1
|
De Simone U, Caloni F, Pignatti P, Gaetano C, Locatelli CA, Coccini T. Human stromal cell-based protocol to generate astrocytes: a straightforward in vitro predictive strategy in neurotoxicology. Toxicol Mech Methods 2025; 35:340-355. [PMID: 39626968 DOI: 10.1080/15376516.2024.2435351] [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: 09/09/2024] [Revised: 11/22/2024] [Accepted: 11/23/2024] [Indexed: 05/04/2025]
Abstract
The inherent adaptability of human mesenchymal stromal cells (hMSCs) to differentiate into neural lineages provides a valuable resource for investigating potential neurotoxicity in humans. By harnessing the ability of hMSCs to transform into astrocytes, we can evaluate the effects of various agents on these vital cells. Our protocol employs hMSCs sourced from umbilical cord tissue, ensuring a readily available supply of high-quality cells. The hMSC-to-neural workflow encompasses six essential steps: hMSC culture, followed by the generation of embryoid bodies (EBs) from these cells on specialized surfaces. Next, EBs and cells are expanded in a growth-promoting medium, directing them toward neural lineages. Subsequent differentiation into immature astrocytes is achieved through the use of specific factors. The process continues with the maturation of EBs/cells into astrocyte-like cells (hALCs) under optimized conditions, culminating in the final development of hALCs in a specialized medium. This methodology yields cells that display astrocyte morphology and express characteristic markers such as GFAP and S100β. The protocol is efficient, requiring roughly 6 weeks to generate hALCs from primary hMSCs without genetic manipulation. The application of hMSCs in evaluating cell damage triggered by neurotoxicants like MeHg and MGO underscores their potential as a valuable component within a more extensive battery of neurotoxicity tests.
Collapse
Affiliation(s)
- Uliana De Simone
- Laboratory of Clinical and Experimental Toxicology, and Pavia Poison Center-National Toxicology Information Center, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Francesca Caloni
- Dipartimento di Scienze e Politiche Ambientali (ESP), Università degli Studi di Milano, Milan, Italy
| | - Patrizia Pignatti
- Allergy and Immunology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Carlo Gaetano
- Laboratory of Epigenetics, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Carlo Alessandro Locatelli
- Laboratory of Clinical and Experimental Toxicology, and Pavia Poison Center-National Toxicology Information Center, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| | - Teresa Coccini
- Laboratory of Clinical and Experimental Toxicology, and Pavia Poison Center-National Toxicology Information Center, Toxicology Unit, Istituti Clinici Scientifici Maugeri IRCCS, Pavia, Italy
| |
Collapse
|
2
|
Smadja DM, Berkane Y, Bentounes NK, Rancic J, Cras A, Pinault C, Ouarne M, Paucod E, Rachidi W, Lellouch AG, Jeljeli M. Immune-privileged cord blood-derived endothelial colony-forming cells: advancing immunomodulation and vascular regeneration. Angiogenesis 2025; 28:19. [PMID: 40047974 PMCID: PMC11885380 DOI: 10.1007/s10456-025-09973-9] [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: 12/11/2024] [Accepted: 02/25/2025] [Indexed: 03/09/2025]
Abstract
Cord blood-derived endothelial colony-forming cells (CB-ECFCs) hold significant promise for regenerative medicine due to their unique vasculogenic and immunomodulatory properties. These cells exhibit a superior proliferative capacity, robust ability to form vascular networks, and lower immunogenicity compared to adult and embryonic stem cell-derived counterparts. The immune-privileged characteristics of CB-ECFCs, including reduced expression of pro-inflammatory mediators and tolerance-inducing molecules such as HLA-G, further enhance their therapeutic potential. Their low immunogenicity minimizes the risk of immune rejection, making them suitable for allogenic cell therapies. Their application extends to complex tissue engineering and organ revascularization, where their ability to integrate into three-dimensional scaffolds and support vascular tree formation represents a significant advancement. Moreover, CB-ECFCs' capability to adapt to inflammatory stimuli and retain immunological memory highlights their functional versatility in dynamic microenvironments. This review highlights the remarkable ontogeny of ECFCs while unveiling the unparalleled potential of CB-ECFCs in revolutionizing regenerative medicine. From pre-vascularizing engineered tissues and organoids to pioneering cell-based therapies for cardiovascular, dermatological, and degenerative diseases, CB-ECFCs stand at the forefront of cutting-edge biomedical advancements, offering unprecedented opportunities for therapeutic innovation. By leveraging their vasculogenic, immune-regulatory, and regenerative capacities, CB-ECFCs offer a robust alternative for addressing the challenges of vascular repair and organ engineering. Future research should focus on unraveling their transcriptomic and functional profiles to optimize clinical applications and advance the field of regenerative medicine.
Collapse
Affiliation(s)
- David M Smadja
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France.
- Hematology Department, AP-HP, Georges Pompidou European Hospital, Paris, F-75015, France.
| | - Yanis Berkane
- Department of Plastic, Reconstructive and Aesthetic Surgery, Hôpital Sud, CHU Rennes, University of Rennes, Rennes, France
- SITI Laboratory, UMR INSERM 1236, Rennes University Hospital, Rennes, France
- Center for Engineering in Medicine and Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Nun K Bentounes
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
- Hematology Department, AP-HP, Georges Pompidou European Hospital, Paris, F-75015, France
| | - Jeanne Rancic
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
- Hematology Department, AP-HP, Georges Pompidou European Hospital, Paris, F-75015, France
| | - Audrey Cras
- Cell Therapy Department, AP-HP, Saint-Louis Hospital, Paris, F-75010, France
| | - Cécile Pinault
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
- Hematology Department, AP-HP, Georges Pompidou European Hospital, Paris, F-75015, France
| | - Marie Ouarne
- Univ. Grenoble Alpes, CEA, INSERM, IRIG-BGE UA13, Grenoble, 38000, France
| | - Elise Paucod
- Univ. Grenoble Alpes, CEA, INSERM, IRIG-BGE UA13, Grenoble, 38000, France
| | - Walid Rachidi
- Univ. Grenoble Alpes, CEA, INSERM, IRIG-BGE UA13, Grenoble, 38000, France
| | - Alexandre G Lellouch
- Université Paris Cité, INSERM U970, Paris Cardiovascular Research Center, Paris, France
- Hematology Department, AP-HP, Georges Pompidou European Hospital, Paris, F-75015, France
- Department of Plastic, Reconstructive and Aesthetic Surgery, Cedars Sinai Hospital, Los Angeles, USA
| | - Maxime Jeljeli
- Department of Plastic, Reconstructive and Aesthetic Surgery, Cedars Sinai Hospital, Los Angeles, USA
| |
Collapse
|
3
|
Lafci Büyükkahraman M, Chen H, Chen-Charpentier BM, Liao J, Kojouharov HV. A Mathematical Exploration of the Effects of Ischemia-Reperfusion Injury After a Myocardial Infarction. Bioengineering (Basel) 2025; 12:177. [PMID: 40001696 PMCID: PMC11851514 DOI: 10.3390/bioengineering12020177] [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: 11/22/2024] [Revised: 01/20/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
INTRODUCTION After myocardial infarction (MI), the heart undergoes necrosis, inflammation, scar formation, and remodeling. While restoring blood flow is crucial, it can cause ischemia-reperfusion (IR) injury, driven by reactive oxygen species (ROSs), which exacerbate cell death and tissue damage. This study introduces a mathematical model capturing key post-MI dynamics, including inflammatory responses, IR injury, cardiac remodeling, and stem cell therapy. The model uses nonlinear ordinary differential equations to simulate these processes under varying conditions, offering a predictive tool to understand MI pathophysiology better and optimize treatments. METHODS After myocardial infarction (MI), left ventricular remodeling progresses through three distinct yet interconnected phases. The first phase captures the immediate dynamics following MI, prior to any medical intervention. This stage is mathematically modeled using the system of ordinary differential equations: The second and third stages of the remodeling process account for the system dynamics of medical treatments, including oxygen restoration and subsequent stem cell injection at the injury site. RESULTS We simulate heart tissue and immune cell dynamics over 30 days for mild and severe MI using the novel mathematical model under medical treatment. The treatment involves no intervention until 2 h post-MI, followed by oxygen restoration and stem cell injection at day 7, which is shown experimentallyand numerically to be optimal. The simulation incorporates a baseline ROS threshold (Rc) where subcritical ROS levels do not cause cell damage. CONCLUSION This study presents a novel mathematical model that extends a previously published framework by incorporating three clinically relevant parameters: oxygen restoration rate (ω), patient risk factors (γ), and neutrophil recruitment profile (δ). The model accounts for post-MI inflammatory dynamics, ROS-mediated ischemia-reperfusion (IR) injury, cardiac remodeling, and stem cell therapy. The model's sensitivity highlights critical clinical insights: while oxygen restoration is vital, excessive rates may exacerbate ROS-driven IR injury. Additionally, heightened patient risk factors (e.g., smoking, obesity) and immunodeficiency significantly impact tissue damage and recovery. This predictive tool offers valuable insights into MI pathology and aids in optimizing treatment strategies to mitigate IR injury and improve post-MI outcomes.
Collapse
Affiliation(s)
| | - Houjia Chen
- Department of Bioengineering, The University of Texas at Arlington, Arlington, TX 76010-0138, USA; (H.C.); (J.L.)
| | - Benito M. Chen-Charpentier
- Department of Mathematics, The University of Texas at Arlington, Arlington, TX 76019-0408, USA; (B.M.C.-C.); (H.V.K.)
| | - Jun Liao
- Department of Bioengineering, The University of Texas at Arlington, Arlington, TX 76010-0138, USA; (H.C.); (J.L.)
| | - Hristo V. Kojouharov
- Department of Mathematics, The University of Texas at Arlington, Arlington, TX 76019-0408, USA; (B.M.C.-C.); (H.V.K.)
| |
Collapse
|
4
|
Arcuri S, Pennarossa G, Prasadani M, Gandolfi F, Brevini TAL. Use of Decellularized Bio-Scaffolds for the Generation of a Porcine Artificial Intestine. Methods Protoc 2024; 7:76. [PMID: 39452790 PMCID: PMC11510128 DOI: 10.3390/mps7050076] [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: 07/29/2024] [Revised: 09/25/2024] [Accepted: 09/26/2024] [Indexed: 10/26/2024] Open
Abstract
In recent years, great interest has been focused on the development of highly reproducible 3D in vitro models that are able to mimic the physiological architecture and functionality of native tissues. To date, a wide range of techniques have been proposed to recreate an intestinal barrier in vitro, including synthetic scaffolds and hydrogels, as well as complex on-a-chip systems and organoids. Here, we describe a novel protocol for the generation of an artificial intestine based on the creation of decellularized bio-scaffolds and their repopulation with intestinal stromal and epithelial cells. Organs collected at the local slaughterhouse are subjected to a decellularization protocol that includes a freezing/thawing step, followed by sequential incubation in 1% SDS for 12 h, 1% Triton X-100 for 12 h, and 2% deoxycholate for 12 h. At the end of the procedure, the generated bio-scaffolds are repopulated with intestinal fibroblasts and then with epithelial cells. The protocol described here represents a promising and novel strategy to generate an in vitro bioengineered intestine platform able to mimic some of the complex functions of the intestinal barrier, thus constituting a promising 3D strategy for nutritional, pharmaceutical, and toxicological studies.
Collapse
Affiliation(s)
- Sharon Arcuri
- Department of Veterinary Medicine, Università degli Studi di Sassari, Via Vienna, 07100 Sassari, Italy;
- Laboratory of Biomedical Embryology and Tissue Engineering, Department of Veterinary Medicine and Animal Sciences, Università degli Studi di Milano, Via dell’Università 6, 26900 Lodi, Italy;
| | - Georgia Pennarossa
- Laboratory of Biomedical Embryology and Tissue Engineering, Department of Veterinary Medicine and Animal Sciences, Università degli Studi di Milano, Via dell’Università 6, 26900 Lodi, Italy;
| | - Madhusha Prasadani
- Institute of Veterinary Medicine and Animal Sciences, Estonian University of Life Sciences, 50411 Tartu, Estonia;
| | - Fulvio Gandolfi
- Department of Agricultural and Environmental Sciences—Production, Landscape, Agroenergy, Università degli Studi di Milano, Via Celoria 2, 20133 Milan, Italy;
| | - Tiziana A. L. Brevini
- Laboratory of Biomedical Embryology and Tissue Engineering, Department of Veterinary Medicine and Animal Sciences, Università degli Studi di Milano, Via dell’Università 6, 26900 Lodi, Italy;
| |
Collapse
|
5
|
Zhou Z, Liu J, Xiong T, Liu Y, Tuan RS, Li ZA. Engineering Innervated Musculoskeletal Tissues for Regenerative Orthopedics and Disease Modeling. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2310614. [PMID: 38200684 DOI: 10.1002/smll.202310614] [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: 11/18/2023] [Revised: 12/28/2023] [Indexed: 01/12/2024]
Abstract
Musculoskeletal (MSK) disorders significantly burden patients and society, resulting in high healthcare costs and productivity loss. These disorders are the leading cause of physical disability, and their prevalence is expected to increase as sedentary lifestyles become common and the global population of the elderly increases. Proper innervation is critical to maintaining MSK function, and nerve damage or dysfunction underlies various MSK disorders, underscoring the potential of restoring nerve function in MSK disorder treatment. However, most MSK tissue engineering strategies have overlooked the significance of innervation. This review first expounds upon innervation in the MSK system and its importance in maintaining MSK homeostasis and functions. This will be followed by strategies for engineering MSK tissues that induce post-implantation in situ innervation or are pre-innervated. Subsequently, research progress in modeling MSK disorders using innervated MSK organoids and organs-on-chips (OoCs) is analyzed. Finally, the future development of engineering innervated MSK tissues to treat MSK disorders and recapitulate disease mechanisms is discussed. This review provides valuable insights into the underlying principles, engineering methods, and applications of innervated MSK tissues, paving the way for the development of targeted, efficacious therapies for various MSK conditions.
