1
|
Lu TY, Ji Y, Lyu C, Shen EN, Sun Y, Xiang Y, Meng-Saccoccio T, Feng GS, Chen S. Bioprinted high cell density liver model with improved hepatic metabolic functions. Biomaterials 2025; 320:123256. [PMID: 40101310 DOI: 10.1016/j.biomaterials.2025.123256] [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: 11/16/2024] [Revised: 02/14/2025] [Accepted: 03/10/2025] [Indexed: 03/20/2025]
Abstract
In vitro liver tissue models are valuable for studying liver function, understanding liver diseases, and screening candidate drugs for toxicity and efficacy. While three-dimensional (3D) bioprinting shows promise in creating various types of functional tissues, current efforts to engineer a functional liver tissue face challenges in replicating native high cell density (HCD) and maintaining long-term cell viability. HCD is crucial for establishing the cell-cell interactions necessary to mimic the liver's metabolic and detoxification functions. However, HCD bioinks exacerbate light scattering in light-based 3D bioprinting. In this study, we incorporated iodixanol into our bioink formulation to minimize light scattering, enabling the fabrication of hepatic tissue constructs with an HCD of 8 × 107 cells/mL while maintaining high cell viability (∼80 %). The printed dense hepatic tissue constructs showed enhanced cell-cell interactions, as evidenced by increased expression of E-cadherin and ZO-1. Furthermore, these constructs promoted albumin secretion, urea production, and P450 metabolic activity. Additionally, HCD hepatic tissue inactivated the YAP/TAZ pathway via cell-cell interactions, preserving primary hepatocyte functions. Further screening revealed that hepatocytes in the dense model were more sensitive to drug treatments than those in a lower-density hepatic model, highlighting the importance of HCD in recapitulating the physiological drug responses. Overall, our approach represents a significant advancement in liver tissue engineering, providing a promising platform for the development of physiologically relevant in vitro liver models for drug screening and toxicity testing.
Collapse
Affiliation(s)
- Ting-Yu Lu
- Program in Materials Science and Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yichun Ji
- Department of Pathology, Department of Molecular Biology, Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Cheng Lyu
- Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Erin Nicole Shen
- Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yazhi Sun
- Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Yi Xiang
- Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Tobias Meng-Saccoccio
- Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, 92093, USA
| | - Gen-Sheng Feng
- Department of Pathology, Department of Molecular Biology, Moores Cancer Center, University of California San Diego, La Jolla, CA, 92093, USA
| | - Shaochen Chen
- Program in Materials Science and Engineering, University of California San Diego, La Jolla, CA, 92093, USA; Department of Chemical and Nano Engineering, University of California San Diego, La Jolla, CA, 92093, USA; Department of Bioengineering, University of California San Diego, La Jolla, CA, 92093, USA.
| |
Collapse
|
2
|
Alnasser SM. From gut to liver: organoids as platforms for next-generation toxicology assessment vehicles for xenobiotics. Stem Cell Res Ther 2025; 16:150. [PMID: 40140938 PMCID: PMC11948905 DOI: 10.1186/s13287-025-04264-y] [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: 11/15/2024] [Accepted: 03/04/2025] [Indexed: 03/28/2025] Open
Abstract
Traditional toxicological assessment relied heavily on 2D cell cultures and animal models of study, which were inadequate for the precise prediction of human response to chemicals. Researchers have now shifted focus on organoids for toxicological assessment. Organoids are 3D structures produced from stem cells that mimic the shape and functionality of human organs and have a number of advantages compared to traditional models of study. They have the capacity to replicate the intricate cellular microenvironment and in vivo interactions. They offer a physiologically pertinent platform that is useful for the researchers to monitor cellular responses in a more realistic manner and evaluate drug toxicity. Additionally, organoids can be created from cells unique to a patient, allowing for individualized toxicological research and providing understanding of the inter-individual heterogeneity in drug responses. Recent developments in the use of gut and liver organoids for assessment of the xenobiotics (environmental toxins and drugs) is reviewed in this article. Gut organoids can reveal potential damage to the digestive system and how xenobiotics affect nutrient absorption and barrier function. Liver is the primary site of detoxification and metabolism of xenobiotics, usually routed from the gut. Hence, these are linked and crucial for evaluating chemical or pollutant induced organ toxicity, forecasting their metabolism and pharmacokinetics. When incorporated into the drug development process, organoid models have the potential to improve the accuracy and efficiency of drug safety assessments, leading to safer and more effective treatments. We also discuss the limitations of using organoid-based toxicological assays, and future prospects, including the need for standardized protocols for overcoming reproducibility issues.
