Material and Engineering-Based Approaches for Organoids

A special issue of Bioengineering (ISSN 2306-5354). This special issue belongs to the section "Regenerative Engineering".

Deadline for manuscript submissions: closed (31 August 2023) | Viewed by 36871

Special Issue Editor


E-Mail Website
Guest Editor
Polymeric Biomaterials Laboratory, University of Pennsylvania, Philadelphia, PA 19104, USA
Interests: biomaterials; hyaluronic acid; tissue engineering; regenerative medicine

Special Issue Information

Dear Colleagues, 

Ex vivo multicellular tissue and organoid models have informed our understanding of tissue morphogenesis and organogenesis; in addition, they have created new avenues for reparative and regenerative therapies. Engineering technologies and biomaterial-based concepts have been shown to direct and guide cellular self-assembly and maturation of ex vivo organoids, regaining certain aspects of tissue structure and functionality. This Special Issue focuses on strategies that maximize the potential of biomaterials and technology in modelling tissue formation and organ development, and applications in drug/small molecule testing, tissue replacement and cancer research. Topics of interest for this Special Issue include, but are not limited to, the following: 

  • Development of material-based multicellular structures such as spheroids, organoids and/or gastruloids;
  • Design of organoid platforms for therapeutic and regenerative applications, drug screening or personalized medicine;
  • Technologies that enhance integration of organoids into biological and clinical workflows, such as biofabrication/additive manufacturing, computational and genome-editing tools;
  • Technologies that enhance long-term growth of complex systems such as bioreactors, co-culture systems and engineered blood vessel or neural networks.

Dr. Claudia Loebel
Guest Editor

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. Bioengineering is an international peer-reviewed open access monthly journal published by MDPI.

Please visit the Instructions for Authors page before submitting a manuscript. The Article Processing Charge (APC) for publication in this open access journal is 2700 CHF (Swiss Francs). 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

  • Biomaterials
  • Hydrogels
  • Organoids
  • 3D tissue models
  • Personalized medicine
  • Tissue engineering
  • Disease modelling
  • Biofabrication

Published Papers (8 papers)

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

Research

Jump to: Review

17 pages, 3661 KiB  
Article
In Vivo Assessment of Laboratory-Grown Kidney Tissue Grafts
by Tinghsien Chuang, Justin Bejar, Zhiwei Yue, Mary Slavinsky, Denise Marciano, Iain Drummond and Leif Oxburgh
Bioengineering 2023, 10(11), 1261; https://doi.org/10.3390/bioengineering10111261 - 29 Oct 2023
Viewed by 1248
Abstract
Directed differentiation of stem cells is an attractive approach to generate kidney tissue for regenerative therapies. Currently, the most informative platform to test the regenerative potential of this tissue is engraftment into kidneys of immunocompromised rodents. Stem cell-derived kidney tissue is vascularized following [...] Read more.
Directed differentiation of stem cells is an attractive approach to generate kidney tissue for regenerative therapies. Currently, the most informative platform to test the regenerative potential of this tissue is engraftment into kidneys of immunocompromised rodents. Stem cell-derived kidney tissue is vascularized following engraftment, but the connection between epithelial tubules that is critical for urine to pass from the graft to the host collecting system has not yet been demonstrated. We show that one significant obstacle to tubule fusion is the accumulation of fibrillar collagens at the interface between the graft and the host. As a screening strategy to identify factors that can prevent this collagen accumulation, we propose encapsulating laboratory-grown kidney tissue in fibrin hydrogels supplemented with candidate compounds such as recombinant proteins, small molecules, feeder cells, and gene therapy vectors to condition the local graft environment. We demonstrate that the AAV-DJ serotype is an efficient gene therapy vector for the subcapsular region and that it is specific for interstitial cells in this compartment. In addition to the histological evaluation of epithelial tubule fusion, we demonstrate the specificity of two urine biomarker assays that can be used to detect human-specific markers of the proximal nephron (CD59) and the distal nephron (uromodulin), and we demonstrate the deposition of human graft-derived urine into the mouse collecting system. Using the testing platform described in this report, it will be possible to systematically screen factors for their potential to promote epithelial fusion of graft and host tissue with a functional intravital read-out. Full article
(This article belongs to the Special Issue Material and Engineering-Based Approaches for Organoids)
Show Figures