Collapse
Affiliation(s)
- Zhilong Zhou
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
| | - Jun Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
| | - Tiandi Xiong
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
| | - Yuwei Liu
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, Guangdong, 518000, P. R. China
| | - Rocky S Tuan
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
| | - Zhong Alan Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Center for Neuromusculoskeletal Restorative Medicine, Hong Kong Science Park, Shatin, NT, Hong Kong SAR, P. R. China
- School of Biomedical Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Key Laboratory of Regenerative Medicine, Ministry of Education, School of Biomedical Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Shatin, NT, Hong Kong SAR, P. R. China
- Shenzhen Research Institute, The Chinese University of Hong Kong, Shenzhen, Guangdong, 518057, P. R. China
| |
Collapse
|
6
|
Liu S, Chen H, Xie H, Liu X, Zhang M. Substrate Stiffness Modulates Stemness and Differentiation of Rabbit Corneal Endothelium Through the Paxillin-YAP Pathway. Invest Ophthalmol Vis Sci 2024; 65:15. [PMID: 38466286 DOI: 10.1167/iovs.65.3.15] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/12/2024] Open
Abstract
Purpose To explore the role of substrate stiffness and the mechanism beneath corneal endothelial cells' (CECs') stemness maintenance and differentiation. Methods CECs were divided into central zone (8 mm trephined boundary) and peripheral zone (8 mm trephined edge with attached limbal). Two zones were analyzed by hematoxylin-eosin staining and scanning electron microscopy for anatomic structure. The elastic modulus of Descemet's membrane (DM) was analyzed by atomic force microscopy. Compressed type I collagen gels with different stiffness were constructed as an in vitro model system to test the role of stiffness on phenotype using cultured rabbit CECs. Cell morphology, expression and intracellular distribution of Yes-associated protein (YAP), differentiation (ZO-1, Na+/K+-ATPase), stemness (FOXD3, CD34, Sox2, Oct3/4), and endothelial-mesenchymal transition (EnMT) markers were analyzed by immunofluorescence, quantitative RT-PCR, and Western blot. Results The results showed that the peripheral area of rabbit and human DM is softer than the central area ex vivo. Using the biomimetic extracellular matrix collagen gels in vitro model, we then demonstrated that soft substrate weakens the differentiation and EnMT in the culture of CECs. It was further proved by the inhibitor experiment that soft substrate enhances stemness maintenance via inhibition of paxillin-YAP signaling, which was activated on a stiff substrate. Conclusions Our findings confirm that substrate stiffness modulates the stemness maintenance and differentiation of CECs and suggest a potential strategy for CEC-based corneal tissue engineering.
Collapse
Affiliation(s)
- Shuting Liu
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Hua Chen
- Senior Department of Ophthalmology, The Third Medical Center of PLA General Hospital, Beijing, China
| | - Huatao Xie
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xin Liu
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Mingchang Zhang
- Department of Ophthalmology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
7
|
Hoveidaei AH, Sadat-Shojai M, Mosalamiaghili S, Salarikia SR, Roghani-Shahraki H, Ghaderpanah R, Ersi MH, Conway JD. Nano-hydroxyapatite structures for bone regenerative medicine: Cell-material interaction. Bone 2024; 179:116956. [PMID: 37951520 DOI: 10.1016/j.bone.2023.116956] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/04/2023] [Revised: 11/04/2023] [Accepted: 11/05/2023] [Indexed: 11/14/2023]
Abstract
Bone tissue engineering holds great promise for the regeneration of damaged or severe bone defects. However, several challenges hinder its translation into clinical practice. To address these challenges, interdisciplinary efforts and advances in biomaterials, cell biology, and bioengineering are required. In recent years, nano-hydroxyapatite (nHA)-based scaffolds have emerged as a promising approach for the development of bone regenerative agents. The unique similarity of nHA with minerals found in natural bones promotes remineralization and stimulates bone growth, which are crucial factors for efficient bone regeneration. Moreover, nHA exhibits desirable properties, such as strong chemical interactions with bone and facilitation of tissue growth, without inducing inflammation or toxicity. It also promotes osteoblast survival, adhesion, and proliferation, as well as increasing alkaline phosphatase activity, osteogenic differentiation, and bone-specific gene expression. However, it is important to note that the effect of nHA on osteoblast behavior is dose-dependent, with cytotoxic effects observed at higher doses. Additionally, the particle size of nHA plays a crucial role, with smaller particles having a more significant impact. Therefore, in this review, we highlighted the potential of nHA for improving bone regeneration processes and summarized the available data on bone cell response to nHA-based scaffolds. In addition, an attempt is made to portray the current status of bone tissue engineering using nHA/polymer hybrids and some recent scientific research in the field.
Collapse
Affiliation(s)
- Amir Human Hoveidaei
- International Center for Limb Lengthening, Rubin Institute for Advanced Orthopedics, Sinai Hospital of Baltimore, Baltimore, MD, USA
| | - Mehdi Sadat-Shojai
- Department of Chemistry, College of Sciences, Shiraz University, Shiraz, Iran
| | - Seyedarad Mosalamiaghili
- Burn and Wound Healing Research Center, Shiraz University of Medical Sciences, Shiraz, Iran; Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | | | | | - Rezvan Ghaderpanah
- Student Research Committee, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mohammad Hamed Ersi
- Evidence Based Medicine Center, Hormozgan University of Medical Sciences, Bandar Abbas, Iran; Clinical Research Development Center of Shahid Mohammadi Hospital, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Janet D Conway
- International Center for Limb Lengthening, Rubin Institute for Advanced Orthopedics, Sinai Hospital of Baltimore, Baltimore, MD, USA.
| |
Collapse
|
8
|
Lu J, Wang M, Wang X, Meng Y, Chen F, Zhuang J, Han Y, Wang H, Liu W. A basement membrane extract-based three-dimensional culture system promotes the neuronal differentiation of cochlear Sox10-positive glial cells in vitro. Mater Today Bio 2024; 24:100937. [PMID: 38269057 PMCID: PMC10805941 DOI: 10.1016/j.mtbio.2023.100937] [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: 10/11/2023] [Revised: 12/14/2023] [Accepted: 12/27/2023] [Indexed: 01/26/2024] Open
Abstract
Spiral ganglion neurons (SGNs) in the mammalian cochleae are essential for the delivery of acoustic information, and damage to SGNs can lead to permanent sensorineural hearing loss as SGNs are not capable of regeneration. Cochlear glial cells (GCs) might be a potential source for SGN regeneration, but the neuronal differentiation ability of GCs is limited and its properties are not clear yet. Here, we characterized the cochlear Sox10-positive (Sox10+) GCs as a neural progenitor population and developed a basement membrane extract-based three-dimensional (BME-3D) culture system to promote its neuronal generation capacity in vitro. Firstly, the purified Sox10+ GCs, isolated from Sox10-creER/tdTomato mice via flow cytometry, were able to form neurospheres after being cultured in the traditional suspension culture system, while significantly more neurospheres were found and the expression of stem cell-related genes was upregulated in the BME-3D culture group. Next, the BME-3D culture system promoted the neuronal differentiation ability of Sox10+ GCs, as evidenced by the increased number, neurite outgrowth, area of growth cones, and synapse density as well as the promoted excitability of newly induced neurons. Notably, the BME-3D culture system also intensified the reinnervation of newly generated neurons with HCs and protected the neurospheres and derived-neurons against cisplatin-induced damage. Finally, transcriptome sequencing analysis was performed to identify the characteristics of the differentiated neurons. These findings suggest that the BME-3D culture system considerably promotes the proliferation capacity and neuronal differentiation efficiency of Sox10+ GCs in vitro, thus providing a possible strategy for the SGN regeneration study.
Collapse
Affiliation(s)
- Junze Lu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China
- Shandong Institute of Otorhinolaryngology, Jinan, 250022, China
| | - Man Wang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China
- Shandong Institute of Otorhinolaryngology, Jinan, 250022, China
| | - Xue Wang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China
- Shandong Institute of Otorhinolaryngology, Jinan, 250022, China
| | - Yu Meng
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China
- Shandong Institute of Otorhinolaryngology, Jinan, 250022, China
| | - Fang Chen
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China
- Shandong Institute of Otorhinolaryngology, Jinan, 250022, China
| | - Jinzhu Zhuang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China
- Shandong Institute of Otorhinolaryngology, Jinan, 250022, China
| | - Yuechen Han
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China
- Shandong Institute of Otorhinolaryngology, Jinan, 250022, China
| | - Haibo Wang
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China
- Shandong Institute of Otorhinolaryngology, Jinan, 250022, China
| | - Wenwen Liu
- Department of Otolaryngology-Head and Neck Surgery, Shandong Provincial ENT Hospital, Shandong University, Jinan, 250022, China
- Shandong Institute of Otorhinolaryngology, Jinan, 250022, China
| |
Collapse
|
9
|
Abuwatfa WH, Pitt WG, Husseini GA. Scaffold-based 3D cell culture models in cancer research. J Biomed Sci 2024; 31:7. [PMID: 38221607 PMCID: PMC10789053 DOI: 10.1186/s12929-024-00994-y] [Citation(s) in RCA: 14] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2023] [Accepted: 01/04/2024] [Indexed: 01/16/2024] Open
Abstract
Three-dimensional (3D) cell cultures have emerged as valuable tools in cancer research, offering significant advantages over traditional two-dimensional (2D) cell culture systems. In 3D cell cultures, cancer cells are grown in an environment that more closely mimics the 3D architecture and complexity of in vivo tumors. This approach has revolutionized cancer research by providing a more accurate representation of the tumor microenvironment (TME) and enabling the study of tumor behavior and response to therapies in a more physiologically relevant context. One of the key benefits of 3D cell culture in cancer research is the ability to recapitulate the complex interactions between cancer cells and their surrounding stroma. Tumors consist not only of cancer cells but also various other cell types, including stromal cells, immune cells, and blood vessels. These models bridge traditional 2D cell cultures and animal models, offering a cost-effective, scalable, and ethical alternative for preclinical research. As the field advances, 3D cell cultures are poised to play a pivotal role in understanding cancer biology and accelerating the development of effective anticancer therapies. This review article highlights the key advantages of 3D cell cultures, progress in the most common scaffold-based culturing techniques, pertinent literature on their applications in cancer research, and the ongoing challenges.
Collapse
Affiliation(s)
- Waad H Abuwatfa
- Materials Science and Engineering Ph.D. Program, College of Arts and Sciences, American University of Sharjah, P.O. Box. 26666, Sharjah, United Arab Emirates
- Department of Chemical and Biological Engineering, College of Engineering, American University of Sharjah, P.O. Box 26666, Sharjah, United Arab Emirates
| | - William G Pitt
- Department of Chemical Engineering, Brigham Young University, Provo, UT, 84602, USA
| | - Ghaleb A Husseini
- Materials Science and Engineering Ph.D. Program, College of Arts and Sciences, American University of Sharjah, P.O. Box. 26666, Sharjah, United Arab Emirates.
- Department of Chemical and Biological Engineering, College of Engineering, American University of Sharjah, P.O. Box 26666, Sharjah, United Arab Emirates.
| |
Collapse
|
10
|
Zhang J, Liu C, Li X, Liu Z, Zhang Z. Application of photo-crosslinkable gelatin methacryloyl in wound healing. Front Bioeng Biotechnol 2023; 11:1303709. [PMID: 38076425 PMCID: PMC10702353 DOI: 10.3389/fbioe.2023.1303709] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2023] [Accepted: 11/10/2023] [Indexed: 02/13/2025] Open
Abstract
Wound healing is a complex and coordinated biological process easily influenced by various internal and external factors. Hydrogels have immense practical importance in wound nursing because of their environmental moisturising, pain-relieving, and cooling effects. As photo-crosslinkable biomaterials, gelatine methacryloyl (GelMA) hydrogels exhibit substantial potential for tissue repair and reconstruction because of their tunable and beneficial properties. GelMA hydrogels have been extensively investigated as scaffolds for cell growth and drug release in various biomedical applications. They also hold great significance in wound healing because of their similarity to the components of the extracellular matrix of the skin and their favourable physicochemical properties. These hydrogels can promote wound healing and tissue remodelling by reducing inflammation, facilitating vascularisation, and supporting cell growth. In this study, we reviewed the applications of GelMA hydrogels in wound healing, including skin tissue engineering, wound dressing, and transdermal drug delivery. We aim to inspire further exploration of their potential for wound healing.
Collapse
Affiliation(s)
- Jinli Zhang
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital (Guangzhou Red Cross Hospital of Jinan University), Guangzhou, China
| | - Changling Liu
- Department of Burns and Plastic Surgery, Guangzhou Red Cross Hospital (Guangzhou Red Cross Hospital of Jinan University), Guangzhou, China
| | - Xiaojian Li
- Department of Burns and Plastic Surgery, Guangzhou Red Cross Hospital (Guangzhou Red Cross Hospital of Jinan University), Guangzhou, China
| | - Zhihe Liu
- Guangzhou Institute of Traumatic Surgery, Guangzhou Red Cross Hospital (Guangzhou Red Cross Hospital of Jinan University), Guangzhou, China
| | - Zhi Zhang
- Department of Burns and Plastic Surgery, Guangzhou Red Cross Hospital (Guangzhou Red Cross Hospital of Jinan University), Guangzhou, China
| |
Collapse
|
11
|
Dimmock RL, Rotherham M, El Haj AJ, Yang Y. Fabrication and Characterisation of Hydrogels with Reversible Wrinkled Surfaces for Limbal Study and Reconstruction. Gels 2023; 9:915. [PMID: 37999005 PMCID: PMC10671082 DOI: 10.3390/gels9110915] [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: 10/21/2023] [Revised: 11/14/2023] [Accepted: 11/15/2023] [Indexed: 11/25/2023] Open
Abstract
In the biomedical field, there is a demand for the development of novel approaches for the investigation of optical epithelial anatomical features with biomimetic materials. These materials are not only required to replicate structures but also enable dynamic modelling for disease states such as limbal stem cell deficiency and ageing. In the present study, the effective generation of reversible wrinkled polydimethylsiloxane (PDMS) substrates was undertaken to mimic the undulating anatomy of the limbal epithelial stem cell niche. This undulating surface pattern was formed through a dual treatment with acid oxidation and plasma using an innovatively designed stretching frame. This system enabled the PDMS substrate to undergo deformation and relaxation, creating a reversible and tuneable wrinkle pattern with cell culture applications. The crypt-like pattern exhibited a width of 70-130 µm and a depth of 17-40 µm, resembling the topography of a limbal epithelial stem cell niche, which is characterised by an undulating anatomy. The cytocompatibility of the patterned substrate was markedly improved using a gelatin methacrylate polymer (GelMa) coating. It was also observed that these wrinkled PDMS surfaces were able to dictate cell growth patterns, showing alignment in motile cells and colony segregation in colony-forming cells when using human and porcine limbal cells, respectively.
