1
|
Karamanos NK, Piperigkou Z, Gourdoupi C, Mangani S, Vivanco MDM. Extracellular matrix matters: matrix-based bioscaffolds in advancing translational cancer research and targeted therapy. Am J Physiol Cell Physiol 2025; 328:C1957-C1963. [PMID: 40033935 DOI: 10.1152/ajpcell.00050.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/07/2025] [Accepted: 02/24/2025] [Indexed: 03/05/2025]
Abstract
The onset, development, and progression of cancer are greatly influenced by the microenvironmental cues originating from diverse elements within the tumor niche. Extracellular matrix (ECM), the complex and dynamic macromolecular three-dimensional network (3-D), governs cell functionality and plays key roles in tumor growth and spreading. This article highlights the significance of ECM-based bioscaffolds in providing a relevant microenvironment not only for studying tumor behavior and drug efficacy but also for narrowing the gap between translational cancer research and targeted cancer treatment. The development of novel and user-friendly platforms that resemble the human tumor microenvironment in early and advanced cancer stages, may help to predict treatment response, thus facilitating the development and testing of new drugs, bridging the gap between in vitro and in vivo models. In addition, we present innovative strategies leveraging ECM bioscaffolds for personalized cancer treatment, including drug delivery systems and tissue engineering approaches. Specific case studies as well as ethical concerns related to the use of ECM bioscaffolds in research and therapy are also presented and critically discussed. By elucidating the intricate interplay between ECM and cancer biology, this article underscores the potential of ECM bioscaffolds as novel platforms for shaping future therapeutic interventions and advancing precision oncology.
Collapse
Affiliation(s)
- Nikos K Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
- Foundation for Research and Technology-Hellas (FORTH), Institute of Chemical Engineering Sciences (ICE-HT), Patras, Greece
| | - Zoi Piperigkou
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Chrisavgi Gourdoupi
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Sylvia Mangani
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, Patras, Greece
| | - Maria dM Vivanco
- CIC bioGUNE, Basque Research and Technology Alliance (BRTA), Derio, Spain
| |
Collapse
|
2
|
Malakpour-Permlid A, Rodriguez MM, Untracht GR, Andersen PE, Oredsson S, Boisen A, Zór K. High-throughput non-homogenous 3D polycaprolactone scaffold for cancer cell and cancer-associated fibroblast mini-tumors to evaluate drug treatment response. Toxicol Rep 2025; 14:101863. [PMID: 39758801 PMCID: PMC11699757 DOI: 10.1016/j.toxrep.2024.101863] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/03/2024] [Accepted: 12/11/2024] [Indexed: 01/07/2025] Open
Abstract
High-throughput screening (HTS) three-dimensional (3D) tumor models are a promising approach for cancer drug discovery, as they more accurately replicate in vivo cell behavior than two-dimensional (2D) models. However, assessing and comparing current 3D models for drug efficacy remains essential, given the significant influence of cellular conditions on treatment response. To develop in vivo mimicking 3D models, we evaluated two HTS 3D models established in 96-well plates with 3D polycaprolactone (PCL) scaffolds fabricated using two distinct methods, resulting in scaffolds with either homogenous or non-homogenous fiber networks. These models, based on human HeLa cervical cancer cells and cancer-associated fibroblasts (CAFs) cultured as mono- or co-cultures within the 3D scaffolds, revealed that anticancer drug paclitaxel (PTX) exhibited consistently higher inhibitory concentration 50 (IC50) in 3D (≥ 1000 nM) compared to 2D (≥ 100 nM), indicating reduced toxicity on cells cultured in 3D. Interestingly, the toxicity of PTX was significantly lower on mini-tumors in non-homogenous 3D (IC50: 600 or 1000 nM) than in homogenous 3D cultures (IC50 exceeding 1000 nM). Microscopic studies revealed that the non-homogenous scaffolds closely resemble the tumor collagen network than their homogeneous counterpart. Both 3D scaffolds offer optimal pore size, facilitating efficient cell infiltration into the depth of 58.1 ± 1.2 µm (homogenous) and 86.4 ± 9.8 µm (non-homogenous) within 3D cultures. Cells cultured in the 3D non-homogenous systems exhibited drug treatment responses closer to in vivo conditions, highlighting the role of scaffold structure and design on cellular response to drug treatment. The PCL-based 3D models provide a robust, tunable, and efficient approach for the HTS of anti-cancer drugs compared to conventional 2D systems.
Collapse
Affiliation(s)
- Atena Malakpour-Permlid
- Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | - Manuel Marcos Rodriguez
- Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | - Gavrielle R. Untracht
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | - Peter E. Andersen
- Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | | | - Anja Boisen
- Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | - Kinga Zór
- Center for Intelligent Drug Delivery and Sensing Using Microcontainers and Nanomechanics (IDUN), Department of Health Technology, Technical University of Denmark, Kongens Lyngby 2800, Denmark
- BioInnovation Institute Foundation, Copenhagen N 2200, Denmark
- Innovation Acta S.r.l., Siena, Via delle 1-53100, Italy
| |
Collapse
|
3
|
Brümmer N, Doll-Nikutta K, Schadzek P, Mikolai C, Kampmann A, Wirth D, Hoffmann A, Pott PC, Karras O, Auer S, Stiesch M. Better models, better treatment? a systematic review of current three dimensional (3D) in vitro models for implant-associated infections. Front Bioeng Biotechnol 2025; 13:1569211. [PMID: 40352353 PMCID: PMC12061920 DOI: 10.3389/fbioe.2025.1569211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2025] [Accepted: 04/04/2025] [Indexed: 05/14/2025] Open
Abstract
Introduction Understanding the biology of implant-associated infections is essential in order to provide adequate detection, prevention and therapeutic strategies. Advanced 3D in vitro models offer valuable insights into the complex interactions between cells and bacteria in the presence of implant materials. This review aims to give a comprehensive overview of current 3D in vitro models that mimic implant-associated infections. Methods The structured literature search initially identified 258 publications, seven of which fitted the inclusion criteria. Results The included 3D models were established either to mimic the in vivo situation (organotypic model) or to investigate future implant materials. In three studies, organotypic models for dental implants were created and one study described an organotypic model containing immune cells. In the remaining three studies, biomaterials for constructing future orthopedic implants were developed and tested. All authors included specific cells and bacteria suitable for the respective implants. The dental implant models used fibroblasts and keratinocytes; the orthopedic implant models used stem cells and fibroblast-like cells; the model containing immune cells incorporated co-cultivation of fibroblasts and THP-1 derived macrophages. For bacterial challenge, most authors used Gram positive bacteria, but three studies employed Gram negative bacterial species. A wide variety of analytical methods of different complexity were applied after co-culture of cells and bacteria and between one and five different methods were used. Discussion All models could be employed to provide answers to specific scientific questions regarding implant-associated infections. Nonetheless, this review reveals the limitations of current 3D models for the investigation of implant-associated infections and highlights the opportunities for further development in this scientific field.
Collapse
Affiliation(s)
- Neele Brümmer
- Hannover Medical School, Clinic of Prosthetic Dentistry and Biomedical Materials Research, Hannover, Germany
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| | - Katharina Doll-Nikutta
- Hannover Medical School, Clinic of Prosthetic Dentistry and Biomedical Materials Research, Hannover, Germany
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| | - Patrik Schadzek
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
- Hannover Medical School, DIAKOVERE Annastift, Department of Orthopedic Surgery, Hannover, Germany
| | - Carina Mikolai
- Hannover Medical School, Clinic of Prosthetic Dentistry and Biomedical Materials Research, Hannover, Germany
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| | - Andreas Kampmann
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
- Hannover Medical School, Clinic for Cranio-Maxillo-Facial Surgery, Hannover, Germany
| | - Dagmar Wirth
- Helmholtz Centre for Infection Research, Braunschweig, Germany
- Hannover Medical School, Department of Experimental Hematology, Hannover, Germany
| | - Andrea Hoffmann
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
- Hannover Medical School, DIAKOVERE Annastift, Department of Orthopedic Surgery, Hannover, Germany
| | - Philipp-Cornelius Pott
- Hannover Medical School, Clinic of Prosthetic Dentistry and Biomedical Materials Research, Hannover, Germany
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| | - Oliver Karras
- TIB - Leibniz Information Centre for Science and Technology, Hannover, Germany
| | - Sören Auer
- TIB - Leibniz Information Centre for Science and Technology, Hannover, Germany
| | - Meike Stiesch
- Hannover Medical School, Clinic of Prosthetic Dentistry and Biomedical Materials Research, Hannover, Germany
- Lower Saxony Center for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Germany
| |
Collapse
|
4
|
Mangani S, Kremmydas S, Karamanos NK. Mimicking the Complexity of Solid Tumors: How Spheroids Could Advance Cancer Preclinical Transformative Approaches. Cancers (Basel) 2025; 17:1161. [PMID: 40227664 PMCID: PMC11987746 DOI: 10.3390/cancers17071161] [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: 02/25/2025] [Revised: 03/27/2025] [Accepted: 03/28/2025] [Indexed: 04/15/2025] Open
Abstract
Traditional 2D cell culture models present significant limitations in replicating the intricate architecture and microenvironment of in vivo solid tumors, which are essential for accurately studying cancer initiation, growth, progression, and metastasis. This underscores the need for the development of advanced preclinical models to accelerate research outcomes. Emerging 3D cell culture systems, particularly spheroid models, provide a more realistic representation of solid tumor properties by capturing the complex interactions occurring within the tumor microenvironment, including the extracellular matrix dynamics that influence cancer progression. Among solid tumors, breast cancer remains the most frequently diagnosed cancer among women globally and a leading cause of cancer-related mortality. Here we emphasize the value of breast cancer cell-derived spheroids in revealing differential molecular characteristics and understanding cancer cell properties during the early stages of invasion into adjacent tissues. Conclusively, this study underscores the urgent need to adopt 3D cell culture platforms, given their significant contributions to advanced cancer research and pharmaceutical targeting. This may well offer a transformative approach for preclinical studies and enhance our ability to test therapeutic efficiency in conditions that closely mimic the growth and progression of in vivo solid tumors.
Collapse
Affiliation(s)
| | | | - Nikos K. Karamanos
- Biochemistry, Biochemical Analysis & Matrix Pathobiology Research Group, Laboratory of Biochemistry, Department of Chemistry, University of Patras, 26504 Patras, Greece
| |
Collapse
|
5
|
Wang Y, Zhang R, Qiao Z, Dou B, Xu H, Meng F, Huang J. Polyacrylamide-Based Hydrogel with Biocompatibility and Tunable Stiffness for Three-Dimensional Cell Culture. ACS APPLIED BIO MATERIALS 2025; 8:2356-2364. [PMID: 39949138 DOI: 10.1021/acsabm.4c01846] [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: 03/18/2025]
Abstract
Three-dimensional (3D) culture of cells has gained increasing popularity because of its enhanced physiological relevance and more accurate representation of in vivo tissues. Matrigel, alginate, hyaluronic acid, and collagen are biocompatible 3D culture platforms with cell biofunctions, while it is difficult to decouple the biofunctions with mechanical properties. Polyacrylamide (PAAm) is a biocompatible but biologically nonfunctional platform heavily used in 2D culture. However, the cytotoxicity of acrylamide (AAm) prevents the application of PAAm as a platform for the 3D culture. Here, through RAFT copolymerization of AAm with a primary amine-bearing functional monomer, followed by postpolymerization modification, we synthesized nontoxic, linear PAAm featuring either multithiol or multinorbornene groups, available in various chain lengths. PAAm networks were fabricated by photoinduced thiol-norbornene coupling. The resulting PAAm hydrogel was biocompatible and structurally homogeneous with highly tunable and reproducible mechanical properties. PAAm hydrogels supported the 3D culture of human umbilical vein endothelial cells (HUVECs), where a higher adhesive ligand density promoted the viability of HUVECs. Furthermore, in combination with Matrigel, the PAAm hydrogel was used in the 3D culture of intestinal organoids, demonstrating that a lower mechanical strength was favorable. In summary, this report paves the way for the use of PAAm hydrogels in 3D culture, which is especially appealing for the decoupling of biological functions and mechanical properties.
