1
|
Ikram M, Mahmud MAP, Kalyar AA, Alomayri T, Almahri A, Hussain D. 3D-bioprinting of MXenes: Developments, medical applications, challenges, and future roadmap. Colloids Surf B Biointerfaces 2025; 251:114568. [PMID: 40020571 DOI: 10.1016/j.colsurfb.2025.114568] [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: 11/18/2024] [Revised: 02/03/2025] [Accepted: 02/15/2025] [Indexed: 03/03/2025]
Abstract
MXenes is a member of 2D transition metals carbides and nitrides with promising application prospects in energy storage, sensing, nanomedicine, tissue engineering, catalysis, and electronics. In the current era, MXenes have been widely applied in biomedical applications due to their unique rheological and electrochemical attributes. They have a larger surface area with more active sites, higher conductivity, lower cytotoxicity, and greater biocompatibility, making them highly suitable candidates for in-vivo biomedical applications. Due to recent advancemnets in MXenes 3D bioprinting, they are widely applied in regenerative medicine to combat challenges in suitable transplantation of tissues and organs. However, 3D bioprinting of MXenes has several complexities based on cell type, cytotoxicity, cell viability, and differentiation. To address these intricacies, surface modifications of MXene materials are done, which makes them highly fascinating for the 3D printing of tissues and organs. In the current review, we summarized recent progress in 3D bioprinting of MXene materials to construct scaffolds with desired rheological and biological properties, focusing on their potential applications in cancer phototherapy, tissue engineering, bone regeneration, and biosensing. We also discussed parameters affecting their biomedical applications and possible solutions by applying surface modifications. In addition, we addressed current challenges and future roadmaps for 3D bioprinting of MXene materials, such as generating high throughput 3D printed tissue constructs, drug delivery, drug discovery, and toxicology.
Collapse
Affiliation(s)
- Muhammad Ikram
- Department of Agricultural and Biosystems Engineering, South Dakota State University, Brookings, SD 57007, United States of America.
| | - M A Parvez Mahmud
- School of Mathematical and Physical Sciences, University of Technology Sydney, Ultimo, NSW 2007, Australia
| | - Amina Akbar Kalyar
- Department of Zoology, Wildlife and Fisheries, University of Agriculture Faisalabad, Pakistan
| | - Thamer Alomayri
- Department of Physics, Faculty of Science, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Albandary Almahri
- Department of Chemistry, College of Science and Humanities, Prince Sattam Bin Abdulaziz University, Al-Kharj 11942, Saudi Arabia
| | - Dilshad Hussain
- HEJ Research Institute of Chemistry, International Center for Chemical and Biological Sciences, University of Karachi, Karachi 75270, Pakistan.
| |
Collapse
|
2
|
Vörösházi J, Mackei M, Sebők C, Tráj P, Márton RA, Neogrády Z, Mátis G. Protective effects of baicalin against deoxynivalenol-induced oxidative and inflammatory damage in chicken-derived hepatic 3D cell cultures. Sci Rep 2025; 15:11180. [PMID: 40169826 PMCID: PMC11962109 DOI: 10.1038/s41598-025-95868-0] [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: 09/10/2024] [Accepted: 03/24/2025] [Indexed: 04/03/2025] Open
Abstract
Deoxynivalenol (DON) is a trichothecene mycotoxin often contaminating grains used in poultry feed production and causing several adverse effects in farm animals. Therefore, it is important to investigate compounds that can be potential candidates to mitigate these effects, such as baicalin. The effects of DON and baicalin were investigated in chicken-derived 3D hepatic cell cultures, and cell viability, LDH activity, oxidative parameters (NRF-2, 8-OHdG) and inflammatory parameters (IL-6, IL-8, IFN-γ) were monitored for 24 and 48 h. Our results suggest that DON reduced cellular metabolic activity but did not prove to be cytotoxic, and baicalin was able to attenuate this adverse effect. The change in extracellular LDH activity suggests that after 48 h the cells have already started to respond to the adverse effects of the toxin and protective mechanisms were induced. Based on the measured oxidative parameters, baicalin showed antioxidant activity, but after longer exposure, our results indicate a prooxidant effect. Baicalin also had an anti-inflammatory effect based on the amount of IL-6 and IL-8, while DON exerted a dose-and time-dependent pleiotropic activity. These results suggest that DON may have an impact on cellular inflammation and oxidative homeostasis, and that baicalin could be able to alleviate these adverse effects.
Collapse
Affiliation(s)
- Júlia Vörösházi
- Division of Biochemistry, Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, 1078, Hungary.
| | - Máté Mackei
- Division of Biochemistry, Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, 1078, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, Budapest, 1078, Hungary
| | - Csilla Sebők
- Division of Biochemistry, Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, 1078, Hungary
| | - Patrik Tráj
- Division of Biochemistry, Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, 1078, Hungary
| | - Rege Anna Márton
- Division of Biochemistry, Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, 1078, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, Budapest, 1078, Hungary
| | - Zsuzsanna Neogrády
- Division of Biochemistry, Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, 1078, Hungary
| | - Gábor Mátis
- Division of Biochemistry, Department of Physiology and Biochemistry, University of Veterinary Medicine, Budapest, 1078, Hungary
- National Laboratory of Infectious Animal Diseases, Antimicrobial Resistance, Veterinary Public Health and Food Chain Safety, University of Veterinary Medicine, Budapest, 1078, Hungary
| |
Collapse
|
3
|
Abdal Dayem A, Bin Jang S, Lim N, Yeo HC, Kwak Y, Lee SH, Shin HJ, Cho SG. Advances in lacrimal gland organoid development: Techniques and therapeutic applications. Biomed Pharmacother 2025; 183:117870. [PMID: 39870025 DOI: 10.1016/j.biopha.2025.117870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 01/11/2025] [Accepted: 01/23/2025] [Indexed: 01/29/2025] Open
Abstract
The human lacrimal gland (LG), located above the outer orbital region within the frontal bone socket, is essential in maintaining eye surface health and lubrication. It is firmly anchored to the orbital periosteum by the connective tissue, and it is vital for protecting and lubricating the eye by secreting lacrimal fluid. Disruption in the production, composition, or secretion of lacrimal fluid can lead to dry eye syndrome, a condition characterized by ocular discomfort and potential eye surface damage. This review explores the recent advancements in LG organoid generation using tissues and stem cells, highlighting cutting-edge techniques in biomaterial-based and scaffold-free technologies. Additionally, we shed light on the complex pathophysiology of LG dysfunction, providing insights into the LG physiological roles while identifying strategies for generating LG organoids and exploring their potential clinical applications. Alterations in LG morphology or secretory function can affect the tear film stability and quality, leading to various ocular pathological conditions. This comprehensive review underlines the critical crosslink of LG organoid development with disease modeling and drug screening, underscoring their potential for advancing therapeutic applications.
Collapse
Affiliation(s)
- Ahmed Abdal Dayem
- Department of Stem Cell and Regenerative Biotechnology, School of Advanced Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, and Institute of Health, Aging & Society, Konkuk University, 120 Neungdong-ro Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Soo Bin Jang
- Department of Stem Cell and Regenerative Biotechnology, School of Advanced Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, and Institute of Health, Aging & Society, Konkuk University, 120 Neungdong-ro Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Nahee Lim
- Department of Stem Cell and Regenerative Biotechnology, School of Advanced Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, and Institute of Health, Aging & Society, Konkuk University, 120 Neungdong-ro Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Han Cheol Yeo
- Department of Stem Cell and Regenerative Biotechnology, School of Advanced Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, and Institute of Health, Aging & Society, Konkuk University, 120 Neungdong-ro Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Yeonjoo Kwak
- Department of Stem Cell and Regenerative Biotechnology, School of Advanced Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, and Institute of Health, Aging & Society, Konkuk University, 120 Neungdong-ro Gwangjin-gu, Seoul 05029, Republic of Korea
| | - Shin-Hyo Lee
- Department of Anatomy, Wonkwang University School of Medicine, Iksan, Republic of Korea; Jesaeng-Euise Clinical Anatomy Center, Wonkwang University School of Medicine, Iksan, Republic of Korea
| | - Hyun Jin Shin
- Konkuk University School of Medicine, Chungju city, Republic of Korea; Department of Ophthalmology, Konkuk University Medical Center, Seoul, Republic of Korea; Research Institute of Medical Science, Konkuk University School of Medicine, Seoul, Republic of Korea; Institute of Biomedical Science & Technology, Konkuk University, Seoul, Republic of Korea.
| | - Sang-Goo Cho
- Department of Stem Cell and Regenerative Biotechnology, School of Advanced Biotechnology, Molecular & Cellular Reprogramming Center, Institute of Advanced Regenerative Science, and Institute of Health, Aging & Society, Konkuk University, 120 Neungdong-ro Gwangjin-gu, Seoul 05029, Republic of Korea; R&D Team, StemExOne Co., Ltd., Seoul, Republic of Korea.
| |
Collapse
|
4
|
Tüçer ME, Tunç N, Tüçer S, Acar R, Usta DD, Salimi K, Konu Ö, Şeker UÖŞ. Transcriptomic investigation of NP toxicity on HepaRG spheroids. Chem Biol Interact 2025; 405:111303. [PMID: 39515631 DOI: 10.1016/j.cbi.2024.111303] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Revised: 10/11/2024] [Accepted: 10/30/2024] [Indexed: 11/16/2024]
Abstract
Metal nanoparticles (NPs) are commonly used nanomaterials, however concerns have been raised about their toxicity. Although a few studies have reported the toxicity of NPs on cells, they have generally been restricted to a limited variety of NPs, inappropriate cell lines, or culture methods. Thus, the adverse effects remain inadequately understood, necessitating further analysis. This study focuses on assessing the impacts of diverse NPs of varying materials and sizes on HepaRG spheroids to determine the genes that respond to acute NP toxicity. HepaRG cells, the most appropriate alternative to primary hepatocytes, were cultured in 3D spheroids to better mimic the cellular microenvironment of the liver. To elucidate the toxicity mechanisms of NPs on HepaRG spheroids, transcriptome analysis was conducted by using RNA sequencing (RNA-seq). Among all NPs, lowest and highest numbers of differentially expressed genes (DEGs) were found for 40 nm AuNP (118 genes) and InP/ZnS (1904 genes), respectively. Remarkably, processes such as drug metabolism, sensitivity to metal ions, oxidative stress, endothelial-mesenchymal transition (EMT) and apoptosis consistently exhibited significant enrichment across all NPs of different materials. Pathways related to stress responses of the cells such as the MAPK, p53 and mTOR pathways are found to be dysregulated upon exposure to various NPs. The genes that are common and unique between DEGs of different NPs were identified. These results provide novel insights into the toxicological mechanisms of NPs on HepaRG spheroids.
Collapse
Affiliation(s)
- Merve Erden Tüçer
- UNAM-Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center, Bilkent University, 06800, Ankara, Turkey
| | - Nazlıcan Tunç
- UNAM-Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center, Bilkent University, 06800, Ankara, Turkey
| | - Suat Tüçer
- UNAM-Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center, Bilkent University, 06800, Ankara, Turkey
| | - Rana Acar
- Bilkent University, Department of Molecular Biology and Genetics, 06800, Ankara, Turkey
| | - Duygu Deniz Usta
- Gazi University, Faculty of Medicine, Department of Medical Biology and Genetics, 06500, Ankara, Turkey
| | - Kouroush Salimi
- Ankara Yildirim Beyazit University, Faculty of Engineering and Natural Sciences, Department of Chemical Engineering, 06010, Ankara, Turkey
| | - Özlen Konu
- UNAM-Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center, Bilkent University, 06800, Ankara, Turkey; Bilkent University, Department of Molecular Biology and Genetics, 06800, Ankara, Turkey
| | - Urartu Özgür Şafak Şeker
- UNAM-Institute of Materials Science and Nanotechnology, National Nanotechnology Research Center, Bilkent University, 06800, Ankara, Turkey.
| |
Collapse
|
5
|
McDonough E, Barroso M, Ginty F, Corr DT. Modeling intratumor heterogeneity in breast cancer. Biofabrication 2024; 17:10.1088/1758-5090/ad9b50. [PMID: 39642392 PMCID: PMC11740194 DOI: 10.1088/1758-5090/ad9b50] [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: 04/25/2024] [Accepted: 12/06/2024] [Indexed: 12/08/2024]
Abstract
Reduced therapy response in breast cancer has been correlated with heterogeneity in biomarker composition, expression level, and spatial distribution of cancer cells within a patient tumor. Thus, there is a need for models to replicate cell-cell, cell-stromal, and cell-microenvironment interactions during cancer progression. Traditional two-dimensional (2D) cell culture models are convenient but cannot adequately represent tumor microenvironment histological organization,in vivo3D spatial/cellular context, and physiological relevance. Recently, three-dimensional (3D)in vitrotumor models have been shown to provide an improved platform for incorporating compositional and spatial heterogeneity and to better mimic the biological characteristics of patient tumors to assess drug response. Advances in 3D bioprinting have allowed the creation of more complex models with improved physiologic representation while controlling for reproducibility and accuracy. This review aims to summarize the advantages and challenges of current 3Din vitromodels for evaluating therapy response in breast cancer, with a particular emphasis on 3D bioprinting, and addresses several key issues for future model development as well as their application to other cancers.