Collapse
Affiliation(s)
- Ryan L. Dimmock
- School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent ST4 7QB, UK
| | - Michael Rotherham
- Healthcare Technologies Institute, Institute of Translational Medicine, School of Chemical Engineering, University of Birmingham, Birmingham B15 2TH, UK
| | - Alicia J. El Haj
- Healthcare Technologies Institute, Institute of Translational Medicine, School of Chemical Engineering, University of Birmingham, Birmingham B15 2TH, UK
| | - Ying Yang
- School of Pharmacy and Bioengineering, Keele University, Stoke-on-Trent ST4 7QB, UK
| |
Collapse
|
12
|
Hao S, Wang M, Yin Z, Jing Y, Bai L, Su J. Microenvironment-targeted strategy steers advanced bone regeneration. Mater Today Bio 2023; 22:100741. [PMID: 37576867 PMCID: PMC10413201 DOI: 10.1016/j.mtbio.2023.100741] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 06/26/2023] [Accepted: 07/19/2023] [Indexed: 08/15/2023] Open
Abstract
Treatment of large bone defects represents a great challenge in orthopedic and craniomaxillofacial surgery. Traditional strategies in bone tissue engineering have focused primarily on mimicking the extracellular matrix (ECM) of bone in terms of structure and composition. However, the synergistic effects of other cues from the microenvironment during bone regeneration are often neglected. The bone microenvironment is a sophisticated system that includes physiological (e.g., neighboring cells such as macrophages), chemical (e.g., oxygen, pH), and physical factors (e.g., mechanics, acoustics) that dynamically interact with each other. Microenvironment-targeted strategies are increasingly recognized as crucial for successful bone regeneration and offer promising solutions for advancing bone tissue engineering. This review provides a comprehensive overview of current microenvironment-targeted strategies and challenges for bone regeneration and further outlines prospective directions of the approaches in construction of bone organoids.
Collapse
Affiliation(s)
- Shuyue Hao
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Mingkai Wang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Zhifeng Yin
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai, 201941, China
| | - Yingying Jing
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Long Bai
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
- Department of Orthopedic Surgery, Xinhua Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200444, China
| |
Collapse
|
13
|
Goldshmid R, Simaan-Yameen H, Ifergan L, Loebel C, Burdick JA, Seliktar D. Modulus-dependent effects on neurogenic, myogenic, and chondrogenic differentiation of human mesenchymal stem cells in three-dimensional hydrogel cultures. J Biomed Mater Res A 2023; 111:1441-1458. [PMID: 37066837 DOI: 10.1002/jbm.a.37545] [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: 09/21/2022] [Revised: 03/23/2023] [Accepted: 03/25/2023] [Indexed: 04/18/2023]
Abstract
Human mesenchymal stromal cells (hMSCs) are of significant interest as a renewable source of therapeutically useful cells. In tissue engineering, hMSCs are implanted within a scaffold to provide enhanced capacity for tissue repair. The present study evaluates how mechanical properties of that scaffold can alter the phenotype and genotype of the cells, with the aim of augmenting hMSC differentiation along the myogenic, neurogenic or chondrogenic linages. The hMSCs were grown three-dimensionally (3D) in a hydrogel comprised of poly(ethylene glycol) (PEG)-conjugated to fibrinogen. The hydrogel's shear storage modulus (G'), which was controlled by increasing the amount of PEG-diacrylate cross-linker in the matrix, was varied in the range of 100-2000 Pascal (Pa). The differentiation into each lineage was initiated by a defined culture medium, and the hMSCs grown in the different modulus hydrogels were characterized using gene and protein expression. Materials having lower storage moduli (G' = 100 Pa) exhibited more hMSCs differentiating to neurogenic lineages. Myogenesis was favored in materials having intermediate modulus values (G' = 500 Pa), whereas chondrogenesis was favored in materials with a higher modulus (G' = 1000 Pa). Enhancing the differentiation pathway of hMSCs in 3D hydrogel scaffolds using simple modifications to mechanical properties represents an important achievement toward the effective application of these cells in tissue engineering.
Collapse
Affiliation(s)
- Revital Goldshmid
- The Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
- The Interdisciplinary Program for Biotechnology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Haneen Simaan-Yameen
- The Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
- The Interdisciplinary Program for Biotechnology, Technion-Israel Institute of Technology, Haifa, Israel
| | - Liaura Ifergan
- The Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| | - Claudia Loebel
- Materials Science & Engineering Department, University of Michigan, Ann Arbor, Michigan, USA
| | - Jason A Burdick
- BioFrontiers Institute and Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado, USA
| | - Dror Seliktar
- The Faculty of Biomedical Engineering, Technion-Israel Institute of Technology, Haifa, Israel
| |
Collapse
|
14
|
Tenreiro MF, Branco MA, Cotovio JP, Cabral JMS, Fernandes TG, Diogo MM. Advancing organoid design through co-emergence, assembly, and bioengineering. Trends Biotechnol 2023; 41:923-938. [PMID: 36653200 DOI: 10.1016/j.tibtech.2022.12.021] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 12/19/2022] [Accepted: 12/30/2022] [Indexed: 01/18/2023]
Abstract
Human adult stem cells and patient-derived induced pluripotent stem cells represent promising tools to understand human biology, development, and disease. Under a permissive environment, stem cell derivatives can self-organize and reconstruct their native milieu, resulting in the creation of organ-like entities known as organoids. Although organoids represent a breakthrough in the stem cell field, there are still considerable shortcomings preventing their widespread use, namely their variability, limited function, and reductionist size. In the past few years, sophisticated methodologies have been proposed to allow the design of organoids with improved biological fidelity and physiological relevance. Here, we summarize these emerging technologies and provide insights into how they can be utilized to fulfill the potential of stem cells.
Collapse
Affiliation(s)
- Miguel F Tenreiro
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Mariana A Branco
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - João P Cotovio
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Joaquim M S Cabral
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Tiago G Fernandes
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal
| | - Maria Margarida Diogo
- Department of Bioengineering and Institute for Bioengineering and Biosciences, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal; Associate Laboratory i4HB-Institute for Health and Bioeconomy, Instituto Superior Técnico, Universidade de Lisboa, Av. Rovisco Pais, 1049-001 Lisbon, Portugal.
| |
Collapse
|
15
|
Ahmed T. Functional biomaterials for biomimetic 3D in vitro tumor microenvironment modeling. IN VITRO MODELS 2023; 2:1-23. [PMID: 39872875 PMCID: PMC11756483 DOI: 10.1007/s44164-023-00043-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 01/11/2023] [Accepted: 01/16/2023] [Indexed: 01/30/2025]
Abstract
The translational potential of promising anticancer medications and treatments may be enhanced by the creation of 3D in vitro models that can accurately reproduce native tumor microenvironments. Tumor microenvironments for cancer treatment and research can be built in vitro using biomaterials. Three-dimensional in vitro cancer models have provided new insights into the biology of cancer. Cancer researchers are creating artificial three-dimensional tumor models based on functional biomaterials that mimic the microenvironment of the real tumor. Our understanding of tumor stroma activity over the course of cancer has improved because of the use of scaffold and matrix-based three-dimensional systems intended for regenerative medicine. Scientists have created synthetic tumor models thanks to recent developments in materials engineering. These models enable researchers to investigate the biology of cancer and assess the therapeutic effectiveness of available medications. The emergence of biomaterial engineering technologies with the potential to hasten treatment outcomes is highlighted in this review, which also discusses the influence of creating in vitro biomimetic 3D tumor microenvironments utilizing functional biomaterials. Future cancer treatments will rely much more heavily on biomaterials engineering.
Collapse
Affiliation(s)
- Tanvir Ahmed
- Department of Pharmaceutical Sciences, North South University, Bashundhara R/A, Dhaka-1229 Dhaka, Bangladesh
| |
Collapse
|
16
|
Zhu M, Wang Q, Gu T, Han Y, Zeng X, Li J, Dong J, Huang H, Qian P. Hydrogel-based microenvironment engineering of haematopoietic stem cells. Cell Mol Life Sci 2023; 80:49. [PMID: 36690903 PMCID: PMC11073069 DOI: 10.1007/s00018-023-04696-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2022] [Revised: 11/06/2022] [Accepted: 01/08/2023] [Indexed: 01/25/2023]
Abstract
Haematopoietic Stem cells (HSCs) have the potential for self-renewal and multilineage differentiation, and their behaviours are finely tuned by the microenvironment. HSC transplantation (HSCT) is widely used in the treatment of haematologic malignancies while limited by the quantity of available HSCs. With the development of tissue engineering, hydrogels have been deployed to mimic the HSC microenvironment in vitro. Engineered hydrogels influence HSC behaviour by regulating mechanical strength, extracellular matrix microstructure, cellular ligands and cytokines, cell-cell interaction, and oxygen concentration, which ultimately facilitate the acquisition of sufficient HSCs. Here, we review recent advances in the application of hydrogel-based microenvironment engineering of HSCs, and provide future perspectives on challenges in basic research and clinical practice.
Collapse
Affiliation(s)
- Meng Zhu
- Center of Stem Cell and Regenerative Medicine, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Qiwei Wang
- Center of Stem Cell and Regenerative Medicine, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Tianning Gu
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China
| | - Yingli Han
- Center of Stem Cell and Regenerative Medicine, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Xin Zeng
- Center of Stem Cell and Regenerative Medicine, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Jinxin Li
- Center of Stem Cell and Regenerative Medicine, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - Jian Dong
- Center of Stem Cell and Regenerative Medicine, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China
| | - He Huang
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China.
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China.
- Bone Marrow Transplantation Center, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, China.
| | - Pengxu Qian
- Center of Stem Cell and Regenerative Medicine, and Bone Marrow Transplantation Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310058, China.
- Liangzhu Laboratory, Zhejiang University Medical Center, 1369 West Wenyi Road, Hangzhou, 311121, China.
- Institute of Hematology, Zhejiang University and Zhejiang Engineering Laboratory for Stem Cell and Immunotherapy, Hangzhou, 310058, China.
| |
Collapse
|
17
|
Geevarghese R, Sajjadi SS, Hudecki A, Sajjadi S, Jalal NR, Madrakian T, Ahmadi M, Włodarczyk-Biegun MK, Ghavami S, Likus W, Siemianowicz K, Łos MJ. Biodegradable and Non-Biodegradable Biomaterials and Their Effect on Cell Differentiation. Int J Mol Sci 2022; 23:ijms232416185. [PMID: 36555829 PMCID: PMC9785373 DOI: 10.3390/ijms232416185] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Biomaterials for tissue scaffolds are key components in modern tissue engineering and regenerative medicine. Targeted reconstructive therapies require a proper choice of biomaterial and an adequate choice of cells to be seeded on it. The introduction of stem cells, and the transdifferentiation procedures, into regenerative medicine opened a new era and created new challenges for modern biomaterials. They must not only fulfill the mechanical functions of a scaffold for implanted cells and represent the expected mechanical strength of the artificial tissue, but furthermore, they should also assure their survival and, if possible, affect their desired way of differentiation. This paper aims to review how modern biomaterials, including synthetic (i.e., polylactic acid, polyurethane, polyvinyl alcohol, polyethylene terephthalate, ceramics) and natural (i.e., silk fibroin, decellularized scaffolds), both non-biodegradable and biodegradable, could influence (tissue) stem cells fate, regulate and direct their differentiation into desired target somatic cells.
Collapse
Affiliation(s)
- Rency Geevarghese
- Biotechnology Center, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Seyedeh Sara Sajjadi
- School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1971653313, Iran
| | - Andrzej Hudecki
- Łukasiewicz Network-Institute of Non-Ferrous Metals, 44-121 Gliwice, Poland
| | - Samad Sajjadi
- School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1971653313, Iran
| | | | - Tayyebeh Madrakian
- Faculty of Chemistry, Bu-Ali Sina University, Hamedan 6516738695, Iran
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| | - Mazaher Ahmadi
- Faculty of Chemistry, Bu-Ali Sina University, Hamedan 6516738695, Iran
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| | - Małgorzata K. Włodarczyk-Biegun
- Biotechnology Center, Silesian University of Technology, 44-100 Gliwice, Poland
- Polymer Science, Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| | - Saeid Ghavami
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada
- Research Institutes of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB R3E 0V9, Canada
- Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
- Faculty of Medicine in Zabrze, University of Technology in Katowice, 41-800 Zabrze, Poland
| | - Wirginia Likus
- Department of Anatomy, Faculty of Health Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland
| | - Krzysztof Siemianowicz
- Department of Biochemistry, Faculty of Medicine in Katowice, Medical University of Silesia, 40-752 Katowice, Poland
- Correspondence: (K.S.); (M.J.Ł.); Tel.: +48-32-237-2913 (M.J.Ł.)
| | - Marek J. Łos
- Biotechnology Center, Silesian University of Technology, 44-100 Gliwice, Poland
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Correspondence: (K.S.); (M.J.Ł.); Tel.: +48-32-237-2913 (M.J.Ł.)
| |
Collapse
|
18
|
Ohlsson E, Galler KM, Widbiller M. A Compilation of Study Models for Dental Pulp Regeneration. Int J Mol Sci 2022; 23:ijms232214361. [PMID: 36430838 PMCID: PMC9695686 DOI: 10.3390/ijms232214361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/12/2022] [Accepted: 11/14/2022] [Indexed: 11/22/2022] Open
Abstract
Efforts to heal damaged pulp tissue through tissue engineering have produced positive results in pilot trials. However, the differentiation between real regeneration and mere repair is not possible through clinical measures. Therefore, preclinical study models are still of great importance, both to gain insights into treatment outcomes on tissue and cell levels and to develop further concepts for dental pulp regeneration. This review aims at compiling information about different in vitro and in vivo ectopic, semiorthotopic, and orthotopic models. In this context, the differences between monolayer and three-dimensional cell cultures are discussed, a semiorthotopic transplantation model is introduced as an in vivo model for dental pulp regeneration, and finally, different animal models used for in vivo orthotopic investigations are presented.
Collapse
Affiliation(s)
- Ella Ohlsson
- Department of Operative Dentistry and Periodontology, Friedrich-Alexander-University Erlangen-Nuernberg, D-91054 Erlangen, Germany
| | - Kerstin M. Galler
- Department of Operative Dentistry and Periodontology, Friedrich-Alexander-University Erlangen-Nuernberg, D-91054 Erlangen, Germany
| | - Matthias Widbiller
- Department of Conservative Dentistry and Periodontology, University Hospital Regensburg, D-93053 Regensburg, Germany
- Correspondence:
| |
Collapse
|
19
|
Thangadurai M, Ajith A, Budharaju H, Sethuraman S, Sundaramurthi D. Advances in electrospinning and 3D bioprinting strategies to enhance functional regeneration of skeletal muscle tissue. BIOMATERIALS ADVANCES 2022; 142:213135. [PMID: 36215745 DOI: 10.1016/j.bioadv.2022.213135] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Revised: 08/31/2022] [Accepted: 09/25/2022] [Indexed: 06/16/2023]
Abstract
Skeletal muscles are essential for body movement, and the loss of motor function due to volumetric muscle loss (VML) limits the mobility of patients. Current therapeutic approaches are insufficient to offer complete functional recovery of muscle damages. Tissue engineering provides viable ways to fabricate scaffolds to regenerate damaged tissues. Hence, tissue engineering options are explored to address existing challenges in the treatment options for muscle regeneration. Electrospinning is a widely employed fabrication technique to make muscle mimetic nanofibrous scaffolds for tissue regeneration. 3D bioprinting has also been utilized to fabricate muscle-like tissues in recent times. This review discusses the anatomy of skeletal muscle, defects, the healing process, and various treatment options for VML. Further, the advanced strategies in electrospinning of natural and synthetic polymers are discussed, along with the recent developments in the fabrication of hybrid scaffolds. Current approaches in 3D bioprinting of skeletal muscle tissues are outlined with special emphasis on the combination of electrospinning and 3D bioprinting towards the development of fully functional muscle constructs. Finally, the current challenges and future perspectives of these convergence techniques are discussed.