Collapse
Affiliation(s)
- Yi Wang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
- SINOPEC Research Institute of Petroleum Engineering Co. Ltd., Beijing 102206, China
| | - Rui Zhang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| | - Ziwen Qiao
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| | - Bohan Dou
- Department of Mechanical Engineering, Tsinghua University, Beijing 100084, China
| | - Hongwei Xu
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| | - Fanlu Meng
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| | - Jianyong Huang
- Department of Mechanics and Engineering Science, College of Engineering, Peking University, Beijing 100871, China
| |
Collapse
|
6
|
Grigoreva TA, Kindt DN, Sagaidak AV, Novikova DS, Tribulovich VG. Cellular Systems for Colorectal Stem Cancer Cell Research. Cells 2025; 14:170. [PMID: 39936962 PMCID: PMC11817814 DOI: 10.3390/cells14030170] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 02/13/2025] Open
Abstract
Oncological diseases consistently occupy leading positions among the most life-threatening diseases, including in highly developed countries. At the same time, the second most common cause of cancer death is colorectal cancer. The current level of research shows that the development of effective therapy, in this case, requires a new grade of understanding processes during the emergence and development of a tumor. In particular, the concept of cancer stem cells that ensure the survival of chemoresistant cells capable of giving rise to new tumors is becoming widespread. To provide adequate conditions that reproduce natural processes typical for tumor development, approaches based on increasingly complex cellular systems are being improved. This review discusses the main strategies that allow for the study of the properties of tumor cells with an emphasis on colorectal cancer stem cells. The features of working with tumor cells and the advantages and disadvantages of 2D and 3D culture systems are considered.
Collapse
Affiliation(s)
- Tatyana A. Grigoreva
- Laboratory of Molecular Pharmacology, St. Petersburg State Institute of Technology (Technical University), 190013 St. Petersburg, Russia (V.G.T.)
| | | | | | | | | |
Collapse
|
7
|
Rainu SK, Singh N. Miniaturized Liver Disease Mimics to Gain Insights into MMP Expression during Disease Progression. ACS Biomater Sci Eng 2025; 11:476-484. [PMID: 39801310 DOI: 10.1021/acsbiomaterials.4c01449] [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: 05/02/2025]
Abstract
Nonalcoholic fatty liver disease (NAFLD) encompasses a spectrum of liver conditions, ranging from hepatic steatosis to steatohepatitis, fibrosis, and severe outcomes such as cirrhosis or cancer. The progression from hepatic steatosis to fibrosis involves significant extracellular matrix (ECM) remodeling, characterized by increased collagen deposition and cross-linking of ECM proteins, causing increased tissue stiffness and altered MMP expression patterns. Dysregulated MMP expression and extracellular acidosis are key contributors to NAFLD progression. Unlike other MMPs, which may be relevant only at specific disease stages, MMP-9 serves as a universal marker, allowing for monitoring of its expression in relation to disease states and ECM parameters. Understanding dysregulated MMP-9 expression across different NAFLD stages can provide crucial insights into disease progression and serve as both a diagnostic and a prognostic biomarker, identifying potential therapeutic targets. This study introduces a three-dimensional (3D) collagen/alginate-based liver disease model designed to investigate how matrix collagen content, elasticity, and diseased cell conditions influence MMP expression and pH levels in situ using nanoprobes. The platform offered an understanding of the relationships between these factors and their role in NAFLD progression, offering valuable insights into disease progression and potential resolution. To examine how various physicochemical and biological factors, particularly MMP expression and collagen deposition, drive NAFLD progression, three 3D NAFLD models were developed, simulating healthy (HL), steatotic (SL), and fibrotic (FL) liver matrices. Additionally, the role of collagenase treatment in the FL matrix in enhancing MMP expression and potentially mitigating fibrosis was also explored. By employing dual-sensitive fluorescent nanoprobes to monitor real-time in situ changes in MMP-9 expression and pH levels, this platform offers a novel approach to understanding the in vitro roles of matrix stiffness, collagen deposition, and diseased cell conditions in NAFLD pathogenesis.
Collapse
Affiliation(s)
- Simran Kaur Rainu
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Neetu Singh
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
- Biomedical Engineering Unit, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| |
Collapse
|
8
|
Abbasi-Malati Z, Khanicheragh P, Narmi MT, Mardi N, Khosrowshahi ND, Hiradfar A, Rezabakhsh A, Sadeghsoltani F, Rashidi S, Chegeni SA, Roozbahani G, Rahbarghazi R. Tumoroids, a valid preclinical screening platform for monitoring cancer angiogenesis. Stem Cell Res Ther 2024; 15:267. [PMID: 39183337 PMCID: PMC11346257 DOI: 10.1186/s13287-024-03880-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Accepted: 08/06/2024] [Indexed: 08/27/2024] Open
Abstract
In recent years, biologists and clinicians have witnessed prominent advances in in vitro 3D culture techniques related to biomimetic human/animal tissue analogs. Numerous data have confirmed that unicellular and multicellular (tumoroids) tumor spheroids with dense native cells in certain matrices are sensitive and valid analytical tools for drug screening, cancer cell dynamic growth, behavior, etc. in laboratory settings. Angiogenesis/vascularization is a very critical biological phenomenon to support oxygen and nutrients to tumor cells within the deep layer of solid masses. It has been shown that endothelial cell (EC)-incorporated or -free spheroid/tumoroid systems provide a relatively reliable biological platform for monitoring the formation of nascent blood vessels in micron/micrometer scales. Besides, the paracrine angiogenic activity of cells within the spheroid/tumoroid systems can be monitored after being treated with different therapeutic approaches. Here, we aimed to collect recent advances and findings related to the monitoring of cancer angiogenesis using unicellular and multicellular tumor spheroids. Vascularized spheroids/tumoroids can help us in the elucidation of mechanisms related to cancer formation, development, and metastasis by monitoring the main influencing factors.
Collapse
Affiliation(s)
- Zahra Abbasi-Malati
- Student Research Committee, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Parisa Khanicheragh
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Narges Mardi
- Biotechnology Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Nafiseh Didar Khosrowshahi
- Stem Cell and Tissue Engineering Research Laboratory, Sahand University of Technology, Tabriz, 51335-1996, Iran
| | - Amirataollah Hiradfar
- Pediatric Health Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | - Aysa Rezabakhsh
- Cardiovascular Research Center, Tabriz University of Medical Sciences, Tabriz, Iran
| | | | - Somayyeh Rashidi
- Department of Medical Biotechnology, Faculty of Medicine, Zanjan University of Medical Sciences, Zanjan, Iran
| | | | - Golbarg Roozbahani
- Department of Plant, Cell and Molecular Biology, Faculty of Natural Sciences, University of Tabriz, Tabriz, Iran
| | - Reza Rahbarghazi
- Stem Cell Research Center, Tabriz University of Medical Sciences, Tabriz, Iran.
- Department of Applied Cell Sciences, Faculty of Advanced Medical Sciences, Tabriz University of Medical Sciences, Tabriz, Iran.
| |
Collapse
|
9
|
Khorsandi D, Yang JW, Foster S, Khosravi S, Hoseinzadeh N, Zarei F, Lee YB, Runa F, Gangrade A, Voskanian L, Adnan D, Zhu Y, Wang Z, Jucaud V, Dokmeci MR, Shen X, Bishehsari F, Kelber JA, Khademhosseini A, de Barros NR. Patient-Derived Organoids as Therapy Screening Platforms in Cancer Patients. Adv Healthc Mater 2024; 13:e2302331. [PMID: 38359321 PMCID: PMC11324859 DOI: 10.1002/adhm.202302331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 11/28/2023] [Indexed: 02/17/2024]
Abstract
Patient-derived organoids (PDOs) developed ex vivo and in vitro are increasingly used for therapeutic screening. They provide a more physiologically relevant model for drug discovery and development compared to traditional cell lines. However, several challenges remain to be addressed to fully realize the potential of PDOs in therapeutic screening. This paper summarizes recent advancements in PDO development and the enhancement of PDO culture models. This is achieved by leveraging materials engineering and microfabrication technologies, including organs-on-a-chip and droplet microfluidics. Additionally, this work discusses the application of PDOs in therapy screening to meet diverse requirements and overcome bottlenecks in cancer treatment. Furthermore, this work introduces tools for data processing and analysis of organoids, along with their microenvironment. These tools aim to achieve enhanced readouts. Finally, this work explores the challenges and future perspectives of using PDOs in drug development and personalized screening for cancer patients.
Collapse
Affiliation(s)
- Danial Khorsandi
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Jia-Wei Yang
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Samuel Foster
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Safoora Khosravi
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Negar Hoseinzadeh
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Fahimeh Zarei
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Yun Bin Lee
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Farhana Runa
- California State University Northridge, 18111 Nordhoff Street, Northridge, California, USA
| | - Ankit Gangrade
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Leon Voskanian
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Darbaz Adnan
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush Medical College, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Yangzhi Zhu
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Zhaohui Wang
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
- Department of Pathology, Duke University School of Medicine, Durham, NC, 27710 USA
| | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Mehmet Remzi Dokmeci
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Xiling Shen
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Faraz Bishehsari
- Rush Center for Integrated Microbiome and Chronobiology Research, Rush Medical College, Rush University Medical Center, Chicago, IL, 60612, USA
- Division of Digestive Diseases, Rush Center for Integrated Microbiome & Chronobiology Research, Rush University Medical Center, Chicago, IL, 60612, USA
| | - Jonathan A. Kelber
- California State University Northridge, 18111 Nordhoff Street, Northridge, California, USA
- Baylor University, 101 Bagby Ave, Waco, Texas, USA
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Natan Roberto de Barros
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| |
Collapse
|
10
|
Yamashita M, Tamamitsu M, Kirisako H, Goda Y, Chen X, Hattori K, Ota S. High-Throughput 3D Imaging Flow Cytometry of Suspended Adherent 3D Cell Cultures. SMALL METHODS 2024; 8:e2301318. [PMID: 38133483 DOI: 10.1002/smtd.202301318] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 11/27/2023] [Indexed: 12/23/2023]
Abstract
3D cell cultures are indispensable in recapitulating in vivo environments. Among the many 3D culture methods, culturing adherent cells on hydrogel beads to form spheroid-like structures is a powerful strategy for maintaining high cell viability and functions in the adherent states. However, high-throughput, scalable technologies for 3D imaging of individual cells cultured on the hydrogel scaffolds are lacking. This study reports the development of a high throughput, scalable 3D imaging flow cytometry platform for analyzing spheroid models. This platform is realized by integrating a single objective fluorescence light-sheet microscopy with a microfluidic device that combines hydrodynamic and acoustofluidic focusing techniques. This integration enabled unprecedentedly high-throughput and scalable optofluidic 3D imaging, processing 1310 spheroids consisting of 28 117 cells min-1. The large dataset obtained enables precise quantification and comparison of the nuclear morphology of adhering and suspended cells, revealing that the adhering cells have smaller nuclei with less rounded surfaces. This platform's high throughput, robustness, and precision for analyzing the morphology of subcellular structures in 3D culture models hold promising potential for various biomedical analyses, including image-based phenotypic screening of drugs with spheroids or organoids.
Collapse
Affiliation(s)
- Minato Yamashita
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Miu Tamamitsu
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Hiromi Kirisako
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Yuki Goda
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Xiaoyao Chen
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Kazuki Hattori
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| | - Sadao Ota
- Research Center for Advanced Science and Technology, The University of Tokyo, 4-6-1 Komaba, Meguro-ku, Tokyo, 153-8904, Japan
| |
Collapse
|
11
|
Alshehri AM, Wilson OC. Biomimetic Hydrogel Strategies for Cancer Therapy. Gels 2024; 10:437. [PMID: 39057460 PMCID: PMC11275631 DOI: 10.3390/gels10070437] [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: 05/27/2024] [Revised: 06/18/2024] [Accepted: 06/28/2024] [Indexed: 07/28/2024] Open
Abstract
Recent developments in biomimetic hydrogel research have expanded the scope of biomedical technologies that can be used to model, diagnose, and treat a wide range of medical conditions. Cancer presents one of the most intractable challenges in this arena due to the surreptitious mechanisms that it employs to evade detection and treatment. In order to address these challenges, biomimetic design principles can be adapted to beat cancer at its own game. Biomimetic design strategies are inspired by natural biological systems and offer promising opportunities for developing life-changing methods to model, detect, diagnose, treat, and cure various types of static and metastatic cancers. In particular, focusing on the cellular and subcellular phenomena that serve as fundamental drivers for the peculiar behavioral traits of cancer can provide rich insights into eradicating cancer in all of its manifestations. This review highlights promising developments in biomimetic nanocomposite hydrogels that contribute to cancer therapies via enhanced drug delivery strategies and modeling cancer mechanobiology phenomena in relation to metastasis and synergistic sensing systems. Creative efforts to amplify biomimetic design research to advance the development of more effective cancer therapies will be discussed in alignment with international collaborative goals to cure cancer.
