug development to deliver new drug candidates against common bone disorders, such as standard two-dimensional (2D) cell culture and animal-based studies, are now being replaced by more relevant diseases modelling, such as three-dimension (3D) cell culture and primary cells under human-focused microphysiological systems (MPS) that can resemble human physiology by mimicking 3D tissue organization and cell microenvironmental cues. In this review, various technological advancements for in vitro bone modeling are discussed, highlighting the progress in biomaterials used as extracellular matrices, stem cell biology, and primary cell culture techniques. With emphasis on examples of modeling healthy and disease-associated bone tissues, this tutorial review aims to survey current approaches of up-to-date bone-on-chips through MPS technology, with special emphasis on the scaffold and chip capabilities for mimicking the bone extracellular matrix as this is the key environment generated for cell crosstalk and interaction. The relevant bone models are studied with critical analysis of the methods employed, aiming to serve as a tool for designing new and translational approaches. Additionally, the features reported in these state-of-the-art studies will be useful for modeling bone pathophysiology, guiding future improvements in personalized bone models that can accelerate drug discovery and clinical translation.
Collapse
Affiliation(s)
- Francisco Verdugo-Avello
- Biotechnology and Biopharmaceuticals Laboratory, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Víctor Lamas 1290, P.O. Box 160-C, Concepción, Chile.
| | | | - Carlos A Villacis-Aguirre
- Biotechnology and Biopharmaceuticals Laboratory, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Víctor Lamas 1290, P.O. Box 160-C, Concepción, Chile.
| | - Matteo D'Este
- AO Research Institute Davos, Clavadelerstrasse 8, 7270, Davos, Switzerland
| | - Jorge R Toledo
- Biotechnology and Biopharmaceuticals Laboratory, Departamento de Fisiopatología, Facultad de Ciencias Biológicas, Universidad de Concepción, Víctor Lamas 1290, P.O. Box 160-C, Concepción, Chile.
| |
Collapse
|
34
|
Lee KY, Oh SY, Lee HJ, Kwon TG, Kim JW, Shin CG, Hong SH, Choi SY. MTMR6 downregulation contributes to cisplatin resistance in oral squamous cell carcinoma. Cancer Cell Int 2025; 25:30. [PMID: 39891222 PMCID: PMC11783708 DOI: 10.1186/s12935-025-03654-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Accepted: 01/16/2025] [Indexed: 02/03/2025] Open
Abstract
BACKGROUND The therapeutic effectiveness of cisplatin, a widely used chemotherapy drug for oral squamous cell carcinoma (OSCC), is often compromised by resistance, making it difficult to predict treatment outcomes. The role of myotubularin and myotubularin-related (MTMR) genes in cisplatin resistance remains unclear. We aimed to elucidate the molecular mechanisms underlying MTMR6 with cisplatin resistance in OSCC. METHODS MTMR6 expression was compared between UMSCC1 and cisplatin-resistant UM-Cis cells. Gain- and loss-of-function experiments involving MTMR6 was performed to evaluate its impact on cisplatin resistance. The regulatory role of hsa-miR-544a on MTMR6 expression was explored via antagomir and miRNA mimic assays. The relationship between MTMR6 protein levels and cisplatin sensitivity was assessed in OSCC patient tissues classified as sensitive or resistant to cisplatin monotherapy. A survival analysis based on The Cancer Genome Atlas (TCGA) head and neck squamous cell carcinoma (HNSCC) dataset was performed to evaluate the correlation between MTMR6 expression and patient outcomes following cisplatin treatment. In vivo cisplatin resistance was examined using mouse xenografts derived from MTMR6-knockdown UMSCC1 cells. RESULTS MTMR6 expression was markedly reduced in cisplatin-resistant UM-Cis cells compared to UMSCC1 cells. Functional analyses revealed that modulating MTMR6 activity alters cisplatin resistance. A luciferase assay confirmed that hsa-miR-544a regulates MTMR6 gene expression. Additionally, antagomir and miRNA mimics demonstrated that hsa-miR-544a enhances cisplatin resistance by suppressing MTMR6 expression. In OSCC patient tissues, higher MTMR6 protein levels were associated with cisplatin sensitivity, while cisplatin-resistant tissues had lower MTMR6 expression. Survival analysis of the TCGA HNSCC dataset indicated that low MTMR6 expression correlates with poorer outcomes in cisplatin-treated patients compared to those with high MTMR6 expression. Mouse xenografts derived from MTMR6-knockdown UMSCC1 cells exhibited increased resistance to cisplatin compared to controls. CONCLUSION Assessing mRNA levels of MTMR6 and has-miR-544a in biopsy samples could help predict cisplatin responsiveness in OSCC.
Collapse
Affiliation(s)
- Kah Young Lee
- Department of Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Su Young Oh
- Department of Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Heon-Jin Lee
- Department of Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Tae-Geon Kwon
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Jin-Wook Kim
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Chang-Geol Shin
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Kyungpook National University, Daegu, South Korea
| | - Su-Hyung Hong
- Department of Microbiology and Immunology, School of Dentistry, Kyungpook National University, Daegu, South Korea.
| | - So-Young Choi
- Department of Oral and Maxillofacial Surgery, School of Dentistry, Kyungpook National University, Daegu, South Korea.