Collapse
Affiliation(s)
- Madhumithra Thangadurai
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, India
| | - Athulya Ajith
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, India
| | - Harshavardhan Budharaju
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, India
| | - Swaminathan Sethuraman
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, India
| | - Dhakshinamoorthy Sundaramurthi
- Tissue Engineering & Additive Manufacturing (TEAM) Lab, Centre for Nanotechnology & Advanced Biomaterials, ABCDE Innovation Centre, School of Chemical & Biotechnology, SASTRA Deemed University, India.
| |
Collapse
|
20
|
Temple J, Velliou E, Shehata M, Lévy R, Gupta P. Current strategies with implementation of three-dimensional cell culture: the challenge of quantification. Interface Focus 2022; 12:20220019. [PMID: 35992772 PMCID: PMC9372643 DOI: 10.1098/rsfs.2022.0019] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2022] [Accepted: 07/20/2022] [Indexed: 12/14/2022] Open
Abstract
From growing cells in spheroids to arranging them on complex engineered scaffolds, three-dimensional cell culture protocols are rapidly expanding and diversifying. While these systems may often improve the physiological relevance of cell culture models, they come with technical challenges, as many of the analytical methods used to characterize traditional two-dimensional (2D) cells must be modified or replaced to be effective. Here we review the advantages and limitations of quantification methods based either on biochemical measurements or microscopy imaging. We focus on the most basic of parameters that one may want to measure, the number of cells. Precise determination of this number is essential for many analytical techniques where measured quantities are only meaningful when normalized to the number of cells (e.g. cytochrome p450 enzyme activity). Thus, accurate measurement of cell number is often a prerequisite to allowing comparisons across different conditions (culturing conditions or drug and treatment screening) or between cells in different spatial states. We note that this issue is often neglected in the literature with little or no information given regarding how normalization was performed, we highlight the pitfalls and complications of quantification and call for more accurate reporting to improve reproducibility.
Collapse
Affiliation(s)
- Jonathan Temple
- Bioscience building, University of Liverpool, Liverpool L69 3BX, UK
| | - Eirini Velliou
- Centre for 3D Models of Health and Disease, University College London, London, UK
| | - Mona Shehata
- Hutchison-MRC Research Centre, University of Cambridge, Cambridge CB2 1TN, UK
| | - Raphaël Lévy
- Bioscience building, University of Liverpool, Liverpool L69 3BX, UK
- Laboratoire for Vascular Translational Science, Université Sorbonne Paris Nord, Bobigny, France
| | - Priyanka Gupta
- Centre for 3D Models of Health and Disease, University College London, London, UK
| |
Collapse
|
21
|
A reductionist approach to determine the effect of cell-cell contact on human epidermal stem cell differentiation. Acta Biomater 2022; 150:265-276. [PMID: 35926780 PMCID: PMC9810539 DOI: 10.1016/j.actbio.2022.07.054] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 07/23/2022] [Accepted: 07/27/2022] [Indexed: 01/07/2023]
Abstract
The balance between stem cell renewal and differentiation is determined by the interplay between intrinsic cellular controls and extrinsic factors presented by the microenvironment, or 'niche'. Previous studies on cultured human epidermis have utilised suspension culture and restricted cell spreading to investigate regulation of differentiation in single keratinocytes. However, keratinocytes are typically adherent to neighbouring cells in vivo. We therefore developed experimental models to investigate the combined effects of cell-ECM adhesion and cell-cell contact. We utilized lipid-modified oligonucleotides to form clusters of keratinocytes which were subsequently placed in suspension to induce terminal differentiation. In this experimental model cell-cell contact had no effect on suspension-induced differentiation of keratinocytes. We next developed a high-throughput platform for robust geometrical confinement of keratinocytes to hexagonal ECM-coated islands permitting direct cell-cell contact between single cells. As in the case of circular islands, differentiation was stimulated on the smallest single hexagonal islands. However, the percentage of involucrin-positive cells on small bowtie islands was significantly lower than on single islands, demonstrating that cell-cell contact reduced differentiation in response to decreased substrate adhesion. None of the small bowtie islands contained two involucrin-positive cells. Rather, if one cell was involucrin-positive the other was involucrin-negative. This suggests that there is intrinsic asymmetry in the effect of cell-cell contact in decreasing differentiation. Thus, our reductionist approaches provide new insights into the effect of the niche on keratinocyte differentiation. STATEMENT OF SIGNIFICANCE: Stem cell behaviour is regulated by a combination of external signals, including the nature of the adhesive substrate and cell-cell interactions. An understanding of how different signals are integrated creates the possibility of developing new biomaterials to promote tissue regeneration and broaden our understanding of skin diseases such as eczema and psoriasis, in which stem cell proliferation and differentiation are perturbed. In this study we have applied two methods to engineer intercellular adhesion of human epidermal stem cells, one involving lipid-modified DNA and the other involving hexagonal micropatterns. We show that the effect of cell-cell adhesion depends on cell-substrate adhesion and uncover evidence that two cells in equivalent environments can nevertheless behave differently.
Collapse
|
22
|
Kim W, Park E, Yoo HS, Park J, Jung YM, Park JH. Recent Advances in Monitoring Stem Cell Status and Differentiation Using Nano-Biosensing Technologies. NANOMATERIALS (BASEL, SWITZERLAND) 2022; 12:2934. [PMID: 36079970 PMCID: PMC9457759 DOI: 10.3390/nano12172934] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/29/2022] [Revised: 08/22/2022] [Accepted: 08/23/2022] [Indexed: 05/14/2023]
Abstract
In regenerative medicine, cell therapies using various stem cells have received attention as an alternative to overcome the limitations of existing therapeutic methods. Clinical applications of stem cells require the identification of characteristics at the single-cell level and continuous monitoring during expansion and differentiation. In this review, we recapitulate the application of various stem cells used in regenerative medicine and the latest technological advances in monitoring the differentiation process of stem cells. Single-cell RNA sequencing capable of profiling the expression of many genes at the single-cell level provides a new opportunity to analyze stem cell heterogeneity and to specify molecular markers related to the branching of differentiation lineages. However, this method is destructive and distorted. In addition, the differentiation process of a particular cell cannot be continuously tracked. Therefore, several spectroscopic methods have been developed to overcome these limitations. In particular, the application of Raman spectroscopy to measure the intrinsic vibration spectrum of molecules has been proposed as a powerful method that enables continuous monitoring of biochemical changes in the process of the differentiation of stem cells. This review provides a comprehensive overview of current analytical methods employed for stem cell engineering and future perspectives of nano-biosensing technologies as a platform for the in situ monitoring of stem cell status and differentiation.
Collapse
Affiliation(s)
- Wijin Kim
- Department of Biomedical Science, Kangwon National University, Chuncheon 24341, Gangwon-do, Korea
| | - Eungyeong Park
- Department of Chemistry, Kangwon National University, Chuncheon 24341, Gangwon-do, Korea
| | - Hyuk Sang Yoo
- Department of Biomedical Science, Kangwon National University, Chuncheon 24341, Gangwon-do, Korea
- Kangwon Radiation Convergence Research Support Center, Kangwon National University, Chuncheon 24341, Gangwon-do, Korea
| | - Jongmin Park
- Department of Chemistry, Kangwon National University, Chuncheon 24341, Gangwon-do, Korea
| | - Young Mee Jung
- Department of Chemistry, Kangwon National University, Chuncheon 24341, Gangwon-do, Korea
- Kangwon Radiation Convergence Research Support Center, Kangwon National University, Chuncheon 24341, Gangwon-do, Korea
| | - Ju Hyun Park
- Department of Biomedical Science, Kangwon National University, Chuncheon 24341, Gangwon-do, Korea
| |
Collapse
|
23
|
Copeland KM, Brazile BL, Butler JR, Cooley J, Brinkman-Ferguson E, Claude A, Lin S, Rais-Rohani S, Welch B, McMahan SR, Nguyen KT, Hong Y, Ramaswamy S, Liu ZP, Bajona P, Peltz M, Liao J. Investigating the Transient Regenerative Potential of Cardiac Muscle Using a Neonatal Pig Partial Apical Resection Model. Bioengineering (Basel) 2022; 9:401. [PMID: 36004926 PMCID: PMC9404987 DOI: 10.3390/bioengineering9080401] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 06/16/2022] [Accepted: 08/16/2022] [Indexed: 11/17/2022] Open
Abstract
Researchers have shown that adult zebrafish have the potential to regenerate 20% of the ventricular muscle within two months of apex resection, and neonatal mice have the capacity to regenerate their heart after apex resection up until day 7 after birth. The goal of this study was to determine if large mammals (porcine heart model) have the capability to fully regenerate a resected portion of the left ventricular apex during the neonatal stage, and if so, how long the regenerative potential persists. A total of 36 piglets were divided into the following groups: 0-day control and surgical groups and seven-day control and surgical groups. For the apex removal groups, each piglet was subjected to a partial wall thickness resection (~30% of the ventricular wall thickness). Heart muscle function was assessed via transthoracic echocardiograms; the seven-day surgery group experienced a decrease in ejection fraction and fractional shortening. Upon gross necropsy, for piglets euthanized four weeks post-surgery, all 0-day-old hearts showed no signs of scarring or any indication of the induced injury. Histological analysis confirmed that piglets in the 0-day surgery group exhibited various degrees of regeneration, with half of the piglets showing full regeneration and the other half showing partial regeneration. However, each piglet in the seven-day surgery group demonstrated epicardial fibrosis along with moderate to severe dissecting interstitial fibrosis, which was accompanied by an abundant collagenous extracellular matrix as the result of a scar formation in the resection site. Histology of one 0-day apex resection piglet (briefly lain on and accidentally killed by the mother sow three days post-surgery) revealed dense, proliferative mesenchymal cells bordering the fibrin and hemorrhage zone and differentiating toward immature cardiomyocytes. We further examined the heart explants at 5-days post-surgery (5D PO) and 1-week post-surgery (1W PO) to assess the repair progression. For the 0-day surgery piglets euthanized at 5D PO and 1W PO, half had abundant proliferating mesenchymal cells, suggesting active regeneration, while the other half showed increased extracellular collagen. The seven-day surgery piglets euthanized at 5D PO, and 1W PO showed evidence of greatly increased extracellular collagen, while some piglets had proliferating mesenchymal cells, suggesting a regenerative effort is ongoing while scar formation seems to predominate. In short, our qualitative findings suggest that the piglets lose the full myocardial regenerative potential by 7 days after birth, but greatly preserve the regenerative potential within 1 day post-partum.
Collapse
Affiliation(s)
- Katherine M. Copeland
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76010, USA
| | - Bryn L. Brazile
- Department of Biological Engineering, College of Veterinary Medicine, Mississippi State University, Starkville, MS 39762, USA
| | - J. Ryan Butler
- Department of Biological Engineering, College of Veterinary Medicine, Mississippi State University, Starkville, MS 39762, USA
| | - Jim Cooley
- Department of Biological Engineering, College of Veterinary Medicine, Mississippi State University, Starkville, MS 39762, USA
| | - Erin Brinkman-Ferguson
- Department of Biological Engineering, College of Veterinary Medicine, Mississippi State University, Starkville, MS 39762, USA
| | - Andrew Claude
- Department of Biological Engineering, College of Veterinary Medicine, Mississippi State University, Starkville, MS 39762, USA
| | - Sallie Lin
- Department of Biological Engineering, College of Veterinary Medicine, Mississippi State University, Starkville, MS 39762, USA
| | - Sammira Rais-Rohani
- Department of Biological Engineering, College of Veterinary Medicine, Mississippi State University, Starkville, MS 39762, USA
| | - Bradley Welch
- Department of Biological Engineering, College of Veterinary Medicine, Mississippi State University, Starkville, MS 39762, USA
| | - Sara R. McMahan
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76010, USA
| | - Kytai T. Nguyen
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76010, USA
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76010, USA
| | - Sharan Ramaswamy
- Department of Biomedical Engineering, Florida International University, Miami, FL 33174, USA
| | - Zhi-Ping Liu
- Department of Cardiovascular and Thoracic Surgery, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Pietro Bajona
- Department of Cardiovascular and Thoracic Surgery, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
- Allegheny Health Network-Drexel University College of Medicine, Pittsburgh, PA 15212, USA
| | - Matthias Peltz
- Department of Cardiovascular and Thoracic Surgery, Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX 75390, USA
| | - Jun Liao
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX 76010, USA
| |
Collapse
|
24
|
Standardised quantitative ultrasound imaging (SQUI) approach for the contact-less three-dimensional analysis of neocartilage formation in hydrogel-based bioscaffolds. Acta Biomater 2022; 147:129-146. [PMID: 35643197 DOI: 10.1016/j.actbio.2022.05.037] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2021] [Revised: 05/18/2022] [Accepted: 05/19/2022] [Indexed: 12/17/2022]
Abstract
In this work we present a standardised quantitative ultrasound imaging (SQUI) approach for the non-destructive three-dimensional imaging and quantification of cartilage formation in hydrogel based bioscaffolds. The standardised concept involves the processing of ultrasound backscatter data with respect to an acellular phantom in combination with the compensation of sound speed mismatch diffraction effects between the bioscaffold and the phantom. As a proof-of-concept, the SQUI approach was tested on a variety of bioscaffolds with varying degree of neocartilage formation. These were composed of Gelatine Methacryloyl (GelMA) hydrogels laden with human adipose-derived stem cells (hADSCs). These were cultured under chondrogenic stimulation following a previously established protocol, where the degree of the neocartilage formation was modulated using different GelMA network densities (6, 8, 10 % w/v) and culture time (0, 14, 28 days). Using the SQUI approach we were able to detect marked acoustic and morphological changes occurring in the bioscaffolds a result of their different chondrogenic outcome. We defined an acoustic neocartilage indicator, the sonomarker, for the selective imaging and quantification of neocartilage formation. The sonomarker, of backscatter intensity logIBC -2.4, was found to correlate with data obtained via standard destructive bioassays. The ultrasonic evaluation of human specimens confirmed the sonomarker as a relevant intensity, although it was found to shift to higher intensity values in proportion to the cartilage condition as inferred from sound speed measurements. This study demonstrates the potential of the SQUI approach for the realization of non-destructive analysis of cartilage regeneration over-time. STATEMENT OF SIGNIFICANCE: As tissue engineering strategies for neocartilage regeneration evolve towards clinical implementation, alternative characterisation approaches that allow the non-destructive monitoring of extracellular matrix formation in implantable hydrogel based bioscaffolds are needed. In this work we present an innovative standardized quantitative ultrasound imaging (SQUI) approach that allows the non-destructive, volumetric, and quantitative evaluation of neocartilage formation in hydrogel based bioscaffolds. The standardised concept aims to provide a robust approach that accounts for the dynamic changes occurring during the conversion from a cellular bioscaffold towards the formation of a neocartilage construct. We believe that the SQUI approach will be of great benefit for the evaluation of constructs developing neocartilage, not only for in-vitro applications but also potentially applicable to in-vivo applications.
