1
|
Evangelista A, Scocozza F, Conti M, Auricchio F, Conti B, Dorati R, Genta I, Benazzo M, Pisani S. Exploring Mechanical Features of 3D Head and Neck Cancer Models. J Funct Biomater 2025; 16:74. [PMID: 40137353 PMCID: PMC11942903 DOI: 10.3390/jfb16030074] [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: 01/28/2025] [Revised: 02/14/2025] [Accepted: 02/17/2025] [Indexed: 03/27/2025] Open
Abstract
Head and neck squamous cell carcinoma (HNSCC) presents significant challenges in oncology due to its complex biology and poor prognosis. Traditional two-dimensional (2D) cell culture models cannot replicate the intricate tumor microenvironment, limiting their usefulness in studying disease mechanisms and testing therapies. In contrast, three-dimensional (3D) in vitro models provide more realistic platforms that better mimic the architecture, mechanical features, and cellular interactions of HNSCC. This review explores the mechanical properties of 3D in vitro models developed for HNSCC research. It highlights key 3D culture techniques, such as spheroids, organoids, and bioprinted tissues, emphasizing their ability to simulate critical tumor characteristics like hypoxia, drug resistance, and metastasis. Particular attention is given to stiffness, elasticity, and dynamic behavior, highlighting how these models emulate native tumor tissues. By enhancing the physiological relevance of in vitro studies, 3D models offer significant potential to revolutionize HNSCC research and facilitate the development of effective, personalized therapeutic strategies. This review bridges the gap between preclinical and clinical applications by summarizing the mechanical properties of 3D models and providing guidance for developing systems that replicate both biological and mechanical characteristics of tumor tissues, advancing innovation in cancer research and therapy.
Collapse
Affiliation(s)
- Aleksandra Evangelista
- Department of Otorhinolaryngology, Fondazione IRCCS Policlinico San Matteo, Via Golgi 19, 27100 Pavia, Italy; (A.E.); (M.B.)
| | - Franca Scocozza
- Department of Civil Engineering and Architecture, University of Pavia, Via Ferrata 3, 27100 Pavia, Italy; (M.C.); (F.A.)
| | - Michele Conti
- Department of Civil Engineering and Architecture, University of Pavia, Via Ferrata 3, 27100 Pavia, Italy; (M.C.); (F.A.)
- 3D and Computer Simulation Laboratory, IRCCS Policlinico San Donato, Piazza Edmondo Malan 2, San Donato Milanese, 20097 Milano, Italy
| | - Ferdinando Auricchio
- Department of Civil Engineering and Architecture, University of Pavia, Via Ferrata 3, 27100 Pavia, Italy; (M.C.); (F.A.)
| | - Bice Conti
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy; (B.C.); (R.D.); (I.G.); (S.P.)
| | - Rossella Dorati
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy; (B.C.); (R.D.); (I.G.); (S.P.)
| | - Ida Genta
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy; (B.C.); (R.D.); (I.G.); (S.P.)
| | - Marco Benazzo
- Department of Otorhinolaryngology, Fondazione IRCCS Policlinico San Matteo, Via Golgi 19, 27100 Pavia, Italy; (A.E.); (M.B.)
| | - Silvia Pisani
- Department of Drug Sciences, University of Pavia, Via Taramelli 12, 27100 Pavia, Italy; (B.C.); (R.D.); (I.G.); (S.P.)
| |
Collapse
|
2
|
Roy NS, Kumari M, Alam K, Bhattacharya A, Kaity S, Kaur K, Ravichandiran V, Roy S. Development of bioengineered 3D patient derived breast cancer organoid model focusing dynamic fibroblast-stem cell reciprocity. PROGRESS IN BIOMEDICAL ENGINEERING (BRISTOL, ENGLAND) 2024; 7:012007. [PMID: 39662055 DOI: 10.1088/2516-1091/ad9dcb] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 12/11/2024] [Indexed: 12/13/2024]
Abstract
Three-dimensional (3D) models, such as tumor spheroids and organoids, are increasingly developed by integrating tissue engineering, regenerative medicine, and personalized therapy strategies. These advanced 3Din-vitromodels are not merely endpoint-driven but also offer the flexibility to be customized or modulated according to specific disease parameters. Unlike traditional 2D monolayer cultures, which inadequately capture the complexities of solid tumors, 3D co-culture systems provide a more accurate representation of the tumor microenvironment. This includes critical interactions with mesenchymal stem/stromal cells (MSCs) and induced pluripotent stem cells (iPSCs), which significantly modulate cancer cell behavior and therapeutic responses. Most of the findings from the co-culture of Michigan Cancer Foundation-7 breast cancer cells and MSC showed the formation of monolayers. Although changes in the plasticity of MSCs and iPSCs caused by other cells and extracellular matrix (ECM) have been extensively researched, the effect of MSCs on cancer stem cell (CSC) aggressiveness is still controversial and contradictory among different research communities. Some researchers have argued that CSCs proliferate more, while others have proposed that cancer spread occurs through dormancy. This highlights the need for further investigation into how these interactions shape cancer aggressiveness. The objective of this review is to explore changes in cancer cell behavior within a 3D microenvironment enriched with MSCs, iPSCs, and ECM components. By describing various MSC and iPSC-derived 3D breast cancer models that replicate tumor biology, we aim to elucidate potential therapeutic targets for breast cancer. A particular focus of this review is the Transwell system, which facilitates understanding how MSCs and iPSCs affect critical processes such as migration, invasion, and angiogenesis. The gradient formed between the two chambers is based on diffusion, as seen in the human body. Once optimized, this Transwell model can serve as a high-throughput screening platform for evaluating various anticancer agents. In the future, primary cell-based and patient-derived 3D organoid models hold promise for advancing personalized medicine and accelerating drug development processes.
Collapse
Affiliation(s)
- Nakka Sharmila Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054 West Bengal, India
| | - Mamta Kumari
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054 West Bengal, India
| | - Kamare Alam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054 West Bengal, India
| | - Anamitra Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054 West Bengal, India
| | - Santanu Kaity
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054 West Bengal, India
| | - Kulwinder Kaur
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine a Health Sciences, Dublin, Ireland
- Department of Anatomy & Regenerative Medicine, Tissue Engineering Research Group, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Velayutham Ravichandiran
- Department of Natural Products, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054 West Bengal, India
| | - Subhadeep Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054 West Bengal, India
| |
Collapse
|
3
|
Ye Z, Sun L, Xiang Q, Hao Y, Liu H, He Q, Yang X, Liao W. Advancements of Biomacromolecular Hydrogel Applications in Food Nutrition and Health. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:23689-23708. [PMID: 39410660 DOI: 10.1021/acs.jafc.4c05903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2024]
Abstract
Hydrogels exhibit remarkable degradability, biocompatibility and functionality, which position them as highly promising materials for applications within the food and pharmaceutical industries. Although many relevant studies on hydrogels have been reported in the chemical industry, materials, and other fields, there have been few reviews on their potential applications in food nutrition and human health. This study aims to address this gap by reviewing the functional properties of hydrogels and assessing their value in terms of food nutrition and human health. The use of hydrogels in preserving bioactive ingredients, food packaging and food distribution is delved into specifically in this review. Hydrogels can serve as cutting-edge materials for food packaging and delivery, ensuring the preservation of nutritional activity within food products, facilitating targeted delivery of bioactive compounds and regulating the digestion and absorption processes in the human body, thereby promoting human health. Moreover, hydrogels find applications in in vitro cell and tissue culture, human tissue repair, as well as chronic disease prevention and treatment. These broad applications have attracted great attention in the fields of human food nutrition and health. Ultimately, this paper serves as a valuable reference for further utilization and exploration of hydrogels in these respective fields.
Collapse
Affiliation(s)
- Zichong Ye
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Linye Sun
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Qianru Xiang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Yuting Hao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Hongji Liu
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Qi He
- Food Safety and Health Research Center, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, P. R. China
| | - Xingfen Yang
- Food Safety and Health Research Center, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, P. R. China
| | - Wenzhen Liao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| |
Collapse
|
4
|
Major G, Ahn M, Cho WW, Santos M, Wise J, Phillips E, Wise SG, Jang J, Rnjak-Kovacina J, Woodfield T, Lim KS. Programming temporal stiffness cues within extracellular matrix hydrogels for modelling cancer niches. Mater Today Bio 2024; 25:101004. [PMID: 38420142 PMCID: PMC10900776 DOI: 10.1016/j.mtbio.2024.101004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 02/13/2024] [Accepted: 02/15/2024] [Indexed: 03/02/2024] Open
Abstract
Extracellular matrix (ECM) stiffening is a common occurrence during the progression of many diseases, such as breast cancer. To accurately mimic the pathophysiological context of disease within 3D in vitro models, there is high demand for smart biomaterials which replicate the dynamic and temporal mechanical cues of diseased states. This study describes a preclinical disease model, using breast cancer as an example, which replicates the dynamic plasticity of the tumour microenvironment by incorporating temporal (3-week progression) biomechanical cues within a tissue-specific hydrogel microenvironment. The composite hydrogel formulation, integrating adipose-derived decellularised ECM (AdECM) and silk fibroin, was initially crosslinked using a visible light-mediated system, and then progressively stiffened through spontaneous secondary structure interactions inherent between the polymer chains (∼10-15 kPa increase, with a final stiffness of 25 kPa). When encapsulated and cultured in vitro, MCF-7 breast cancer cells initially formed numerous, large spheroids (>1000 μm2 in area), however, with progressive temporal stiffening, cells demonstrated growth arrest and underwent phenotypic changes resulting in intratumoral heterogeneity. Unlike widely-investigated static mechanical models, this stiffening hydrogel allowed for progressive phenotypic changes to be observed, and fostered the development of mature organoid-like spheroids, which mimicked both the organisation and acinar-structures of mature breast epithelium. The spheroids contained a central population of cells which expressed aggressive cellular programs, evidenced by increased fibronectin expression and reduction of E-cadherin. The phenotypic heterogeneity observed using this model is more reflective of physiological tumours, demonstrating the importance of establishing temporal cues within preclinical models in future work. Overall, the developed model demonstrated a novel strategy to uncouple ECM biomechanical properties from the cellular complexities of the disease microenvironment and offers the potential for wide applicability in other 3D in vitro disease models through addition of tissue-specific dECM materials.
