1
|
Song C, Yang J, Gu Z. Latest developments of microphysiological systems (MPS) in aging-related and geriatric diseases research: A review. Ageing Res Rev 2025; 107:102728. [PMID: 40058462 DOI: 10.1016/j.arr.2025.102728] [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: 12/02/2024] [Revised: 03/04/2025] [Accepted: 03/06/2025] [Indexed: 03/16/2025]
Abstract
Aging is a gradual and irreversible process accompanied by the decline in tissue function and a significantly increased risk of various aging-related and geriatric diseases. Especially in the paradoxical context of accelerated global aging and the widespread emergence of pandemics, aging-related and geriatric diseases have become leading causes of individual mortality and disability, drawing increasing attention from researchers and investors alike. Despite the utility of current in vitro systems and in vivo animal models for studying aging, these approaches are limited by insurmountable inherent constraints. In response, microphysiological systems (MPS), leveraging advances in tissue engineering and microfluidics, have emerged as highly promising platforms. MPS are capable of replicating key features of the tissue microenvironment within microfabricated devices, offering biomimetic tissue culture conditions that enhance the in vitro simulation of intact or precise human body structure and function. This capability improves the predictability of clinical trial outcomes while reducing time and cost. In this review, we focus on recent advancements in MPS used to study age-related and geriatric diseases, with particular emphasis on the application of organoids and organ-on-a-chip technologies in understanding cardiovascular diseases, cerebrovascular diseases, neurodegenerative diseases, fibrotic diseases, locomotor and sensory degenerative disorders, and rare diseases. And we aim to provide readers with critical guidelines and an overview of examples for modeling age-related and geriatric diseases using MPS, exploring mechanisms, treatments, drug screening, and other subsequent applications, from a physiopathological perspective, emphasizing the characteristic of age-related and geriatric diseases and their established correlations with the aging process. We also discuss the limitations of current models and propose future directions for MPS in aging research, highlighting the potential of interdisciplinary approaches to address unresolved challenges in the field.
Collapse
Affiliation(s)
- Chao Song
- State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, China; School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Jiachen Yang
- State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, China; School of Biological Science & Medical Engineering, Southeast University, Nanjing, China
| | - Zhongze Gu
- State Key Laboratory of Digital Medical Engineering, Southeast University, Nanjing, China; School of Biological Science & Medical Engineering, Southeast University, Nanjing, China.
| |
Collapse
|
2
|
Bessot A, Röhl J, Emmerich M, Klotz A, Ravichandran A, Meinert C, Waugh D, McGovern J, Gunter J, Bock N. ECM-mimicking hydrogel models of human adipose tissue identify deregulated lipid metabolism in the prostate cancer-adipocyte crosstalk under antiandrogen therapy. Mater Today Bio 2025; 30:101424. [PMID: 39866784 PMCID: PMC11764633 DOI: 10.1016/j.mtbio.2024.101424] [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: 09/09/2024] [Revised: 12/19/2024] [Accepted: 12/22/2024] [Indexed: 01/28/2025] Open
Abstract
Antiandrogen therapies are effectively used to treat advanced prostate cancer, but eventually cancer adaptation drives unresolved metastatic castration-resistant prostate cancer (mCRPC). Adipose tissue influences metabolic reprogramming in cancer and was proposed as a contributor to therapy resistance. Using extracellular matrix (ECM)-mimicking hydrogel coculture models of human adipocytes and prostate cancer cells, we show that adipocytes from subcutaneous or bone marrow fat have dissimilar responses under the antiandrogen Enzalutamide. We demonstrate that androgen receptor (AR)-dependent cancer cells (LNCaP) are more influenced by human adipocytes than AR-independent cells (C4-2B), with altered lipid metabolism and adipokine secretion. This response changes under Enzalutamide, with increased AR expression and adipogenic and lipogenic genes in cancer cells and decreased lipid content and gene dysregulation associated with insulin resistance in adipocytes. This is in line with the metabolic syndrome that men with mCRPC under Enzalutamide experience. The all-human, all-3D, models presented here provide a significant advance to dissect the role of fat in therapy response for mCRPC.
Collapse
Affiliation(s)
- Agathe Bessot
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia
- Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia
- Max Planck Queensland Centre, Brisbane, QLD, 4000, Australia
- Australian Prostate Cancer Research Centre (APCRC-Q), QUT, Brisbane, QLD, 4102, Australia
| | - Joan Röhl
- Faculty of Health Sciences and Medicine, Bond University, Robina, QLD, 4226, Australia
| | - Maria Emmerich
- School of Computation, Information and Technology, Technical University of Munich (TUM), Munich, Germany
| | - Anton Klotz
- Division of Gastroenterology and Hepatology, Department of Internal Medicine 3, Medical University of Vienna, Vienna, Austria
| | - Akhilandeshwari Ravichandran
- Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia
- School of Mechanical, Medical and Process Engineering, Faculty of Engineering, QUT, Brisbane, QLD 4000, Australia
- Australian Research Council (ARC) Training Centre for Cell and Tissue Engineering Technologies (CTET), QUT, Brisbane, QLD 4000, Australia
| | | | - David Waugh
- Centre for Cancer Biology, University of South Australia, Adelaide, SA 5005, Australia
| | - Jacqui McGovern
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia
- Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia
- Max Planck Queensland Centre, Brisbane, QLD, 4000, Australia
- Australian Research Council (ARC) Training Centre for Cell and Tissue Engineering Technologies (CTET), QUT, Brisbane, QLD 4000, Australia
| | - Jenni Gunter
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia
- Australian Prostate Cancer Research Centre (APCRC-Q), QUT, Brisbane, QLD, 4102, Australia
| | - Nathalie Bock
- School of Biomedical Sciences, Faculty of Health, and Translational Research Institute (TRI), Queensland University of Technology (QUT), Brisbane, QLD, 4102, Australia
- Centre for Biomedical Technologies, QUT, Brisbane, QLD, 4000, Australia
- Max Planck Queensland Centre, Brisbane, QLD, 4000, Australia
- Australian Prostate Cancer Research Centre (APCRC-Q), QUT, Brisbane, QLD, 4102, Australia
- Australian Research Council (ARC) Training Centre for Multiscale 3D Imaging, Modelling, and Manufacturing (M3D Innovation), Queensland University of Technology, Brisbane, QLD 4000, Australia
| |
Collapse
|
3
|
Chou LS, Zhang J, Jildeh TR. Metabolic Functions of the Infrapatellar Fat Pad: Implications for Knee Health and Pathology. JBJS Rev 2024; 12:01874474-202410000-00001. [PMID: 39361777 DOI: 10.2106/jbjs.rvw.24.00110] [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: 10/05/2024]
Abstract
» Despite being historically viewed as a vestigial structure, the infrapatellar fat pad (IPFP) is now recognized as a metabolically active structure, influencing knee health through cytokine production and metabolic pathways.» With distinct anatomical regions, the IPFP contains diverse cell types including adipocytes, fibroblasts, and immune cells, influencing its functional roles, pathology, and contributions to knee disorders.» The IPFP acts as an endocrine organ by releasing adipokines such as adiponectin, leptin, and tumor necrosis factor α, regulating energy balance, immune responses, and tissue remodelling, with implications for knee joint health.» The IPFP's metabolic interactions with neighboring tissues influence joint health, clinical conditions such as knee pain, osteoarthritis, postoperative complications, and ganglion cysts, highlighting its therapeutic potential and clinical relevance.» Understanding the multifaceted metabolic role of the IPFP opens avenues for collaborative approaches that integrate orthopaedics, endocrinology, and immunology to develop innovative therapeutic strategies targeting the intricate connections between adipokines, joint health, and immune responses.