| |
Collapse
|
35
|
Martorana A, Puleo G, Miceli GC, Cancilla F, Licciardi M, Pitarresi G, Tranchina L, Marrale M, Palumbo FS. Redox/NIR dual-responsive glutathione extended polyurethane urea electrospun membranes for synergistic chemo-photothermal therapy. Int J Pharm 2025; 669:125108. [PMID: 39708849 DOI: 10.1016/j.ijpharm.2024.125108] [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: 10/09/2024] [Revised: 12/18/2024] [Accepted: 12/18/2024] [Indexed: 12/23/2024]
Abstract
Despite advancements in cancer treatments, therapies frequently exhibit high cytotoxicity, and surgery remains the predominant method for treating most solid tumors, often with limited success in preventing post-surgical recurrence. Implantable biomaterials, designed to release drugs at a localised site in response to specific stimuli, represent a promising approach for enhancing tumour therapy. In this study, a redox-responsive glutathione extended polyurethane urea (PolyCEGS) was used to produce paclitaxel (PTX) and gold nanorods (AuNRs) loaded electrospun membranes for combined redox/near-infrared (NIR) light-responsive release chemotherapy and hyperthermic effect. Electrospinning conditions were optimized to fabricate AuNR-loaded scaffolds, at three different AuNRs concentrations. The obtained membranes were characterized by scanning electron microscopy (SEM) analyses and photothermal profiles were evaluated by a thermocamera, showing a temperature increase, up to 42.5 °C, when exposed to NIR light (810 nm) at 3 W/cm2. The AuNRs/PTX loaded scaffolds exhibited sustained PTX release, with 15 % released over 30 days and almost 1.8 times more in a simulated reductive environment. Moreover, their excellent photothermal effects and NIR light-triggered release led to significant synergic cytotoxicity in human colon cancer (HCT-116) and human breast cancer (MCF-7) cell lines. This system potentially enables controllable locoregional PTX release at the tumour site post-surgery, preventing recurrence and enhancing cytotoxicity through combined drug and PTT effects, highlighting its potential for future anticancer treatments.
Collapse
Affiliation(s)
- Annalisa Martorana
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, Italy; Fondazione Ri.MED, c/o IRCCS ISMETT, via E. Tricomi 5, 90127, Palermo, Italy(2)
| | - Giorgia Puleo
- Department of Biological, Chemical and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Viale delle Scienze, Edificio 18, Palermo, Italy; Department of Pharmacy, University of Copenhagen, Universitetsparken 2, Copenhagen, 2100, Denmark
| | - Giovanni Carlo Miceli
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, Italy; Department of Bioengineering, Imperial College London, London, SW7 2BX, UK(2)
| | - Francesco Cancilla
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, Italy
| | - Mariano Licciardi
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, Italy
| | - Giovanna Pitarresi
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, Italy
| | - Luigi Tranchina
- Advanced Technologies Network (ATeN) Center, University of Palermo, Viale delle Scienze, edificio 18a, Palermo, 90128, Italy
| | - Maurizio Marrale
- Department of Physics and Chemistry "Emilio Segrè", University of Palermo, Viale delle Scienze, edificio 18, Palermo, 90128, Italy; National Institute for Nuclear Physics (INFN), Catania Division, Via Santa Sofia,64, Catania, 95123, Italy
| | - Fabio Salvatore Palumbo
- Department of Biological, Chemical, and Pharmaceutical Sciences and Technologies (STEBICEF), University of Palermo, Via Archirafi 32, Palermo, Italy; Istituto per la Ricerca e Innovazione Biomedica (IRIB), CNR, Via Ugo La Malfa, 153, 90146, Palermo, Italy.
| |
Collapse
|
36
|
Ladd B, Gräslund T, Chotteau V. Harnessing cell aggregates for enhanced adeno-associated virus manufacturing: Cultivation strategies and scale-up considerations. Biotechnol Prog 2025:e3522. [PMID: 39846514 DOI: 10.1002/btpr.3522] [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: 07/08/2024] [Revised: 11/11/2024] [Accepted: 11/20/2024] [Indexed: 01/24/2025]
Abstract
The possibility to produce recombinant adeno-associated virus (rAAV) by adherent HEK293T cells was studied in a stirred tank bioreactor (STR) culture of cell aggregates. A proof-of-concept of rAAV production was successfully demonstrated in a process where single cells were first expanded, then cell aggregates were formed by dilution into a different medium 1 day before triple plasmid transfection was conducted. An alternative approach for the STR inoculation using a seed taken from a high cell density perfusion (HCDP) culture was also investigated. It was, however, found that the spent medium of the HCDP inhibited the transfection of HEK293T cell aggregates, which was confirmed when testing with single-cell suspension culture. The formation of aggregates in shaken multi-well plates was also investigated to develop a screening system using the average power input as a scale-down criterion, which revealed that cell aggregates could be generated in 12-well plates, however with a larger size than in a STR. Taking into account the reported higher rAAV production of adherent cells in comparison with single cells for triple-plasmid transfection, HEK293T cell aggregates can possibly surpass single-cell suspension in space-time rAAV yield. The formation of HEK293T cell aggregates in a STR system offers a promising approach for scaling up and intensifying rAAV production by triple-plasmid transfection, in comparison with traditional 2D scale-up methods.
