1
|
Bhattacharya T, Kumari M, Kaur K, Kaity S, Arumugam S, Ravichandiran V, Roy S. Decellularized extracellular matrix-based bioengineered 3D breast cancer scaffolds for personalized therapy and drug screening. J Mater Chem B 2024; 12:8843-8867. [PMID: 39162395 DOI: 10.1039/d4tb00680a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/21/2024]
Abstract
Breast cancer (BC) is the second deadliest cancer after lung cancer. Similar to all cancers, it is also driven by a 3D microenvironment. The extracellular matrix (ECM) is an essential component of the 3D tumor micro-environment, wherein it functions as a scaffold for cells and provides metabolic support. BC is characterized by alterations in the ECM. Various studies have attempted to mimic BC-specific ECMs using artificial materials, such as Matrigel. Nevertheless, research has proven that naturally derived decellularized extracellular matrices (dECMs) are superior in providing the essential in vivo-like cues needed to mimic a cancer-like environment. Developing in vitro 3-D BC models is not straightforward and requires extensive analysis of the data established by researchers. For the benefit of researchers, in this review, we have tried to highlight all developmental studies that have been conducted by various scientists so far. The analysis of the conclusions drawn from these studies is also discussed. The advantages and drawbacks of the decellularization methods employed for generating BC scaffolds will be covered, and the review will shed light on how dECM scaffolds help develop a BC environment. The later stages of the article will also focus on immunogenicity issues arising from decellularization and the origin of the tissue. Finally, this review will also discuss the biofabrication of matrices, which is the core part of the bioengineering process.
Collapse
Affiliation(s)
- Teeshyo 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.
| | - Mamta Kumari
- 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
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Santanu Kaity
- Department of Pharmaceutics, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India
| | - Somasundaram Arumugam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Chunilal Bhawan, 168 Maniktala Main Road, Kolkata, 700054, West Bengal, India.
| | - 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
|
2
|
Shishparenok AN, Furman VV, Dobryakova NV, Zhdanov DD. Protein Immobilization on Bacterial Cellulose for Biomedical Application. Polymers (Basel) 2024; 16:2468. [PMID: 39274101 PMCID: PMC11397966 DOI: 10.3390/polym16172468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/20/2024] [Accepted: 08/21/2024] [Indexed: 09/16/2024] Open
Abstract
New carriers for protein immobilization are objects of interest in various fields of biomedicine. Immobilization is a technique used to stabilize and provide physical support for biological micro- and macromolecules and whole cells. Special efforts have been made to develop new materials for protein immobilization that are non-toxic to both the body and the environment, inexpensive, readily available, and easy to modify. Currently, biodegradable and non-toxic polymers, including cellulose, are widely used for protein immobilization. Bacterial cellulose (BC) is a natural polymer with excellent biocompatibility, purity, high porosity, high water uptake capacity, non-immunogenicity, and ease of production and modification. BC is composed of glucose units and does not contain lignin or hemicellulose, which is an advantage allowing the avoidance of the chemical purification step before use. Recently, BC-protein composites have been developed as wound dressings, tissue engineering scaffolds, three-dimensional (3D) cell culture systems, drug delivery systems, and enzyme immobilization matrices. Proteins or peptides are often added to polymeric scaffolds to improve their biocompatibility and biological, physical-chemical, and mechanical properties. To broaden BC applications, various ex situ and in situ modifications of native BC are used to improve its properties for a specific application. In vivo studies showed that several BC-protein composites exhibited excellent biocompatibility, demonstrated prolonged treatment time, and increased the survival of animals. Today, there are several patents and commercial BC-based composites for wounds and vascular grafts. Therefore, further research on BC-protein composites has great prospects. This review focuses on the major advances in protein immobilization on BC for biomedical applications.
Collapse
Affiliation(s)
| | - Vitalina V Furman
- The Center for Chemical Engineering, ITMO University, 197101 Saint Petersburg, Russia
| | | | - Dmitry D Zhdanov
- Institute of Biomedical Chemistry, 10/8 Pogodinskaya St., 119121 Moscow, Russia
- Department of Biochemistry, People's Friendship University of Russia Named after Patrice Lumumba (RUDN University), Miklukho-Maklaya St. 6, 117198 Moscow, Russia
| |
Collapse
|
3
|
Keller CR, Martinez SR, Keltz A, Chen M, Li W. Lactate Oxidase Disrupts Lactate-Activated RAS and PI3K Oncogenic Signaling. Cancers (Basel) 2024; 16:2817. [PMID: 39199589 PMCID: PMC11353192 DOI: 10.3390/cancers16162817] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 07/31/2024] [Accepted: 08/07/2024] [Indexed: 09/01/2024] Open
Abstract
LOX was recently shown to inhibit cancer cell proliferation and tumor growth. The mechanism of this inhibition, however, has been exclusively attributed to LOX depletion of TME lactate, a cancer cell energy source, and production of H2O2, an oxidative stressor. We report that TME lactate triggers the assembly of the lactate receptor hydroxycarboxylic acid receptor 1 (HCAR1)-associated protein complex, which includes GRB2, SOS1, KRAS, GAB1, and PI3K, for the activation of both the RAS and the PI3K oncogenic signaling pathways in breast cancer (BCa) cells. LOX treatment decreased the levels of the proteins in the protein complex via induction of their proteasomal degradation. In addition, LOX inhibited lactate-stimulated expression of the lactate transporters MCT1 and MCT4. Our data suggest that HCAR1 activation by lactate is crucial for the assembly and function of the RAS and PI3K signaling nexus. Shutting down lactate signaling by disrupting this nexus could be detrimental to cancer cells. HCAR1 is therefore a promising target for the control of the RAS and the PI3K signaling required for BCa progression. Thus, our study provides insights into lactate signaling regulation of cancer progression and extends our understanding of LOX's functional mechanisms that are fundamental for exploring its therapeutic potential.
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
| | - Steve R. Martinez
- Department of Surgery, The Everett Clinic, Part of Optum, Everett, WA 98201, USA
- Providence Regional Cancer Partnership, Providence Regional Medical Center, Everett, WA 98201, USA
- Department of Medical Education and Clinical Sciences, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA
| | - Alexys Keltz
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA
- Eastern Washington University, Cheney, WA 99004, USA
| | - Michelle Chen
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA
- Ferris High School, Spokane, WA 99223, USA
| | - Weimin Li
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA
| |
Collapse
|
4
|
Gao X, Caruso BR, Li W. Advanced Hydrogels in Breast Cancer Therapy. Gels 2024; 10:479. [PMID: 39057502 PMCID: PMC11276203 DOI: 10.3390/gels10070479] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Revised: 07/13/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Breast cancer is the most common malignancy among women and is the second leading cause of cancer-related death for women. Depending on the tumor grade and stage, breast cancer is primarily treated with surgery and antineoplastic therapy. Direct or indirect side effects, emotional trauma, and unpredictable outcomes accompany these traditional therapies, calling for therapies that could improve the overall treatment and recovery experiences of patients. Hydrogels, biomimetic materials with 3D network structures, have shown great promise for augmenting breast cancer therapy. Hydrogel implants can be made with adipogenic and angiogenic properties for tissue integration. 3D organoids of malignant breast tumors grown in hydrogels retain the physical and genetic characteristics of the native tumors, allowing for post-surgery recapitulation of the diseased tissues for precision medicine assessment of the responsiveness of patient-specific cancers to antineoplastic treatment. Hydrogels can also be used as carrier matrices for delivering chemotherapeutics and immunotherapeutics or as post-surgery prosthetic scaffolds. The hydrogel delivery systems could achieve localized and controlled medication release targeting the tumor site, enhancing efficacy and minimizing the adverse effects of therapeutic agents delivered by traditional procedures. This review aims to summarize the most recent advancements in hydrogel utilization for breast cancer post-surgery tissue reconstruction, tumor modeling, and therapy and discuss their limitations in clinical translation.
Collapse
Affiliation(s)
- Xiangyu Gao
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA
- Doctor of Medicine Program, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA;
| | - Benjamin R. Caruso
- Doctor of Medicine Program, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA;
| | - Weimin Li
- Department of Translational Medicine and Physiology, Elson S. Floyd College of Medicine, Washington State University, Spokane, WA 99202, USA
| |
Collapse
|
5
|
García-Astrain C, Henriksen-Lacey M, Lenzi E, Renero-Lecuna C, Langer J, Piñeiro P, Molina-Martínez B, Plou J, Jimenez de Aberasturi D, Liz-Marzán LM. A Scaffold-Assisted 3D Cancer Cell Model for Surface-Enhanced Raman Scattering-Based Real-Time Sensing and Imaging. ACS NANO 2024; 18:11257-11269. [PMID: 38632933 PMCID: PMC11064228 DOI: 10.1021/acsnano.4c00543] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/02/2024] [Accepted: 04/09/2024] [Indexed: 04/19/2024]
Abstract
Despite recent advances in the development of scaffold-based three-dimensional (3D) cell models, challenges persist in imaging and monitoring cell behavior within these complex structures due to their heterogeneous cell distribution and geometries. Incorporating sensors into 3D scaffolds provides a potential solution for real-time, in situ sensing and imaging of biological processes such as cell growth and disease development. We introduce a 3D printed hydrogel-based scaffold capable of supporting both surface-enhanced Raman scattering (SERS) biosensing and imaging of 3D breast cancer cell models. The scaffold incorporates plasmonic nanoparticles and SERS tags, for sensing and imaging, respectively. We demonstrate the scaffold's adaptability and modularity in supporting breast cancer spheroids, thereby enabling spatial and temporal monitoring of tumor evolution.
Collapse
Affiliation(s)
- Clara García-Astrain
- CIC
biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Centro
de Investigación Biomédica en Red, Bioingeniería,
Biomateriales y Nanomedicina (CIBER-BBN), 20014 Donostia-San Sebastián, Spain
| | - Malou Henriksen-Lacey
- CIC
biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Centro
de Investigación Biomédica en Red, Bioingeniería,
Biomateriales y Nanomedicina (CIBER-BBN), 20014 Donostia-San Sebastián, Spain
| | - Elisa Lenzi
- CIC
biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
| | - Carlos Renero-Lecuna
- CIC
biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Cinbio,
University of Vigo, 36310 Vigo, Spain
| | - Judith Langer
- CIC
biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
| | - Paula Piñeiro
- CIC
biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Department
of Applied Chemistry, University of the
Basque Country (UPV-EHU), 20018 Donostia-San Sebastián, Spain
| | - Beatriz Molina-Martínez
- CIC
biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
| | - Javier Plou
- CIC
biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
| | - Dorleta Jimenez de Aberasturi
- CIC
biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Centro
de Investigación Biomédica en Red, Bioingeniería,
Biomateriales y Nanomedicina (CIBER-BBN), 20014 Donostia-San Sebastián, Spain
- Ikerbasque,
Basque Foundation for Science, 48009 Bilbao, Spain
| | - Luis M. Liz-Marzán
- CIC
biomaGUNE, Basque Research and Technology Alliance (BRTA), 20014 Donostia-San
Sebastián, Spain
- Centro
de Investigación Biomédica en Red, Bioingeniería,
Biomateriales y Nanomedicina (CIBER-BBN), 20014 Donostia-San Sebastián, Spain
- Cinbio,
University of Vigo, 36310 Vigo, Spain
- Ikerbasque,
Basque Foundation for Science, 48009 Bilbao, Spain
| |
Collapse
|
6
|
Quoniou R, Moreau E, Cachin F, Miot-Noirault E, Chautard E, Peyrode C. 3D Coculture between Cancer Cells and Macrophages: From Conception to Experimentation. ACS Biomater Sci Eng 2024; 10:313-325. [PMID: 38110331 DOI: 10.1021/acsbiomaterials.3c01437] [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] [Indexed: 12/20/2023]
Abstract
A tumor is a complex cluster with many types of cells in the microenvironment that help it grow. Macrophages, immune cells whose main role is to maintain body homeostasis, represent in the majority of cancers the most important cell population. In this context, they are called tumor-associated macrophages (TAMs) because of their phenotype, which contributes to tumor growth. In order to limit the use of animals, there is a real demand for the creation of in vitro models able to represent more specifically the complexity of the tumor microenvironment (TME) in order to characterize it and evaluate new treatments. However, the two-dimensional (2D) culture, which has been used for a long time, has shown many limitations, especially in terms of tumor representation. The three-dimensional (3D) models, developed over the last 20 years, have made it possible to get closer to what happens in vivo in terms of phenotypic and functional characteristics. Due to their architectural similarity to in vivo tissues, they provide a more physiologically relevant in vitro system. Most recently, it is the development of 3D coculture models in which two or three cell lines are cultured together that has allowed a better representation of TME with cell-cell interactions. Unfortunately, there is no clear direction for the design of these models at this time. In this Review on the coculture between cancer cells and TAMs, an in-depth analysis is performed to answer multiple questions on the conception of these models: Which models to use, and with which material and cancer lineage? Which monocyte or macrophage lines should be added to the coculture? And how can these models be exploited?