Collapse
Affiliation(s)
- Lee S Chou
- Department of Orthopaedic Surgery, Michigan State University, East Lansing, Michigan
| | | | | |
Collapse
|
4
|
Yue S, Zhai G, Zhao S, Liang X, Liu Y, Zheng J, Chen X, Dong Y. The biphasic role of the infrapatellar fat pad in osteoarthritis. Biomed Pharmacother 2024; 179:117364. [PMID: 39226725 DOI: 10.1016/j.biopha.2024.117364] [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: 06/26/2024] [Revised: 08/23/2024] [Accepted: 08/26/2024] [Indexed: 09/05/2024] Open
Abstract
Osteoarthritis (OA) is a progressive degenerative disease resulting in joint deterioration. It is a whole organ disease characterized by cartilage degeneration and varying degrees of synovitis, involving pathological changes in all joint tissues, such as cartilage, subchondral bone, ligaments, meniscus, synovium, and infrapatellar fat pad (IPFP). IPFP is the largest adipose tissue structure in the knee joint and is composed of fat cells, immune cells and blood vessels. Moreover, IPFP is located close to the cartilage and bone surface so that it may reduce the impact of loading and absorb forces generated through the knee joint, and may have a protective role in joint health. IPFP has been shown to release various cytokines and adipokines that play pro-inflammatory and pro-catabolic roles in cartilage, promoting OA progression. Intra-articular injections of IPFP-derived mesenchymal stem cells and exosomes have been shown to reduce pain and prevent OA progression in patients with knee OA. Previous studies have shown that IPFP has a biphasic effect on OA progression. This article reviews the latest research progress of IPFP, discusses the role and mechanism of IPFP in OA, provide new intervention strategies for the treatment of OA. This article will also discuss the handling of IPFP during the procedure of total knee arthroplasty.
Collapse
Affiliation(s)
- Songkai Yue
- Department of Orthopedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University People's Hospital, Zhengzhou 450003, China
| | - Ganggang Zhai
- Department of Orthopedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University People's Hospital, Zhengzhou 450003, China
| | - Siyu Zhao
- Department of Orthopedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University People's Hospital, Zhengzhou 450003, China
| | - Xiaming Liang
- Department of Orthopedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University People's Hospital, Zhengzhou 450003, China
| | - Yunke Liu
- Department of Orthopedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University People's Hospital, Zhengzhou 450003, China
| | - Jia Zheng
- Department of Orthopedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University People's Hospital, Zhengzhou 450003, China
| | - Xiaoyang Chen
- Department of Orthopedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University People's Hospital, Zhengzhou 450003, China
| | - Yonghui Dong
- Department of Orthopedics, Henan Provincial People's Hospital, People's Hospital of Zhengzhou University, Henan University People's Hospital, Zhengzhou 450003, China.
| |
Collapse
|
5
|
Luca T, Pezzino S, Puleo S, Castorina S. Lesson on obesity and anatomy of adipose tissue: new models of study in the era of clinical and translational research. J Transl Med 2024; 22:764. [PMID: 39143643 PMCID: PMC11323604 DOI: 10.1186/s12967-024-05547-3] [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: 03/10/2024] [Accepted: 07/28/2024] [Indexed: 08/16/2024] Open
Abstract
Obesity is a serious global illness that is frequently associated with metabolic syndrome. Adipocytes are the typical cells of adipose organ, which is composed of at least two different tissues, white and brown adipose tissue. They functionally cooperate, interconverting each other under physiological conditions, but differ in their anatomy, physiology, and endocrine functions. Different cellular models have been proposed to study adipose tissue in vitro. They are also useful for elucidating the mechanisms that are responsible for a pathological condition, such as obesity, and for testing therapeutic strategies. Each cell model has its own characteristics, culture conditions, advantages and disadvantages. The choice of one model rather than another depends on the specific study the researcher is conducting. In recent decades, three-dimensional cultures, such as adipose spheroids, have become very attractive because they more closely resemble the phenotype of freshly isolated cells. The use of such models has developed in parallel with the evolution of translational research, an interdisciplinary branch of the biomedical field, which aims to learn a scientific translational approach to improve human health and longevity. The focus of the present review is on the growing body of data linking the use of new cell models and the spread of translational research. Also, we discuss the possibility, for the future, to employ new three-dimensional adipose tissue cell models to promote the transition from benchside to bedsite and vice versa, allowing translational research to become routine, with the final goal of obtaining clinical benefits in the prevention and treatment of obesity and related disorders.
Collapse
Affiliation(s)
- Tonia Luca
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Via Santa Sofia, 87, Catania, 95123, Italy.
| | | | - Stefano Puleo
- Mediterranean Foundation "GB Morgagni", Catania, Italy
| | - Sergio Castorina
- Department of Medical, Surgical Sciences and Advanced Technologies "G.F. Ingrassia", University of Catania, Via Santa Sofia, 87, Catania, 95123, Italy
- Mediterranean Foundation "GB Morgagni", Catania, Italy
| |
Collapse
|
6
|
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
|
7
|
Liu X, Yang J, Yan Y, Li Q, Huang RL. Unleashing the potential of adipose organoids: A revolutionary approach to combat obesity-related metabolic diseases. Theranostics 2024; 14:2075-2098. [PMID: 38505622 PMCID: PMC10945346 DOI: 10.7150/thno.93919] [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: 01/05/2024] [Accepted: 02/15/2024] [Indexed: 03/21/2024] Open
Abstract
Obesity-related metabolic diseases, including obesity, diabetes, hyperlipidemia, and non-alcoholic fatty liver diseases pose a significant threat to health. However, comprehensive pathogenesis exploration and effective therapy development are impeded by the limited availability of human models. Notably, advances in organoid technology enable the generation of adipose organoids that recapitulate structures and functions of native human adipose tissues to investigate mechanisms and develop corresponding treatments for obesity-related metabolic diseases. Here, we review the general principles, sources, and three-dimensional techniques for engineering adipose organoids, along with strategies to promote maturation. We also outline the application of white adipose organoids, primarily for disease modeling and drug screening, and highlight the therapeutic potential of thermogenic beige and brown adipose organoids in promoting weight loss and glucose and lipid metabolic homeostasis. We also discuss the challenges and prospects in the establishment and bench-to-bedside of adipose organoids, as well as their potential applications.
Collapse
Affiliation(s)
- Xingran Liu
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute for Plastic and Reconstructive Surgery, Shanghai, China
| | - Jing Yang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute for Plastic and Reconstructive Surgery, Shanghai, China
| | - Yuxin Yan
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute for Plastic and Reconstructive Surgery, Shanghai, China
| | - Qingfeng Li
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute for Plastic and Reconstructive Surgery, Shanghai, China
| | - Ru-Lin Huang
- Department of Plastic and Reconstructive Surgery, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- Shanghai Institute for Plastic and Reconstructive Surgery, Shanghai, China
| |
Collapse
|
8
|
Vasella M, Arnke K, Dranseikiene D, Guzzi E, Melega F, Reid G, Klein HJ, Schweizer R, Tibbitt MW, Kim BS. Methacrylated Gelatin as a Scaffold for Mechanically Isolated Stromal Vascular Fraction for Cutaneous Wound Repair. Int J Mol Sci 2023; 24:13944. [PMID: 37762247 PMCID: PMC10530931 DOI: 10.3390/ijms241813944] [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: 08/03/2023] [Revised: 09/05/2023] [Accepted: 09/08/2023] [Indexed: 09/29/2023] Open
Abstract
Mechanically processed stromal vascular fraction (mSVF) is a highly interesting cell source for regenerative purposes, including wound healing, and a practical alternative to enzymatically isolated SVF. In the clinical context, SVF benefits from scaffolds that facilitate viability and other cellular properties. In the present work, the feasibility of methacrylated gelatin (GelMA), a stiffness-tunable, light-inducible hydrogel with high biocompatibility is investigated as a scaffold for SVF in an in vitro setting. Lipoaspirates from elective surgical procedures were collected and processed to mSVF and mixed with GelMA precursor solutions. Non-encapsulated mSVF served as a control. Viability was measured over 21 days. Secreted basic fibroblast growth factor (bFGF) levels were measured on days 1, 7 and 21 by ELISA. IHC was performed to detect VEGF-A, perilipin-2, and CD73 expression on days 7 and 21. The impact of GelMA-mSVF on human dermal fibroblasts was measured in a co-culture assay by the same viability assay. The viability of cultured GelMA-mSVF was significantly higher after 21 days (p < 0.01) when compared to mSVF alone. Also, GelMA-mSVF secreted stable levels of bFGF over 21 days. While VEGF-A was primarily expressed on day 21, perilipin-2 and CD73-positive cells were observed on days 7 and 21. Finally, GelMA-mSVF significantly improved fibroblast viability as compared with GelMA alone (p < 0.01). GelMA may be a promising scaffold for mSVF as it maintains cell viability and proliferation with the release of growth factors while facilitating adipogenic differentiation, stromal cell marker expression and fibroblast proliferation.
