3D Cell Culture in Disease Modeling and Tissue Regeneration

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

Deadline for manuscript submissions: closed (30 September 2023) | Viewed by 26535

Special Issue Editors


E-Mail Website
Guest Editor
Department of Medical and Surgical Sciences, “Alma Mater Studiorum” Università di Bologna, Bologna, Italy
Interests: stem cells; histological modeling of human tissues; three-dimensional (3D) cultures; perinatal stem cells; spheroids; organoids; antimicrobial factors; natural compounds; regenerative medicine; biomaterials
Special Issues, Collections and Topics in MDPI journals
1. Department of Surgery, School of Medicine, University of California, Davis, CA 95616, USA
2. Department of Biomedical Engineering, College of Engineering, University of California, Davis, CA 95616, USA
Interests: stem cell biology; molecular, cellular and tissue engineering; biomaterials; regenerative medicine; neovascularization congenital anomalies; developmental biology
Special Issues, Collections and Topics in MDPI journals

E-Mail Website
Guest Editor
Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong, China
Interests: dental stem cell
Special Issues, Collections and Topics in MDPI journals

Special Issue Information

Dear Colleagues,

The introduction of the third dimension in cell culture has completely changed the way in which cells and stem cells simulate diseases and facilitate in vitro fabrication to replace tissue blocks. The preservation of histological features, such as the presence of surrounding stromal niche, intercellular heterotypic connections, and mechanical stimulation, is essential for mimicking organ functionality. New techniques, biomaterials, and emerging platforms that improve 3D reconstruction of the in vivo environment represent a tremendous advantage for achieving further progress in biomedicine and the bioengineering future. The application of physiologically relevant culture models that have been invented and tested by researchers has potential in the development of more effective therapeutic treatments. These cell–cell or cell–biomaterial systems offer a cost-effective option for testing pharmaceutical and cosmetic products.
This Special Issue aims to collect articles detailing the contribution of 3D cell culture to uncovering novel insights into the molecular mechanisms regulating pathogenesis in the advancement of cell-based biofabrication and in enabling regenerative medicine strategies. Original research articles, review articles, and short communications are welcome.

Potential topics include but are not limited to the following:

  • Development or application of in vitro 3D disease models
  • Novel co-culture protocols and multicellular systems
  • Evaluation of tumor/cancer spheroids (tumoroids)
  • Three-dimensional culture of stem cells
  • Organoids and organotypic cultures
  • New tissue engineering strategies
  • Bioprinted cell-based models
  • Biomimetic scaffold development and biocompatibility
  • Novel biomaterials used in preclinical or clinical studies
  • Validation of drugs and compounds in 3D in vitro systems
  • Testing of natural compounds in 3D models
  • Organ-on-a-chip and cell-based sensors
  • Fluidic and label-free approaches for 3D modeling
  • Standardization of 3D models

Dr. Pasquale Marrazzo
Dr. Aijun Wang
Prof. Dr. Chengfei Zhang
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. 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

  • 3D cell culture
  • 3D cell model
  • tissue engineering
  • stem cells
  • biomaterials
  • disease model
  • spheroids
  • organoids
  • scaffold

Published Papers (13 papers)

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

Research

Jump to: Review, Other

17 pages, 5675 KiB  
Article
Extracellular Mechanical Stimuli Alters the Metastatic Progression of Prostate Cancer Cells within 3D Tissue Matrix
by Maggie Ditto, Diego Jacho, Kathryn M. Eisenmann and Eda Yildirim-Ayan
Bioengineering 2023, 10(11), 1271; https://doi.org/10.3390/bioengineering10111271 - 31 Oct 2023
Viewed by 1169
Abstract
This study aimed to understand extracellular mechanical stimuli’s effect on prostate cancer cells’ metastatic progression within a three-dimensional (3D) bone-like microenvironment. In this study, a mechanical loading platform, EQUicycler, has been employed to create physiologically relevant static and cyclic mechanical stimuli to a [...] Read more.
This study aimed to understand extracellular mechanical stimuli’s effect on prostate cancer cells’ metastatic progression within a three-dimensional (3D) bone-like microenvironment. In this study, a mechanical loading platform, EQUicycler, has been employed to create physiologically relevant static and cyclic mechanical stimuli to a prostate cancer cell (PC-3)-embedded 3D tissue matrix. Three mechanical stimuli conditions were applied: control (no loading), cyclic (1% strain at 1 Hz), and static mechanical stimuli (1% strain). The changes in prostate cancer cells’ cytoskeletal reorganization, polarity (elongation index), proliferation, expression level of N-Cadherin (metastasis-associated gene), and migratory potential within the 3D collagen structures were assessed upon mechanical stimuli. The results have shown that static mechanical stimuli increased the metastasis progression factors, including cell elongation (p < 0.001), cellular F-actin accumulation (p < 0.001), actin polymerization (p < 0.001), N-Cadherin gene expression, and invasion capacity of PC-3 cells within a bone-like microenvironment compared to its cyclic and control loading counterparts. This study established a novel system for studying metastatic cancer cells within bone and enables the creation of biomimetic in vitro models for cancer research and mechanobiology. Full article
(This article belongs to the Special Issue 3D Cell Culture in Disease Modeling and Tissue Regeneration)
Show Figures