Collapse
Affiliation(s)
- Gretel Major
- Department of Orthopaedic Surgery and Musculoskeletal Medicine, Centre for Bioengineering & Nanomedicine, University of Otago, Christchurch, New Zealand
| | - Minjun Ahn
- Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Won-Woo Cho
- Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Miguel Santos
- Applied Materials Group, School of Medical Sciences, University of Sydney, Sydney, Australia
| | - Jessika Wise
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Elisabeth Phillips
- Mackenzie Cancer Research Group, Department of Pathology and Biomedical Science, University of Otago, Christchurch, New Zealand
| | - Steven G Wise
- Applied Materials Group, School of Medical Sciences, University of Sydney, Sydney, Australia
| | - Jinah Jang
- Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Jelena Rnjak-Kovacina
- Graduate School of Biomedical Engineering, University of New South Wales, Sydney, Australia
- Tyree Institute of Health Engineering, University of New South Wales, Sydney, NSW, 2052, Australia
| | - Tim Woodfield
- Department of Orthopaedic Surgery and Musculoskeletal Medicine, Centre for Bioengineering & Nanomedicine, University of Otago, Christchurch, New Zealand
| | - Khoon S Lim
- Department of Orthopaedic Surgery and Musculoskeletal Medicine, Centre for Bioengineering & Nanomedicine, University of Otago, Christchurch, New Zealand
- Light-Activated Materials Group, School of Medical Sciences, University of Sydney, Australia
| |
Collapse
|
5
|
Suryavanshi P, Bodas D. Knockout cancer by nano-delivered immunotherapy using perfusion-aided scaffold-based tumor-on-a-chip. Nanotheranostics 2024; 8:380-400. [PMID: 38751938 PMCID: PMC11093718 DOI: 10.7150/ntno.87818] [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: 12/01/2023] [Accepted: 01/20/2024] [Indexed: 05/18/2024] Open
Abstract
Cancer is a multifactorial disease produced by mutations in the oncogenes and tumor suppressor genes, which result in uncontrolled cell proliferation and resistance to cell death. Cancer progresses due to the escape of altered cells from immune monitoring, which is facilitated by the tumor's mutual interaction with its microenvironment. Understanding the mechanisms involved in immune surveillance evasion and the significance of the tumor microenvironment might thus aid in developing improved therapies. Although in vivo models are commonly utilized, they could be better for time, cost, and ethical concerns. As a result, it is critical to replicate an in vivo model and recreate the cellular and tissue-level functionalities. A 3D cell culture, which gives a 3D architecture similar to that found in vivo, is an appropriate model. Furthermore, numerous cell types can be cocultured, establishing cellular interactions between TME and tumor cells. Moreover, microfluidics perfusion can provide precision flow rates, thus simulating tissue/organ function. Immunotherapy can be used with the perfused 3D cell culture technique to help develop successful therapeutics. Immunotherapy employing nano delivery can target the spot and silence the responsible genes, ensuring treatment effectiveness while minimizing adverse effects. This study focuses on the importance of 3D cell culture in understanding the pathophysiology of 3D tumors and TME, the function of TME in drug resistance, tumor progression, and the development of advanced anticancer therapies for high-throughput drug screening.
Collapse
Affiliation(s)
- Pooja Suryavanshi
- Nanobioscience Group, Agharkar Research Institute, G.G. Agarkar Road, Pune 411 004 India
- Savitribai Phule Pune University, Ganeshkhind Road, Pune 411 007 India
| | - Dhananjay Bodas
- Nanobioscience Group, Agharkar Research Institute, G.G. Agarkar Road, Pune 411 004 India
- Savitribai Phule Pune University, Ganeshkhind Road, Pune 411 007 India
| |
Collapse
|
6
|
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
|
7
|
Jackson CE, Doyle I, Khan H, Williams SF, Aldemir Dikici B, Barajas Ledesma E, Bryant HE, English WR, Green NH, Claeyssens F. Gelatin-containing porous polycaprolactone PolyHIPEs as substrates for 3D breast cancer cell culture and vascular infiltration. Front Bioeng Biotechnol 2024; 11:1321197. [PMID: 38260750 PMCID: PMC10800367 DOI: 10.3389/fbioe.2023.1321197] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2023] [Accepted: 12/13/2023] [Indexed: 01/24/2024] Open
Abstract
Tumour survival and growth are reliant on angiogenesis, the formation of new blood vessels, to facilitate nutrient and waste exchange and, importantly, provide a route for metastasis from a primary to a secondary site. Whilst current models can ensure the transport and exchange of nutrients and waste via diffusion over distances greater than 200 μm, many lack sufficient vasculature capable of recapitulating the tumour microenvironment and, thus, metastasis. In this study, we utilise gelatin-containing polymerised high internal phase emulsion (polyHIPE) templated polycaprolactone-methacrylate (PCL-M) scaffolds to fabricate a composite material to support the 3D culture of MDA-MB-231 breast cancer cells and vascular ingrowth. Firstly, we investigated the effect of gelatin within the scaffolds on the mechanical and chemical properties using compression testing and FTIR spectroscopy, respectively. Initial in vitro assessment of cell metabolic activity and vascular endothelial growth factor expression demonstrated that gelatin-containing PCL-M polyHIPEs are capable of supporting 3D breast cancer cell growth. We then utilised the chick chorioallantoic membrane (CAM) assay to assess the angiogenic potential of cell-seeded gelatin-containing PCL-M polyHIPEs, and vascular ingrowth within cell-seeded, surfactant and gelatin-containing scaffolds was investigated via histological staining. Overall, our study proposes a promising composite material to fabricate a substrate to support the 3D culture of cancer cells and vascular ingrowth.
Collapse
Affiliation(s)
- Caitlin E. Jackson
- The Kroto Research Institute, Materials Science and Engineering, University of Sheffield, Sheffield, United Kingdom
- Insigneo Institute for in Silico Medicine, The Pam Liversidge Building, University of Sheffield, Sheffield, United Kingdom
| | - Iona Doyle
- The Kroto Research Institute, Materials Science and Engineering, University of Sheffield, Sheffield, United Kingdom
| | - Hamood Khan
- The Kroto Research Institute, Materials Science and Engineering, University of Sheffield, Sheffield, United Kingdom
| | - Samuel F. Williams
- Department of Infection, Immunity and Cardiovascular Disease, Royal Hallamshire Hospital, The University of Sheffield, Sheffield, United Kingdom
| | | | | | - Helen E. Bryant
- School of Medicine and Population Health, University of Sheffield, Sheffield, United Kingdom
| | - William R. English
- Norwich Medical School, University of East Anglia, Norwich, United Kingdom
| | - Nicola H. Green
- The Kroto Research Institute, Materials Science and Engineering, University of Sheffield, Sheffield, United Kingdom
- Insigneo Institute for in Silico Medicine, The Pam Liversidge Building, University of Sheffield, Sheffield, United Kingdom
| | - Frederik Claeyssens
- The Kroto Research Institute, Materials Science and Engineering, University of Sheffield, Sheffield, United Kingdom
- Insigneo Institute for in Silico Medicine, The Pam Liversidge Building, University of Sheffield, Sheffield, United Kingdom
| |
Collapse
|
8
|
Bhattacharya A, Alam K, Roy NS, Kaur K, Kaity S, Ravichandiran V, Roy S. Exploring the interaction between extracellular matrix components in a 3D organoid disease model to replicate the pathophysiology of breast cancer. J Exp Clin Cancer Res 2023; 42:343. [PMID: 38102637 PMCID: PMC10724947 DOI: 10.1186/s13046-023-02926-4] [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: 10/20/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023] Open
Abstract
In vitro models are necessary to study the pathophysiology of the disease and the development of effective, tailored treatment methods owing to the complexity and heterogeneity of breast cancer and the large population affected by it. The cellular connections and tumor microenvironments observed in vivo are often not recapitulated in conventional two-dimensional (2D) cell cultures. Therefore, developing 3D in vitro models that mimic the complex architecture and physiological circumstances of breast tumors is crucial for advancing our understanding of the illness. A 3D scaffold-free in vitro disease model mimics breast cancer pathophysiology by allowing cells to self-assemble/pattern into 3D structures, in contrast with other 3D models that rely on artificial scaffolds. It is possible that this model, whether applied to breast tumors using patient-derived primary cells (fibroblasts, endothelial cells, and cancer cells), can accurately replicate the observed heterogeneity. The complicated interactions between different cell types are modelled by integrating critical components of the tumor microenvironment, such as the extracellular matrix, vascular endothelial cells, and tumor growth factors. Tissue interactions, immune cell infiltration, and the effects of the milieu on drug resistance can be studied using this scaffold-free 3D model. The scaffold-free 3D in vitro disease model for mimicking tumor pathophysiology in breast cancer is a useful tool for studying the molecular basis of the disease, identifying new therapeutic targets, and evaluating treatment modalities. It provides a more physiologically appropriate high-throughput platform for screening large compound library in a 96-384 well format. We critically discussed the rapid development of personalized treatment strategies and accelerated drug screening platforms to close the gap between traditional 2D cell culture and in vivo investigations.
Collapse
Affiliation(s)
- Anamitra Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Kamare Alam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Nakka Sharmila Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Kulwinder Kaur
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine a Health Sciences, Dublin, Ireland
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Santanu Kaity
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Velayutham Ravichandiran
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Subhadeep Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India.
| |
Collapse
|
9
|
Tofani LB, Luiz MT, Paes Dutra JA, Abriata JP, Chorilli M. Three-dimensional culture models: emerging platforms for screening the antitumoral efficacy of nanomedicines. Nanomedicine (Lond) 2023; 18:633-647. [PMID: 37183804 DOI: 10.2217/nnm-2022-0205] [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] [Indexed: 05/16/2023] Open
Abstract
Nanomedicines have been investigated for delivering drugs to tumors due to their ability to accumulate in the tumor tissues. 2D in vitro cell culture has been used to investigate the antitumoral potential of nanomedicines. However, a 2D model cannot adequately mimic the in vivo tissue conditions because of the lack of cell-cell interaction, a gradient of nutrients and the expression of genes. To overcome this limitation, 3D cell culture models have emerged as promising platforms that better replicate the complexity of native tumors. For this purpose, different techniques can be used to produce 3D models, including scaffold-free, scaffold-based and microfluidic-based models. This review addresses the principles, advantages and limitations of these culture methods for evaluating the antitumoral efficacy of nanomedicines.