Collapse
Affiliation(s)
- Mauro Vasella
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (M.V.); (G.R.)
| | - Kevin Arnke
- Center for Preclinical Development, University Hospital Zurich, 8091 Zurich, Switzerland;
| | - Dalia Dranseikiene
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, 8092 Zurich, Switzerland; (D.D.); (E.G.); (M.W.T.)
| | - Elia Guzzi
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, 8092 Zurich, Switzerland; (D.D.); (E.G.); (M.W.T.)
| | - Francesca Melega
- Institute of Pathology and Molecular Pathology, University Hospital Zurich, 8091 Zurich, Switzerland;
| | - Gregory Reid
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (M.V.); (G.R.)
| | - Holger Jan Klein
- Department of Plastic Surgery and Hand Surgery, Cantonal Hospital Aarau, 5001 Aarau, Switzerland;
| | - Riccardo Schweizer
- Department of Plastic, Reconstructive and Aesthetic Surgery, Regional Hospital Lugano, 6900 Lugano, Switzerland;
| | - Mark W. Tibbitt
- Macromolecular Engineering Laboratory, Department of Mechanical and Process Engineering, ETH Zurich, 8092 Zurich, Switzerland; (D.D.); (E.G.); (M.W.T.)
| | - Bong-Sung Kim
- Department of Plastic Surgery and Hand Surgery, University Hospital Zurich, 8091 Zurich, Switzerland; (M.V.); (G.R.)
| |
Collapse
|
9
|
Al-Ghadban S, Walczak SG, Isern SU, Martin EC, Herbst KL, Bunnell BA. Enhanced Angiogenesis in HUVECs Preconditioned with Media from Adipocytes Differentiated from Lipedema Adipose Stem Cells In Vitro. Int J Mol Sci 2023; 24:13572. [PMID: 37686378 PMCID: PMC10487727 DOI: 10.3390/ijms241713572] [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: 08/01/2023] [Revised: 08/25/2023] [Accepted: 08/31/2023] [Indexed: 09/10/2023] Open
Abstract
Lipedema is a connective tissue disorder characterized by increased dilated blood vessels (angiogenesis), inflammation, and fibrosis of the subcutaneous adipose tissue. This project aims to gain insights into the angiogenic processes in lipedema using human umbilical vein endothelial cells (HUVECs) as an in vitro model. HUVECs were cultured in conditioned media (CM) collected from healthy (non-lipedema, AQH) and lipedema adipocytes (AQL). The impacts on the expression levels of multiple endothelial and angiogenic markers [CD31, von Willebrand Factor (vWF), angiopoietin 2 (ANG2), hepatocyte growth factor (HGF), vascular endothelial growth factor (VEGF), matrix metalloproteinase (MMPs), NOTCH and its ligands] in HUVECs were investigated. The data demonstrate an increased expression of CD31 and ANG2 at both the gene and protein levels in HUVECs treated with AQL CM in 2D monolayer and 3D cultures compared to untreated cells. Furthermore, the expression of the vWF, NOTCH 4, and DELTA-4 genes decreased. In contrast, increased VEGF, MMP9, and HGF gene expression was detected in HUVECs treated with AQL CM cultured in a 2D monolayer. In addition, the results of a tube formation assay indicate that the number of formed tubes increased in lipedema-treated HUVECs cultured in a 2D monolayer. Together, the data indicate that lipedema adipocyte-CM promotes angiogenesis through paracrine-driven mechanisms.
Collapse
Affiliation(s)
- Sara Al-Ghadban
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (S.G.W.); (S.U.I.)
| | - Samantha G. Walczak
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (S.G.W.); (S.U.I.)
| | - Spencer U. Isern
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (S.G.W.); (S.U.I.)
| | - Elizabeth C. Martin
- Department of Medicine, Section of Hematology and Oncology, Tulane University, New Orleans, LA 70118, USA;
| | | | - Bruce A. Bunnell
- Department of Microbiology, Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA; (S.G.W.); (S.U.I.)
| |
Collapse
|
10
|
Bessot A, Gunter J, Waugh D, Clements JA, Hutmacher DW, McGovern J, Bock N. GelMA and Biomimetic Culture Allow the Engineering of Mineralized, Adipose, and Tumor Tissue Human Microenvironments for the Study of Advanced Prostate Cancer In Vitro and In Vivo. Adv Healthc Mater 2023; 12:e2201701. [PMID: 36708740 PMCID: PMC11469108 DOI: 10.1002/adhm.202201701] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2022] [Revised: 12/21/2022] [Indexed: 01/30/2023]
Abstract
Increasing evidence shows bone marrow (BM)-adipocytes as a potentially important contributor in prostate cancer (PCa) bone metastases. However, a lack of relevant models has prevented the full understanding of the effects of human BM-adipocytes in this microenvironment. It is hypothesized that the combination of tunable gelatin methacrylamide (GelMA)-based hydrogels with the biomimetic culture of human cells would offer a versatile 3D platform to engineer human bone tumor microenvironments containing BM-adipocytes. Human osteoprogenitors, adipocytes, and PCa cells are individually cultured in vitro in GelMA hydrogels, leading to mineralized, adipose, and PCa tumor 3D microtissues, respectively. Osteoblast mineralization and tumor spheroid formation are tailored by hydrogel stiffness with lower stiffnesses correlating with increased mineralization and tumor spheroid size. Upon coculture with tumor cells, BM-adipocytes undergo morphological changes and delipidation, suggesting reciprocal interactions between the cell types. When brought in vivo, the mineralized and adipose microtissues successfully form a humanized fatty bone microenvironment, presenting, for the first time, with human adipocytes. Using this model, an increase in tumor burden is observed when human adipocytes are present, suggesting that adipocytes support early bone tumor growth. The advanced platform presented here combines natural aspects of the microenvironment with tunable properties useful for bone tumor research.
Collapse
Affiliation(s)
- Agathe Bessot
- School of Biomedical SciencesFaculty of Health, and Translational Research Institute (TRI)Queensland University of Technology (QUT)BrisbaneQLD4102Australia
- Australian Prostate Cancer Research Centre ‐ Queensland (APCRC‐Q)QUTBrisbaneQLD4102Australia
- Centre for Biomedical TechnologiesQUTBrisbaneQLD4000Australia
- Max Planck Queensland CentreBrisbaneQLD4059Australia
| | - Jennifer Gunter
- School of Biomedical SciencesFaculty of Health, and Translational Research Institute (TRI)Queensland University of Technology (QUT)BrisbaneQLD4102Australia
- Australian Prostate Cancer Research Centre ‐ Queensland (APCRC‐Q)QUTBrisbaneQLD4102Australia
- Centre for Genomics and Personalised HealthQUTBrisbaneQLD4102Australia
| | - David Waugh
- School of Biomedical SciencesFaculty of Health, and Translational Research Institute (TRI)Queensland University of Technology (QUT)BrisbaneQLD4102Australia
| | - Judith A. Clements
- School of Biomedical SciencesFaculty of Health, and Translational Research Institute (TRI)Queensland University of Technology (QUT)BrisbaneQLD4102Australia
- Australian Prostate Cancer Research Centre ‐ Queensland (APCRC‐Q)QUTBrisbaneQLD4102Australia
| | - Dietmar W. Hutmacher
- School of MechanicalMedical and Process EngineeringEngineering FacultyQUTBrisbaneQLD4000Australia
- Max Planck Queensland CentreBrisbaneQLD4059Australia
| | - Jacqui McGovern
- School of Biomedical SciencesFaculty of Health, and Translational Research Institute (TRI)Queensland University of Technology (QUT)BrisbaneQLD4102Australia
- Centre for Biomedical TechnologiesQUTBrisbaneQLD4000Australia
- Max Planck Queensland CentreBrisbaneQLD4059Australia
| | - Nathalie Bock
- School of Biomedical SciencesFaculty of Health, and Translational Research Institute (TRI)Queensland University of Technology (QUT)BrisbaneQLD4102Australia
- Australian Prostate Cancer Research Centre ‐ Queensland (APCRC‐Q)QUTBrisbaneQLD4102Australia
- Centre for Biomedical TechnologiesQUTBrisbaneQLD4000Australia
- Max Planck Queensland CentreBrisbaneQLD4059Australia
| |
Collapse
|
11
|
Lee H, Hong HJ, Ahn S, Kim D, Kang SH, Cho K, Koh WG. One-Pot Synthesis of Double-Network PEG/Collagen Hydrogel for Enhanced Adipogenic Differentiation and Retrieval of Adipose-Derived Stem Cells. Polymers (Basel) 2023; 15:polym15071777. [PMID: 37050391 PMCID: PMC10098799 DOI: 10.3390/polym15071777] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 03/27/2023] [Accepted: 03/27/2023] [Indexed: 04/05/2023] Open
Abstract
Hydrogels are widely used in stem cell therapy due to their extensive tunability and resemblance to the extracellular matrix (ECM), which has a three-dimensional (3D) structure. These features enable various applications that enhance stem cell maintenance and function. However, fast and simple hydrogel fabrication methods are desirable for stem cells for efficient encapsulation and to reduce adverse effects on the cells. In this study, we present a one-pot double-crosslinked hydrogel consisting of polyethylene glycol (PEG) and collagen, which can be prepared without the multi-step sequential synthesis of each network, by using bio-orthogonal chemistry. To enhance the adipogenic differentiation efficiency of adipose-derived stem cells (ADSCs), we added degradable components within the hydrogel to regulate matrix stiffness through cell-mediated degradation. Bio-orthogonal reactions used for hydrogel gelation allow rapid gel formation for efficient cell encapsulation without toxic by-products. Furthermore, the hybrid network of synthetic (PEG) and natural (collagen) components demonstrated adequate mechanical strength and higher cell adhesiveness. Therefore, ADSCs grown within this hybrid hydrogel proliferated and functioned better than those grown in the single-crosslinked hydrogel. The degradable elements further improved adipogenesis in ADSCs with dynamic changes in modulus during culture and enabled the retrieval of differentiated cells for potential future applications.