Collapse
Affiliation(s)
- Elizabeth McDonough
- Department of Biomedical Engineering, Rensselaer
Polytechnic Institute, 110 Eighth Street, Troy, New York 12180, United States
- GE HealthCare Technology & Innovation Center, 1
Research Circle, Niskayuna, New York 12309, United States
| | - Margarida Barroso
- Department of Molecular and Cellular Physiology, Albany
Medical College, Albany, NY 12208, United States
| | - Fiona Ginty
- GE HealthCare Technology & Innovation Center, 1
Research Circle, Niskayuna, New York 12309, United States
| | - David T. Corr
- Department of Biomedical Engineering, Rensselaer
Polytechnic Institute, 110 Eighth Street, Troy, New York 12180, United States
| |
Collapse
|
6
|
Wang X, Zhang D, Singh YP, Yeo M, Deng G, Lai J, Chen F, Ozbolat IT, Yu Y. Progress in Organ Bioprinting for Regenerative Medicine. ENGINEERING 2024; 42:121-142. [DOI: 10.1016/j.eng.2024.04.023] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/05/2025]
|
7
|
Okamoto N, Taniura N, Nakayama T, Tanaka E, Kageyama Y, Noujima M, Kushima R, Mukaisho KI. Three-Dimensional Culture of Glioblastoma Cells Using a Tissueoid Cell Culture System. Acta Histochem Cytochem 2024; 57:149-155. [PMID: 39552931 PMCID: PMC11565224 DOI: 10.1267/ahc.24-00043] [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: 08/06/2024] [Accepted: 08/12/2024] [Indexed: 11/19/2024] Open
Abstract
In classical cell culture techniques, cancer cells typically proliferate in a single layer by adhering to the undersurface of laboratory vessels. Consequently, concerns have been raised regarding the fidelity of the morphological and functional characteristics of these cultured cancer cells compared to those of their in vivo counterparts. Our previous studies have investigated various epithelial malignant tumors utilizing the Tissueoid cell culture system, a three-dimensional (3D) cultivation method employing Cellbed-a nonwoven sheet composed of high-purity silica fibers as a scaffold. In this investigation, we have achieved successful 3D culturing of glioblastoma cells (A172 and T98G), which are non-epithelial in nature. As such our focus is to juxtapose their morphological features against that of those cultivated via conventional two-dimensional (2D) methods. Our findings will be elucidated using immunostaining, immunofluorescence staining, and scanning electron microscopy, substantiated with accompanying imaging. Notably, cells cultured in the 3D environment exhibited distinct morphological attributes compared to those of their 2D counterparts, notably featuring pronounced cellular protrusions. We envisage the continued utilization of the 3D culture platform to facilitate diverse avenues of research, encompassing the exploration of novel therapeutic modalities for glioblastoma cells and beyond.
Collapse
Affiliation(s)
- Natsume Okamoto
- Division of Human Pathology, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Naoko Taniura
- Division of Human Pathology, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
- Education Center for Medicine and Nursing, Shiga University of Medical Science, Otsu, Japan
| | - Takahisa Nakayama
- Division of Human Pathology, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Eri Tanaka
- Division of Human Pathology, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Yusuke Kageyama
- Division of Human Pathology, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Mai Noujima
- Division of Human Pathology, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Ryoji Kushima
- Division of Human Pathology, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Ken-ichi Mukaisho
- Education Center for Medicine and Nursing, Shiga University of Medical Science, Otsu, Japan
| |
Collapse
|
8
|
Wang G, Mao X, Wang W, Wang X, Li S, Wang Z. Bioprinted research models of urological malignancy. EXPLORATION (BEIJING, CHINA) 2024; 4:20230126. [PMID: 39175884 PMCID: PMC11335473 DOI: 10.1002/exp.20230126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Accepted: 01/08/2024] [Indexed: 08/24/2024]
Abstract
Urological malignancy (UM) is among the leading threats to health care worldwide. Recent years have seen much investment in fundamental UM research, including mechanistic investigation, early diagnosis, immunotherapy, and nanomedicine. However, the results are not fully satisfactory. Bioprinted research models (BRMs) with programmed spatial structures and functions can serve as powerful research tools and are likely to disrupt traditional UM research paradigms. Herein, a comprehensive review of BRMs of UM is presented. It begins with a brief introduction and comparison of existing UM research models, emphasizing the advantages of BRMs, such as modeling real tissues and organs. Six kinds of mainstream bioprinting techniques used to fabricate such BRMs are summarized with examples. Thereafter, research advances in the applications of UM BRMs, such as culturing tumor spheroids and organoids, modeling cancer metastasis, mimicking the tumor microenvironment, constructing organ chips for drug screening, and isolating circulating tumor cells, are comprehensively discussed. At the end of this review, current challenges and future development directions of BRMs and UM are highlighted from the perspective of interdisciplinary science.
Collapse
Affiliation(s)
- Guanyi Wang
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
- Department of Biomedical Engineering and Hubei Province Key Laboratory of Allergy and Immune Related DiseaseTaiKang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhanChina
| | - Xiongmin Mao
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Wang Wang
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Xiaolong Wang
- Lewis Katz School of MedicineTemple UniversityPhiladelphiaPennsylvaniaUSA
| | - Sheng Li
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
| | - Zijian Wang
- Department of UrologyCancer Precision Diagnosis and Treatment and Translational Medicine Hubei Engineering Research CenterZhongnan Hospital of Wuhan UniversityWuhanChina
- Department of Biomedical Engineering and Hubei Province Key Laboratory of Allergy and Immune Related DiseaseTaiKang Medical School (School of Basic Medical Sciences)Wuhan UniversityWuhanChina
| |
Collapse
|
9
|
Ogire E, Perrin-Cocon L, Figl M, Kundlacz C, Jacquemin C, Hubert S, Aublin-Gex A, Toesca J, Ramière C, Vidalain PO, Mathieu C, Lotteau V, Diaz O. Dengue Virus dependence on glucokinase activity and glycolysis Confers Sensitivity to NAD(H) biosynthesis inhibitors. Antiviral Res 2024; 228:105939. [PMID: 38909960 DOI: 10.1016/j.antiviral.2024.105939] [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: 02/01/2024] [Revised: 05/20/2024] [Accepted: 06/17/2024] [Indexed: 06/25/2024]
Abstract
Viruses have developed sophisticated strategies to control metabolic activity of infected cells in order to supply replication machinery with energy and metabolites. Dengue virus (DENV), a mosquito-borne flavivirus responsible for dengue fever, is no exception. Previous reports have documented DENV interactions with metabolic pathways and shown in particular that glycolysis is increased in DENV-infected cells. However, underlying molecular mechanisms are still poorly characterized and dependence of DENV on this pathway has not been investigated in details yet. Here, we identified an interaction between the non-structural protein 3 (NS3) of DENV and glucokinase regulator protein (GCKR), a host protein that inhibits the liver-specific hexokinase GCK. NS3 expression was found to increase glucose consumption and lactate secretion in hepatic cell line expressing GCK. Interestingly, we observed that GCKR interaction with GCK decreases DENV replication, indicating the dependence of DENV to GCK activity and supporting the role of NS3 as an inhibitor of GCKR function. Accordingly, in the same cells, DENV replication both induces and depends on glycolysis. By targeting NAD(H) biosynthesis with the antimetabolite 6-Amino-Nicotinamide (6-AN), we decreased cellular glycolytic activity and inhibited DENV replication in hepatic cells. Infection of primary organotypic liver cultures (OLiC) from hamsters was also inhibited by 6-AN. Altogether, our results show that DENV has evolved strategies to control glycolysis in the liver, which could account for hepatic dysfunctions associated to infection. Besides, our findings suggest that lowering intracellular availability of NAD(H) could be a valuable therapeutic strategy to control glycolysis and inhibit DENV replication in the liver.
Collapse
Affiliation(s)
- Eva Ogire
- CIRI, Centre International de Recherche en Infectiologie, NeuroInvasion TROpism and VIRal Encephalitis Team, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 21 Avenue Tony Garnier, F-69007, Lyon, France
| | - Laure Perrin-Cocon
- CIRI, Centre International de Recherche en Infectiologie, VIRal Infection Metabolism and Immunity Team, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 21 Avenue Tony Garnier, F-69007, Lyon, France
| | - Marianne Figl
- CIRI, Centre International de Recherche en Infectiologie, VIRal Infection Metabolism and Immunity Team, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 21 Avenue Tony Garnier, F-69007, Lyon, France
| | - Cindy Kundlacz
- CIRI, Centre International de Recherche en Infectiologie, VIRal Infection Metabolism and Immunity Team, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 21 Avenue Tony Garnier, F-69007, Lyon, France
| | - Clémence Jacquemin
- CIRI, Centre International de Recherche en Infectiologie, VIRal Infection Metabolism and Immunity Team, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 21 Avenue Tony Garnier, F-69007, Lyon, France
| | - Sophie Hubert
- CIRI, Centre International de Recherche en Infectiologie, VIRal Infection Metabolism and Immunity Team, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 21 Avenue Tony Garnier, F-69007, Lyon, France
| | - Anne Aublin-Gex
- CIRI, Centre International de Recherche en Infectiologie, VIRal Infection Metabolism and Immunity Team, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 21 Avenue Tony Garnier, F-69007, Lyon, France
| | - Johan Toesca
- CIRI, Centre International de Recherche en Infectiologie, VIRal Infection Metabolism and Immunity Team, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 21 Avenue Tony Garnier, F-69007, Lyon, France
| | - Christophe Ramière
- CIRI, Centre International de Recherche en Infectiologie, VIRal Infection Metabolism and Immunity Team, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 21 Avenue Tony Garnier, F-69007, Lyon, France; Laboratoire de Virologie, Hôpital de la Croix-Rousse, Hospices Civils de Lyon, Lyon, France
| | - Pierre-Olivier Vidalain
- CIRI, Centre International de Recherche en Infectiologie, VIRal Infection Metabolism and Immunity Team, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 21 Avenue Tony Garnier, F-69007, Lyon, France
| | - Cyrille Mathieu
- CIRI, Centre International de Recherche en Infectiologie, NeuroInvasion TROpism and VIRal Encephalitis Team, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 21 Avenue Tony Garnier, F-69007, Lyon, France
| | - Vincent Lotteau
- CIRI, Centre International de Recherche en Infectiologie, VIRal Infection Metabolism and Immunity Team, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 21 Avenue Tony Garnier, F-69007, Lyon, France; Laboratoire P4-Jean Mérieux, INSERM, Lyon, France
| | - Olivier Diaz
- CIRI, Centre International de Recherche en Infectiologie, VIRal Infection Metabolism and Immunity Team, Univ Lyon, Inserm, U1111, Université Claude Bernard Lyon 1, CNRS, UMR5308, ENS de Lyon, 21 Avenue Tony Garnier, F-69007, Lyon, France.
| |
Collapse
|
10
|
Aye KTN, Ferreira JN, Chaweewannakorn C, Souza GR. Advances in the application of iron oxide nanoparticles (IONs and SPIONs) in three-dimensional cell culture systems. SLAS Technol 2024; 29:100132. [PMID: 38582355 DOI: 10.1016/j.slast.2024.100132] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2023] [Revised: 03/22/2024] [Accepted: 04/04/2024] [Indexed: 04/08/2024]
Abstract
BACKGROUND The field of tissue engineering has remarkably progressed through the integration of nanotechnology and the widespread use of magnetic nanoparticles. These nanoparticles have resulted in innovative methods for three-dimensional (3D) cell culture platforms, including the generation of spheroids, organoids, and tissue-mimetic cultures, where they play a pivotal role. Notably, iron oxide nanoparticles and superparamagnetic iron oxide nanoparticles have emerged as indispensable tools for non-contact manipulation of cells within these 3D environments. The variety and modification of the physical and chemical properties of magnetic nanoparticles have profound impacts on cellular mechanisms, metabolic processes, and overall biological function. This review article focuses on the applications of magnetic nanoparticles, elucidating their advantages and potential pitfalls when integrated into 3D cell culture systems. This review aims to shed light on the transformative potential of magnetic nanoparticles in terms of tissue engineering and their capacity to improve the cultivation and manipulation of cells in 3D environments.
Collapse
Affiliation(s)
- Khin The Nu Aye
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Department of Research Affairs, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Joao N Ferreira
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Department of Research Affairs, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Chayanit Chaweewannakorn
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Department of Research Affairs, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand; Department of Occlusion, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.
| | - Glauco R Souza
- Greiner Bio-One North America, Inc., 4238 Capital Drive, Monroe, NC 28110, USA
| |
Collapse
|
11
|
de Araújo TBS, Nogueira RLR, Siquara da Rocha LDO, Bastos IN, Dias RB, Souza BSDF, Lambert DW, Coletta RD, Silva VAO, Gurgel Rocha CA. Enhancing scaffold-free spheroid models: 3D cell bioprinting method for metastatic HSC3-Oral squamous carcinoma cell line. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2024; 29:100158. [PMID: 38852983 DOI: 10.1016/j.slasd.2024.100158] [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: 02/11/2024] [Revised: 04/22/2024] [Accepted: 04/29/2024] [Indexed: 06/11/2024]
Abstract
3D in vitro systems offer advantages over the shortcomings of two-dimensional models by simulating the morphological and functional features of in vivo-like environments, such as cell-cell and cell-extracellular matrix interactions, as well as the co-culture of different cell types. Nevertheless, these systems present technical challenges that limit their potential in cancer research requiring cell line- and culture-dependent standardization. This protocol details the use of a magnetic 3D bioprinting method and other associated techniques (cytotoxicity assay and histological analysis) using oral squamous cell carcinoma cell line, HSC3, which offer advantages compared to existing widely used approaches. This protocol is particularly timely, as it validates magnetic bioprinting as a method for the rapid deployment of 3D cultures as a tool for compound screening and development of heterotypic cultures such as co-culture of oral squamous cell carcinoma cells with cancer-associated fibroblasts (HSC3/CAFs).