Collapse
Affiliation(s)
- Brian Ladd
- AdBIOPRO, VINNOVA Competence Centre for Advanced Bioproduction by Continuous Processing, Royal Institute of Technology (KTH), Stockholm, Sweden
- Department of Industrial Biotechnology, School of Engineering Sciences in Chemistry, Biotechnology and Health, Royal Institute of Technology (KTH), Stockholm, Sweden
| | - Torbjörn Gräslund
- AdBIOPRO, VINNOVA Competence Centre for Advanced Bioproduction by Continuous Processing, Royal Institute of Technology (KTH), Stockholm, Sweden
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, Royal Institute of Technology (KTH), Stockholm, Sweden
| | - Véronique Chotteau
- AdBIOPRO, VINNOVA Competence Centre for Advanced Bioproduction by Continuous Processing, Royal Institute of Technology (KTH), Stockholm, Sweden
- Department of Industrial Biotechnology, School of Engineering Sciences in Chemistry, Biotechnology and Health, Royal Institute of Technology (KTH), Stockholm, Sweden
| |
Collapse
|
37
|
Birjandi AA, Sharpe P. The Secretome of the Inductive Tooth Germ Exhibits Signals Required for Tooth Development. Bioengineering (Basel) 2025; 12:96. [PMID: 40001617 PMCID: PMC11851894 DOI: 10.3390/bioengineering12020096] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2024] [Revised: 12/19/2024] [Accepted: 12/26/2024] [Indexed: 02/27/2025] Open
Abstract
Teeth develop from reciprocal signaling between inductive and receptive cells. The inductive signals for tooth development are initially in the epithelium of the developing branchial arch, but later shift to the underlying mesenchyme of a developing tooth germ. The inductive signals that are needed for tooth development have not yet been fully identified. Our lab previously provided a basis for bioengineering new teeth by separating the tooth germ's epithelial and mesenchyme cells into a single cell population and recombing them. This approach, however, is not clinically applicable as the cells lose their inductive ability when expanded in vitro. In this study, we investigate whether the secretome and small extracellular vehicles (sEV) derived from inductive tooth germ mesenchyme can contribute to inductive signals required for tooth development. To address this, small extracellular vesicles and secretome were purified from inductive tooth germ mesenchyme and characterized. We investigated the proteome of sEV and proteome of inductive tooth germ mesenchyme and the impact of the culture condition and duration on the proteome. Additionally, we investigated the transcriptomic changes in tooth germ epithelium after treatment with sEV from inductive tooth germ mesenchyme. We show that culture duration of inductive tooth germ mesenchyme has an impact on the proteome of sEV purified from these cells. Similarly, culturing these cells in 2D and 3D environments results in different protein content. Proteome unique to sEV derived from inductive shows an association with multiple signaling pathways related to tooth development. Our RNASeq results show that treatment of tooth germ epithelial cells with small extracellular vesicles derived from inductive tooth germ mesenchyme results in an increased expression of some of the known odontogenic genes. Whilst further analysis is required to harvest the full potential of these sEV, our results suggests that extracellular vehicles contribute to signals required during tooth development, potentially through modulation of cellular metabolism and ECM organization.
Collapse
Affiliation(s)
| | - Paul Sharpe
- Centre for Craniofacial and Regenerative Biology, Faculty of Dentistry, Oral & Craniofacial Sciences, King’s College London, London SE1 9RT, UK
| |
Collapse
|
38
|
Shahbazi AS, Irandoost F, Mahdavian R, Shojaeilangari S, Allahvardi A, Naderi-Manesh H. A multi-stage weakly supervised design for spheroid segmentation to explore mesenchymal stem cell differentiation dynamics. BMC Bioinformatics 2025; 26:20. [PMID: 39825265 PMCID: PMC11742216 DOI: 10.1186/s12859-024-06031-x] [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: 05/13/2024] [Accepted: 12/27/2024] [Indexed: 01/20/2025] Open
Abstract
There is a growing interest in utilizing 3D culture models for stem cell and cancer cell research due to their closer resemblance to in vivo environments. In this study, human mesenchymal stem cells (MSCs) were cultured using adipocytes and osteocytes as differentiative mediums on varying concentrations of chitosan substrate. Light microscopy was employed to capture cell images from the first day to the 21st day of differentiation. Accurate image segmentation is crucial for analyzing the morphological features of the spheroids during the experimental period and for understanding MSC differentiation dynamics for therapeutic applications. Therefore, we developed an innovative, weakly supervised model, aided by convolutional neural networks, to perform label-free spheroid segmentation. Since obtaining pixel-level ground truth labels through manual annotation is labor-intensive, our approach improves the overall quality of the ground-truth map by incorporating a multi-stage process within a weakly supervised learning framework. Additionally, we developed a robust learning scheme for spheroid detection, providing a reliable foundation to study MSC differentiation dynamics. The proposed framework was systematically evaluated using low-resolution microscopic data and challenging, noisy backgrounds. The experimental results demonstrate the effectiveness of our segmentation approach in accurately separating the spheroid from the background. Furthermore, it achieves performance comparable to fully supervised state-of-the-art approaches. To quantitatively evaluate our algorithm, extensive experiments were conducted using available annotated data, confirming the reliability and robustness of our method. Our computationally extracted features can confirm the experimental results regarding alterations in MSC viability, attachment, and differentiation dynamics among the substrates with three concentrations of chitosan used. We observed the formation of more compact spheroids with higher solidity and convex area, resulting improved cell attachment and viability on the 2% chitosan substrate. Additionally, this substrate exhibited a higher propensity for differentiation into osteocytes, as evidenced by the formation of smaller and more ellipsoid spheroids. "Chitosan biofilms mimic in vivo environments for stem cell culture, advancing therapeutic and fundamental applications.” "Innovative weakly supervised model enables label-free spheroid segmentation in stem cell differentiation studies.” "Robust learning scheme achieves accurate spheroid separation, comparable to state-of-the-art approaches.”