Collapse
Affiliation(s)
- Sulaiman Mohammed Alnasser
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, 51452, Buraydah, Qassim, Saudi Arabia.
| |
Collapse
|
3
|
Nguyen CT, Le VP, Le TH, Kim JS, Back SH, Koo KI. Sacrificing Alginate in Decellularized Extracellular Matrix Scaffolds for Implantable Artificial Livers. J Funct Biomater 2025; 16:35. [PMID: 39852591 PMCID: PMC11766338 DOI: 10.3390/jfb16010035] [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: 11/25/2024] [Revised: 01/11/2025] [Accepted: 01/15/2025] [Indexed: 01/26/2025] Open
Abstract
This research introduced a strategy to fabricate sub-millimeter-diameter artificial liver tissue by extruding a combination of a liver decellularized extracellular matrix (dECM), alginate, endothelial cells, and hepatocytes. Vascularization remains a critical challenge in liver tissue engineering, as replicating the liver's intricate vascular network is essential for sustaining cellular function and viability. Seven scaffold groups were evaluated, incorporating different cell compositions, scaffold materials, and structural configurations. The hepatocyte and endothelial cell scaffold treated with alginate lyase demonstrated the highest diffusion rate, along with enhanced albumin secretion (2.8 µg/mL) and urea synthesis (220 µg/mL) during the same period by day 10. A dense and interconnected endothelial cell network was observed as early as day 4 in the lyased coculture group. Furthermore, three-week implantation studies in rats showed a stable integration to the host with no adverse effects. This approach offers significant potential for advancing functional liver tissue replacements, combining accelerated diffusion, enhanced albumin secretion, improved urea synthesis, dense vascular network formation, and stable implantation outcomes.
Collapse
Affiliation(s)
- Chanh-Trung Nguyen
- Department of Electrical, Electronic and Computer Engineering, University of Ulsan, Ulsan 44610, Republic of Korea; (C.-T.N.)
| | - Van Phu Le
- Department of Electrical, Electronic and Computer Engineering, University of Ulsan, Ulsan 44610, Republic of Korea; (C.-T.N.)
| | - Thi Huong Le
- Department of Electrical, Electronic and Computer Engineering, University of Ulsan, Ulsan 44610, Republic of Korea; (C.-T.N.)
| | - Jeong Sook Kim
- Department of Obstetrics and Gynecology, Ulsan University Hospital, University of Ulsan College of Medicine, Ulsan 44033, Republic of Korea;
| | - Sung Hoon Back
- School of Biological Sciences, University of Ulsan, Ulsan 44610, Republic of Korea
| | - Kyo-in Koo
- Department of Electrical, Electronic and Computer Engineering, University of Ulsan, Ulsan 44610, Republic of Korea; (C.-T.N.)
- Basic-Clinical Convergence Research Institute, University of Ulsan, Ulsan 44610, Republic of Korea
| |
Collapse
|
4
|
Zhang Y, Li L, Dong L, Cheng Y, Huang X, Xue B, Jiang C, Cao Y, Yang J. Hydrogel-Based Strategies for Liver Tissue Engineering. CHEM & BIO ENGINEERING 2024; 1:887-915. [PMID: 39975572 PMCID: PMC11835278 DOI: 10.1021/cbe.4c00079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/15/2024] [Accepted: 09/15/2024] [Indexed: 02/21/2025]
Abstract
The liver's role in metabolism, detoxification, and immune regulation underscores the urgency of addressing liver diseases, which claim millions of lives annually. Due to donor shortages in liver transplantation, liver tissue engineering (LTE) offers a promising alternative. Hydrogels, with their biocompatibility and ability to mimic the liver's extracellular matrix (ECM), support cell survival and function in LTE. This review analyzes recent advances in hydrogel-based strategies for LTE, including decellularized liver tissue hydrogels, natural polymer-based hydrogels, and synthetic polymer-based hydrogels. These materials are ideal for in vitro cell culture and obtaining functional hepatocytes. Hydrogels' tunable properties facilitate creating artificial liver models, such as organoids, 3D bioprinting, and liver-on-a-chip technologies. These developments demonstrate hydrogels' versatility in advancing LTE's applications, including hepatotoxicity testing, liver tissue regeneration, and treating acute liver failure. This review highlights the transformative potential of hydrogels in LTE and their implications for future research and clinical practice.