Collapse
Affiliation(s)
- Awatef M. Alshehri
- Department of Biomedical Engineering, The Catholic University of America, Washington, DC 20064, USA
- Department of Nanomedicine, King Abdullah International Medical Research Center (KAIMRC), King Saud bin Abdelaziz University for Health Sciences (KSAU-HS), Ministry of National Guard-Health Affairs (MNGHA), Riyadh 11426, Saudi Arabia;
| | - Otto C. Wilson
- Department of Biomedical Engineering, The Catholic University of America, Washington, DC 20064, USA
| |
Collapse
|
12
|
Kashkooli FM, Jakhmola A, A Ferrier G, Sathiyamoorthy K, Tavakkoli J(J, C Kolios M. Development of an ultrasound-mediated nano-sized drug-delivery system for cancer treatment: from theory to experiment. Nanomedicine (Lond) 2024; 19:1167-1189. [PMID: 38722104 PMCID: PMC11418290 DOI: 10.2217/nnm-2023-0259] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2023] [Accepted: 03/06/2024] [Indexed: 09/21/2024] Open
Abstract
Aim: To establish a methodology for understanding how ultrasound (US) induces drug release from nano-sized drug-delivery systems (NSDDSs) and enhances drug penetration and uptake in tumors. This aims to advance cancer treatment strategies.Materials & methods: We developed a multi-physics mathematical model to elucidate and understand the intricate mechanisms governing drug release, transport and delivery. Unique in vitro models (monolayer, multilayer, spheroid) and a tailored US exposure setup were introduced to evaluate drug penetration and uptake.Results: The results highlight the potential advantages of US-mediated NSDDSs over conventional NSDDSs and chemotherapy, notably in enhancing drug release and inducing cell death.Conclusion: Our sophisticated numerical and experimental methods aid in determining and quantifying drug penetration and uptake into solid tumors.
Collapse
Affiliation(s)
| | - Anshuman Jakhmola
- Department of Physics, Toronto Metropolitan University, Toronto, ON, Canada
| | - Graham A Ferrier
- Department of Physics, Toronto Metropolitan University, Toronto, ON, Canada
| | | | - Jahangir (Jahan) Tavakkoli
- Department of Physics, Toronto Metropolitan University, Toronto, ON, Canada
- Institute for Biomedical Engineering, Science & Technology (iBEST), Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada
| | - Michael C Kolios
- Department of Physics, Toronto Metropolitan University, Toronto, ON, Canada
- Institute for Biomedical Engineering, Science & Technology (iBEST), Keenan Research Centre for Biomedical Science, St. Michael's Hospital, Toronto, ON, Canada
| |
Collapse
|
13
|
Rainu SK, Singh N. Dual-Sensitive Fluorescent Nanoprobes for Simultaneously Monitoring In Situ Changes in pH and Matrix Metalloproteinase Expression in Stiffness-Tunable Three-Dimensional In Vitro Scaffolds. ACS APPLIED MATERIALS & INTERFACES 2024; 16:12175-12187. [PMID: 38420964 DOI: 10.1021/acsami.3c16334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/02/2024]
Abstract
A tumor microenvironment often presents altered physicochemical characteristics of the extracellular matrix (ECM) including changes in matrix composition, stiffness, protein expression, pH, temperature, or the presence of certain stromal and immune cells. Of these, overexpression of matrix metalloproteinases (MMPs) and extracellular acidosis are the two major hallmarks of cancer that can be exploited for tumor detection. The change in matrix stiffness and the release of certain cytokines (TNF-α) in the tumor microenvironment play major roles in inducing MMP-9 expression in cancerous cells. This study highlights the role of mechanical cues in upregulating MMP-9 expression in cancerous cells using stiffness-tunable matrix compositions and dual-sensitive fluorescent nanoprobes. Ionically cross-linked 3D alginate/gelatin (AG) scaffolds with three stiffnesses were chosen to reflect the ECM stiffnesses corresponding to healthy and pathological tissues. Moreover, a dual-sensitive nanoprobe, an MMP-sensitive peptide conjugated to carbon nanoparticles with intrinsic pH fluorescence properties, was utilized for in situ monitoring of the two cancer hallmarks in the 3D scaffolds. This platform was further utilized for designing a 3D core-shell platform for spatially mapping tumor margins and for visualizing TNF-α-induced MMP-9 expression in cancerous cells.
Collapse
Affiliation(s)
- Simran Kaur Rainu
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
| | - Neetu Singh
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi 110016, India
- Biomedical Engineering Unit, All India Institute of Medical Sciences, Ansari Nagar, New Delhi 110029, India
| |
Collapse
|
14
|
Azhari Rad R, Naghdi Y, Majidi Jamalabadi M, Masoumi S, Rezakhani L, Alizadeh M. Tissue Engineering Scaffolds Loaded With a Variety of Plant Extracts: Novel Model in Breast Cancer Therapy. Breast Cancer (Auckl) 2024; 18:11782234241236358. [PMID: 38476474 PMCID: PMC10929036 DOI: 10.1177/11782234241236358] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Accepted: 02/14/2024] [Indexed: 03/14/2024] Open
Abstract
Despite recent improvements in detecting and managing breast cancer (BC), it continues to be a major worldwide health concern that annually affects millions of people. Exploring the anti-BC potentials of natural compounds has received a lot of scientific attention due to their multi-target mode of action and good safety profiles because of these unmet needs. Drugs made from herbs are secure and have a lot fewer negative effects than those made from synthetic materials. Early stage patients benefit from breast-conserving surgery, but the risk of local recurrence remains, necessitating implanted scaffolds. These scaffolds provide residual cancer cell killing and tailored drug delivery. This review looks at plant extract-infused tissue engineering scaffolds, which provide a novel approach to treating BC. By offering patient individualized, safer treatments, these scaffolds could completely change how BC is treated.
Collapse
Affiliation(s)
- Reyhaneh Azhari Rad
- Student Research Committee, School of Paramedicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Yasaman Naghdi
- Student Research Committee, School of Paramedicine, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Mobina Majidi Jamalabadi
- Student Research Committee, School of Nursing and Midwifery, Shahroud University of Medical Sciences, Shahroud, Iran
| | - Sima Masoumi
- Graduate of Faculty of Veterinary Sciences, Sanandaj Branch, Islamic Azad University, Sanandaj, Iran
| | - Leila Rezakhani
- Fertility and Infertility Research Center, Health Technology Institute, Kermanshah University of Medical Sciences, Kermanshah, Iran
- Department of Tissue Engineering, School of Medicine, Kermanshah University of Medical Sciences, Kermanshah, Iran
| | - Morteza Alizadeh
- Department of Tissue Engineering, School of Medicine, Shahroud University of Medical Sciences, Shahroud, Iran
| |
Collapse
|
15
|
Tevlek A. The role of decellularized cell derived extracellular matrix in the establishment and culture of in vitrobreast cancer tumor model. Biomed Mater 2024; 19:025037. [PMID: 38286003 DOI: 10.1088/1748-605x/ad2378] [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: 11/18/2023] [Accepted: 01/29/2024] [Indexed: 01/31/2024]
Abstract
Decades of research have shown that two-dimensional cell culture studies are insufficient for preclinical cancer diagnosis and treatment, and that cancer cells in three-dimensional (3D) culture systems have better cell-cell and cell-matrix interactions, gene expression, heterogeneity, and structural complexity that more closely resemblein vivotumors. Researchers are still optimizing 3D culturing settings for different cancers. Despite promising tumor spheroid research, tumor cell-only aggregates lack the tumor microenvironment and cannot model tumors. Here, MCF-7 breast cancer cell derived decellularized extracellular matrix (CD-dECMs) were obtained and converted into autologous, biologically active, biocompatible, and non-immunogenic hydrogels to be used as micro-environment in both organoid formation and culture. For the production of organoids, CD-dECM doping concentrations ranging from 0.1 mg ml-1to 1.5 mg ml-1were evaluated, and the lowest concentration was found to be the most effective. For organoid culture, 8 mg ml-1CD-dECM, 4 mg ml-1rat tendon collagen type I (Col I) (4 mg ml-1) and a 1:1 (v/v) mixture of these two were used and the most viable and the biggest organoids were discovered in CD-dECM/Col I (1:1) group. The results show that autologous CD-dECM can replace hydrogels in tumor organoid generation and culture at low and high concentrations, respectively.
Collapse
Affiliation(s)
- Atakan Tevlek
- Middle East Technical University (METU), MEMS Research and Application Center, 06530 Ankara, Turkey
| |
Collapse
|
16
|
Liu Y, Liu H, Zhang C, Kong Y, Chen C, Gao W, Xi X, Deng L. Preparation and investigation of a novel antibacterial collagen-based material loaded with gentamicin following surface modification with citric acid for corneal tissue engineering. Int J Biol Macromol 2023; 253:126791. [PMID: 37683748 DOI: 10.1016/j.ijbiomac.2023.126791] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/04/2023] [Accepted: 09/05/2023] [Indexed: 09/10/2023]
Abstract
Corneal disease is an important clinical problem that affects millions of blind people and keratoplasty is currently the most successful treatment for corneal blindness. Unfortunately, there is a very high risk of bacterial infection during corneal transplantation. In this study, we proposed a novel synthetic collagen-based film for corneal therapy, and we effectively incorporated aminoglycoside gentamicin molecules onto the surface of the collagen film. We anticipate that this collagen-based substance will be antimicrobial and repair corneal tissue damage. Three steps were used to create this gentamicin-modified carboxylated collagen film, including: (i) Cross-link the collagen molecules with 1-ethyl-3- (3-dimethylaminopropyl) carbodiimide and n-hydroxysuccinimide to create a collagen (Col) film. (ii) Citric acid was used to modify the Col film's surface in order to increase the number of carboxyl groups there (ColCA). (iii) Gentamicin molecules were grafted onto the surface of ColCA film by forming amide bonds (ColCA-GM). We discovered that this ColCA-GM film has good physicochemical properties and excellent biocompatibility. Furthermore, it was demonstrated that treating collagen films with citric acid significantly improved the antibacterial properties of ColCA-GM film. The outcomes point to a variety of potential applications for this novel film in corneal tissue engineering.