Collapse
Affiliation(s)
- Rohan Quoniou
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, 63000 Clermont-Ferrand, France
| | - Emmanuel Moreau
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, 63000 Clermont-Ferrand, France
| | - Florent Cachin
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, 63000 Clermont-Ferrand, France
- Service de Médecine Nucléaire, Centre Jean Perrin, 63000 Clermont-Ferrand, France
| | - Elisabeth Miot-Noirault
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, 63000 Clermont-Ferrand, France
| | - Emmanuel Chautard
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, 63000 Clermont-Ferrand, France
- Service de Pathologie, Centre Jean Perrin, 63000 Clermont-Ferrand, France
| | - Caroline Peyrode
- Imagerie Moléculaire et Stratégies Théranostiques, UMR1240, Université Clermont Auvergne, INSERM, 63000 Clermont-Ferrand, France
| |
Collapse
|
7
|
Wei Z, Liu J, Jia Y, Lei M, Zhang S, Xi P, Ma Y, Zhang M, Ma J, Wang L, Guo H, Xu F. Fiber Microarchitecture in Interpenetrating Collagen-Alginate Hydrogel with Tunable Mechanical Plasticity Regulates Tumor Cell Migration. Adv Healthc Mater 2023; 12:e2301586. [PMID: 37506713 DOI: 10.1002/adhm.202301586] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2023] [Revised: 07/13/2023] [Indexed: 07/30/2023]
Abstract
The fiber structures of tumor microenvironment (TME) are well-known in regulating tumor cell behaviors, and the plastic remolding of TME has recently been suggested to enhance tumor metastasis as well. However, the interrelationship between the fiber microarchitecture and matrix plasticity is inextricable by existing in vitro models. The individual roles of fiber microarchitecture and matrix plasticity in tuning tumor cell behaviors remain elusive. This study develops an interpenetrating collagen-alginate hydrogel platform with independently tunable matrix plasticity and fiber microarchitecture through an interpenetrating strategy of alginate networks and collagen I networks. With this hydrogel platform, it is demonstrated that tumor cells in high plasticity hydrogels are more extensive and aggressive than in low plasticity hydrogels and fiber structures only have influence in high plasticity hydrogels. The study further elucidates the underlying mechanisms through analyzing the distribution of forces within the matrix and tracking the focal adhesions (FAs) and finds that highly plastic hydrogels can activate the FAs formation, whereas the maturation and stability of FAs are dominated by fiber dispersion. This study not only establishes new ideas on how cells interact with TME cues but also would help to further finely tailor engineered hydrogel platforms for studying tumor behaviors in vitro.
Collapse
Affiliation(s)
- Zhao Wei
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Jingyi Liu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Yuanbo Jia
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Meng Lei
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Songbai Zhang
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Department of General Dentistry and Emergency, School of Stomatology, Fourth Military Medical University, No. 145 West Changle Road, Xi'an, 710032, P. R. China
| | - Pan Xi
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Yufei Ma
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| | - Min Zhang
- State Key Laboratory of Military Stomatology and National Clinical Research Center for Oral Diseases and Shaanxi International Joint Research Center for Oral Diseases, Department of General Dentistry and Emergency, School of Stomatology, Fourth Military Medical University, No. 145 West Changle Road, Xi'an, 710032, P. R. China
| | - Jinlu Ma
- Department of Radiation Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Lin Wang
- College of Medicine, Xi'an International University, Xi'an, Shaanxi, 710077, China
- Engineering Research Center of Personalized Anti-aging Health Product Development and Transformation, Universities of Shaanxi Province, Xi'an, Shaanxi, 710077, China
| | - Hui Guo
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
- Department of Medical Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an, 710061, P. R. China
| | - Feng Xu
- The Key Laboratory of Biomedical Information Engineering of Ministry of Education, School of Life Science and Technology, Xi'an Jiaotong University, Xi'an, 710049, China
- Bioinspired Engineering and Biomechanics Center (BEBC), Xi'an Jiaotong University, Xi'an, 710049, P. R. China
| |
Collapse
|
8
|
Blanchard R, Adjei I. Engineering the glioblastoma microenvironment with bioactive nanoparticles for effective immunotherapy. RSC Adv 2023; 13:31411-31425. [PMID: 37901257 PMCID: PMC10603567 DOI: 10.1039/d3ra01153d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Accepted: 09/27/2023] [Indexed: 10/31/2023] Open
Abstract
While immunotherapies have revolutionized treatment for other cancers, glioblastoma multiforme (GBM) patients have not shown similar positive responses. The limited response to immunotherapies is partly due to the unique challenges associated with the GBM tumor microenvironment (TME), which promotes resistance to immunotherapies, causing many promising therapies to fail. There is, therefore, an urgent need to develop strategies that make the TME immune permissive to promote treatment efficacy. Bioactive nano-delivery systems, in which the nanoparticle, due to its chemical composition, provides the pharmacological function, have recently emerged as an encouraging option for enhancing the efficacy of immunotherapeutics. These systems are designed to overcome immunosuppressive mechanisms in the TME to improve the efficacy of a therapy. This review will discuss different aspects of the TME and how they impede therapy success. Then, we will summarize recent developments in TME-modifying nanotherapeutics and the in vitro models utilized to facilitate these advances.
Collapse
Affiliation(s)
- Ryan Blanchard
- Department of Biomedical Engineering, Texas A&M University TX USA
| | - Isaac Adjei
- Department of Biomedical Engineering, Texas A&M University TX USA
| |
Collapse
|
9
|
Farahani MK, Gharibshahian M, Rezvani A, Vaez A. Breast cancer brain metastasis: from etiology to state-of-the-art modeling. J Biol Eng 2023; 17:41. [PMID: 37386445 DOI: 10.1186/s13036-023-00352-w] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Accepted: 05/02/2023] [Indexed: 07/01/2023] Open
Abstract
Currently, breast carcinoma is the most common form of malignancy and the main cause of cancer mortality in women worldwide. The metastasis of cancer cells from the primary tumor site to other organs in the body, notably the lungs, bones, brain, and liver, is what causes breast cancer to ultimately be fatal. Brain metastases occur in as many as 30% of patients with advanced breast cancer, and the 1-year survival rate of these patients is around 20%. Many researchers have focused on brain metastasis, but due to its complexities, many aspects of this process are still relatively unclear. To develop and test novel therapies for this fatal condition, pre-clinical models are required that can mimic the biological processes involved in breast cancer brain metastasis (BCBM). The application of many breakthroughs in the area of tissue engineering has resulted in the development of scaffold or matrix-based culture methods that more accurately imitate the original extracellular matrix (ECM) of metastatic tumors. Furthermore, specific cell lines are now being used to create three-dimensional (3D) cultures that can be used to model metastasis. These 3D cultures satisfy the requirement for in vitro methodologies that allow for a more accurate investigation of the molecular pathways as well as a more in-depth examination of the effects of the medication being tested. In this review, we talk about the latest advances in modeling BCBM using cell lines, animals, and tissue engineering methods.
Collapse
Affiliation(s)
| | - Maliheh Gharibshahian
- Student Research Committee, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Alireza Rezvani
- Hematology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Ahmad Vaez
- Department of Tissue Engineering and Applied Cell Sciences, School of Advanced Medical Sciences and Technologies, Shiraz University of Medical Sciences, Shiraz, Iran.
| |
Collapse
|
10
|
Chai W, Kong Y, Escalona MB, Hu C, Balajee AS, Huang Y. Evaluation of Low-dose Radiation-induced DNA Damage and Repair in 3D Printed Human Cellular Constructs. HEALTH PHYSICS 2023; Publish Ahead of Print:00004032-990000000-00091. [PMID: 37294952 DOI: 10.1097/hp.0000000000001709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
ABSTRACT DNA double-strand breaks (DSBs) induced by ionizing radiation (IR) are considered to be the most critical lesion that when unrepaired or misrepaired leads to genomic instability or cell death depending on the radiation exposure dose. The potential health risks associated with exposures of low-dose radiation are of concern since they are being increasingly used in diverse medical and non-medical applications. Here, we have used a novel human tissue-like 3-dimensional bioprint to evaluate low-dose radiation-induced DNA damage response. For the generation of 3-dimensional tissue-like constructs, human hTERT immortalized foreskin fibroblast BJ1 cells were extrusion printed and further enzymatically gelled in a gellan microgel-based support bath. Low-dose radiation-induced DSBs and repair were analyzed in the tissue-like bioprints by indirect immunofluorescence using a well-known DSB surrogate marker, 53BP1, at different post-irradiation times (0.5 h, 6 h, and 24 h) after treatment with various doses of γ rays (50 mGy, 100 mGy, and 200 mGy). The 53BP1 foci showed a dose dependent induction in the tissue bioprints after 30 min of radiation exposure and subsequently declined at 6 h and 24 h in a dose-dependent manner. The residual 53BP1 foci number observed at 24 h post-irradiation time for the γ-ray doses of 50 mGy, 100 mGy, and 200 mGy was not statistically different from mock treated bioprints illustrative of an efficient DNA repair response at these low-dose exposures. Similar results were obtained for yet another DSB surrogate marker, γ-H2AX (phosphorylated form of histone H2A variant) in the human tissue-like constructs. Although we have primarily used foreskin fibroblasts, our bioprinting approach-mimicking a human tissue-like microenvironment-can be extended to different organ-specific cell types for evaluating the radio-response at low-dose and dose-rates of IR.
Collapse
Affiliation(s)
- Wenxuan Chai
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611
| | - Yunfan Kong
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611
| | - Maria B Escalona
- Cytogenetic Biodosimetry Laboratory, Radiation Emergency Assistance Center/Training Site, Oak Ridge Associated Universities, 1299 Bethel Valley Road, Oak Ridge, TN 37830
| | - Chunshan Hu
- Department of Mechanical and Aerospace Engineering, University of Florida, Gainesville, FL 32611
| | - Adayabalam S Balajee
- Cytogenetic Biodosimetry Laboratory, Radiation Emergency Assistance Center/Training Site, Oak Ridge Associated Universities, 1299 Bethel Valley Road, Oak Ridge, TN 37830
| | | |
Collapse
|
11
|
Marques JROF, González-Alva P, Yu-Tong Lin R, Ferreira Fernandes B, Chaurasia A, Dubey N. Advances in tissue engineering of cancer microenvironment-from three-dimensional culture to three-dimensional printing. SLAS Technol 2023; 28:152-164. [PMID: 37019216 DOI: 10.1016/j.slast.2023.03.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2022] [Revised: 02/27/2023] [Accepted: 03/28/2023] [Indexed: 04/05/2023]
Abstract
Cancer treatment development is a complex process, with tumor heterogeneity and inter-patient variations limiting the success of therapeutic intervention. Traditional two-dimensional cell culture has been used to study cancer metabolism, but it fails to capture physiologically relevant cell-cell and cell-environment interactions required to mimic tumor-specific architecture. Over the past three decades, research efforts in the field of 3D cancer model fabrication using tissue engineering have addressed this unmet need. The self-organized and scaffold-based model has shown potential to study the cancer microenvironment and eventually bridge the gap between 2D cell culture and animal models. Recently, three-dimensional (3D) bioprinting has emerged as an exciting and novel biofabrication strategy aimed at developing a 3D compartmentalized hierarchical organization with the precise positioning of biomolecules, including living cells. In this review, we discuss the advancements in 3D culture techniques for the fabrication of cancer models, as well as their benefits and limitations. We also highlight future directions associated with technological advances, detailed applicative research, patient compliance, and regulatory challenges to achieve a successful bed-to-bench transition.