Collapse
Affiliation(s)
- Taís Bacelar Sacramento de Araújo
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil; Department of Propaedeutics, School of Dentistry of the Federal University of Bahia, Salvador 40110-150, Bahia, Brazil
| | - Raphael Luís Rocha Nogueira
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil; Department of Pathology, School of Medicine of the Federal University of Bahia, Salvador 40110-909, Bahia, Brazil
| | - Leonardo de Oliveira Siquara da Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil; Department of Pathology, School of Medicine of the Federal University of Bahia, Salvador 40110-909, Bahia, Brazil
| | - Iasmin Nogueira Bastos
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil; Department of Propaedeutics, School of Dentistry of the Federal University of Bahia, Salvador 40110-150, Bahia, Brazil
| | - Rosane Borges Dias
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil; Department of Propaedeutics, School of Dentistry of the Federal University of Bahia, Salvador 40110-150, Bahia, Brazil
| | - Bruno Solano De Freitas Souza
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil; Department of Pathology, School of Medicine of the Federal University of Bahia, Salvador 40110-909, Bahia, Brazil; Center for Biotechnology and Cell Therapy, D'Or Institute for Research and Education (IDOR), São Rafael Hospital, Salvador 41253-190, Brazil
| | | | - Ricardo D Coletta
- Department of Oral Diagnosis, School of Dentistry, University of Campinas, Piracicaba 13414-903, São Paulo, Brazil; Graduate Program in Oral Biology, School of Dentistry University of Campinas, Piracicaba 13414-903, São Paulo, Brazil
| | - Viviane Aline Oliveira Silva
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil; Department of Pathology, School of Medicine of the Federal University of Bahia, Salvador 40110-909, Bahia, Brazil; Center for Biotechnology and Cell Therapy, D'Or Institute for Research and Education (IDOR), São Rafael Hospital, Salvador 41253-190, Brazil; Molecular Oncology Research Center, Barretos Cancer Hospital, Barretos 14784-400, São Paulo, Brazil.
| | - Clarissa A Gurgel Rocha
- Gonçalo Moniz Institute, Oswaldo Cruz Foundation (IGM-FIOCRUZ/BA), Salvador 40296-710, Bahia, Brazil; Department of Propaedeutics, School of Dentistry of the Federal University of Bahia, Salvador 40110-150, Bahia, Brazil; Department of Pathology, School of Medicine of the Federal University of Bahia, Salvador 40110-909, Bahia, Brazil; Center for Biotechnology and Cell Therapy, D'Or Institute for Research and Education (IDOR), São Rafael Hospital, Salvador 41253-190, Brazil.
| |
Collapse
|
12
|
Klangprapan J, Souza GR, Ferreira JN. Bioprinting salivary gland models and their regenerative applications. BDJ Open 2024; 10:39. [PMID: 38816372 PMCID: PMC11139920 DOI: 10.1038/s41405-024-00219-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 04/29/2024] [Accepted: 04/30/2024] [Indexed: 06/01/2024] Open
Abstract
OBJECTIVE Salivary gland (SG) hypofunction is a common clinical condition arising from radiotherapy to suppress head and neck cancers. The radiation often destroys the SG secretory acini, and glands are left with limited regenerative potential. Due to the complex architecture of SG acini and ducts, three-dimensional (3D) bioprinting platforms have emerged to spatially define these in vitro epithelial units and develop mini-organs or organoids for regeneration. Due to the limited body of evidence, this comprehensive review highlights the advantages and challenges of bioprinting platforms for SG regeneration. METHODS SG microtissue engineering strategies such as magnetic 3D bioassembly of cells and microfluidic coaxial 3D bioprinting of cell-laden microfibers and microtubes have been proposed to replace the damaged acinar units, avoid the use of xenogeneic matrices (like Matrigel), and restore salivary flow. RESULTS Replacing the SG damaged organ is challenging due to its complex architecture, which combines a ductal network with acinar epithelial units to facilitate a unidirectional flow of saliva. Our research group was the first to develop 3D bioassembly SG epithelial functional organoids with innervation to respond to both cholinergic and adrenergic stimulation. More recently, microtissue engineering using coaxial 3D bioprinting of hydrogel microfibers and microtubes could also supported the formation of viable epithelial units. Both bioprinting approaches could overcome the need for Matrigel by facilitating the assembly of adult stem cells, such as human dental pulp stem cells, and primary SG cells into micro-sized 3D constructs able to produce their own matrix and self-organize into micro-modular tissue clusters with lumenized areas. Furthermore, extracellular vesicle (EV) therapies from organoid-derived secretome were also designed and validated ex vivo for SG regeneration after radiation damage. CONCLUSION Magnetic 3D bioassembly and microfluidic coaxial bioprinting platforms have the potential to create SG mini-organs for regenerative applications via organoid transplantation or organoid-derived EV therapies.
Collapse
Affiliation(s)
- Jutapak Klangprapan
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Faculty of Dentistry, Chulalongkorn University, 34 Henri-Dunant Road, Pathumwan, Bangkok, 10330, Thailand
| | - Glauco R Souza
- Greiner Bio-one North America Inc., 4238 Capital Drive, Monroe, NC, 28110, USA
| | - João N Ferreira
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Faculty of Dentistry, Chulalongkorn University, 34 Henri-Dunant Road, Pathumwan, Bangkok, 10330, Thailand.
| |
Collapse
|
13
|
Huang G, Zhao Y, Chen D, Wei L, Hu Z, Li J, Zhou X, Yang B, Chen Z. Applications, advancements, and challenges of 3D bioprinting in organ transplantation. Biomater Sci 2024; 12:1425-1448. [PMID: 38374788 DOI: 10.1039/d3bm01934a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/21/2024]
Abstract
To date, organ transplantation remains an effective method for treating end-stage diseases of various organs. In recent years, despite the continuous development of organ transplantation technology, a variety of problems restricting its progress have emerged one after another, and the shortage of donors is at the top of the list. Bioprinting is a very useful tool that has huge application potential in many fields of life science and biotechnology, among which its use in medicine occupies a large area. With the development of bioprinting, advances in medicine have focused on printing cells and tissues for tissue regeneration and reconstruction of viable human organs, such as the heart, kidneys, and bones. In recent years, with the development of organ transplantation, three-dimensional (3D) bioprinting has played an increasingly important role in this field, giving rise to many unsolved problems, including a shortage of organ donors. This review respectively introduces the development of 3D bioprinting as well as its working principles and main applications in the medical field, especially in the applications, and advancements and challenges of 3D bioprinting in organ transplantation. With the continuous update and progress of printing technology and its deeper integration with the medical field, many obstacles will have new solutions, including tissue repair and regeneration, organ reconstruction, etc., especially in the field of organ transplantation. 3D printing technology will provide a better solution to the problem of donor shortage.
Collapse
Affiliation(s)
- Guobin Huang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, No. 1095 Jiefang Avenue, Wuhan 430030, China.
| | - Yuanyuan Zhao
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, No. 1095 Jiefang Avenue, Wuhan 430030, China.
| | - Dong Chen
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, No. 1095 Jiefang Avenue, Wuhan 430030, China.
| | - Lai Wei
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, No. 1095 Jiefang Avenue, Wuhan 430030, China.
| | - Zhiping Hu
- Department of Pathology, School of Medicine, University of Pittsburgh, Pittsburgh, USA
| | - Junbo Li
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, No. 1095 Jiefang Avenue, Wuhan 430030, China.
| | - Xi Zhou
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, No. 1095 Jiefang Avenue, Wuhan 430030, China.
| | - Bo Yang
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, No. 1095 Jiefang Avenue, Wuhan 430030, China.
| | - Zhishui Chen
- Institute of Organ Transplantation, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology; Key Laboratory of Organ Transplantation, Ministry of Education; NHC Key Laboratory of Organ Transplantation; Key Laboratory of Organ Transplantation, Chinese Academy of Medical Sciences, No. 1095 Jiefang Avenue, Wuhan 430030, China.
| |
Collapse
|
14
|
Srisongkram T, Syahid NF, Piyasawetkul T, Thirawatthanasak P, Khamtang P, Sawasnopparat N, Tookkane D, Weerapreeyakul N, Puthongking P. Prediction of Spheroid Cell Death Using Fluorescence Staining and Convolutional Neural Networks. Chem Res Toxicol 2023; 36:1980-1989. [PMID: 38052002 DOI: 10.1021/acs.chemrestox.3c00257] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/07/2023]
Abstract
Three-dimensional (3D) cell culture is emerging for drug design and drug screening. Skin toxicity is one of the most important assays for determining the toxicity of a compound before being used in skin application. Much work has been done to find an alternative assay without animal experiments. 3D cell culture is one of the methods that provides clinically relevant models with superior clinical translation compared to that of 2D cell culture. In this study, we developed a spheroid toxicity assay using keratinocyte HaCaT cells with propidium iodide and calcein AM. We also applied the transfer learning-containing convolutional neural network (CNN) to further determine spheroid cell death with fluorescence labeling. Our result shows that the morphologies of the spheroid are the key features in determining the apoptosis cell death of the HaCaT spheroid. Our CNN model provided good statistical measurement in terms of accuracy, precision, and recall in both validation and external test data sets. One can predict keratinocyte spheroid cell death if that spheroid image contains the fluorescence signals from propidium iodide and calcein AM. The CNN model can be accessed in the web application at https://qsarlabs.com/#spheroiddeath.
Collapse
Affiliation(s)
- Tarapong Srisongkram
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Nur Fadhilah Syahid
- Graduate School in the Program of Pharmaceutical Chemistry and Natural Products, Pharmaceutical Sciences, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Thanawat Piyasawetkul
- Doctor of Pharmacy Program, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Pannaphat Thirawatthanasak
- Doctor of Pharmacy Program, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Patcharapa Khamtang
- Doctor of Pharmacy Program, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Nathida Sawasnopparat
- Doctor of Pharmacy Program, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Dheerapat Tookkane
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Natthida Weerapreeyakul
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
| | - Ploenthip Puthongking
- Division of Pharmaceutical Chemistry, Faculty of Pharmaceutical Sciences, Khon Kaen University, Khon Kaen 40002, Thailand
| |
Collapse
|
15
|
Zakaria NH, Saad N, Che Abdullah CA, Mohd Esa N. The Antiproliferative Effect of Chloroform Fraction of Eleutherine bulbosa (Mill.) Urb. on 2D- and 3D-Human Lung Cancer Cells (A549) Model. Pharmaceuticals (Basel) 2023; 16:936. [PMID: 37513848 PMCID: PMC10384492 DOI: 10.3390/ph16070936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Revised: 06/21/2023] [Accepted: 06/26/2023] [Indexed: 07/30/2023] Open
Abstract
Since lung cancer is the leading cause of cancer-related death worldwide, research is being conducted to discover anticancer agents as its treatment. Eleutherine bulbosa, a Dayak folklore medicine, exhibited anticancer effects against several cancer cells; however, its anticancer potency against lung cancer cells has not been explored yet. This study aims to determine the anticancer potency of E. bulbosa bulbs against lung cancer cells (A549) using 2D and 3D culture models, as well as determine its active compounds using gas chromatography-mass spectrometry (GC-MS) analysis. Three fractions of E. bulbosa bulbs, namely chloroform, n-hexane, and ethyl acetate, were tested for cytotoxicity using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium-bromide (MTT) and CellTiter-Glo. The antiproliferative effects of the most cytotoxic fraction against the 2D culture model were determined by a clonogenic survival assay and propidium iodide/Hoechst 33342 double staining, whereas the effects against the 3D culture model were determined by microscopy, flow cytometry, and gene expression analysis. The chloroform fraction is the most cytotoxic against A549 cells than other fractions, and it inhibited colony formation and induced apoptosis of A549 cells. The chloroform fraction also inhibited the growth of the A549 spheroid by suppressing the spheroid size, inducing apoptosis, reducing the proportion of CD44 lung cancer stem cells, causing arrest at the S phase of the cell cycle, and suppressing the expression of the SOX2 and MYC genes. Furthermore, the GC-MS analysis detected 20 active compounds in the chloroform fraction, including the major compounds of eleutherine and isoeleutherine. In conclusion, the chloroform fraction of E. bulbosa bulbs exhibit its antiproliferative effect on 2D and 3D culture models of A549 cells, suggesting it could be a lung cancer chemopreventive agent.
Collapse
Affiliation(s)
- Nur Hannan Zakaria
- UPM-MAKNA Cancer Research Laboratory (CANRES), Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
- Natural Medicine and Product Research Laboratory (NaturMeds), Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Norazalina Saad
- UPM-MAKNA Cancer Research Laboratory (CANRES), Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Che Azurahanim Che Abdullah
- UPM-MAKNA Cancer Research Laboratory (CANRES), Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
- Department of Physics, Faculty of Science, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
- Materials Synthesis and Characterization Laboratory (MSCL), Institute of Nanoscience and Nanotechnology (ION2), Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| | - Norhaizan Mohd Esa
- Natural Medicine and Product Research Laboratory (NaturMeds), Institute of Bioscience, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
- Department of Nutrition, Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, 43400 Serdang, Selangor, Malaysia
| |
Collapse
|
16
|
Phan TV, Oo Y, Ahmed K, Rodboon T, Rosa V, Yodmuang S, Ferreira JN. Salivary gland regeneration: from salivary gland stem cells to three-dimensional bioprinting. SLAS Technol 2023; 28:199-209. [PMID: 37019217 DOI: 10.1016/j.slast.2023.03.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2022] [Revised: 03/13/2023] [Accepted: 03/28/2023] [Indexed: 04/05/2023]
Abstract
Hyposalivation and severe dry mouth syndrome are the most common complications in patients with head and neck cancer (HNC) after receiving radiation therapy. Conventional treatment for hyposalivation relies on the use of sialogogues such as pilocarpine; however, their efficacy is constrained by the limited number of remnant acinar cells after radiation. After radiotherapy, the salivary gland (SG) secretory parenchyma is largely destroyed, and due to the reduced stem cell niche, this gland has poor regenerative potential. To tackle this, researchers must be able to generate highly complex cellularized 3D constructs for clinical transplantation via technologies, including those that involve bioprinting of cells and biomaterials. A potential stem cell source with promising clinical outcomes to reserve dry mouth is adipose mesenchymal stem cells (AdMSC). MSC-like cells like human dental pulp stem cells (hDPSC) have been tested in novel magnetic bioprinting platforms using nanoparticles that can bind cell membranes by electrostatic interaction, as well as their paracrine signals arising from extracellular vesicles. Both magnetized cells and their secretome cues were found to increase epithelial and neuronal growth of in vitro and ex vivo irradiated SG models. Interestingly, these magnetic bioprinting platforms can be applied as a high-throughput drug screening system due to the consistency in structure and functions of their organoids. Recently, exogenous decellularized porcine ECM was added to this magnetic platform to stimulate an ideal environment for cell tethering, proliferation, and/or differentiation. The combination of these SG tissue biofabrication strategies will promptly allow for in vitro organoid formation and establishment of cellular senescent organoids for aging models, but challenges remain in terms of epithelial polarization and lumen formation for unidirectional fluid flow. Current magnetic bioprinting nanotechnologies can provide promising functional and aging features to in vitro craniofacial exocrine gland organoids, which can be utilized for novel drug discovery and/or clinical transplantation.