Collapse
Affiliation(s)
- Larissa Bueno Tofani
- School of Pharmaceutical Science of Ribeirao Preto, University of Sao Paulo (USP), Ribeirao Preto, Sao Paulo, 14040-903, Brazil
| | - Marcela Tavares Luiz
- School of Pharmaceutical Science of Sao Paulo State University (UNESP), Araraquara, Sao Paulo, 14800-903, Brazil
| | - Jessyca Aparecida Paes Dutra
- School of Pharmaceutical Science of Sao Paulo State University (UNESP), Araraquara, Sao Paulo, 14800-903, Brazil
| | - Juliana Palma Abriata
- School of Pharmaceutical Science of Ribeirao Preto, University of Sao Paulo (USP), Ribeirao Preto, Sao Paulo, 14040-903, Brazil
| | - Marlus Chorilli
- School of Pharmaceutical Science of Sao Paulo State University (UNESP), Araraquara, Sao Paulo, 14800-903, Brazil
| |
Collapse
|
10
|
Jackson CE, Ramos-Rodriguez DH, Farr NTH, English WR, Green NH, Claeyssens F. Development of PCL PolyHIPE Substrates for 3D Breast Cancer Cell Culture. Bioengineering (Basel) 2023; 10:bioengineering10050522. [PMID: 37237592 DOI: 10.3390/bioengineering10050522] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2023] [Revised: 04/12/2023] [Accepted: 04/23/2023] [Indexed: 05/28/2023] Open
Abstract
Cancer is a becoming a huge social and economic burden on society, becoming one of the most significant barriers to life expectancy in the 21st century. In particular, breast cancer is one of the leading causes of death for women. One of the most significant difficulties to finding efficient therapies for specific cancers, such as breast cancer, is the efficiency and ease of drug development and testing. Tissue-engineered (TE) in vitro models are rapidly developing as an alternative to animal testing for pharmaceuticals. Additionally, porosity included within these structures overcomes the diffusional mass transfer limit whilst enabling cell infiltration and integration with surrounding tissue. Within this study, we investigated the use of high-molecular-weight polycaprolactone methacrylate (PCL-M) polymerised high-internal-phase emulsions (polyHIPEs) as a scaffold to support 3D breast cancer (MDA-MB-231) cell culture. We assessed the porosity, interconnectivity, and morphology of the polyHIPEs when varying mixing speed during formation of the emulsion, successfully demonstrating the tunability of these polyHIPEs. An ex ovo chick chorioallantoic membrane assay identified the scaffolds as bioinert, with biocompatible properties within a vascularised tissue. Furthermore, in vitro assessment of cell attachment and proliferation showed promising potential for the use of PCL polyHIPEs to support cell growth. Our results demonstrate that PCL polyHIPEs are a promising material to support cancer cell growth with tuneable porosity and interconnectivity for the fabrication of perfusable 3D cancer models.
Collapse
Affiliation(s)
- Caitlin E Jackson
- Materials Science and Engineering, The Kroto Research Institute, University of Sheffield, Sheffield S3 7HQ, UK
- Insigneo Institute for In Silico Medicine, The Pam Liversidge Building, University of Sheffield, Sheffield S1 3JD, UK
| | | | - Nicholas T H Farr
- Materials Science and Engineering, The Kroto Research Institute, University of Sheffield, Sheffield S3 7HQ, UK
- Insigneo Institute for In Silico Medicine, The Pam Liversidge Building, University of Sheffield, Sheffield S1 3JD, UK
| | - William R English
- Norwich Medical School, University of East Anglia, Norwich NR3 7TJ, UK
| | - Nicola H Green
- Materials Science and Engineering, The Kroto Research Institute, University of Sheffield, Sheffield S3 7HQ, UK
- Insigneo Institute for In Silico Medicine, The Pam Liversidge Building, University of Sheffield, Sheffield S1 3JD, UK
| | - Frederik Claeyssens
- Materials Science and Engineering, The Kroto Research Institute, University of Sheffield, Sheffield S3 7HQ, UK
- Insigneo Institute for In Silico Medicine, The Pam Liversidge Building, University of Sheffield, Sheffield S1 3JD, UK
| |
Collapse
|
11
|
Nair L, Mukherjee S, Kaur K, Murphy CM, Ravichandiran V, Roy S, Singh M. Multi compartmental 3D breast cancer disease model–recapitulating tumor complexity in in-vitro. Biochim Biophys Acta Gen Subj 2023; 1867:130361. [PMID: 37019341 DOI: 10.1016/j.bbagen.2023.130361] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2023] [Revised: 03/26/2023] [Accepted: 03/29/2023] [Indexed: 04/05/2023]
Abstract
Breast cancer is the most common ailment among women. In 2020, it had the highest incidence of any type of cancer. Many Phase II and III anti-cancer drugs fail due to efficacy, durability, and side effects. Thus, accelerated drug screening models must be accurate. In-vivo models have been used for a long time, but delays, inconsistent results, and a greater sense of responsibility among scientists toward wildlife have led to the search for in-vitro alternatives. Stromal components support breast cancer growth and survival. Multi-compartment Transwell models may be handy instruments. Co-culturing breast cancer cells with endothelium and fibroblasts improves modelling. The extracellular matrix (ECM) supports native 3D hydrogels in natural and polymeric forms. 3D Transwell cultured tumor spheroids mimicked in-vivo pathological conditions. Tumor invasion, migration, Trans-endothelial migration, angiogenesis, and spread are studied using comprehensive models. Transwell models can create a cancer niche and conduct high-throughput drug screening, promising future applications. Our comprehensive shows how 3D in-vitro multi compartmental models may be useful in producing breast cancer stroma in Transwell culture.
Collapse
Affiliation(s)
- Lakshmi Nair
- Department of Pharmaceutical Sciences, Assam Central University, Silchar, Assam 788011, India
| | - Souvik Mukherjee
- Department of Pharmaceutical Sciences, Guru Ghasidas University, Koni, Bilaspur,(C.G 495009, India
| | - Kulwinder Kaur
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, Royal College of Surgeons (RCSI), Dublin D02YN77, Ireland
| | - Ciara M Murphy
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, Royal College of Surgeons (RCSI), Dublin D02YN77, Ireland; Trinity Centre for Biomedical Engineering, Trinity College Dublin (TCD), Dublin D02YN77, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), RCSI and TCD, Dublin, Ireland
| | - Velayutham Ravichandiran
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal 700054, India
| | - Subhadeep Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal 700054, India.
| | - Manjari Singh
- Department of Pharmaceutical Sciences, Assam Central University, Silchar, Assam 788011, India.
| |
Collapse
|
12
|
The Use of Biomaterials in Three-Dimensional Culturing of Cancer Cells. Curr Issues Mol Biol 2023; 45:1100-1112. [PMID: 36826018 PMCID: PMC9954970 DOI: 10.3390/cimb45020073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/24/2023] [Accepted: 01/27/2023] [Indexed: 02/03/2023] Open
Abstract
Cell culture is an important tool in biological research. Most studies use 2D cell culture, but cells grown in 2D cell culture have drawbacks, including limited cell and cell-extracellular matrix interactions, which make it inaccurate to model conditions in vivo. Anticancer drug screening is an important research and development process for developing new drugs. As an experiment to mimic the cancer environment in vivo, several studies have been carried out on 3-dimensional (3D) cell cultures with added biomaterials. The use of hydrogel in 3D culture cells is currently developing. The type of hydrogel used might influence cell morphology, viability, and drug screening outcome. Therefore, this review discusses 3D cell culture research regarding the addition of biomaterials.
Collapse
|
13
|
Londoño-Berrio M, Castro C, Cañas A, Ortiz I, Osorio M. Advances in Tumor Organoids for the Evaluation of Drugs: A Bibliographic Review. Pharmaceutics 2022; 14:pharmaceutics14122709. [PMID: 36559203 PMCID: PMC9784359 DOI: 10.3390/pharmaceutics14122709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2022] [Revised: 11/25/2022] [Accepted: 11/27/2022] [Indexed: 12/11/2022] Open
Abstract
Tumor organoids are defined as self-organized three-dimensional assemblies of heterogeneous cell types derived from patient samples that mimic the key histopathological, genetic, and phenotypic characteristics of the original tumor. This technology is proposed as an ideal candidate for the evaluation of possible therapies against cancer, presenting advantages over other models which are currently used. However, there are no reports in the literature that relate the techniques and material development of tumor organoids or that emphasize in the physicochemical and biological properties of materials that intent to biomimicry the tumor extracellular matrix. There is also little information regarding the tools to identify the correspondence of native tumors and tumoral organoids (tumoroids). Moreover, this paper relates the advantages of organoids compared to other models for drug evaluation. A growing interest in tumoral organoids has arisen from 2009 to the present, aimed at standardizing the process of obtaining organoids, which more accurately resemble patient-derived tumor tissue. Likewise, it was found that the characteristics to consider for the development of organoids, and therapeutic responses of them, are cell morphology, physiology, the interaction between cells, the composition of the cellular matrix, and the genetic, phenotypic, and epigenetic characteristics. Currently, organoids have been used for the evaluation of drugs for brain, lung, and colon tumors, among others. In the future, tumor organoids will become closer to being considered a better model for studying cancer in clinical practice, as they can accurately mimic the characteristics of tumors, in turn ensuring that the therapeutic response aligns with the clinical response of patients.