Collapse
Affiliation(s)
- Joana Rita Oliveira Faria Marques
- Oral Biology and Biochemistry Research Group (GIBBO), Unidade de Investigação em Ciências Orais e Biomédicas (UICOB), Faculdade de Medicina Dentária, Universidade de Lisboa, Lisboa, Portugal
| | - Patricia González-Alva
- Tissue Bioengineering Laboratory, Postgraduate Studies and Research Division, Faculty of Dentistry, National Autonomous University of Mexico (UNAM), 04510, Mexico, CDMX, Mexico
| | - Ruby Yu-Tong Lin
- Faculty of Dentistry, National University of Singapore, Singapore
| | - Beatriz Ferreira Fernandes
- Oral Biology and Biochemistry Research Group (GIBBO), Unidade de Investigação em Ciências Orais e Biomédicas (UICOB), Faculdade de Medicina Dentária, Universidade de Lisboa, Lisboa, Portugal
| | - Akhilanand Chaurasia
- Department of Oral Medicine, Faculty of Dental Sciences, King George's Medical University, Lucknow, Uttar Pradesh, India
| | - Nileshkumar Dubey
- Faculty of Dentistry, National University of Singapore, Singapore; ORCHIDS: Oral Care Health Innovations and Designs Singapore, National University of Singapore, Singapore.
| |
Collapse
|
12
|
Kim H, Wirasaputra A, Mohammadi F, Kundu AN, Esteves JAE, Heiser LM, Meyer AS, Peyton SR. Live Cell Lineage Tracing of Dormant Cancer Cells. Adv Healthc Mater 2023; 12:e2202275. [PMID: 36625629 PMCID: PMC10238615 DOI: 10.1002/adhm.202202275] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2022] [Revised: 12/02/2022] [Indexed: 01/11/2023]
Abstract
Breast cancer is a leading cause of global cancer-related deaths, and metastasis is the overwhelming culprit of poor patient prognosis. The most nefarious aspect of metastasis is dormancy, a prolonged period between primary tumor resection and relapse. Current therapies are insufficient at killing dormant cells; thus, they can remain quiescent in the body for decades until eventually undergoing a phenotypic switch, resulting in metastases that are more adaptable and drug resistant. Unfortunately, dormancy has few in vitro models, largely because lab-derived cell lines are highly proliferative. Existing models address tumor dormancy, not cellular dormancy, because tracking individual cells is technically challenging. To combat this problem, a live cell lineage approach to find and track individual dormant cells, distinguishing them from proliferative and dying cells over multiple days, is adapted. This approach is applied across a range of different in vitro microenvironments. This approach reveals that the proportion of cells that exhibit long-term quiescence is regulated by both cell intrinsic and extrinsic factors, with the most dormant cells found in 3D collagen gels. This paper envisions that this approach will prove useful to biologists and bioengineers in the dormancy community to identify, quantify, and study dormant tumor cells.
Collapse
Affiliation(s)
- Hyuna Kim
- Molecular and Cell Biology Graduate Program, University of Massachusetts, Amherst, MA, 01002, USA
| | - Anna Wirasaputra
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, 01002, USA
| | - Farnaz Mohammadi
- Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA
| | - Aritra Nath Kundu
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, 01002, USA
| | - Jennifer A E Esteves
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, 01002, USA
| | - Laura M Heiser
- Department of Biomedical Engineering, Knight Cancer Institute, Oregon Health & Science University, Portland, OR, 97239, USA
| | - Aaron S Meyer
- Department of Bioengineering, University of California, Los Angeles, CA, 90095, USA
- Jonsson Comprehensive Cancer Center, University of California, Los Angeles, CA, 90095, USA
| | - Shelly R Peyton
- Molecular and Cell Biology Graduate Program, University of Massachusetts, Amherst, MA, 01002, USA
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, 01002, USA
- Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, 01002, USA
| |
Collapse
|
13
|
Ahmadian M, Hosseini S, Alipour A, Jahanfar M, Farrokhi N, Homaeigohar S, Shahsavarani H. In vitro modeling of hepatocellular carcinoma niche on decellularized tomato thorny leaves: a novel natural three-dimensional (3D) scaffold for liver cancer therapeutics. Front Bioeng Biotechnol 2023; 11:1189726. [PMID: 37251569 PMCID: PMC10212619 DOI: 10.3389/fbioe.2023.1189726] [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: 03/19/2023] [Accepted: 04/24/2023] [Indexed: 05/31/2023] Open
Abstract
Liver cancer is now one of the main causes leading to death worldwide. To achieve reliable therapeutic effects, it is crucial to develop efficient approaches to test novel anticancer drugs. Considering the significant contribution of tumor microenvironment to cell's response to medications, in vitro 3D bioinspiration of cancer cell niches can be regarded as an advanced strategy to improve the accuracy and reliability of the drug-based treatment. In this regard, decellularized plant tissues can perform as suitable 3D scaffolds for mammalian cell culture to create a near-to-real condition to test drug efficacy. Here, we developed a novel 3D natural scaffold made from decellularized tomato hairy leaves (hereafter called as DTL) to mimic the microenvironment of human hepatocellular carcinoma (HCC) for pharmaceutical purposes. The surface hydrophilicity, mechanical properties, and topography measurement and molecular analyses revealed that the 3D DTL scaffold is an ideal candidate for liver cancer modeling. The cells exhibited a higher growth and proliferation rate within the DTL scaffold, as verified by quantifying the expression of related genes, DAPI staining, and SEM imaging of the cells. Moreover, prilocaine, an anticancer drug, showed a higher effectiveness against the cancer cells cultured on the 3D DTL scaffold, compared to a 2D platform. Taken together, this new cellulosic 3D scaffold can be confidently proposed for chemotherapeutic testing of drugs on hepatocellular carcinoma.
Collapse
Affiliation(s)
- Mariye Ahmadian
- Department of Cell and Molecular Biology, Faculty of Life Science and Biotechnology, Shahid Beheshti University, Tehran, Iran
- Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, National Cell Bank, Tehran, Iran
| | - Saadi Hosseini
- Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, National Cell Bank, Tehran, Iran
| | - Atefeh Alipour
- Department of Nanobiotechnology, Pasteur Institute of Iran, Tehran, Iran
| | - Mehdi Jahanfar
- Department of Cell and Molecular Biology, Faculty of Life Science and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Naser Farrokhi
- Department of Cell and Molecular Biology, Faculty of Life Science and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Shahin Homaeigohar
- School of Science and Engineering, University of Dundee, Dundee, United Kingdom
| | - Hosein Shahsavarani
- Department of Cell and Molecular Biology, Faculty of Life Science and Biotechnology, Shahid Beheshti University, Tehran, Iran
- Laboratory of Regenerative Medicine and Biomedical Innovations, Pasteur Institute of Iran, National Cell Bank, Tehran, Iran
| |
Collapse
|
14
|
Identification of the Collagen Types Essential for Mammalian Breast Acinar Structures. Gels 2022; 8:gels8120837. [PMID: 36547361 PMCID: PMC9777629 DOI: 10.3390/gels8120837] [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: 11/10/2022] [Revised: 12/12/2022] [Accepted: 12/16/2022] [Indexed: 12/23/2022] Open
Abstract
Modeling human breast tissue architecture is essential to study the pathophysiological conditions of the breast. We report that normal mammary epithelial cells grown in human breast extracellular matrix (ECM) hydrogel formed acini structurally similar to those of human and pig mammary tissues. Type I, II, III and V collagens were commonly identified in human, pig, and mouse breast ECM. Mammary epithelial cells formed acini on certain types or combinations of the four collagens at normal levels of breast tissue elasticity. Comparison of the collagen species in mouse normal breast and breast tumor ECM revealed common and distinct sets of collagens within the two types of tissues. Elevated expression of collagen type I alpha 1 chain (Col1a1) was found in mouse and human breast cancers. Collagen type XXV alpha 1 chain (Col25a1) was identified in mouse breast tumors but not in normal breast tissues. Our data provide strategies for modeling human breast pathophysiological structures and functions using native tissue-derived hydrogels and offer insight into the potential contributions of different collagen types in breast cancer development.
Collapse
|
15
|
Xie D, Jia S, Ping D, Wang D, Cao L. Scaffold-based three-dimensional cell model of pancreatic cancer is more suitable than scaffold-free three-dimensional cell model of pancreatic cancer for drug discovery. Cytotechnology 2022; 74:657-667. [PMID: 36389286 PMCID: PMC9652184 DOI: 10.1007/s10616-022-00553-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Accepted: 10/03/2022] [Indexed: 11/03/2022] Open
Abstract
Pancreatic cancer is one of the deadliest malignancies. Three-dimensional (3D) pancreatic cancer cell models for drug screening have been established to improve treatment for pancreatic cancer. However, few studies focus on different drug responses and drug-related molecular mechanisms in various types of 3D cell models. In this study, we constructed 3D scaffold-free cell models and 3D scaffold-based cell models of pancreatic cancer, evaluated chemotherapeutic drug responses in different 3D models, assessed clinical relevance of the models, and investigated molecular mechanisms of chemoresistance and drug pathways in different 3D models. Both types of 3D models showed resistance to chemotherapeutic drugs, and scaffold-based pancreatic cancer models could better reflect in vivo drug efficacy than 2D and scaffold-free pancreatic cancer models did. Increased cell adhesion, extracellular matrix (ECM) synthesis and drug transport were essential for drug resistance in 3D models, and anti-apoptosis might contribute to extreme chemoresistance in scaffold-free models. Moreover, scaffold-based pancreatic cancer models were more suitable than scaffold-free models for drug pathway research.
Collapse
Affiliation(s)
- Dafei Xie
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000 China
- Department of General Surgery, Zhejiang Hospital, Hangzhou, 310000 China
| | - Shengnan Jia
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000 China
| | - Dongnan Ping
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000 China
| | - Dong Wang
- Department of General Surgery, Zhejiang Hospital, Hangzhou, 310000 China
| | - Liping Cao
- Department of General Surgery, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, 310000 China
| |
Collapse
|
16
|
Grilli F, Pitton M, Altomare L, Farè S. Decellularized fennel and dill leaves as possible 3D channel network in GelMA for the development of an in vitro adipose tissue model. Front Bioeng Biotechnol 2022; 10:984805. [DOI: 10.3389/fbioe.2022.984805] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2022] [Accepted: 10/17/2022] [Indexed: 11/13/2022] Open
Abstract
The development of 3D scaffold-based models would represent a great step forward in cancer research, offering the possibility of predicting the potential in vivo response to targeted anticancer or anti-angiogenic therapies. As regards, 3D in vitro models require proper materials, which faithfully recapitulated extracellular matrix (ECM) properties, adequate cell lines, and an efficient vascular network. The aim of this work is to investigate the possible realization of an in vitro 3D scaffold-based model of adipose tissue, by incorporating decellularized 3D plant structures within the scaffold. In particular, in order to obtain an adipose matrix capable of mimicking the composition of the adipose tissue, methacrylated gelatin (GelMA), UV photo-crosslinkable, was selected. Decellularized fennel, wild fennel and, dill leaves have been incorporated into the GelMA hydrogel before crosslinking, to mimic a 3D channel network. All leaves showed a loss of pigmentation after the decellularization with channel dimensions ranging from 100 to 500 µm up to 3 μm, comparable with those of human microcirculation (5–10 µm). The photo-crosslinking process was not affected by the embedded plant structures in GelMA hydrogels. In fact, the weight variation test, performed on hydrogels with or without decellularized leaves showed a weight loss in the first 96 h, followed by a stability plateau up to 5 weeks. No cytotoxic effects were detected comparing the three prepared GelMA/D-leaf structures; moreover, the ability of the samples to stimulate differentiation of 3T3-L1 preadipocytes in mature adipocytes was investigated, and cells were able to grow and proliferate in the structure, colonizing the entire microenvironment and starting to differentiate. The developed GelMA hydrogels mimicked adipose tissue together with the incorporated plant structures seem to be an adequate solution to ensure an efficient vascular system for a 3D in vitro model. The obtained results showed the potentiality of the innovative proposed approach to mimic the tumoral microenvironment in 3D scaffold-based models.