Collapse
Affiliation(s)
- Toan V Phan
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Department of Research Affairs, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand; International Graduate Program in Oral Biology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Yamin Oo
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Department of Research Affairs, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Khurshid Ahmed
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Department of Research Affairs, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand; Department of Industrial Biotechnology, Faculty of Agro-Industry, Prince of Songkla University, Songkhla, Thailand
| | - Teerapat Rodboon
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Department of Research Affairs, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand; Department of Clinical Pathology, Faculty of Medicine, Navamindradhiraj University, Bangkok, Thailand
| | - Vinicius Rosa
- Faculty of Dentistry, National University of Singapore, Singapore, Singapore; Centre for Advanced 2D Materials, National University of Singapore, Singapore, Singapore; Department of Materials Science and Engineering, College of Design and Engineering, National University of Singapore, Singapore, Singapore; ORCHIDS: Oral Care Health Innovations and Designs Singapore, National University of Singapore, Singapore, Singapore
| | - Supansa Yodmuang
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Department of Research Affairs, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand; Department of Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Joao N Ferreira
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Department of Research Affairs, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand.
| |
Collapse
|
17
|
Wang T, Desmet J, Pérez-Albaladejo E, Porte C. Development of fish liver PLHC-1 spheroids and its applicability to investigate the toxicity of plastic additives. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2023; 259:115016. [PMID: 37196525 DOI: 10.1016/j.ecoenv.2023.115016] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/20/2023] [Revised: 05/10/2023] [Accepted: 05/13/2023] [Indexed: 05/19/2023]
Abstract
Fish liver cell lines are valuable tools to understand the toxicity of chemicals in aquatic vertebrates. While conventional 2D cell cultures grown in monolayers are well established, they fail to emulate toxic gradients and cellular functions as in in-vivo conditions. To overcome these limitations, this work focuses on the development of Poeciliopsis lucida (PLHC-1) spheroids as a testing platform to evaluate the toxicity of a mixture of plastic additives. The growth of spheroids was monitored over a period of 30 days, and spheroids 2-8 days old and sized between 150 and 250 µm were considered optimal for conducting toxicity tests due to their excellent viability and metabolic activity. Eight-day-old spheroids were selected for lipidomic characterization. Compared to 2D-cells, the lipidome of spheroids was relatively enriched in highly unsaturated phosphatidylcholines (PCs), sphingosines (SPBs), sphingomyelins (SMs) and cholesterol esters (CEs). When exposed to a mixture of plastic additives, spheroids were less responsive in terms of decreased cell viability and generation of reactive oxygen species (ROS), but were more sensitive than cells growing in monolayers for lipidomic responses. The lipid profile of 3D-spheroids was similar to a liver-like phenotype and it was strongly modulated by exposure to plastic additives. The development of PLHC-1 spheroids represents an important step towards the application of more realistic in-vitro methods in aquatic toxicity studies.
Collapse
Affiliation(s)
- Tiantian Wang
- Environmental Chemistry Department, IDAEA -CSIC-, C/ Jordi Girona, 18-26, 08034 Barcelona, Spain.
| | - Judith Desmet
- Environmental Chemistry Department, IDAEA -CSIC-, C/ Jordi Girona, 18-26, 08034 Barcelona, Spain
| | | | - Cinta Porte
- Environmental Chemistry Department, IDAEA -CSIC-, C/ Jordi Girona, 18-26, 08034 Barcelona, Spain
| |
Collapse
|
18
|
Pushparaj K, Balasubramanian B, Pappuswamy M, Anand Arumugam V, Durairaj K, Liu WC, Meyyazhagan A, Park S. Out of Box Thinking to Tangible Science: A Benchmark History of 3D Bio-Printing in Regenerative Medicine and Tissues Engineering. Life (Basel) 2023; 13:954. [PMID: 37109483 PMCID: PMC10145662 DOI: 10.3390/life13040954] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/31/2023] [Accepted: 04/04/2023] [Indexed: 04/09/2023] Open
Abstract
Advancements and developments in the 3D bioprinting have been promising and have met the needs of organ transplantation. Current improvements in tissue engineering constructs have enhanced their applications in regenerative medicines and other medical fields. The synergistic effects of 3D bioprinting have brought technologies such as tissue engineering, microfluidics, integrated tissue organ printing, in vivo bioprinted tissue implants, artificial intelligence and machine learning approaches together. These have greatly impacted interventions in medical fields, such as medical implants, multi-organ-on-chip models, prosthetics, drug testing tissue constructs and much more. This technological leap has offered promising personalized solutions for patients with chronic diseases, and neurodegenerative disorders, and who have been in severe accidents. This review discussed the various standing printing methods, such as inkjet, extrusion, laser-assisted, digital light processing, and stereolithographic 3D bioprinter models, adopted for tissue constructs. Additionally, the properties of natural, synthetic, cell-laden, dECM-based, short peptides, nanocomposite and bioactive bioinks are briefly discussed. Sequels of several tissue-laden constructs such as skin, bone and cartilage, liver, kidney, smooth muscles, cardiac and neural tissues are briefly analyzed. Challenges, future perspectives and the impact of microfluidics in resolving the limitations in the field, along with 3D bioprinting, are discussed. Certainly, a technology gap still exists in the scaling up, industrialization and commercialization of this technology for the benefit of stakeholders.
Collapse
Affiliation(s)
- Karthika Pushparaj
- Department of Zoology, School of Biosciences, Avinashilingam Institute for Home Science and Higher Education for Women, Coimbatore 641 043, Tamil Nadu, India;
| | | | - Manikantan Pappuswamy
- Department of Life Science, CHRIST (Deemed to be University), Bengaluru 560 076, Karnataka, India
| | - Vijaya Anand Arumugam
- Department of Human Genetics and Molecular Biology, Bharathiar University, Coimbatore 641 046, Tamil Nadu, India
| | - Kaliannan Durairaj
- Department of Infection Biology, School of Medicine, Wonkwang University, lksan 54538, Republic of Korea
| | - Wen-Chao Liu
- Department of Animal Science, College of Coastal Agricultural Sciences, Guangdong Ocean University, Zhanjiang 524088, China
| | - Arun Meyyazhagan
- Department of Life Science, CHRIST (Deemed to be University), Bengaluru 560 076, Karnataka, India
| | - Sungkwon Park
- Department of Food Science and Biotechnology, College of Life Science, Sejong University, Seoul 05006, Republic of Korea;
| |
Collapse
|
19
|
Wolff A, Frank M, Staehlke S, Springer A, Hahn O, Meyer J, Peters K. 3D Spheroid Cultivation Alters the Extent and Progression of Osteogenic Differentiation of Mesenchymal Stem/Stromal Cells Compared to 2D Cultivation. Biomedicines 2023; 11:biomedicines11041049. [PMID: 37189667 DOI: 10.3390/biomedicines11041049] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Revised: 03/08/2023] [Accepted: 03/17/2023] [Indexed: 03/31/2023] Open
Abstract
Mesenchymal stem/stromal cells (MSC) are capable of progenitor cell fraction renewal or tissue-specific differentiation. These properties are maintained during in vitro cultivation, making them an interesting model system for testing biological and pharmacological compounds. Cell cultivation in 2D is commonly used to study cellular responses, but the 2D environment does not reflect the structural situation of most cell types. Therefore, 3D culture systems have been developed to provide a more accurate physiological environment in terms of cell–cell interactions. Since knowledge about the effects of 3D culture on specific differentiation processes is limited, we studied the effects on osteogenic differentiation and the release of factors affecting bone metabolism for up to 35 days and compared them with the effects in 2D culture. We demonstrated that the selected 3D model allowed the rapid and reliable formation of spheroids that were stable over several weeks and both accelerated and enhanced osteogenic differentiation compared with the 2D culture. Thus, our experiments provide new insights into the effects of cell arrangement of MSC in 2D and 3D. However, due to the different culture dimensions, various detection methods had to be chosen, which in principle limits the explanatory power of the comparison between 2D and 3D cultures.
Collapse
|
20
|
Tumor Spheroids as Model to Design Acoustically Mediated Drug Therapies: A Review. Pharmaceutics 2023; 15:pharmaceutics15030806. [PMID: 36986667 PMCID: PMC10056013 DOI: 10.3390/pharmaceutics15030806] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Revised: 02/22/2023] [Accepted: 02/27/2023] [Indexed: 03/06/2023] Open
Abstract
Tumor spheroids as well as multicellular tumor spheroids (MCTSs) are promising 3D in vitro tumor models for drug screening, drug design, drug targeting, drug toxicity, and validation of drug delivery methods. These models partly reflect the tridimensional architecture of tumors, their heterogeneity and their microenvironment, which can alter the intratumoral biodistribution, pharmacokinetics, and pharmacodynamics of drugs. The present review first focuses on current spheroid formation methods and then on in vitro investigations exploiting spheroids and MCTS for designing and validating acoustically mediated drug therapies. We discuss the limitations of the current studies and future perspectives. Various spheroid formation methods enable the easy and reproducible generation of spheroids and MCTSs. The development and assessment of acoustically mediated drug therapies have been mainly demonstrated in spheroids made up of tumor cells only. Despite the promising results obtained with these spheroids, the successful evaluation of these therapies will need to be addressed in more relevant 3D vascular MCTS models using MCTS-on-chip platforms. These MTCSs will be generated from patient-derived cancer cells and nontumor cells, such as fibroblasts, adipocytes, and immune cells.
Collapse
|
21
|
Wychowaniec JK, Brougham DF. Emerging Magnetic Fabrication Technologies Provide Controllable Hierarchically-Structured Biomaterials and Stimulus Response for Biomedical Applications. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2022; 9:e2202278. [PMID: 36228106 PMCID: PMC9731717 DOI: 10.1002/advs.202202278] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Revised: 09/12/2022] [Indexed: 06/16/2023]
Abstract
Multifunctional nanocomposites which exhibit well-defined physical properties and encode spatiotemporally-controlled responses are emerging as components for advanced responsive systems. For biomedical applications magnetic nanocomposite materials have attracted significant attention due to their ability to respond to spatially and temporally varying magnetic fields. The current state-of-the-art in development and fabrication of magnetic hydrogels toward biomedical applications is described. There is accelerating progress in the field due to advances in manufacturing capabilities. Three categories can be identified: i) Magnetic hydrogelation, DC magnetic fields are used during solidification/gelation for aligning particles; ii) additive manufacturing of magnetic materials, 3D printing technologies are used to develop spatially-encoded magnetic properties, and more recently; iii) magnetic additive manufacturing, magnetic responses are applied during the printing process to develop increasingly complex structural arrangement that may recapitulate anisotropic tissue structure and function. The magnetic responsiveness of conventionally and additively manufactured magnetic hydrogels are described along with recent advances in soft magnetic robotics, and the categorization is related to final architecture and emergent properties. Future challenges and opportunities, including the anticipated role of combinatorial approaches in developing 4D-responsive functional materials for tackling long-standing problems in biomedicine including production of 3D-specified responsive cell scaffolds are discussed.
Collapse
Affiliation(s)
- Jacek K. Wychowaniec
- School of ChemistryUniversity College DublinBelfieldDublin 4Ireland
- AO Research Institute DavosClavadelerstrasse 8Davos7270Switzerland
| | | |
Collapse
|
22
|
Banerjee D, Singh YP, Datta P, Ozbolat V, O'Donnell A, Yeo M, Ozbolat IT. Strategies for 3D bioprinting of spheroids: A comprehensive review. Biomaterials 2022; 291:121881. [DOI: 10.1016/j.biomaterials.2022.121881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Revised: 10/04/2022] [Accepted: 10/23/2022] [Indexed: 11/17/2022]
|
23
|
Shakeri R, Savari B, Sheikholeslami MN, Radjabian T, Khorshidi J, Safavi M. Untargeted Metabolomics Analysis of Crocus cancellatus subsp. damascenus (Herb.) B. Mathew Stigmas and Their Anticarcinogenic Effect on Breast Cancer Cells. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE : ECAM 2022; 2022:3861783. [PMID: 36016682 PMCID: PMC9398734 DOI: 10.1155/2022/3861783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 06/01/2022] [Accepted: 06/24/2022] [Indexed: 11/17/2022]
Abstract
Safranal, crocin, crocetin, and picrocrocin are major known compounds in the stigma extract of Crocus sativus with various medicinal properties. Crocus cancellatus is another Crocus species that grows extensively in Iran's various regions, such as the Kurdistan province. The predominant metabolites and biological properties of C. cancellatus have not yet been investigated. The ingredients of the stigma ethanol extract of C. cancellatus were investigated using gas chromatography-mass spectrometry (GC-MS) and liquid chromatography with tandem mass spectrometry (LC-MS). The ROIMCR approach was performed to analyze the LC-MS full scan data sets. This method searches the MS regions of interest (ROI) data in the m/z domain and analyses the results using the multivariate curve-resolution alternating least squares (MCR-ALS) chemometrics technique for simultaneous resolution of two extracts. Also, the antiproliferative properties of C. cancellatus against MDA-MB-231 and MCF-7 cancer cells were examined by MTT, dual acridine orange/ethidium bromide test, Annexin V-FITC/PI, and zymography. The GC-MS and LC-MS untargeted metabolomics data analysis of the extract indicated the presence of cytotoxic agents including safranal, crocin, picrocrocin, and crocetin in the stigma ethanol extract of C. cancellatus. Biological tests showed that the viability of MDA-MB-231 and MCF-7 cancer cells is decreased following C. cancellatus treatment in a time- and dose-dependent way in both monolayer and 3D cell cultures. The MCF-7 cell spheroids had greater resistance to the cytotoxic activity of the extract in 3D cell culture than the MDA-MB-231 cell spheroids. The morphological changes of the cells treated with C. cancellatus stigmas extract were indicative of apoptosis. Zymography analysis revealed a similar trend of matrix metallopeptidase-2 (MMP-2) and matrix metallopeptidase-9 (MMP-9) activity in the treated cells with C. cancellatus extract in comparison with doxorubicin treatment as a positive control. The findings of this research indicate that the ethanolic extract of C. cancellatus stigmas was a good source of bioactive metabolites with anticancer activity.