Collapse
Affiliation(s)
- Yang Liu
- Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou 213164, China; School of Pharmacy, School of Biology and Food Engineering, Changzhou University, Changzhou 213164, China; School of Medical and Health Engineering, Changzhou University, Changzhou 213164, China.
| | - Huiyu Liu
- Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou 213164, China; School of Pharmacy, School of Biology and Food Engineering, Changzhou University, Changzhou 213164, China
| | - Chuanlei Zhang
- Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou 213164, China; School of Pharmacy, School of Biology and Food Engineering, Changzhou University, Changzhou 213164, China
| | - Yanhui Kong
- Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou 213164, China; School of Pharmacy, School of Biology and Food Engineering, Changzhou University, Changzhou 213164, China
| | - Cheng Chen
- Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou 213164, China; School of Pharmacy, School of Biology and Food Engineering, Changzhou University, Changzhou 213164, China
| | - Wenyu Gao
- Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou 213164, China; School of Pharmacy, School of Biology and Food Engineering, Changzhou University, Changzhou 213164, China
| | - Xiaowei Xi
- Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou 213164, China; School of Pharmacy, School of Biology and Food Engineering, Changzhou University, Changzhou 213164, China
| | - Linhong Deng
- Institute of Biomedical Engineering and Health Sciences, Changzhou University, Changzhou 213164, China; School of Pharmacy, School of Biology and Food Engineering, Changzhou University, Changzhou 213164, China; School of Medical and Health Engineering, Changzhou University, Changzhou 213164, China
| |
Collapse
|
17
|
Moriwaki T, Tani H, Haga K, Morita-Umei Y, Soma Y, Umei TC, Sekine O, Takatsuna K, Kishino Y, Kanazawa H, Fujita J, Fukuda K, Tohyama S, Ieda M. Scalable production of homogeneous cardiac organoids derived from human pluripotent stem cells. CELL REPORTS METHODS 2023; 3:100666. [PMID: 38113855 PMCID: PMC10753388 DOI: 10.1016/j.crmeth.2023.100666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 08/24/2023] [Accepted: 11/16/2023] [Indexed: 12/21/2023]
Abstract
Three-dimensional (3D) cultures are known to more closely mimic in vivo conditions compared with 2D cultures. Cardiac spheroids (CSs) and organoids (COs) are useful for 3D tissue engineering and are advantageous for their simplicity and mass production for regenerative therapy and drug discovery. Herein, we describe a large-scale method for producing homogeneous human induced pluripotent stem cell (hiPSC)-derived CSs (hiPSC-CSs) and COs without scaffolds using a porous 3D microwell substratum with a suction system. Our method has many advantages, such as increased efficiency and improved functionality, homogeneity, and sphericity of hiPSC-CSs. Moreover, we have developed a substratum on a clinically relevant large scale for regenerative therapy and have succeeded in producing approximately 40,000 hiPSC-CSs with high sphericity at once. Furthermore, we efficiently produced a fused CO model consisting of hiPSC-derived atrial and ventricular cardiomyocytes localized on opposite sides of one organoid. This method will facilitate progress toward hiPSC-based clinical applications.
Collapse
Affiliation(s)
- Taijun Moriwaki
- Department of Cardiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Hidenori Tani
- Department of Cardiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan; Joint Research Laboratory for Medical Innovation in Heart Disease, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Kotaro Haga
- Department of Cardiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Yuika Morita-Umei
- Department of Cardiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan; Kanagawa Institute of Industrial Science and Technology (KISTEC), Kawasaki, Kanagawa, Japan
| | - Yusuke Soma
- Department of Cardiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Tomohiko C Umei
- Department of Cardiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Otoya Sekine
- Department of Cardiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Kaworu Takatsuna
- Department of Cardiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Yoshikazu Kishino
- Department of Cardiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Hideaki Kanazawa
- Department of Cardiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Jun Fujita
- Department of Cardiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan; Department of Pathology & Immunology, Baylor College of Medicine, Houston, TX, USA
| | - Keiichi Fukuda
- Department of Cardiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| | - Shugo Tohyama
- Department of Cardiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan.
| | - Masaki Ieda
- Department of Cardiology, Keio University School of Medicine, Shinjuku, Tokyo, Japan
| |
Collapse
|
18
|
Bhattacharya A, Alam K, Roy NS, Kaur K, Kaity S, Ravichandiran V, Roy S. Exploring the interaction between extracellular matrix components in a 3D organoid disease model to replicate the pathophysiology of breast cancer. J Exp Clin Cancer Res 2023; 42:343. [PMID: 38102637 PMCID: PMC10724947 DOI: 10.1186/s13046-023-02926-4] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023] Open
Abstract
In vitro models are necessary to study the pathophysiology of the disease and the development of effective, tailored treatment methods owing to the complexity and heterogeneity of breast cancer and the large population affected by it. The cellular connections and tumor microenvironments observed in vivo are often not recapitulated in conventional two-dimensional (2D) cell cultures. Therefore, developing 3D in vitro models that mimic the complex architecture and physiological circumstances of breast tumors is crucial for advancing our understanding of the illness. A 3D scaffold-free in vitro disease model mimics breast cancer pathophysiology by allowing cells to self-assemble/pattern into 3D structures, in contrast with other 3D models that rely on artificial scaffolds. It is possible that this model, whether applied to breast tumors using patient-derived primary cells (fibroblasts, endothelial cells, and cancer cells), can accurately replicate the observed heterogeneity. The complicated interactions between different cell types are modelled by integrating critical components of the tumor microenvironment, such as the extracellular matrix, vascular endothelial cells, and tumor growth factors. Tissue interactions, immune cell infiltration, and the effects of the milieu on drug resistance can be studied using this scaffold-free 3D model. The scaffold-free 3D in vitro disease model for mimicking tumor pathophysiology in breast cancer is a useful tool for studying the molecular basis of the disease, identifying new therapeutic targets, and evaluating treatment modalities. It provides a more physiologically appropriate high-throughput platform for screening large compound library in a 96-384 well format. We critically discussed the rapid development of personalized treatment strategies and accelerated drug screening platforms to close the gap between traditional 2D cell culture and in vivo investigations.
Collapse
Affiliation(s)
- Anamitra Bhattacharya
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Kamare Alam
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Nakka Sharmila Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Kulwinder Kaur
- School of Pharmacy and Biomolecular Sciences, RCSI University of Medicine a Health Sciences, Dublin, Ireland
- Tissue Engineering Research Group, Department of Anatomy & Regenerative Medicine, RCSI University of Medicine and Health Sciences, Dublin, Ireland
| | - Santanu Kaity
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Velayutham Ravichandiran
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India
| | - Subhadeep Roy
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Kolkata, West Bengal, 700054, India.
| |
Collapse
|
19
|
Ngo TKN, Yang SJ, Mao BH, Nguyen TKM, Ng QD, Kuo YL, Tsai JH, Saw SN, Tu TY. A deep learning-based pipeline for analyzing the influences of interfacial mechanochemical microenvironments on spheroid invasion using differential interference contrast microscopic images. Mater Today Bio 2023; 23:100820. [PMID: 37810748 PMCID: PMC10558776 DOI: 10.1016/j.mtbio.2023.100820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Revised: 07/16/2023] [Accepted: 09/24/2023] [Indexed: 10/10/2023] Open
Abstract
Metastasis is the leading cause of cancer-related deaths. During this process, cancer cells are likely to navigate discrete tissue-tissue interfaces, enabling them to infiltrate and spread throughout the body. Three-dimensional (3D) spheroid modeling is receiving more attention due to its strengths in studying the invasive behavior of metastatic cancer cells. While microscopy is a conventional approach for investigating 3D invasion, post-invasion image analysis, which is a time-consuming process, remains a significant challenge for researchers. In this study, we presented an image processing pipeline that utilized a deep learning (DL) solution, with an encoder-decoder architecture, to assess and characterize the invasion dynamics of tumor spheroids. The developed models, equipped with feature extraction and measurement capabilities, could be successfully utilized for the automated segmentation of the invasive protrusions as well as the core region of spheroids situated within interfacial microenvironments with distinct mechanochemical factors. Our findings suggest that a combination of the spheroid culture and DL-based image analysis enable identification of time-lapse migratory patterns for tumor spheroids above matrix-substrate interfaces, thus paving the foundation for delineating the mechanism of local invasion during cancer metastasis.
Collapse
Affiliation(s)
- Thi Kim Ngan Ngo
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Sze Jue Yang
- Department of Artificial Intelligence, Faculty of Computer Science and Information Technology, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Bin-Hsu Mao
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Thi Kim Mai Nguyen
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, Tainan, 70101, Taiwan
| | - Qi Ding Ng
- Department of Artificial Intelligence, Faculty of Computer Science and Information Technology, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Yao-Lung Kuo
- Department of Surgery, College of Medicine, National Cheng Kung University, Tainan, 70101, Taiwan
- Department of Surgery, National Cheng Kung University Hospital, Tainan, 70101, Taiwan
| | - Jui-Hung Tsai
- Department of Internal Medicine, National Cheng Kung University Hospital, Tainan, 70101, Taiwan
| | - Shier Nee Saw
- Department of Artificial Intelligence, Faculty of Computer Science and Information Technology, University of Malaya, 50603, Kuala Lumpur, Malaysia
| | - Ting-Yuan Tu
- Department of Biomedical Engineering, College of Engineering, National Cheng Kung University, Tainan, 70101, Taiwan
- Medical Device Innovation Center, National Cheng Kung University, Tainan, 70101, Taiwan
| |
Collapse
|
20
|
Demir D, Bolgen N, Vaseashta A. Electrospun Nanofibers for Biomedical, Sensing, and Energy Harvesting Functions. Polymers (Basel) 2023; 15:4253. [PMID: 37959933 PMCID: PMC10648854 DOI: 10.3390/polym15214253] [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: 10/01/2023] [Revised: 10/26/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
The process of electrospinning is over a century old, yet novel material and method achievements, and later the addition of nanomaterials in polymeric solutions, have spurred a significant increase in research innovations with several unique applications. Significant improvements have been achieved in the development of electrospun nanofibrous matrices, which include tailoring compositions of polymers with active agents, surface functionalization with nanoparticles, and encapsulation of functional materials within the nanofibers. Recently, sequentially combining fabrication of nanofibers with 3D printing was reported by our group and the synergistic process offers fiber membrane functionalities having the mechanical strength offered by 3D printed scaffolds. Recent developments in electrospun nanofibers are enumerated here with special emphasis on biomedical technologies, chemical and biological sensing, and energy harvesting aspects in the context of e-textile and tactile sensing. Energy harvesting offers significant advantages in many applications, such as biomedical technologies and critical infrastructure protection by using the concept of finite state machines and edge computing. Many other uses of devices using electrospun nanofibers, either as standalone or conjoined with 3D printed materials, are envisaged. The focus of this review is to highlight selected novel applications in biomedical technologies, chem.-bio sensing, and broadly in energy harvesting for use in internet of things (IoT) devices. The article concludes with a brief projection of the future direction of electrospun nanofibers, limitations, and how synergetic combination of the two processes will open pathways for future discoveries.
Collapse
Affiliation(s)
- Didem Demir
- Chemistry and Chemical Process Technologies Department, Mersin Tarsus Organized Industrial Zone Technical Sciences Vocational School, Tarsus University, Mersin 33100, Türkiye;
| | - Nimet Bolgen
- Chemical Engineering Department, Faculty of Engineering, Mersin University, Mersin 33110, Türkiye;
| | - Ashok Vaseashta
- Applied Research, International Clean Water Institute, Manassas, VA 20110, USA
- Institute of Biomedical Engineering and Nanotechnologies, Riga Technical University, LV 1048 Riga, Latvia
| |
Collapse
|
21
|
Shi W, Mirza S, Kuss M, Liu B, Hartin A, Wan S, Kong Y, Mohapatra B, Krishnan M, Band H, Band V, Duan B. Embedded Bioprinting of Breast Tumor Cells and Organoids Using Low-Concentration Collagen-Based Bioinks. Adv Healthc Mater 2023; 12:e2300905. [PMID: 37422447 PMCID: PMC10592394 DOI: 10.1002/adhm.202300905] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 06/30/2023] [Accepted: 07/03/2023] [Indexed: 07/10/2023]
Abstract
Bioinks for 3D bioprinting of tumor models should not only meet printability requirements but also accurately maintain and support phenotypes of tumor surrounding cells to recapitulate key tumor hallmarks. Collagen is a major extracellular matrix protein for solid tumors, but low viscosity of collagen solution has made 3D bioprinted cancer models challenging. This work produces embedded, bioprinted breast cancer cells and tumor organoid models using low-concentration collagen I based bioinks. The biocompatible and physically crosslinked silk fibroin hydrogel is used to generate the support bath for the embedded 3D printing. The composition of the collagen I based bioink is optimized with a thermoresponsive hyaluronic acid-based polymer to maintain the phenotypes of both the noninvasive epithelial and invasive breast cancer cells, as well as cancer-associated fibroblasts. Mouse breast tumor organoids are bioprinted using optimized collagen bioink to mimic in vivo tumor morphology. A vascularized tumor model is also created using a similar strategy, with significantly enhanced vasculature formation under hypoxia. This study shows the great potential of embedded bioprinted breast tumor models utilizing a low-concentration collagen-based bioink for advancing the understanding of tumor cell biology and facilitating drug discovery research.