Collapse
Affiliation(s)
- Arash Shahbazpoor Shahbazi
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, 14115-111, Iran
| | - Farzin Irandoost
- Department of Physics, Shahid Beheshti University (SBU Physics), Tehran, Iran
| | - Reza Mahdavian
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, 14115-111, Iran
| | - Seyedehsamaneh Shojaeilangari
- Biomedical Engineering Group, Department of Electrical and Information Technology, Iranian Research Organization for Science and Technology (IROST), Tehran, 33535111, Iran.
| | - Abdollah Allahvardi
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, 14115-111, Iran
| | - Hossein Naderi-Manesh
- Department of Biophysics, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, 14115-111, Iran.
| |
Collapse
|
39
|
Wallace J, McElroy MC, Klausner M, Corley R, Ayehunie S. Two- and Three-Dimensional Culture Systems: Respiratory In Vitro Tissue Models for Chemical Screening and Risk-Based Decision Making. Pharmaceuticals (Basel) 2025; 18:113. [PMID: 39861174 PMCID: PMC11768377 DOI: 10.3390/ph18010113] [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: 12/20/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Risk of lung damage from inhaled chemicals or substances has long been assessed using animal models. However, New Approach Methodologies (NAMs) that replace, reduce, and/or refine the use of animals in safety testing such as 2D and 3D cultures are increasingly being used to understand human-relevant toxicity responses and for the assessment of hazard identification. Here we review 2D and 3D lung models in terms of their application for inhalation toxicity assessment. We highlight a key case study for the Organization for Economic Cooperation and Development (OECD), in which a 3D model was used to assess human toxicity and replace the requirement for a 90-day inhalation toxicity study in rats. Finally, we consider the regulatory guidelines for the application of NAMs and potential use of different lung models for aerosol toxicity studies depending on the regulatory requirement/context of use.
Collapse
Affiliation(s)
| | | | | | - Richard Corley
- Greek Creek Toxicokinetics Consulting LLC, Boise, ID 83714, USA;
| | | |
Collapse
|
40
|
Lee Y, Choi Y, Chun JL, Kim HB, Kim S, Kim ES, Park S. High-throughput microfluidic spheroid technology for early detection of colistin-induced nephrotoxicity with gradient-based analysis. LAB ON A CHIP 2025; 25:275-284. [PMID: 39691981 DOI: 10.1039/d4lc00782d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/19/2024]
Abstract
Colistin is essential for treating multidrug-resistant Gram-negative bacterial infections but has significant nephrotoxic side effects. Traditional approaches for studying colistin's nephrotoxicity are challenged by the rapid metabolism of its prodrug, colistin methanesulfonate and the difficulty of obtaining adequate plasma from critically ill patients. To address these challenges, we developed the Spheroid Nephrotoxicity Assessing Platform (SNAP), a microfluidic device that efficiently detects colistin-induced toxicity in renal proximal tubular epithelial cell (RPTEC) spheroids within 48 hours using just 200 μL of patient plasma. Our findings demonstrate that SNAP not only promotes higher expression of kidney-specific markers aquaporin-1 (AQP1) and low-density lipoprotein receptor-related protein 2 (LRP2) compared to traditional two-dimensional (2D) cultures, but also exhibits increased sensitivity to colistin, with significant toxicity detected at concentrations of 50 μg ml-1 and above. Notably, SNAP's non-invasive method did not identify nephrotoxicity in plasma from healthy donors, thereby confirming its physiological relevance and showcasing superior sensitivity over 2D cultures, which yielded false-positive results. In clinical validation, SNAP accurately identified patients at risk of colistin-induced nephrotoxicity with 100% accuracy for both early and late onset and demonstrated a 75% accuracy rate in predicting the non-occurrence of nephrotoxicity. These results underline the potential of SNAP in personalized medicine, offering a non-invasive, precise and efficient tool for the assessment of antibiotic-induced nephrotoxicity, thus enhancing the safety and efficacy of treatments against resistant bacterial infections.
Collapse
Affiliation(s)
- Yugyeong Lee
- Department of Biomedical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Korea.
| | - Yunsang Choi
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, 13620, Korea.
| | - Ju Lan Chun
- National Institute of Animal Science, Rural Development Administration, Wanjugun, Jeollabukdo, 55365, Korea
| | - Hong Bin Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, 13620, Korea.
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Sejoong Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, 13620, Korea.
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Eu Suk Kim
- Department of Internal Medicine, Seoul National University Bundang Hospital, Seongnam, 13620, Korea.
- Department of Internal Medicine, Seoul National University College of Medicine, Seoul, 03080, Korea
| | - Sungsu Park
- Department of Biomedical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Korea.
- School of Mechanical Engineering, Sungkyunkwan University (SKKU), Suwon, 16419, Korea
- Department of Biophysics, Institute of Quantum Biophysics (IQB), Sungkyunkwan University (SKKU), Suwon, 16419, Korea
| |
Collapse
|
41
|
Black V, Bafligil C, Greaves E, Zondervan KT, Becker CM, Hellner K. Modelling Endometriosis Using In Vitro and In Vivo Systems. Int J Mol Sci 2025; 26:580. [PMID: 39859296 PMCID: PMC11766166 DOI: 10.3390/ijms26020580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/03/2025] [Accepted: 01/09/2025] [Indexed: 01/27/2025] Open
Abstract
Endometriosis is a chronic inflammatory condition characterised by the presence of endometrium-like tissue outside the uterus. Despite its high prevalence and recent advances in molecular science, many aspects of endometriosis and its pathophysiology are still poorly understood. Previously, in vitro and in vivo modelling have been instrumental in establishing our current understanding of endometriosis. As the field of molecular science and the advance towards personalised medicine is ever increasing, more sophisticated models are continually being developed. These hold great potential to provide more intricate knowledge of the underlying pathophysiology and facilitate investigations into potential future approaches to diagnosis and treatment. This review provides an overview of different in vitro and in vivo models of endometriosis that are pertinent to establishing our current understanding. Moreover, we discuss new cross-cutting approaches to endometriosis modelling, such as the use of microfluidic cultures and 3D printing, which have the potential to shape the future of endometriosis research.