Collapse
Affiliation(s)
- Yu Zhang
- National
Laboratory of Solid State Microstructures, Department of Physics, Nanjing University, Nanjing 210093, China
- Jinan
Microecological Biomedicine Shandong Laboratory, Jinan 250021, China
| | - Luofei Li
- National
Laboratory of Solid State Microstructures, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Liang Dong
- National
Laboratory of Solid State Microstructures, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Yuanqi Cheng
- National
Laboratory of Solid State Microstructures, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Xiaoyu Huang
- National
Laboratory of Solid State Microstructures, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Bin Xue
- National
Laboratory of Solid State Microstructures, Department of Physics, Nanjing University, Nanjing 210093, China
| | - Chunping Jiang
- Jinan
Microecological Biomedicine Shandong Laboratory, Jinan 250021, China
| | - Yi Cao
- National
Laboratory of Solid State Microstructures, Department of Physics, Nanjing University, Nanjing 210093, China
- Jinan
Microecological Biomedicine Shandong Laboratory, Jinan 250021, China
| | - Jiapeng Yang
- National
Laboratory of Solid State Microstructures, Department of Physics, Nanjing University, Nanjing 210093, China
- Jinan
Microecological Biomedicine Shandong Laboratory, Jinan 250021, China
| |
Collapse
|
5
|
Guo K, van den Beucken T. Advances in drug-induced liver injury research: in vitro models, mechanisms, omics and gene modulation techniques. Cell Biosci 2024; 14:134. [PMID: 39488681 PMCID: PMC11531151 DOI: 10.1186/s13578-024-01317-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Accepted: 10/21/2024] [Indexed: 11/04/2024] Open
Abstract
Drug-induced liver injury (DILI) refers to drug-mediated damage to the structure and function of the liver, ranging from mild elevation of liver enzymes to severe hepatic insufficiency, and in some cases, progressing to liver failure. The mechanisms and clinical symptoms of DILI are diverse due to the varying combination of drugs, making clinical treatment and prevention complex. DILI has significant public health implications and is the primary reason for post-marketing drug withdrawals. The search for reliable preclinical models and validated biomarkers to predict and investigate DILI can contribute to a more comprehensive understanding of adverse effects and drug safety. In this review, we examine the progress of research on DILI, enumerate in vitro models with potential benefits, and highlight cellular molecular perturbations that may serve as biomarkers. Additionally, we discuss omics approaches frequently used to gather comprehensive datasets on molecular events in response to drug exposure. Finally, three commonly used gene modulation techniques are described, highlighting their application in identifying causal relationships in DILI. Altogether, this review provides a thorough overview of ongoing work and approaches in the field of DILI.
Collapse
Affiliation(s)
- Kaidi Guo
- Department of Toxicogenomics, GROW - Research Institute for Oncology & Reproduction, Maastricht University, Maastricht, 6200, MD, The Netherlands.
| | - Twan van den Beucken
- Department of Toxicogenomics, GROW - Research Institute for Oncology & Reproduction, Maastricht University, Maastricht, 6200, MD, The Netherlands
| |
Collapse
|
6
|
Romaldini A, Spanò R, Veronesi M, Grimaldi B, Bandiera T, Sabella S. Human Multi-Lineage Liver Organoid Model Reveals Impairment of CYP3A4 Expression upon Repeated Exposure to Graphene Oxide. Cells 2024; 13:1542. [PMID: 39329726 PMCID: PMC11429598 DOI: 10.3390/cells13181542] [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: 07/08/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/28/2024] Open
Abstract
Three-dimensional hepatic cell cultures can provide an important advancement in the toxicity assessment of nanomaterials with respect to 2D models. Here, we describe liver organoids (LOs) obtained by assembling multiple cell lineages in a fixed ratio 1:1:0.2. These are upcyte® human hepatocytes, UHHs, upcyte® liver sinusoidal endothelial cells, LSECs, and human bone marrow-derived mesenchymal stromal cells, hbmMSCs. The structural and functional analyses indicated that LOs reached size stability upon ca. 10 days of cultivation (organoid maturation), showing a surface area of approximately 10 mm2 and the hepatic cellular lineages, UHHs and LSECs, arranged to form both primitive biliary networks and sinusoid structures, alike in vivo. LOs did not show signs of cellular apoptosis, senescence, or alteration of hepatocellular functions (e.g., dis-regulation of CYP3A4 or aberrant production of Albumin) for the entire culture period (19 days since organoid maturation). After that, LOs were repeatedly exposed for 19 days to a single or repeated dose of graphene oxide (GO: 2-40 µg/mL). We observed that the treatment did not induce any macroscopic signs of tissue damage, apoptosis activation, and alteration of cell viability. However, in the repeated dose regimen, we observed a down-regulation of CYP3A4 gene expression. Notably, these findings are in line with recent in vivo data, which report a similar impact on CYP3A4 when mice were repeatedly exposed to GO. Taken together, these findings warn of the potential detrimental effects of GO in real-life exposure (e.g., occupational scenario), where its progressive accumulation is likely expected. More in general, this study highlights that LOs formed by many cell lineages can enable repeated exposure regimens (suitable to mimic accumulation); thus, they can be suitably considered alternative or complementary in vitro systems to animal models.
Collapse
Affiliation(s)
- Alessio Romaldini
- Nanoregulatory Group, D3 PharmaChemistry, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy; (A.R.); (R.S.)
| | - Raffaele Spanò
- Nanoregulatory Group, D3 PharmaChemistry, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy; (A.R.); (R.S.)
| | - Marina Veronesi
- Structural Biophysics Facility, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy;
- D3 PharmaChemistry, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Benedetto Grimaldi
- Molecular Medicine, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Tiziano Bandiera
- Nanoregulatory Group, D3 PharmaChemistry, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy; (A.R.); (R.S.)
| | - Stefania Sabella
- Nanoregulatory Group, D3 PharmaChemistry, Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy; (A.R.); (R.S.)