Collapse
Affiliation(s)
- Wen Shi
- Mary and Dick Holland Regenerative Medicine ProgramUniversity of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Sameer Mirza
- Department of GeneticsCell Biology and AnatomyCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
- Fred and Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNE68198USA
- Department of ChemistryCollege of ScienceUnited Arab Emirates UniversityAbu DhabiUnited Arab Emirates
| | - Mitchell Kuss
- Mary and Dick Holland Regenerative Medicine ProgramUniversity of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Bo Liu
- Mary and Dick Holland Regenerative Medicine ProgramUniversity of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Andrew Hartin
- Mary and Dick Holland Regenerative Medicine ProgramUniversity of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Shibiao Wan
- Department of GeneticsCell Biology and AnatomyCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
- Fred and Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Yunfan Kong
- Mary and Dick Holland Regenerative Medicine ProgramUniversity of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Bhopal Mohapatra
- Department of GeneticsCell Biology and AnatomyCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
- Fred and Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Mena Krishnan
- Mary and Dick Holland Regenerative Medicine ProgramUniversity of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Hamid Band
- Eppley InstituteUniversity of Nebraska Medical CenterOmahaNE68198USA
- Fred and Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Vimla Band
- Department of GeneticsCell Biology and AnatomyCollege of MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
- Fred and Pamela Buffett Cancer CenterUniversity of Nebraska Medical CenterOmahaNE68198USA
| | - Bin Duan
- Mary and Dick Holland Regenerative Medicine ProgramUniversity of Nebraska Medical CenterOmahaNE68198USA
- Division of CardiologyDepartment of Internal MedicineUniversity of Nebraska Medical CenterOmahaNE68198USA
- Department of SurgeryUniversity of Nebraska Medical CenterOmahaNE68198USA
- Department of Mechanical EngineeringUniversity of Nebraska–LincolnLincolnNE68588USA
| |
Collapse
|
22
|
Pasquier E, Rosendahl J, Solberg A, Ståhlberg A, Håkansson J, Chinga-Carrasco G. Polysaccharides and Structural Proteins as Components in Three-Dimensional Scaffolds for Breast Cancer Tissue Models: A Review. Bioengineering (Basel) 2023; 10:682. [PMID: 37370613 PMCID: PMC10295496 DOI: 10.3390/bioengineering10060682] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2023] [Revised: 05/26/2023] [Accepted: 05/30/2023] [Indexed: 06/29/2023] Open
Abstract
Breast cancer is the most common cancer among women, and even though treatments are available, efficiency varies with the patients. In vitro 2D models are commonly used to develop new treatments. However, 2D models overestimate drug efficiency, which increases the failure rate in later phase III clinical trials. New model systems that allow extensive and efficient drug screening are thus required. Three-dimensional printed hydrogels containing active components for cancer cell growth are interesting candidates for the preparation of next generation cancer cell models. Macromolecules, obtained from marine- and land-based resources, can form biopolymers (polysaccharides such as alginate, chitosan, hyaluronic acid, and cellulose) and bioactive components (structural proteins such as collagen, gelatin, and silk fibroin) in hydrogels with adequate physical properties in terms of porosity, rheology, and mechanical strength. Hence, in this study attention is given to biofabrication methods and to the modification with biological macromolecules to become bioactive and, thus, optimize 3D printed structures that better mimic the cancer cell microenvironment. Ink formulations combining polysaccharides for tuning the mechanical properties and bioactive polymers for controlling cell adhesion is key to optimizing the growth of the cancer cells.
Collapse
Affiliation(s)
- Eva Pasquier
- RISE PFI AS, Høgskoleringen 6b, NO-7491 Trondheim, Norway; (E.P.); (A.S.)
| | - Jennifer Rosendahl
- RISE Unit of Biological Function, Division Materials and Production, RISE Research Institutes of Sweden, Box 857, 50115 Borås, Sweden; (J.R.); (J.H.)
| | - Amalie Solberg
- RISE PFI AS, Høgskoleringen 6b, NO-7491 Trondheim, Norway; (E.P.); (A.S.)
| | - Anders Ståhlberg
- Sahlgrenska Center for Cancer Research, Department of Laboratory Medicine, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, 41390 Gothenburg, Sweden;
- Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, 41390 Gothenburg, Sweden
- Department of Clinical Genetics and Genomics, Sahlgrenska University Hospital, 41345 Gothenburg, Sweden
| | - Joakim Håkansson
- RISE Unit of Biological Function, Division Materials and Production, RISE Research Institutes of Sweden, Box 857, 50115 Borås, Sweden; (J.R.); (J.H.)
- Department of Laboratory Medicine, Institute of Biomedicine, University of Gothenburg, 40530 Gothenburg, Sweden
- Department of Chemistry and Molecular Biology, University of Gothenburg, 40530 Gothenburg, Sweden
| | | |
Collapse
|
23
|
Ricci C, Azimi B, Panariello L, Antognoli B, Cecchini B, Rovelli R, Rustembek M, Cinelli P, Milazzo M, Danti S, Lazzeri A. Assessment of Electrospun Poly(ε-caprolactone) and Poly(lactic acid) Fiber Scaffolds to Generate 3D In Vitro Models of Colorectal Adenocarcinoma: A Preliminary Study. Int J Mol Sci 2023; 24:9443. [PMID: 37298394 PMCID: PMC10253282 DOI: 10.3390/ijms24119443] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2023] [Revised: 05/16/2023] [Accepted: 05/24/2023] [Indexed: 06/12/2023] Open
Abstract
Three-dimensional scaffold-based culture has been increasingly gaining influence in oncology as a therapeutic strategy for tumors with a high relapse percentage. This study aims to evaluate electrospun poly(ε-caprolactone) (PCL) and poly(lactic acid) (PLA) scaffolds to create a 3D model of colorectal adenocarcinoma. Specifically, the physico-mechanical and morphological properties of PCL and PLA electrospun fiber meshes collected at different drum velocities, i.e., 500 rpm, 1000 rpm and 2500 rpm, were assessed. Fiber size, mesh porosity, pore size distribution, water contact angle and tensile mechanical properties were investigated. Caco-2 cells were cultured on the produced PCL and PLA scaffolds for 7 days, demonstrating good cell viability and metabolic activity in all the scaffolds. A cross-analysis of the cell-scaffold interactions with morphological, mechanical and surface characterizations of the different electrospun fiber meshes was carried out, showing an opposite trend of cell metabolic activity in PLA and PCL scaffolds regardless of the fiber alignment, which increased in PLA and decreased in PCL. The best samples for Caco-2 cell culture were PCL500 (randomly oriented fibers) and PLA2500 (aligned fibers). Caco-2 cells had the highest metabolic activity in these scaffolds, with Young's moduli in the range of 8.6-21.9 MPa. PCL500 showed Young's modulus and strain at break close to those of the large intestine. Advancements in 3D in vitro models of colorectal adenocarcinoma could move forward the development of therapies for this cancer.
Collapse
Affiliation(s)
- Claudio Ricci
- Department of Civil and Industrial Engineering, University of Pisa, Largo L. Lazzarino 2, 56126 Pisa, Italy
| | - Bahareh Azimi
- Department of Civil and Industrial Engineering, University of Pisa, Largo L. Lazzarino 2, 56126 Pisa, Italy
| | - Luca Panariello
- Department of Civil and Industrial Engineering, University of Pisa, Largo L. Lazzarino 2, 56126 Pisa, Italy
| | - Benedetta Antognoli
- Department of Civil and Industrial Engineering, University of Pisa, Largo L. Lazzarino 2, 56126 Pisa, Italy
| | - Beatrice Cecchini
- Department of Civil and Industrial Engineering, University of Pisa, Largo L. Lazzarino 2, 56126 Pisa, Italy
| | - Roberta Rovelli
- Department of Civil and Industrial Engineering, University of Pisa, Largo L. Lazzarino 2, 56126 Pisa, Italy
| | - Meruyert Rustembek
- Department of Civil and Industrial Engineering, University of Pisa, Largo L. Lazzarino 2, 56126 Pisa, Italy
| | - Patrizia Cinelli
- Department of Civil and Industrial Engineering, University of Pisa, Largo L. Lazzarino 2, 56126 Pisa, Italy
- Centre for Instrumentation Sharing of University of Pisa (CISUP), Lungarno Pacinotti 43/44, 56126 Pisa, Italy
| | - Mario Milazzo
- Department of Civil and Industrial Engineering, University of Pisa, Largo L. Lazzarino 2, 56126 Pisa, Italy
- Centre for Instrumentation Sharing of University of Pisa (CISUP), Lungarno Pacinotti 43/44, 56126 Pisa, Italy
| | - Serena Danti
- Department of Civil and Industrial Engineering, University of Pisa, Largo L. Lazzarino 2, 56126 Pisa, Italy
- Centre for Instrumentation Sharing of University of Pisa (CISUP), Lungarno Pacinotti 43/44, 56126 Pisa, Italy
| | - Andrea Lazzeri
- Department of Civil and Industrial Engineering, University of Pisa, Largo L. Lazzarino 2, 56126 Pisa, Italy
- Centre for Instrumentation Sharing of University of Pisa (CISUP), Lungarno Pacinotti 43/44, 56126 Pisa, Italy
| |
Collapse
|
24
|
Goel R, Gulwani D, Upadhyay P, Sarangthem V, Singh TD. Unsung versatility of elastin-like polypeptide inspired spheroid fabrication: A review. Int J Biol Macromol 2023; 234:123664. [PMID: 36791934 DOI: 10.1016/j.ijbiomac.2023.123664] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2022] [Revised: 01/23/2023] [Accepted: 02/09/2023] [Indexed: 02/15/2023]
Abstract
Lately, 3D cell culture technique has gained a lot of appreciation as a research model. Augmented with technological advancements, the area of 3D cell culture is growing rapidly with a diverse array of scaffolds being tested. This is especially the case for spheroid cultures. The culture of cells as spheroids provides opportunities for unanticipated vision into biological phenomena with its application to drug discovery, metabolic profiling, stem cell research as well as tumor, and disease biology. Spheroid fabrication techniques are broadly categorised into matrix-dependent and matrix-independent techniques. While there is a profusion of spheroid fabrication substrates with substantial biological relevance, an economical, modular, and bio-compatible substrate for high throughput production of spheroids is lacking. In this review, we posit the prospects of elastin-like polypeptides (ELPs) as a broad-spectrum spheroid fabrication platform. Elastin-like polypeptides are nature inspired, size-tunable genetically engineered polymers with wide applicability in various arena of biological considerations, has been employed for spheroid culture with profound utility. The technology offers a cheap, high-throughput, reproducible alternative for spheroid culture with exquisite adaptability. Here, we will brief the applicability of 3D cultures as compared to 2D cultures with spheroids being the focal point of the review. Common approaches to spheroid fabrication are discussed with existential limitations. Finally, the versatility of elastin-like polypeptide inspired substrates for spheroid culture has been discussed.
Collapse
Affiliation(s)
- Ridhima Goel
- Department of Medical Oncology Laboratory, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Deepak Gulwani
- Department of Medical Oncology Laboratory, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Priyanka Upadhyay
- Department of Medical Oncology Laboratory, All India Institute of Medical Sciences, New Delhi 110029, India
| | - Vijaya Sarangthem
- Department of Pathology, All India Institute of Medical Sciences, New Delhi 110029, India.
| | - Thoudam Debraj Singh
- Department of Medical Oncology Laboratory, All India Institute of Medical Sciences, New Delhi 110029, India.
| |
Collapse
|
25
|
Mitrakas AG, Tsolou A, Didaskalou S, Karkaletsou L, Efstathiou C, Eftalitsidis E, Marmanis K, Koffa M. Applications and Advances of Multicellular Tumor Spheroids: Challenges in Their Development and Analysis. Int J Mol Sci 2023; 24:ijms24086949. [PMID: 37108113 PMCID: PMC10138394 DOI: 10.3390/ijms24086949] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Revised: 03/31/2023] [Accepted: 04/06/2023] [Indexed: 04/29/2023] Open
Abstract
Biomedical research requires both in vitro and in vivo studies in order to explore disease processes or drug interactions. Foundational investigations have been performed at the cellular level using two-dimensional cultures as the gold-standard method since the early 20th century. However, three-dimensional (3D) cultures have emerged as a new tool for tissue modeling over the last few years, bridging the gap between in vitro and animal model studies. Cancer has been a worldwide challenge for the biomedical community due to its high morbidity and mortality rates. Various methods have been developed to produce multicellular tumor spheroids (MCTSs), including scaffold-free and scaffold-based structures, which usually depend on the demands of the cells used and the related biological question. MCTSs are increasingly utilized in studies involving cancer cell metabolism and cell cycle defects. These studies produce massive amounts of data, which demand elaborate and complex tools for thorough analysis. In this review, we discuss the advantages and disadvantages of several up-to-date methods used to construct MCTSs. In addition, we also present advanced methods for analyzing MCTS features. As MCTSs more closely mimic the in vivo tumor environment, compared to 2D monolayers, they can evolve to be an appealing model for in vitro tumor biology studies.