Collapse
Affiliation(s)
- Verity Black
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, John Radcliffe Hospital, Women’s Centre, Oxford OX3 9DU, UK; (V.B.); (K.T.Z.); (C.M.B.)
| | - Cemsel Bafligil
- Botnar Research Centre, NIHR Biomedical Research Unit Oxford, Nuffield Department of Orthopaedics, Rheumatology and Musculoskeletal Sciences, University of Oxford, Oxford OX3 7LD, UK
| | - Erin Greaves
- Division of Biomedical Sciences, Warwick Medical School, University of Warwick, Coventry CV4 7AL, UK;
| | - Krina T. Zondervan
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, John Radcliffe Hospital, Women’s Centre, Oxford OX3 9DU, UK; (V.B.); (K.T.Z.); (C.M.B.)
- Wellcome Trust Centre for Human Genetics, University of Oxford, Roosevelt Drive, Oxford OX3 7BN, UK
| | - Christian M. Becker
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, John Radcliffe Hospital, Women’s Centre, Oxford OX3 9DU, UK; (V.B.); (K.T.Z.); (C.M.B.)
| | - Karin Hellner
- Nuffield Department of Obstetrics and Gynaecology, University of Oxford, John Radcliffe Hospital, Women’s Centre, Oxford OX3 9DU, UK; (V.B.); (K.T.Z.); (C.M.B.)
| |
Collapse
|
42
|
Lederer AK, Görrissen N, Nguyen TT, Kreutz C, Rasel H, Bartsch F, Lang H, Endres K. Exploring the effects of gut microbiota on cholangiocarcinoma progression by patient-derived organoids. J Transl Med 2025; 23:34. [PMID: 39789543 PMCID: PMC11716211 DOI: 10.1186/s12967-024-06012-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: 08/21/2024] [Accepted: 12/19/2024] [Indexed: 01/12/2025] Open
Abstract
BACKGROUND Recent research indicates a role of gut microbiota in development and progression of life-threatening diseases such as cancer. Carcinomas of the biliary ducts, the so-called cholangiocarcinomas, are known for their aggressive tumor biology, implying poor prognosis of affected patients. An impact of the gut microbiota on cholangiocarcinoma development and progression is plausible due to the enterohepatic circulation and is therefore the subject of scientific debate, however evidence is still lacking. This review aimed to discuss the suitability of complex cell culture models to investigate the role of gut microbiota in cholangiocarcinoma progression. MAIN BODY Clinical research in this area is challenging due to poor comparability of patients and feasibility reasons, which is why translational models are needed to understand the basis of tumor progression in cholangiocarcinoma. A promising approach to investigate the influence of gut microbiota could be an organoid model. Organoids are 3D cell models cultivated in a modifiable and controlled condition, which can be grown from tumor tissue. 3D cell models are able to imitate physiological and pathological processes in the human body and thus contribute to a better understanding of health and disease. CONCLUSION The use of complex cell cultures such as organoids and organoid co-cultures might be powerful and valuable tools to study not only the growth behavior and growth of cholangiocarcinoma cells, but also the interaction with the tumor microenvironment and with components of the gut microbiota.
Collapse
Affiliation(s)
- Ann-Kathrin Lederer
- Department of General, Visceral and Transplantation Surgery, University Medical Center Mainz, 55131, Mainz, Germany.
- Center for Complementary Medicine, Department of Medicine II, Faculty of Medicine, Medical Center-University of Freiburg, University of Freiburg, 79106, Freiburg, Germany.