| |
Collapse
|
7
|
Park SY, Koh WG, Lee HJ. Enhanced hepatotoxicity assessment through encapsulated HepG2 spheroids in gelatin hydrogel matrices: Bridging the gap from 2D to 3D culture. Eur J Pharm Biopharm 2024; 202:114417. [PMID: 39013493 DOI: 10.1016/j.ejpb.2024.114417] [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: 04/30/2024] [Revised: 07/07/2024] [Accepted: 07/13/2024] [Indexed: 07/18/2024]
Abstract
Conventional 2D drug screening often fails to accurately predict clinical outcomes. We present an innovative approach to improve hepatotoxicity assessment by encapsulating HepG2 spheroids in gelatin hydrogel matrices with different mechanical properties. Encapsulated spheroids exhibit sustained liver-specific functionality, enhanced expression of drug-metabolizing enzymes, and increased drug sensitivity compared to 2D cultures. The platform detects critical variations in drug response, with significant differences in IC50 values between 2D and spheroid cultures ranging from 1.3-fold to > 13-fold, particularly for acetaminophen. Furthermore, drug-metabolizing enzyme expression varies across hydrogel concentrations, suggesting a role for matrix mechanical properties in modulating hepatocyte function. This novel spheroid-hydrogel platform offers a transformative approach to hepatotoxicity assessment, providing increased sensitivity, improved prediction, and a more physiologically relevant environment. The use of such advanced in vitro models can accelerate drug development, reduce animal testing, and contribute to improved patient safety and clinical outcomes.
Collapse
Affiliation(s)
- Se Yeon Park
- School of Chemical, Biological and Battery Engineering, Gachon University, 1342 Seongnam-daero, Seongnam-si, Gyeonggi-do 13120, Republic of Korea
| | - Won-Gun Koh
- Department of Chemical and Biomolecular Engineering, Yonsei University, 50 Yonsei-ro, Seodaemun-gu, Seoul 03722, Republic of Korea.
| | - Hyun Jong Lee
- School of Chemical, Biological and Battery Engineering, Gachon University, 1342 Seongnam-daero, Seongnam-si, Gyeonggi-do 13120, Republic of Korea.
| |
Collapse
|
8
|
Khan MUA, Aslam MA, Abdullah MFB, Gul H, Stojanović GM, Abdal-Hay A, Hasan A. Microneedle system for tissue engineering and regenerative medicines: a smart and efficient therapeutic approach. Biofabrication 2024; 16:042005. [PMID: 39121888 DOI: 10.1088/1758-5090/ad6d90] [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: 04/26/2024] [Accepted: 08/09/2024] [Indexed: 08/12/2024]
Abstract
The global demand for an enhanced quality of life and extended lifespan has driven significant advancements in tissue engineering and regenerative medicine. These fields utilize a range of interdisciplinary theories and techniques to repair structurally impaired or damaged tissues and organs, as well as restore their normal functions. Nevertheless, the clinical efficacy of medications, materials, and potent cells used at the laboratory level is always constrained by technological limitations. A novel platform known as adaptable microneedles has been developed to address the abovementioned issues. These microneedles offer a solution for the localized distribution of various cargos while minimizing invasiveness. Microneedles provide favorable patient compliance in clinical settings due to their effective administration and ability to provide a painless and convenient process. In this review article, we summarized the most recent development of microneedles, and we started by classifying various microneedle systems, advantages, and fundamental properties. Subsequently, it provides a comprehensive overview of different types of microneedles, the material used to fabricate microneedles, the fundamental properties of ideal microneedles, and their applications in tissue engineering and regenerative medicine, primarily focusing on preserving and restoring impaired tissues and organs. The limitations and perspectives have been discussed by concluding their future therapeutic applications in tissue engineering and regenerative medicines.