Collapse
Affiliation(s)
- Raheleh Shakeri
- Department of Biological Science and Biotechnology, Faculty of Science, University of Kurdistan, Sanandaj, Iran
| | - Bahram Savari
- Department of Biological Science and Biotechnology, Faculty of Science, University of Kurdistan, Sanandaj, Iran
| | - Mahsa N. Sheikholeslami
- Center of Excellence in Electrochemistry, Faculty of Chemistry, University of Tehran, Tehran 1417614418, Iran
| | | | - Jalal Khorshidi
- Department of Horticultural Science and Engineering, Research Center of Medicinal Plants Breeding and Development, University of Kurdistan, Sanandaj, Iran
| | - Maliheh Safavi
- Department of Biotechnology, Iranian Research Organization for Science and Technology, Tehran 13353-5111, Iran
| |
Collapse
|
24
|
Henrique RBL, Lima RRM, Monteiro CAP, Oliveira WF, Pereira G, Cabral Filho PE, Fontes A. Advances in the study of spheroids as versatile models to evaluate biological interactions of inorganic nanoparticles. Life Sci 2022; 302:120657. [PMID: 35609631 DOI: 10.1016/j.lfs.2022.120657] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2022] [Revised: 05/10/2022] [Accepted: 05/18/2022] [Indexed: 12/26/2022]
Abstract
Spheroids are in vitro three-dimensional multicellular microstructures able to mimic the biological microenvironment, including the complexity of tumor architecture. Therefore, results closer to those expected for in vivo organisms can be reached using spheroids compared to the cell culture monolayer model. Inorganic nanoparticles (NPs) have also been playing relevant roles in the comprehension of biological processes. Moreover, they have been probed as novel diagnostic and therapeutical nanosystems. In this context, in this review, we present applications, published in the last five years, which show that spheroids can be versatile models to study and evaluate biological interactions involving inorganic NPs. Applications of spheroids associated with (i) basic studies to assess the penetration profile of nanostructures, (ii) the evaluation of NP toxicity, and (iii) NP-based therapeutical approaches are described. Fundamentals of spheroids and their formation methods are also included. We hope that this review can be a reference and guide future investigations related to this interesting three-dimensional biological model, favoring advances to Nanobiotechnology.
Collapse
Affiliation(s)
- Rafaella B L Henrique
- Departamento de Biofísica e Radiobiologia, Centro de Biociências, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Rennan R M Lima
- Departamento de Biofísica e Radiobiologia, Centro de Biociências, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Camila A P Monteiro
- Departamento de Biofísica e Radiobiologia, Centro de Biociências, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Weslley F Oliveira
- Departamento de Bioquímica, Centro de Biociências, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Goreti Pereira
- Departamento de Química Fundamental, Centro de Ciências Exatas e da Natureza, Universidade Federal de Pernambuco, Recife, PE, Brazil
| | - Paulo E Cabral Filho
- Departamento de Biofísica e Radiobiologia, Centro de Biociências, Universidade Federal de Pernambuco, Recife, PE, Brazil.
| | - Adriana Fontes
- Departamento de Biofísica e Radiobiologia, Centro de Biociências, Universidade Federal de Pernambuco, Recife, PE, Brazil.
| |
Collapse
|
25
|
Moshe Halamish H, Zlotver I, Sosnik A. Polymeric nanoparticles surface-complexed with boric acid actively target solid tumors overexpressing sialic acid. J Colloid Interface Sci 2022; 626:916-929. [PMID: 35835042 DOI: 10.1016/j.jcis.2022.07.027] [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: 05/04/2022] [Revised: 06/22/2022] [Accepted: 07/04/2022] [Indexed: 11/16/2022]
Abstract
Sialic acid is a fundamental component of the tumor microenvironment, modulates cell-cell and cell-extracellular matrix interactions and is associated with bad prognosis and clinical outcomes in different cancers. Capitalizing on the ability of boric acid to form cyclic esters with diols, in this work, we design self-assembled multi-micellar colloidal systems of an amphiphilic poly(vinyl alcohol)-g-poly(methyl methacrylate) copolymer surface-modified with boric acid for the active targeting of solid tumors that overexpress sialic acid. Nanoparticles display sizes in the 100-200 nm range and a spherical morphology, as determined by dynamic light scattering and high resolution-scanning electron microscopy, respectively. The uptake and anti-proliferative activity are assessed in 2D and 3D models of rhabdomyosarcoma in vitro. Surface boration increases the nanoparticle permeability and uptake, especially in rhabdomyosarcoma spheroids that overexpress sialic acid to a greater extent than 2D cultures. The biodistribution of non-borated and borated nanoparticles upon intravenous injection to a subcutaneous rhabdomyosarcoma murine xenograft model confirm a statistically significant increase in the intertumoral accumulation of the modified nanocarriers with respect to the unmodified counterparts and a sharp decrease in major clearance organs such as the liver. Overall, our results highlight the promise of these borated nanomaterials to actively target hypersialylated solid tumors.
Collapse
Affiliation(s)
- Hen Moshe Halamish
- Laboratory of Pharmaceutical Nanomaterials Science, Department of Materials Science and Engineering, Technion-Israel Institute of Technology, De-Jur Building, Office 607, Technion City 3200003 Haifa, Israel
| | - Ivan Zlotver
- Laboratory of Pharmaceutical Nanomaterials Science, Department of Materials Science and Engineering, Technion-Israel Institute of Technology, De-Jur Building, Office 607, Technion City 3200003 Haifa, Israel
| | - Alejandro Sosnik
- Laboratory of Pharmaceutical Nanomaterials Science, Department of Materials Science and Engineering, Technion-Israel Institute of Technology, De-Jur Building, Office 607, Technion City 3200003 Haifa, Israel.
| |
Collapse
|
26
|
Jeong Y, Tin A, Irudayaraj J. Flipped Well-Plate Hanging-Drop Technique for Growing Three-Dimensional Tumors. Front Bioeng Biotechnol 2022; 10:898699. [PMID: 35860331 PMCID: PMC9289396 DOI: 10.3389/fbioe.2022.898699] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2022] [Accepted: 05/25/2022] [Indexed: 11/24/2022] Open
Abstract
Three-dimensional (3D) tumor culture techniques are gaining popularity as in vitro models of tumoral tissue analogues. Despite the widespread interest, need, and present-day effort, most of the 3D tumor culturing methodologies have not gone beyond the inventors’ laboratories. This, in turn, limits their applicability and standardization. In this study, we introduce a straightforward and user-friendly approach based on standard 96-well plates with basic amenities for growing 3D tumors in a scaffold-free/scaffold-based format. Hanging drop preparation can be easily employed by flipping a universal 96-well plate. The droplets of the medium generated by the well-plate flip (WPF) method can be easily modified to address various mechanisms and processes in cell biology, including cancer. To demonstrate the applicability and practicality of the conceived approach, we utilized human colorectal carcinoma cells (HCT116) to first show the generation of large scaffold-free 3D tumor spheroids over 1.5 mm in diameter in single-well plates. As a proof-of-concept, we also demonstrate matrix-assisted tumor culture techniques in advancing the broader use of 3D culture systems. The conceptualized WPF approach can be adapted for a range of applications in both basic and applied biological/engineering research.
Collapse
Affiliation(s)
- Yoon Jeong
- Department of Bioengineering, University of Illinois at Urbana‐Champaign, Urbana, IL, United States
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Ashley Tin
- Department of Computer Science, University of Illinois at Urbana‐Champaign, Urbana, IL, United States
| | - Joseph Irudayaraj
- Department of Bioengineering, University of Illinois at Urbana‐Champaign, Urbana, IL, United States
- Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- Carl R. Woese Institute for Genomic Biology, University of Illinois at Urbana-Champaign, Urbana, IL, United States
- *Correspondence: Joseph Irudayaraj,
| |
Collapse
|
27
|
Paškevičiūtė M, Petrikaitė V. Effect of natural flavonoids to reverse P-glycoprotein-related multidrug resistance in breast cancer cell cultures. Am J Cancer Res 2022; 12:2526-2538. [PMID: 35812069 PMCID: PMC9251692] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Accepted: 05/17/2022] [Indexed: 06/15/2023] Open
Abstract
The aim of the research was to evaluate the influence of two P-glycoprotein (P-gp) inhibitors silymarin and quercetin on anticancer drug doxorubicin (DOX) and pegylated liposomal doxorubicin (PLD) delivery into breast cancer cells (2D cultures) and cancer cell spheroids (3D cultures) at different pH. The cytotoxicity of the compounds was assessed using MTT assay. Spheroids were generated using magnetic 3D Bioprinting method. The uptake of DOX and PLD into monolayer-cultured cells and spheroids was assessed by fluorescence microscopy. Both tested flavonoids did not increase DOX and PLD levels into monolayer-cultured 4T1 cells and 4T1 cell spheroids. However, both silymarin and quercetin enhanced DOX and PLD uptake into JC cell cultures. Silymarin and quercetin may modulate DOX and PLD transport into monolayer-cultured cells and three-dimensional cancer cell cultures depending on P-gp activity.
Collapse
Affiliation(s)
- Miglė Paškevičiūtė
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health SciencesSukilėlių pr. 13, LT-50162, Kaunas, Lithuania
| | - Vilma Petrikaitė
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health SciencesSukilėlių pr. 13, LT-50162, Kaunas, Lithuania
- Institute of Physiology and Pharmacology, Faculty of Medicine, Lithuanian University of Health SciencesA. Mickevičiaus g. 9, LT-44307, Kaunas, Lithuania
| |
Collapse
|
28
|
Rodboon T, Yodmuang S, Chaisuparat R, Ferreira JN. Development of high-throughput lacrimal gland organoid platforms for drug discovery in dry eye disease. SLAS DISCOVERY : ADVANCING LIFE SCIENCES R & D 2022; 27:151-158. [PMID: 35058190 DOI: 10.1016/j.slasd.2021.11.002] [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] [Indexed: 06/14/2023]
Abstract
Dysfunction and damage of the lacrimal gland (LG) results in ocular discomfort and dry eye disease (DED). Current therapies for DED do not fully replenish the necessary lubrication to rescue optimal vision. New drug discovery for DED has been limited perhaps because in vitro models cannot mimic the biology of the native LG. The existing platforms for LG organoid culture are scarce and still not ready for consistency and scale up production towards drug screening. The magnetic three-dimensional (3D) bioprinting (M3DB) is a novel system for 3D in vitro biofabrication of cellularized tissues using magnetic nanoparticles to bring cells together. M3DB provides a scalable platform for consistent handling of spheroid-like cell cultures facilitating consistent biofabrication of organoids. Previously, we successfully generated innervated secretory epithelial organoids from human dental pulp stem cells with M3DB and found that this platform is feasible for epithelial organoid bioprinting. Research targeting LG organogenesis, drug discovery for DED has extensively used mouse models. However, certain inter-species differences between mouse and human must be considered. Porcine LG appear to have more similarities to human LG than the mouse counterparts. We have conducted preliminary studies with the M3DB for fabricating LG organoids from primary cells isolated from murine and porcine LG, and found that this platform provides robust LG organoids for future potential high-throughput analysis and drug discovery. The LG organoid holds promise to be a functional model of tearing, a platform for drug screening, and may offer clinical applications for DED.
Collapse
Affiliation(s)
- Teerapat Rodboon
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Supansa Yodmuang
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand; Department of Research Affairs, Faculty of Medicine, Chulalongkorn University, Bangkok, Thailand
| | - Risa Chaisuparat
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand; Department of Oral Pathology, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand
| | - Joao N Ferreira
- Avatar Biotechnologies for Oral Health and Healthy Longevity Research Unit, Faculty of Dentistry, Chulalongkorn University, Bangkok, Thailand; Faculty of Dentistry, National University of Singapore, Singapore.
| |
Collapse
|
29
|
Filippi M, Garello F, Yasa O, Kasamkattil J, Scherberich A, Katzschmann RK. Engineered Magnetic Nanocomposites to Modulate Cellular Function. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2104079. [PMID: 34741417 DOI: 10.1002/smll.202104079] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/13/2021] [Indexed: 06/13/2023]
Abstract
Magnetic nanoparticles (MNPs) have various applications in biomedicine, including imaging, drug delivery and release, genetic modification, cell guidance, and patterning. By combining MNPs with polymers, magnetic nanocomposites (MNCs) with diverse morphologies (core-shell particles, matrix-dispersed particles, microspheres, etc.) can be generated. These MNCs retain the ability of MNPs to be controlled remotely using external magnetic fields. While the effects of these biomaterials on the cell biology are still poorly understood, such information can help the biophysical modulation of various cellular functions, including proliferation, adhesion, and differentiation. After recalling the basic properties of MNPs and polymers, and describing their coassembly into nanocomposites, this review focuses on how polymeric MNCs can be used in several ways to affect cell behavior. A special emphasis is given to 3D cell culture models and transplantable grafts, which are used for regenerative medicine, underlining the impact of MNCs in regulating stem cell differentiation and engineering living tissues. Recent advances in the use of MNCs for tissue regeneration are critically discussed, particularly with regard to their prospective involvement in human therapy and in the construction of advanced functional materials such as magnetically operated biomedical robots.