| | - Nele Görrissen
- Department of General, Visceral and Transplantation Surgery, University Medical Center Mainz, 55131, Mainz, Germany
| | - Tinh Thi Nguyen
- Department of Psychiatry and Psychotherapy, University Medical Center Mainz, 55131, Mainz, Germany
- Institute of Molecular Biology (IMB), 55128, Mainz, Germany
| | - Clemens Kreutz
- Institute of Medical Biometry and Statistics (IMBI), Faculty of Medicine and Medical Center, 79106, Freiburg, Germany
| | - Hannah Rasel
- Department of General, Visceral and Transplantation Surgery, University Medical Center Mainz, 55131, Mainz, Germany
| | - Fabian Bartsch
- Department of General, Visceral and Transplantation Surgery, University Medical Center Mainz, 55131, Mainz, Germany
| | - Hauke Lang
- Department of General, Visceral and Transplantation Surgery, University Medical Center Mainz, 55131, Mainz, Germany
| | - Kristina Endres
- Department of Psychiatry and Psychotherapy, University Medical Center Mainz, 55131, Mainz, Germany
- Faculty of Computer Sciences and Microsystems Technology, University of Applied Sciences Kaiserslautern, 66482, Zweibrücken, Germany
| |
Collapse
|
43
|
Yee MMF, Chin KY, Ima-Nirwana S, Alias E, Chua KH, Wong SK. Evaluation of bone-protecting effects of palm carotene mixture in two- and three-dimensional osteoblast/osteoclast co-culture systems. Int J Med Sci 2025; 22:585-603. [PMID: 39898246 PMCID: PMC11783079 DOI: 10.7150/ijms.103445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Accepted: 12/19/2024] [Indexed: 02/04/2025] Open
Abstract
Background: Carotene exists naturally in a complex mixture consisting of alpha (α), beta (β), and gamma (γ)-isoforms. Previous studies investigated the effects of individual carotene isomers on bone rather than their actions in a mixture. Purpose: This study explored the bone-protective properties of palm carotene mixture using both two- and three-dimensional co-culture systems. Study design: The viability of human foetal osteoblasts (hFOB 1.19), viability of human monocytic cell line (THP-1), osteoblast differentiation, osteoclast maturation, bone quality and strength were assessed in two- and three-dimensional co-culture system after treatment of palm carotene mixture. Methods: The viability of hFOB 1.19 and THP-1 was determined on day 1, 3, and 6 following treatment of palm carotene mixture. The osteoblast-osteoclast co-culture (ratio of hFOB 1.19 to THP-1 = 2:1) was treated with palm carotene mixture as well as subjected to alkaline phosphatase (ALP) and tartrate resistant acid phosphatase (TRAP) staining on day 21 to assess the osteoblast proliferation and osteoclast maturation. Dual-energy X-ray absorptiometry, micro-computed tomography, universal testing machine, and bone histomorphometry were used to assess the bone parameters of scaffolds co-cultured with osteoblasts and osteoclasts. Results: Palm carotene mixture (3.13 - 50 μg/mL) increased osteoblast viability. Monocyte viability decreased in lower concentration (3.13 - 12.5 μg/mL) but increased in higher concentration (25 - 50 μg/mL) of palm carotene mixture. Treatment with palm carotene mixture (12.5 µg/mL) demonstrated earlier peak for the ALP-positive area on day 14 but decreased total number of TRAP-positive multinucleated cells on day 21. Palm carotene mixture also increased bone volume and osteoblast number in the three-dimensional co-culture system. Conclusion: Palm carotene mixture potentially exhibits beneficial effects on bone by accelerating osteoblast proliferation and suppressing osteoclast maturation. The findings of current study serve as the basis for the further validation through animal experiments and human trials.
Collapse
Affiliation(s)
- Michelle Min-Fang Yee
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Kok-Yong Chin
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Soelaiman Ima-Nirwana
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Ekram Alias
- Department of Biochemistry, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Kien Hui Chua
- Department of Physiology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Malaysia
| | - Sok Kuan Wong
- Department of Pharmacology, Faculty of Medicine, Universiti Kebangsaan Malaysia, Jalan Yaacob Latif, Bandar Tun Razak, 56000 Cheras, Kuala Lumpur, Malaysia
| |
Collapse
|
44
|
Akagi S, Ando H, Matsuo CNA, Tajima K, Takata H, Matsushima T, Kusano T, Ishida T. A 3D Cell-Culture System That Uses Nano-Fibrillated Bacterial Cellulose to Prepare a Spherical Formulation of Culture Cells. Biol Pharm Bull 2025; 48:23-32. [PMID: 39864853 DOI: 10.1248/bpb.b24-00804] [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: 01/28/2025]
Abstract
A 3-dimensional (3D) cell culture is now being actively pursued to accomplish the in vivo-like cellular morphology and biological functions in cell culture. We recently obtained nano-fibrillated bacterial cellulose (NFBC). In this study, we developed a novel NFBC-based 3D cell-culture system, the OnGel method, and the Suspension method. HepG2 human liver cancer cells were cultured via these methods and formed spherical formulations in the optimized condition, 1.0% (w/v) of NFBC in the OnGel method, and 0.06-0.10% (w/v) of NFBC in the Suspension method. Non-cancerous cells such as human-induced pluripotent stem (iPS) cells and human mesenchymal stem cells (MSCs) also formed spherical formulations. It is noteworthy that both the size and cell viability of spheroids prepared via these methods were comparable to those cultured using commercially available 3D cell-culture systems. Both OnGel and Suspension methods are less complicated than the existing 3D cell-culture systems, which is an invaluable advantage for the preparation of cancer spheroids. The NFBC-based 3D cell-culture systems introduced here show great promise as a tool to prepare cultures for cell-derived spheroids for the progress of both in vitro and in vivo studies of the biological functioning of cells.
Collapse
Affiliation(s)
- Shunsuke Akagi
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Hidenori Ando
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
- Innovative Research Center for Drug Delivery System, Institute of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Cristina Nana Amorim Matsuo
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | - Kenji Tajima
- Faculty of Engineering, Hokkaido University, Sapporo 060-8628, Japan
| | - Haruka Takata
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
- Innovative Research Center for Drug Delivery System, Institute of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
| | | | | | - Tatsuhiro Ishida
- Department of Pharmacokinetics and Biopharmaceutics, Institute of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
- Innovative Research Center for Drug Delivery System, Institute of Biomedical Sciences, Tokushima University, Tokushima 770-8505, Japan
| |
Collapse
|
45
|
Anandi L, Garcia J, Ros M, Janská L, Liu J, Carmona-Fontaine C. Direct visualization of emergent metastatic features within an ex vivo model of the tumor microenvironment. Life Sci Alliance 2025; 8:e202403053. [PMID: 39419548 PMCID: PMC11487089 DOI: 10.26508/lsa.202403053] [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: 09/19/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 10/19/2024] Open
Abstract
Ischemic conditions such as hypoxia and nutrient starvation, together with interactions with stromal cells, are critical drivers of metastasis. These conditions arise deep within tumor tissues, and thus, observing nascent metastases is exceedingly challenging. We thus developed the 3MIC-an ex vivo model of the tumor microenvironment-to study the emergence of metastatic features in tumor cells in a 3-dimensional (3D) context. Here, tumor cells spontaneously create ischemic-like conditions, allowing us to study how tumor spheroids migrate, invade, and interact with stromal cells under different metabolic conditions. Consistent with previous data, we show that ischemia increases cell migration and invasion, but the 3MIC allowed us to directly observe and perturb cells while they acquire these pro-metastatic features. Interestingly, our results indicate that medium acidification is one of the strongest pro-metastatic cues and also illustrate using the 3MIC to test anti-metastatic drugs on cells experiencing different metabolic conditions. Overall, the 3MIC can help dissecting the complexity of the tumor microenvironment for the direct observation and perturbation of tumor cells during the early metastatic process.