Graphical abstract

23 pages, 4305 KiB  
Article
Human-Derived Cortical Neurospheroids Coupled to Passive, High-Density and 3D MEAs: A Valid Platform for Functional Tests
by Lorenzo Muzzi, Donatella Di Lisa, Matteo Falappa, Sara Pepe, Alessandro Maccione, Laura Pastorino, Sergio Martinoia and Monica Frega
Bioengineering 2023, 10(4), 449; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering10040449 - 06 Apr 2023
Viewed by 2524
Abstract
With the advent of human-induced pluripotent stem cells (hiPSCs) and differentiation protocols, methods to create in-vitro human-derived neuronal networks have been proposed. Although monolayer cultures represent a valid model, adding three-dimensionality (3D) would make them more representative of an in-vivo environment. Thus, human-derived [...] Read more.
With the advent of human-induced pluripotent stem cells (hiPSCs) and differentiation protocols, methods to create in-vitro human-derived neuronal networks have been proposed. Although monolayer cultures represent a valid model, adding three-dimensionality (3D) would make them more representative of an in-vivo environment. Thus, human-derived 3D structures are becoming increasingly used for in-vitro disease modeling. Achieving control over the final cell composition and investigating the exhibited electrophysiological activity is still a challenge. Thence, methodologies to create 3D structures with controlled cellular density and composition and platforms capable of measuring and characterizing the functional aspects of these samples are needed. Here, we propose a method to rapidly generate neurospheroids of human origin with control over cell composition that can be used for functional investigations. We show a characterization of the electrophysiological activity exhibited by the neurospheroids by using micro-electrode arrays (MEAs) with different types (i.e., passive, C-MOS, and 3D) and number of electrodes. Neurospheroids grown in free culture and transferred on MEAs exhibited functional activity that can be chemically and electrically modulated. Our results indicate that this model holds great potential for an in-depth study of signal transmission to drug screening and disease modeling and offers a platform for in-vitro functional testing. Full article
(This article belongs to the Special Issue 3D Cell Culture in Disease Modeling and Tissue Regeneration)
Show Figures

Figure 1

20 pages, 9084 KiB  
Article
Reduced Fibroblast Activation on Electrospun Polycaprolactone Scaffolds
by Joe P. Woodley, Daniel W. Lambert and Ilida Ortega Asencio
Bioengineering 2023, 10(3), 348; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering10030348 - 11 Mar 2023
Cited by 2 | Viewed by 1517
Abstract
In vivo, quiescent fibroblasts reside in three-dimensional connective tissues and are activated in response to tissue injury before proliferating rapidly and becoming migratory and contractile myofibroblasts. When deregulated, chronic activation drives fibrotic disease. Fibroblasts cultured on stiff 2D surfaces display a partially activated [...] Read more.
In vivo, quiescent fibroblasts reside in three-dimensional connective tissues and are activated in response to tissue injury before proliferating rapidly and becoming migratory and contractile myofibroblasts. When deregulated, chronic activation drives fibrotic disease. Fibroblasts cultured on stiff 2D surfaces display a partially activated phenotype, whilst many 3D environments limit fibroblast activation. Cell mechanotransduction, spreading, polarity, and integrin expression are controlled by material mechanical properties and micro-architecture. Between 3D culture systems, these features are highly variable, and the challenge of controlling individual properties without altering others has led to an inconsistent picture of fibroblast behaviour. Electrospinning offers greater control of mechanical properties and microarchitecture making it a valuable model to study fibroblast activation behaviour in vitro. Here, we present a comprehensive characterisation of the activation traits of human oral fibroblasts grown on a microfibrous scaffold composed of electrospun polycaprolactone. After over 7 days in the culture, we observed a reduction in proliferation rates compared to cells cultured in 2D, with low KI67 expression and no evidence of cellular senescence. A-SMA mRNA levels fell, and the expression of ECM protein-coding genes also decreased. Electrospun fibrous scaffolds, therefore, represent a tuneable platform to investigate the mechanisms of fibroblast activation and their roles in fibrotic disease. Full article
(This article belongs to the Special Issue 3D Cell Culture in Disease Modeling and Tissue Regeneration)
Show Figures