Collapse
Affiliation(s)
- Miriam Filippi
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Francesca Garello
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Via Nizza 52, Torino, 10126, Italy
| | - Oncay Yasa
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| | - Jesil Kasamkattil
- Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, Basel, 4031, Switzerland
| | - Arnaud Scherberich
- Department of Biomedicine, University Hospital Basel, Hebelstrasse 20, Basel, 4031, Switzerland
- Department of Biomedical Engineering, University of Basel, Gewerbestrasse 14, Allschwil, 4123, Switzerland
| | - Robert K Katzschmann
- Soft Robotics Laboratory, ETH Zurich, Tannenstrasse 3, Zurich, 8092, Switzerland
| |
Collapse
|
30
|
Three-dimensional models: a novel approach for lymphoma research. J Cancer Res Clin Oncol 2022; 148:753-765. [DOI: 10.1007/s00432-021-03897-9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Accepted: 12/21/2021] [Indexed: 12/12/2022]
|
31
|
Dermal Delivery of Lipid Nanoparticles: Effects on Skin and Assessment of Absorption and Safety. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2022; 1357:83-114. [DOI: 10.1007/978-3-030-88071-2_4] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
32
|
Barbosa MAG, Xavier CPR, Pereira RF, Petrikaitė V, Vasconcelos MH. 3D Cell Culture Models as Recapitulators of the Tumor Microenvironment for the Screening of Anti-Cancer Drugs. Cancers (Basel) 2021; 14:190. [PMID: 35008353 PMCID: PMC8749977 DOI: 10.3390/cancers14010190] [Citation(s) in RCA: 124] [Impact Index Per Article: 31.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2021] [Revised: 12/23/2021] [Accepted: 12/29/2021] [Indexed: 12/12/2022] Open
Abstract
Today, innovative three-dimensional (3D) cell culture models have been proposed as viable and biomimetic alternatives for initial drug screening, allowing the improvement of the efficiency of drug development. These models are gaining popularity, given their ability to reproduce key aspects of the tumor microenvironment, concerning the 3D tumor architecture as well as the interactions of tumor cells with the extracellular matrix and surrounding non-tumor cells. The development of accurate 3D models may become beneficial to decrease the use of laboratory animals in scientific research, in accordance with the European Union's regulation on the 3R rule (Replacement, Reduction, Refinement). This review focuses on the impact of 3D cell culture models on cancer research, discussing their advantages, limitations, and compatibility with high-throughput screenings and automated systems. An insight is also given on the adequacy of the available readouts for the interpretation of the data obtained from the 3D cell culture models. Importantly, we also emphasize the need for the incorporation of additional and complementary microenvironment elements on the design of 3D cell culture models, towards improved predictive value of drug efficacy.
Collapse
Affiliation(s)
- Mélanie A. G. Barbosa
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (M.A.G.B.); (C.P.R.X.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
| | - Cristina P. R. Xavier
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (M.A.G.B.); (C.P.R.X.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
| | - Rúben F. Pereira
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
- Biofabrication Group, INEB—Instituto de Engenharia Biomédica, Universidade do Porto, 4200-135 Porto, Portugal
- ICBAS—Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, 4050-313 Porto, Portugal
| | - Vilma Petrikaitė
- Laboratory of Drug Targets Histopathology, Institute of Cardiology, Lithuanian University of Health Sciences, A. Mickevičiaus g 9, LT-44307 Kaunas, Lithuania;
- Institute of Biotechnology, Life Sciences Center, Vilnius University, Saulėtekio al. 7, LT-10257 Vilnius, Lithuania
| | - M. Helena Vasconcelos
- Cancer Drug Resistance Group, IPATIMUP—Institute of Molecular Pathology and Immunology, University of Porto, 4200-135 Porto, Portugal; (M.A.G.B.); (C.P.R.X.)
- i3S—Instituto de Investigação e Inovação em Saúde, Universidade do Porto, 4200-135 Porto, Portugal;
- Department of Biological Sciences, FFUP—Faculty of Pharmacy of the University of Porto, 4050-313 Porto, Portugal
| |
Collapse
|
33
|
Caleffi JT, Aal MCE, Gallindo HDOM, Caxali GH, Crulhas BP, Ribeiro AO, Souza GR, Delella FK. Magnetic 3D cell culture: State of the art and current advances. Life Sci 2021; 286:120028. [PMID: 34627776 DOI: 10.1016/j.lfs.2021.120028] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 09/24/2021] [Accepted: 10/02/2021] [Indexed: 02/07/2023]
Abstract
Cell culture is an important tool for the understanding of cell biology and behavior. In vitro cultivation has been increasingly indispensable for biomedical, pharmaceutical, and biotechnology research. Nevertheless, with the demand for in vitro experimentation strategies more representative of in vivo conditions, tridimensional (3D) cell culture models have been successfully developed. Although these 3D models are efficient and address critical questions from different research areas, there are considerable differences between the existing techniques regarding both elaboration and cost. In light of this, this review describes the construction of 3D spheroids using magnetization while bringing the most recent updates in this field. Magnetic 3D cell culture consists of magnetizing cells using an assembly of gold and iron oxide nanoparticles cross-linked with poly-l-lysine nanoparticles. Then, 3D culture formation in special plates with the assistance of magnets for levitation or bioprinting. Here, we discuss magnetic 3D cell culture advancements, including tumor microenvironment, tissue reconstruction, blood vessel engineering, toxicology, cytotoxicity, and 3D culture of cardiomyocytes, bronchial and pancreatic cells.
Collapse
Affiliation(s)
- Juliana Trindade Caleffi
- São Paulo State University (UNESP), Institute of Biosciences, Department of Structural and Functional Biology, Botucatu, São Paulo, Brazil
| | - Mirian Carolini Esgoti Aal
- São Paulo State University (UNESP), Institute of Biosciences, Department of Structural and Functional Biology, Botucatu, São Paulo, Brazil
| | | | - Gabriel Henrique Caxali
- São Paulo State University (UNESP), Institute of Biosciences, Department of Structural and Functional Biology, Botucatu, São Paulo, Brazil
| | | | - Amanda Oliveira Ribeiro
- São Paulo State University (UNESP), Institute of Biosciences, Department of Structural and Functional Biology, Botucatu, São Paulo, Brazil
| | - Glauco R Souza
- University of Texas Health Sciences Center at Houston, Houston, TX, USA
| | - Flávia Karina Delella
- São Paulo State University (UNESP), Institute of Biosciences, Department of Structural and Functional Biology, Botucatu, São Paulo, Brazil.
| |
Collapse
|
34
|
Wu Y, Zhou Y, Qin X, Liu Y. From cell spheroids to vascularized cancer organoids: Microfluidic tumor-on-a-chip models for preclinical drug evaluations. BIOMICROFLUIDICS 2021; 15:061503. [PMID: 34804315 PMCID: PMC8589468 DOI: 10.1063/5.0062697] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/07/2021] [Accepted: 10/16/2021] [Indexed: 05/14/2023]
Abstract
Chemotherapy is one of the most effective cancer treatments. Starting from the discovery of new molecular entities, it usually takes about 10 years and 2 billion U.S. dollars to bring an effective anti-cancer drug from the benchtop to patients. Due to the physiological differences between animal models and humans, more than 90% of drug candidates failed in phase I clinical trials. Thus, a more efficient drug screening system to identify feasible compounds and pre-exclude less promising drug candidates is strongly desired. For their capability to accurately construct in vitro tumor models derived from human cells to reproduce pathological and physiological processes, microfluidic tumor chips are reliable platforms for preclinical drug screening, personalized medicine, and fundamental oncology research. This review summarizes the recent progress of the microfluidic tumor chip and highlights tumor vascularization strategies. In addition, promising imaging modalities for enhancing data acquisition and machine learning-based image analysis methods to accurately quantify the dynamics of tumor spheroids are introduced. It is believed that the microfluidic tumor chip will serve as a high-throughput, biomimetic, and multi-sensor integrated system for efficient preclinical drug evaluation in the future.
Collapse
Affiliation(s)
- Yue Wu
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, USA
| | - Yuyuan Zhou
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, USA
| | - Xiaochen Qin
- Department of Bioengineering, Lehigh University, Bethlehem, Pennsylvania 18015, USA
| | - Yaling Liu
- Author to whom correspondence should be addressed:
| |
Collapse
|
35
|
Abstract
Organoids have complex three-dimensional structures that exhibit functionalities and feature architectures similar to those of in vivo organs and are developed from adult stem cells, embryonic stem cells, and pluripotent stem cells through a self-organization process. Organoids derived from adult epithelial stem cells are the most mature and extensive. In recent years, using organoid culture techniques, researchers have established various adult human tissue-derived epithelial organoids, including intestinal, colon, lung, liver, stomach, breast, and oral mucosal organoids, all of which exhibit strong research and application prospects. Studies have shown that epithelial organoids are mainly applied in drug discovery, personalized drug response testing, disease mechanism research, and regenerative medicine. In this review, we mainly discuss current organoid culture systems and potential applications of this technique with human epithelial tissue.
Collapse
Affiliation(s)
- Fengjiao Li
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Peng Zhang
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China.,Key Laboratory of Chemical Biology and Traditional Chinese Medicine Research (Hunan Normal University), Ministry of Education, College of Chemistry & Chemical Engineering, Changsha, Hunan 410081, China
| | - Saizhi Wu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| | - Lianwen Yuan
- Department of Geriatric Surgery, The Second Xiangya Hospital of Central South University, Changsha, Hunan 410011, China
| | - Zhonghua Liu
- The National and Local Joint Engineering Laboratory of Animal Peptide Drug Development, College of Life Sciences, Hunan Normal University, Changsha, Hunan 410081, China
| |
Collapse
|
36
|
Zhuang P, Chiang YH, Fernanda MS, He M. Using Spheroids as Building Blocks Towards 3D Bioprinting of Tumor Microenvironment. Int J Bioprint 2021; 7:444. [PMID: 34805601 PMCID: PMC8600307 DOI: 10.18063/ijb.v7i4.444] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Accepted: 10/02/2021] [Indexed: 12/12/2022] Open
Abstract
Cancer still ranks as a leading cause of mortality worldwide. Although considerable efforts have been dedicated to anticancer therapeutics, progress is still slow, partially due to the absence of robust prediction models. Multicellular tumor spheroids, as a major three-dimensional (3D) culture model exhibiting features of avascular tumors, gained great popularity in pathophysiological studies and high throughput drug screening. However, limited control over cellular and structural organization is still the key challenge in achieving in vivo like tissue microenvironment. 3D bioprinting has made great strides toward tissue/organ mimicry, due to its outstanding spatial control through combining both cells and materials, scalability, and reproducibility. Prospectively, harnessing the power from both 3D bioprinting and multicellular spheroids would likely generate more faithful tumor models and advance our understanding on the mechanism of tumor progression. In this review, the emerging concept on using spheroids as a building block in 3D bioprinting for tumor modeling is illustrated. We begin by describing the context of the tumor microenvironment, followed by an introduction of various methodologies for tumor spheroid formation, with their specific merits and drawbacks. Thereafter, we present an overview of existing 3D printed tumor models using spheroids as a focus. We provide a compilation of the contemporary literature sources and summarize the overall advancements in technology and possibilities of using spheroids as building blocks in 3D printed tissue modeling, with a particular emphasis on tumor models. Future outlooks about the wonderous advancements of integrated 3D spheroidal printing conclude this review.
Collapse
Affiliation(s)
- Pei Zhuang
- Department of Pharmaceutics, University of Florida, Gainesville, Florida, 32610, USA
| | - Yi-Hua Chiang
- Department of Pharmaceutics, University of Florida, Gainesville, Florida, 32610, USA
| | | | - Mei He
- Department of Pharmaceutics, University of Florida, Gainesville, Florida, 32610, USA
| |
Collapse
|
37
|
Celikkin N, Presutti D, Maiullari F, Fornetti E, Agarwal T, Paradiso A, Volpi M, Święszkowski W, Bearzi C, Barbetta A, Zhang YS, Gargioli C, Rizzi R, Costantini M. Tackling Current Biomedical Challenges With Frontier Biofabrication and Organ-On-A-Chip Technologies. Front Bioeng Biotechnol 2021; 9:732130. [PMID: 34604190 PMCID: PMC8481890 DOI: 10.3389/fbioe.2021.732130] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Accepted: 08/31/2021] [Indexed: 12/13/2022] Open
Abstract
In the last decades, biomedical research has significantly boomed in the academia and industrial sectors, and it is expected to continue to grow at a rapid pace in the future. An in-depth analysis of such growth is not trivial, given the intrinsic multidisciplinary nature of biomedical research. Nevertheless, technological advances are among the main factors which have enabled such progress. In this review, we discuss the contribution of two state-of-the-art technologies-namely biofabrication and organ-on-a-chip-in a selection of biomedical research areas. We start by providing an overview of these technologies and their capacities in fabricating advanced in vitro tissue/organ models. We then analyze their impact on addressing a range of current biomedical challenges. Ultimately, we speculate about their future developments by integrating these technologies with other cutting-edge research fields such as artificial intelligence and big data analysis.
