ijms-logo

Journal Browser

Journal Browser

Organoid Culture and Characterization Systems for Tissue Engineering Applications

A special issue of International Journal of Molecular Sciences (ISSN 1422-0067). This special issue belongs to the section "Molecular Biology".

Deadline for manuscript submissions: 30 September 2024 | Viewed by 9454

Special Issue Editors


E-Mail Website
Guest Editor
Pathology Department, Faculty of Medicine and Odonthology, University of Valencia, Blasco Ibanez Avenue, 17, 46010 Valencia, Spain
Interests: tissue engineering; cell culture; molecular biology; cartilage regeneration; lung cancer

E-Mail Website
Guest Editor
Pathology Department, Faculty of Medicine and Odonthology, University of Valencia, Blasco Ibanez Avenue, 17, 46010 Valencia, Spain
Interests: human histology; experimental pathology; cell culture; tissue engineering; artificial organs

Special Issue Information

Dear Colleagues,

Recently, the use of organoids is gaining great interest in areas as different from tissue engineering as the manufacture of substitutes for different tissues, like cartilage or skin, or the construction of models for the study of pathophysiological aspects in complex diseases such as cancer or Alzheimer's. The literature shows great heterogeneity in terms of the methodology used for the isolation and/or construction of organoids (using stem cells, human induced pluripotent stem cell (hiPSC) and/or isolated stromal cells), as well as in the different matrices (native decellularized or manufactured) used. The selection of different study parameters (spheroid morphology, matrix generation, angiogenesis, gene expression, etc.) also vary enormously in relation to the final objective of each investigation. Each specific application of organoids has its particular conditions, so it is convenient to have an idea of what is the best approach when generating study systems based on organoids. The objective of this Special Issue is, on the one hand, to define some suitable models for the construction of organoid-based culture systems for specific applications such as, for example, the regeneration of skeletal tissues, the study of cancer or the design of platforms for drug screening for precision medicine approaches. On the other hand, we propose to define which parameters need to be analyzed and by which methodologies, for each specific application.

Prof. Dr. Manuel Mata
Dr. María Sancho-Tello
Guest Editors

Manuscript Submission Information

Manuscripts should be submitted online at www.mdpi.com by registering and logging in to this website. Once you are registered, click here to go to the submission form. Manuscripts can be submitted until the deadline. All submissions that pass pre-check are peer-reviewed. Accepted papers will be published continuously in the journal (as soon as accepted) and will be listed together on the special issue website. Research articles, review articles as well as short communications are invited. For planned papers, a title and short abstract (about 100 words) can be sent to the Editorial Office for announcement on this website.

Submitted manuscripts should not have been published previously, nor be under consideration for publication elsewhere (except conference proceedings papers). All manuscripts are thoroughly refereed through a single-blind peer-review process. A guide for authors and other relevant information for submission of manuscripts is available on the Instructions for Authors page. International Journal of Molecular Sciences is an international peer-reviewed open access semimonthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. There is an Article Processing Charge (APC) for publication in this open access journal. For details about the APC please see here. Submitted papers should be well formatted and use good English. Authors may use MDPI's English editing service prior to publication or during author revisions.

Keywords

  • organoid manufacture
  • organoids isolation
  • 3D culture
  • precision medicine
  • disease modeling
  • stem cells
  • hiPSC
  • decellularized extracellular matrix

Published Papers (6 papers)

Order results
Result details
Select all
Export citation of selected articles as:

Research

Jump to: Review

21 pages, 3728 KiB  
Article
Collagen Lattice Model, Populated with Heterogeneous Cancer-Associated Fibroblasts, Facilitates Advanced Reconstruction of Pancreatic Cancer Microenvironment
by Xiaoyu Song, Yuma Nihashi, Yukiko Imai, Nobuhito Mori, Noritaka Kagaya, Hikaru Suenaga, Kazuo Shin-ya, Masamichi Yamamoto, Daiki Setoyama, Yuya Kunisaki and Yasuyuki S. Kida
Int. J. Mol. Sci. 2024, 25(7), 3740; https://doi.org/10.3390/ijms25073740 - 27 Mar 2024
Viewed by 516
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is a solid-tumor malignancy. To enhance the treatment landscape of PDAC, a 3D model optimized for rigorous drug screening is essential. Within the PDAC tumor microenvironment, a dense stroma comprising a large extracellular matrix and cancer-associated fibroblasts (CAFs) is [...] Read more.
Pancreatic ductal adenocarcinoma (PDAC) is a solid-tumor malignancy. To enhance the treatment landscape of PDAC, a 3D model optimized for rigorous drug screening is essential. Within the PDAC tumor microenvironment, a dense stroma comprising a large extracellular matrix and cancer-associated fibroblasts (CAFs) is well-known for its vital role in modulating tumor growth, cellular heterogeneity, bidirectional paracrine signaling, and chemoresistance. In this study, we employed a fibroblast-populated collagen lattice (FPCL) modeling approach that has the ability to replicate fibroblast contractility in the collagenous matrix to build dense stroma. This FPCL model allows CAF differentiation by facilitating multifaceted cell–cell interactions between cancer cells and CAFs, with the differentiation further influenced by mechanical forces and hypoxia carried within the 3D structure. Our FPCL models displayed hallmark features, including ductal gland structures and differentiated CAFs with spindle shapes. Through morphological explorations alongside in-depth transcriptomic and metabolomic profiling, we identified substantial molecular shifts from the nascent to mature model stages and potential metabolic biomarkers, such as proline. The initial pharmacological assays highlighted the effectiveness of our FPCL model in screening for improved therapeutic strategies. In conclusion, our PDAC modeling platform mirrors complex tumor microenvironmental dynamics and offers an unparalleled perspective for therapeutic exploration. Full article
Show Figures

Figure 1

15 pages, 5413 KiB  
Article
Adipose Tissue-Derived Mesenchymal Stem Cells Extend the Lifespan and Enhance Liver Function in Hepatocyte Organoids
by Sun A Ock, Seo-Yeon Kim, Won Seok Ju, Young-Im Kim, Ha-Yeon Wi and Poongyeon Lee
Int. J. Mol. Sci. 2023, 24(20), 15429; https://doi.org/10.3390/ijms242015429 - 21 Oct 2023
Viewed by 1610
Abstract
In this study, we generated hepatocyte organoids (HOs) using frozen-thawed primary hepatocytes (PHs) within a three-dimensional (3D) Matrigel dome culture in a porcine model. Previously studied hepatocyte organoid analogs, spheroids, or hepatocyte aggregates created using PHs in 3D culture systems have limitations in [...] Read more.
In this study, we generated hepatocyte organoids (HOs) using frozen-thawed primary hepatocytes (PHs) within a three-dimensional (3D) Matrigel dome culture in a porcine model. Previously studied hepatocyte organoid analogs, spheroids, or hepatocyte aggregates created using PHs in 3D culture systems have limitations in their in vitro lifespans. By co-culturing adipose tissue-derived mesenchymal stem cells (A-MSCs) with HOs within a 3D Matrigel dome culture, we achieved a 3.5-fold increase in the in vitro lifespan and enhanced liver function compared to a conventional two-dimensional (2D) monolayer culture, i.e., more than twice that of the HO group cultured alone, reaching up to 126 d. Although PHs were used to generate HOs, we identified markers associated with cholangiocyte organoids such as cytokeratin 19 and epithelial cellular adhesion molecule (EPCAM). Co-culturing A-MSCs with HOs increased the secretion of albumin and urea and glucose consumption compared to HOs cultured alone. After more than 100 d, we observed the upregulation of tumor protein P53 (TP53)-P21 and downregulation of EPCAM, albumin (ALB), and cytochrome P450 family 3 subfamily A member 29 (CYP3A29). Therefore, HOs with function and longevity improved through co-culturing with A-MSCs can be used to create large-scale human hepatotoxicity testing models and precise livestock nutrition assessment tools. Full article
Show Figures