Collapse
Affiliation(s)
- Maritza Londoño-Berrio
- Systems Biology Research Group, Pontifical Bolivarian University (Universidad Pontificia Bolivariana), Carrera 78B No. 72a-109, Medellin 050034, Colombia
| | - Cristina Castro
- New Materials Research Group, School of Engineering, Pontifical Bolivarian University, Circular 1 No. 70-01, Medellin 050031, Colombia
| | - Ana Cañas
- Corporation for Biological Research, Medical, and Experimental Research Group, Carrera 72A # 78b-141, Medellin 050034, Colombia
| | - Isabel Ortiz
- Systems Biology Research Group, Pontifical Bolivarian University (Universidad Pontificia Bolivariana), Carrera 78B No. 72a-109, Medellin 050034, Colombia
| | - Marlon Osorio
- Systems Biology Research Group, Pontifical Bolivarian University (Universidad Pontificia Bolivariana), Carrera 78B No. 72a-109, Medellin 050034, Colombia
- New Materials Research Group, School of Engineering, Pontifical Bolivarian University, Circular 1 No. 70-01, Medellin 050031, Colombia
- Correspondence:
| |
Collapse
|
14
|
Sevinyan L, Gupta P, Velliou E, Madhuri TK. The Development of a Three-Dimensional Platform for Patient-Derived Ovarian Cancer Tissue Models: A Systematic Literature Review. Cancers (Basel) 2022; 14:5628. [PMID: 36428724 PMCID: PMC9688222 DOI: 10.3390/cancers14225628] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2022] [Accepted: 11/09/2022] [Indexed: 11/18/2022] Open
Abstract
There is an unmet biomedical need for ex vivo tumour models that would predict drug responses and in turn help determine treatment regimens and potentially predict resistance before clinical studies. Research has shown that three dimensional models of ovarian cancer (OvCa) are more realistic than two dimensional in vitro systems as they are able to capture patient in vivo conditions in more accurate manner. The vast majority of studies aiming to recapitulate the ovarian tumour morphology, behaviors, and study chemotherapy responses have been using ovarian cancer cell lines. However, despite the advantages of utilising cancer cell lines to set up a platform, they are not as informative as systems applying patient derived cells, as cell lines are not able to recapitulate differences between each individual patient characteristics. In this review we discussed the most recent advances in the creation of 3D ovarian cancer models that have used patient derived material, the challenges to overcome and future applications.
Collapse
Affiliation(s)
- Lusine Sevinyan
- Department of Gynaecological Oncology, Royal Surrey NHS Foundation Trust, Guildford GU2 7XX, UK
- Cancer Research, School of Applied Sciences, University of Brighton, Brighton BN2 4HQ, UK
| | - Priyanka Gupta
- Centre for 3D Models of Health and Disease, Division of Surgery and Interventional Science, University College London, London WC1E 6BT, UK
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford GU2 7XH, UK
| | - Eirini Velliou
- Centre for 3D Models of Health and Disease, Division of Surgery and Interventional Science, University College London, London WC1E 6BT, UK
- Bioprocess and Biochemical Engineering Group (BioProChem), Department of Chemical and Process Engineering, University of Surrey, Guildford GU2 7XH, UK
| | - Thumuluru Kavitha Madhuri
- Department of Gynaecological Oncology, Royal Surrey NHS Foundation Trust, Guildford GU2 7XX, UK
- Cancer Research, School of Applied Sciences, University of Brighton, Brighton BN2 4HQ, UK
| |
Collapse
|
15
|
Revia RA, Wagner B, James M, Zhang M. High-Throughput Dispensing of Viscous Solutions for Biomedical Applications. MICROMACHINES 2022; 13:1730. [PMID: 36296083 PMCID: PMC9609595 DOI: 10.3390/mi13101730] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 10/03/2022] [Accepted: 10/07/2022] [Indexed: 06/16/2023]
Abstract
Cells cultured in three-dimensional scaffolds express a phenotype closer to in vivo cells than cells cultured in two-dimensional containers. Natural polymers are suitable materials to make three-dimensional scaffolds to develop disease models for high-throughput drug screening owing to their excellent biocompatibility. However, natural polymer solutions have a range of viscosities, and none of the currently available liquid dispensers are capable of dispensing highly viscous polymer solutions. Here, we report the development of an automated scaffold dispensing system for rapid, reliable, and homogeneous creation of scaffolds in well-plate formats. We employ computer-controlled solenoid valves to regulate air pressure impinging upon a syringe barrel filled with scaffold solution to be dispensed. Automated dispensing of scaffold solution is achieved via a programmable software interface that coordinates solution extrusion and the movement of a dispensing head. We show that our pneumatically actuated dispensing system can evenly distribute high-viscosity, chitosan-based polymer solutions into 96- and 384-well plates to yield highly uniform three-dimensional scaffolds after lyophilization. We provide a proof-of-concept demonstration of high-throughput drug screening by culturing glioblastoma cells in scaffolds and exposing them to temozolomide. This work introduces a device that can hasten the creation of three-dimensional cell scaffolds and their application to high-throughput testing.
Collapse
Affiliation(s)
- Richard A. Revia
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Brandon Wagner
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Matthew James
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
| | - Miqin Zhang
- Department of Materials Science and Engineering, University of Washington, Seattle, WA 98195, USA
- Department of Neurological Surgery, University of Washington, Seattle, WA 98195, USA
| |
Collapse
|
16
|
Keller CR, Hu Y, Ruud KF, VanDeen AE, Martinez SR, Kahn BT, Zhang Z, Chen RK, Li W. Human Breast Extracellular Matrix Microstructures and Protein Hydrogel 3D Cultures of Mammary Epithelial Cells. Cancers (Basel) 2021; 13:cancers13225857. [PMID: 34831010 PMCID: PMC8616054 DOI: 10.3390/cancers13225857] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2021] [Revised: 11/10/2021] [Accepted: 11/17/2021] [Indexed: 01/21/2023] Open
Abstract
Simple Summary Human breast tissue extracellular matrix (ECM) is a microenvironment essential for the survival and biological activities of mammary epithelial cells. The ECM structural features of human breast tissues remain poorly defined. In this study, we identified the structural and mechanical properties of human normal breast and invasive ductal carcinoma tissue ECM using histological methods and atomic force microscopy. Additionally, a protein hydrogel was generated using human breast tissue ECM and defined for its microstructural features using immunofluorescence imaging and machine learning. Furthermore, we examined the three-dimensional growth of normal mammary epithelial cells or breast cancer cells cultured on the ECM protein hydrogel, where the cells exhibited biological phenotypes like those seen in native breast tissues. Our data provide novel insights into cancer cell biology, tissue microenvironment mimicry and engineering, and native tissue ECM-based biomedical and pharmaceutical applications. Abstract Tissue extracellular matrix (ECM) is a structurally and compositionally unique microenvironment within which native cells can perform their natural biological activities. Cells grown on artificial substrata differ biologically and phenotypically from those grown within their native tissue microenvironment. Studies examining human tissue ECM structures and the biology of human tissue cells in their corresponding tissue ECM are lacking. Such investigations will improve our understanding about human pathophysiological conditions for better clinical care. We report here human normal breast tissue and invasive ductal carcinoma tissue ECM structural features. For the first time, a hydrogel was successfully fabricated using whole protein extracts of human normal breast ECM. Using immunofluorescence staining of type I collagen (Col I) and machine learning of its fibrous patterns in the polymerized human breast ECM hydrogel, we have defined the microstructural characteristics of the hydrogel and compared the microstructures with those of other native ECM hydrogels. Importantly, the ECM hydrogel supported 3D growth and cell-ECM interaction of both normal and cancerous mammary epithelial cells. This work represents further advancement toward full reconstitution of the human breast tissue microenvironment, an accomplishment that will accelerate the use of human pathophysiological tissue-derived matrices for individualized biomedical research and therapeutic development.
Collapse
Affiliation(s)
- Chandler R. Keller
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; (C.R.K.); (K.F.R.)
| | - Yang Hu
- Department of Crop and Soil Sciences, College of Agricultural, Human, and Natural Resources Sciences, Washington State University, Pullman, WA 99164, USA; (Y.H.); (Z.Z.)
| | - Kelsey F. Ruud
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; (C.R.K.); (K.F.R.)
| | - Anika E. VanDeen
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164, USA; (A.E.V.); (R.K.C.)
| | - Steve R. Martinez
- Department of Surgery, The Everett Clinic and Providence Regional Cancer Partnership, Everett, WA 98201, USA;
- Department of Medical Education and Clinical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA
| | - Barry T. Kahn
- CellNetix Pathology & Laboratories, Seattle, WA 98104, USA;
- Providence Regional Medical Center, Everett, WA 98201, USA
| | - Zhiwu Zhang
- Department of Crop and Soil Sciences, College of Agricultural, Human, and Natural Resources Sciences, Washington State University, Pullman, WA 99164, USA; (Y.H.); (Z.Z.)
| | - Roland K. Chen
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164, USA; (A.E.V.); (R.K.C.)
| | - Weimin Li
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA; (C.R.K.); (K.F.R.)
- Correspondence:
| |
Collapse
|
17
|
Rijal G. Understanding the Role of Fibroblasts following a 3D Tumoroid Implantation for Breast Tumor Formation. Bioengineering (Basel) 2021; 8:bioengineering8110163. [PMID: 34821729 PMCID: PMC8615023 DOI: 10.3390/bioengineering8110163] [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: 09/29/2021] [Revised: 10/22/2021] [Accepted: 10/24/2021] [Indexed: 11/16/2022] Open
Abstract
An understanding of the participation and modulation of fibroblasts during tumor formation and growth is still unclear. Among many speculates, one might be the technical challenge to reveal the versatile function of fibroblasts in tissue complexity, and another is the dynamics in tissue physiology and cell activity. The histology of most solid tumors shows a predominant presence of fibroblasts, suggesting that tumor cells recruit fibroblasts for breast tumor growth. In this review paper, therefore, the migration, activation, differentiation, secretion, and signaling systems that are associated with fibroblasts and cancer-associated fibroblasts (CAFs) after implantation of a breast tumoroid, i.e., a lab-generated tumor tissue into an animal, are discussed.
Collapse
Affiliation(s)
- Girdhari Rijal
- Department of Medical Laboratory Sciences and Public Health, Tarleton State University, a Member of Texas A & M University System, Fort Worth, TX 76104, USA
| |
Collapse
|
18
|
Estrada‐Villaseñor E, Valdés‐Flores M, Meneses‐García A, Silva‐Bermudez P, Pichardo‐Bahena R, Ostoa‐Saloma P, Mercado‐Celis G, Delgado‐Cedillo ED, Olivos‐Meza A, Landa‐Solís C. A novel model to culture cells from giant cell tumor of bone using three-dimensional (3D) polycaprolactone scaffold. Eng Life Sci 2021; 21:539-543. [PMID: 34584518 PMCID: PMC8456323 DOI: 10.1002/elsc.202100020] [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: 02/05/2021] [Revised: 06/09/2021] [Accepted: 06/21/2021] [Indexed: 11/09/2022] Open
Abstract
Two-dimensional (2D) culture of cells from giant cell tumor of bone (GCTB) is affected by loss of the multinucleated giant cells in subsequent passages. Therefore, there is limited time to study GCTB with all its histological components in 2D culture. Here, we explored the possibility of culturing GCTB cells on a polycaprolactone (PCL)-printed scaffold. We also evaluated the viability of the cultured cells and their adherence to the PCL scaffold at day 14 days using immunofluorescence analysis with calcein, vinculin, and phalloidin. Using the histological technique with hematoxylin and eosin staining, we observed all the histological components of GCTB in this 3D model. Immunohistochemical assays with cathepsin K, p63, and receptor activator of nuclear factor (NF)-κB ligand (RANKL) yielded positive results in this construct, which allowed us to confirm that the seeded cells maintained the expression of GCTB markers. Based on these findings, we concluded that the PCL scaffold is an efficient model to culture GCTB cells, and the cell viability and adherence to the scaffold can be preserved for up to 14 days. Moreover, this model can also be used in subsequent studies to assess in vitro cell-cell interactions and antineoplastic efficacy of certain agents to establish a treatment against GCTB.