Graphical abstract

15 pages, 4310 KiB  
Article
Alginate Core–Shell Capsules for 3D Cultivation of Adipose-Derived Mesenchymal Stem Cells
by Sabrina Nebel, Manuel Lux, Sonja Kuth, Faina Bider, Wolf Dietrich, Dominik Egger, Aldo R. Boccaccini and Cornelia Kasper
Bioengineering 2022, 9(2), 66; https://doi.org/10.3390/bioengineering9020066 - 06 Feb 2022
Cited by 9 | Viewed by 9722
Abstract
Mesenchymal stem cells (MSCs) are primary candidates in tissue engineering and stem cell therapies due to their intriguing regenerative and immunomodulatory potential. Their ability to self-assemble into three-dimensional (3D) aggregates further improves some of their therapeutic properties, e.g., differentiation potential, secretion of cytokines, [...] Read more.
Mesenchymal stem cells (MSCs) are primary candidates in tissue engineering and stem cell therapies due to their intriguing regenerative and immunomodulatory potential. Their ability to self-assemble into three-dimensional (3D) aggregates further improves some of their therapeutic properties, e.g., differentiation potential, secretion of cytokines, and homing capacity after administration. However, high hydrodynamic shear forces and the resulting mechanical stresses within commercially available dynamic cultivation systems can decrease their regenerative properties. Cells embedded within a polymer matrix, however, lack cell-to-cell interactions found in their physiological environment. Here, we present a “semi scaffold-free” approach to protect the cells from high shear forces by a physical barrier, but still allow formation of a 3D structure with in vivo-like cell-to-cell contacts. We highlight a relatively simple method to create core–shell capsules by inverse gelation. The capsules consist of an outer barrier made from sodium alginate, which allows for nutrient and waste diffusion and an inner compartment for direct cell-cell interactions. Next to capsule characterization, a harvesting procedure was established and viability and proliferation of human adipose-derived MSCs were investigated. In the future, this encapsulation and cultivation technique might be used for MSC-expansion in scalable dynamic bioreactor systems, facilitating downstream procedures, such as cell harvest and differentiation into mature tissue grafts. Full article
(This article belongs to the Special Issue Material and Engineering-Based Approaches for Organoids)
Show Figures

Graphical abstract

21 pages, 4607 KiB  
Article
Alginate Hydrogel Microtubes for Salivary Gland Cell Organization and Cavitation
by Matthew Jorgensen, Pujhitha Ramesh, Miriam Toro, Emily Evans, Nicholas Moskwa, Xulang Zhang, Susan T. Sharfstein, Melinda Larsen and Yubing Xie
Bioengineering 2022, 9(1), 38; https://doi.org/10.3390/bioengineering9010038 - 15 Jan 2022
Cited by 10 | Viewed by 3370
Abstract
Understanding the different regulatory functions of epithelial and mesenchymal cell types in salivary gland development and cellular organization is essential for proper organoid formation and salivary gland tissue regeneration. Here, we demonstrate a biocompatible platform using pre-formed alginate hydrogel microtubes to facilitate direct [...] Read more.
Understanding the different regulatory functions of epithelial and mesenchymal cell types in salivary gland development and cellular organization is essential for proper organoid formation and salivary gland tissue regeneration. Here, we demonstrate a biocompatible platform using pre-formed alginate hydrogel microtubes to facilitate direct epithelial–mesenchymal cell interaction for 3D salivary gland cell organization, which allows for monitoring cellular organization while providing a protective barrier from cell-cluster loss during medium changes. Using mouse salivary gland ductal epithelial SIMS cells as the epithelial model cell type and NIH 3T3 fibroblasts or primary E16 salivary mesenchyme cells as the stromal model cell types, self-organization from epithelial–mesenchymal interaction was examined. We observed that epithelial and mesenchymal cells undergo aggregation on day 1, cavitation by day 4, and generation of an EpCAM-expressing epithelial cell layer as early as day 7 of the co-culture in hydrogel microtubes, demonstrating the utility of hydrogel microtubes to facilitate heterotypic cell–cell interactions to form cavitated organoids. Thus, pre-formed alginate microtubes are a promising co-culture method for further understanding epithelial and mesenchymal interaction during tissue morphogenesis and for future practical applications in regenerative medicine. Full article
(This article belongs to the Special Issue Material and Engineering-Based Approaches for Organoids)
Show Figures