Collapse
Affiliation(s)
- Libi Anandi
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Jeremy Garcia
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Manon Ros
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Libuše Janská
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Josephine Liu
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| | - Carlos Carmona-Fontaine
- Center for Genomics & Systems Biology, Department of Biology, New York University, New York, NY, USA
| |
Collapse
|
46
|
Maity S, Bhuyan T, Jewell C, Kawakita S, Sharma S, Nguyen HT, Najafabadi AH, Ermis M, Falcone N, Chen J, Mandal K, Khorsandi D, Yilgor C, Choroomi A, Torres E, Mecwan M, John JV, Akbari M, Wang Z, Moniz-Garcia D, Quiñones-Hinojosa A, Jucaud V, Dokmeci MR, Khademhosseini A. Recent Developments in Glioblastoma-On-A-Chip for Advanced Drug Screening Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2405511. [PMID: 39535474 PMCID: PMC11719323 DOI: 10.1002/smll.202405511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/08/2024] [Indexed: 11/16/2024]
Abstract
Glioblastoma (GBM) is an aggressive form of cancer, comprising ≈80% of malignant brain tumors. However, there are no effective treatments for GBM due to its heterogeneity and the presence of the blood-brain barrier (BBB), which restricts the delivery of therapeutics to the brain. Despite in vitro models contributing to the understanding of GBM, conventional 2D models oversimplify the complex tumor microenvironment. Organ-on-a-chip (OoC) models have emerged as promising platforms that recapitulate human tissue physiology, enabling disease modeling, drug screening, and personalized medicine. There is a sudden increase in GBM-on-a-chip models that can significantly advance the knowledge of GBM etiology and revolutionize drug development by reducing animal testing and enhancing translation to the clinic. In this review, an overview of GBM-on-a-chip models and their applications is reported for drug screening and discussed current challenges and potential future directions for GBM-on-a-chip models.
Collapse
Affiliation(s)
- Surjendu Maity
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
- Department of Orthopedic Surgery, Duke University School of
Medicine, Duke University, Durham, NC 27705
| | - Tamanna Bhuyan
- Department of Applied Biology, School of Biological
Sciences, University of Science & Technology Meghalaya, Meghalaya, 793101,
India
| | - Christopher Jewell
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Satoru Kawakita
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Saurabh Sharma
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Huu Tuan Nguyen
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | | | - Menekse Ermis
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
- Center of Excellence in Biomaterials and Tissue
Engineering, Middle East Technical University, Ankara, Turkey
| | - Natashya Falcone
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Junjie Chen
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Kalpana Mandal
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Danial Khorsandi
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Can Yilgor
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Auveen Choroomi
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Emily Torres
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Marvin Mecwan
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Johnson V. John
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Mohsen Akbari
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
- Laboratoryfor Innovations in Micro Engineering (LiME),
Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2,
Canada
- Biotechnology Center, Silesian University of Technology,
Akademicka 2A, 44-100 Gliwice, Poland
| | - Zhaohui Wang
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | | | | | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | | | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| |
Collapse
|
47
|
Da Silva K, Kumar P, Choonara YE. The paradigm of stem cell secretome in tissue repair and regeneration: Present and future perspectives. Wound Repair Regen 2025; 33:e13251. [PMID: 39780313 PMCID: PMC11711308 DOI: 10.1111/wrr.13251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 12/04/2024] [Accepted: 12/13/2024] [Indexed: 01/11/2025]
Abstract
As the number of patients requiring organ transplants continues to rise exponentially, there is a dire need for therapeutics, with repair and regenerative properties, to assist in alleviating this medical crisis. Over the past decade, there has been a shift from conventional stem cell treatments towards the use of the secretome, the protein and factor secretions from cells. These components may possess novel druggable targets and hold the key to profoundly altering the field of regenerative medicine. Despite the progress in this field, clinical translation of secretome-containing products is limited by several challenges including but not limited to ensuring batch-to-batch consistency, the prevention of further heterogeneity, production of sufficient secretome quantities, product registration, good manufacturing practice protocols and the pharmacokinetic/pharmacodynamic profiles of all the components. Despite this, the secretome may hold the key to unlocking the regenerative blockage scientists have encountered for years. This review critically analyses the secretome derived from different cell sources and used in several tissues for tissue regeneration. Furthermore, it provides an overview of the current delivery strategies and the future perspectives for the secretome as a potential therapeutic. The success and possible shortcomings of the secretome are evaluated.