Collapse
Affiliation(s)
- Achilleas G Mitrakas
- Cell Biology Lab, Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Avgi Tsolou
- Cell Biology Lab, Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Stylianos Didaskalou
- Cell Biology Lab, Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Lito Karkaletsou
- Cell Biology Lab, Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Christos Efstathiou
- Cell Biology Lab, Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Evgenios Eftalitsidis
- Cell Biology Lab, Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Konstantinos Marmanis
- Cell Biology Lab, Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| | - Maria Koffa
- Cell Biology Lab, Department of Molecular Biology and Genetics, Democritus University of Thrace, 68100 Alexandroupolis, Greece
| |
Collapse
|
26
|
Chen YS, Ng HY, Chen YW, Cho DY, Ho CC, Chen CY, Chiu SC, Jhong YR, Shie MY. Additive manufacturing of Schwann cell-laden collagen/alginate nerve guidance conduits by freeform reversible embedding regulate neurogenesis via exosomes secretion towards peripheral nerve regeneration. BIOMATERIALS ADVANCES 2023; 146:213276. [PMID: 36640522 DOI: 10.1016/j.bioadv.2022.213276] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2022] [Revised: 12/09/2022] [Accepted: 12/29/2022] [Indexed: 01/02/2023]
Abstract
Peripheral nerve injury is a common clinical problem that could be debilitating to one's quality of life. The complex nerve guidance conduits (NGCs) with cells in order to improve nerve regeneration. Therefore, we used freeform reversible embedding of suspended hydrogels to fabricate Schwann cells (SCs)-laden collagen/alginate (Col/Alg) NGCs. First, we evaluated Col influence on the characteristics of NGCs. After which, Wharton's jelly mesenchymal stem cells (WJMSC) are seeded onto the inner channel of NGCs and evaluated neural regeneration behaviors. Results indicated the SCs-laden NGCs with 2.5 % Col found the highest proliferation and secretion of neurotrophic protein. Furthermore, co-culture of SCs promoted differentiation of WJMSC as seen from the increased neurogenic-related protein in NGCs. To determine the molecular mechanism between SCs and WJMSC, we demonstrated the neurotrophic factors secreted by SCs act on tropomyosin receptor kinase A (TrkA) receptors of WJMSC to promote nerve regeneration. In addition, our study demonstrated SCs-derived exosomes had a critical role in regulating neural differentiation of WJMSC. Taken together, this study demonstrates the fabrication of SCs-laden Col/Alg NGCs for nerve regeneration and understanding regarding the synergistic regenerative mechanisms of different cells could bring us a step closer for clinical treatment of large nerve defects.
Collapse
Affiliation(s)
- Yueh-Sheng Chen
- School of Chinese Medicine, China Medical University, Taichung 40447, Taiwan; Department of Bioinformatics and Medical Engineering, Asia University, Taichung City 41354, Taiwan
| | - Hooi Yee Ng
- Department of Education, China Medical University Hospital, Taichung 404332, Taiwan
| | - Yi-Wen Chen
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung City 406040, Taiwan; x-Dimension Center for Medical Research and Translation, China Medical University Hospital, Taichung City 404332, Taiwan
| | - Der-Yang Cho
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung City 406040, Taiwan; Department of Neurosurgery, China Medical University Hospital, Taichung, Taiwan; Translational Cell Therapy Center, China Medical University Hospital, Taichung, Taiwan
| | - Chia-Che Ho
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung City 41354, Taiwan; High Performance Materials Institute for x-Dimensional Printing, Asia University, Taichung City 41354, Taiwan
| | - Cheng-Yu Chen
- x-Dimension Center for Medical Research and Translation, China Medical University Hospital, Taichung City 404332, Taiwan
| | - Shao-Chih Chiu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung City 406040, Taiwan; Translational Cell Therapy Center, China Medical University Hospital, Taichung, Taiwan
| | - Yi-Rong Jhong
- x-Dimension Center for Medical Research and Translation, China Medical University Hospital, Taichung City 404332, Taiwan
| | - Ming-You Shie
- Department of Bioinformatics and Medical Engineering, Asia University, Taichung City 41354, Taiwan; x-Dimension Center for Medical Research and Translation, China Medical University Hospital, Taichung City 404332, Taiwan; School of Dentistry, China Medical University, Taichung City 406040, Taiwan.
| |
Collapse
|
27
|
Redmond J, McCarthy HO, Buchanan P, Levingstone TJ, Dunne NJ. Development and characterisation of 3D collagen-gelatin based scaffolds for breast cancer research. BIOMATERIALS ADVANCES 2022; 142:213157. [PMID: 36279748 DOI: 10.1016/j.bioadv.2022.213157] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/20/2022] [Accepted: 10/12/2022] [Indexed: 06/16/2023]
Abstract
While 2D culture presents a useful tool for cancer research, it fails to replicate the tumor microenvironment as it lacks proper three-dimensional cell-cell/cell-matrix interactions, often resulting in exaggerated responses to therapeutic agents. 3D models that aim to overcome the issues associated with 2D culture research offer a new frontier for cancer research with cell growth, morphology and genetic properties that more closely match in vivo cancers. Herein, we aim to develop a collagen-based scaffold that supports the attachment and proliferation of breast cancer (BC) cells as a 3D culture model. Scaffolds were produced on a repeatable basis using a freeze-drying procedure. The constructs were highly porous (>99%) with homogenous pore sizes (150-300 μm) and an interconnected structure. The application of 1-Ethyl-3-(3-dimethylaminopropyl)carbodiimide (EDAC) crosslinking resulted in scaffolds with elastic moduli in the range of 1-2 kPa, mimicking cancerous breast tissue stiffness. Furthermore, the incorporation of gelatin into the scaffolds enabled the porosity, pore size and mechanical properties to be tailored, resulting in scaffolds with stiffness values that accurately replicate the stiffness of human BC extracellular matrix (ECM) (1.3-1.7 kPa). Scaffolds displayed high in vitro stability with 90% of mass remaining after 14 days of culture. The scaffolds were shown to be highly biocompatible, and capable of supporting the attachment, infiltration and proliferation of MCF7 breast cancer (BC) cells over +14 days. These results confirm the suitability of these scaffolds as culture models for BC cells. These collagen-based scaffolds offer significant potential for the exploration of aspects of BC, such as gene expression profiles and patterns, and for the assessment of the efficacy of therapeutic agents in treating BC.
Collapse
Affiliation(s)
- John Redmond
- School of Mechanical and Manufacturing Engineering, Dublin City University, Collins Avenue, Dublin 9, Ireland; Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, Stokes Building, Collins Avenue, Dublin 9, Ireland.
| | - Helen O McCarthy
- School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom; School of Chemical Sciences, Dublin City University, Collins Avenue, Dublin 9, Ireland
| | - Paul Buchanan
- School of Nursing and Human Science, Dublin City University, Collins Avenue, Dublin, Ireland; National Institute of Cellular Biotechnology, Dublin City University, Dublin 9, Ireland
| | - Tanya J Levingstone
- School of Mechanical and Manufacturing Engineering, Dublin City University, Collins Avenue, Dublin 9, Ireland; Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, Stokes Building, Collins Avenue, Dublin 9, Ireland; Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; Advanced Manufacturing Research Centre (I-Form), School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; Advanced Processing Technology Research Centre, Dublin City University, Dublin 9, Ireland; Biodesign Europe, Dublin City University, Dublin 9, Ireland.
| | - Nicholas J Dunne
- School of Mechanical and Manufacturing Engineering, Dublin City University, Collins Avenue, Dublin 9, Ireland; Centre for Medical Engineering Research, School of Mechanical and Manufacturing Engineering, Dublin City University, Stokes Building, Collins Avenue, Dublin 9, Ireland; School of Pharmacy, Queen's University Belfast, 97 Lisburn Road, Belfast BT9 7BL, United Kingdom; Trinity Centre for Biomedical Engineering, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin 2, Ireland; Department of Mechanical and Manufacturing Engineering, School of Engineering, Trinity College Dublin, Dublin 2, Ireland; Advanced Materials and Bioengineering Research Centre (AMBER), Royal College of Surgeons in Ireland and Trinity College Dublin, Dublin 2, Ireland; Advanced Manufacturing Research Centre (I-Form), School of Mechanical and Manufacturing Engineering, Dublin City University, Dublin 9, Ireland; Advanced Processing Technology Research Centre, Dublin City University, Dublin 9, Ireland; Biodesign Europe, Dublin City University, Dublin 9, Ireland.
| |
Collapse
|
28
|
Azimian Zavareh V, Rafiee L, Sheikholeslam M, Shariati L, Vaseghi G, Savoji H, Haghjooy Javanmard S. Three-Dimensional in Vitro Models: A Promising Tool To Scale-Up Breast Cancer Research. ACS Biomater Sci Eng 2022; 8:4648-4672. [PMID: 36260561 DOI: 10.1021/acsbiomaterials.2c00277] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Common models used in breast cancer studies, including two-dimensional (2D) cultures and animal models, do not precisely model all aspects of breast tumors. These models do not well simulate the cell-cell and cell-stromal interactions required for normal tumor growth in the body and lake tumor like microenvironment. Three-dimensional (3D) cell culture models are novel approaches to studying breast cancer. They do not have the restrictions of these conventional models and are able to recapitulate the structural architecture, complexity, and specific function of breast tumors and provide similar in vivo responses to therapeutic regimens. These models can be a link between former traditional 2D culture and in vivo models and are necessary for further studies in cancer. This review attempts to summarize the most common 3D in vitro models used in breast cancer studies, including scaffold-free (spheroid and organoid), scaffold-based, and chip-based models, particularly focused on the basic and translational application of these 3D models in drug screening and the tumor microenvironment in breast cancer.
Collapse
Affiliation(s)
- Vajihe Azimian Zavareh
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 81746 73461, Iran.,Core Research Facilities (CRF), Isfahan University of Medical Sciences, Isfahan 81746 73461, Iran
| | - Laleh Rafiee
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 81746 73461, Iran
| | - Mohammadali Sheikholeslam
- Department of Biomaterials, Nanotechnology and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan 81746 73461, Iran.,Biosensor Research Center, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan 81746 73461, Iran
| | - Laleh Shariati
- Department of Biomaterials, Nanotechnology and Tissue Engineering, School of Advanced Technologies in Medicine, Isfahan University of Medical Sciences, Isfahan 81746 73461, Iran.,Cancer Prevention Research Center, Omid Hospital, Isfahan University of Medical Sciences, Isfahan 81746 73461, Iran
| | - Golnaz Vaseghi
- Isfahan Cardiovascular Research Center, Isfahan Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 81746 73461, Iran
| | - Houman Savoji
- Institute of Biomedical Engineering, Department of Pharmacology and Physiology, Faculty of Medicine, University of Montreal, Montreal, QC H3T 1J4, Canada.,Research Center, Centre Hospitalier Universitaire Sainte-Justine, Montreal, QC H3T 1C5, Canada.,Montreal TransMedTech Institute, Montreal, QC H3T 1J4, Canada
| | - Shaghayegh Haghjooy Javanmard
- Applied Physiology Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan 81746 73461, Iran
| |
Collapse
|
29
|
Sencha LM, Dobrynina OE, Pospelov AD, Guryev EL, Peskova NN, Brilkina AA, Cherkasova EI, Balalaeva IV. Real-Time Fluorescence Visualization and Quantitation of Cell Growth and Death in Response to Treatment in 3D Collagen-Based Tumor Model. Int J Mol Sci 2022; 23:ijms23168837. [PMID: 36012102 PMCID: PMC9408454 DOI: 10.3390/ijms23168837] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 08/07/2022] [Accepted: 08/08/2022] [Indexed: 11/24/2022] Open
Abstract
The use of 3D in vitro tumor models has become a common trend in cancer biology studies as well as drug screening and preclinical testing of drug candidates. The transition from 2D to 3D matrix-based cell cultures requires modification of methods for assessing tumor growth. We propose the method for assessing the growth of tumor cells in a collagen hydrogel using macro-scale registration and quantification of the gel epi-fluorescence. The technique does not require gel destruction, can be used for real-time observation of fast (in seconds) cellular responses and demonstrates high agreement with cell counting approaches or measuring total DNA content. The potency of the method was proven in experiments aimed at testing cytotoxic activity of chemotherapeutic drug (cisplatin) and recombinant targeted toxin (DARPin-LoPE) against two different tumor cell lines genetically labelled with fluorescent proteins. Moreover, using fluorescent proteins with sensor properties allows registration of dynamic changes in cells’ metabolism, which was shown for the case of sensor of caspase 3 activity.