Collapse
Affiliation(s)
- Muhammad Umar Aslam Khan
- Department of Mechanical and Industrial Engineering, Qatar University, Doha 2713, Qatar
- Biomedical Research Center, Qatar University, Doha 2713, Qatar
| | - Muhammad Azhar Aslam
- Department of Physics, University of Engineering and Technology, Lahore 39161, Pakistan
| | - Mohd Faizal Bin Abdullah
- Oral and Maxillofacial Surgery Unit, School of Dental Sciences, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kota Bharu, Kelantan, Malaysia
- Oral and Maxillofacial Surgery Unit, Hospital Universiti Sains Malaysia, Universiti Sains Malaysia, Health Campus, 16150 Kubang Kerian, Kota Bharu, Kelantan, Malaysia
| | - Hilal Gul
- Department of Biomedical Engineering, Faculty of Engineering, University of Alberta, Edmonton, Alberta, Canada
| | - Goran M Stojanović
- Department of Electronics, Faculty of Technical Sciences, University of Novi Sad, 21000 Novi Sad, Serbia
| | - Abdalla Abdal-Hay
- School of Dentistry, University of Queensland, 288 Herston Road, Herston, QLD 4006, Australia
- Department of Mechanical Engineering, Faculty of Engineering, South Valley University, Qena 83523, Egypt
- Faculty of Industry and Energy Technology, Mechatronics Technology Program, New Cairo Technological University, New Cairo-Fifth Settlement, Cairo 11835, Egypt
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, Qatar University, Doha 2713, Qatar
- Biomedical Research Center, Qatar University, Doha 2713, Qatar
| |
Collapse
|
9
|
Hu W, Bei HP, Jiang H, Wu D, Yu X, Zhou X, Sun Q, Lu Q, Du Q, Wang L, Luo Z, Wu G, Zhao X, Wang S. DLM-GelMA/tumor slice sandwich structured tumor on a chip for drug efficacy testing. LAB ON A CHIP 2024; 24:3718-3727. [PMID: 38953554 DOI: 10.1039/d4lc00278d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/04/2024]
Abstract
The in vitro recapitulation of tumor microenvironment is of great interest to preclinical screening of drugs. Compared with culture of cell lines, tumor organ slices can better preserve the complex tumor architecture and phenotypic activity of native cells, but are limited by their exposure to fluid shear and gradual degradation under perfusion culture. Here, we established a decellularized liver matrix (DLM)-GelMA "sandwich" structure and a perfusion-based microfluidic platform to support long-term culture of tumor slices with excellent structural integrity and cell viability over 7 days. The DLM-GelMA was able to secrete cytokines and growth factors while providing shear protection to the tumor slice via the sandwich structure, leading to the preservation of the tumor microenvironment where immune cells (CD3, CD8, CD68), tumor-associated fibroblasts (α-SMA), and extracellular matrix components (collagen I, fibronectin) were well maintained. Furthermore, this chip presented anti-tumor efficacy at cisplatin (20 μM) on tumor patients, demonstrating our platform's efficacy to design patient-specific treatment regimens. Taken together, the successful development of this DLM-GelMA sandwich structure on the chip could faithfully reflect the tumor microenvironment and immune response, accelerating the screening process of drug molecules and providing insights for practical medicine.
Collapse
Affiliation(s)
- Wenqi Hu
- Department of Respiratory and Critical Care Medicine, Provincial Clinical Research Center for Respiratory Diseases, West China Hospital, Sichuan University, Chengdu, 610065, People's Republic of China.
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
- Tianfu Jincheng Laboratory, City of Future Medicine, Chengdu, 641400, China
| | - Ho-Pan Bei
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, 999077, China.
| | - Hongwei Jiang
- Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China.
| | - Di Wu
- Department of Respiratory and Critical Care Medicine, Provincial Clinical Research Center for Respiratory Diseases, West China Hospital, Sichuan University, Chengdu, 610065, People's Republic of China.
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
- Tianfu Jincheng Laboratory, City of Future Medicine, Chengdu, 641400, China
| | - Xiaorui Yu
- Department of Respiratory and Critical Care Medicine, Provincial Clinical Research Center for Respiratory Diseases, West China Hospital, Sichuan University, Chengdu, 610065, People's Republic of China.
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
- Tianfu Jincheng Laboratory, City of Future Medicine, Chengdu, 641400, China
| | - Xintong Zhou
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, 999077, China.
| | - Qiuwan Sun
- Sichuan Diya BioTechnology Group Company, Chengdu, 641400, China
| | - Qinrui Lu
- Sichuan Diya BioTechnology Group Company, Chengdu, 641400, China
| | - Qijun Du
- Department of Respiratory and Critical Care Medicine, Provincial Clinical Research Center for Respiratory Diseases, West China Hospital, Sichuan University, Chengdu, 610065, People's Republic of China.
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
- Tianfu Jincheng Laboratory, City of Future Medicine, Chengdu, 641400, China
| | - Liangwen Wang
- Department of Interventional Radiology, Zhongshan Hospital, Fudan University, Shanghai, 200032, China
| | - Zhi Luo
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, 518055, China
| | - Guohua Wu
- Henan Key Laboratory of Rare Diseases, Endocrinology and Metabolism Center, The First Affiliated Hospital and College of Clinical Medicine of Henan University of Science and Technology, Luoyang, 471003, China.
| | - Xin Zhao
- Department of Applied Biology and Chemical Technology, The Hong Kong Polytechnic University, Hung Hom, Hong Kong, 999077, China.
- Research Institute for Intelligent Wearable Systems, the Hong Kong Polytechnic University, Hung Hom, Kowloon, Hong Kong, 999077, China
| | - Shuqi Wang
- Department of Respiratory and Critical Care Medicine, Provincial Clinical Research Center for Respiratory Diseases, West China Hospital, Sichuan University, Chengdu, 610065, People's Republic of China.