Collapse
|
17
|
Tiwari A, Trivedi R, Lin SY. Tumor microenvironment: barrier or opportunity towards effective cancer therapy. J Biomed Sci 2022; 29:83. [PMID: 36253762 PMCID: PMC9575280 DOI: 10.1186/s12929-022-00866-3] [Citation(s) in RCA: 120] [Impact Index Per Article: 60.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 10/01/2022] [Indexed: 12/24/2022] Open
Abstract
Tumor microenvironment (TME) is a specialized ecosystem of host components, designed by tumor cells for successful development and metastasis of tumor. With the advent of 3D culture and advanced bioinformatic methodologies, it is now possible to study TME’s individual components and their interplay at higher resolution. Deeper understanding of the immune cell’s diversity, stromal constituents, repertoire profiling, neoantigen prediction of TMEs has provided the opportunity to explore the spatial and temporal regulation of immune therapeutic interventions. The variation of TME composition among patients plays an important role in determining responders and non-responders towards cancer immunotherapy. Therefore, there could be a possibility of reprogramming of TME components to overcome the widely prevailing issue of immunotherapeutic resistance. The focus of the present review is to understand the complexity of TME and comprehending future perspective of its components as potential therapeutic targets. The later part of the review describes the sophisticated 3D models emerging as valuable means to study TME components and an extensive account of advanced bioinformatic tools to profile TME components and predict neoantigens. Overall, this review provides a comprehensive account of the current knowledge available to target TME.
Collapse
Affiliation(s)
- Aadhya Tiwari
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| | - Rakesh Trivedi
- Department of Translational Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Shiaw-Yih Lin
- Department of Systems Biology, The University of Texas MD Anderson Cancer Center, Houston, TX, USA.
| |
Collapse
|
18
|
Wu ST, Chen PC, Tseng YH, Chen TH, Wang YJ, Tsai ZL, Lin EC. Assessment of cellular responses in three-dimensional cell cultures through chemical exchange saturation transfer and 1 H MRS. NMR IN BIOMEDICINE 2022; 35:e4757. [PMID: 35510307 DOI: 10.1002/nbm.4757] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 04/21/2022] [Accepted: 05/02/2022] [Indexed: 06/14/2023]
Abstract
Metabolic responses to physiological changes have been detected using chemical exchange saturation transfer (CEST) imaging in clinical settings. Similarly to other MRI techniques, the CEST technique was based originally on phantoms from buffer solutions and was then further developed through animal experiments. However, CEST imaging can capture certain dynamics of metabolism that solution phantoms cannot model. Cell culture phantoms can fill the gap between buffer phantoms and animal models. In this study, we used 1 H NMR and CEST in a B0 field of 9.4 T to investigate HEK293T cells from two-dimensional (2D) cultures, three-dimensional (3D) cultures, and 3D cultures seeded with cell spheroids. Two CEST dips were observed: the magnitude of the amine dip at 2.8 ppm increased during the incubation period, whereas the hydroxyl dip at 1.2 ppm remained approximately the same or modestly increased. We also observed a CEST dip at 2.8 ppm from the 2D culture responding dramatically to doxorubicin treatment. By cross-validating with pH values and the concentrations of amine and hydroxyl protons extracted through 1 H NMR, we observed that they did not correspond to an increase in the amine pool. We believe that the denaturation or degradation of proteins from the fetal bovine serum increased the size of the amine pool. Although 3D culture conditions can be further improved, our study suggests that 3D cultures have the potential to bridge studies of solution phantoms and those on animals.
Collapse
Affiliation(s)
- Ssu-Ting Wu
- Department of Chemistry and Biochemistry, National Chung Cheng University, Chiayi, Taiwan
| | - Pin-Chen Chen
- Department of Chemistry and Biochemistry, National Chung Cheng University, Chiayi, Taiwan
| | - Yu-Hsien Tseng
- Department of Chemistry and Biochemistry, National Chung Cheng University, Chiayi, Taiwan
| | - Ting-Hao Chen
- Department of Chemistry and Biochemistry, National Chung Cheng University, Chiayi, Taiwan
| | - Yi-Jiun Wang
- Department of Chemistry and Biochemistry, National Chung Cheng University, Chiayi, Taiwan
| | - Zong-Lin Tsai
- Department of Chemistry and Biochemistry, National Chung Cheng University, Chiayi, Taiwan
| | - Eugene C Lin
- Department of Chemistry and Biochemistry, National Chung Cheng University, Chiayi, Taiwan
- Center for Nano Bio-detection, National Chung Cheng University, Chiayi, Taiwan
| |
Collapse
|
19
|
A Tumor Accelerator Based on Multicomponent Bone Scaffolds and Cancer Cell Homing. Polymers (Basel) 2022; 14:polym14163340. [PMID: 36015599 PMCID: PMC9416103 DOI: 10.3390/polym14163340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2022] [Revised: 08/15/2022] [Accepted: 08/15/2022] [Indexed: 11/17/2022] Open
Abstract
Bone tissue attracts cancer cell homing biologically, mechanically, or chemically. It is difficult and time consuming to identify their complex cross-talk using existed methods. In this study, a multi-component bone matrix was fabricated using gelatin, hydroxyapatite (HAp), and epidermal growth factor (EGF) as raw materials to investigate how “acellular” bone matrix affects cancer cell homing in bone. Then, EGF-responsive cancer cells were cultured with the scaffold in a dynamical bioreactor. For different culture periods, the effects of HAp, gelatin, and EGF on the cell adhesion, proliferation, 3D growth, and migration of cancer were evaluated. The results indicated that a small amount of calcium ion released from the scaffolds accelerated cancer MDA-MB-231 adhesion on the surface of inner pores. Moreover, degradable gelatin key caused cancer cell growth on the scaffold surface to turn into a 3D aggregation. Despite this, the formation of cancer spheroids was slow, and required 14 days of dynamic culture. Thankfully, EGF promoted cancer cell adhesion, proliferation, and migration, and cancer spheroids were observed only after 3-day culture. We concluded that the combination of the multiple components in this scaffold allows cancer cells to meet multiple requirements of cancer dynamic progression.
Collapse
|
20
|
Salinas-Vera YM, Valdés J, Pérez-Navarro Y, Mandujano-Lazaro G, Marchat LA, Ramos-Payán R, Nuñez-Olvera SI, Pérez-Plascencia C, López-Camarillo C. Three-Dimensional 3D Culture Models in Gynecological and Breast Cancer Research. Front Oncol 2022; 12:826113. [PMID: 35692756 PMCID: PMC9177953 DOI: 10.3389/fonc.2022.826113] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Accepted: 04/20/2022] [Indexed: 12/12/2022] Open
Abstract
Traditional two-dimensional (2D) monolayer cell cultures have long been the gold standard for cancer biology research. However, their ability to accurately reflect the molecular mechanisms of tumors occurring in vivo is limited. Recent development of three-dimensional (3D) cell culture models facilitate the possibility to better recapitulate several of the biological and molecular characteristics of tumors in vivo, such as cancer cells heterogeneity, cell-extracellular matrix interactions, development of a hypoxic microenvironment, signaling pathway activities depending on contacts with extracellular matrix, differential growth kinetics, more accurate drugs response, and specific gene expression and epigenetic patterns. In this review, we discuss the utilization of different types of 3D culture models including spheroids, organotypic models and patient-derived organoids in gynecologic cancers research, as well as its potential applications in oncological research mainly for screening drugs with major physiological and clinical relevance. Moreover, microRNAs regulation of cancer hallmarks in 3D cell cultures from different types of cancers is discussed.
Collapse
Affiliation(s)
- Yarely M. Salinas-Vera
- Departamento de Bioquímica, Centro de Investigación de Estudios Avanzados (CINVESTAV-IPN), Ciudad de Mexico, Mexico
| | - Jesús Valdés
- Departamento de Bioquímica, Centro de Investigación de Estudios Avanzados (CINVESTAV-IPN), Ciudad de Mexico, Mexico
| | - Yussel Pérez-Navarro
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de Mexico, Ciudad de Mexico, Mexico
| | - Gilberto Mandujano-Lazaro
- Programa en Biomedicina Molecular y Red de Biotecnología, Instituto Politécnico Nacional, Ciudad de Mexico, Mexico
| | - Laurence A. Marchat
- Programa en Biomedicina Molecular y Red de Biotecnología, Instituto Politécnico Nacional, Ciudad de Mexico, Mexico
| | - Rosalio Ramos-Payán
- Facultad de Ciencias Químico Biológicas, Universidad Autónoma de Sinaloa, Culiacán Sinaloa, Mexico
| | - Stephanie I. Nuñez-Olvera
- Departamento de Biología Celular y Fisiología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Mexico
| | | | - César López-Camarillo
- Posgrado en Ciencias Genómicas, Universidad Autónoma de la Ciudad de Mexico, Ciudad de Mexico, Mexico
- *Correspondence: César López-Camarillo, ; orcid.org/0000-0002-9417-2609
| |
Collapse
|
21
|
Precision Medicine in Head and Neck Cancers: Genomic and Preclinical Approaches. J Pers Med 2022; 12:jpm12060854. [PMID: 35743639 PMCID: PMC9224778 DOI: 10.3390/jpm12060854] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 05/11/2022] [Accepted: 05/19/2022] [Indexed: 02/07/2023] Open
Abstract
Head and neck cancers (HNCs) represent the sixth most widespread malignancy worldwide. Surgery, radiotherapy, chemotherapeutic and immunotherapeutic drugs represent the main clinical approaches for HNC patients. Moreover, HNCs are characterised by an elevated mutational load; however, specific genetic mutations or biomarkers have not yet been found. In this scenario, personalised medicine is showing its efficacy. To study the reliability and the effects of personalised treatments, preclinical research can take advantage of next-generation sequencing and innovative technologies that have been developed to obtain genomic and multi-omic profiles to drive personalised treatments. The crosstalk between malignant and healthy components, as well as interactions with extracellular matrices, are important features which are responsible for treatment failure. Preclinical research has constantly implemented in vitro and in vivo models to mimic the natural tumour microenvironment. Among them, 3D systems have been developed to reproduce the tumour mass architecture, such as biomimetic scaffolds and organoids. In addition, in vivo models have been changed over the last decades to overcome problems such as animal management complexity and time-consuming experiments. In this review, we will explore the new approaches aimed to improve preclinical tools to study and apply precision medicine as a therapeutic option for patients affected by HNCs.
Collapse
|
22
|
Chinga-Carrasco G, Rosendahl J, Catalán J. Nanocelluloses - Nanotoxicology, Safety Aspects and 3D Bioprinting. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1357:155-177. [PMID: 35583644 DOI: 10.1007/978-3-030-88071-2_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Nanocelluloses have good rheological properties that facilitate the extrusion of nanocellulose gels in micro-extrusion systems. It is considered a highly relevant characteristic that makes it possible to use nanocellulose as an ink component for 3D bioprinting purposes. The nanocelluloses assessed in this book chapter include wood nanocellulose (WNC), bacterial nanocellulose (BNC), and tunicate nanocellulose (TNC), which are often assumed to be non-toxic. Depending on various chemical and mechanical processes, both cellulose nanofibrils (CNF) and cellulose nanocrystals (CNC) can be obtained from the three mentioned nanocelluloses (WNC, BNC, and TNC). Pre/post-treatment processes (chemical and mechanical) cause modifications regarding surface chemistry and nano-morphology. Hence, it is essential to understand whether physicochemical properties may affect the toxicological profile of nanocelluloses. In this book chapter, we provide an overview of nanotoxicology and safety aspects associated with nanocelluloses. Relevant regulatory requirements are considered. We also discuss hazard assessment strategies based on tiered approaches for safety testing, which can be applied in the early stages of the innovation process. Ensuring the safe development of nanocellulose-based 3D bioprinting products will enable full market use of these sustainable resources throughout their life cycle.