Collapse
|
30
|
Enhanced Adipogenic Differentiation of Human Dental Pulp Stem Cells in Enzymatically Decellularized Adipose Tissue Solid Foams. BIOLOGY 2022; 11:biology11081099. [PMID: 35892955 PMCID: PMC9394288 DOI: 10.3390/biology11081099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 07/18/2022] [Accepted: 07/20/2022] [Indexed: 12/03/2022]
Abstract
Simple Summary Obesity shortens human lifespan and represents one of the most important public health problems causing significant economic and societal consequences worldwide. However, the current development of physiological human 3D adipose tissue models for in vitro research on preclinical personalized medicine is limited and expensive. Here, we designed, produced, and characterized 3D solid foams using a mixture of bovine collagen I and decellularized human adipose tissue to serve as a 3D matrix mimicking in vivo adipose microenvironment for cell culture purposes. Furthermore, we sought to validate its compatibility for the culture of human mesenchymal stem cells isolated from the dental pulp. We demonstrated that 3D solid foams are able to integrate the stem cells from the dental pulp and provide the appropriate cues to differentiate them into mature adipocytes. The results represent an advance of in vitro 3D models using a human extracellular matrix derived material for future personalized stem cell therapies. Abstract Engineered 3D human adipose tissue models and the development of physiological human 3D in vitro models to test new therapeutic compounds and advance in the study of pathophysiological mechanisms of disease is still technically challenging and expensive. To reduce costs and develop new technologies to study human adipogenesis and stem cell differentiation in a controlled in vitro system, here we report the design, characterization, and validation of extracellular matrix (ECM)-based materials of decellularized human adipose tissue (hDAT) or bovine collagen-I (bCOL-I) for 3D adipogenic stem cell culture. We aimed at recapitulating the dynamics, composition, and structure of the native ECM to optimize the adipogenic differentiation of human mesenchymal stem cells. hDAT was obtained by a two-enzymatic step decellularization protocol and post-processed by freeze-drying to produce 3D solid foams. These solid foams were employed either as pure hDAT, or combined with bCOL-I in a 3:1 proportion, to recreate a microenvironment compatible with stem cell survival and differentiation. We sought to investigate the effect of the adipogenic inductive extracellular 3D-microenvironment on human multipotent dental pulp stem cells (hDPSCs). We found that solid foams supported hDPSC viability and proliferation. Incubation of hDPSCs with adipogenic medium in hDAT-based solid foams increased the expression of mature adipocyte LPL and c/EBP gene markers as determined by RT-qPCR, with respect to bCOL-I solid foams. Moreover, hDPSC capability to differentiate towards adipocytes was assessed by PPAR-γ immunostaining and Oil-red lipid droplet staining. We found out that both hDAT and mixed 3:1 hDAT-COL-I solid foams could support adipogenesis in 3D-hDPSC stem cell cultures significantly more efficiently than solid foams of bCOL-I, opening the possibility to obtain hDAT-based solid foams with customized properties. The combination of human-derived ECM biomaterials with synthetic proteins can, thus, be envisaged to reduce fabrication costs, thus facilitating the widespread use of autologous stem cells and biomaterials for personalized medicine.
Collapse
|
31
|
Wang Z, Yang Y, Gao Y, Xu Z, Yang S, Jin M. Establishing a novel 3D printing bioinks system with recombinant human collagen. Int J Biol Macromol 2022; 211:400-409. [PMID: 35577188 DOI: 10.1016/j.ijbiomac.2022.05.088] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/08/2022] [Accepted: 05/10/2022] [Indexed: 11/19/2022]
Abstract
Bioinks are one of the key elements in realizing three-dimensional (3D) bioprinting. However, bioinks prepared from conventional collagen are hindered to their further applications due to concerns of collagen purity, unstable mechanical properties, and low solubility under neutralized conditions. This study aimed to develop a reliable UV-curable bioink system from a novel water-soluble recombinant human collagen (RHC). RHC was modified by methacrylic anhydride (MAA) and later crosslinked by 1-ethyl-3-(3-dimethylaminopropyl) carbodiimide hydrochloride (EDC)/N-hydroxysuccinimide (NHS) to obtain Pro-RHCMA. 1H nuclear magnetic resonance (1H NMR) confirmed the methacryloyl grafts, Fourier transform-infrared spectroscopy (FT-IR) illustrated the chemical crosslinking in producing the Pro-RHCMA. Internal morphology, mechanical properties and degradation of UV cured boinks were MAA and EDC/NHS modification-dependent. Photorheological properties and printability of the bioinks were determined. Cellular bioactivities were sustained within the printed bioinks, validating the bioinks biocompatibility in vitro. Finally, qRT-PCR revealed that the Pro-RHCMA bioinks provided a cell-friendly microenvironment for human umbilical vein endothelial cells (HUVECs) and human foreskin fibroblasts (HFFs), by supporting the expression of extracellular matrix (ECM) and angiogenesis-associated proteins, respectively. Taken together, this novel RHC-based bioink system shows great potential in tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Zixun Wang
- School of Environmental and Biological Engineering, Nanjing University of Science & Technology, Nanjing 210094, PR China
| | - Yang Yang
- School of Environmental and Biological Engineering, Nanjing University of Science & Technology, Nanjing 210094, PR China; Biomanufacturing Center, Department of Mechanical Engineering, Tsinghua University, Beijing 100084, PR China.
| | - Yunbo Gao
- Beijing Tongren Hospital, Capital Medical University, Beijing 100730, PR China
| | - Zhaoxian Xu
- School of Environmental and Biological Engineering, Nanjing University of Science & Technology, Nanjing 210094, PR China
| | - Shulin Yang
- School of Environmental and Biological Engineering, Nanjing University of Science & Technology, Nanjing 210094, PR China
| | - Mingjie Jin
- School of Environmental and Biological Engineering, Nanjing University of Science & Technology, Nanjing 210094, PR China
| |
Collapse
|
32
|
Sun M, Huang K, Luo X, Li H. Templated Three-Dimensional Engineered Bone Matrix as a Model for Breast Cancer Osteolytic Bone Metastasis Process. Int J Nanomedicine 2022; 16:8391-8403. [PMID: 35002234 PMCID: PMC8727640 DOI: 10.2147/ijn.s338609] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2021] [Accepted: 12/13/2021] [Indexed: 11/30/2022] Open
Abstract
Purpose Bone metastasis is one of the common causes of death relative to breast cancer. However, the evolvement of bone niche in cancer progression remains poorly understood. A three-dimensional (3D) engineered bone matrix was developed as an effective biomimetic model to explore the mechanism relative to bone cancer metastasis. Methods In the study, a 3D engineered bone matrix was developed via cell biomineralization templated by a biomimetic collagen template. The process of bone metastasis relative to breast cancer was investigated by co-culturing breast cancer MDA-MB-231-GFP cells with pre-osteogenic MC3T3-E1 cells on the 3D bone matrix. Results A typical bone matrix was obtained, where mineralized collagen fibers were packed into the bundle to form a 3D engineered bone matrix. As the cancer cells were invading along the way vertical to the alignment of mineralized collagen fiber, the bone matrix gradually became thinner, accompanied with the erosion of Col I and the loss of calcium and phosphorus. As a result, the disassembled structure of mineralized collagen fiber was observed, which may be attributed to osteolytic bone metastasis. Conclusion An engineered 3D bone-like matrix was successfully prepared via cell mineralization, which can act as a model for bone metastasis process. The study revealed mineralized collagen fiber disassembled at nanoscale relative to breast cancer cells.
Collapse
Affiliation(s)
- Manman Sun
- Department of Materials Science and Engineering, College of Chemistry and Materials Science, Jinan University, Guangzhou, Guangdong, People's Republic of China
| | - Ke Huang
- Department of Materials Science and Engineering, College of Chemistry and Materials Science, Jinan University, Guangzhou, Guangdong, People's Republic of China
| | - Xueshi Luo
- The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, Guangdong, People's Republic of China.,Department of Chemistry, College of Chemistry and Materials Science, Jinan University, Guangzhou, Guangdong, People's Republic of China
| | - Hong Li
- Department of Materials Science and Engineering, College of Chemistry and Materials Science, Jinan University, Guangzhou, Guangdong, People's Republic of China
| |
Collapse
|
33
|
Herring B, Jang S, Whitt J, Goliwas K, Aburjania Z, Dudeja V, Ren B, Berry J, Bibb J, Frost A, Chen H, Rose JB, Jaskula-Sztul R. Ex Vivo Modeling of Human Neuroendocrine Tumors in Tissue Surrogates. Front Endocrinol (Lausanne) 2021; 12:710009. [PMID: 35002949 PMCID: PMC8734644 DOI: 10.3389/fendo.2021.710009] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 11/10/2021] [Indexed: 12/24/2022] Open
Abstract
Few models exist for studying neuroendocrine tumors (NETs), and there are mounting concerns that the currently available array of cell lines is not representative of NET biology. The lack of stable patient-derived NET xenograft models further limits the scientific community's ability to make conclusions about NETs and their response to therapy in patients. To address these limitations, we propose the use of an ex vivo 3D flow-perfusion bioreactor system for culturing and studying patient-derived NET surrogates. Herein, we demonstrate the utility of the bioreactor system for culturing NET surrogates and provide methods for evaluating the efficacy of therapeutic agents on human NET cell line xenograft constructs and patient-derived NET surrogates. We also demonstrate that patient-derived NET tissues can be propagated using the bioreactor system and investigate the near-infrared (NIR) dye IR-783 for its use in monitoring their status within the bioreactor. The results indicate that the bioreactor system and similar 3D culture models may be valuable tools for culturing patient-derived NETs and monitoring their response to therapy ex vivo.
Collapse
Affiliation(s)
- Brendon Herring
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Samuel Jang
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Jason Whitt
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Kayla Goliwas
- Department of Medicine, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Zviadi Aburjania
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Vikas Dudeja
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Bin Ren
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Joel Berry
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL, United States
| | - James Bibb
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Andra Frost
- Department of Pathology, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Herbert Chen
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - John Bart Rose
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Renata Jaskula-Sztul
- Department of Surgery, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
34
|
Wu D, Berg J, Arlt B, Röhrs V, Al-Zeer MA, Deubzer HE, Kurreck J. Bioprinted Cancer Model of Neuroblastoma in a Renal Microenvironment as an Efficiently Applicable Drug Testing Platform. Int J Mol Sci 2021; 23:ijms23010122. [PMID: 35008547 PMCID: PMC8745467 DOI: 10.3390/ijms23010122] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 12/10/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022] Open
Abstract
Development of new anticancer drugs with currently available animal models is hampered by the fact that human cancer cells are embedded in an animal-derived environment. Neuroblastoma is the most common extracranial solid malignancy of childhood. Major obstacles include managing chemotherapy-resistant relapses and resistance to induction therapy, leading to early death in very-high-risk patients. Here, we present a three-dimensional (3D) model for neuroblastoma composed of IMR-32 cells with amplified genes of the myelocytomatosis viral related oncogene MYCN and the anaplastic lymphoma kinase (ALK) in a renal environment of exclusively human origin, made of human embryonic kidney 293 cells and primary human kidney fibroblasts. The model was produced with two pneumatic extrusion printheads using a commercially available bioprinter. Two drugs were exemplarily tested in this model: While the histone deacetylase inhibitor panobinostat selectively killed the cancer cells by apoptosis induction but did not affect renal cells in the therapeutically effective concentration range, the peptidyl nucleoside antibiotic blasticidin induced cell death in both cell types. Importantly, differences in sensitivity between two-dimensional (2D) and 3D cultures were cell-type specific, making the therapeutic window broader in the bioprinted model and demonstrating the value of studying anticancer drugs in human 3D models. Altogether, this cancer model allows testing cytotoxicity and tumor selectivity of new anticancer drugs, and the open scaffold design enables the free exchange of tumor and microenvironment by any cell type.