Figure 1

18 pages, 5985 KiB  
Article
Electrospun Poly(L-lactide-co-ε-caprolactone) Scaffold Potentiates C2C12 Myoblast Bioactivity and Acts as a Stimulus for Cell Commitment in Skeletal Muscle Myogenesis
by Serafina Pacilio, Roberta Costa, Valentina Papa, Maria Teresa Rodia, Carlo Gotti, Giorgia Pagnotta, Giovanna Cenacchi and Maria Letizia Focarete
Bioengineering 2023, 10(2), 239; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering10020239 - 11 Feb 2023
Cited by 2 | Viewed by 1772
Abstract
Tissue engineering combines a scaffold, cells and regulatory signals, reproducing a biomimetic extracellular matrix capable of supporting cell attachment and proliferation. We examined the role of an electrospun scaffold made of a biocompatible polymer during the myogenesis of skeletal muscle (SKM) as an [...] Read more.
Tissue engineering combines a scaffold, cells and regulatory signals, reproducing a biomimetic extracellular matrix capable of supporting cell attachment and proliferation. We examined the role of an electrospun scaffold made of a biocompatible polymer during the myogenesis of skeletal muscle (SKM) as an alternative approach to tissue regeneration. The engineered nanostructure was obtained by electrospinning poly(L-lactide-co-ε-caprolactone) (PLCL) in the form of a 3D porous nanofibrous scaffold further coated with collagen. C2C12 were cultured on the PLCL scaffold, and cell morphology and differentiation pathways were thoroughly investigated. The functionalized PLCL scaffold recreated the SKM nanostructure and performed its biological functions, guiding myoblast morphogenesis and promoting cell differentiation until tissue formation. The scaffold enabled cell–cell interactions through the development of cellular adhesions that were fundamental during myoblast fusion and myotube formation. Expression of myogenic regulatory markers and muscle-specific proteins at different stages of myogenesis suggested that the PLCL scaffold enhanced myoblast differentiation within a shorter time frame. The functionalized PLCL scaffold impacts myoblast bioactivity and acts as a stimulus for cell commitment, surpassing traditional 2D cell culture techniques. We developed a screening model for tissue development and a device for tissue restoration. Full article
(This article belongs to the Special Issue 3D Cell Culture in Disease Modeling and Tissue Regeneration)
Show Figures