Collapse
Affiliation(s)
| | - Jennifer Rosendahl
- RISE, Division Materials and Production, Department Chemistry, Biomaterials and Textiles, Section Biological Function, Borås, Sweden
| | - Julia Catalán
- Occupational Safety, Finnish Institute of Occupational Health, Helsinki, Finland
- Department of Anatomy, Embryology and Genetics, University of Zaragoza, Zaragoza, Spain
| |
Collapse
|
23
|
Kato-Negishi M, Sawayama J, Kawahara M, Takeuchi S. Cell fiber-based 3D tissue array for drug response assay. Sci Rep 2022; 12:7870. [PMID: 35552465 PMCID: PMC9098497 DOI: 10.1038/s41598-022-11670-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2021] [Accepted: 04/22/2022] [Indexed: 11/17/2022] Open
Abstract
For the establishment of a reproducible and sensitive assay system for three-dimensional (3D) tissue-based drug screening, it is essential to develop 3D tissue arrays with uniform shapes and high cell numbers that prevent cell death in the center of the tissue. In recent years, 3D tissue arrays based on spheroids have attracted increased attention. However, they have only been used in specific tissues with hypoxic regions, such as cancer tissues, because nutrient deprivation and hypoxic regions are formed in the core as spheroids grow. Herein, we propose a method to array cell-encapsulated tube-like tissue (cell fiber (CF)) with diameters < 150 μm to prevent nutrient deprivation and hypoxia using a device that can fix the CFs, section them in uniform sizes, and transfer them to a 96-well plate. We fabricated the arrays of CF fragments from cell lines (GT1-7), cancer cells (HeLa), mouse neural stem cells (mNSCs) and differentiated mNSCs, and performed drug response assays. The array of CF fragments assessed the drug response differences among different cell types and drug responses specific to 3D tissues. The array of CF fragments may be used as a versatile drug screening system to detect drug sensitivities in various types of tissues.
Collapse
Affiliation(s)
- Midori Kato-Negishi
- Department of Bio-Analytical Chemistry, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, 1-1-20 Shinmachi, Nishitokyo, Tokyo, 202-8585, Japan
| | - Jun Sawayama
- Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan
| | - Masahiro Kawahara
- Department of Bio-Analytical Chemistry, Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, 1-1-20 Shinmachi, Nishitokyo, Tokyo, 202-8585, Japan
| | - Shoji Takeuchi
- Institute of Industrial Science, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8505, Japan. .,Department of Mechano-Informatics, Graduate School of Information Science and Technology, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan. .,International Research Center for Neurointelligence (WPI-IRCN), The University of Tokyo Institutes for Advanced Study (UTIAS), The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, 113-8656, Japan.
| |
Collapse
|
24
|
Models of Head and Neck Squamous Cell Carcinoma Using Bioengineering Approaches. Crit Rev Oncol Hematol 2022; 175:103724. [DOI: 10.1016/j.critrevonc.2022.103724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2022] [Revised: 04/24/2022] [Accepted: 05/18/2022] [Indexed: 11/21/2022] Open
|
25
|
Ertekin Ö, Monavari M, Krüger R, Fuentes-Chandía M, Parma B, Letort G, Tripal P, Boccaccini AR, Bosserhoff AK, Ceppi P, Kappelmann-Fenzl M, Leal-Egaña A. 3D hydrogel-based microcapsules as an in vitro model to study tumorigenicity, cell migration and drug resistance. Acta Biomater 2022; 142:208-220. [PMID: 35167953 DOI: 10.1016/j.actbio.2022.02.010] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/01/2021] [Revised: 02/06/2022] [Accepted: 02/09/2022] [Indexed: 02/06/2023]
Abstract
In this work, we analyzed the reliability of alginate-gelatin microcapsules as artificial tumor model. These tumor-like scaffolds are characterized by their composition and stiffness (∼25 kPa), and their capability to restrict -but not hinder- cell migration, proliferation and release from confinement. Hydrogel-based microcapsules were initially utilized to detect differences in mechano-sensitivity between MCF7 and MDA-MB-231 breast cancer cells, and the endothelial cell line EA.hy926. Additionally, we used RNA-seq and transcriptomic methods to determine how the culture strategy (i.e. 2D v/s 3D) may pre-set the expression of genes involved in multidrug resistance, being then validated by performing cytotoxicological tests and assays of cell morphology. Our results show that both breast cancer cells can generate elongated multicellular spheroids inside the microcapsules, prior being released (mimicking intravasation stages), a behavior which was not observed in endothelial cells. Further, we demonstrate that cells isolated from 3D scaffolds show resistance to cisplatin, a process which seems to be strongly influenced by mechanical stress, instead of hypoxia. We finally discuss the role played by aneuploidy in malignancy and resistance to anticancer drugs, based on the increased number of polynucleated cells found within these microcapsules. Overall, our outcomes demonstrate that alginate-gelatin microcapsules represent a simple, yet very accurate tumor-like model, enabling us to mimic the most relevant malignant hints described in vivo, suggesting that confinement and mechanical stress need to be considered when studying pathogenicity and drug resistance of cancer cells in vitro. STATEMENT OF SIGNIFICANCE: In this work, we analyzed the reliability of alginate-gelatin microcapsules as an artificial tumor model. These scaffolds are characterized by their composition, elastic properties, and their ability to restrict cell migration, proliferation, and release from confinement. Our results demonstrate four novel outcomes: (i) studying cell migration and proliferation in 3D enabled discrimination between malignant and non-pathogenic cells, (ii) studying the cell morphology of cancer aggregates entrapped in alginate-gelatin microcapsules enabled determination of malignancy degree in vitro, (iii) determination that confinement and mechanical stress, instead of hypoxia, are required to generate clones resistant to anticancer drugs (i.e. cisplatin), and (iv) evidence that resistance to anticancer drugs could be due to the presence of polynucleated cells localized inside polymer-based artificial tumors.
Collapse
Affiliation(s)
- Özlem Ertekin
- Institute of Biomaterials, Friedrich-Alexander University Erlangen-Nürnberg, Cauerstraße 6, Erlangen 91058, Germany; Diagno Biotechnology, Marmara Technopark, Gebze, Kocaeli, Turkey
| | - Mahshid Monavari
- Institute of Biomaterials, Friedrich-Alexander University Erlangen-Nürnberg, Cauerstraße 6, Erlangen 91058, Germany; Department of Pharmaceutical Biomaterials and Medical Biomaterials Research Centre, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran 1417614411, Iran
| | - René Krüger
- Department of Nephrology and Hypertension, Friedrich-Alexander University Erlangen-Nürnberg, and University Clinics Erlangen, Erlangen 91054, Germany
| | - Miguel Fuentes-Chandía
- Institute of Biomaterials, Friedrich-Alexander University Erlangen-Nürnberg, Cauerstraße 6, Erlangen 91058, Germany; Department of Biology, Skeletal Research Center, Case Western Reserve University, Cleveland, OH, USA
| | - Beatrice Parma
- Interdisciplinary Center for Clinical Research (IZKF), Friedrich-Alexander Universität Erlangen-Nürnberg Glueckstrasse 6, Erlangen 91054, Germany
| | - Gaelle Letort
- Center for Interdisciplinary Research in Biology, Collège de France UMR7241/U1050, 11, Place Marcelin Berthelot, Paris 75231 CEDEX 05, France
| | - Philipp Tripal
- Optical Imaging Centre Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Cauerstraße 3, Erlangen 91058, Germany
| | - Aldo R Boccaccini
- Institute of Biomaterials, Friedrich-Alexander University Erlangen-Nürnberg, Cauerstraße 6, Erlangen 91058, Germany
| | - Anja K Bosserhoff
- Institute of Biochemistry, Emil-Fischer-Zentrum, Friedrich-Alexander Universität Erlangen-Nürnberg, Fahrstraße 17, Erlangen 91054, Germany
| | - Paolo Ceppi
- Interdisciplinary Center for Clinical Research (IZKF), Friedrich-Alexander Universität Erlangen-Nürnberg Glueckstrasse 6, Erlangen 91054, Germany; Department of Biochemistry and Molecular Biology, University of Southern Denmark, Campusvej 55, Odense DK-5230, Denmark
| | - Melanie Kappelmann-Fenzl
- Institute of Biochemistry, Emil-Fischer-Zentrum, Friedrich-Alexander Universität Erlangen-Nürnberg, Fahrstraße 17, Erlangen 91054, Germany; Faculty of Applied Informatics, University of Applied Science Deggendorf, Deggendorf 94469, Germany
| | - Aldo Leal-Egaña
- Institute of Biomaterials, Friedrich-Alexander University Erlangen-Nürnberg, Cauerstraße 6, Erlangen 91058, Germany; Institute for Molecular Systems Engineering, University of Heidelberg. INF 253, Heidelberg 69120, Germany.
| |
Collapse
|
26
|
Hakim M, Kermanshah L, Abouali H, Hashemi HM, Yari A, Khorasheh F, Alemzadeh I, Vossoughi M. Unraveling Cancer Metastatic Cascade Using Microfluidics-based Technologies. Biophys Rev 2022; 14:517-543. [PMID: 35528034 PMCID: PMC9043145 DOI: 10.1007/s12551-022-00944-8] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 03/14/2022] [Indexed: 12/24/2022] Open
Abstract
Cancer has long been a leading cause of death. The primary tumor, however, is not the main cause of death in more than 90% of cases. It is the complex process of metastasis that makes cancer deadly. The invasion metastasis cascade is the multi-step biological process of cancer cell dissemination to distant organ sites and adaptation to the new microenvironment site. Unraveling the metastasis process can provide great insight into cancer death prevention or even treatment. Microfluidics is a promising platform, that provides a wide range of applications in metastasis-related investigations. Cell culture microfluidic technologies for in vitro modeling of cancer tissues with fluid flow and the presence of mechanical factors have led to the organ-on-a-chip platforms. Moreover, microfluidic systems have also been exploited for capturing and characterization of circulating tumor cells (CTCs) that provide crucial information on the metastatic behavior of a tumor. We present a comprehensive review of the recent developments in the application of microfluidics-based systems for analysis and understanding of the metastasis cascade from a wider perspective.
Collapse
Affiliation(s)
- Maziar Hakim
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Leyla Kermanshah
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Hesam Abouali
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Hanieh Mohammad Hashemi
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Alireza Yari
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Farhad Khorasheh
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Iran Alemzadeh
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| | - Manouchehr Vossoughi
- Department of Chemical and Petroleum Engineering, Sharif University of Technology, Tehran, Iran
| |
Collapse
|
27
|
Head T, Cady NC. Monitoring and modulation of the tumor microenvironment for enhanced cancer modeling. Exp Biol Med (Maywood) 2022; 247:598-613. [PMID: 35088603 PMCID: PMC9014523 DOI: 10.1177/15353702221074293] [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] [Indexed: 11/16/2022] Open
Abstract
Cancer treatments utilizing biologic or cytotoxic drugs compose the frontline of therapy, and though gains in treatment efficacy have been persistent in recent decades, much work remains in understanding cancer progression and treatment. Compounding this situation is the low rate of success when translating preclinical drug candidates to the clinic, which raises costs and development timelines. This underperformance is due in part to the poor recapitulation of the tumor microenvironment, a critical component of cancer biology, in cancer model systems. New technologies capable of both accurately observing and manipulating the tumor microenvironment are needed to effectively model cancer response to treatment. In this review, conventional cancer models are summarized, and a primer on emerging techniques for monitoring and modulating the tumor microenvironment is presented and discussed.