Collapse
Affiliation(s)
- Hwajung Lee
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Hye Jin Hong
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Sujeong Ahn
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Dohyun Kim
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Shin Hyuk Kang
- Departments of Plastic and Reconstructive Surgery, Chung-Ang University Hospital, Chung-Ang University College of Medicine, Seoul 06973, Republic of Korea
| | - Kanghee Cho
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea
| | - Won-Gun Koh
- Department of Chemical and Biomolecular Engineering, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
12
|
Li ZA, Sant S, Cho SK, Goodman SB, Bunnell BA, Tuan RS, Gold MS, Lin H. Synovial joint-on-a-chip for modeling arthritis: progress, pitfalls, and potential. Trends Biotechnol 2023; 41:511-527. [PMID: 35995600 PMCID: PMC9938846 DOI: 10.1016/j.tibtech.2022.07.011] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2022] [Revised: 07/14/2022] [Accepted: 07/25/2022] [Indexed: 12/30/2022]
Abstract
Disorders of the synovial joint, such as osteoarthritis (OA) and rheumatoid arthritis (RA), afflict a substantial proportion of the global population. However, current clinical management has not been focused on fully restoring the native function of joints. Organ-on-chip (OoC), also called a microphysiological system, which typically accommodates multiple human cell-derived tissues/organs under physiological culture conditions, is an emerging platform that potentially overcomes the limitations of current models in developing therapeutics. Herein, we review major steps in the generation of OoCs for studying arthritis, discuss the challenges faced when these novel platforms enter the next phase of development and application, and present the potential for OoC technology to investigate the pathogenesis of joint diseases and the development of efficacious therapies.
Collapse
Affiliation(s)
- Zhong Alan Li
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Shilpa Sant
- Department of Pharmaceutical Sciences, University of Pittsburgh School of Pharmacy, Pittsburgh, PA 15261, USA; Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15260, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA
| | - Sung Kwon Cho
- Department of Mechanical Engineering and Materials Science, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15261, USA
| | - Stuart B Goodman
- Departments of Orthopaedic Surgery and Bioengineering, Stanford University, Stanford, CA 94305, USA
| | - Bruce A Bunnell
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, Fort Worth, TX 76107, USA
| | - Rocky S Tuan
- McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Hong Kong, SAR 999077, China
| | - Michael S Gold
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | - Hang Lin
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA; Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA 15260, USA; McGowan Institute for Regenerative Medicine, University of Pittsburgh School of Medicine, Pittsburgh, PA 15219, USA.
| |
Collapse
|
13
|
Wen J, Zhao Z, Fang F, Xiao J, Wang L, Cheng J, Wu J, Miao Y. Prussian Blue Nanoparticle-Entrapped GelMA Gels Laden with Mesenchymal Stem Cells as Prospective Biomaterials for Pelvic Floor Tissue Repair. Int J Mol Sci 2023; 24:ijms24032704. [PMID: 36769027 PMCID: PMC9916949 DOI: 10.3390/ijms24032704] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 01/13/2023] [Accepted: 01/26/2023] [Indexed: 02/04/2023] Open
Abstract
Pelvic organ prolapse (POP) seriously affects elderly patients' quality of life, and new repair materials are urgently needed. To solve this problem, we synthesized methacrylated gelatin (GelMA) hydrogels and incorporated photothermally active Prussian blue nanoparticles (PBNPs) to synthesize PBNP@GelMA. Then, MSCs were encapsulated in the PBNP@GelMA and exposed to a 1.0 W/cm2 of 808 nm laser for 10 min to perform heat shock pretreatment for the implantation of mesenchymal stem cells (MSCs). Next, we tested the repair efficacy of scaffold-cell complexes both in vitro and in vivo. Our results reveal that the heat shock treatment induced by PBNP@GelMA improved the viability of MSCs, and the underlying mechanism may be related to HSP70. Furthermore, 2 weeks after implantation in the SD rat model, the collagen content increased in the MSC implantation group and PBNP@GelMA implantation group. However, the muscle regeneration at the implanting position was mostly enhanced after the implantation of the heat-shock-pretreated MSCs, which illustrates that heat shock treatment can further promote the MSC-mediated muscle regeneration. Therefore, manipulating the cell environment and providing proper heat stimulus by using PBNP@GelMA with NIR is a novel strategy to enhance the regenerative potential of MSCs and to promote pelvic tissue repair.
Collapse
Affiliation(s)
- Jirui Wen
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Deep Underground Space Medical Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Zhiwei Zhao
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Fei Fang
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Jingyue Xiao
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, China
| | - Ling Wang
- Deep Underground Space Medical Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Juan Cheng
- Deep Underground Space Medical Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jiang Wu
- Deep Underground Space Medical Center, West China Hospital, Sichuan University, Chengdu 610041, China
- Correspondence: (J.W.); (Y.M.)
| | - Yali Miao
- Department of Obstetrics and Gynecology, Key Laboratory of Birth Defects and Related Diseases of Women and Children of MOE, West China Second University Hospital, Sichuan University, Chengdu 610041, China
- Correspondence: (J.W.); (Y.M.)
| |
Collapse
|
14
|
Alternative Methods as Tools for Obesity Research: In Vitro and In Silico Approaches. LIFE (BASEL, SWITZERLAND) 2022; 13:life13010108. [PMID: 36676057 PMCID: PMC9860640 DOI: 10.3390/life13010108] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 12/22/2022] [Accepted: 12/23/2022] [Indexed: 01/04/2023]
Abstract
The study of adipogenesis is essential for understanding and treating obesity, a multifactorial problem related to body fat accumulation that leads to several life-threatening diseases, becoming one of the most critical public health problems worldwide. In this review, we propose to provide the highlights of the adipogenesis study based on in vitro differentiation of human mesenchymal stem cells (hMSCs). We list in silico methods, such as molecular docking for identification of molecular targets, and in vitro approaches, from 2D, more straightforward and applied for screening large libraries of substances, to more representative physiological models, such as 3D and bioprinting models. We also describe the development of physiological models based on microfluidic systems applied to investigate adipogenesis in vitro. We intend to identify the main alternative models for adipogenesis evaluation, contributing to the direction of preclinical research in obesity. Future directions indicate the association of in silico and in vitro techniques to bring a clear picture of alternative methods based on adipogenesis as a tool for obesity research.