Figure 1

11 pages, 2403 KiB  
Article
The Medium Obtained from the Culture of Hodgkin Lymphoma Cells Affects the Biophysical Characteristics of a Fibroblast Cell Model
by Maura Rossi, Francesco Alviano, Barie Myrtaj, Silvia Zia, Simona Righi, Valeria Pizzuti, Francesca Paris, Barbara Roda, Andrea Zattoni, Laura Bonsi, Elena Sabattini and Claudio Agostinelli
Bioengineering 2023, 10(2), 197; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering10020197 - 03 Feb 2023
Cited by 1 | Viewed by 1281
Abstract
The neoplastic Hodgkin-Reed-Sternberg (HRS) cells in Hodgkin lymphoma (HL) represent only 1–10% of cells and are surrounded by an inflammatory microenvironment. The HL cytokine network is a key point for the proliferation of HRS cells and for the maintenance of an advantageous microenvironment [...] Read more.
The neoplastic Hodgkin-Reed-Sternberg (HRS) cells in Hodgkin lymphoma (HL) represent only 1–10% of cells and are surrounded by an inflammatory microenvironment. The HL cytokine network is a key point for the proliferation of HRS cells and for the maintenance of an advantageous microenvironment for HRS survival. In the tumor microenvironment (TME), the fibroblasts are involved in crosstalk with HRS cells. The aim of this work was to study the effect of lymphoma cell conditioned medium on a fibroblast cell population and evaluate modifications of cell morphology and proliferation. Hodgkin lymphoma-derived medium was used to obtain a population of “conditioned” fibroblasts (WS-1 COND). Differences in biophysical parameters were detected by the innovative device Celector®. Fibroblast-HL cells interactions were reproduced in 3D co-culture spheroids. WS-1 COND showed a different cellular morphology with an enlarged cytoplasm and enhanced metabolism. Area and diameter cell values obtained by Celector® measurement were increased. Co-culture spheroids created with WS-1 COND showed a tighter aggregation than those with non-conditioned WS-1. The presence of soluble factors derived from HRS cells in the conditioned medium was adequate for the proliferation of fibroblasts and conditioned fibroblasts in a 3D HL model allowed to develop a representative model of the in vivo TME. Full article
(This article belongs to the Special Issue 3D Cell Culture in Disease Modeling and Tissue Regeneration)
Show Figures

Figure 1

18 pages, 10407 KiB  
Article
Characterization of Perinatal Stem Cell Spheroids for the Development of Cell Therapy Strategy
by Francesca Paris, Pasquale Marrazzo, Valeria Pizzuti, Cosetta Marchionni, Maura Rossi, Martina Michelotti, Biljana Petrovic, Elisabetta Ciani, Giuliana Simonazzi, Andrea Pession, Laura Bonsi and Francesco Alviano
Bioengineering 2023, 10(2), 189; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering10020189 - 02 Feb 2023
Cited by 4 | Viewed by 2643
Abstract
Type 1 diabetes mellitus (T1DM) is a complex metabolic disease characterized by a massive loss of insulin-producing cells due to an autoimmune reaction. Currently, daily subcutaneous administration of exogenous insulin is the only effective treatment. Therefore, in recent years considerable interest has been [...] Read more.
Type 1 diabetes mellitus (T1DM) is a complex metabolic disease characterized by a massive loss of insulin-producing cells due to an autoimmune reaction. Currently, daily subcutaneous administration of exogenous insulin is the only effective treatment. Therefore, in recent years considerable interest has been given to stem cell therapy and in particular to the use of three-dimensional (3D) cell cultures to better reproduce in vivo conditions. The goal of this study is to provide a reliable cellular model that could be investigated for regenerative medicine applications for the replacement of insulin-producing cells in T1DM. To pursue this aim we create a co-culture spheroid of amniotic epithelial cells (AECs) and Wharton’s jelly mesenchymal stromal cells (WJ-MSCs) in a one-to-one ratio. The resulting co-culture spheroids were analyzed for viability, extracellular matrix production, and hypoxic state in both early- and long-term cultures. Our results suggest that co-culture spheroids are stable in long-term culture and are still viable with a consistent extracellular matrix production evaluated with immunofluorescence staining. These findings suggest that this co-culture may potentially be differentiated into endo-pancreatic cells for regenerative medicine applications in T1DM. Full article
(This article belongs to the Special Issue 3D Cell Culture in Disease Modeling and Tissue Regeneration)
Show Figures