Collapse
Affiliation(s)
| | - Margarita Valdés‐Flores
- Genetics LaboratoryNational Institute of Rehabilitation Luis Guillermo Ibarra IbarraMexico CityMexico
| | | | - Phaedra Silva‐Bermudez
- Tissue Engineering and Cell Therapy UnitNational Institute of Rehabilitation Luis Guillermo Ibarra IbarraMexico CityMexico
| | - Raul Pichardo‐Bahena
- Pathology ServiceNational Institute of Rehabilitation Luis Guillermo Ibarra IbarraMexico CityMexico
| | - Pedro Ostoa‐Saloma
- Department of ImmunologyInstitute of Biomedical ResearchNational Autonomous University of MexicoMexico CityMexico
| | - Gabriela Mercado‐Celis
- Laboratory of Clinical GenomicsDivision of Graduate Studies and ResearchFaculty of OdontologyNational Autonomous University of MexicoMexico CityMexico
| | | | - Anell Olivos‐Meza
- Sports Orthopedics and ArthroscopyNational Institute of Rehabilitation Luis Guillermo Ibarra IbarraMexico CityMexico
| | - Carlos Landa‐Solís
- Tissue Engineering and Cell Therapy UnitNational Institute of Rehabilitation Luis Guillermo Ibarra IbarraMexico CityMexico
| |
Collapse
|
19
|
Liu C, Li M, Dong ZX, Jiang D, Li X, Lin S, Chen D, Zou X, Zhang XD, Luker GD. Heterogeneous microenvironmental stiffness regulates pro-metastatic functions of breast cancer cells. Acta Biomater 2021; 131:326-340. [PMID: 34246802 PMCID: PMC8784164 DOI: 10.1016/j.actbio.2021.07.009] [Citation(s) in RCA: 70] [Impact Index Per Article: 17.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 06/24/2021] [Accepted: 07/02/2021] [Indexed: 12/22/2022]
Abstract
Besides molecular and phenotypic variations observed in cancer cells, intratumoral heterogeneity also occurs in the tumor microenvironment. Correlative stiffness maps of different intratumor locations in breast tumor biopsies show that stiffness increases from core to periphery. However, how different local ECM stiffness regulates key functions of cancer cells in tumor progression remains unclear. Although increased tissue stiffness is an established driver of breast cancer progression, conclusions from 2D cultures do not correspond with newer data from cancer cells in 3D environments. Many past studies of breast cancer in 3D culture fail to recapitulate the stiffness of a real breast tumor or the various local stiffnesses present in a tumor microenvironment. In this study, we developed a series of collagen/alginate hybrid hydrogels with adjustable stiffness to match the core, middle, and peripheral zones of a breast tumor. We used this hydrogel system to investigate effects of different local stiffness on morphology, proliferation, and migration of breast cancer cells. RNA sequencing of cells in hydrogels with different stiffness revealed changes in multiple cellular processes underlying cancer progression, including angiogenesis and metabolism. We discovered that tumor cells in a soft environment enriched YAP1 and AP1 signaling related genes, whereas tumor cells in a stiff environment became more pro-angiogenic by upregulating fibronectin 1 (FN1) and matrix metalloproteinase 9 (MMP9) expression. This systematic study defines how the range of environmental stiffnesses present in a breast tumor regulates cancer cells, providing new insights into tumorigenesis and disease progression at the tumor-stroma interface. STATEMENT OF SIGNIFICANCE: Applied a well-defined hybrid hydrogel system to mimic the tumor microenvironment with heterogeneous local stiffness. Breast cancer cells tended to proliferate in soft core environment while migrate in stiff peripheral environment. Breast cancer cells shift from glycolysis to OXPHOS and fatty acid metabolism responding to stiff matrix microenvironment. The transcriptomic profile of breast cancer cells altered due to microenvironmental stiffness changes.
Collapse
Affiliation(s)
- Chun Liu
- Orthopedic Research Institute/Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, China.
| | - Miao Li
- The First Affiliated Hospital of Shenzhen University, Shenzhen Second People's Hospital, Shenzhen, China; Guangdong Key Laboratory for Biomedical Measurements and Ultrasound Imaging, School of Biomedical Engineering, Shenzhen University Health Science Center, Shenzhen, China
| | - Zhao-Xia Dong
- Department of Pharmacology, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University, Shenzhen, China
| | - Dong Jiang
- Orthopedic Research Institute/Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, China
| | - Xiaojing Li
- Guangdong Provincial Key Laboratory of Biomedical Imaging, The Fifth Affiliated Hospital of Sun Yat-sen University, Zhuhai, China
| | - Shuibin Lin
- Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Demeng Chen
- Center for Translational Medicine, Precision Medicine Institute, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Xuenong Zou
- Orthopedic Research Institute/Department of Spine Surgery, The First Affiliated Hospital of Sun Yat-sen University, Guangzhou, China; Guangdong Provincial Key Laboratory of Orthopedics and Traumatology, Guangzhou, China
| | - Xing-Ding Zhang
- Department of Pharmacology, Molecular Cancer Research Center, School of Medicine, Sun Yat-sen University, Shenzhen, China.
| | - Gary D Luker
- Center for Molecular Imaging, Department of Radiology, University of Michigan Medical School, Ann Arbor, MI 48109, United States; Department of Biomedical Engineering, University of Michigan Medical School, Ann Arbor, MI 48109, United States; Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, MI 48109, United States.
| |
Collapse
|
20
|
Joo G, Sultana T, Rahaman S, Bae SH, Jung HI, Lee BT. Polycaprolactone-gelatin membrane as a sealant biomaterial efficiently prevents postoperative anastomotic leakage with promoting tissue repair. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2021; 32:1530-1547. [PMID: 33849401 DOI: 10.1080/09205063.2021.1917107] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Anastomotic leakage due to post-surgical suture line disruption is one of the crucial factors affecting patient's survival and quality of life. To resolve the poor healing of surgical anastomosis and protect suture sites leakage, fibrous membrane sealing patch was developed using a synthetic polymer (polycaprolactone (PCL)) and biopolymer (gelatin). Electrospinning was used to develop fibrous architecture of membranes fabricated in different ratios (15% (w/v) PCL: 15% (w/v) gelatin mixing ratio of 1:1, 1:2, 1:3 and 1:4). Experimental findings suggested that, higher gelatin content in the membranes reduced the fiber diameter and contact angle, leading to a more hydrophilic scaffold facilitating attachment to the defect site. The degradation rate of various PCL-gelatin membranes (P1G1, P1G2, P1G3 and P1G4) was proportional to the gelatin content. Cytocompatibility was assessed using L929 cells while the P1G4 (PCL: gelatin 1:4 ratio) scaffold exhibited optimum outcome. From in vivo study, the wound site healed significantly without any leakage when the sutured area of rat caecum was covered with P1G4 membrane whereas rats in the control group (suture only) showed leakage after two weeks of surgery. In summary, the P1G4 membrane has potential to be applied as a post-surgical leakage-preventing tissue repair biomaterial.
Collapse
Affiliation(s)
- Gyeongjin Joo
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea
| | - Tamanna Sultana
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea.,Institute of Tissue Regeneration, Soonchunhyang University, Cheonan, Republic of Korea
| | - Sohanur Rahaman
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea
| | - Sang Ho Bae
- Institute of Tissue Regeneration, Soonchunhyang University, Cheonan, Republic of Korea.,Department of General Surgery, Soonchunhyang University Hospital 31, Cheonan, Republic of Korea
| | - Hae Il Jung
- Institute of Tissue Regeneration, Soonchunhyang University, Cheonan, Republic of Korea.,Department of General Surgery, Soonchunhyang University Hospital 31, Cheonan, Republic of Korea
| | - Byong-Taek Lee
- Department of Regenerative Medicine, College of Medicine, Soonchunhyang University, Cheonan, Republic of Korea.,Institute of Tissue Regeneration, Soonchunhyang University, Cheonan, Republic of Korea
| |
Collapse
|
21
|
Turtoi M, Anghelache M, Bucatariu SM, Deleanu M, Voicu G, Safciuc F, Manduteanu I, Fundueanu G, Simionescu M, Calin M. A novel platform for drug testing: Biomimetic three-dimensional hyaluronic acid-based scaffold seeded with human hepatocarcinoma cells. Int J Biol Macromol 2021; 185:604-619. [PMID: 34216662 DOI: 10.1016/j.ijbiomac.2021.06.174] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2021] [Revised: 06/09/2021] [Accepted: 06/26/2021] [Indexed: 12/26/2022]
Abstract
Hepatic cancer is one of the most widespread maladies worldwide that requires urgent therapies and thus reliable means for testing anti-cancer drugs. The switch from two-dimensional (2D) to three-dimensional (3D) cell cultures produced an improvement in the in vitro outcomes for testing anti-cancer drugs. We aimed to develop a novel hyaluronic acid (HA)-based 3D cell model of human hepatocellular carcinoma (HepG2 cells) for drug testing and to assess comparatively in 3D vs. 2D, the cytotoxicity and the apoptotic response to the anti-tumor agent, cisplatin. The 3D model was developed by seeding HepG2 cells in a HA/poly(methylvinylether-alt-maleic acid) (HA3P50)-based scaffold. Compared to 2D, the cells grown in the HA3P50 scaffold proliferate into larger-cellular aggregates that exhibit liver-like functions by controlling the release of hepatocyte-specific biomarkers (albumin, urea, bile acids, transaminases) and the synthesis of cytochrome-P450 (CYP)7A1 enzyme. Also, growing the cells in the scaffold sensitize the hepatocytes to the anti-tumor effect of cisplatin, by a mechanism involving the activation of ERK/p38α-MAPK and dysregulation of NF-kB/STAT3/Bcl-2 pathways. In conclusion, the newly developed HA-based 3D model is suitable for chemotherapeutic drug testing on hepatocellular carcinoma. Moreover, the system can be adapted and employed as experimental platform functioning as a proper tissue/tumor surrogate.