Collapse
|
15
|
Li Y, Liu D, Tan F, Yin W, Li Z. Umbilical cord derived mesenchymal stem cell-GelMA microspheres for accelerated wound healing. Biomed Mater 2022; 18. [PMID: 36541452 DOI: 10.1088/1748-605x/aca947] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2022] [Accepted: 12/06/2022] [Indexed: 12/12/2022]
Abstract
Mesenchymal stem cells (MSCs) are an ideal seed cell for tissue engineering and stem cell transplantation. MSCs combined with biological scaffolds play an important role in promoting the repair of cutaneous wound. However, direct administration of MSCs is challenging for MSCs survival and integration into tissues. Providing MSCs with a biocompatible scaffold can improve MSCs survival, but the effect of gelatin methacrylate (GelMA) loaded MSCs from umbilical cord MSCs (UC-MSCs) in wound healing remains unknown. Here, we investigated the ability of GelMA with UC-MSCs complexes to promote migration and proliferation and the effect on wound healing in mouse models. We discovered that UC-MSCs attached to GelMA and promoted the proliferation and migration of fibroblasts. Both UC-MSCs and UC-MSCs-derived extracellular vesicles accelerated wound healing. MSC + Gelatin methacrylate microspheres (GMs) application decreased expression of transforming growth factor-β(TGF-β) and Type III collagen (Col3)in vivo, leading to new collagen deposition and angiogenesis, and accelerate wound healing and skin tissue regeneration. Taken together, these findings indicate MSC + GMs can promote wound healing by regulating wound healing-related factors in the paracrine. Therefore, our research proves that GelMA is an ideal scaffold for the top management of UC-MSCs in wound healing medical practice.
Collapse
Affiliation(s)
- Yanqun Li
- Dongguan Enlife Stem Cell Biotechnology Institute, Zheshang Building, #430 Dongguan Ave., Dongguan, Guangdong 523000, People's Republic of China
| | - Dongyu Liu
- Dongguan Enlife Stem Cell Biotechnology Institute, Zheshang Building, #430 Dongguan Ave., Dongguan, Guangdong 523000, People's Republic of China
| | - Fengming Tan
- Dongguan Enlife Stem Cell Biotechnology Institute, Zheshang Building, #430 Dongguan Ave., Dongguan, Guangdong 523000, People's Republic of China
| | - Wenling Yin
- Dongguan Enlife Stem Cell Biotechnology Institute, Zheshang Building, #430 Dongguan Ave., Dongguan, Guangdong 523000, People's Republic of China
| | - Zhihuan Li
- Dongguan Enlife Stem Cell Biotechnology Institute, Zheshang Building, #430 Dongguan Ave., Dongguan, Guangdong 523000, People's Republic of China
| |
Collapse
|
16
|
Dashtarzheneh AK, Afrashtehpour A, Ramesh BS, Loizidou M. Harvestable tumour spheroids initiated in a gelatin-carboxymethyl cellulose hydrogel for cancer targeting and imaging with fluorescent gold nanoclusters. IN VITRO MODELS 2022; 1:437-446. [PMID: 39872615 PMCID: PMC11756458 DOI: 10.1007/s44164-022-00033-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/19/2022] [Revised: 08/11/2022] [Accepted: 08/18/2022] [Indexed: 01/30/2025]
Abstract
Cancer cell spheroids are the simplest 3D in vitro cancer models and have been extensively used for cancer research. More recently, models have been becoming complex, with the introduction of a matrix and non-cancer cell types to mimic specific tumour aspects. However, applying drugs or agents in matrix-embedded cancer spheroids can be problematic. Most matrices can impede and also bind drugs or visualizing agents non-specifically, in the vicinity of the embedded spheroids. This may interfere with imaging or further analysis without breaking apart the 3D model into its constituents. Here, we developed a combined gelatin-carboxymethyl cellulose (G-CMC) hydrogel for initiating cancer spheroids that enabled intact harvesting pre/post treatment for further investigation, such as targeting and imaging. We combined CMC (1.25%) and gelatin (2.5%) at 25 °C and initiated polymerisation after autoclaving (121 °C) to obtain a mechanical strength (sheer stress) of 38 Pas versus 1.28 Pas for CMC alone. These matrix conditions facilitated separation of the spheroids from the G-CMC, using low centrifugation (100 g). We described growth of colorectal and breast cancer spheroids within the G-CMC matrix (with average diameters of 220 mm and 180 μm for representative cell lines HT29 and MCF7 at 10 days, respectively). As the cancer cells express the surface biomarker calreticulin (CRT), we manufactured anti-calreticulin IgG (anti-CRT) conjugated to fluorescent gold nanoclusters (anti-CRT-AuNC) as a probe. We harvested cancer spheroids and incubated live with the nanoclusters. Imaging demonstrated strong binding of CRT-targeted AuNCs compared to control AuNCs. This novel model preserves cancer spheroid integrity upon isolation and is well suited for targeted imaging and drug delivery of cancer in 3D.
Collapse
Affiliation(s)
- Ashkan Kamali Dashtarzheneh
- Department of Surgical Biotechnology, Division of Surgery and Interventional Science, University College London, Royal Free Campus, Rowland Hill Street, London, UK
| | - Amir Afrashtehpour
- Department of Surgical Biotechnology, Division of Surgery and Interventional Science, University College London, Royal Free Campus, Rowland Hill Street, London, UK
| | - Bala Subramaniyam Ramesh
- Department of Surgical Biotechnology, Division of Surgery and Interventional Science, University College London, Royal Free Campus, Rowland Hill Street, London, UK
| | - Marilena Loizidou
- Department of Surgical Biotechnology, Division of Surgery and Interventional Science, University College London, Royal Free Campus, Rowland Hill Street, London, UK
| |
Collapse
|
17
|
Albrecht FB, Schmidt FF, Volz AC, Kluger PJ. Bioprinting of 3D Adipose Tissue Models Using a GelMA-Bioink with Human Mature Adipocytes or Human Adipose-Derived Stem Cells. Gels 2022; 8:gels8100611. [PMID: 36286112 PMCID: PMC9601941 DOI: 10.3390/gels8100611] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2022] [Revised: 09/21/2022] [Accepted: 09/21/2022] [Indexed: 12/31/2022] Open
Abstract
Adipose tissue is related to the development and manifestation of multiple diseases, demonstrating the importance of suitable in vitro models for research purposes. In this study, adipose tissue lobuli were explanted, cultured, and used as an adipose tissue control to evaluate in vitro generated adipose tissue models. During culture, lobule exhibited a stable weight, lactate dehydrogenase, and glycerol release over 15 days. For building up in vitro adipose tissue models, we adapted the biomaterial gelatin methacryloyl (GelMA) composition and handling to homogeneously mix and bioprint human primary mature adipocytes (MA) and adipose-derived stem cells (ASCs), respectively. Accelerated cooling of the bioink turned out to be essential for the homogeneous distribution of lipid-filled MAs in the hydrogel. Last, we compared manual and bioprinted GelMA hydrogels with MA or ASCs and the explanted lobules to evaluate the impact of the printing process and rate the models concerning the physiological reference. The viability analyses demonstrated no significant difference between the groups due to additive manufacturing. The staining of intracellular lipids and perilipin A suggest that GelMA is well suited for ASCs and MA. Therefore, we successfully constructed physiological in vitro models by bioprinting MA-containing GelMA bioinks.