Graphical abstract

19 pages, 3902 KiB  
Article
TARTESSUS: A Customized Electrospun Drug Delivery System Loaded with Irinotecan for Local and Sustained Chemotherapy Release in Pancreatic Cancer
by Carmen Cepeda-Franco, Oihane Mitxelena-Iribarren, Francisco José Calero-Castro, Malen Astigarraga, Juan M. Castillo-Tuñon, Iman Laga, Sheila Pereira, Sergio Arana, Maite Mujika and Javier Padillo-Ruiz
Bioengineering 2023, 10(2), 183; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering10020183 - 01 Feb 2023
Viewed by 1689
Abstract
Post-surgical chemotherapy in pancreatic cancer has notorious side effects due to the high dose required. Multiple devices have been designed to tackle this aspect and achieve a delayed drug release. This study aimed to explore the controlled and sustained local delivery of a [...] Read more.
Post-surgical chemotherapy in pancreatic cancer has notorious side effects due to the high dose required. Multiple devices have been designed to tackle this aspect and achieve a delayed drug release. This study aimed to explore the controlled and sustained local delivery of a reduced drug dose from an irinotecan-loaded electrospun nanofiber membrane (named TARTESSUS) that can be placed on the patients’ tissue after tumor resection surgery. The drug delivery system formulation was made of polycaprolactone (PCL). The mechanical properties and the release kinetics of the drug were adjusted by the electrospinning parameters and by the polymer ratio between 10 w.t.% and 14 w.t.% of PCL in formic acid:acetic acid:chloroform (47.5:47.5:5). The irinotecan release analysis was performed and three different release periods were obtained, depending on the concentration of the polymer in the dissolution. The TARTESSUS device was tested in 2D and 3D cell cultures and it demonstrated a decrease in cell viability in 2D culture between 72 h and day 7 from the start of treatment. In 3D culture, a decrease in viability was seen between 72 h, day 7 (p < 0.001), day 10 (p < 0.001), 14 (p < 0.001), and day 17 (p = 0.003) as well as a decrease in proliferation between 72 h and day 10 (p = 0.030) and a reduction in spheroid size during days 10 (p = 0.001), 14 (p < 0.001), and 17 (p < 0.001). In conclusion, TARTESSUS showed a successful encapsulation of a chemotherapeutic drug and a sustained and delayed release with an adjustable releasing period to optimize the therapeutic effect in pancreatic cancer treatment. Full article
(This article belongs to the Special Issue 3D Cell Culture in Disease Modeling and Tissue Regeneration)
Show Figures

Figure 1

10 pages, 1602 KiB  
Article
Bioengineering of Hair Follicle-like Structure for Validation of Hair Growth Promoting Compounds
by Hyun Woo Joo, Min Kyu Kim, Soon Sun Bak and Young Kwan Sung
Bioengineering 2022, 9(11), 645; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering9110645 - 03 Nov 2022
Cited by 1 | Viewed by 2374
Abstract
We aimed to establish screening and efficacy test techniques for use in the development of hair-promoting agents. To this end, we used the dermal papilla cell (DPc)-derived immortalized cell line (SV40T-hTERT DPc) and neonatal foreskin-derived keratinocyte cell line (Ker-CT) to form an immortalized [...] Read more.
We aimed to establish screening and efficacy test techniques for use in the development of hair-promoting agents. To this end, we used the dermal papilla cell (DPc)-derived immortalized cell line (SV40T-hTERT DPc) and neonatal foreskin-derived keratinocyte cell line (Ker-CT) to form an immortalized cell-based hair follicle-like structure. The SV40T-hTERT DPc spheroids exhibited a higher cell ratio in the spheroids than primary DPc spheroids, and SV40T-hTERT DPc aggregated with spheroids larger in diameter than primary DPc when the same cell number was seeded into the low-adhesion plate. Microscopic imaging and fluorescence staining results indicated that both primary and immortalized cell combinations form a hair follicle-like structure with a long-stretched keratinocyte layer under the condition that the spheroids have the same diameter as that of in vivo dermal papillary tissue in the hair follicle. The hair follicle-like structure elongation was increased upon treatment with three known hair follicle growth-promoting compounds (minoxidil, tofacitinib, and ascorbic acid) compared with that in the control group. Therefore, using immortalized cells to generate a coherent follicle-like structure, we have developed models for screening and evaluating hair-care materials commonly used in the industry. Full article
(This article belongs to the Special Issue 3D Cell Culture in Disease Modeling and Tissue Regeneration)
Show Figures