Collapse
Affiliation(s)
- Mihaela Turtoi
- "Medical and Pharmaceutical Bionanotechnologies" Laboratory, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 050568 Bucharest, Romania
| | - Maria Anghelache
- "Medical and Pharmaceutical Bionanotechnologies" Laboratory, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 050568 Bucharest, Romania
| | - Sanda-Maria Bucatariu
- Department of Natural Polymers, Bioactive and Biocompatible Materials, "Petru Poni" Institute of Macromolecular Chemistry, 700487 Iassy, Romania
| | - Mariana Deleanu
- "Liquid and Gas Chromatography" Laboratory, Department of Lipidomics, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 050568 Bucharest, Romania; Faculty of Biotechnologies, University of Agronomic Sciences and Veterinary Medicine of Bucharest (UASVM), 050568 Bucharest, Romania
| | - Geanina Voicu
- "Medical and Pharmaceutical Bionanotechnologies" Laboratory, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 050568 Bucharest, Romania
| | - Florentina Safciuc
- "Medical and Pharmaceutical Bionanotechnologies" Laboratory, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 050568 Bucharest, Romania
| | - Ileana Manduteanu
- "Medical and Pharmaceutical Bionanotechnologies" Laboratory, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 050568 Bucharest, Romania
| | - Gheorghe Fundueanu
- Department of Natural Polymers, Bioactive and Biocompatible Materials, "Petru Poni" Institute of Macromolecular Chemistry, 700487 Iassy, Romania
| | - Maya Simionescu
- "Medical and Pharmaceutical Bionanotechnologies" Laboratory, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 050568 Bucharest, Romania
| | - Manuela Calin
- "Medical and Pharmaceutical Bionanotechnologies" Laboratory, Institute of Cellular Biology and Pathology "Nicolae Simionescu" of the Romanian Academy, 050568 Bucharest, Romania.
| |
Collapse
|
22
|
Dey M, Ayan B, Yurieva M, Unutmaz D, Ozbolat IT. Studying Tumor Angiogenesis and Cancer Invasion in a Three-Dimensional Vascularized Breast Cancer Micro-Environment. Adv Biol (Weinh) 2021; 5:e2100090. [PMID: 33857356 PMCID: PMC8574137 DOI: 10.1002/adbi.202100090] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2021] [Revised: 02/28/2021] [Indexed: 12/19/2022]
Abstract
Metastatic breast cancer is one of the deadliest forms of malignancy, primarily driven by its characteristic micro-environment comprising cancer cells interacting with stromal components. These interactions induce genetic and metabolic alterations creating a conducive environment for tumor growth. In this study, a physiologically relevant 3D vascularized breast cancer micro-environment is developed comprising of metastatic MDA-MB-231 cells and human umbilical vein endothelial cells loaded in human dermal fibroblasts laden fibrin, representing the tumor stroma. The matrix, as well as stromal cell density, impacts the transcriptional profile of genes involved in tumor angiogenesis and cancer invasion, which are hallmarks of cancer. Cancer-specific canonical pathways and activated upstream regulators are also identified by the differential gene expression signatures of these composite cultures. Additionally, a tumor-associated vascular bed of capillaries is established exhibiting dilated vessel diameters, representative of in vivo tumor physiology. Further, employing aspiration-assisted bioprinting, cancer-endothelial crosstalk, in the form of collective angiogenesis of tumor spheroids bioprinted at close proximity, is identified. Overall, this bottom-up approach of tumor micro-environment fabrication provides an insight into the potential of in vitro tumor models and enables the identification of novel therapeutic targets as a preclinical drug screening platform.
Collapse
Affiliation(s)
- Madhuri Dey
- Department of Chemistry, Penn State University, University Park, PA, 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA
| | - Bugra Ayan
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA
| | - Marina Yurieva
- The Jackson Laboratory for Genomic Medicine and University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Derya Unutmaz
- The Jackson Laboratory for Genomic Medicine and University of Connecticut Health Center, Farmington, CT 06032, USA
| | - Ibrahim T Ozbolat
- Engineering Science and Mechanics Department, Penn State University, University Park, PA 16802, USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA 16802, USA
- Biomedical Engineering Department, Penn State University, University Park, PA 16802, USA
- Materials Research Institute, Penn State University, University Park, PA 16802, USA
| |
Collapse
|
23
|
Kim J, Lyu HZ, Jung C, Lee KM, Han SH, Lee JH, Cha M. Osteogenic Response of MC3T3-E1 and Raw264.7 in the 3D-Encapsulated Co-Culture Environment. Tissue Eng Regen Med 2021; 18:387-397. [PMID: 33415675 PMCID: PMC8169729 DOI: 10.1007/s13770-020-00321-0] [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: 08/17/2020] [Revised: 10/05/2020] [Accepted: 11/05/2020] [Indexed: 10/22/2022] Open
Abstract
BACKGROUND Three-dimensional (3D) in vitro cultures recapitulate the physiological microenvironment and exhibit high concordance with in vivo conditions. Improving co-culture models with different kind of cell types cultured on a 3D scaffold can closely mimic the in vivo environment. In this study, we examined the osteogenic response of pre-osteoblast MC3T3-E1 cells and Raw264.7 mouse monocytes in a 3D-encapsulated co-culture environment composed of the Cellrix® 3D culture system, which provides a physiologically relevant environment. METHODS The Cellrix® 3D Bio-Gel scaffolds were used to individually culture or co-culture two type cells in 3D microenvironment. Under 3D culture conditions, osteoblastic behavior was evaluated with an ALP assay and staining. ACP assay and TRAP staining were used as osteoclastic behavior indicator. RESULTS Treatment with osteoblastic induction factors (+3F) and RANKL had on positively effect on alkaline phosphatase activity but significantly inhibited to acid phosphatase activity during osteoclastic differentiation in 3D co-culture. Interestingly, alkaline phosphatase activity or acid phosphatase activity in 3D co-culture was stimulated with opposite differentiation factors at an early stage of differentiation. We guess that these effects may be related to RANK-RANKL signaling, which is important in osteoblast regulation of osteoclasts. CONCLUSION In this study, the osteogenic response of 3D encapsulated pre-osteoblast MC3T3-E1 cells and mouse monocyte Raw264.7 cells was successfully demonstrated. Our 3D culture conditions will be able to provide a foundation for developing a high-throughput in vitro bone model to study the effects of various drugs and other agents on molecular pathways.
Collapse
Affiliation(s)
- Jungju Kim
- Research Institute of Biotechnology, Medifab Co, Ltd., 70, Dusan-ro, Doksan-dong, Geumcheon-gu, Seoul, 08584, South Korea
| | - Hao-Zhen Lyu
- Department of Orthopedic Surgery, College of Medicine, Seoul National University, Daehak-ro 103, Jongno-gu, Seoul, 03080, South Korea
| | - Chisung Jung
- Research Institute of Biotechnology, Medifab Co, Ltd., 70, Dusan-ro, Doksan-dong, Geumcheon-gu, Seoul, 08584, South Korea
| | - Kyung Mee Lee
- Department of Orthopedic Surgery, College of Medicine, Seoul National University, Daehak-ro 103, Jongno-gu, Seoul, 03080, South Korea
| | - Shi Huan Han
- Department of Orthopedic Surgery, College of Medicine, Seoul National University, Daehak-ro 103, Jongno-gu, Seoul, 03080, South Korea
| | - Jae Hyup Lee
- Department of Orthopedic Surgery, College of Medicine, Seoul National University, Daehak-ro 103, Jongno-gu, Seoul, 03080, South Korea.
- Department of Orthopedic Surgery, SMG-SNU Boramae Medical Center, Boramae-ro 5-gil 20, Dongjak-gu, Seoul, 07061, South Korea.
| | - Misun Cha
- Research Institute of Biotechnology, Medifab Co, Ltd., 70, Dusan-ro, Doksan-dong, Geumcheon-gu, Seoul, 08584, South Korea.
| |
Collapse
|
24
|
Sharifi M, Bai Q, Babadaei MMN, Chowdhury F, Hassan M, Taghizadeh A, Derakhshankhah H, Khan S, Hasan A, Falahati M. 3D bioprinting of engineered breast cancer constructs for personalized and targeted cancer therapy. J Control Release 2021; 333:91-106. [PMID: 33774120 DOI: 10.1016/j.jconrel.2021.03.026] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 03/21/2021] [Accepted: 03/22/2021] [Indexed: 12/12/2022]
Abstract
The bioprinting technique with specialized tissue production allows the study of biological, physiological, and behavioral changes of cancerous and non-cancerous tissues in response to pharmacological compounds in personalized medicine. To this end, to evaluate the efficacy of anticancer drugs before entering the clinical setting, tissue engineered 3D scaffolds containing breast cancer and derived from the especially patient, similar to the original tissue architecture, can potentially be used. Despite recent advances in the manufacturing of 3D bioprinted breast cancer tissue (BCT), many studies still suffer from reproducibility primarily because of the uncertainty of the materials used in the scaffolds and lack of printing methods. In this review, we present an overview of the breast cancer environment to optimize personalized treatment by examining and identifying the physiological and biological factors that mimic BCT. We also surveyed the materials and techniques related to 3D bioprinting, i.e, 3D bioprinting systems, current strategies for fabrication of 3D bioprinting tissues, cell adhesion and migration in 3D bioprinted BCT, and 3D bioprinted breast cancer metastasis models. Finally, we emphasized on the prospective future applications of 3D bioprinted cancer models for rapid and accurate drug screening in breast cancer.