Graphical abstract

10 pages, 2638 KiB  
Article
Human Peripheral Nerve-Derived Pluripotent Cells Can Be Stimulated by In Vitro Bone Morphogenetic Protein-2
by Renyi Sun, Tanghong Jia, Bradley Dart, Sunaina Shrestha, Morgan Bretches, Michael H. Heggeness and Shang-You Yang
Bioengineering 2021, 8(10), 132; https://doi.org/10.3390/bioengineering8100132 - 26 Sep 2021
Cited by 2 | Viewed by 2833
Abstract
We have recently identified a population of cells within the peripheral nerves of adult rodent animals (mice and rats) that can respond to Bone Morphogenetic Protein-2 (BMP-2) exposure or physical injury to rapidly proliferate. More importantly, these cells exhibited embryonic differentiation potentials that [...] Read more.
We have recently identified a population of cells within the peripheral nerves of adult rodent animals (mice and rats) that can respond to Bone Morphogenetic Protein-2 (BMP-2) exposure or physical injury to rapidly proliferate. More importantly, these cells exhibited embryonic differentiation potentials that could be induced into osteoblastic and endothelial cells in vitro. The current study examined human nerve specimens to compare and characterize the cells after BMP-2 stimulation. Fresh pieces of human nerve tissue were minced and treated with either BMP-2 (750 ng/mL) or a PBS vehicle for 12 h at 37 °C, before being digested in 0.2% collagenase and 0.05% trypsin-EDTA. Isolated cells were cultured in a restrictive stem cell medium. Significantly more cells were obtained from the nerve pieces with the BMP-2 treatment in comparison with the PBS vehicle controls. Cell colonies started to form at Day 3. Expressions of the four transcription factors, namely, Klf4, c-Myc, Sox2, and Oct4, were confirmed at both the transcriptional and translational levels. The cells can be maintained in the stem cell culture medium for at least 6 weeks without changing their morphology. When the cells were transferred to a fibroblast growth medium, dispersed spindle-shaped motile cells were noted and became fibroblast activated protein-α (FAP) positive with immunocytochemistry staining. The data suggest that human peripheral nerve tissue also contains a population of cells that can respond to BMP-2 and express Klf4, Sox2, cMyc, and Oct4—the four transcription factors driving cell pluripotency. These cells are able to differentiate into FAP-positive fibroblasts. In summary, in human peripheral nerves also reside a population of quiescent cells with pluripotency potential that may be the same cells as rodent nerve-derived adult stem (NEDAPS) cells. It is proposed that these cells are possibly at the core of a previously unknown natural mechanism for healing an injury. Full article
(This article belongs to the Special Issue Material and Engineering-Based Approaches for Organoids)
Show Figures

Figure 1

Review

Jump to: Research

13 pages, 2076 KiB  
Review
Organoid Cultures In Silico: Tools or Toys?
by Torsten Thalheim, Gabriela Aust and Joerg Galle
Bioengineering 2023, 10(1), 50; https://doi.org/10.3390/bioengineering10010050 - 30 Dec 2022
Cited by 2 | Viewed by 2017
Abstract
The implementation of stem-cell-based organoid culture more than ten years ago started a development that created new avenues for diagnostic analyses and regenerative medicine. In parallel, computational modelling groups realized the potential of this culture system to support their theoretical approaches to study [...] Read more.
The implementation of stem-cell-based organoid culture more than ten years ago started a development that created new avenues for diagnostic analyses and regenerative medicine. In parallel, computational modelling groups realized the potential of this culture system to support their theoretical approaches to study tissues in silico. These groups developed computational organoid models (COMs) that enabled testing consistency between cell biological data and developing theories of tissue self-organization. The models supported a mechanistic understanding of organoid growth and maturation and helped linking cell mechanics and tissue shape in general. What comes next? Can we use COMs as tools to complement the equipment of our biological and medical research? While these models already support experimental design, can they also quantitatively predict tissue behavior? Here, we review the current state of the art of COMs and discuss perspectives for their application. Full article
(This article belongs to the Special Issue Material and Engineering-Based Approaches for Organoids)
Show Figures