Graphical abstract

16 pages, 4275 KiB  
Article
Formation of Three-Dimensional Spheres Enhances the Neurogenic Potential of Stem Cells from Apical Papilla
by Mohammed S. Basabrain, Jialin Zhong, Haiyun Luo, Junqing Liu, Baicheng Yi, Ahmed Zaeneldin, Junhao Koh, Ting Zou and Chengfei Zhang
Bioengineering 2022, 9(11), 604; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering9110604 - 22 Oct 2022
Cited by 3 | Viewed by 1468
Abstract
Cell-based neural regeneration is challenging due to the difficulty in obtaining sufficient neural stem cells with clinical applicability. Stem cells from apical papilla (SCAPs) originating from embryonic neural crests with high neurogenic potential could be a promising cell source for neural regeneration. This [...] Read more.
Cell-based neural regeneration is challenging due to the difficulty in obtaining sufficient neural stem cells with clinical applicability. Stem cells from apical papilla (SCAPs) originating from embryonic neural crests with high neurogenic potential could be a promising cell source for neural regeneration. This study aimed to investigate whether the formation of 3D spheres can promote SCAPs’ neurogenic potential. Material and methods: Three-dimensional SCAP spheres were first generated in a 256-well agarose microtissue mold. The spheres and single cells were individually cultured on collagen I-coated μ-slides. Cell morphological changes, neural marker expression, and neurite outgrowth were evaluated by confocal microscope, ELISA, and RT-qPCR. Results: Pronounced morphological changes were noticed in a time-dependent manner. The migrating cells’ morphology changed from fibroblast-like cells to neuron-like cells. Compared to the 2D culture, neurite length, number, and the expression of multiple progenitors, immature and mature neural markers were significantly higher in the 3D spheres. BDNF and NGF-β may play a significant role in the neural differentiation of SCAP spheres. Conclusion: The formation of 3D spheres enhanced the neurogenic potential of SCAPs, suggesting the advantage of using the 3D spheres of SCAPs for treating neural diseases. Full article
(This article belongs to the Special Issue 3D Cell Culture in Disease Modeling and Tissue Regeneration)
Show Figures

Graphical abstract

16 pages, 5629 KiB  
Article
Therapeutic Effect of Biomimetic Scaffold Loaded with Human Amniotic Epithelial Cell-Derived Neural-like Cells for Spinal Cord Injury
by Chen Qiu, Yuan Sun, Jinying Li, Yuchen Xu, Jiayi Zhou, Cong Qiu, Shaomin Zhang, Yong He and Luyang Yu
Bioengineering 2022, 9(10), 535; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering9100535 - 09 Oct 2022
Cited by 5 | Viewed by 1670
Abstract
Spinal cord injury (SCI) results in devastating consequences for the motor and sensory function of patients due to neuronal loss and disrupted neural circuits, confronting poor prognosis and lack of effective therapies. A new therapeutic strategy is urgently required. Here, human amniotic epithelial [...] Read more.
Spinal cord injury (SCI) results in devastating consequences for the motor and sensory function of patients due to neuronal loss and disrupted neural circuits, confronting poor prognosis and lack of effective therapies. A new therapeutic strategy is urgently required. Here, human amniotic epithelial cells (hAEC), featured with immunocompatibility, non-tumorgenicity and no ethical issues, were induced into neural-like cells by a compound cocktail, as evidenced with morphological change and the expression of neural cell markers. Interestingly, the hAEC-neural-like cells maintain the characteristic of low immunogenicity as hAEC. Aiming at SCI treatment in vivo, we constructed a 3D-printed GelMA hydrogel biomimetic spinal cord scaffold with micro-channels, in which hAEC-neural-like cells were well-induced and grown. In a rat full transection SCI model, hAEC-neural-like cell scaffolds that were implanted in the lesion demonstrated significant therapeutic effects; the neural circuit and hindlimb locomotion were partly recovered compared to little affection in the SCI rats receiving an empty scaffold or a sham implantation operation. Thus, the establishment of hAEC-neural-like cell biomimetic scaffolds may provide a safe and effective treatment strategy for SCI. Full article
(This article belongs to the Special Issue 3D Cell Culture in Disease Modeling and Tissue Regeneration)
Show Figures