Collapse
Affiliation(s)
- Majid Sharifi
- Department of Anesthesiology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China; Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Science, Shahroud, Iran; Department of Animal Science, Faculty of Agriculture, University of Tabriz, Tabriz, Iran
| | - Qian Bai
- Department of Anesthesiology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Mohammad Mahdi Nejadi Babadaei
- Department of Molecular Genetics, Faculty of Biological Science, North Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Farhan Chowdhury
- Department of Mechanical Engineering and Energy Processes, Southern Illinois University Carbondale, Carbondale, IL 62901, USA
| | - Mahbub Hassan
- The University of Sydney, School of Chemical and Biomolecular Engineering, NSW 2006, Australia
| | - Akbar Taghizadeh
- Department of Animal Science, Faculty of Agriculture, University of Tabriz, Tabriz, Iran
| | - Hossein Derakhshankhah
- Pharmaceutical Sciences Research Center, Health Institute, Kermanshah University of Medical Sciences, Kermanshah 6714415153, Iran
| | - Suliman Khan
- Department of Anesthesiology, The Second Affiliated Hospital of Zhengzhou University, Zhengzhou, China.
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, Doha 2713, Qatar; Biomedical Research Center, Qatar University, Doha 2713, Qatar.
| | - Mojtaba Falahati
- Department of Nanotechnology, Faculty of Advanced Sciences and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Iran.
| |
Collapse
|
25
|
Janagama D, Hui SK. 3-D Cell Culture Systems in Bone Marrow Tissue and Organoid Engineering, and BM Phantoms as In Vitro Models of Hematological Cancer Therapeutics-A Review. MATERIALS 2020; 13:ma13245609. [PMID: 33316977 PMCID: PMC7763362 DOI: 10.3390/ma13245609] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2020] [Revised: 10/24/2020] [Accepted: 10/29/2020] [Indexed: 12/15/2022]
Abstract
We review the state-of-the-art in bone and marrow tissue engineering (BMTE) and hematological cancer tissue engineering (HCTE) in light of the recent interest in bone marrow environment and pathophysiology of hematological cancers. This review focuses on engineered BM tissue and organoids as in vitro models of hematological cancer therapeutics, along with identification of BM components and their integration as synthetically engineered BM mimetic scaffolds. In addition, the review details interaction dynamics of various BM and hematologic cancer (HC) cell types in co-culture systems of engineered BM tissues/phantoms as well as their relation to drug resistance and cytotoxicity. Interaction between hematological cancer cells and their niche, and the difference with respect to the healthy niche microenvironment narrated. Future perspectives of BMTE for in vitro disease models, BM regeneration and large scale ex vivo expansion of hematopoietic and mesenchymal stem cells for transplantation and therapy are explained. We conclude by overviewing the clinical application of biomaterials in BM and HC pathophysiology and its challenges and opportunities.
Collapse
|
26
|
A new sponge-type hydrogel based on hyaluronic acid and poly(methylvinylether-alt-maleic acid) as a 3D platform for tumor cell growth. Int J Biol Macromol 2020; 165:2528-2540. [PMID: 33098901 DOI: 10.1016/j.ijbiomac.2020.10.095] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2020] [Revised: 09/29/2020] [Accepted: 10/13/2020] [Indexed: 12/15/2022]
Abstract
A new sponge-type hydrogel was obtained by cross-linking hyaluronic acid (HA) and poly(methylvinylether-alt-maleic acid) P(MVE-alt-MA) through a solvent-free thermal method. The sponge-type hydrogel was characterized and checked as a support for cell growth. The influence of concentration and weight ratio of polymers on the morphology and hydrogel stability was investigated. The total polymers concentration of 3% (w/w) and the weight ratio of 1:1 were optimal for the synthesis of a stable hydrogel (HA3P50) and to promote cell proliferation. The swelling measurements revealed a high-water absorption capacity of the hydrogel in basic medium. Diphenhydramine (DPH), lidocaine (Lid) and propranolol (Prop) were loaded within the hydrogel as a model drugs to investigate the ability of drug transport and release. In vitro studies revealed that HA3P50 hydrogel promoted the adhesion and proliferation of human hepatocellular carcinoma cell line HepG2, providing a good support for 3D cell culture to obtain surrogate tumor scaffold suitable for preclinical anti-cancer drug screening.
Collapse
|
27
|
Gregory E, Dugan R, David G, Song YH. The biology and engineered modeling strategies of cancer-nerve crosstalk. Biochim Biophys Acta Rev Cancer 2020; 1874:188406. [PMID: 32827578 DOI: 10.1016/j.bbcan.2020.188406] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2020] [Revised: 07/23/2020] [Accepted: 07/26/2020] [Indexed: 02/06/2023]
Abstract
A recent finding critical to cancer aggravation is the interaction between cancer cells and nerves. There exist two main modes of cancer-nerve interaction: perineural invasion (PNI) and tumor innervation. PNI occurs when cancer cells infiltrate the adjacent nerves, and its relative opposite, tumor innervation, occurs when axons extend into tumor bodies. Like most cancer studies, these crosstalk interactions have mostly been observed in patient samples and animal models at this point, making it difficult to understand the mechanisms in a controlled manner. As such, in recent years in vitro studies have emerged that have helped identify various microenvironmental factors responsible for cancer-nerve crosstalk, including but not limited to neurotrophic factors, neurotransmitters, chemokines, cancer-derived exosomes, and Schwann cells. The versatility of in vitro systems warrants continuous development to increase physiological relevance to study PNI and tumor innervation, for example by utilizing biomimetic three-dimensional (3D) culture systems. Despite the wealth of 3D in vitro cancer models, comparatively there exists a lack of 3D in vitro models of nerve, PNI, and tumor innervation. Native-like 3D in vitro models of cancer-nerve interactions may further help develop therapeutic strategies to curb nerve-mediated cancer aggravation. As such, we provide an overview of the key players of cancer-nerve crosstalk and current in vitro models of the crosstalk, as well as cancer and nerve models. We also discuss a few future directions in cancer-nerve crosstalk research.
Collapse
Affiliation(s)
- Emory Gregory
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR 72701, United States of America.
| | - Reagan Dugan
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR 72701, United States of America.
| | - Gabriel David
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR 72701, United States of America.
| | - Young Hye Song
- Department of Biomedical Engineering, University of Arkansas, Fayetteville, AR 72701, United States of America.
| |
Collapse
|
28
|
3D-microenvironments initiate TCF4 expression rescuing nuclear β-catenin activity in MCF-7 breast cancer cells. Acta Biomater 2020; 103:153-164. [PMID: 31843716 DOI: 10.1016/j.actbio.2019.12.008] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 11/20/2019] [Accepted: 12/09/2019] [Indexed: 12/19/2022]
Abstract
Mechanical cues sensed by tumor cells in their microenvironment can influence important mechanisms including adhesion, invasion and proliferation. However, a common mechanosensitive protein and/or pathway can be regulated in different ways among diverse types of tumors. Of particular interest are human breast epithelial cancers, which markedly exhibit a heterogeneous pattern of nuclear β-catenin localization, a protein known to be involved in both mechanotransduction and tumorigenesis. β-catenin can be aberrantly accumulated in the nucleus wherein it binds to and activates lymphoid enhancer factor/T cell factor (LEF/TCF) transcription factors. At present, little is known about how mechanical cues are integrated into breast cancer cells harboring impaired mechanisms of β-catenin's nuclear uptake and/or retention. This prompted us to investigate the influence of mechanical cues on MCF-7 human breast cancer cells which are known to fail in relocating β-catenin into the nucleus due to very low baseline levels of LEF/TCFs. Exploiting three-dimensional (3D) microscaffolds realized by two-photon lithography, we show that surrounding MCF-7 cells have not only a nuclear pool of β-catenin, but also rescue from their defective expression of TCF4 and boost invasiveness. Together with heightened amounts of vimentin, a β-catenin/TCF-target gene regulator of proliferation and invasiveness, such 3D-elicited changes indicate an epithelial-to-mesenchymal phenotypic switch of MCF-7 cells. This is also consistent with an increased in situ MCF-7 cell proliferation that can be abrogated by blocking β-catenin/TCF-transcription activity. Collectively, these data suggest that 3D microenvironments are per se sufficient to prime a TCF4-dependent rescuing of β-catenin nuclear activity in MCF-7 cells. The employed methodology could, therefore, provide a mechanism-based rationale to dissect further aspects of mechanotranscription in breast cancerogenesis, somewhat independent of β-catenin's nuclear accumulation. More importantly, by considering the heterogeneity of β-catenin signaling pathway in breast cancer patients, these data may open alternative avenues for personalized disease management and prevention. STATEMENT OF SIGNIFICANCE: Mechanical cues play a critical role in cancer pathogenesis. Little is known about their influence in breast cancer cells harboring impaired mechanisms of β-catenin's nuclear uptake and/or retention, involved in both mechanotransduction and tumorigenesis. We engineered 3D scaffold, by two-photon lithography, to study the influence of mechanical cues on MCF-7 cells which are known to fail in relocating β-catenin into the nucleus. We found that 3D microenvironments are per se sufficient to prime a TCF4-dependent rescuing of β-catenin nuclear activity that boost cell proliferation and invasiveness. Thus, let us suggest that our system could provide a mechanism-based rationale to further dissect key aspects of mechanotranscription in breast cancerogenesis and progression, somewhat independent of β-catenin's nuclear accumulation.
Collapse
|
29
|
Bhartiya A, Madi K, Disney CM, Courtois L, Jupe A, Zhang F, Bodey AJ, Lee P, Rau C, Robinson IK, Yusuf M. Phase-contrast 3D tomography of HeLa cells grown in PLLA polymer electrospun scaffolds using synchrotron X-rays. JOURNAL OF SYNCHROTRON RADIATION 2020; 27:158-163. [PMID: 31868748 DOI: 10.1107/s1600577519015583] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/09/2019] [Accepted: 11/18/2019] [Indexed: 06/10/2023]
Abstract
Advanced imaging is useful for understanding the three-dimensional (3D) growth of cells. X-ray tomography serves as a powerful noninvasive, nondestructive technique that can fulfill these purposes by providing information about cell growth within 3D platforms. There are a limited number of studies taking advantage of synchrotron X-rays, which provides a large field of view and suitable resolution to image cells within specific biomaterials. In this study, X-ray synchrotron radiation microtomography at Diamond Light Source and advanced image processing were used to investigate cellular infiltration of HeLa cells within poly L-lactide (PLLA) scaffolds. This study demonstrates that synchrotron X-rays using phase contrast is a useful method to understand the 3D growth of cells in PLLA electrospun scaffolds. Two different fiber diameter (2 and 4 µm) scaffolds with different pore sizes, grown over 2, 5 and 8 days in vitro, were examined for infiltration and cell connectivity. After performing visualization by segmentation of the cells from the fibers, the results clearly show deeper cell growth and higher cellular interconnectivity in the 4 µm fiber diameter scaffold. This indicates the potential for using such 3D technology to study cell-scaffold interactions for future medical use.