Collapse
Affiliation(s)
- Nehar Celikkin
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Dario Presutti
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| | - Fabio Maiullari
- Istituto Nazionale Genetica Molecolare INGM “Romeo Ed Enrica Invernizzi”, Milan, Italy
| | | | - Tarun Agarwal
- Department of Biotechnology, Indian Institute of Technology Kharagpur, Kharagpur, India
| | - Alessia Paradiso
- Faculty of Materials Science and Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Marina Volpi
- Faculty of Materials Science and Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Wojciech Święszkowski
- Faculty of Materials Science and Engineering, Warsaw University of Technology, Warsaw, Poland
| | - Claudia Bearzi
- Istituto Nazionale Genetica Molecolare INGM “Romeo Ed Enrica Invernizzi”, Milan, Italy
- Institute of Genetic and Biomedical Research, National Research Council of Italy (IRGB-CNR), Milan, Italy
| | - Andrea Barbetta
- Department of Chemistry, Sapienza University of Rome, Rome, Italy
| | - Yu Shrike Zhang
- Division of Engineering in Medicine, Department of Medicine, Brigham and Women’s Hospital and Harvard Medical School, Cambridge, MA, United States
| | - Cesare Gargioli
- Department of Biology, Rome University Tor Vergata, Rome, Italy
| | - Roberto Rizzi
- Istituto Nazionale Genetica Molecolare INGM “Romeo Ed Enrica Invernizzi”, Milan, Italy
- Institute of Genetic and Biomedical Research, National Research Council of Italy (IRGB-CNR), Milan, Italy
- Institute of Biomedical Technologies, National Research Council of Italy (ITB-CNR), Milan, Italy
| | - Marco Costantini
- Institute of Physical Chemistry, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
38
|
Oliveira C, Forster C, Feitosa V, Baby A, Léo P, Rangel-Yagui C. Catalase-loaded polymersomes as a promising safe ingredient to active photoprotection. JOURNAL OF PHOTOCHEMISTRY AND PHOTOBIOLOGY 2021. [DOI: 10.1016/j.jpap.2021.100056] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
|
39
|
Kotze LA, Beltran CGG, Lang D, Loxton AG, Cooper S, Meiring M, Koegelenberg CFN, Allwood BW, Malherbe ST, Hiemstra AM, Glanzmann B, Kinnear C, Walzl G, du Plessis N. Establishment of a Patient-Derived, Magnetic Levitation-Based, Three-Dimensional Spheroid Granuloma Model for Human Tuberculosis. mSphere 2021; 6:e0055221. [PMID: 34287004 PMCID: PMC8386456 DOI: 10.1128/msphere.00552-21] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Accepted: 06/17/2021] [Indexed: 12/18/2022] Open
Abstract
Tuberculous granulomas that develop in response to Mycobacterium tuberculosis (M. tuberculosis) infection are highly dynamic entities shaped by the host immune response and disease kinetics. Within this microenvironment, immune cell recruitment, polarization, and activation are driven not only by coexisting cell types and multicellular interactions but also by M. tuberculosis-mediated changes involving metabolic heterogeneity, epigenetic reprogramming, and rewiring of the transcriptional landscape of host cells. There is an increased appreciation of the in vivo complexity, versatility, and heterogeneity of the cellular compartment that constitutes the tuberculosis (TB) granuloma and the difficulty in translating findings from animal models to human disease. Here, we describe a novel biomimetic in vitro three-dimensional (3D) human lung spheroid granuloma model, resembling early "innate" and "adaptive" stages of the TB granuloma spectrum, and present results of histological architecture, host transcriptional characterization, mycobacteriological features, cytokine profiles, and spatial distribution of key immune cells. A range of manipulations of immune cell populations in these spheroid granulomas will allow the study of host/pathogen pathways involved in the outcome of infection, as well as pharmacological interventions. IMPORTANCE TB is a highly infectious disease, with granulomas as its hallmark. Granulomas play an important role in the control of M. tuberculosis infection and as such are crucial indicators for our understanding of host resistance to TB. Correlates of risk and protection to M. tuberculosis are still elusive, and the granuloma provides the perfect environment in which to study the immune response to infection and broaden our understanding thereof; however, human granulomas are difficult to obtain, and animal models are costly and do not always faithfully mimic human immunity. In fact, most TB research is conducted in vitro on immortalized or primary immune cells and cultured in two dimensions on flat, rigid plastic, which does not reflect in vivo characteristics. We have therefore conceived a 3D, human in vitro spheroid granuloma model which allows researchers to study features of granuloma-forming diseases in a 3D structural environment resembling in vivo granuloma architecture and cellular orientation.
Collapse
Affiliation(s)
- Leigh A. Kotze
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Caroline G. G. Beltran
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Dirk Lang
- Confocal and Light Microscopy Imaging Facility, University of Cape Town, Cape Town, South Africa
| | - Andre G. Loxton
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Susan Cooper
- Confocal and Light Microscopy Imaging Facility, University of Cape Town, Cape Town, South Africa
| | - Maynard Meiring
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Coenraad F. N. Koegelenberg
- Division of Pulmonology, Department of Medicine, Stellenbosch University and Tygerberg Academic Hospital, Cape Town, South Africa
| | - Brian W. Allwood
- Division of Pulmonology, Department of Medicine, Stellenbosch University and Tygerberg Academic Hospital, Cape Town, South Africa
| | - Stephanus T. Malherbe
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Andriette M. Hiemstra
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Brigitte Glanzmann
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council Genomics Centre, Cape Town, South Africa
| | - Craig Kinnear
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council Genomics Centre, Cape Town, South Africa
| | - Gerhard Walzl
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Nelita du Plessis
- DST-NRF Centre of Excellence for Biomedical Tuberculosis Research, South African Medical Research Council Centre for Tuberculosis Research, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| |
Collapse
|
40
|
Jeon S, Lee SH, Ahmed SB, Han J, Heo SJ, Kang HW. 3D cell aggregate printing technology and its applications. Essays Biochem 2021; 65:467-480. [PMID: 34223609 PMCID: PMC11293493 DOI: 10.1042/ebc20200128] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/14/2021] [Accepted: 06/08/2021] [Indexed: 12/27/2022]
Abstract
Various cell aggregate culture technologies have been developed and actively applied to tissue engineering and organ-on-a-chip. However, the conventional culture technologies are labor-intensive, and their outcomes are highly user dependent. In addition, the technologies cannot be used to produce three-dimensional (3D) complex tissues. In this regard, 3D cell aggregate printing technology has attracted increased attention from many researchers owing to its 3D processability. The technology allows the fabrication of 3D freeform constructs using multiple types of cell aggregates in an automated manner. Technological advancement has resulted in the development of a printing technology with a high resolution of approximately 20 μm in 3D space. A high-speed printing technology that can print a cell aggregate in milliseconds has also been introduced. The developed aggregate printing technologies are being actively applied to produce various types of engineered tissues. Although various types of high-performance printing technologies have been developed, there are still some technical obstacles in the fabrication of engineered tissues that mimic the structure and function of native tissues. This review highlights the central importance and current technical level of 3D cell aggregate printing technology, and their applications to tissue/disease models, artificial tissues, and drug-screening platforms. The paper also discusses the remaining hurdles and future directions of the printing processes.
Collapse
Affiliation(s)
- Seunggyu Jeon
- School of Life Sciences, Ulsan National Institute of Science and Technology, 50, UNIST-gil, Ulju-gun 44919, Ulsan, South Korea
| | - Se-Hwan Lee
- School of Life Sciences, Ulsan National Institute of Science and Technology, 50, UNIST-gil, Ulju-gun 44919, Ulsan, South Korea
- Department of Orthopaedic Surgery, University of Pennsylvania, 36 Street and Hamilton Walk, Philadelphia, PA 19104, U.S.A
| | - Saeed B. Ahmed
- Department of Orthopaedic Surgery, University of Pennsylvania, 36 Street and Hamilton Walk, Philadelphia, PA 19104, U.S.A
| | - Jonghyeuk Han
- School of Life Sciences, Ulsan National Institute of Science and Technology, 50, UNIST-gil, Ulju-gun 44919, Ulsan, South Korea
| | - Su-Jin Heo
- Department of Orthopaedic Surgery, University of Pennsylvania, 36 Street and Hamilton Walk, Philadelphia, PA 19104, U.S.A
| | - Hyun-Wook Kang
- School of Life Sciences, Ulsan National Institute of Science and Technology, 50, UNIST-gil, Ulju-gun 44919, Ulsan, South Korea
| |
Collapse
|
41
|
Shrestha S, Lekkala VKR, Acharya P, Siddhpura D, Lee MY. Recent advances in microarray 3D bioprinting for high-throughput spheroid and tissue culture and analysis. Essays Biochem 2021; 65:481-489. [PMID: 34296737 PMCID: PMC9270997 DOI: 10.1042/ebc20200150] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2021] [Revised: 06/29/2021] [Accepted: 06/30/2021] [Indexed: 12/26/2022]
Abstract
Three-dimensional (3D) cell culture in vitro has proven to be more physiologically relevant than two-dimensional (2D) culture of cell monolayers, thus more predictive in assessing efficacy and toxicity of compounds. There have been several 3D cell culture techniques developed, which include spheroid and multicellular tissue cultures. Cell spheroids have been generated from single or multiple cell types cultured in ultralow attachment (ULA) well plates and hanging droplet plates. In general, cell spheroids are formed in a relatively short period of culture, in the absence of extracellular matrices (ECMs), via gravity-driven self-aggregation, thus having limited ability to self-organization in layered structure. On the other hand, multicellular tissue cultures including miniature tissues derived from pluripotent stem cells and adult stem cells (a.k.a. 'organoids') and 3D bioprinted tissue constructs require biomimetic hydrogels or ECMs and show highly ordered structure due to spontaneous self-organization of cells during differentiation and maturation processes. In this short review article, we summarize traditional methods of spheroid and multicellular tissue cultures as well as their technical challenges, and introduce how droplet-based, miniature 3D bioprinting ('microarray 3D bioprinting') can be used to improve assay throughput and reproducibility for high-throughput, predictive screening of compounds. Several platforms including a micropillar chip and a 384-pillar plate developed to facilitate miniature spheroid and tissue cultures via microarray 3D bioprinting are introduced. We excluded microphysiological systems (MPSs) in this article although they are important tissue models to simulate multiorgan interactions.
Collapse
Affiliation(s)
- Sunil Shrestha
- Department of Biomedical Engineering, University of North Texas, 1155 Union Circle, Denton, Texas 76203, United States
| | - Vinod Kumar Reddy Lekkala
- Department of Biomedical Engineering, University of North Texas, 1155 Union Circle, Denton, Texas 76203, United States
| | - Prabha Acharya
- Department of Biomedical Engineering, University of North Texas, 1155 Union Circle, Denton, Texas 76203, United States
| | - Darshita Siddhpura
- Department of Chemical and Biomedical Engineering, Cleveland State University, 2121 Euclid Ave, Cleveland, Ohio 44115, United States
| | - Moo-Yeal Lee
- Department of Biomedical Engineering, University of North Texas, 1155 Union Circle, Denton, Texas 76203, United States
| |
Collapse
|
42
|
Transcending toward Advanced 3D-Cell Culture Modalities: A Review about an Emerging Paradigm in Translational Oncology. Cells 2021; 10:cells10071657. [PMID: 34359827 PMCID: PMC8304089 DOI: 10.3390/cells10071657] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2021] [Revised: 06/27/2021] [Accepted: 06/28/2021] [Indexed: 02/06/2023] Open
Abstract
Cancer is a disorder characterized by an uncontrollable overgrowth and a fast-moving spread of cells from a localized tissue to multiple organs of the body, reaching a metastatic state. Throughout years, complexity of cancer progression and invasion, high prevalence and incidence, as well as the high rise in treatment failure cases leading to a poor patient prognosis accounted for continuous experimental investigations on animals and cellular models, mainly with 2D- and 3D-cell culture. Nowadays, these research models are considered a main asset to reflect the physiological events in many cancer types in terms of cellular characteristics and features, replication and metastatic mechanisms, metabolic pathways, biomarkers expression, and chemotherapeutic agent resistance. In practice, based on research perspective and hypothesis, scientists aim to choose the best model to approach their understanding and to prove their hypothesis. Recently, 3D-cell models are seen to be highly incorporated as a crucial tool for reflecting the true cancer cell microenvironment in pharmacokinetic and pharmacodynamics studies, in addition to the intensity of anticancer drug response in pharmacogenomics trials. Hence, in this review, we shed light on the unique characteristics of 3D cells favoring its promising usage through a comparative approach with other research models, specifically 2D-cell culture. Plus, we will discuss the importance of 3D models as a direct reflector of the intrinsic cancer cell environment with the newest multiple methods and types available for 3D-cells implementation.
Collapse
|
43
|
Tumor spheroid-based microtumor models for preclinical evaluation of anticancer nanomedicines. JOURNAL OF PHARMACEUTICAL INVESTIGATION 2021. [DOI: 10.1007/s40005-021-00534-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
44
|
Rosette KA, Lander SM, VanOpstall C, Looyenga BD. Three-dimensional coculture provides an improved in vitro model for papillary renal cell carcinoma. Am J Physiol Renal Physiol 2021; 321:F33-F46. [PMID: 34029144 DOI: 10.1152/ajprenal.00141.2021] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Papillary renal cell carcinoma (pRCC) represents the second most common kidney cancer and can be distinguished from other types based on its unique histological architecture and specific pattern of genomic alterations. Sporadic type 1 pRCC is almost universally driven by focal or chromosomal amplification of the receptor tyrosine kinase MET, although the specific mode of its activation is unclear. Although the MET receptors found in human tumor specimens appear highly active, those found on the surface of in vitro-cultured tumor cells are only weakly activated in the absence of exogenous hepatocyte growth factor ligand. Furthermore, pRCC cells cultured in standard two-dimensional conditions with serum fail to respond functionally to MET knockdown or the selective MET inhibitor capmatinib despite clear evidence of kinase inhibition at the molecular level. To better model pRCC in vitro, we developed a three-dimensional coculture system in which renal tumor cells are layered on top of primary fibroblasts in a fashion that mimics the papillary architecture of human tumors. In this three-dimensional spheroid model, the tumor cells survive and proliferate in the absence of serum due to trophic support of hepatocyte growth factor-producing fibroblasts. Unlike tumor cells grown in monoculture, the proliferation of cocultured tumor cells is sensitive to capmatinib and parallels inhibition of MET kinase activity. These findings demonstrate the importance of stromal fibroblasts in pRCC and indicate that accurate in vitro representation of this disease requires the presence of both tumor and fibroblast cells in a structured coculture model.NEW & NOTEWORTHY Two-dimensional monoculture of papillary renal cancer cells fails to replicate several features of the disease found in humans. We hypothesized that this discordance results from lack of trophic support from renal fibroblasts, which are involved in the architecture of human papillary renal tumors. We found that three-dimensional layering of renal cancer cells on top of a fibroblast core using magnetic bioprinting produces a structured spheroid that more faithfully mimics the behavior of human tumors.