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, 610065, China
- College of Biomedical Engineering, Sichuan University, Chengdu, 610065, China
- Tianfu Jincheng Laboratory, City of Future Medicine, Chengdu, 641400, China
| |
Collapse
|
10
|
Jadalannagari S, Ewart L. Beyond the hype and toward application: liver complex in vitro models in preclinical drug safety. Expert Opin Drug Metab Toxicol 2024; 20:607-619. [PMID: 38465923 DOI: 10.1080/17425255.2024.2328794] [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: 01/15/2024] [Accepted: 03/06/2024] [Indexed: 03/12/2024]
Abstract
INTRODUCTION Drug induced Liver-Injury (DILI) is a leading cause of drug attrition and complex in vitro models (CIVMs), including three dimensional (3D) spheroids, 3D bio printed tissues and flow-based systems, could improve preclinical prediction. Although CIVMs have demonstrated good sensitivity and specificity in DILI detection their adoption remains limited. AREAS COVERED This article describes DILI, the challenges with its prediction and the current strategies and models that are being used. It reviews data from industry-FDA collaborations and strategic partnerships and finishes with an outlook of CIVMs in preclinical toxicity testing. Literature searches were performed using PubMed and Google Scholar while product information was collected from manufacturer websites. EXPERT OPINION Liver CIVMs are promising models for predicting DILI although, a decade after their introduction, routine use by the pharmaceutical industry is limited. To accelerate their adoption, several industry-regulator-developer partnerships or consortia have been established to guide the development and qualification. Beyond this, liver CIVMs should continue evolving to capture greater immunological mimicry while partnering with computational approaches to deliver systems that change the paradigm of predicting DILI.
Collapse
Affiliation(s)
| | - Lorna Ewart
- Department of Bioinnovations, Emulate Inc, Boston, MA, USA
| |
Collapse
|
11
|
Wang T, Desmet J, Porte C. Protective role of fetal bovine serum on PLHC-1 spheroids exposed to a mixture of plastic additives: A lipidomic perspective. Toxicol In Vitro 2024; 96:105771. [PMID: 38182034 DOI: 10.1016/j.tiv.2024.105771] [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: 07/13/2023] [Revised: 12/20/2023] [Accepted: 01/02/2024] [Indexed: 01/07/2024]
Abstract
The use of fetal bovine serum (FBS) in cell culture is being questioned for scientific and ethical reasons, prompting the exploration of alternative approaches. Nevertheless, the influence of FBS on cell functioning, especially in fish cells, has not been comprehensively examined. This study aims to evaluate the impact of FBS on the lipidome of PLHC-1 spheroids and investigate cellular and molecular responses to plastic additives in the presence/absence of FBS. Lipidomic analyses were conducted on PLHC-1 cell spheroids using liquid chromatography coupled with a high-resolution quadrupole time-of-flight mass spectrometer (HRMS-QToF). The removal of FBS from the culture medium for 24 h significantly changed the lipid profile of spheroids, resulting in a depletion of cholesterol esters (CEs), phosphatidylcholines (PCs) and lyso-phosphatidylcholines (LPCs), while ceramides and certain glycerophospholipids slightly increased. Additionally, the exclusion of FBS from the medium led to increased cytotoxicity caused by a mixture of plastic additives and increased lipidomic alterations, including an elevation of ceramides. This study emphasizes the protective role of serum components in fish liver spheroids against a mixture of plastic additives and underscores the importance of considering exposure conditions when studying metabolomic and lipidomic responses to toxicants.
Collapse
Affiliation(s)
- Tiantian Wang
- Environmental Chemistry Department, IDAEA -CSIC-, C/ Jordi Girona, 18-26, 08034 Barcelona, Spain; PhD student at the University of Barcelona, 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
|
12
|
De S, Chauhan R, Singh M, Singh N. Ubiquitin specific peptidase (USP37) mediated effects in microscaffold-encapsulated cells: a comprehensive study on growth, proliferation and EMT. RSC Adv 2024; 14:5461-5471. [PMID: 38352690 PMCID: PMC10862100 DOI: 10.1039/d3ra08786g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2023] [Accepted: 01/14/2024] [Indexed: 02/16/2024] Open
Abstract
Though significant advances have been made in developing therapeutic strategies for cancer, suitable in vitro models for mechanistically identifying relevant drug targets and understanding disease progression are still lacking. Most studies are generally performed using two-dimensional (2D) models, since these models can be readily established and allow high throughput assays. However, these models have also been reported as the reason for unreliable pre-clinical information. To avoid this discrepancy, three-dimensional (3D) cell culture models have been established and have demonstrated the potential to provide alternative ways to study tissue behavior. However, most of these models first require optimization and cell cultures with a certain density, thus adding a prepping step in the platform before it can be used for any studies. This limits their use in studies where the fundamental understanding of biological processes must be carried out in a short time frame. In this study, we developed a 3D cell culture system that tests a less explored cancer therapeutic target-the deubiquitinating enzyme ubiquitin specific peptidase 37 (USP37)-in different cancer cell lines using sensitive carbon dot pH nanosensors, which provides a rapid model for studies compared to the parallel model available commercially. This enzyme is found to be elevated in different cancers and has been reported to play a role in cell cycle regulation, oncogenesis and metastasis. However, the confirmation of the role of USP37 downregulation in cellular proliferation via appropriate in vitro 3D models has not been demonstrated. To establish the applicability of the developed 3D platform in studying such oncogenes, classical 2D models have been used in this study for identifying the role of USP37 in tumor progression and metastasis. The data clearly suggests that this ingeniously developed 3D cell culture system is a better alternative to 2D models to study the growth and migration of different cancer cell lines on depletion of oncogenic proteins like USP37 and its effect on epithelial-mesenchymal transition (EMT) markers, and it can further be targeted as a viable therapeutic option.