Graphical abstract

13 pages, 1854 KiB  
Article
An Optical and Chemiluminescence Assay for Assessing the Cytotoxicity of Balamuthia mandrillaris against Human Neurospheroids
by Worakamol Pengsart and Kasem Kulkeaw
Bioengineering 2022, 9(7), 330; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering9070330 - 20 Jul 2022
Cited by 5 | Viewed by 1866
Abstract
A spheroid is a cell aggregate in a three-dimensional context; thereby, it recapitulates the cellular architecture in human tissue. However, the utility of spheroids as an assay for host–parasite interactions remains unexplored. This study demonstrates the potential use of neurospheroids for assessing the [...] Read more.
A spheroid is a cell aggregate in a three-dimensional context; thereby, it recapitulates the cellular architecture in human tissue. However, the utility of spheroids as an assay for host–parasite interactions remains unexplored. This study demonstrates the potential use of neurospheroids for assessing the cytotoxicity of the life-threatening pathogenic amoeba Balamuthia mandrillaris. The neuroblastoma SH-SY5Y cells formed a spheroid in a hanging drop of culture medium. Cellular damage caused by B. mandrillaris trophozoites on human neuronal spheroids was observed using microscopic imaging and ATP detection. B. mandrillaris trophozoites rapidly caused a decrease in ATP production in the spheroid, leading to loss of neurospheroid integrity. Moreover, 3D confocal microscopy imaging revealed interactions between the trophozoites and SH-SY5Y neuronal cells in the outer layer of the neurospheroid. In conclusion, the neurospheroid allows the assessment of host cell damage in a simple and quantitative manner. Full article
(This article belongs to the Special Issue 3D Cell Culture in Disease Modeling and Tissue Regeneration)
Show Figures

Graphical abstract

Review

Jump to: Research, Other

26 pages, 2792 KiB  
Review
Vascularized Tissue Organoids
by Hannah A. Strobel, Sarah M. Moss and James B. Hoying
Bioengineering 2023, 10(2), 124; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering10020124 - 17 Jan 2023
Cited by 9 | Viewed by 3326
Abstract
Tissue organoids hold enormous potential as tools for a variety of applications, including disease modeling and drug screening. To effectively mimic the native tissue environment, it is critical to integrate a microvasculature with the parenchyma and stroma. In addition to providing a means [...] Read more.
Tissue organoids hold enormous potential as tools for a variety of applications, including disease modeling and drug screening. To effectively mimic the native tissue environment, it is critical to integrate a microvasculature with the parenchyma and stroma. In addition to providing a means to physiologically perfuse the organoids, the microvasculature also contributes to the cellular dynamics of the tissue model via the cells of the perivascular niche, thereby further modulating tissue function. In this review, we discuss current and developing strategies for vascularizing organoids, consider tissue-specific vascularization approaches, discuss the importance of perfusion, and provide perspectives on the state of the field. Full article
(This article belongs to the Special Issue 3D Cell Culture in Disease Modeling and Tissue Regeneration)
Show Figures

Figure 1

Other

Jump to: Research, Review

10 pages, 2816 KiB  
Brief Report
Generation of an Adequate Perfusion Network within Dense Collagen Hydrogels Using Thermoplastic Polymers as Sacrificial Matrix to Promote Cell Viability
by Marie Camman, Pierre Marquaille, Pierre Joanne, Onnik Agbulut and Christophe Hélary
Bioengineering 2022, 9(7), 313; https://0-doi-org.brum.beds.ac.uk/10.3390/bioengineering9070313 - 14 Jul 2022
Cited by 3 | Viewed by 1465
Abstract
Dense collagen hydrogels are promising biomaterials for several tissue-engineering applications. They exhibit high mechanical properties, similar to physiological extracellular matrices, and do not shrink under cellular activity. However, they suffer from several drawbacks, such as weak nutrient and O2 diffusion, impacting cell [...] Read more.
Dense collagen hydrogels are promising biomaterials for several tissue-engineering applications. They exhibit high mechanical properties, similar to physiological extracellular matrices, and do not shrink under cellular activity. However, they suffer from several drawbacks, such as weak nutrient and O2 diffusion, impacting cell survival. Here, we report a novel strategy to create a perfusion system within dense and thick collagen hydrogels to promote cell viability. The 3D printing of a thermoplastic filament (high-impact polystyrene, HIPS) with a three-wave shape is used to produce an appropriate sacrificial matrix. The HIPS thermoplastic polymer allows for good shape fidelity of the filament and does not collapse under the mechanical load of the collagen solution. After the collagen gels around the filament and dissolves, a channel is generated, allowing for adequate and rapid hydrogel perfusion. The dissolution process does not alter the collagen hydrogel’s physical or chemical properties, and the perfusion is associated with an increased fibroblast survival. Here, we report the novel utilization of thermoplastics to generate a perfusion network within biomimetic collagen hydrogels. Full article
(This article belongs to the Special Issue 3D Cell Culture in Disease Modeling and Tissue Regeneration)
Show Figures

Figure 1

Back to TopTop