Figure 1

12 pages, 2551 KiB  
Article
A Strainer-Based Platform for the Collection and Immunolabeling of Mouse Intestinal Organoids
by Jinlong Tan, Yinju Liu, Weike Li, Guohua Chen, Yongxiang Fang, Xiaobing He, Baoquan Fu and Zhizhong Jing
Int. J. Mol. Sci. 2023, 24(17), 13568; https://doi.org/10.3390/ijms241713568 - 01 Sep 2023
Cited by 1 | Viewed by 879
Abstract
Intestinal organoids have emerged as powerful model systems for studying the complex structure and function of the intestine. However, there is a lack of widely applicable methods for the collection, labeling, and imaging of intestinal organoids. In this study, we developed a novel [...] Read more.
Intestinal organoids have emerged as powerful model systems for studying the complex structure and function of the intestine. However, there is a lack of widely applicable methods for the collection, labeling, and imaging of intestinal organoids. In this study, we developed a novel method for loading and labeling intestinal organoids, a method that efficiently collects the organoids and facilitates imaging of their three-dimensional (3D) structure. Based on this strainer platform, mouse intestinal organoids were adequately collected and immobilized, facilitating the immunolabeling workflow to target proteins of the organoids. After evaluation, the strainer size of 40 μm was considered to be more conducive to the collection and labeling of mouse intestinal organoids. More extensive research on organoids of multiple types and species origins will contribute to broadening the applicability of the methodology. Overall, our study proposes an innovative workflow for loading and analyzing intestinal organoids. The combination of a strainer-based collection method, fluorescent labeling, and 3D reconstruction provides valuable insights into the organization and complexity of these tissue models, thereby offering new avenues for investigating intestinal development, disease modeling, and drug discovery. Full article
Show Figures

Figure 1

23 pages, 9468 KiB  
Article
Partner, Neighbor, Housekeeper and Dimension: 3D versus 2D Glomerular Co-Cultures Reveal Drawbacks of Currently Used Cell Culture Models
by Anna Rederer, Victoria Rose, René Krüger, Linda Schmittutz, Izabela Swierzy, Lena Fischer, Ingo Thievessen, Julian Bauer, Oliver Friedrich, Mario Schiffer and Janina Müller-Deile
Int. J. Mol. Sci. 2023, 24(12), 10384; https://doi.org/10.3390/ijms241210384 - 20 Jun 2023
Cited by 2 | Viewed by 1918
Abstract
Signaling-pathway analyses and the investigation of gene responses to different stimuli are usually performed in 2D monocultures. However, within the glomerulus, cells grow in 3D and are involved in direct and paracrine interactions with different glomerular cell types. Thus, the results from 2D [...] Read more.
Signaling-pathway analyses and the investigation of gene responses to different stimuli are usually performed in 2D monocultures. However, within the glomerulus, cells grow in 3D and are involved in direct and paracrine interactions with different glomerular cell types. Thus, the results from 2D monoculture experiments must be taken with caution. We cultured glomerular endothelial cells, podocytes and mesangial cells in 2D/3D monocultures and 2D/3D co-cultures and analyzed cell survival, self-assembly, gene expression, cell–cell interaction, and gene pathways using live/dead assay, time-lapse analysis, bulk-RNA sequencing, qPCR, and immunofluorescence staining. Without any need for scaffolds, 3D glomerular co-cultures self-organized into spheroids. Podocyte- and glomerular endothelial cell-specific markers and the extracellular matrix were increased in 3D co-cultures compared to 2D co-cultures. Housekeeping genes must be chosen wisely, as many genes used for the normalization of gene expression were themselves affected in 3D culture conditions. The transport of podocyte-derived VEGFA to glomerular endothelial cells confirmed intercellular crosstalk in the 3D co-culture models. The enhanced expression of genes important for glomerular function in 3D, compared to 2D, questions the reliability of currently used 2D monocultures. Hence, glomerular 3D co-cultures might be more suitable in the study of intercellular communication, disease modelling and drug screening ex vivo. Full article
Show Figures