Collapse
Affiliation(s)
- A Bhartiya
- London Centre for Nanotechnology, University College London, London WC1H 0AH, UK
| | - K Madi
- 3DMagination Ltd, Atlas Building, Fermi Avenue, Harwell, Didcot OX11 0QX, UK
| | - C M Disney
- School of Mechanical, Aerospace and Civil Engineering, University of Manchester, Manchester, UK
| | - L Courtois
- 3DMagination Ltd, Atlas Building, Fermi Avenue, Harwell, Didcot OX11 0QX, UK
| | - A Jupe
- Department of Applied Computing, The University of Buckingham, UK
| | - F Zhang
- London Centre for Nanotechnology, University College London, London WC1H 0AH, UK
| | - A J Bodey
- Diamond Light Source, Oxfordshire OX11 0DE, UK
| | - P Lee
- Mechanical Engineering, University College London, London WC1E 7JE, UK
| | - C Rau
- Diamond Light Source, Oxfordshire OX11 0DE, UK
| | - I K Robinson
- London Centre for Nanotechnology, University College London, London WC1H 0AH, UK
| | - M Yusuf
- London Centre for Nanotechnology, University College London, London WC1H 0AH, UK
| |
Collapse
|
30
|
Dynamic in vitro models for tumor tissue engineering. Cancer Lett 2019; 449:178-185. [PMID: 30763717 DOI: 10.1016/j.canlet.2019.01.043] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2018] [Revised: 01/24/2019] [Accepted: 01/29/2019] [Indexed: 01/04/2023]
Abstract
Cancer research uses in vitro studies for controllable analysis of tumor behavior and preclinical testing of therapeutics. Shortcomings of basic cell culture systems in recreating in vivo interactions have driven the development of more efficient and biomimetic in vitro environments for cancer research. Assimilation of certain developments in tissue engineering will accelerate and improve the design of these environments. With the continual improvement of the tumor engineering field, the next step is towards macroscopic systems such as scaffold-supported, flow-perfused macroscale tumor bioreactors. Surface modifications of synthetic scaffolds allow for targeted cell adhesion and improved ECM development. Flow perfusion has emerged as means to expose cancerous tissues to critical biomechanical forces for tumor progression while simultaneously improving nutrient and waste transport. Macroscale perfusable systems allow for non-destructive real-time monitoring using biosensors capable of improving understanding of in vitro tumor development at reduced cost and waste. The combination of macroscale perfusable systems, surface-modified synthetic scaffolds, and non-destructive real-time monitoring will provide advanced platforms for in vitro modeling of tumor development, with broad applications in basic tumor research and preclinical drug development.
Collapse
|
31
|
Sempertegui ND, Narkhede AA, Thomas V, Rao SS. A combined compression molding, heating, and leaching process for fabrication of micro-porous poly(ε-caprolactone) scaffolds. JOURNAL OF BIOMATERIALS SCIENCE-POLYMER EDITION 2018; 29:1978-1993. [DOI: 10.1080/09205063.2018.1498719] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Affiliation(s)
- Nicole D. Sempertegui
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL, USA
| | - Akshay A. Narkhede
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL, USA
| | - Vinoy Thomas
- Department of Materials Science and Engineering, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Shreyas S. Rao
- Department of Chemical and Biological Engineering, The University of Alabama, Tuscaloosa, AL, USA
| |
Collapse
|
32
|
Polonio-Alcalá E, Rabionet M, Guerra AJ, Yeste M, Ciurana J, Puig T. Screening of Additive Manufactured Scaffolds Designs for Triple Negative Breast Cancer 3D Cell Culture and Stem-Like Expansion. Int J Mol Sci 2018; 19:E3148. [PMID: 30322103 PMCID: PMC6213377 DOI: 10.3390/ijms19103148] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2018] [Revised: 10/10/2018] [Accepted: 10/11/2018] [Indexed: 12/31/2022] Open
Abstract
Breast cancer stem cells (BCSCs) are tumor-initiating cells responsible for metastasis and tumor reappearance, but their research is limited by the impossibility to cultivate them in a monolayer culture. Scaffolds are three-dimensional (3D) cell culture systems which avoid problems related with culturing BCSC. However, a standardized scaffold for enhancing a BCSC population is still an open issue. The main aim of this study is to establish a suitable poly (lactic acid) (PLA) scaffold which will produce BCSC enrichment, thus allowing them to be studied. Different 3D printing parameters were analyzed using Taguchi experimental design methods. Several PLA scaffold architectures were manufactured using a Fused Filament Fabrication (FFF) 3D printer. They were then evaluated by cell proliferation assay and the configurations with the highest growth rates were subjected to BCSC quantification by ALDH activity. The design SS1 (0.2 mm layer height, 70% infill density, Zigzag infill pattern, 45° infill direction, and 100% flow) obtained the highest proliferation rate and was capable of enhancing a ALDH+ cell population compared to 2D cell culture. In conclusion, the data obtained endorse the PLA porous scaffold as useful for culturing breast cancer cells in a microenvironment similar to in vivo and increasing the numbers of BCSCs.
Collapse
Affiliation(s)
- Emma Polonio-Alcalá
- New Therapeutic Targets Laboratory (TargetsLab)-Oncology Unit, Department of Medical Sciences, Faculty of Medicine, University of Girona, Emili Grahit 77, 17003 Girona, Spain.
- Product, Process and Production Engineering Research Group (GREP), Department of Mechanical Engineering and Industrial Construction, University of Girona, Maria Aurèlia Capmany 61, 17003 Girona, Spain.
| | - Marc Rabionet
- New Therapeutic Targets Laboratory (TargetsLab)-Oncology Unit, Department of Medical Sciences, Faculty of Medicine, University of Girona, Emili Grahit 77, 17003 Girona, Spain.
- Product, Process and Production Engineering Research Group (GREP), Department of Mechanical Engineering and Industrial Construction, University of Girona, Maria Aurèlia Capmany 61, 17003 Girona, Spain.
| | - Antonio J Guerra
- Product, Process and Production Engineering Research Group (GREP), Department of Mechanical Engineering and Industrial Construction, University of Girona, Maria Aurèlia Capmany 61, 17003 Girona, Spain.
| | - Marc Yeste
- Biotechnology of Animal and Human Reproduction (TechnoSperm), Department of Biology, Institute of Food and Agricultural Technology, University of Girona, Pic de Peguera 15, 17003 Girona, Spain.
| | - Joaquim Ciurana
- Product, Process and Production Engineering Research Group (GREP), Department of Mechanical Engineering and Industrial Construction, University of Girona, Maria Aurèlia Capmany 61, 17003 Girona, Spain.
| | - Teresa Puig
- New Therapeutic Targets Laboratory (TargetsLab)-Oncology Unit, Department of Medical Sciences, Faculty of Medicine, University of Girona, Emili Grahit 77, 17003 Girona, Spain.
| |
Collapse
|
33
|
Rijal G, Wang J, Yu I, Gang DR, Chen RK, Li W. Porcine Breast Extracellular Matrix Hydrogel for Spatial Tissue Culture. Int J Mol Sci 2018; 19:ijms19102912. [PMID: 30257480 PMCID: PMC6213433 DOI: 10.3390/ijms19102912] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2018] [Revised: 09/22/2018] [Accepted: 09/22/2018] [Indexed: 02/07/2023] Open
Abstract
Porcine mammary fatty tissues represent an abundant source of natural biomaterial for generation of breast-specific extracellular matrix (ECM). Here we report the extraction of total ECM proteins from pig breast fatty tissues, the fabrication of hydrogel and porous scaffolds from the extracted ECM proteins, the structural properties of the scaffolds (tissue matrix scaffold, TMS), and the applications of the hydrogel in human mammary epithelial cell spatial cultures for cell surface receptor expression, metabolomics characterization, acini formation, proliferation, migration between different scaffolding compartments, and in vivo tumor formation. This model system provides an additional option for studying human breast diseases such as breast cancer.
Collapse
Affiliation(s)
- Girdhari Rijal
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA.
| | - Jing Wang
- Tissue Imaging and Proteomics Laboratory, Washington State University, Pullman, WA 99164, USA.
| | - Ilhan Yu
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164, USA.
| | - David R Gang
- Tissue Imaging and Proteomics Laboratory, Washington State University, Pullman, WA 99164, USA.
| | - Roland K Chen
- School of Mechanical and Materials Engineering, Washington State University, Pullman, WA 99164, USA.
| | - Weimin Li
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA.
| |
Collapse
|
34
|
Rijal G, Li W. Native-mimicking in vitro microenvironment: an elusive and seductive future for tumor modeling and tissue engineering. J Biol Eng 2018; 12:20. [PMID: 30220913 PMCID: PMC6136168 DOI: 10.1186/s13036-018-0114-7] [Citation(s) in RCA: 43] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2018] [Accepted: 08/30/2018] [Indexed: 12/15/2022] Open
Abstract
Human connective tissues are complex physiological microenvironments favorable for optimal survival, function, growth, proliferation, differentiation, migration, and death of tissue cells. Mimicking native tissue microenvironment using various three-dimensional (3D) tissue culture systems in vitro has been explored for decades, with great advances being achieved recently at material, design and application levels. These achievements are based on improved understandings about the functionalities of various tissue cells, the biocompatibility and biodegradability of scaffolding materials, the biologically functional factors within native tissues, and the pathophysiological conditions of native tissue microenvironments. Here we discuss these continuously evolving physical aspects of tissue microenvironment important for human disease modeling, with a focus on tumors, as well as for tissue repair and regeneration. The combined information about human tissue spaces reflects the necessities of considerations when configuring spatial microenvironments in vitro with native fidelity to culture cells and regenerate tissues that are beyond the formats of 2D and 3D cultures. It is important to associate tissue-specific cells with specific tissues and microenvironments therein for a better understanding of human biology and disease conditions and for the development of novel approaches to treat human diseases.
Collapse
Affiliation(s)
- Girdhari Rijal
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99210 USA
| | - Weimin Li
- Department of Biomedical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99210 USA
| |
Collapse
|