Collapse
Affiliation(s)
- Kylee A Rosette
- Department of Chemistry and Biochemistry, Calvin University, Grand Rapids, Michigan
| | - Stephen M Lander
- Department of Chemistry and Biochemistry, Calvin University, Grand Rapids, Michigan
| | - Calvin VanOpstall
- Department of Chemistry and Biochemistry, Calvin University, Grand Rapids, Michigan
| | - Brendan D Looyenga
- Department of Chemistry and Biochemistry, Calvin University, Grand Rapids, Michigan
| |
Collapse
|
45
|
Kapoor S, Gustafson T, Zhang M, Chen YS, Li J, Nguyen N, Perez JET, Dashwood WM, Rajendran P, Dashwood RH. Deacetylase Plus Bromodomain Inhibition Downregulates ERCC2 and Suppresses the Growth of Metastatic Colon Cancer Cells. Cancers (Basel) 2021; 13:cancers13061438. [PMID: 33809839 PMCID: PMC8004213 DOI: 10.3390/cancers13061438] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 03/19/2021] [Indexed: 01/14/2023] Open
Abstract
There is growing evidence that DNA repair factors have clinical value for cancer treatment. Nucleotide excision repair (NER) proteins, including excision repair cross-complementation group 2 (ERCC2), play a critical role in maintaining genome integrity. Here, we examined ERCC2 expression following epigenetic combination drug treatment. Attention was drawn to ERCC2 for three reasons. First, from online databases, colorectal cancer (CRC) patients exhibited significantly reduced survival when ERCC2 was overexpressed in colon tumors. Second, ERCC2 was the most highly downregulated RNA transcript in human colon cancer cells, plus Ercc2 in rat tumors, after treatment with the histone deacetylase 3 (HDAC3) inhibitor sulforaphane (SFN) plus JQ1, which is an inhibitor of the bromodomain and extraterminal domain (BET) family. Third, as reported here, RNA-sequencing of polyposis in rat colon (Pirc) polyps following treatment of rats with JQ1 plus 6-methylsulfinylhexyl isothiocyanate (6-SFN) identified Ercc2 as the most highly downregulated gene. The current work also defined promising second-generation epigenetic drug combinations with enhanced synergy and efficacy, especially in metastasis-lineage colon cancer cells cultured as 3D spheroids and xenografts. This investigation adds to the growing interest in combination approaches that target epigenetic 'readers', 'writers', and 'erasers' that are deregulated in cancer and other pathologies, providing new avenues for precision oncology and cancer interception.
Collapse
Affiliation(s)
- Sabeeta Kapoor
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
| | - Trace Gustafson
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
| | - Mutian Zhang
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
| | - Ying-Shiuan Chen
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
| | - Jia Li
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
| | - Nhung Nguyen
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
| | - Jorge Enrique Tovar Perez
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
| | - Wan Mohaiza Dashwood
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
| | - Praveen Rajendran
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
- Antibody & Biopharmaceuticals Core, Texas A&M Health, Houston, TX 77030, USA
- Correspondence: (P.R.); (R.H.D.)
| | - Roderick H. Dashwood
- Center for Epigenetics & Disease Prevention, Texas A&M Health, Department of Translational Medical Sciences, Texas A&M College of Medicine, Houston, TX 77030, USA; (S.K.); (T.G.); (M.Z.); (Y.-S.C.); (J.L.); (N.N.); (J.E.T.P.); (W.M.D.)
- Correspondence: (P.R.); (R.H.D.)
| |
Collapse
|
46
|
Academic collaborative models fostering the translation of physiological in vitro systems from basic research into drug discovery. Drug Discov Today 2021; 26:1369-1381. [PMID: 33677144 DOI: 10.1016/j.drudis.2021.02.024] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 02/15/2021] [Accepted: 02/23/2021] [Indexed: 12/30/2022]
Abstract
The success of preclinical drug discovery strongly relies on the ability of experimental models to resemble human pathophysiology. The number of compounds receiving approval for clinical use is limited, and this has led to the development of more physiologically relevant cellular models aimed at making preclinical results more prone to be successfully translated into clinical use. In this review, we summarize the technologies available in the field of high-throughput screening (HTS) using complex cellular models, and describe collaborative initiatives, such as EU-OPENSCREEN, which can efficiently support researchers to easily access state-of-the-art chemical biology platforms for improving the drug discovery process.
Collapse
|
47
|
Sánchez-Salazar MG, Álvarez MM, Trujillo-de Santiago G. Advances in 3D bioprinting for the biofabrication of tumor models. ACTA ACUST UNITED AC 2021. [DOI: 10.1016/j.bprint.2020.e00120] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
48
|
Soto F, Guimarães CF, Reis RL, Franco W, Rizvi I, Demirci U. Emerging biofabrication approaches for gastrointestinal organoids towards patient specific cancer models. Cancer Lett 2021; 504:116-124. [PMID: 33577978 DOI: 10.1016/j.canlet.2021.01.023] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 01/14/2021] [Accepted: 01/23/2021] [Indexed: 01/12/2023]
Abstract
Tissue engineered organoids are simple biomodels that can emulate the structural and functional complexity of specific organs. Here, we review developments in three-dimensional (3D) artificial cell constructs to model gastrointestinal dynamics towards cancer diagnosis. We describe bottom-up approaches to fabricate close-packed cell aggregates, from the use of biochemical and physical cues to guide the self-assembly of organoids, to the use of engineering approaches, including 3D printing/additive manufacturing and external field-driven protocols. Finally, we outline the main challenges and possible risks regarding the potential translation of gastrointestinal organoids from laboratory settings to patient-specific models in clinical applications.
Collapse
Affiliation(s)
- Fernando Soto
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Department of Radiology, School of Medicine Stanford University, Palo Alto, California, 94304-5427, USA
| | - Carlos F Guimarães
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Department of Radiology, School of Medicine Stanford University, Palo Alto, California, 94304-5427, USA; 3B's Research Group, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's, PT Government Associate Laboratory, University of Minho, Braga/Guimarães, Portugal
| | - Rui L Reis
- 3B's Research Group, Research Institute on Biomaterials, Biodegradables and Biomimetics, University of Minho, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, AvePark, Parque de Ciência e Tecnologia, Zona Industrial da Gandra, 4805-017 Barco, Guimarães, Portugal; ICVS/3B's, PT Government Associate Laboratory, University of Minho, Braga/Guimarães, Portugal
| | - Walfre Franco
- Department of Biomedical Engineering, University of Massachusetts, Lowell, 01854, MA, USA; Wellman Center for Photomedicine, Massachusetts General Hospital, Boston, 02114, MA, USA
| | - Imran Rizvi
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, NC and North Carolina State University, Raleigh, NC, 27599, USA; Lineberger Comprehensive Cancer Center, University of North Carolina School of Medicine, Chapel Hill, NC, 27599, USA
| | - Utkan Demirci
- Canary Center at Stanford for Cancer Early Detection, Bio-Acoustic MEMS in Medicine (BAMM) Laboratory, Department of Radiology, School of Medicine Stanford University, Palo Alto, California, 94304-5427, USA.
| |
Collapse
|
49
|
Abreu TR, Biscaia M, Gonçalves N, Fonseca NA, Moreira JN. In Vitro and In Vivo Tumor Models for the Evaluation of Anticancer Nanoparticles. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1295:271-299. [PMID: 33543464 DOI: 10.1007/978-3-030-58174-9_12] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Multiple studies about tumor biology have revealed the determinant role of the tumor microenvironment in cancer progression, resulting from the dynamic interactions between tumor cells and surrounding stromal cells within the extracellular matrix. This malignant microenvironment highly impacts the efficacy of anticancer nanoparticles by displaying drug resistance mechanisms, as well as intrinsic physical and biochemical barriers, which hamper their intratumoral accumulation and biological activity.Currently, two-dimensional cell cultures are used as the initial screening method in vitro for testing cytotoxic nanocarriers. However, this fails to mimic the tumor heterogeneity, as well as the three-dimensional tumor architecture and pathophysiological barriers, leading to an inaccurate pharmacological evaluation.Biomimetic 3D in vitro tumor models, on the other hand, are emerging as promising tools for more accurately assessing nanoparticle activity, owing to their ability to recapitulate certain features of the tumor microenvironment and thus provide mechanistic insights into nanocarrier intratumoral penetration and diffusion rates.Notwithstanding, in vivo validation of nanomedicines remains irreplaceable at the preclinical stage, and a vast variety of more advanced in vivo tumor models is currently available. Such complex animal models (e.g., genetically engineered mice and patient-derived xenografts) are capable of better predicting nanocarrier clinical efficiency, as they closely resemble the heterogeneity of the human tumor microenvironment.Herein, the development of physiologically more relevant in vitro and in vivo tumor models for the preclinical evaluation of anticancer nanoparticles will be discussed, as well as the current limitations and future challenges in clinical translation.
Collapse
Affiliation(s)
- Teresa R Abreu
- CNC - Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, Coimbra, Portugal.,UC - University of Coimbra, CIBB, Faculty of Pharmacy, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, Portugal
| | - Mariana Biscaia
- CNC - Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, Coimbra, Portugal
| | - Nélio Gonçalves
- CNC - Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, Coimbra, Portugal
| | - Nuno A Fonseca
- CNC - Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, Coimbra, Portugal.,TREAT U, SA, Parque Industrial de Taveiro, Lote 44, Coimbra, Portugal
| | - João Nuno Moreira
- CNC - Center for Neurosciences and Cell Biology, Center for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Faculty of Medicine (Polo 1), Rua Larga, Coimbra, Portugal. .,UC - University of Coimbra, CIBB, Faculty of Pharmacy, Pólo das Ciências da Saúde, Azinhaga de Santa Comba, Coimbra, Portugal.
| |
Collapse
|
50
|
Murakami S, Tanaka H, Nakayama T, Taniura N, Miyake T, Tani M, Kushima R, Yamamoto G, Sugihara H, Mukaisho KI. Similarities and differences in metabolites of tongue cancer cells among two- and three-dimensional cultures and xenografts. Cancer Sci 2020; 112:918-931. [PMID: 33244783 PMCID: PMC7894009 DOI: 10.1111/cas.14749] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Revised: 11/09/2020] [Accepted: 11/16/2020] [Indexed: 12/22/2022] Open
Abstract
Metabolic programming of cancer cells is an essential step in transformation and tumor growth. We established two-dimensional (2D) monolayer and three-dimensional (3D) cultures, the latter called a "tissueoid cell culture system", using four types of tongue cancer cell lines. We also undertook a comprehensive metabolome analysis of three groups that included xenografts created by transplanting the cell lines into nude mice. In addition, we undertook a functional analysis of the mitochondria, which plays a key role in cancer metabolism. Principal component analysis revealed the plots of the four cell lines to be much narrower in 2D culture than in 3D culture and xenograft groups. Moreover, compared to xenografts, the 2D culture had significantly lower levels of most metabolites. These results suggest that the unique characteristics of each cell disappeared in 2D culture, and a type of metabolism unique to monolayer culture took over. Conversely, ATP production, biomass synthesis, and maintenance of redox balance were shown in 3D culture using sufficient nutrients, which closely resembled the metabolic activity in the xenografts. However, there were several differences between the metabolic activity in the 3D culture and xenografts. In vivo, the cancer tissue had blood flow with stromal cells present around the cancer cells. In the xenografts, we detected metabolized and degraded products in the liver and other organs of the host mice. Furthermore, the 3D system did not show impairment of mitochondrial function in the cancer cells, suggesting that cancer cells produce energy simultaneously through mitochondria, as well as aerobic glycolysis.
Collapse
Affiliation(s)
- Shoko Murakami
- Division of Human Pathology, Department of Pathology, Shiga University of Medical Science, Otsu, Japan.,Department of Oral and Maxillofacial Surgery, Shiga University of Medical Science, Otsu, Japan
| | - Hiroyuki Tanaka
- Department of Biochemistry and Molecular Biology, Shiga University of Medical Science, Otsu, Japan
| | - Takahisa Nakayama
- Division of Human Pathology, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Naoko Taniura
- Division of Human Pathology, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Toru Miyake
- Department of Surgery, Shiga University of Medical Science, Otsu, Japan
| | - Masaji Tani
- Department of Surgery, Shiga University of Medical Science, Otsu, Japan
| | - Ryoji Kushima
- Division of Human Pathology, Department of Pathology, Shiga University of Medical Science, Otsu, Japan.,Division of Clinical Laboratory Medicine, Shiga University of Medical Science, Otsu, Japan
| | - Gaku Yamamoto
- Department of Oral and Maxillofacial Surgery, Shiga University of Medical Science, Otsu, Japan
| | - Hiroyuki Sugihara
- Division of Human Pathology, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| | - Ken-Ichi Mukaisho
- Division of Human Pathology, Department of Pathology, Shiga University of Medical Science, Otsu, Japan
| |
Collapse
|