Collapse
|
25
|
Application of Bio-Active Elastin-like Polypeptide on Regulation of Human Mesenchymal Stem Cell Behavior. Biomedicines 2022; 10:biomedicines10051151. [PMID: 35625887 PMCID: PMC9138580 DOI: 10.3390/biomedicines10051151] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2022] [Revised: 02/24/2022] [Accepted: 02/24/2022] [Indexed: 02/01/2023] Open
Abstract
Regenerative medicine using stem cells offers promising strategies for treating a variety of degenerative diseases. Regulation of stem cell behavior and rejuvenate senescence are required for stem cells to be clinically effective. The extracellular matrix (ECM) components have a significant impact on the stem cell’s function and fate mimicking the local environment to maintain cells or generate a distinct phenotype. Here, human elastin-like polypeptide-based ECM-mimic biopolymer was designed by incorporating various cell-adhesion ligands, such as RGD and YIGSR. The significant effects of bioactive fusion ELPs named R-ELP, Y-ELP, and RY-ELP were analyzed for human bone-marrow-derived stem cell adhesion, proliferation, maintenance of stemness properties, and differentiation. Multivalent presentation of variable cell-adhesive ligands on RY-ELP polymers indeed promote efficient cell attachment and proliferation of human fibroblast cells dose-dependently. Similarly, surface modified with RY-ELP promoted strong mesenchymal stem cell (MSCs) attachment with greater focal adhesion (FA) complex formation at 6 h post-incubation. The rate of cell proliferation, migration, population doubling time, and collagen I deposition were significantly enhanced in the presence of RY-ELP compared with other fusion ELPs. Together, the expression of multipotent markers and differentiation capacity of MSCs remained unaffected, clearly demonstrating that stemness properties of MSCs were well preserved when cultured on a RY-ELP-modified surface. Hence, bioactive RY-ELP offers an anchorage support system and effectively induces stimulatory response to support stem cell proliferation.
Collapse
|
26
|
Ding M, Koroma KE, Wendt D, Martin I, Martinetti R, Jespersen S, Schrøder HD, Overgaard S. Efficacy of bioreactor-activated bone substitute with bone marrow nuclear cells on fusion rate and fusion mass microarchitecture in sheep. J Biomed Mater Res B Appl Biomater 2022; 110:1862-1875. [PMID: 35233920 DOI: 10.1002/jbm.b.35044] [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: 04/09/2020] [Revised: 04/02/2021] [Accepted: 02/22/2022] [Indexed: 11/07/2022]
Abstract
Bioreactors have been used for bone graft engineering in pre-clinical investigations over the past 15 years. The ability of bioreactor-incubated bone marrow nuclear cells (BMNCs) to enhance bone-forming potential varies significantly, and the three-dimensional (3D) distribution of BMNCs within the scaffold is largely unknown. The aims of this study were (1) to investigate the efficacy of a carbonated hydroxyapatite (CHA) with/without BMNCs on spine fusion rate and fusion mass microarchitecture using a highly challenging two-level posterolateral spine fusion without instrumentation; and (2) to evaluate 3D distribution of BMNCs within scaffolds characterized by immunohistochemistry. Fusion rate and fusion mass were quantified by micro-CT, microarchitectural analysis, and histology. While the homogenous 3D distribution of BMNCs was not observed, BMNCs were found to migrate towards a substitute core. In the autograft group, the healing rate was 83.3%, irrespective of the presence of BMNCs. In the CHA group, also 83.3% was fused in the presence of BMNCs, and 66.7% fused without BMNCs. A significant decrease in the fusion mass porosity (p = .001) of the CHA group suggested the deposition of mineralized bone. The autograft group revealed more bone, thicker trabeculae, and better trabecular orientation but less connection compared to the CHA group. Immunohistochemistry confirmed the ability of bioreactors to incubate a large-sized substitute coated with viable BMNCs with the potential for proliferation and differentiation. These findings suggested that a bioreactor-activated substitute is comparable to autograft on spine fusion and that new functional bone regeneration could be achieved by a combination of BMNCs, biomaterials, and bioreactors.
Collapse
Affiliation(s)
- Ming Ding
- Orthopaedic Research Laboratory, Department of Orthopaedic Surgery and Traumatology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Kariatta Esther Koroma
- Orthopaedic Research Laboratory, Department of Orthopaedic Surgery and Traumatology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - David Wendt
- Department of Surgery and of Biomedicine, University Hospital Basel, Basel, Switzerland
| | - Ivan Martin
- Department of Surgery and of Biomedicine, University Hospital Basel, Basel, Switzerland
| | | | - Stig Jespersen
- Orthopaedic Research Laboratory, Department of Orthopaedic Surgery and Traumatology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark
| | - Henrik Daa Schrøder
- Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Pathology, Odense University Hospital, Odense, Denmark
| | - Søren Overgaard
- Orthopaedic Research Laboratory, Department of Orthopaedic Surgery and Traumatology, Odense University Hospital, Odense, Denmark.,Department of Clinical Research, University of Southern Denmark, Odense, Denmark.,Department of Orthopaedic Surgery & Traumatology, Copenhagen University Hospital, Bispebjerg, Copenhagen, Denmark.,Department of Clinical Medicine, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
27
|
Maji S, Lee H. Engineering Hydrogels for the Development of Three-Dimensional In Vitro Models. Int J Mol Sci 2022; 23:2662. [PMID: 35269803 PMCID: PMC8910155 DOI: 10.3390/ijms23052662] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2021] [Revised: 02/25/2022] [Accepted: 02/26/2022] [Indexed: 02/06/2023] Open
Abstract
The superiority of in vitro 3D cultures over conventional 2D cell cultures is well recognized by the scientific community for its relevance in mimicking the native tissue architecture and functionality. The recent paradigm shift in the field of tissue engineering toward the development of 3D in vitro models can be realized with its myriad of applications, including drug screening, developing alternative diagnostics, and regenerative medicine. Hydrogels are considered the most suitable biomaterial for developing an in vitro model owing to their similarity in features to the extracellular microenvironment of native tissue. In this review article, recent progress in the use of hydrogel-based biomaterial for the development of 3D in vitro biomimetic tissue models is highlighted. Discussions of hydrogel sources and the latest hybrid system with different combinations of biopolymers are also presented. The hydrogel crosslinking mechanism and design consideration are summarized, followed by different types of available hydrogel module systems along with recent microfabrication technologies. We also present the latest developments in engineering hydrogel-based 3D in vitro models targeting specific tissues. Finally, we discuss the challenges surrounding current in vitro platforms and 3D models in the light of future perspectives for an improved biomimetic in vitro organ system.
Collapse
Affiliation(s)
- Somnath Maji
- Department of Mechanical and Biomedical Engineering, Kangwon National University (KNU), Chuncheon 24341, Korea;
| | - Hyungseok Lee
- Department of Mechanical and Biomedical Engineering, Kangwon National University (KNU), Chuncheon 24341, Korea;
- Department of Smart Health Science and Technology, Kangwon National University (KNU), Chuncheon 24341, Korea
| |
Collapse
|
28
|
Low-Cost Devices for Three-Dimensional Cell Aggregation, Real-Time Monitoring Microscopy, Microfluidic Immunostaining, and Deconvolution Analysis. Bioengineering (Basel) 2022; 9:bioengineering9020060. [PMID: 35200413 PMCID: PMC8869754 DOI: 10.3390/bioengineering9020060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2021] [Revised: 01/25/2022] [Accepted: 01/28/2022] [Indexed: 11/23/2022] Open
Abstract
The wide use of 3D-organotypic cell models is imperative for advancing our understanding of basic cell biological mechanisms. For this purpose, easy-to-use enabling technology is required, which should optimally link standardized assessment methods to those used for the formation, cultivation, and evaluation of cell aggregates or primordial tissue. We thus conceived, manufactured, and tested devices which provide the means for cell aggregation and online monitoring within a hanging drop. We then established a workflow for spheroid manipulation and immune phenotyping. This described workflow conserves media and reagent, facilitates the uninterrupted tracking of spheroid formation under various conditions, and enables 3D-marker analysis by means of 3D epifluorescence deconvolution microscopy. We provide a full description of the low-cost manufacturing process for the fluidic devices and microscopic assessment tools, and the detailed blueprints and building instructions are disclosed. Conclusively, the presented compilation of methods and techniques promotes a quick and barrier-free entry into 3D cell biology.
Collapse
|
29
|
Liu Y, Peng L, Li L, Huang C, Shi K, Meng X, Wang P, Wu M, Li L, Cao H, Wu K, Zeng Q, Pan H, Lu WW, Qin L, Ruan C, Wang X. 3D-bioprinted BMSC-laden biomimetic multiphasic scaffolds for efficient repair of osteochondral defects in an osteoarthritic rat model. Biomaterials 2021; 279:121216. [PMID: 34739982 DOI: 10.1016/j.biomaterials.2021.121216] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Revised: 10/13/2021] [Accepted: 10/20/2021] [Indexed: 01/09/2023]
Abstract
Osteochondral defect repair in osteoarthritis (OA) remains an unsolved clinical problem due to the lack of enough seed cells in the defect and chronic inflammation in the joint. To address this clinical need, we designed a bone marrow-derived mesenchymal stem cell (BMSC)-laden 3D-bioprinted multilayer scaffold with methacrylated hyaluronic acid (MeHA)/polycaprolactone incorporating kartogenin and β-TCP for osteochondral defect repair within each region. BMSC-laden MeHA was designed to actively introduce BMSCs in situ, and diclofenac sodium (DC)-incorporated matrix metalloproteinase-sensitive peptide-modified MeHA was induced on the BMSC-laden scaffold as an anti-inflammatory strategy. BMSCs in the scaffolds survived, proliferated, and produced large amounts of cartilage-specific extracellular matrix in vitro. The effect of BMSC-laden scaffolds on osteochondral defect repair was investigated in an animal model of medial meniscectomy-induced OA. BMSC-laden scaffolds facilitated chondrogenesis by promoting collagen II and suppressed interleukin 1β in osteochondral defects of the femoral trochlea. Congruently, BMSC-laden scaffolds significantly improved joint function of the injured leg with respect to the ground support force, paw grip force, and walk gait parameters. Therefore, this research demonstrates the potential of 3D-bioprinted BMSC-laden scaffolds to simultaneously inhibit joint inflammation and promote cartilage defect repair in OA joints.
Collapse
Affiliation(s)
- Yanzhi Liu
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Guangdong Key Laboratory for Research and Development of Natural Drugs, Marine Medical Research Institute, Guangdong Medical University, Zhanjiang, 524023, China
| | - Liuqi Peng
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Lingli Li
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Cuishan Huang
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Keda Shi
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Xiangbo Meng
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Pinpin Wang
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Mingming Wu
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Ling Li
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Huijuan Cao
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Kefeng Wu
- Guangdong Key Laboratory for Research and Development of Natural Drugs, Marine Medical Research Institute, Guangdong Medical University, Zhanjiang, 524023, China
| | - Qingqiang Zeng
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Haobo Pan
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - William Weijia Lu
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Department of Orthopaedic and Traumatology, The University of Hong Kong, 21 Sassoon Road, Pokfulam, Hong Kong SAR, China
| | - Ling Qin
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China; Musculoskeletal Research Laboratory of Department of Orthopaedics & Traumatology and Innovative Orthopaedic Biomaterial and Drug Translational Research Laboratory of Li Ka Shing Institute of Health, The Chinese University of Hong Kong, Hong Kong SAR, China.
| | - Changshun Ruan
- Research Center for Human Tissue and Organs Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| | - Xinluan Wang
- Translational Medicine R&D Center, Institute of Biomedical and Health Engineering, Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
30
|
Lee SY, Ma J, Khoo TS, Abdullah N, Nik Md Noordin Kahar NNF, Abdul Hamid ZA, Mustapha M. Polysaccharide-Based Hydrogels for Microencapsulation of Stem Cells in Regenerative Medicine. Front Bioeng Biotechnol 2021; 9:735090. [PMID: 34733829 PMCID: PMC8558675 DOI: 10.3389/fbioe.2021.735090] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 09/27/2021] [Indexed: 12/29/2022] Open
Abstract
Stem cell-based therapy appears as a promising strategy to induce regeneration of damaged and diseased tissues. However, low survival, poor engraftment and a lack of site-specificity are major drawbacks. Polysaccharide hydrogels can address these issues and offer several advantages as cell delivery vehicles. They have become very popular due to their unique properties such as high-water content, biocompatibility, biodegradability and flexibility. Polysaccharide polymers can be physically or chemically crosslinked to construct biomimetic hydrogels. Their resemblance to living tissues mimics the native three-dimensional extracellular matrix and supports stem cell survival, proliferation and differentiation. Given the intricate nature of communication between hydrogels and stem cells, understanding their interaction is crucial. Cells are incorporated with polysaccharide hydrogels using various microencapsulation techniques, allowing generation of more relevant models and further enhancement of stem cell therapies. This paper provides a comprehensive review of human stem cells and polysaccharide hydrogels most used in regenerative medicine. The recent and advanced stem cell microencapsulation techniques, which include extrusion, emulsion, lithography, microfluidics, superhydrophobic surfaces and bioprinting, are described. This review also discusses current progress in clinical translation of stem-cell encapsulated polysaccharide hydrogels for cell delivery and disease modeling (drug testing and discovery) with focuses on musculoskeletal, nervous, cardiac and cancerous tissues.