Collapse
Affiliation(s)
- Franziska B. Albrecht
- Reutlingen Research Institute, Reutlingen University, 72762 Reutlingen, Germany
- Faculty of Natural Science, University of Hohenheim, 70599 Stuttgart, Germany
| | - Freia F. Schmidt
- Reutlingen Research Institute, Reutlingen University, 72762 Reutlingen, Germany
| | - Ann-Cathrin Volz
- Reutlingen Research Institute, Reutlingen University, 72762 Reutlingen, Germany
| | - Petra J. Kluger
- Faculty of Applied Chemistry, Reutlingen University, 72762 Reutlingen, Germany
- Correspondence: ; Tel.: +49-7121-271-2061
| |
Collapse
|
18
|
Sugii S, Wong CYQ, Lwin AKO, Chew LJM. Reassessment of adipocyte technology for cellular agriculture of alternative fat. Compr Rev Food Sci Food Saf 2022; 21:4146-4163. [PMID: 36018497 DOI: 10.1111/1541-4337.13021] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2022] [Revised: 06/24/2022] [Accepted: 07/18/2022] [Indexed: 01/28/2023]
Abstract
Alternative proteins, such as cultivated meat, have recently attracted significant attention as novel and sustainable food. Fat tissue/cell is an important component of meat that makes organoleptic and nutritional contributions. Although adipocyte biology is relatively well investigated, there is limited focus on the specific techniques and strategies to produce cultivated fat from agricultural animals. In the assumed standard workflow, stem/progenitor cell lines are derived from tissues of animals, cultured for expansion, and differentiated into mature adipocytes. Here, we compile information from literature related to cell isolation, growth, differentiation, and analysis from bovine, porcine, chicken, other livestock, and seafood species. A diverse range of tissue sources, cell isolation methods, cell types, growth media, differentiation cocktails, and analytical methods for measuring adipogenic levels were used across species. Based on our analysis, we identify opportunities and challenges in advancing new technology era toward producing "alternative fat" that is suitable for human consumption.
Collapse
Affiliation(s)
- Shigeki Sugii
- Bioengineering Systems Division, Institute of Bioengineering and Bioimaging (IBB), A*STAR, Singapore.,Program of Cardiovascular and Metabolic Disorders, Duke-NUS Medical School, Singapore
| | - Cheryl Yeh Qi Wong
- Bioengineering Systems Division, Institute of Bioengineering and Bioimaging (IBB), A*STAR, Singapore
| | - Angela Khin Oo Lwin
- Bioengineering Systems Division, Institute of Bioengineering and Bioimaging (IBB), A*STAR, Singapore
| | - Lamony Jian Ming Chew
- Bioengineering Systems Division, Institute of Bioengineering and Bioimaging (IBB), A*STAR, Singapore
| |
Collapse
|
19
|
Aberrant Expression of COX-2 and FOXG1 in Infrapatellar Fat Pad-Derived ASCs from Pre-Diabetic Donors. Cells 2022; 11:cells11152367. [PMID: 35954211 PMCID: PMC9367583 DOI: 10.3390/cells11152367] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2022] [Revised: 07/25/2022] [Accepted: 07/26/2022] [Indexed: 12/10/2022] Open
Abstract
Osteoarthritis (OA) is a degenerative joint disease resulting in limited mobility and severe disability. Type II diabetes mellitus (T2D) is a weight-independent risk factor for OA, but a link between the two diseases has not been elucidated. Adipose stem cells (ASCs) isolated from the infrapatellar fat pad (IPFP) may be a viable regenerative cell for OA treatment. This study analyzed the expression profiles of inflammatory and adipokine-related genes in IPFP-ASCs of non-diabetic (Non-T2D), pre-diabetic (Pre-T2D), and T2D donors. Pre-T2D ASCs exhibited a substantial decrease in levels of mesenchymal markers CD90 and CD105 with no change in adipogenic differentiation compared to Non-T2D and T2D IPFP-ASCs. In addition, Cyclooxygenase-2 (COX-2), Forkhead box G1 (FOXG1) expression and prostaglandin E2 (PGE2) secretion were significantly increased in Pre-T2D IPFP-ASCs upon stimulation by interleukin-1 beta (IL-1β). Interestingly, M1 macrophages exhibited a significant reduction in expression of pro-inflammatory markers TNFα and IL-6 when co-cultured with Pre-T2D IPFP-ASCs. These data suggest that the heightened systemic inflammation associated with untreated T2D may prime the IPFP-ASCs to exhibit enhanced anti-inflammatory characteristics via suppressing the IL-6/COX-2 signaling pathway. In addition, the elevated production of PGE2 by the Pre-T2D IPFP-ASCs may also suggest the contribution of pre-diabetic conditions to the onset and progression of OA.
Collapse
|
20
|
Knežić T, Janjušević L, Djisalov M, Yodmuang S, Gadjanski I. Using Vertebrate Stem and Progenitor Cells for Cellular Agriculture, State-of-the-Art, Challenges, and Future Perspectives. Biomolecules 2022; 12:699. [PMID: 35625626 PMCID: PMC9138761 DOI: 10.3390/biom12050699] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2022] [Revised: 05/01/2022] [Accepted: 05/07/2022] [Indexed: 12/19/2022] Open
Abstract
Global food systems are under significant pressure to provide enough food, particularly protein-rich foods whose demand is on the rise in times of crisis and inflation, as presently existing due to post-COVID-19 pandemic effects and ongoing conflict in Ukraine and resulting in looming food insecurity, according to FAO. Cultivated meat (CM) and cultivated seafood (CS) are protein-rich alternatives for traditional meat and fish that are obtained via cellular agriculture (CA) i.e., tissue engineering for food applications. Stem and progenitor cells are the building blocks and starting point for any CA bioprocess. This review presents CA-relevant vertebrate cell types and procedures needed for their myogenic and adipogenic differentiation since muscle and fat tissue are the primary target tissues for CM/CS production. The review also describes existing challenges, such as a need for immortalized cell lines, or physical and biochemical parameters needed for enhanced meat/fat culture efficiency and ways to address them.
Collapse
Affiliation(s)
- Teodora Knežić
- Center for Biosystems, BioSense Institute, University of Novi Sad, Dr. Zorana Djindjica 1, 21000 Novi Sad, Serbia; (T.K.); (L.J.); (M.D.)
| | - Ljiljana Janjušević
- Center for Biosystems, BioSense Institute, University of Novi Sad, Dr. Zorana Djindjica 1, 21000 Novi Sad, Serbia; (T.K.); (L.J.); (M.D.)
| | - Mila Djisalov
- Center for Biosystems, BioSense Institute, University of Novi Sad, Dr. Zorana Djindjica 1, 21000 Novi Sad, Serbia; (T.K.); (L.J.); (M.D.)
| | - Supansa Yodmuang
- Research Affairs, Faculty of Medicine, Chulalongkorn University, 1873 Rama 4 Rd, Pathumwan, Bangkok 10330, Thailand;
| | - Ivana Gadjanski
- Center for Biosystems, BioSense Institute, University of Novi Sad, Dr. Zorana Djindjica 1, 21000 Novi Sad, Serbia; (T.K.); (L.J.); (M.D.)
| |
Collapse
|
21
|
A Comparative Study on the Adipogenic Differentiation of Mesenchymal Stem/Stromal Cells in 2D and 3D Culture. Cells 2022; 11:cells11081313. [PMID: 35455993 PMCID: PMC9029885 DOI: 10.3390/cells11081313] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/07/2022] [Accepted: 04/08/2022] [Indexed: 02/04/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSC) are capable of renewing the progenitor cell fraction or differentiating in a tissue-specific manner. Adipogenic differentiation of adipose-tissue-derived MSC (adMSC) is important in various pathological processes. Adipocytes and their progenitors are metabolically active and secrete molecules (adipokines) that have both pro- and anti-inflammatory properties. Cell culturing in 2D is commonly used to study cellular responses, but the 2D environment does not reflect the structural situation for most cell types. Therefore, 3D culture systems have been developed to create an environment considered more physiological. Since knowledge about the effects of 3D cultivation on adipogenic differentiation is limited, we investigated its effects on adipogenic differentiation and adipokine release of adMSC (up to 28 days) and compared these with the effects in 2D. We demonstrated that cultivation conditions are crucial for cell behavior: in both 2D and 3D culture, adipogenic differentiation occurred only after specific stimulation. While the size and structure of adipogenically stimulated 3D spheroids remained stable during the experiment, the unstimulated spheroids showed signs of disintegration. Adipokine release was dependent on culture dimensionality; we found upregulated adiponectin and downregulated pro-inflammatory factors. Our findings are relevant for cell therapeutic applications of adMSC in complex, three-dimensionally arranged tissues.