Collapse
Affiliation(s)
- Dongwei Wu
- Institute of Biotechnology, Chair of Applied Biochemistry, Technische Universität Berlin, 13355 Berlin, Germany; (D.W.); (J.B.); (V.R.); (M.A.A.-Z.)
| | - Johanna Berg
- Institute of Biotechnology, Chair of Applied Biochemistry, Technische Universität Berlin, 13355 Berlin, Germany; (D.W.); (J.B.); (V.R.); (M.A.A.-Z.)
| | - Birte Arlt
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany; (B.A.); (H.E.D.)
| | - Viola Röhrs
- Institute of Biotechnology, Chair of Applied Biochemistry, Technische Universität Berlin, 13355 Berlin, Germany; (D.W.); (J.B.); (V.R.); (M.A.A.-Z.)
| | - Munir A. Al-Zeer
- Institute of Biotechnology, Chair of Applied Biochemistry, Technische Universität Berlin, 13355 Berlin, Germany; (D.W.); (J.B.); (V.R.); (M.A.A.-Z.)
| | - Hedwig E. Deubzer
- Department of Pediatric Hematology and Oncology, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany; (B.A.); (H.E.D.)
- Neuroblastoma Research Group, Experimental and Clinical Research Center (ECRC) of the Charité and the Max-Delbrück-Center for Molecular Medicine (MDC) in the Helmholtz Association, 13125 Berlin, Germany
- German Cancer Consortium (Deutsches Konsortium für Translationale Krebsforschung, DKTK), Partner Site Berlin, 10115 Berlin, Germany
- Berliner Institut für Gesundheitsforschung (BIH), 10178 Berlin, Germany
| | - Jens Kurreck
- Institute of Biotechnology, Chair of Applied Biochemistry, Technische Universität Berlin, 13355 Berlin, Germany; (D.W.); (J.B.); (V.R.); (M.A.A.-Z.)
- Correspondence: ; Tel.: +49-30-314-27-582; Fax: +49-30-314-27-502
| |
Collapse
|
35
|
Habanjar O, Diab-Assaf M, Caldefie-Chezet F, Delort L. 3D Cell Culture Systems: Tumor Application, Advantages, and Disadvantages. Int J Mol Sci 2021; 22:12200. [PMID: 34830082 PMCID: PMC8618305 DOI: 10.3390/ijms222212200] [Citation(s) in RCA: 256] [Impact Index Per Article: 64.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2021] [Revised: 11/05/2021] [Accepted: 11/07/2021] [Indexed: 01/09/2023] Open
Abstract
The traditional two-dimensional (2D) in vitro cell culture system (on a flat support) has long been used in cancer research. However, this system cannot be fully translated into clinical trials to ideally represent physiological conditions. This culture cannot mimic the natural tumor microenvironment due to the lack of cellular communication (cell-cell) and interaction (cell-cell and cell-matrix). To overcome these limitations, three-dimensional (3D) culture systems are increasingly developed in research and have become essential for tumor research, tissue engineering, and basic biology research. 3D culture has received much attention in the field of biomedicine due to its ability to mimic tissue structure and function. The 3D matrix presents a highly dynamic framework where its components are deposited, degraded, or modified to delineate functions and provide a platform where cells attach to perform their specific functions, including adhesion, proliferation, communication, and apoptosis. So far, various types of models belong to this culture: either the culture based on natural or synthetic adherent matrices used to design 3D scaffolds as biomaterials to form a 3D matrix or based on non-adherent and/or matrix-free matrices to form the spheroids. In this review, we first summarize a comparison between 2D and 3D cultures. Then, we focus on the different components of the natural extracellular matrix that can be used as supports in 3D culture. Then we detail different types of natural supports such as matrigel, hydrogels, hard supports, and different synthetic strategies of 3D matrices such as lyophilization, electrospiding, stereolithography, microfluid by citing the advantages and disadvantages of each of them. Finally, we summarize the different methods of generating normal and tumor spheroids, citing their respective advantages and disadvantages in order to obtain an ideal 3D model (matrix) that retains the following characteristics: better biocompatibility, good mechanical properties corresponding to the tumor tissue, degradability, controllable microstructure and chemical components like the tumor tissue, favorable nutrient exchange and easy separation of the cells from the matrix.
Collapse
Affiliation(s)
- Ola Habanjar
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France; (O.H.); (F.C.-C.)
| | - Mona Diab-Assaf
- Equipe Tumorigénèse Pharmacologie Moléculaire et Anticancéreuse, Faculté des Sciences II, Université Libanaise Fanar, Beyrouth 1500, Liban;
| | - Florence Caldefie-Chezet
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France; (O.H.); (F.C.-C.)
| | - Laetitia Delort
- Université Clermont-Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH-Auvergne, 63000 Clermont-Ferrand, France; (O.H.); (F.C.-C.)
| |
Collapse
|
36
|
Wang J, Xu W, Qian J, Wang Y, Hou G, Suo A. Photo-crosslinked hyaluronic acid hydrogel as a biomimic extracellular matrix to recapitulate in vivo features of breast cancer cells. Colloids Surf B Biointerfaces 2021; 209:112159. [PMID: 34687973 DOI: 10.1016/j.colsurfb.2021.112159] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2021] [Revised: 10/01/2021] [Accepted: 10/10/2021] [Indexed: 11/26/2022]
Abstract
2D cell culture is widely utilized to develop anti-cancer drugs and to explore the mechanisms of cancer tumorigenesis and development. However, the findings obtained from 2D culture often fail to provide guidance for clinical tumor treatments since it cannot precisely replicate the features of real tumors. 3D tumor models capable of recapitulating native tumor microenvironments have been proved to be a promising alternative technique. Herein, we constructed a breast tumor model from novel hyaluronic acid (HA) hydrogel which was prepared through photocrosslinking of methacrylated HA. The hydrogel was used as a biomimetic extracellular matrix to incubate MCF-7 cells. It was found that methacrylation degree had great effects on hydrogel's microstructure, mechanical performances, and liquid-absorbing and degradation abilities. Optimized hydrogel exhibited highly porous morphology, high equilibrium swelling ratio, suitable mechanical properties, and hyaluronidase-responsive degradation behavior. The results demonstrated that the HA hydrogel facilitated MCF-7 cell proliferation and growth in an aggregation manner. Furthermore, 3D-cultured MCF-7 cells not only up-regulated the expression of VEGF, bFGF and interleukin-8 but exhibited greater invasion and tumorigenesis capabilities compared with 2D-cultured cells. Therefore, the HA hydrogel is a reliable substitute for tumor model construction.
Collapse
Affiliation(s)
- Jinlei Wang
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an 710049, China; Northwest Institute for Non-ferrous Metal Research, Xi'an 710016, China
| | - Weijun Xu
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an 710049, China
| | - Junmin Qian
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an 710049, China.
| | - Yaping Wang
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an 710049, China
| | - Guanghui Hou
- State Key Laboratory for Mechanical Behavior of Materials, Xi'an Jiaotong University, Xi'an 710049, China
| | - Aili Suo
- Department of Oncology, The First Affiliated Hospital of Xi'an Jiaotong University, Xi'an 710061, China.
| |
Collapse
|
37
|
Cimen Z, Babadag S, Odabas S, Altuntas S, Demirel G, Demirel GB. Injectable and Self-Healable pH-Responsive Gelatin–PEG/Laponite Hybrid Hydrogels as Long-Acting Implants for Local Cancer Treatment. ACS APPLIED POLYMER MATERIALS 2021; 3:3504-3518. [DOI: 10.1021/acsapm.1c00419] [Citation(s) in RCA: 46] [Impact Index Per Article: 11.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
Affiliation(s)
- Zeynep Cimen
- Department of Chemistry, Polatlı Faculty of Arts and Sciences, Ankara Hacı Bayram Veli University, Ankara 06900, Turkey
- Department of Chemistry, Institute of Natural and Applied Sciences, Gazi University, Ankara 06560, Turkey
| | - Sena Babadag
- Biomaterials and Tissue Engineering Laboratory (bteLAB), Department of Chemistry, Faculty of Science, Ankara University, Ankara 06560, Turkey
- Interdisciplinary Research Unit for Advanced Materials (INTRAM), Ankara University, Ankara 06560, Turkey
| | - Sedat Odabas
- Biomaterials and Tissue Engineering Laboratory (bteLAB), Department of Chemistry, Faculty of Science, Ankara University, Ankara 06560, Turkey
- Interdisciplinary Research Unit for Advanced Materials (INTRAM), Ankara University, Ankara 06560, Turkey
| | - Sevde Altuntas
- Tissue Engineering Department, University of Health Sciences Turkey, Istanbul 34662, Turkey
- Experimental Medicine Research and Application Center, Validebag Research
Park, University of Health Sciences, Istanbul 34662, Turkey
| | - Gokhan Demirel
- Department of Chemistry, Faculty of Science, Gazi University, Ankara 06500, Turkey
| | - Gokcen Birlik Demirel
- Department of Chemistry, Polatlı Faculty of Arts and Sciences, Ankara Hacı Bayram Veli University, Ankara 06900, Turkey
| |
Collapse
|
38
|
Puertas-Bartolomé M, Mora-Boza A, García-Fernández L. Emerging Biofabrication Techniques: A Review on Natural Polymers for Biomedical Applications. Polymers (Basel) 2021; 13:1209. [PMID: 33918049 PMCID: PMC8069319 DOI: 10.3390/polym13081209] [Citation(s) in RCA: 53] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 04/01/2021] [Accepted: 04/03/2021] [Indexed: 12/21/2022] Open
Abstract
Natural polymers have been widely used for biomedical applications in recent decades. They offer the advantages of resembling the extracellular matrix of native tissues and retaining biochemical cues and properties necessary to enhance their biocompatibility, so they usually improve the cellular attachment and behavior and avoid immunological reactions. Moreover, they offer a rapid degradability through natural enzymatic or chemical processes. However, natural polymers present poor mechanical strength, which frequently makes the manipulation processes difficult. Recent advances in biofabrication, 3D printing, microfluidics, and cell-electrospinning allow the manufacturing of complex natural polymer matrixes with biophysical and structural properties similar to those of the extracellular matrix. In addition, these techniques offer the possibility of incorporating different cell lines into the fabrication process, a revolutionary strategy broadly explored in recent years to produce cell-laden scaffolds that can better mimic the properties of functional tissues. In this review, the use of 3D printing, microfluidics, and electrospinning approaches has been extensively investigated for the biofabrication of naturally derived polymer scaffolds with encapsulated cells intended for biomedical applications (e.g., cell therapies, bone and dental grafts, cardiovascular or musculoskeletal tissue regeneration, and wound healing).
Collapse
Affiliation(s)
- María Puertas-Bartolomé
- INM—Leibniz Institute for New Materials, Campus D2 2, 66123 Saarbrücken, Germany
- Saarland University, 66123 Saarbrücken, Germany
| | - Ana Mora-Boza
- Woodruff School of Mechanical Engineering and Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, 315 Ferst Drive, 2310 IBB Building, Atlanta, GA 30332-0363, USA
- Institute of Polymer Science and Technology (ICTP-CSIC), Juan de la Cierva 3, 28006 Madrid, Spain
| | - Luis García-Fernández
- Institute of Polymer Science and Technology (ICTP-CSIC), Juan de la Cierva 3, 28006 Madrid, Spain
- Networking Biomedical Research Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Monforte de Lemos 3-5, Pabellón 11, 28029 Madrid, Spain
| |
Collapse
|