Collapse
Affiliation(s)
- Tristen Head
- College of Nanoscale Science & Engineering,
State University of New York Polytechnic Institute, Albany, NY 12203, USA
| | - Nathaniel C Cady
- College of Nanoscale Science & Engineering,
State University of New York Polytechnic Institute, Albany, NY 12203, USA
| |
Collapse
|
28
|
Weigel T, Malkmus C, Weigel V, Wußmann M, Berger C, Brennecke J, Groeber-Becker F, Hansmann J. Fully Synthetic 3D Fibrous Scaffolds for Stromal Tissues-Replacement of Animal-Derived Scaffold Materials Demonstrated by Multilayered Skin. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2106780. [PMID: 34933407 DOI: 10.1002/adma.202106780] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 12/14/2021] [Indexed: 06/14/2023]
Abstract
The extracellular matrix (ECM) of soft tissues in vivo has remarkable biological and structural properties. Thereby, the ECM provides mechanical stability while it still can be rearranged via cellular remodeling during tissue maturation or healing processes. However, modern synthetic alternatives fail to provide these key features among basic properties. Synthetic matrices are usually completely degraded or are inert regarding cellular remodeling. Based on a refined electrospinning process, a method is developed to generate synthetic scaffolds with highly porous fibrous structures and enhanced fiber-to-fiber distances. Since this approach allows for cell migration, matrix remodeling, and ECM synthesis, the scaffold provides an ideal platform for the generation of soft tissue equivalents. Using this matrix, an electrospun-based multilayered skin equivalent composed of a stratified epidermis, a dermal compartment, and a subcutis is able to be generated without the use of animal matrix components. The extension of classical dense electrospun scaffolds with high porosities and motile fibers generates a fully synthetic and defined alternative to collagen-gel-based tissue models and is a promising system for the construction of tissue equivalents as in vitro models or in vivo implants.
Collapse
Affiliation(s)
- Tobias Weigel
- Translational Center for Regenerative Therapies (TLC-RT), Fraunhofer Institute for Silicate Research (ISC), 97082, Würzburg, Germany
- Department for Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, 97070, Würzburg, Germany
| | - Christoph Malkmus
- Department for Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, 97070, Würzburg, Germany
| | - Verena Weigel
- Translational Center for Regenerative Therapies (TLC-RT), Fraunhofer Institute for Silicate Research (ISC), 97082, Würzburg, Germany
- Department for Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, 97070, Würzburg, Germany
| | - Maximiliane Wußmann
- Translational Center for Regenerative Therapies (TLC-RT), Fraunhofer Institute for Silicate Research (ISC), 97082, Würzburg, Germany
| | - Constantin Berger
- Department for Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, 97070, Würzburg, Germany
| | - Julian Brennecke
- Department for Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, 97070, Würzburg, Germany
| | - Florian Groeber-Becker
- Translational Center for Regenerative Therapies (TLC-RT), Fraunhofer Institute for Silicate Research (ISC), 97082, Würzburg, Germany
- Department for Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, 97070, Würzburg, Germany
| | - Jan Hansmann
- Department for Tissue Engineering and Regenerative Medicine, University Hospital Würzburg, 97070, Würzburg, Germany
- Faculty of Electrical Engineering, University of Applied Sciences Würzburg-Schweinfurt, 97421, Schweinfurt, Germany
| |
Collapse
|
29
|
Fabiano E, Zhang J, Reinhart-King C. Tissue density in the progression of breast cancer: Bedside to bench and back again. CURRENT OPINION IN BIOMEDICAL ENGINEERING 2022; 22. [DOI: 10.1016/j.cobme.2022.100383] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
30
|
Wan J, Wu T, Liu Y, Yang M, Fichna J, Guo Y, Yin L, Chen C. Mast Cells Tryptase Promotes Intestinal Fibrosis in Natural Decellularized Intestinal Scaffolds. Tissue Eng Regen Med 2022; 19:717-726. [PMID: 35218507 PMCID: PMC9294124 DOI: 10.1007/s13770-022-00433-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 12/18/2021] [Accepted: 01/08/2022] [Indexed: 11/24/2022] Open
Abstract
BACKGROUND Standard two-dimensional (2D) culture has confirmed the mechanism of mast cells (MCs) in the pathogenesis of inflammatory bowel disease (IBD), but the regulation of signaling responses of MCs may well differ in three-dimensional (3D) microenvironments. The aim of the study was to develop a 3D culture model based on decellularized intestinal scaffolds (DIS) and verify how MCs influenced fibroblasts phenotype in the 3D model. METHODS DIS were achieved using the detergent technique and extracellular matrix (ECM) components were verified by histologic analysis, quantification and scanning electron microscope. After human colon fibroblasts recellularized into the scaffolds and activated by MCs tryptase and TGFβ1, the changes in genes and signaling pathways during fibroblasts activation in 3D were studied and compared with the changes in 2D cell culture on plastic plates. RESULTS Decellularization process effectively removed native cell debris while retaining natural ECM components and structure. The engrafted fibroblasts could penetrate into the scaffolds and maintain its phenotype. No matter whether fibroblasts were cultured in 2D or 3D, MCs tryptase and transforming growth factor β1 (TGF-β1) could promote the differentiation of fibroblasts into fibrotic-phenotype myofibroblasts through Akt and Smad2/3 signaling pathways. Furthermore, the pro-collagen1α1 and fibronectin synthesis of myofibroblasts in 3D was higher than in 2D culture. CONCLUSION Our results demonstrated that the DIS can be used as a bioactive microenvironment for the study of intestinal fibrosis, providing an innovative platform for future intestinal disease modeling and screening of genes and signaling pathways.
Collapse
Affiliation(s)
- Jian Wan
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 China
| | - Tianqi Wu
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 China
| | - Ying Liu
- Department of General Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 China
| | - Muqing Yang
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People’s Hospital, Tongji University School of Medicine, Shanghai, 200072 China
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 6/8, 92-215 Lodz, Poland
| | - Yibing Guo
- Research Center of Clinical Medicine, Affiliated Hospital of Nantong University, Nantong, 226000 China
| | - Lu Yin
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| | - Chunqiu Chen
- Center for Difficult and Complicated Abdominal Surgery, Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai, 200072, China.
| |
Collapse
|
31
|
Das R, Fernandez JG. Biomaterials for Mimicking and Modelling Tumor Microenvironment. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1379:139-170. [DOI: 10.1007/978-3-031-04039-9_6] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
32
|
Rezaeeyazdi M, Colombani T, Eggermont LJ, Bencherif SA. Engineering hyaluronic acid-based cryogels for CD44-mediated breast tumor reconstruction. Mater Today Bio 2022; 13:100207. [PMID: 35198956 PMCID: PMC8844817 DOI: 10.1016/j.mtbio.2022.100207] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 01/19/2022] [Accepted: 01/22/2022] [Indexed: 01/03/2023] Open
Abstract
Breast cancer is a major health concern worldwide and is the leading cause of cancer-related death among American women. Traditional therapies, such as surgery, chemotherapy, and radiotherapy, are usually ineffective. Furthermore, cancer recurrence following targeted therapy often results from acquired drug resistance. Therefore, more realistic tumor models than monolayer cell culture for drug screening and discovery in an in vitro setting would facilitate the development of new therapeutic strategies. Toward this goal, we first developed a simple, rapid, low-cost, and high-throughput method for generating uniform multi-cellular tumor spheroids (MCTS) with controllable size. Next, biomimetic cryogel scaffolds fabricated from hyaluronic acid (HA) were utilized as a platform to reconstruct breast tumor microtissues with aspects of the complex tumor microenvironment in three dimensions. Finally, we investigated the interactions between the HA-based cryogels and CD44-positive breast tumor cells, individually or as MCTS. We found that incorporating the adhesive RGD peptide in cryogels led to the formation of a monolayer of tumor cells on the polymer walls, whereas MCTS cultured on RGD-free HA cryogels resulted in the growth of large and dense microtumors, more similar to native tumor masses. As a result, the MCTS-laden HA cryogel system induced a highly aggressive and chemotherapy drug-resistant tumor model. RGD-free HA-based cryogels represent an effective starting point for designing tumor models for preclinical research, therapeutic drug screening, and early cancer diagnosis.
Collapse
Affiliation(s)
| | - Thibault Colombani
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Loek J. Eggermont
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Sidi A. Bencherif
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
- Department of Bioengineering, Northeastern University, Boston, MA, 02115, USA
- Harvard John A. Paulson School of Engineering and Applied Sciences, Harvard University, Cambridge, MA, 02138, USA
| |
Collapse
|
33
|
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
|
34
|
Ravichandran A, Clegg J, Adams MN, Hampson M, Fielding A, Bray LJ. 3D Breast Tumor Models for Radiobiology Applications. Cancers (Basel) 2021; 13:5714. [PMID: 34830869 PMCID: PMC8616164 DOI: 10.3390/cancers13225714] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Revised: 10/28/2021] [Accepted: 11/07/2021] [Indexed: 12/17/2022] Open
Abstract
Breast cancer is a leading cause of cancer-associated death in women. The clinical management of breast cancers is normally carried out using a combination of chemotherapy, surgery and radiation therapy. The majority of research investigating breast cancer therapy until now has mainly utilized two-dimensional (2D) in vitro cultures or murine models of disease. However, there has been significant uptake of three-dimensional (3D) in vitro models by cancer researchers over the past decade, highlighting a complimentary model for studies of radiotherapy, especially in conjunction with chemotherapy. In this review, we underline the effects of radiation therapy on normal and malignant breast cells and tissues, and explore the emerging opportunities that pre-clinical 3D models offer in improving our understanding of this treatment modality.
Collapse
Affiliation(s)
- Akhilandeshwari Ravichandran
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; (A.R.); (J.C.); (M.H.)
- ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia;
| | - Julien Clegg
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; (A.R.); (J.C.); (M.H.)
- ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia;
| | - Mark N. Adams
- ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia;
- School of Biomedical Sciences, Queensland University of Technology, Brisbane, QLD 4000, Australia
| | - Madison Hampson
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; (A.R.); (J.C.); (M.H.)
| | - Andrew Fielding
- School of Chemistry and Physics, Queensland University of Technology, Brisbane, QLD 4000, Australia;
| | - Laura J. Bray
- School of Mechanical, Medical and Process Engineering, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia; (A.R.); (J.C.); (M.H.)
- ARC Training Centre for Cell and Tissue Engineering Technologies, Queensland University of Technology (QUT), Brisbane, QLD 4000, Australia;
| |
Collapse
|
35
|
Chick Embryo Experimental Platform for Micrometastases Research in a 3D Tissue Engineering Model: Cancer Biology, Drug Development, and Nanotechnology Applications. Biomedicines 2021; 9:biomedicines9111578. [PMID: 34829808 PMCID: PMC8615510 DOI: 10.3390/biomedicines9111578] [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: 09/05/2021] [Revised: 10/06/2021] [Accepted: 10/16/2021] [Indexed: 12/31/2022] Open
Abstract
Colonization of distant organs by tumor cells is a critical step of cancer progression. The initial avascular stage of this process (micrometastasis) remains almost inaccessible to study due to the lack of relevant experimental approaches. Herein, we introduce an in vitro/in vivo model of organ-specific micrometastases of triple-negative breast cancer (TNBC) that is fully implemented in a cost-efficient chick embryo (CE) experimental platform. The model was built as three-dimensional (3D) tissue engineering constructs (TECs) combining human MDA-MB-231 cells and decellularized CE organ-specific scaffolds. TNBC cells colonized CE organ-specific scaffolds in 2–3 weeks, forming tissue-like structures. The feasibility of this methodology for basic cancer research, drug development, and nanomedicine was demonstrated on a model of hepatic micrometastasis of TNBC. We revealed that MDA-MB-231 differentially colonize parenchymal and stromal compartments of the liver-specific extracellular matrix (LS-ECM) and become more resistant to the treatment with molecular doxorubicin (Dox) and Dox-loaded mesoporous silica nanoparticles than in monolayer cultures. When grafted on CE chorioallantoic membrane, LS-ECM-based TECs induced angiogenic switch. These findings may have important implications for the diagnosis and treatment of TNBC. The methodology established here is scalable and adaptable for pharmacological testing and cancer biology research of various metastatic and primary tumors.