Collapse
Affiliation(s)
- Kate Da Silva
- Wits Advanced Drug Delivery Platform (WADDP) Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Pradeep Kumar
- Wits Advanced Drug Delivery Platform (WADDP) Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| | - Yahya E. Choonara
- Wits Advanced Drug Delivery Platform (WADDP) Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health SciencesUniversity of the WitwatersrandJohannesburgSouth Africa
| |
Collapse
|
48
|
Han S, Park S. Spheroid Formation and Easy and Stable Control of Stromal Cells Using Multi-inlet Spheroid Generator. Methods Mol Biol 2025; 2924:217-222. [PMID: 40307645 DOI: 10.1007/978-1-0716-4530-7_15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025]
Abstract
Tumor spheroids have been used as a powerful tool for testing anti-cancer drugs or understanding tumors in vivo due to their structural and physiological recapitulation of human solid tumors. Furthermore, when co-cultured with stromal cells, it is much more relevant to tumor in vivo than cancer cell alone, because stromal cells in tumor microenvironment play major roles in cancer progression and drug resistance. This protocol describes how to generate a spheroid with a controlled stromal cell ratio using a multi-inlet spheroid generator.
Collapse
Affiliation(s)
- Seokgyu Han
- School of Mechanical Engineering, Sungkyunkwan University, Suwon, South Korea
| | - Sungsu Park
- School of Mechanical Engineering, Sungkyunkwan University, Suwon, South Korea
| |
Collapse
|
49
|
Murkar RS, Wiese-Rischke C, Weigel T, Kopp S, Walles H. Developing human upper, lower, and deep lung airway models: Combining different scaffolds and developing complex co-cultures. J Tissue Eng 2025; 16:20417314241299076. [PMID: 39885949 PMCID: PMC11780661 DOI: 10.1177/20417314241299076] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Accepted: 10/27/2024] [Indexed: 02/01/2025] Open
Abstract
Advanced in vitro models are crucial for studying human airway biology. Our objective was the development and optimization of 3D in vitro models representing diverse airway regions, including deep lung alveolar region. This initiative was aimed at assessing the influence of selective scaffold materials on distinct airway co-culture models. While PET membranes (30 µm thickness) were unsuitable for alveolar models due to their stiffness and relatively high Young's modulus, a combination of collagenous scaffolds seeded with Calu-3 cells and fibroblasts, showed increased mucus production going from week 1 to week 4 of air lift culture. Meanwhile standard electrospun polymer membrane (50-60 µm thick), which possesses a considerably low modulus of elasticity, offered higher flexibility and supported co-cultures of primary alveolar epithelial (huAEC) and endothelial cells (hEC) in concert with lung biopsy-derived fibroblasts which enhanced maturation of the tissue model. As published, designing human alveolar in vitro models require thin scaffold to mimic the required ultra-thin ECM, in addition to assuring right balanced AT1/AT2 ratio for biomimetic representation. We concluded that co-cultivation of primary/stem cells or cell lines has a higher influence on the function of the airway tissue models than the applied scaffolds.
Collapse
Affiliation(s)
- Rasika S Murkar
- Core Facility Tissue Engineering, Institute of Chemistry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Cornelia Wiese-Rischke
- University Clinic for Cardiac and Thoracic Surgery, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Tobias Weigel
- Fraunhofer Translational Center for Regenerative Medicine, Fraunhofer ISC, Wuerzburg, Germany
| | - Sascha Kopp
- Core Facility Tissue Engineering, Institute of Chemistry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| | - Heike Walles
- Core Facility Tissue Engineering, Institute of Chemistry, Otto-von-Guericke-University Magdeburg, Magdeburg, Germany
| |
Collapse
|
50
|
Zigan C, Benito Alston C, Chatterjee A, Solorio L, Chan DD. Characterization of Composite Agarose-Collagen Hydrogels for Chondrocyte Culture. Ann Biomed Eng 2025; 53:120-132. [PMID: 39277549 PMCID: PMC11782374 DOI: 10.1007/s10439-024-03613-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: 05/24/2024] [Accepted: 09/01/2024] [Indexed: 09/17/2024]
Abstract
To elucidate the mechanisms of cellular mechanotransduction, it is necessary to employ biomaterials that effectively merge biofunctionality with appropriate mechanical characteristics. Agarose and collagen separately are common biopolymers used in cartilage mechanobiology and mechanotransduction studies but lack features that make them ideal for functional engineered cartilage. In this study, agarose is blended with collagen type I to create hydrogels with final concentrations of 4% w/v or 2% w/v agarose with 2 mg/mL collagen. We hypothesized that the addition of collagen into a high-concentration agarose hydrogel does not diminish mechanical properties. Acellular and cell-laden studies were completed to assess rheologic and compressive properties, contraction, and structural homogeneity in addition to cell proliferation and sulfated glycosaminoglycan production. Over 21 days in culture, cellular 4% agarose-2 mg/mL collagen I hydrogels seeded with primary murine chondrocytes displayed structural and bulk mechanical behaviors that did not significantly alter from 4% agarose-only hydrogels, cell proliferation, and continual glycosaminoglycan production, indicating promise toward the development of an effective hydrogel for chondrocyte mechanotransduction and mechanobiology studies.
Collapse
Affiliation(s)
- Clarisse Zigan
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | | | - Aritra Chatterjee
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
- Department of Mechanical Engineering, Birla Institute of Technology and Science, Pilani, Hyderabad Campus, Hyderabad, Telangana, India
| | - Luis Solorio
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA
| | - Deva D Chan
- Weldon School of Biomedical Engineering, Purdue University, West Lafayette, IN, USA.
- School of Mechanical Engineering, Purdue University, West Lafayette, IN, USA.
| |
Collapse
|