Collapse
Affiliation(s)
- Shreemoyee De
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi Hauz Khas New Delhi 110016 India
| | - Ravi Chauhan
- Department of Medical Oncology (Lab), All India Institute of Medical Sciences New Delhi India
| | - Mayank Singh
- Department of Medical Oncology (Lab), All India Institute of Medical Sciences New Delhi India
| | - Neetu Singh
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi Hauz Khas New Delhi 110016 India
- Biomedical Engineering Unit, All India Institute of Medical Sciences Ansari Nagar New Delhi 110029 India
| |
Collapse
|
13
|
Ali ASM, Berg J, Roehrs V, Wu D, Hackethal J, Braeuning A, Woelken L, Rauh C, Kurreck J. Xeno-Free 3D Bioprinted Liver Model for Hepatotoxicity Assessment. Int J Mol Sci 2024; 25:1811. [PMID: 38339088 PMCID: PMC10855587 DOI: 10.3390/ijms25031811] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2023] [Revised: 01/26/2024] [Accepted: 01/30/2024] [Indexed: 02/12/2024] Open
Abstract
Three-dimensional (3D) bioprinting is one of the most promising methodologies that are currently in development for the replacement of animal experiments. Bioprinting and most alternative technologies rely on animal-derived materials, which compromises the intent of animal welfare and results in the generation of chimeric systems of limited value. The current study therefore presents the first bioprinted liver model that is entirely void of animal-derived constituents. Initially, HuH-7 cells underwent adaptation to a chemically defined medium (CDM). The adapted cells exhibited high survival rates (85-92%) after cryopreservation in chemically defined freezing media, comparable to those preserved in standard medium (86-92%). Xeno-free bioink for 3D bioprinting yielded liver models with high relative cell viability (97-101%), akin to a Matrigel-based liver model (83-102%) after 15 days of culture. The established xeno-free model was used for toxicity testing of a marine biotoxin, okadaic acid (OA). In 2D culture, OA toxicity was virtually identical for cells cultured under standard conditions and in CDM. In the xeno-free bioprinted liver model, 3-fold higher concentrations of OA than in the respective monolayer culture were needed to induce cytotoxicity. In conclusion, this study describes for the first time the development of a xeno-free 3D bioprinted liver model and its applicability for research purposes.
Collapse
Affiliation(s)
- Ahmed S. M. Ali
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, TIB 4/3-2, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Johanna Berg
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, TIB 4/3-2, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Viola Roehrs
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, TIB 4/3-2, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | - Dongwei Wu
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, TIB 4/3-2, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| | | | - Albert Braeuning
- Department Food Safety, German Federal Institute for Risk Assessment (BfR), 10589 Berlin, Germany;
| | - Lisa Woelken
- Department of Food Biotechnology and Food Process Engineering, Technische Universität Berlin, 14195 Berlin, Germany (C.R.)
| | - Cornelia Rauh
- Department of Food Biotechnology and Food Process Engineering, Technische Universität Berlin, 14195 Berlin, Germany (C.R.)
| | - Jens Kurreck
- Department of Applied Biochemistry, Institute of Biotechnology, Technische Universität Berlin, TIB 4/3-2, Gustav-Meyer-Allee 25, 13355 Berlin, Germany
| |
Collapse
|
14
|
De S, Vasudevan A, Tripathi DM, Kaur S, Singh N. A decellularized matrix enriched collagen microscaffold for a 3D in vitro liver model. J Mater Chem B 2024; 12:772-783. [PMID: 38167699 DOI: 10.1039/d3tb01652h] [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: 01/05/2024]
Abstract
The development of liver scaffolds retaining their three-dimensional (3D) structure and extra-cellular matrix (ECM) composition is essential for the advancement of liver tissue engineering. We report the design and validation of an alginate-based platform using a combination of decellularized matrices and collagen to preserve the functionality of liver cells. The scaffolds were characterized using SEM and fluorescence microscopy techniques. The proliferation and functional behaviours of hepatocellular carcinoma HuH7 cells were observed. It was found that the decellularized skin scaffold with collagen was better for maintaining the growth of cells in comparison to other decellularized matrices. In addition, we observed a significant increase in the functional profile once exogenous collagen was added to the liver matrix. Our study also suggests that a cirrhotic liver model should have a different matrix composition as compared to a healthy liver model. When primary rat hepatocytes were used for developing a healthy liver model, the proliferation studies with hepatocytes showed a decellularized skin matrix as the better option, but the functionality was only maintained in a decellularized liver matrix with addition of exogenous collagen. We further checked if these platforms can be used for studying drug induced toxicity observed in the liver by studying the activation of cytochrome P450 upon drug exposure of the cells growing in our model. We observed a significant induction of the CYP1A1 gene on administering the drugs for 6 days. Thus, this platform could be used for drug-toxicity screening studies using primary hepatocytes in a short span of time. Being a microscaffold based system, this platform offers some advantages, such as smaller volumes of samples, analysing multiple samples simultaneously and a minimal amount of decellularized matrix in the matrix composition, making it an economical option compared to a completely dECM based platform.
