Next Issue
Volume 2, March
Previous Issue
Volume 1, September
 
 

Organoids, Volume 1, Issue 2 (December 2022) – 5 articles

  • Issues are regarded as officially published after their release is announced to the table of contents alert mailing list.
  • You may sign up for e-mail alerts to receive table of contents of newly released issues.
  • PDF is the official format for papers published in both, html and pdf forms. To view the papers in pdf format, click on the "PDF Full-text" link, and use the free Adobe Reader to open them.
Order results
Result details
Select all
Export citation of selected articles as:
16 pages, 4069 KiB  
Article
Pancreatic Cancer 3D Cell Line Organoids (CLOs) Maintain the Phenotypic Characteristics of Organoids and Accurately Reflect the Cellular Architecture and Heterogeneity In Vivo
by Sara Noorani, Shannon R. Nelson, Neil T. Conlon, Justine Meiller, Ekaterina Shcheglova, Alice Usai, Jojanneke Stoof, Letizia Palanga, Fiona O’Neill, Sandra Roche, Maura B. Cotter, Niall Swan and Naomi Walsh
Organoids 2022, 1(2), 168-183; https://doi.org/10.3390/organoids1020013 - 12 Dec 2022
Cited by 1 | Viewed by 3702
Abstract
Pancreatic cancer is a highly lethal disease. Therapeutic resistance to chemotherapy is a major cause of treatment failure and recurrence in pancreatic cancer. Organoids derived from cancer stem cells (CSC) are promising models for the advancement of personalised therapeutic responses to inform clinical [...] Read more.
Pancreatic cancer is a highly lethal disease. Therapeutic resistance to chemotherapy is a major cause of treatment failure and recurrence in pancreatic cancer. Organoids derived from cancer stem cells (CSC) are promising models for the advancement of personalised therapeutic responses to inform clinical decisions. However, scaling-up of 3D organoids for high-throughput screening is time-consuming and costly. Here, we successfully developed organoid-derived cell lines (2.5D) from 3D organoids; the cells were then expanded and recapitulated back into organoids known as cell line organoids (CLOs). The 2.5D lines were cultured long term into 2D established cell lines for downstream comparison analysis. Experimental characterisation of the models revealed that the proliferation of CLOs was slightly faster than that of parental organoids. The therapeutic response to chemotherapeutic agents in 3D CLOs and organoids showed a similar responsive profile. Compared to 3D CLOs and organoids, 2D cell lines tended to be less responsive to all the drugs tested. Stem cell marker expression was higher in either 3D CLOs or organoids compared to 2D cell lines. An in vivo tumorigenicity study found CLOs form tumours at a similar rate to organoids and retain enhanced CSC marker expression, indicating the plasticity of CSCs within the in vivo microenvironment. Full article
(This article belongs to the Topic Human Current and Future Model Systems)
Show Figures

Figure 1

19 pages, 4088 KiB  
Review
3D Tumor Spheroid and Organoid to Model Tumor Microenvironment for Cancer Immunotherapy
by Yichen Zhu, Elliot Kang, Matthew Wilson, Taylor Basso, Evelynn Chen, Yanqi Yu and Yan-Ruide Li
Organoids 2022, 1(2), 149-167; https://doi.org/10.3390/organoids1020012 - 05 Dec 2022
Cited by 12 | Viewed by 7357
Abstract
The intricate microenvironment in which malignant cells reside is essential for the progression of tumor growth. Both the physical and biochemical features of the tumor microenvironment (TME) play a critical role in promoting the differentiation, proliferation, invasion, and metastasis of cancer cells. It [...] Read more.
The intricate microenvironment in which malignant cells reside is essential for the progression of tumor growth. Both the physical and biochemical features of the tumor microenvironment (TME) play a critical role in promoting the differentiation, proliferation, invasion, and metastasis of cancer cells. It is therefore essential to understand how malignant cells interact and communicate with an assortment of supportive tumor-associated cells including macrophages, fibroblasts, endothelial cells, and other immune cells. To study the complex mechanisms behind cancer progression, 3D spheroid and organoid models are widely in favor because they replicate the stromal environment and multicellular structure present within an in vivo tumor. It provides more precise data about the cell–cell interactions, tumor characteristics, drug discovery, and metabolic profile of cancer cells compared to oversimplified 2D systems and unrepresentative animal models. This review provides a description of the key elements of the tumor microenvironment as well as early research using cell-line derived, 3D spheroid tumor models that paved the way for the adoption of patient-derived spheroid and organoid models. In particular, 3D spheroid and organoid models provide a method for drug screening with a particular emphasis on influence of the TME in cancer immunotherapy. Full article
(This article belongs to the Special Issue Feature Papers in Organoids)
Show Figures