Collapse
|
36
|
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
|
37
|
Berger Fridman I, Kostas J, Gregus M, Ray S, Sullivan MR, Ivanov AR, Cohen S, Konry T. High-throughput microfluidic 3D biomimetic model enabling quantitative description of the human breast tumor microenvironment. Acta Biomater 2021; 132:473-488. [PMID: 34153511 PMCID: PMC8434998 DOI: 10.1016/j.actbio.2021.06.025] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 05/31/2021] [Accepted: 06/14/2021] [Indexed: 12/15/2022]
Abstract
Cancer is driven by both genetic aberrations in the tumor cells and fundamental changes in the tumor microenvironment (TME). These changes offer potential targets for novel therapeutics, yet lack of in vitro 3D models recapitulating this complex microenvironment impedes such progress. Here, we generated several tumor-stroma scaffolds reflecting the dynamic in vivo breast TME, using a high throughput microfluidic system. Alginate (Alg) or alginate-alginate sulfate (Alg/Alg-S) hydrogels were used as ECM-mimics, enabling the encapsulation and culture of tumor cells, fibroblasts and immune cells (macrophages and T cells, of the innate and adaptive immune systems, respectively). Specifically, Alg/Alg-S was shown capable of capturing and presenting growth factors and cytokines with binding affinity that is comparable to heparin. Viability and cytotoxicity were shown to strongly correlate with the dynamics of cellular milieu, as well as hydrogel type. Using on-chip immunofluorescence, production of reactive oxygen species and apoptosis were imaged and quantitatively analyzed. We then show how macrophages in our microfluidic system were shifted from a proinflammatory to an immunosuppressive phenotype when encapsulated in Alg/Alg-S, reflecting in vivo TME dynamics. LC-MS proteomic profiling of tumor cells sorted from the TME scaffolds revealed upregulation of proteins involved in cell-cell interactions and immunomodulation in Alg/Alg-S scaffolds, correlating with in vivo findings and demonstrating the appropriateness of Alg/Alg-S as an ECM biomimetic. Finally, we show the formation of large tumor-derived vesicles, formed exclusively in Alg/Alg-S scaffolds. Altogether, our system offers a robust platform for quantitative description of the breast TME that successfully recapitulates in vivo patterns. STATEMENT OF SIGNIFICANCE: Cancer progression is driven by profound changes in both tumor cells and surrounding stroma. Here, we present a high throughput microfluidic system for the generation and analysis of dynamic tumor-stroma scaffolds, that mimic the complex in vivo TME cell proportions and compositions, constructing robust in vitro models for the study of the TME. Utilizing Alg/Alg-S as a bioinspired ECM, mimicking heparin's in vivo capabilities of capturing and presenting signaling molecules, we show how Alg/Alg-S induces complex in vivo-like responses in our models. Alg/Alg-S is shown here to promote dynamic protein expression patterns, that can serve as potential therapeutic targets for breast cancer treatment. Formation of large tumor-derived vesicles, observed exclusively in the Alg/Alg-S scaffolds suggests a mechanism for tumor survival.
Collapse
Affiliation(s)
- Ilana Berger Fridman
- Department of Pharmaceutical Sciences, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA; Avram and Stella Goldstein-Goren Department of Biotechnology Engineering and Regenerative Medicine and Stem Cell Center, Ben-Gurion University of the Negev, POB 653, Beer-Sheva 84105, Israel.
| | - James Kostas
- Department of Chemistry and Chemical Biology, Barnett Institute of Chemical and Biological Analysis, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA.
| | - Michal Gregus
- Department of Chemistry and Chemical Biology, Barnett Institute of Chemical and Biological Analysis, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA.
| | - Somak Ray
- Department of Chemistry and Chemical Biology, Barnett Institute of Chemical and Biological Analysis, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA.
| | - Matthew R Sullivan
- Department of Pharmaceutical Sciences, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA.
| | - Alexander R Ivanov
- Department of Chemistry and Chemical Biology, Barnett Institute of Chemical and Biological Analysis, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA.
| | - Smadar Cohen
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering and Regenerative Medicine and Stem Cell Center, Ben-Gurion University of the Negev, POB 653, Beer-Sheva 84105, Israel.
| | - Tania Konry
- Department of Pharmaceutical Sciences, Northeastern University, 360 Huntington Avenue, Boston, MA 02115, USA.
| |
Collapse
|
38
|
Costard LS, Hosn RR, Ramanayake H, O'Brien FJ, Curtin CM. Influences of the 3D microenvironment on cancer cell behaviour and treatment responsiveness: A recent update on lung, breast and prostate cancer models. Acta Biomater 2021; 132:360-378. [PMID: 33484910 DOI: 10.1016/j.actbio.2021.01.023] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2020] [Revised: 01/13/2021] [Accepted: 01/14/2021] [Indexed: 12/21/2022]
Abstract
The majority of in vitro studies assessing cancer treatments are performed in two-dimensional (2D) monolayers and are subsequently validated in in vivo animal models. However, 2D models fail to accurately model the tumour microenvironment. Furthermore, animal models are not directly applicable to mimic the human scenario. Three-dimensional (3D) culture models may help to address the discrepancies of 2D and animal models. When cancer cells escape the primary tumour, they can invade at distant organs building secondary tumours, called metastasis. The development of metastasis leads to a dramatic decrease in the life expectancy of patients. Therefore, 3D systems to model the microenvironment of metastasis have also been developed. Several studies have demonstrated changes in cell behaviour and gene expression when cells are cultured in 3D compared to 2D and concluded a better comparability to cells in vivo. Of special importance is the effect seen in response to anti-cancer treatments as models are built primarily to serve as drug-testing platforms. This review highlights these changes between cancer cells grown in 2D and 3D models for some of the most common cancers including lung, breast and prostate tumours. In addition to models aiming to mimic the primary tumour site, the effects of 3D cell culturing in bone metastasis models are also described. STATEMENT OF SIGNIFICANCE: Most in vitro studies in cancer research are performed in 2D and are subsequently validated in in vivo animal models. However, both models possess numerous limitations: 2D models fail to accurately model the tumour microenvironment while animal models are expensive, time-consuming and can differ considerably from humans. It is accepted that the cancer microenvironment plays a critical role in the disease, thus, 3D models have been proposed as a potential solution to address the discrepancies of 2D and animal models. This review highlights changes in cell behaviour, including proliferation, gene expression and chemosensitivity, between cancer cells grown in 2D and 3D models for some of the most common cancers including lung, breast and prostate cancer as well as bone metastasis.
Collapse
|
39
|
Hot or cold: Bioengineering immune contextures into in vitro patient-derived tumor models. Adv Drug Deliv Rev 2021; 175:113791. [PMID: 33965462 DOI: 10.1016/j.addr.2021.05.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/02/2021] [Accepted: 05/04/2021] [Indexed: 02/06/2023]
Abstract
In the past decade, immune checkpoint inhibitors (ICI) have proven to be tremendously effective for a subset of cancer patients. However, it is difficult to predict the response of individual patients and efforts are now directed at understanding the mechanisms of ICI resistance. Current models of patient tumors poorly recapitulate the immune contexture, which describe immune parameters that are associated with patient survival. In this Review, we discuss parameters that influence the induction of different immune contextures found within tumors and how engineering strategies may be leveraged to recapitulate these contextures to develop the next generation of immune-competent patient-derived in vitro models.
Collapse
|
40
|
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: 7.0] [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
|
41
|
Lobo DA, Ginestra P, Ceretti E, Miquel TP, Ciurana J. Cancer Cell Direct Bioprinting: A Focused Review. MICROMACHINES 2021; 12:764. [PMID: 34203530 PMCID: PMC8305105 DOI: 10.3390/mi12070764] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/23/2021] [Accepted: 06/25/2021] [Indexed: 12/24/2022]
Abstract
Three-dimensional printing technologies allow for the fabrication of complex parts with accurate geometry and less production time. When applied to biomedical applications, two different approaches, known as direct or indirect bioprinting, may be performed. The classical way is to print a support structure, the scaffold, and then culture the cells. Due to the low efficiency of this method, direct bioprinting has been proposed, with or without the use of scaffolds. Scaffolds are the most common technology to culture cells, but bioassembly of cells may be an interesting methodology to mimic the native microenvironment, the extracellular matrix, where the cells interact between themselves. The purpose of this review is to give an updated report about the materials, the bioprinting technologies, and the cells used in cancer research for breast, brain, lung, liver, reproductive, gastric, skin, and bladder associated cancers, to help the development of possible treatments to lower the mortality rates, increasing the effectiveness of guided therapies. This work introduces direct bioprinting to be considered as a key factor above the main tissue engineering technologies.
Collapse
Affiliation(s)
- David Angelats Lobo
- Department of Mechanical and Industrial Engineering, University of Brescia, V. Branze 38, 25123 Brescia, Italy; (D.A.L.); (E.C.)
- New Therapeutic Targets Laboratory (TargetsLab), Oncology Unit, Department of Medical Sciences, Girona Institute for Biomedical Research, University of Girona, Emili Grahit 77, 17003 Girona, Spain;
| | - Paola Ginestra
- Department of Mechanical and Industrial Engineering, University of Brescia, V. Branze 38, 25123 Brescia, Italy; (D.A.L.); (E.C.)
| | - Elisabetta Ceretti
- Department of Mechanical and Industrial Engineering, University of Brescia, V. Branze 38, 25123 Brescia, Italy; (D.A.L.); (E.C.)
| | - Teresa Puig Miquel
- New Therapeutic Targets Laboratory (TargetsLab), Oncology Unit, Department of Medical Sciences, Girona Institute for Biomedical Research, University of Girona, Emili Grahit 77, 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;
| |
Collapse
|
42
|
Hydroxyethyl chitosan hydrogels for enhancing breast cancer cell tumorigenesis. Int J Biol Macromol 2021; 184:768-775. [PMID: 34174305 DOI: 10.1016/j.ijbiomac.2021.06.110] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2021] [Revised: 05/28/2021] [Accepted: 06/16/2021] [Indexed: 12/22/2022]
Abstract
Polysaccharide hydrogels are promising candidate matrices for recapitulating the characteristics of extracellular matrix (ECM) in breast tumors in terms of their structure and composition. Herein, to obtain an ECM-mimetic matrix, hydroxyethyl chitosan (HECS) hydrogels were prepared through Schiff-base crosslinking reaction using dialdehyde hyaluronic acid as crosslinker. The obtained HECS hydrogels displayed a highly porous structure, a stiffness comparable to that of breast tissue, and a fast water-absorption speed. The amount of crosslinker had great effects on the swelling and rheological behaviors of the HECS hydrogels. Preliminary results from in vitro biological assessments confirmed that MCF-7 cells incubated within HECS hydrogels preferred to grow into three-dimensional spheroids. Importantly, the cells displayed enhanced migrative capability and upregulated expression levels of MMP-2, TGF-β and VEGF in comparison to two-dimension cultured cells. Hence, the HECS hydrogels show great promise as a biomimetic ECM in constructing breast tumor models.