Figure 1

18 pages, 10184 KiB  
Article
Morphological Characterization of Human Lung Cancer Organoids Cultured in Type I Collagen Hydrogels: A Histological Approach
by Irene Monleón-Guinot, Lara Milian, Patricia Martínez-Vallejo, María Sancho-Tello, Mauro Llop-Miguel, José Marcelo Galbis, Antonio Cremades, Carmen Carda and Manuel Mata
Int. J. Mol. Sci. 2023, 24(12), 10131; https://doi.org/10.3390/ijms241210131 - 14 Jun 2023
Cited by 4 | Viewed by 2024
Abstract
The malignity of lung cancer is conditioned by the tumor microenvironment (TME), in which cancer-associated fibroblasts (CAFs) are relevant. In this work, we generated organoids by combining A549 cells with CAFs and normal fibroblasts (NF) isolated from adenocarcinoma tumors. We optimized the conditions [...] Read more.
The malignity of lung cancer is conditioned by the tumor microenvironment (TME), in which cancer-associated fibroblasts (CAFs) are relevant. In this work, we generated organoids by combining A549 cells with CAFs and normal fibroblasts (NF) isolated from adenocarcinoma tumors. We optimized the conditions for their manufacture in a short time. We evaluated the morphology of organoids using confocal microscopy analysis of F-actin, vimentin and pankeratin. We determined the ultrastructure of the cells in the organoids via transmission electron microscopy and the expression of CDH1, CDH2 and VIM via RT-PCR. The addition of stromal cells induces the self-organization of the organoids, which acquired a bowl morphology, as well as their growth and the generation of cell processes. They also influenced the expression of genes related to epithelial mesenchymal transition (EMT). CAFs potentiated these changes. All cells acquired a characteristic secretory phenotype, with cohesive cells appearing inside the organoids. In the periphery, many cells acquired a migratory phenotype, especially in organoids that incorporated CAFs. The deposit of abundant extracellular matrix could also be observed. The results presented here reinforce the role of CAFs in the progression of lung tumors and could lay the foundation for a useful in vitro pharmacological model. Full article
Show Figures

Figure 1

Review

Jump to: Research

27 pages, 3408 KiB  
Review
Addressing Key Questions in Organoid Models: Who, Where, How, and Why?
by María Gómez-Álvarez, Marcos Agustina-Hernández, Emilio Francés-Herrero, Adolfo Rodríguez-Eguren, Clara Bueno-Fernandez and Irene Cervelló
Int. J. Mol. Sci. 2023, 24(21), 16014; https://doi.org/10.3390/ijms242116014 - 06 Nov 2023
Viewed by 1857
Abstract
Organoids are three-dimensional cellular structures designed to recreate the biological characteristics of the body’s native tissues and organs in vitro. There has been a recent surge in studies utilizing organoids due to their distinct advantages over traditional two-dimensional in vitro approaches. However, there [...] Read more.
Organoids are three-dimensional cellular structures designed to recreate the biological characteristics of the body’s native tissues and organs in vitro. There has been a recent surge in studies utilizing organoids due to their distinct advantages over traditional two-dimensional in vitro approaches. However, there is no consensus on how to define organoids. This literature review aims to clarify the concept of organoids and address the four fundamental questions pertaining to organoid models: (i) What constitutes organoids?—The cellular material. (ii) Where do organoids grow?—The extracellular scaffold. (iii) How are organoids maintained in vitro?—Via the culture media. (iv) Why are organoids suitable in vitro models?—They represent reproducible, stable, and scalable models for biological applications. Finally, this review provides an update on the organoid models employed within the female reproductive tract, underscoring their relevance in both basic biology and clinical applications. Full article
Show Figures

Figure 1

Back to TopTop