Figure 1

14 pages, 5301 KiB  
Article
Articular Tissue-Mimicking Organoids Derived from Mesenchymal Stem Cells and Induced Pluripotent Stem Cells
by Zhong Alan Li, Jiangyinzi Shang, Shiqi Xiang, Eileen N. Li, Haruyo Yagi, Kanyakorn Riewruja, Hang Lin and Rocky S. Tuan
Organoids 2022, 1(2), 135-148; https://doi.org/10.3390/organoids1020011 - 14 Nov 2022
Cited by 6 | Viewed by 2611
Abstract
Organoids offer a promising strategy for articular tissue regeneration, joint disease modeling, and development of precision medicine. In this study, two types of human stem cells—primary mesenchymal stem cells (MSCs) and induced pluripotent stem cells (iPSCs)—were employed to engineer organoids that mimicked bone, [...] Read more.
Organoids offer a promising strategy for articular tissue regeneration, joint disease modeling, and development of precision medicine. In this study, two types of human stem cells—primary mesenchymal stem cells (MSCs) and induced pluripotent stem cells (iPSCs)—were employed to engineer organoids that mimicked bone, cartilage and adipose tissue, three key tissue components in articular joints. Prior to organoidogenesis, the iPSCs were first induced into mesenchymal progenitor cells (iMPCs). After characterizing the MSCs and iMPCs, they were used to generate cell-embedded extracellular matrix (ECM) constructs, which then underwent self-aggregation and lineage-specific differentiation in different induction media. Hydroxyapatite nanorods, an osteoinductive bioceramic, were leveraged to generate bone and osteochondral organoids, which effectively enhanced mineralization. The phenotypes of the generated organoids were confirmed on the basis of gene expression profiling and histology. Our findings demonstrate the feasibility and potential of generating articular tissue-recapitulating organoids from MSCs and iPSCs. Full article
(This article belongs to the Special Issue Organoids Mimicking Articular Tissue)
Show Figures

Figure 1

19 pages, 1597 KiB  
Review
Meniscus Repair: From In Vitro Research to Patients
by Hélène Vignes, Guillaume Conzatti, Guoqiang Hua and Nadia Benkirane-Jessel
Organoids 2022, 1(2), 116-134; https://doi.org/10.3390/organoids1020010 - 02 Nov 2022
Cited by 2 | Viewed by 4340
Abstract
Walking, running, jumping, or even just standing up are habits that we all have to perform in our everyday lives. However, defects in tissues composing the knee joint can drastically alter our ability to complete those simple actions. The knee joint is made [...] Read more.
Walking, running, jumping, or even just standing up are habits that we all have to perform in our everyday lives. However, defects in tissues composing the knee joint can drastically alter our ability to complete those simple actions. The knee joint is made up of the interaction between bones (femur, tibia, and patella), tendons, ligaments, and the two menisci (lateral and medial) in order to ensure smooth body movements. The meniscus corresponds to a crescent-shaped fibrocartilaginous tissue, which is found in the knee joint between the femoral condyles and the tibial plateau. It plays a key role in the stability of the knee joint. However, it is quite vulnerable and therefore tears can occur within this tissue and compromise the proper function of the knee. Recently, numerous efforts have been made in order to find solutions to repair and regenerate the meniscus, supported by both bioengineering researchers and orthopedic surgeons. However, due to its poor healing capacity and its complex structure, the reconstruction of the meniscus remains particularly challenging. In this review, the current treatment options will be explained and the possibility of using organoids as building blocks for implant formation or as an in vitro three-dimensional model will be highlighted. Full article
(This article belongs to the Special Issue Organoids Mimicking Articular Tissue)
Show Figures

Figure 1

10 pages, 1806 KiB  
Communication
Single-Cell Atlas of Patient-Derived Trophoblast Organoids in Ongoing Pregnancies
by Olivier J. M. Schäffers, Catherine Dupont, Eric M. Bindels, Diane Van Opstal, Dick H. W. Dekkers, Jeroen A. A. Demmers, Joost Gribnau and Bas B. van Rijn
Organoids 2022, 1(2), 106-115; https://doi.org/10.3390/organoids1020009 - 02 Oct 2022
Cited by 2 | Viewed by 2757
Abstract
Trophoblast organoids (TOs) hold great promise for elucidating human placental development and function. By deriving TOs in ongoing pregnancies using chorionic villus sampling (CVS), we established a platform to study trophoblast differentiation and function in early pregnancy, including pregnancies with different fetal genetic [...] Read more.
Trophoblast organoids (TOs) hold great promise for elucidating human placental development and function. By deriving TOs in ongoing pregnancies using chorionic villus sampling (CVS), we established a platform to study trophoblast differentiation and function in early pregnancy, including pregnancies with different fetal genetic abnormalities. We addressed cellular heterogeneity of CVS-derived TOs by providing a single-cell transcriptomic atlas and showed that CVS-TOs recapitulate key aspects of the human placenta, including syncytial fusion and hormone synthesis. This study demonstrates the utility of trophoblast organoids for investigating genetic defects in the placenta and describes an experimental platform for future personalized placental medicine approaches, including genotype–phenotype mapping. Full article
Show Figures

Graphical abstract

Previous Issue
Next Issue
Back to TopTop