Collapse
|
22
|
BASKAN OZNUR, OZCIVICI ENGIN. VIABILITY OF 3T3-L1 PREADIPOCYTES IS MODULATED BY THE APPLIED FREQUENCY BUT NOT THE EXPOSURE DURATION OF LOW INTENSITY VIBRATORY STIMULATION. J MECH MED BIOL 2022. [DOI: 10.1142/s0219519422500063] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Mechanical forces are the integral determinants in cell and tissue homeostasis and regeneration, and they can affect numerous biological process from proliferation to fate determination. Mechanical forces that possess low magnitude and high frequency characteristics are also known as low intensity vibrations (LIVs). These signals were studied widely on many cell types for regenerative purposes, however most of these studies select components of LIV signals (e.g., magnitude, frequency, duration, etc.) arbitrarily. Here, we addressed the effect of LIV applied frequency, LIV daily exposure time and fate induction on the viability of preadipocyte 3T3-L1 cells. For this, we performed a frequency sweep that was ranging from 30[Formula: see text]Hz to 120[Formula: see text]Hz with 15[Formula: see text]Hz increments applied for 5, 10 or 20[Formula: see text]min during quiescent growth or adipogenesis for up to 10 days. Results suggest that the applied frequency and fate induction was an important determinant of cell viability while daily exposure time had no effect. These findings contribute to the effort of optimizing a relevant mechanical stimulus that can inhibit adipogenesis.
Collapse
Affiliation(s)
- OZNUR BASKAN
- Department of Bioengineering, Izmir Institute of Technology Urla, Izmir 35430, Turkey
| | - ENGIN OZCIVICI
- Department of Bioengineering, Izmir Institute of Technology Urla, Izmir 35430, Turkey
| |
Collapse
|
23
|
Additive-Free Gelatine-Based Devices for Chondral Tissue Regeneration: Shaping Process Comparison among Mould Casting and Three-Dimensional Printing. Polymers (Basel) 2022; 14:polym14051036. [PMID: 35267859 PMCID: PMC8915043 DOI: 10.3390/polym14051036] [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: 12/10/2021] [Revised: 02/12/2022] [Accepted: 03/02/2022] [Indexed: 02/07/2023] Open
Abstract
Gelatine is a well-known and extensively studied biopolymer, widely used in recent decades to create biomaterials in many different ways, exploiting its molecular resemblance with collagen, the main constituent of the extra-cellular matrix, from which it is derived. Many have employed this biopolymer in tissue engineering and chemically modified (e.g., gelatin methacryloyl) or blended it with other polymers (e.g., alginate) to modulate or increase its performances and printability. Nevertheless, little is reported about its use as a stand-alone material. Moreover, despite the fact that multiple works have been reported on the realization of mould-casted and three-dimensional printed scaffolds in tissue engineering, a clear comparison among these two shaping processes, towards a comparable workflow starting from the same material, has never been published. Herein, we report the use of gelatine as stand-alone material, not modified, blended, or admixed to be processed or crosslinked, for the realization of suitable scaffolds for tissue engineering, towards the two previously mentioned shaping processes. To make the comparison reliable, the same pre-process (e.g., the gelatin solution preparation) and post-process (e.g., freeze-drying and crosslinking) steps were applied. In this study, gelatine solution was firstly rheologically characterized to find a formulation suitable for being processed with both the shaping processes selected. The realized scaffolds were then morphologically, phisico-chemically, mechanically, and biologically characterized to determine and compare their performances. Despite the fact that the same starting material was employed, as well as the same pre- and post-process steps, the two groups resulted, for most aspects, in diametrically opposed characteristics. The mould-casted scaffolds that resulted were characterized by small, little-interconnected, and random porosity, high resistance to compression and slow cell colonization, while the three-dimensional printed scaffolds displayed big, well-interconnected, and geometrically defined porosity, high elasticity and recover ability after compression, as well as fast and deep cell colonization.
Collapse
|
24
|
Al-Ghadban S, Artiles M, Bunnell BA. Adipose Stem Cells in Regenerative Medicine: Looking Forward. Front Bioeng Biotechnol 2022; 9:837464. [PMID: 35096804 PMCID: PMC8792599 DOI: 10.3389/fbioe.2021.837464] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Accepted: 12/27/2021] [Indexed: 12/16/2022] Open
Abstract
Over the last decade, stem cell-based regenerative medicine has progressed to clinical testing and therapeutic applications. The applications range from infusions of autologous and allogeneic stem cells to stem cell-derived products. Adult stem cells from adipose tissue (ASCs) show significant promise in treating autoimmune and neurodegenerative diseases, vascular and metabolic diseases, bone and cartilage regeneration and wound defects. The regenerative capabilities of ASCs in vivo are primarily orchestrated by their secretome of paracrine factors and cell-matrix interactions. More recent developments are focused on creating more complex structures such as 3D organoids, tissue elements and eventually fully functional tissues and organs to replace or repair diseased or damaged tissues. The current and future applications for ASCs in regenerative medicine are discussed here.
Collapse
Affiliation(s)
| | | | - Bruce A. Bunnell
- Department of Microbiology Immunology and Genetics, University of North Texas Health Science Center, Fort Worth, TX, United States
| |
Collapse
|
25
|
Widera D. Recent Advances in Translational Adipose-Derived Stem Cell Biology. Biomolecules 2021; 11:biom11111660. [PMID: 34827658 PMCID: PMC8615724 DOI: 10.3390/biom11111660] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2021] [Accepted: 11/05/2021] [Indexed: 12/30/2022] Open
Affiliation(s)
- Darius Widera
- Stem Cell Biology and Regenerative Medicine Group, School of Pharmacy, University of Reading, Reading RG6 6UB, UK
| |
Collapse
|
26
|
Xiang S, Li Z, Fritch MR, Li L, Velankar S, Liu Y, Sohn J, Baker N, Lin H, Tuan RS. Caveolin-1 mediates soft scaffold-enhanced adipogenesis of human mesenchymal stem cells. Stem Cell Res Ther 2021; 12:347. [PMID: 34127047 PMCID: PMC8201886 DOI: 10.1186/s13287-021-02356-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2021] [Accepted: 04/26/2021] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Human bone marrow-derived mesenchymal stem cells (hBMSCs) can differentiate into adipocytes upon stimulation and are considered an appropriate cell source for adipose tissue engineering. In addition to biochemical cues, the stiffness of a substrate that cells attach to has also been shown to affect hBMSC differentiation potential. Of note, most current studies are conducted on monolayer cultures which do not directly inform adipose tissue engineering, where 3-dimensional (3D) scaffolds are often used to create proper tissue architecture. In this study, we aim to examine the adipogenic differentiation of hBMSCs within soft or stiff scaffolds and investigate the molecular mechanism mediating the response of hBMSCs to substrate stiffness in 3D culture, specifically the involvement of the integral membrane protein, caveolin-1 (CAV1), known to regulate signaling in MSCs via compartmentalizing and concentrating signaling molecules. METHODS By adjusting the photo-illumination time, photocrosslinkable gelatin scaffolds with the same polymer concentration but different stiffnesses were created. hBMSCs were seeded within soft and stiff scaffolds, and their response to adipogenic induction under different substrate mechanical conditions was characterized. The functional involvement of CAV1 was assessed by suppressing its expression level using CAV1-specific siRNA. RESULTS The soft and stiff scaffolds used in this study had a compressive modulus of ~0.5 kPa and ~23.5 kPa, respectively. hBMSCs showed high viability in both scaffold types, but only spread out in the soft scaffolds. hBMSCs cultured in soft scaffolds displayed significantly higher adipogenesis, as revealed by histology, qRT-PCR, and immunostaining. Interestingly, a lower CAV1 level was observed in hBMSCs in the soft scaffolds, concomitantly accompanied by increased levels of Yes-associated protein (YAP) and decreased YAP phosphorylation, when compared to cells seeded in the stiff scaffolds. Interestingly, reducing CAV1 expression with siRNA was shown to further enhance hBMSC adipogenesis, which may function through activation of the YAP signaling pathway. CONCLUSIONS Soft biomaterials support superior adipogenesis of encapsulated hBMSCs in 3D culture, which is partially mediated by the CAV1-YAP axis. Suppressing CAV1 expression levels represents a robust method in the promotion of hBMSC adipogenesis.