Figure 1

15 pages, 2150 KiB  
Review
Emerging Paradigms in Bioengineering the Lungs
by Raxshanaa Mohgan, Mayuren Candasamy, Jayashree Mayuren, Sachin Kumar Singh, Gaurav Gupta, Kamal Dua and Dinesh Kumar Chellappan
Bioengineering 2022, 9(5), 195; https://doi.org/10.3390/bioengineering9050195 - 01 May 2022
Cited by 3 | Viewed by 3467
Abstract
In end-stage lung diseases, the shortage of donor lungs for transplantation and long waiting lists are the main culprits in the significantly increasing number of patient deaths. New strategies to curb this issue are being developed with the help of recent advancements in [...] Read more.
In end-stage lung diseases, the shortage of donor lungs for transplantation and long waiting lists are the main culprits in the significantly increasing number of patient deaths. New strategies to curb this issue are being developed with the help of recent advancements in bioengineering technology, with the generation of lung scaffolds as a steppingstone. There are various types of lung scaffolds, namely, acellular scaffolds that are developed via decellularization and recellularization techniques, artificial scaffolds that are synthesized using synthetic, biodegradable, and low immunogenic materials, and hybrid scaffolds which combine the advantageous properties of materials in the development of a desirable lung scaffold. There have also been advances in the design of bioreactors in terms of providing an optimal regenerative environment for the maturation of functional lung tissue over time. In this review, the emerging paradigms in the field of lung tissue bioengineering will be discussed. Full article
(This article belongs to the Special Issue Material and Engineering-Based Approaches for Organoids)
Show Figures

Figure 1

23 pages, 3054 KiB  
Review
Alzheimer’s Disease: Current Perspectives and Advances in Physiological Modeling
by E. Josephine Boder and Ipsita A. Banerjee
Bioengineering 2021, 8(12), 211; https://doi.org/10.3390/bioengineering8120211 - 12 Dec 2021
Cited by 7 | Viewed by 4076
Abstract
Though Alzheimer’s disease (AD) is the most common cause of dementia, complete disease-modifying treatments are yet to be fully attained. Until recently, transgenic mice constituted most in vitro model systems of AD used for preclinical drug screening; however, these models have so far [...] Read more.
Though Alzheimer’s disease (AD) is the most common cause of dementia, complete disease-modifying treatments are yet to be fully attained. Until recently, transgenic mice constituted most in vitro model systems of AD used for preclinical drug screening; however, these models have so far failed to adequately replicate the disease’s pathophysiology. However, the generation of humanized APOE4 mouse models has led to key discoveries. Recent advances in stem cell differentiation techniques and the development of induced pluripotent stem cells (iPSCs) have facilitated the development of novel in vitro devices. These “microphysiological” systems—in vitro human cell culture systems designed to replicate in vivo physiology—employ varying levels of biomimicry and engineering control. Spheroid-based organoids, 3D cell culture systems, and microfluidic devices or a combination of these have the potential to replicate AD pathophysiology and pathogenesis in vitro and thus serve as both tools for testing therapeutics and models for experimental manipulation. Full article
(This article belongs to the Special Issue Material and Engineering-Based Approaches for Organoids)
Show Figures

Figure 1

16 pages, 1152 KiB  
Review
3D Hepatic Organoid-Based Advancements in LIVER Tissue Engineering
by Amit Panwar, Prativa Das and Lay Poh Tan
Bioengineering 2021, 8(11), 185; https://doi.org/10.3390/bioengineering8110185 - 14 Nov 2021
Cited by 13 | Viewed by 8638
Abstract
Liver-associated diseases and tissue engineering approaches based on in vitro culture of functional Primary human hepatocytes (PHH) had been restricted by the rapid de-differentiation in 2D culture conditions which restricted their usability. It was proven that cells growing in 3D format can better [...] Read more.
Liver-associated diseases and tissue engineering approaches based on in vitro culture of functional Primary human hepatocytes (PHH) had been restricted by the rapid de-differentiation in 2D culture conditions which restricted their usability. It was proven that cells growing in 3D format can better mimic the in vivo microenvironment, and thus help in maintaining metabolic activity, phenotypic properties, and longevity of the in vitro cultures. Again, the culture method and type of cell population are also recognized as important parameters for functional maintenance of primary hepatocytes. Hepatic organoids formed by self-assembly of hepatic cells are microtissues, and were able to show long-term in vitro maintenance of hepato-specific characteristics. Thus, hepatic organoids were recognized as an effective tool for screening potential cures and modeling liver diseases effectively. The current review summarizes the importance of 3D hepatic organoid culture over other conventional 2D and 3D culture models and its applicability in Liver tissue engineering. Full article
(This article belongs to the Special Issue Material and Engineering-Based Approaches for Organoids)
Show Figures

Figure 1

Back to TopTop