Collapse
Affiliation(s)
- Si-Yuen Lee
- Department of Medicine, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| | - Jingyi Ma
- Duke-NUS Medical School, Singapore, Singapore
| | - Tze Sean Khoo
- UKM Medical Molecular Biology Institute, National University of Malaysia, Bangi, Malaysia
| | - Norfadhilatuladha Abdullah
- Advanced Membrane Technology Research Centre, Faculty of Chemical and Energy Engineering, Universiti Teknologi Malaysia, Skudai, Malaysia
| | | | - Zuratul Ain Abdul Hamid
- School of Materials and Mineral Resources Engineering, Universiti Sains Malaysia, Nibong Tebal, Malaysia
| | - Muzaimi Mustapha
- Department of Neurosciences, School of Medical Sciences, Universiti Sains Malaysia, Kota Bharu, Malaysia
| |
Collapse
|
31
|
Comparative Transcriptome Analysis of Human Adipose-Derived Stem Cells Undergoing Osteogenesis in 2D and 3D Culture Conditions. Int J Mol Sci 2021; 22:ijms22157939. [PMID: 34360705 PMCID: PMC8347556 DOI: 10.3390/ijms22157939] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/18/2021] [Accepted: 07/21/2021] [Indexed: 01/03/2023] Open
Abstract
Human adipose-derived stem cells (hADSCs) are types of mesenchymal stem cells (MSCs) that have been used as tissue engineering models for bone, cartilage, muscle, marrow stroma, tendon, fat and other connective tissues. Tissue regeneration materials composed of hADSCs have the potential to play an important role in reconstituting damaged tissue or diseased mesenchymal tissue. In this study, we assessed and investigated the osteogenesis of hADSCs in both two-dimensional (2D) and three-dimensional (3D) culture conditions. We confirmed that the hADSCs successfully differentiated into bone tissues by ARS staining and quantitative RT–PCR. To gain insight into the detailed biological difference between the two culture conditions, we profiled the overall gene expression by analyzing the whole transcriptome sequencing data using various bioinformatic methods. We profiled the overall gene expression through RNA-Seq and further analyzed this using various bioinformatic methods. During differential gene expression testing, significant differences in the gene expressions between hADSCs cultured in 2D and 3D conditions were observed. The genes related to skeletal development, bone development and bone remodeling processes were overexpressed in the 3D culture condition as compared to the 2D culture condition. In summary, our RNA-Seq-based study proves effective in providing new insights that contribute toward achieving a genome-wide understanding of gene regulation in mesenchymal stem cell osteogenic differentiation and bone tissue regeneration within the 3D culture system.
Collapse
|
32
|
Firipis K, Boyd-Moss M, Long B, Dekiwadia C, Hoskin W, Pirogova E, Nisbet DR, Kapsa RMI, Quigley AF, Williams RJ. Tuneable Hybrid Hydrogels via Complementary Self-Assembly of a Bioactive Peptide with a Robust Polysaccharide. ACS Biomater Sci Eng 2021; 7:3340-3350. [PMID: 34125518 DOI: 10.1021/acsbiomaterials.1c00675] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Synthetic materials designed for improved biomimicry of the extracellular matrix must contain fibrous, bioactive, and mechanical cues. Self-assembly of low molecular weight gelator (LMWG) peptides Fmoc-DIKVAV (Fmoc-aspartic acid-isoleucine-lysine-valine-alanine-valine) and Fmoc-FRGDF (Fmoc-phenylalanine-arginine-glycine-aspartic acid-phenylalanine) creates fibrous and bioactive hydrogels. Polysaccharides such as agarose are biocompatible, degradable, and non-toxic. Agarose and these Fmoc-peptides have both demonstrated efficacy in vitro and in vivo. These materials have complementary properties; agarose has known mechanics in the physiological range but is inert and would benefit from bioactive and topographical cues found in the fibrous, protein-rich extracellular matrix. Fmoc-DIKVAV and Fmoc-FRGDF are synthetic self-assembling peptides that present bioactive cues "IKVAV" and "RGD" designed from the ECM proteins laminin and fibronectin. The work presented here demonstrates that the addition of agarose to Fmoc-DIKVAV and Fmoc-FRGDF results in physical characteristics that are dependent on agarose concentration. The networks are peptide-dominated at low agarose concentrations, and agarose-dominated at high agarose concentrations, resulting in distinct changes in structural morphology. Interestingly, at mid-range agarose concentration, a hybrid network is formed with structural similarities to both peptide and agarose systems, demonstrating reinforced mechanical properties. Bioactive-LMWG polysaccharide hydrogels demonstrate controllable microenvironmental properties, providing the ability for tissue-specific biomaterial design for tissue engineering and 3D cell culture.
Collapse
Affiliation(s)
- Kate Firipis
- Biofab3D, Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia.,Biomedical and Electrical Engineering, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia
| | - Mitchell Boyd-Moss
- Biofab3D, Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia.,Biomedical and Electrical Engineering, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia.,Institute of Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia
| | - Benjamin Long
- Faculty of Science and Technology, Federation University, Mt. Helen, VIC 3350, Australia
| | - Chaitali Dekiwadia
- RMIT Microscopy and MicroAnalysis Facility (RMMF), RMIT University, Melbourne, Vic 3000, Australia
| | - William Hoskin
- Faculty of Science and Technology, Federation University, Mt. Helen, VIC 3350, Australia
| | - Elena Pirogova
- Biomedical and Electrical Engineering, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia
| | - David R Nisbet
- Laboratory of Advanced Biomaterials, The Australian National University, Acton, Canberra 2601, Australia
| | - Robert M I Kapsa
- Biofab3D, Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia.,Biomedical and Electrical Engineering, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia.,ARC Centre of Excellence in Electromaterials Science, Department of Medicine, Melbourne University, St Vincent's Hospital Melbourne, Fitzroy, Victoria 3065, Australia.,Department of Medicine, St Vincent's Hospital Melbourne, University of Melbourne, Fitzroy, Vic 3065, Australia
| | - Anita F Quigley
- Biofab3D, Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia.,Biomedical and Electrical Engineering, School of Engineering, RMIT University, Melbourne, VIC 3000, Australia.,ARC Centre of Excellence in Electromaterials Science, Department of Medicine, Melbourne University, St Vincent's Hospital Melbourne, Fitzroy, Victoria 3065, Australia.,Department of Medicine, St Vincent's Hospital Melbourne, University of Melbourne, Fitzroy, Vic 3065, Australia
| | - Richard J Williams
- Biofab3D, Aikenhead Centre for Medical Discovery, St Vincent's Hospital Melbourne, Fitzroy, VIC 3065, Australia.,Institute of Mental and Physical Health and Clinical Translation, School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia
| |
Collapse
|
33
|
Xie YH, Tang CQ, Huang ZZ, Zhou SC, Yang YW, Yin Z, Heng BC, Chen WS, Chen X, Shen WL. ECM remodeling in stem cell culture: a potential target for regulating stem cell function. TISSUE ENGINEERING PART B-REVIEWS 2021; 28:542-554. [PMID: 34082581 DOI: 10.1089/ten.teb.2021.0066] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Stem cells (SCs) hold great potential for regenerative medicine, tissue engineering and cell therapy. The clinical applications of SCs require both high quality and quantity of transplantable cells. However, during conventional in vitro expansion, SCs tend to lose properties that make them amenable for cell therapies. Extracellular matrix (ECM) serves an essential regulatory part in the growth, differentiation and homeostasis of all cells in vivo. when signals transmitted to cells, they do not respond passively. Many cell types can remodel pericellular matrix to meet their specific needs. This reciprocal cell-ECM interaction is crucial for the conservation of cell and tissue functions and homeostasis. In vitro ECM remodeling also plays a key role in regulating the lineage fate of SCs. A deeper understanding of in vitro ECM remodeling may provide new perspectives for the maintenance of SC function. In this review, we critically examined three ways that cells can be used to influence the pericellular matrix: (i) exerting tensile force on the ECM, (ii) secreting a variety of ECM proteins, and (iii) degrading the surrounding matrix, and its impact on SC lineage fate. Finally, we describe the deficiencies of current studies and what needs to be done next to further understand the role of ECM remodeling in ex vivo SC cultures.
Collapse
Affiliation(s)
- Yuan-Hao Xie
- Zhejiang University School of Medicine Second Affiliated Hospital, 89681, Department of Orthopedic Surgery, Hangzhou, Zhejiang, China;
| | - Chen-Qi Tang
- Zhejiang University School of Medicine Second Affiliated Hospital, 89681, Department of Orthopedic Surgery, Hangzhou, Zhejiang, China;
| | - Zi-Zhan Huang
- Zhejiang University School of Medicine Second Affiliated Hospital, 89681, Department of Orthopedic Surgery, Hangzhou, Zhejiang, China;
| | - Si-Cheng Zhou
- Zhejiang University School of Medicine Second Affiliated Hospital, 89681, Hangzhou, Zhejiang, China;
| | - Yu-Wei Yang
- Zhejiang University School of Medicine, 26441, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Hangzhou, Zhejiang, China;
| | - Zi Yin
- Zhejiang University School of Medicine, 26441, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Hangzhou, Zhejiang, China;
| | - Boon Chin Heng
- Peking University School of Stomatology, 159460, Beijing, Beijing, China;
| | - Wei-Shan Chen
- Zhejiang University School of Medicine Second Affiliated Hospital, 89681, Department of Orthopedic Surgery, Hangzhou, Zhejiang, China;
| | - Xiao Chen
- Zhejiang University School of Medicine, 26441, Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cells and Regenerative Medicine, Hangzhou, Zhejiang, China;
| | - Wei-Liang Shen
- Zhejiang University School of Medicine Second Affiliated Hospital, 89681, Department of Orthopedic Surgery, Hangzhou, Zhejiang, China;
| |
Collapse
|
34
|
Saska S, Pilatti L, Silva ESDS, Nagasawa MA, Câmara D, Lizier N, Finger E, Dyszkiewicz Konwińska M, Kempisty B, Tunchel S, Blay A, Shibli JA. Polydioxanone-Based Membranes for Bone Regeneration. Polymers (Basel) 2021; 13:polym13111685. [PMID: 34064251 PMCID: PMC8196877 DOI: 10.3390/polym13111685] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2021] [Revised: 05/15/2021] [Accepted: 05/17/2021] [Indexed: 01/14/2023] Open
Abstract
Resorbable synthetic and natural polymer-based membranes have been extensively studied for guided tissue regeneration. Alloplastic biomaterials are often used for tissue regeneration due to their lower immunoreactivity when compared with allogeneic and xenogeneic materials. Plenum® Guide is a synthetic membrane material based on polydioxanone (PDO), whose surface morphology closely mimics the extracellular matrix. In this study, Plenum® Guide was compared with collagen membranes as a barrier material for bone-tissue regeneration in terms of acute and subchronic systemic toxicity. Moreover, characterizations such as morphology, thermal analysis (Tm = 107.35 °C and crystallinity degree = 52.86 ± 2.97 %, final product), swelling (thickness: 0.25 mm ≅ 436% and 0.5 mm ≅ 425% within 24 h), and mechanical tests (E = 30.1 ± 6.25 MPa; σ = 3.92 ± 0.28 MPa; ε = 287.96 ± 34.68%, final product) were performed. The in vivo results revealed that the PDO membranes induced a slightly higher quantity of newly formed bone tissue than the control group (score: treated group = 15, control group = 13) without detectable systemic toxicity (clinical signs and evaluation of the membranes after necropsy did not result in differences between groups, i.e., non-reaction -> tissue-reaction index = 1.3), showing that these synthetic membranes have the essential characteristics for an effective tissue regeneration. Human adipose-derived stem cells (hASCs) were seeded on PDO membranes; results demonstrated efficient cell migration, adhesion, spread, and proliferation, such that there was a slightly better hASC osteogenic differentiation on PDO than on collagen membranes. Hence, Plenum® Guide membranes are a safe and efficient alternative for resorbable membranes for tissue regeneration.
Collapse
Affiliation(s)
- Sybele Saska
- M3 Health Ind. Com. de Prod. Med. Odont. e Correlatos S.A., 640 Ain Ata, Jundiaí 13212-213, Brazil; (L.P.); (E.S.d.S.S.); (M.A.N.); (S.T.); (A.B.)
- Correspondence: (S.S.); (J.A.S.); Tel.: +55-11-3109-9045 (J.A.S.)
| | - Livia Pilatti
- M3 Health Ind. Com. de Prod. Med. Odont. e Correlatos S.A., 640 Ain Ata, Jundiaí 13212-213, Brazil; (L.P.); (E.S.d.S.S.); (M.A.N.); (S.T.); (A.B.)
| | - Edvaldo Santos de Sousa Silva
- M3 Health Ind. Com. de Prod. Med. Odont. e Correlatos S.A., 640 Ain Ata, Jundiaí 13212-213, Brazil; (L.P.); (E.S.d.S.S.); (M.A.N.); (S.T.); (A.B.)
| | - Magda Aline Nagasawa
- M3 Health Ind. Com. de Prod. Med. Odont. e Correlatos S.A., 640 Ain Ata, Jundiaí 13212-213, Brazil; (L.P.); (E.S.d.S.S.); (M.A.N.); (S.T.); (A.B.)
- Department of Periodontology and Oral Implantology, Dental Research Division, University of Guarulhos, Guarulhos 07023-070, Brazil
| | - Diana Câmara
- Nicell—Pesquisa e Desenvolvimento Ltd.a, 2721 Av. Indianápolis, São Paulo 04063-005, Brazil;
| | - Nelson Lizier
- CCB—Centro de Criogenia Brasil, 1861 Av. Indianápolis, São Paulo 04063-003, Brazil;
| | - Eduardo Finger
- Hospital Israelita Albert Einstein, 627 Av. Albert Einstein, São Paulo 05652-900, Brazil;
| | | | - Bartosz Kempisty
- Department of Histology and Embryology, Poznań University of Medical Sciences, 60-781 Poznan, Poland;
- Department of Veterinary Surgery, Institute of Veterinary Medicine, Nicolaus Copernicus University in Toruń, 87-100 Torun, Poland
- Prestage Department of Poultry Science, North Carolina State University, Raleigh, NC 27695-7608, USA
| | - Samy Tunchel
- M3 Health Ind. Com. de Prod. Med. Odont. e Correlatos S.A., 640 Ain Ata, Jundiaí 13212-213, Brazil; (L.P.); (E.S.d.S.S.); (M.A.N.); (S.T.); (A.B.)
| | - Alberto Blay
- M3 Health Ind. Com. de Prod. Med. Odont. e Correlatos S.A., 640 Ain Ata, Jundiaí 13212-213, Brazil; (L.P.); (E.S.d.S.S.); (M.A.N.); (S.T.); (A.B.)
| | - Jamil Awad Shibli
- M3 Health Ind. Com. de Prod. Med. Odont. e Correlatos S.A., 640 Ain Ata, Jundiaí 13212-213, Brazil; (L.P.); (E.S.d.S.S.); (M.A.N.); (S.T.); (A.B.)