Collapse
|
43
|
Leiva MC, Garre E, Gustafsson A, Svanström A, Bogestål Y, Håkansson J, Ståhlberg A, Landberg G. Breast cancer patient-derived scaffolds as a tool to monitor chemotherapy responses in human tumor microenvironments. J Cell Physiol 2021; 236:4709-4724. [PMID: 33368325 PMCID: PMC8049042 DOI: 10.1002/jcp.30191] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2020] [Revised: 11/04/2020] [Accepted: 11/20/2020] [Indexed: 12/21/2022]
Abstract
Breast cancer is a heterogeneous disease where the tumor microenvironment, including extracellular components, plays a crucial role in tumor progression, potentially modulating treatment response. Different approaches have been used to develop three-dimensional models able to recapitulate the complexity of the extracellular matrix. Here, we use cell-free patient-derived scaffolds (PDSs) generated from breast cancer samples that were recellularized with cancer cell lines as an in vivo-like culture system for drug testing. We show that PDS cultured MCF7 cancer cells increased their resistance against the front-line chemotherapy drugs 5-fluorouracil, doxorubicin and paclitaxel in comparison to traditional two-dimensional cell cultures. The gene expression of the environmentally adapted cancer cells was modulated in different ways depending on the drug and the concentration used. High doses of doxorubicin reduced cancer stem cell features, whereas 5-fluorouracil increased stemness and decreased the proliferative phenotype. By using PDSs repopulated with other breast cancer cell lines, T-47D and MDA-MB-231, we observed both general and cell line specific drug responses. In summary, PDSs can be used to examine the extracellular matrix influence on cancer drug responses and for testing novel compounds in in vivo-like microenvironments.
Collapse
Affiliation(s)
- Maria Carmen Leiva
- Department of Laboratory Medicine, Sahlgrenska Center for Cancer Research, Institute of Biomedicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Elena Garre
- Department of Laboratory Medicine, Sahlgrenska Center for Cancer Research, Institute of Biomedicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Anna Gustafsson
- Department of Laboratory Medicine, Sahlgrenska Center for Cancer Research, Institute of Biomedicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Andreas Svanström
- Department of Laboratory Medicine, Sahlgrenska Center for Cancer Research, Institute of Biomedicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| | - Yalda Bogestål
- Department of Biological FunctionRISE Research Institutes of SwedenBoråsSweden
| | - Joakim Håkansson
- Department of Laboratory Medicine, Sahlgrenska Center for Cancer Research, Institute of Biomedicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Department of Biological FunctionRISE Research Institutes of SwedenBoråsSweden
| | - Anders Ståhlberg
- Department of Laboratory Medicine, Sahlgrenska Center for Cancer Research, Institute of Biomedicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
- Wallenberg Center for Molecular and Translational MedicineUniversity of GothenburgGothenburgSweden
- Department of Clinical Genetics and GenomicsSahlgrenska University HospitalGothenburgSweden
| | - Göran Landberg
- Department of Laboratory Medicine, Sahlgrenska Center for Cancer Research, Institute of Biomedicine, Sahlgrenska AcademyUniversity of GothenburgGothenburgSweden
| |
Collapse
|
44
|
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: 6] [Impact Index Per Article: 2.0] [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
|
45
|
Identification of extracellular matrix proteins secreted by human dermal fibroblasts cultured in 3D electrospun scaffolds. Sci Rep 2021; 11:6655. [PMID: 33758206 PMCID: PMC7988018 DOI: 10.1038/s41598-021-85742-0] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Accepted: 03/01/2021] [Indexed: 02/07/2023] Open
Abstract
The appreciation that cell interactions in tissues is dependent on their three dimensional (3D) distribution has stimulated the development of 3D cell culture models. We constructed an artificial 3D tumour by culturing human breast cancer JIMT-1 cells and human dermal fibroblasts (HDFs) in a 3D network of electrospun polycaprolactone fibres. Here, we investigate ECM components produced by the cells in the artificial 3D tumour, which is an important step in validating the model. Immunostaining and confocal fluorescence microscopy show that the ECM proteins fibronectin, collagen I, and laminin are deposited throughout the entire 3D structure. Secreted soluble factors including matrix metalloproteinases (MMPs) and interleukine-6 (IL-6) were analysed in collected medium and were found to be mainly derived from the HDFs. Treatment with transforming growth factor-β1 (TGF-β1), a major cytokine found in a tumour, significantly alters the MMP activity and IL-6 concentration. In addition, TGF-β1 treatment, changes the morphology of the HDFs to become more elongated and with increased linearized actin filaments compared to non-treated HDFs. Collectively, these novel findings suggest that the artificial 3D tumour displays a clear cell distribution and ECM deposition that resembles a tumour environment in vivo, suggesting an innovative biological model to study a human tumour.
Collapse
|
46
|
Rodrigues J, Heinrich MA, Teixeira LM, Prakash J. 3D In Vitro Model (R)evolution: Unveiling Tumor–Stroma Interactions. Trends Cancer 2021; 7:249-264. [DOI: 10.1016/j.trecan.2020.10.009] [Citation(s) in RCA: 99] [Impact Index Per Article: 33.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2020] [Revised: 10/19/2020] [Accepted: 10/26/2020] [Indexed: 12/22/2022]
|
47
|
Jing L, Wang X, Leng B, Zhan N, Liu H, Wang S, Lu Y, Sun J, Huang D. Engineered Nanotopography on the Microfibers of 3D-Printed PCL Scaffolds to Modulate Cellular Responses and Establish an In Vitro Tumor Model. ACS APPLIED BIO MATERIALS 2021; 4:1381-1394. [PMID: 35014489 DOI: 10.1021/acsabm.0c01243] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Scaffold-based three-dimensional (3D) cell culture systems have gained increased interest in cell biology, tissue engineering, and drug screening fields as a replacement of two-dimensional (2D) monolayer cell culture and as a way to provide biomimetic extracellular matrix environments. In this study, microscale fibrous scaffolds were fabricated via electrohydrodynamic printing, and nanoscale features were created on the fiber surface by simply leaching gliadin of poly(ε-caprolactone) (PCL)/gliadin composites in ethanol solution. The microstructure of the printed scaffolds could be precisely controlled by printing parameters, and the surface nanotopography of the printed fiber could be tuned by varying the PCL/gliadin ratios. By seeding mouse embryonic fibroblast (NIH/3T3) cells and human nonsmall cell lung cancer (A549) cells on the printed scaffolds, the cellular responses showed that the fiber nanotopography on printed scaffolds efficiently favored cell adhesion, migration, proliferation, and tissue formation. Quantitative analysis of the transcript expression levels of A549 cells seeded on nanoporous scaffolds further revealed the upregulation of integrin-β1, focal adhesion kinase, Ki-67, E-cadherin, and epithelial growth factor receptors over what was observed in the cells grown on the pure PCL scaffold. Furthermore, a significant difference was found in the relevant biomarker expression on the developed scaffolds compared with that in the monolayer culture, demonstrating the potential of cancer cell-seeded scaffolds as 3D in vitro tumor models for cancer research and drug screening.
Collapse
Affiliation(s)
- Linzhi Jing
- National University of Singapore (Suzhou) Research Institute, 377 Linquan Street, Suzhou, Jiangsu 215123, China.,Department of Food Science and Technology, National University of Singapore, 3 Science Drive 2, 117542, Singapore
| | - Xiang Wang
- National University of Singapore (Suzhou) Research Institute, 377 Linquan Street, Suzhou, Jiangsu 215123, China
| | - Bin Leng
- Department of Food Science and Technology, National University of Singapore, 3 Science Drive 2, 117542, Singapore
| | - Ningping Zhan
- Department of Food Science and Technology, National University of Singapore, 3 Science Drive 2, 117542, Singapore
| | - Hang Liu
- National University of Singapore (Suzhou) Research Institute, 377 Linquan Street, Suzhou, Jiangsu 215123, China.,Department of Food Science and Technology, National University of Singapore, 3 Science Drive 2, 117542, Singapore
| | - Shifei Wang
- Department of Food Science and Technology, National University of Singapore, 3 Science Drive 2, 117542, Singapore
| | - Yuyun Lu
- Department of Food Science and Technology, National University of Singapore, 3 Science Drive 2, 117542, Singapore
| | - Jie Sun
- Department of Mechatronics and Robotics, Xi'an Jiaotong-Liverpool University, 111 Ren'ai Road, Suzhou, Jiangsu 215123, China
| | - Dejian Huang
- National University of Singapore (Suzhou) Research Institute, 377 Linquan Street, Suzhou, Jiangsu 215123, China.,Department of Food Science and Technology, National University of Singapore, 3 Science Drive 2, 117542, Singapore
| |
Collapse
|
48
|
Golchin A, Farzaneh S, Porjabbar B, Sadegian F, Estaji M, Ranjbarvan P, Kanafimahbob M, Ranjbari J, Salehi-Nik N, Hosseinzadeh S. Regenerative Medicine Under the Control of 3D Scaffolds: Current State and Progress of Tissue Scaffolds. Curr Stem Cell Res Ther 2021; 16:209-229. [DOI: 10.2174/1574888x15666200720115519] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2020] [Revised: 06/17/2020] [Accepted: 06/18/2020] [Indexed: 11/22/2022]
Abstract
Currently, combining stem cells (SCs) with biomaterial scaffolds provides a promising strategy
for the future of biomedicine and regenerative medicine (RG). The cells need similar substrates of
the extracellular matrix (ECM) for normal tissue development, which signifies the importance of
three dimensional (3D) scaffolds to determine cell fate. Herein, the importance and positive contributions
of corresponding 3D scaffolds on cell functions, including cell interactions, cell migrations,
and nutrient delivery, are presented. Furthermore, the synthesis techniques which are recruited to
fabricate the 3D scaffolds are discussed, and the related studies of 3D scaffold for different tissues
are also reported in this paper. This review focuses on 3D scaffolds that have been used for tissue
engineering purposes and directing stem cell fate as a means of producing replacements for biomedical
applications.
Collapse
Affiliation(s)
- Ali Golchin
- Department of Clinical Biochemistry and Applied Cell Science, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Sina Farzaneh
- Department of Tissue engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Bahareh Porjabbar
- Department of Tissue engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Fatemeh Sadegian
- Department of Tissue engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Masoumeh Estaji
- Department of Tissue engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Parviz Ranjbarvan
- Department of Clinical Biochemistry and Applied Cell Science, School of Medicine, Urmia University of Medical Sciences, Urmia, Iran
| | - Mohammad Kanafimahbob
- Department of Tissue engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Javad Ranjbari
- Department of Biotechnology, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Science, Tehran, Iran
| | - Nasim Salehi-Nik
- Department of Biomechanical Engineering, University of Twente, Enschede, Netherlands
| | - Simzar Hosseinzadeh
- Department of Tissue engineering and Applied Cell Sciences, School of Advanced Technologies in Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
49
|
Fernando K, Kwang LG, Lim JTC, Fong ELS. Hydrogels to engineer tumor microenvironments in vitro. Biomater Sci 2021; 9:2362-2383. [DOI: 10.1039/d0bm01943g] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Illustration of engineered hydrogel to recapitulate aspects of the tumor microenvironment.
Collapse
Affiliation(s)
- Kanishka Fernando
- Department of Biomedical Engineering
- National University of Singapore
- Singapore
| | - Leng Gek Kwang
- Department of Biomedical Engineering
- National University of Singapore
- Singapore
| | - Joanne Tze Chin Lim
- Department of Biomedical Engineering
- National University of Singapore
- Singapore
| | - Eliza Li Shan Fong
- Department of Biomedical Engineering
- National University of Singapore
- Singapore
- The N.1 Institute for Health
- National University of Singapore
| |
Collapse
|
50
|
Abstract
Defined by its potential for self-renewal, differentiation and tumorigenicity, cancer stem cells (CSCs) are considered responsible for drug resistance and relapse. To understand the behavior of CSC, the effects of the microenvironment in each tissue are a matter of great concerns for scientists in cancer biology. However, there are many complicated obstacles in the mimicking the microenvironment of CSCs even with current advanced technology. In this context, novel biomaterials have widely been assessed as in vitro platforms for their ability to mimic cancer microenvironment. These efforts should be successful to identify and characterize various CSCs specific in each type of cancer. Therefore, extracellular matrix scaffolds made of biomaterial will modulate the interactions and facilitate the investigation of CSC associated with biological phenomena simplifying the complexity of the microenvironment. In this review, we summarize latest advances in biomaterial scaffolds, which are exploited to mimic CSC microenvironment, and their chemical and biological requirements with discussion. The discussion includes the possible effects on both cells in tumors and microenvironment to propose what the critical factors are in controlling the CSC microenvironment focusing the future investigation. Our insights on their availability in drug screening will also follow the discussion.
Collapse
|