Collapse
Affiliation(s)
- Shiqi Xiang
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Department of Orthopaedics, The Second Xiangya Hospital of Central South University, Changsha, Hunan, China
| | - Zhong Li
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Madalyn R Fritch
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - La Li
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sachin Velankar
- Department of Chem/Petroleum Engineering and Mechanical Engineering & Materials Science, University of Pittsburgh Swanson School of Engineering, Pittsburgh, Pennsylvania, USA
| | - Yuwei Liu
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Jihee Sohn
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Present Address: Biogen, Boston, Massachusetts, USA
| | - Natasha Baker
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.,Present Address: Department of Oral Biology, University of Pittsburgh School of Dental Medicine, Pittsburgh, Pennsylvania, USA
| | - Hang Lin
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA. .,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA. .,Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, Pennsylvania, USA.
| | - Rocky S Tuan
- Center for Cellular and Molecular Engineering, Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA. .,McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, Pennsylvania, USA. .,Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, Pennsylvania, USA. .,Present Address: Institute for Tissue Engineering and Regenerative Medicine, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China.
| |
Collapse
|
27
|
Gibler P, Gimble J, Hamel K, Rogers E, Henderson M, Wu X, Olesky S, Frazier T. Human Adipose-Derived Stromal/Stem Cell Culture and Analysis Methods for Adipose Tissue Modeling In Vitro: A Systematic Review. Cells 2021; 10:1378. [PMID: 34204869 PMCID: PMC8227575 DOI: 10.3390/cells10061378] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2021] [Revised: 05/25/2021] [Accepted: 05/26/2021] [Indexed: 12/11/2022] Open
Abstract
Human adipose-derived stromal/stem cells (hASC) are widely used for in vitro modeling of physiologically relevant human adipose tissue. These models are useful for the development of tissue constructs for soft tissue regeneration and 3-dimensional (3D) microphysiological systems (MPS) for drug discovery. In this systematic review, we report on the current state of hASC culture and assessment methods for adipose tissue engineering using 3D MPS. Our search efforts resulted in the identification of 184 independent records, of which 27 were determined to be most relevant to the goals of the present review. Our results demonstrate a lack of consensus on methods for hASC culture and assessment for the production of physiologically relevant in vitro models of human adipose tissue. Few studies have assessed the impact of different 3D culture conditions on hASC adipogenesis. Additionally, there has been a limited use of assays for characterizing the functionality of adipose tissue in vitro. Results from this study suggest the need for more standardized culture methods and further analysis on in vitro tissue functionality. These will be necessary to validate the utility of 3D MPS as an in vitro model to reduce, refine, and replace in vivo experiments in the drug discovery regulatory process.
Collapse
Affiliation(s)
- Peyton Gibler
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Jeffrey Gimble
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
- Department of Structural and Cell Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Department of Medicine, Tulane University School of Medicine, New Orleans, LA 70112, USA
- Department of Surgery, Tulane University School of Medicine, New Orleans, LA 70112, USA
| | - Katie Hamel
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Emma Rogers
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Michael Henderson
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Xiying Wu
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Spencer Olesky
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
| | - Trivia Frazier
- Obatala Sciences Inc., New Orleans, LA 70148, USA; (P.G.); (K.H.); (E.R.); (M.H.); (X.W.); (S.O.); (T.F.)
- Department of Structural and Cell Biology, Tulane University School of Medicine, New Orleans, LA 70112, USA
| |
Collapse
|
28
|
Frazier T, Williams C, Henderson M, Duplessis T, Rogers E, Wu X, Hamel K, Martin EC, Mohiuddin O, Shaik S, Devireddy R, Rowan BG, Hayes DJ, Gimble JM. Breast Cancer Reconstruction: Design Criteria for a Humanized Microphysiological System. Tissue Eng Part A 2021; 27:479-488. [PMID: 33528293 PMCID: PMC8196546 DOI: 10.1089/ten.tea.2020.0372] [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: 12/12/2020] [Accepted: 01/21/2021] [Indexed: 11/12/2022] Open
Abstract
International regulatory agencies such as the Food and Drug Administration have mandated that the scientific community develop humanized microphysiological systems (MPS) as an in vitro alternative to animal models in the near future. While the breast cancer research community has long appreciated the importance of three-dimensional growth dynamics in their experimental models, there are remaining obstacles preventing a full conversion to humanized MPS for drug discovery and pathophysiological studies. This perspective evaluates the current status of human tissue-derived cells and scaffolds as building blocks for an "idealized" breast cancer MPS based on bioengineering design principles. It considers the utility of adipose tissue as a potential source of endothelial, lymphohematopoietic, and stromal cells for the support of breast cancer epithelial cells. The relative merits of potential MPS scaffolds derived from adipose tissue, blood components, and synthetic biomaterials is evaluated relative to the current "gold standard" material, Matrigel, a murine chondrosarcoma-derived basement membrane-enriched hydrogel. The advantages and limitations of a humanized breast cancer MPS are discussed in the context of in-process and destructive read-out assays. Impact statement Regulatory authorities have highlighted microphysiological systems as an emerging tool in breast cancer research. This has been led by calls for more predictive human models and reduced animal experimentation. This perspective describes how human-derived cells, extracellular matrices, and hydrogels will provide the building blocks to create breast cancer models that accurately reflect diversity at multiple levels, that is, patient ethnicity, pathophysiology, and metabolic status.
Collapse
Affiliation(s)
| | - Christopher Williams
- Division of Basic Pharmaceutical Sciences, Xavier University of Louisiana, New Orleans, Louisiana, USA
| | | | - Tamika Duplessis
- Department of Physical Sciences, Delgado Community College, New Orleans, Louisiana, USA
| | - Emma Rogers
- Obatala Sciences, Inc., New Orleans, Louisiana, USA
| | - Xiying Wu
- Obatala Sciences, Inc., New Orleans, Louisiana, USA
| | - Katie Hamel
- Obatala Sciences, Inc., New Orleans, Louisiana, USA
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Elizabeth C. Martin
- Department of Biological and Agricultural Engineering, Louisiana State University, Baton Rouge, Louisiana, USA
| | - Omair Mohiuddin
- Panjwani Center for Molecular Medicine and Drug Research, International Center for Chemical and Biological Science, University of Karachi, Karachi, Pakistan
| | - Shahensha Shaik
- Cell and Molecular Biology Core Laboratory, Xavier University of Louisiana, New Orleans, Louisiana, USA
| | - Ram Devireddy
- Department of Mechanical Engineering, Louisiana State University, New Orleans, Louisiana, USA
| | - Brian G. Rowan
- Department of Structural and Cellular Biology, Tulane University School of Medicine, New Orleans, Louisiana, USA
| | - Daniel J. Hayes
- Department of Biomedical Engineering, Pennsylvania State University, State College, Pennsylvania, USA
| | | |
Collapse
|
29
|
3D Spheroids Derived from Human Lipedema ASCs Demonstrated Similar Adipogenic Differentiation Potential and ECM Remodeling to Non-Lipedema ASCs In Vitro. Int J Mol Sci 2020; 21:ijms21218350. [PMID: 33171717 PMCID: PMC7664323 DOI: 10.3390/ijms21218350] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2020] [Revised: 11/01/2020] [Accepted: 11/05/2020] [Indexed: 02/08/2023] Open
Abstract
The growth and differentiation of adipose tissue-derived stem cells (ASCs) is stimulated and regulated by the adipose tissue (AT) microenvironment. In lipedema, both inflammation and hypoxia influence the expansion and differentiation of ASCs, resulting in hypertrophic adipocytes and deposition of collagen, a primary component of the extracellular matrix (ECM). The goal of this study was to characterize the adipogenic differentiation potential and assess the levels of expression of ECM-remodeling markers in 3D spheroids derived from ASCs isolated from both lipedema and healthy individuals. The data showed an increase in the expression of the adipogenic genes (ADIPOQ, LPL, PPAR-γ and Glut4), a decrease in matrix metalloproteinases (MMP2, 9 and 11), with no significant changes in the expression of ECM markers (collagen and fibronectin), or integrin A5 in 3D differentiated lipedema spheroids as compared to healthy spheroids. In addition, no statistically significant changes in the levels of expression of inflammatory genes were detected in any of the samples. However, immunofluorescence staining showed a decrease in fibronectin and increase in laminin and Collagen VI expression in the 3D differentiated spheroids in both groups. The use of 3D ASC spheroids provide a functional model to study the cellular and molecular characteristics of lipedema AT.
Collapse
|