- Department of Periodontology and Oral Implantology, Dental Research Division, University of Guarulhos, Guarulhos 07023-070, Brazil
- Correspondence: (S.S.); (J.A.S.); Tel.: +55-11-3109-9045 (J.A.S.)
| |
Collapse
|
35
|
Brazile BL, Butler JR, Patnaik SS, Claude A, Prabhu R, Williams LN, Perez KL, Nguyen KT, Zhang G, Bajona P, Peltz M, Yang Y, Hong Y, Liao J. Biomechanical properties of acellular scar ECM during the acute to chronic stages of myocardial infarction. J Mech Behav Biomed Mater 2021; 116:104342. [PMID: 33516128 PMCID: PMC8245054 DOI: 10.1016/j.jmbbm.2021.104342] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2020] [Revised: 01/14/2021] [Accepted: 01/17/2021] [Indexed: 02/08/2023]
Abstract
After myocardial infarction (MI), the infarcted tissue undergoes dynamic and time-dependent changes. Previous knowledge on MI biomechanical alterations has been obtained by studying the explanted scar tissues. In this study, we decellularized MI scar tissue and characterized the biomechanics of the obtained pure scar ECM. By thoroughly removing the cellular content in the MI scar tissue, we were able to avoid its confounding effects. Rat MI hearts were obtained from a reliable and reproducible model based on permanent left coronary artery ligation (PLCAL). MI heart explants at various time points (15 min, 1 week, 2 weeks, 4 weeks, and 12 weeks) were subjected to decellularization with 0.1% sodium dodecyl sulfate solution for ~1-2 weeks to obtain acellular scar ECM. A biaxial mechanical testing system was used to characterize the acellular scar ECM under physiologically relevant loading conditions. After decellularization, large decrease in wall thickness was observed in the native heart ECM and 15 min scar ECM, implying the collapse of cardiomyocyte lacunae after removal of heart muscle fibers. For scar ECM 1 week, 2 weeks, and 4 weeks post infarction, the decrease in wall thickness after decellularization was small. For scar ECM 12 weeks post infarction, the reduction amount of wall thickness due to decellularization was minimal. We found that the scar ECM preserved the overall mechanical anisotropy of the native ventricle wall and MI scar tissue, in which the longitudinal direction is more extensible. Acellular scar ECM from 15 min to 12 weeks post infarction showed an overall stiffening trend in biaxial behavior, in which longitudinal direction was mostly affected and manifested with a decreased extensibility and increased modulus. This reduction trend of longitudinal extensibility also led to a decreased anisotropy index in the scar ECM from the acute to chronic stages of MI. The post-MI change in biomechanical properties of the scar ECM reflected the alterations of collagen fiber network, confirmed by the histology of scar ECM. In short, the reported structure-property relationship reveals how scar ECM biophysical properties evolve from the acute to chronic stages of MI. The obtained information will help establish a knowledge basis about the dynamics of scar ECM to better understand post-MI cardiac remodeling.
Collapse
Affiliation(s)
- Bryn L Brazile
- Department of Biological Engineering and College of Veterinary Medicine, Mississippi State University, MS, 39762, USA
| | - J Ryan Butler
- Department of Biological Engineering and College of Veterinary Medicine, Mississippi State University, MS, 39762, USA
| | - Sourav S Patnaik
- Department of Biological Engineering and College of Veterinary Medicine, Mississippi State University, MS, 39762, USA
| | - Andrew Claude
- Department of Biological Engineering and College of Veterinary Medicine, Mississippi State University, MS, 39762, USA
| | - Raj Prabhu
- Department of Biological Engineering and College of Veterinary Medicine, Mississippi State University, MS, 39762, USA
| | - Lakiesha N Williams
- Department of Biological Engineering and College of Veterinary Medicine, Mississippi State University, MS, 39762, USA; Department of Biomedical Engineering, University of Florida, Gainesville, FL, 32611, USA
| | - Karla L Perez
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, 76019, USA
| | - Kytai T Nguyen
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, 76019, USA
| | - Ge Zhang
- Department of Biomedical Engineering, University of Akron, Akron, OH, 44325, USA
| | - Pietro Bajona
- Department of Cardiovascular and Thoracic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Matthias Peltz
- Department of Cardiovascular and Thoracic Surgery, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| | - Yong Yang
- Department of Biomedical Engineering, University of Northern Texas, Denton, TX, 76203, USA
| | - Yi Hong
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, 76019, USA
| | - Jun Liao
- Department of Bioengineering, University of Texas at Arlington, Arlington, TX, 76019, USA.
| |
Collapse
|
36
|
Digital Twins for Tissue Culture Techniques—Concepts, Expectations, and State of the Art. Processes (Basel) 2021. [DOI: 10.3390/pr9030447] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Techniques to provide in vitro tissue culture have undergone significant changes during the last decades, and current applications involve interactions of cells and organoids, three-dimensional cell co-cultures, and organ/body-on-chip tools. Efficient computer-aided and mathematical model-based methods are required for efficient and knowledge-driven characterization, optimization, and routine manufacturing of tissue culture systems. As an alternative to purely experimental-driven research, the usage of comprehensive mathematical models as a virtual in silico representation of the tissue culture, namely a digital twin, can be advantageous. Digital twins include the mechanistic of the biological system in the form of diverse mathematical models, which describe the interaction between tissue culture techniques and cell growth, metabolism, and the quality of the tissue. In this review, current concepts, expectations, and the state of the art of digital twins for tissue culture concepts will be highlighted. In general, DT’s can be applied along the full process chain and along the product life cycle. Due to the complexity, the focus of this review will be especially on the design, characterization, and operation of the tissue culture techniques.
Collapse
|
37
|
Rmaidi A, Zelzer M, Sindji L, Dima R, Boury F, Delorme N, Montero-Menei CN. Impact of the physico-chemical properties of polymeric microspheres functionalized with cell adhesion molecules on the behavior of mesenchymal stromal cells. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2021; 121:111852. [DOI: 10.1016/j.msec.2020.111852] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/01/2020] [Revised: 12/17/2020] [Accepted: 12/28/2020] [Indexed: 12/12/2022]
|
38
|
Pennarossa G, Arcuri S, De Iorio T, Gandolfi F, Brevini TAL. Current Advances in 3D Tissue and Organ Reconstruction. Int J Mol Sci 2021; 22:E830. [PMID: 33467648 PMCID: PMC7830719 DOI: 10.3390/ijms22020830] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2020] [Revised: 12/31/2020] [Accepted: 01/13/2021] [Indexed: 12/11/2022] Open
Abstract
Bi-dimensional culture systems have represented the most used method to study cell biology outside the body for over a century. Although they convey useful information, such systems may lose tissue-specific architecture, biomechanical effectors, and biochemical cues deriving from the native extracellular matrix, with significant alterations in several cellular functions and processes. Notably, the introduction of three-dimensional (3D) platforms that are able to re-create in vitro the structures of the native tissue, have overcome some of these issues, since they better mimic the in vivo milieu and reduce the gap between the cell culture ambient and the tissue environment. 3D culture systems are currently used in a broad range of studies, from cancer and stem cell biology, to drug testing and discovery. Here, we describe the mechanisms used by cells to perceive and respond to biomechanical cues and the main signaling pathways involved. We provide an overall perspective of the most recent 3D technologies. Given the breadth of the subject, we concentrate on the use of hydrogels, bioreactors, 3D printing and bioprinting, nanofiber-based scaffolds, and preparation of a decellularized bio-matrix. In addition, we report the possibility to combine the use of 3D cultures with functionalized nanoparticles to obtain highly predictive in vitro models for use in the nanomedicine field.
Collapse
Affiliation(s)
- Georgia Pennarossa
- Laboratory of Biomedical Embryology, Department of Health, Animal Science and Food Safety and Center for Stem Cell Research, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy; (G.P.); (S.A.); (T.D.I.)
| | - Sharon Arcuri
- Laboratory of Biomedical Embryology, Department of Health, Animal Science and Food Safety and Center for Stem Cell Research, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy; (G.P.); (S.A.); (T.D.I.)
| | - Teresina De Iorio
- Laboratory of Biomedical Embryology, Department of Health, Animal Science and Food Safety and Center for Stem Cell Research, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy; (G.P.); (S.A.); (T.D.I.)
| | - Fulvio Gandolfi
- Department of Agricultural and Environmental Sciences—Production, Landscape, Agroenergy and Center for Stem Cell Research, Università degli Studi di Milano, Via Celoria 2, 20133 Milan, Italy;
| | - Tiziana A. L. Brevini
- Laboratory of Biomedical Embryology, Department of Health, Animal Science and Food Safety and Center for Stem Cell Research, Università degli Studi di Milano, Via Celoria 10, 20133 Milan, Italy; (G.P.); (S.A.); (T.D.I.)
| |
Collapse
|
39
|
Modulation of Human Mesenchymal Stem Cells by Electrical Stimulation Using an Enzymatic Biofuel Cell. Catalysts 2021. [DOI: 10.3390/catal11010062] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Enzymatic biofuel cells (EBFCs) have excellent potential as components in bioelectronic devices, especially as active biointerfaces to regulate stem cell behavior for regenerative medicine applications. However, it remains unclear to what extent EBFC-generated electrical stimulation can regulate the functional behavior of human adipose-derived mesenchymal stem cells (hAD-MSCs) at the morphological and gene expression levels. Herein, we investigated the effect of EBFC-generated electrical stimulation on hAD-MSC cell morphology and gene expression using next-generation RNA sequencing. We tested three different electrical currents, 127 ± 9, 248 ± 15, and 598 ± 75 nA/cm2, in mesenchymal stem cells. We performed transcriptome profiling to analyze the impact of EBFC-derived electrical current on gene expression using next generation sequencing (NGS). We also observed changes in cytoskeleton arrangement and analyzed gene expression that depends on the electrical stimulation. The electrical stimulation of EBFC changes cell morphology through cytoskeleton re-arrangement. In particular, the results of whole transcriptome NGS showed that specific gene clusters were up- or down-regulated depending on the magnitude of applied electrical current of EBFC. In conclusion, this study demonstrates that EBFC-generated electrical stimulation can influence the morphological and gene expression properties of stem cells; such capabilities can be useful for regenerative medicine applications such as bioelectronic devices.
Collapse
|
40
|
James BD, Guerin P, Allen JB. Let's Talk About Sex-Biological Sex Is Underreported in Biomaterial Studies. Adv Healthc Mater 2021; 10:e2001034. [PMID: 33043626 PMCID: PMC7791002 DOI: 10.1002/adhm.202001034] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 08/26/2020] [Indexed: 12/13/2022]
Abstract
Precision medicine aims to better individualize healthcare. It requires that biomaterials be designed for the physiological characteristics of a specific patient. To make this a reality, biomaterials research and development must address differences of biological sex. More specifically, biomaterials should be designed with properties optimized and appropriate for male and female patients. In analyzing research articles from seven prominent biomaterials journals, sex as a biological variable is missing from an overwhelming majority of in vitro biomaterial studies. From the survey, the reporting of the sex of primary cell cultures happened only 10.3% of the time. Contributing to this trend is that commercial vendors bias cell lines toward one sex or another by not disclosing information of cell line sex at the time of purchase; researchers do not communicate this pertinent information in published studies; and many journal policies have little to no requirements for reporting cell line characteristics. Omitting this valuable information leads to a gap in the understanding of sex-specific cell-biomaterial interactions and it creates a bias in research findings towards one sex or another. To curb this concerning trend and make precision biomaterials a reality will require the biomaterials field to "talk about sex" by reporting cell sex more broadly.
Collapse
Affiliation(s)
- Bryan D James
- Department of Materials Science and Engineering, University of Florida, 206 Rhines Hall, PO Box 116400, Gainesville, FL, 32611-6400, USA
| | - Paxton Guerin
- Department of Materials Science and Engineering, University of Florida, 206 Rhines Hall, PO Box 116400, Gainesville, FL, 32611-6400, USA
| | - Josephine B Allen
- Department of Materials Science and Engineering, University of Florida, 206 Rhines Hall, PO Box 116400, Gainesville, FL, 32611-6400, USA
| |
Collapse
|
41
|
Barrett P, Quick TJ, Mudera V, Player DJ. Generating intrafusal skeletal muscle fibres in vitro: Current state of the art and future challenges. J Tissue Eng 2020; 11:2041731420985205. [PMID: 34956586 PMCID: PMC8693220 DOI: 10.1177/2041731420985205] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2020] [Accepted: 12/12/2020] [Indexed: 01/18/2023] Open
Abstract
Intrafusal fibres are a specialised cell population in skeletal muscle, found within the muscle spindle. These fibres have a mechano-sensory capacity, forming part of the monosynaptic stretch-reflex arc, a key component responsible for proprioceptive function. Impairment of proprioception and associated dysfunction of the muscle spindle is linked with many neuromuscular diseases. Research to-date has largely been undertaken in vivo or using ex vivo preparations. These studies have provided a foundation for our understanding of muscle spindle physiology, however, the cellular and molecular mechanisms which underpin physiological changes are yet to be fully elucidated. Therefrom, the use of in vitro models has been proposed, whereby intrafusal fibres can be generated de novo. Although there has been progress, it is predominantly a developing and evolving area of research. This narrative review presents the current state of art in this area and proposes the direction of future work, with the aim of providing novel pre-clinical and clinical applications.
Collapse
Affiliation(s)
- Philip Barrett
- Centre for 3D Models of Health and Disease, Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London, London, UK
| | - Tom J Quick
- Peripheral Nerve Injury Research Unit, Royal National Orthopaedic Hospital, Stanmore, UK
- UCL Centre for Nerve Engineering, University College London, London, UK
| | - Vivek Mudera
- Centre for 3D Models of Health and Disease, Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London, London, UK
| | - Darren J Player
- Centre for 3D Models of Health and Disease, Division of Surgery and Interventional Science, Faculty of Medical Sciences, University College London, London, UK
| |
Collapse
|
42
|
Cell-Laden Gelatin Methacryloyl Bioink for the Fabrication of Z-Stacked Hydrogel Scaffolds for Tissue Engineering. Polymers (Basel) 2020; 12:polym12123027. [PMID: 33348811 PMCID: PMC7767119 DOI: 10.3390/polym12123027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 12/12/2020] [Accepted: 12/15/2020] [Indexed: 11/27/2022] Open
Abstract
Hydrogel-based scaffolds have been widely used to fabricate artificial tissues capable of replacing tissues and organs. However, several challenges inherent in fabricating tissues of large size and complex morphology using such scaffolds while ensuring cell viability remain. To address this problem, we synthesized gelatin methacryloyl (GelMA) based bioink with cells for fabricating a scaffold with superior characteristics. The bioink was grafted onto a Z-stacking bioprinter that maintained the cells at physiological temperature during the printing process, without exerting any physical pressure on the cells. Various parameters, such as the bioink composition and light exposure time, were optimized. The printing accuracy of the scaffolds was evaluated using photorheological studies. The internal morphology of the scaffolds at different time points was analyzed using electron microscopy. The Z-stacked scaffolds were fabricated using high-speed printing, with the conditions optimized to achieve high model reproducibility. Stable adhesion and high proliferation of cells encapsulated within the scaffold were confirmed. We introduced various strategies to improve the accuracy and reproducibility of Z-stack GelMA bioprinting while ensuring that the scaffolds facilitated cell adhesion, encapsulation, and proliferation. Our results demonstrate the potential of the present method for various applications in tissue engineering.
Collapse
|
43
|
Nie J, Fu J, He Y. Hydrogels: The Next Generation Body Materials for Microfluidic Chips? SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2020; 16:e2003797. [PMID: 33103353 DOI: 10.1002/smll.202003797] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/20/2020] [Indexed: 05/27/2023]
Abstract
The integration of microfluidics with biomedical research is confronted with considerable limitations due to its body materials. With high content of water, hydrogels own superior biocompatibility and degradability. Can hydrogels become another material choice for the construction of microfluidic chips, particularly biofluidics? The present review aims to systematically establish the concept of hydrogel-based microfluidic chips (HMCs) and address three main concerns: i) why choosing hydrogels? ii) how to fabricate HMCs?, and iii) in which fields to apply HMCs? It is envisioned that hydrogels may be used increasingly as substitute for traditional materials and gradually act as the body material for microfluidic chips. The modifications of conventional process are highlighted to overcome issues arising from the incompatibility between the construction methods and hydrogel materials. Specifically targeting at the "soft and wet" hydrogels, an efficient flowchart of "i) high resolution template printing; ii) damage-free demolding; iii) twice-crosslinking bonding" is proposed. Accordingly, a broader microfluidic chip concept is proposed in terms of form and function. Potential biomedical applications of HMCs are discussed. This review also highlights the challenges arising from the material replacement, as well as the future directions of the proposed concept. Finally, the authors' viewpoints and perspectives for this emerging field are discussed.
Collapse
Affiliation(s)
- Jing Nie
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Jianzhong Fu
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of 3D Printing Process and Equipment of Zhejiang Province, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, China
- Key Laboratory of Materials Processing and Mold, Zhengzhou University, Zhengzhou, 450002, China
| |
Collapse
|
44
|
Sekuła-Stryjewska M, Noga S, Dźwigońska M, Adamczyk E, Karnas E, Jagiełło J, Szkaradek A, Chytrosz P, Boruczkowski D, Madeja Z, Kotarba A, Lipińska L, Zuba-Surma EK. Graphene-based materials enhance cardiomyogenic and angiogenic differentiation capacity of human mesenchymal stem cells in vitro - Focus on cardiac tissue regeneration. MATERIALS SCIENCE & ENGINEERING. C, MATERIALS FOR BIOLOGICAL APPLICATIONS 2020; 119:111614. [PMID: 33321657 DOI: 10.1016/j.msec.2020.111614] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Revised: 08/28/2020] [Accepted: 10/05/2020] [Indexed: 12/22/2022]
Abstract
Cell-based therapies have recently emerged as promising strategies for the treatment of cardiovascular disease. Mesenchymal stem cells (MSCs) are a promising cell type that represent a class of adult stem cells characterized by multipotency, high proliferative capacity, paracrine activity, and low immunogenicity. To improve the functional and therapeutic efficacy of MSCs, novel biomaterials are considered as scaffolds/surfaces that promote MSCs growth and differentiation. One of them are graphene-based materials, including graphene oxide (GO) and reduced graphene oxide (rGO). Due to the unique physical, chemical, and biological properties of graphene, scaffolds comprising GO/rGO have been examined as novel platforms to improve the differentiation potential of human MSCs in vitro. We verified different i) size of GO flakes, ii) reduction level, and iii) layer thickness to select the most suitable artificial niche for MSCs culture. The results revealed that graphene-based substrates constitute non-toxic substrates for MSCs. Surfaces with large flakes of GO as well as low reduced rGO are the most biocompatible for MSCs propagation and do not affect their proliferation and survival. Interestingly, small GO flakes and highly reduced rGO decreased MSCs proliferation and induced their apoptosis. We also found that GO and rGO substrates did not alter the MSCs phenotype, cell cycle progression and might modulate the adhesive capabilities of these cells. Importantly, we demonstrated that both materials promoted the cardiomyogenic and angiogenic differentiation capacity of MSCs in vitro. Thus, our data indicates that graphene-based surfaces represent promising materials that may influence the therapeutic application of MSCs via supporting their pro-regenerative potential.
Collapse
Affiliation(s)
- Małgorzata Sekuła-Stryjewska
- Laboratory of Stem Cell Biotechnology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland
| | - Sylwia Noga
- Laboratory of Stem Cell Biotechnology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland; Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Monika Dźwigońska
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Edyta Adamczyk
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Elżbieta Karnas
- Laboratory of Stem Cell Biotechnology, Malopolska Centre of Biotechnology, Jagiellonian University, Krakow, Poland; Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Joanna Jagiełło
- Department of Chemical Synthesis and Flake Graphene, Łukasiewicz Research Network - Institute of Electronic Materials Technology, Warsaw, Poland
| | - Agnieszka Szkaradek
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | | | | | - Zbigniew Madeja
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - Andrzej Kotarba
- Faculty of Chemistry, Jagiellonian University, Krakow, Poland
| | - Ludwika Lipińska
- Department of Chemical Synthesis and Flake Graphene, Łukasiewicz Research Network - Institute of Electronic Materials Technology, Warsaw, Poland
| | - Ewa K Zuba-Surma
- Department of Cell Biology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland.
| |
Collapse
|
45
|
Santini L, Palandri C, Nediani C, Cerbai E, Coppini R. Modelling genetic diseases for drug development: Hypertrophic cardiomyopathy. Pharmacol Res 2020; 160:105176. [DOI: 10.1016/j.phrs.2020.105176] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/22/2020] [Revised: 08/16/2020] [Accepted: 08/22/2020] [Indexed: 12/13/2022]
|
46
|
Dame K, Ribeiro AJ. Microengineered systems with iPSC-derived cardiac and hepatic cells to evaluate drug adverse effects. Exp Biol Med (Maywood) 2020; 246:317-331. [PMID: 32938227 PMCID: PMC7859673 DOI: 10.1177/1535370220959598] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Hepatic and cardiac drug adverse effects are among the leading causes of attrition in drug development programs, in part due to predictive failures of current animal or in vitro models. Hepatocytes and cardiomyocytes differentiated from human induced pluripotent stem cells (iPSCs) hold promise for predicting clinical drug effects, given their human-specific properties and their ability to harbor genetically determined characteristics that underlie inter-individual variations in drug response. Currently, the fetal-like properties and heterogeneity of hepatocytes and cardiomyocytes differentiated from iPSCs make them physiologically different from their counterparts isolated from primary tissues and limit their use for predicting clinical drug effects. To address this hurdle, there have been ongoing advances in differentiation and maturation protocols to improve the quality and use of iPSC-differentiated lineages. Among these are in vitro hepatic and cardiac cellular microsystems that can further enhance the physiology of cultured cells, can be used to better predict drug adverse effects, and investigate drug metabolism, pharmacokinetics, and pharmacodynamics to facilitate successful drug development. In this article, we discuss how cellular microsystems can establish microenvironments for these applications and propose how they could be used for potentially controlling the differentiation of hepatocytes or cardiomyocytes. The physiological relevance of cells is enhanced in cellular microsystems by simulating properties of tissue microenvironments, such as structural dimensionality, media flow, microfluidic control of media composition, and co-cultures with interacting cell types. Recent studies demonstrated that these properties also affect iPSC differentiations and we further elaborate on how they could control differentiation efficiency in microengineered devices. In summary, we describe recent advances in the field of cellular microsystems that can control the differentiation and maturation of hepatocytes and cardiomyocytes for drug evaluation. We also propose how future research with iPSCs within engineered microenvironments could enable their differentiation for scalable evaluations of drug effects.
Collapse
Affiliation(s)
- Keri Dame
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translation Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| | - Alexandre Js Ribeiro
- Division of Applied Regulatory Science, Office of Clinical Pharmacology, Office of Translation Sciences, Center for Drug Evaluation and Research, US Food and Drug Administration, Silver Spring, MD 20993, USA
| |
Collapse
|
47
|
Castro N, Ribeiro S, Fernandes MM, Ribeiro C, Cardoso V, Correia V, Minguez R, Lanceros‐Mendez S. Physically Active Bioreactors for Tissue Engineering Applications. ACTA ACUST UNITED AC 2020; 4:e2000125. [DOI: 10.1002/adbi.202000125] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 07/15/2020] [Indexed: 01/09/2023]
Affiliation(s)
- N. Castro
- BCMaterials, Basque Centre for Materials, Applications and Nanostructures University of the Basque Country UPV/EHU Science Park Leioa E‐48940 Spain
| | - S. Ribeiro
- Physics Centre University of Minho Campus de Gualtar Braga 4710‐057 Portugal
- Centre of Molecular and Environmental Biology (CBMA) University of Minho Campus de Gualtar Braga 4710‐057 Portugal
| | - M. M. Fernandes
- Physics Centre University of Minho Campus de Gualtar Braga 4710‐057 Portugal
- CEB – Centre of Biological Engineering University of Minho Braga 4710‐057 Portugal
| | - C. Ribeiro
- Physics Centre University of Minho Campus de Gualtar Braga 4710‐057 Portugal
- CEB – Centre of Biological Engineering University of Minho Braga 4710‐057 Portugal
| | - V. Cardoso
- CMEMS‐UMinho Universidade do Minho Campus de Azurém Guimarães 4800‐058 Portugal
| | - V. Correia
- Algoritmi Research Centre University of Minho Campus de Azurém Guimarães 4800‐058 Portugal
| | - R. Minguez
- Department of Graphic Design and Engineering Projects University of the Basque Country UPV/EHU Bilbao E‐48013 Spain
| | - S. Lanceros‐Mendez
- BCMaterials, Basque Centre for Materials, Applications and Nanostructures University of the Basque Country UPV/EHU Science Park Leioa E‐48940 Spain
- IKERBASQUE Basque Foundation for Science Bilbao E‐48013 Spain
| |
Collapse
|
48
|
Kang Y, Datta P, Shanmughapriya S, Ozbolat IT. 3D Bioprinting of Tumor Models for Cancer Research. ACS APPLIED BIO MATERIALS 2020; 3:5552-5573. [DOI: 10.1021/acsabm.0c00791] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- Youngnam Kang
- The Huck Institutes of the Life Sciences, Penn State University, University Park, Pennsylvania 16802, United States
- Engineering Science and Mechanics Department, Penn State University, University Park, Pennsylvania 16802, United States
| | - Pallab Datta
- Centre for Healthcare Science and Technology, Indian Institute of Engineering Science and Technology Shibpur, Howrah, West Bengal 711103, India
| | - Santhanam Shanmughapriya
- Department of Medicine, Penn State University, College of Medicine, Hershey, Pennsylvania 17033, United States
- Department of Cellular and Molecular Physiology, Penn State University, College of Medicine, Hershey, Pennsylvania 17033, United States
- Heart and Vascular Institute, Penn State University, College of Medicine, Hershey, Pennsylvania 17033, United States
| | - Ibrahim T. Ozbolat
- The Huck Institutes of the Life Sciences, Penn State University, University Park, Pennsylvania 16802, United States
- Engineering Science and Mechanics Department, Penn State University, University Park, Pennsylvania 16802, United States
- Biomedical Engineering Department, Penn State University, University Park, Pennsylvania 16802, United States
- Materials Research Institute, Penn State University, University Park, Pennsylvania 16802, United States
- Department of Neurosurgery, Penn State University, Hershey, Pennsylvania 17033, United States
| |
Collapse
|
49
|
Wechsler ME, Shevchuk M, Peppas NA. Developing a Multidisciplinary Approach for Engineering Stem Cell Organoids. Ann Biomed Eng 2020; 48:1895-1904. [PMID: 31659603 PMCID: PMC7186139 DOI: 10.1007/s10439-019-02391-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2019] [Accepted: 10/22/2019] [Indexed: 01/21/2023]
Abstract
Recent advances in stem cell biology, synthetic biology, bioengineering, and biotechnology have included significant work leading to the development of stem cell-derived organoids. The growing popularity of organoid research and use of organoids is widely due to the fact that these three-dimensional cellular structures better model human physiology compared to traditional in vitro and in vivo methods by recapitulating many biologically relevant parameters. Organoids show great promise for a wide range of applications, such as for use in disease modeling, drug discovery, and regenerative medicine. However, many challenges associated with reproducibility and scale up still remain. Identification of the conditions which generate a robust environment that predictably promotes cellular self-assembly and organization leading to organoid formation is critical and requires a multidisciplinary approach. To accomplish this we need to identify a cellular source, engineer a matrix to stimulate cell-cell and cell-matrix interactions, and provide the biochemical and biophysical cues which mimic that of the in vivo environment. Discussion of the components needed for organoid development and formation is reviewed herein, as well as specific organoid examples and the promise of this research for the future.
Collapse
Affiliation(s)
- Marissa E Wechsler
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
| | - Mariya Shevchuk
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA
| | - Nicholas A Peppas
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA.
- Institute for Biomaterials, Drug Delivery, and Regenerative Medicine, The University of Texas at Austin, Austin, TX, USA.
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, USA.
- Division of Molecular Pharmaceutics and Drug Delivery, College of Pharmacy, The University of Texas at Austin, Austin, TX, USA.
- Department of Surgery and Perioperative Care, and Department of Pediatrics, Dell Medical School, The University of Texas at Austin, Austin, TX, USA.
| |
Collapse
|
50
|
Aisenbrey EA, Murphy WL. Synthetic alternatives to Matrigel. NATURE REVIEWS. MATERIALS 2020; 5:539-551. [PMID: 32953138 PMCID: PMC7500703 DOI: 10.1038/s41578-020-0199-8] [Citation(s) in RCA: 531] [Impact Index Per Article: 106.2] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/31/2020] [Indexed: 05/19/2023]
Abstract
Matrigel, a basement-membrane matrix extracted from Engelbreth-Holm-Swarm mouse sarcomas, has been used for more than four decades for a myriad of cell culture applications. However, Matrigel is limited in its applicability to cellular biology, therapeutic cell manufacturing and drug discovery owing to its complex, ill-defined and variable composition. Variations in the mechanical and biochemical properties within a single batch of Matrigel - and between batches - have led to uncertainty in cell culture experiments and a lack of reproducibility. Moreover, Matrigel is not conducive to physical or biochemical manipulation, making it difficult to fine-tune the matrix to promote intended cell behaviours and achieve specific biological outcomes. Recent advances in synthetic scaffolds have led to the development of xenogenic-free, chemically defined, highly tunable and reproducible alternatives. In this Review, we assess the applications of Matrigel in cell culture, regenerative medicine and organoid assembly, detailing the limitations of Matrigel and highlighting synthetic scaffold alternatives that have shown equivalent or superior results. Additionally, we discuss the hurdles that are limiting a full transition from Matrigel to synthetic scaffolds and provide a brief perspective on the future directions of synthetic scaffolds for cell culture applications.
Collapse
Affiliation(s)
| | - William L. Murphy
- Department of Biomedical Engineering, University of Wisconsin–Madison, WI, USA
- Department of Orthopedics and Rehabilitation, University of Wisconsin–Madison, WI, USA
| |
Collapse
|