Collapse
Affiliation(s)
- Shreemoyee De
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi-110016, India.
| | - Ashwini Vasudevan
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, D1, Vasant Kunj Marg, New Delhi, Delhi 110070, India.
| | - Dinesh M Tripathi
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, D1, Vasant Kunj Marg, New Delhi, Delhi 110070, India.
| | - Savneet Kaur
- Department of Molecular and Cellular Medicine, Institute of Liver and Biliary Sciences, D1, Vasant Kunj Marg, New Delhi, Delhi 110070, India.
| | - Neetu Singh
- Centre for Biomedical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi-110016, India.
- Biomedical Engineering Unit, All India Institute of Medical Sciences, Ansari Nagar, New Delhi-110029, India
| |
Collapse
|
15
|
Carvalho AM, Bansal R, Barrias CC, Sarmento B. The Material World of 3D-Bioprinted and Microfluidic-Chip Models of Human Liver Fibrosis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307673. [PMID: 37961933 DOI: 10.1002/adma.202307673] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/06/2023] [Indexed: 11/15/2023]
Abstract
Biomaterials are extensively used to mimic cell-matrix interactions, which are essential for cell growth, function, and differentiation. This is particularly relevant when developing in vitro disease models of organs rich in extracellular matrix, like the liver. Liver disease involves a chronic wound-healing response with formation of scar tissue known as fibrosis. At early stages, liver disease can be reverted, but as disease progresses, reversion is no longer possible, and there is no cure. Research for new therapies is hampered by the lack of adequate models that replicate the mechanical properties and biochemical stimuli present in the fibrotic liver. Fibrosis is associated with changes in the composition of the extracellular matrix that directly influence cell behavior. Biomaterials could play an essential role in better emulating the disease microenvironment. In this paper, the recent and cutting-edge biomaterials used for creating in vitro models of human liver fibrosis are revised, in combination with cells, bioprinting, and/or microfluidics. These technologies have been instrumental to replicate the intricate structure of the unhealthy tissue and promote medium perfusion that improves cell growth and function, respectively. A comprehensive analysis of the impact of material hints and cell-material interactions in a tridimensional context is provided.
Collapse
Affiliation(s)
- Ana Margarida Carvalho
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Ruchi Bansal
- Translational Liver Research, Department of Medical Cell Biophysics, Technical Medical Center, Faculty of Science and Technology, University of Twente, Enschede, 7522 NB, The Netherlands
| | - Cristina C Barrias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Rua de Jorge Viterbo Ferreira 228, Porto, 4050-313, Portugal
| | - Bruno Sarmento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Rua Alfredo Allen 208, Porto, 4200-135, Portugal
- IUCS - Instituto Universitário de Ciências da Saúde, CESPU, Rua Central de Gandra 1317, Gandra, 4585-116, Portugal
| |
Collapse
|
16
|
Aghajanshakeri S, Salmanmahiny A, Aghajanshakeri S, Babaei A, Alishahi F, Babayani E, Shokrzadeh M. Modulatory effect of amifostine (WR-1065) against genotoxicity and oxidative stress induced by methotrexate in human umbilical vein endothelial cells (HUVECs). Toxicol Mech Methods 2023; 33:755-765. [PMID: 37537746 DOI: 10.1080/15376516.2023.2238069] [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: 05/31/2023] [Accepted: 07/11/2023] [Indexed: 08/05/2023]
Abstract
Amifostine is used in chemotherapy and radiotherapy as a cytoprotective adjuvant alongside DNA-binding chemotherapeutic agents. It functions by reducing free radicals and detoxifying harmful metabolites. Methotrexate, as an antimetabolite drug has been considered for treating various cancers and autoimmune diseases. However, the cytotoxic effects of methotrexate extend beyond tumor cells to crucial organs, including the heart. This study applied the HUVEC cell line as a reference in vitro model for researching the characteristics of vascular endothelium and cardiotoxicity. The current study aimed to assess amifostine's potential cytoprotective properties against methotrexate-induced cellular damage. Cytotoxicity was measured using the MTT assay. Apoptotic rates were evaluated by Annexin V-FITC/PI staining via flow cytometry. The genoprotective effect of amifostine was determined using the comet assay. Cells were exposed to various amifostine doses (10-200 μg/mL) and methotrexate (2.5 μM) in pretreatment culture condition. Methotrexate at 2.5 μM revealed cytotoxicity, apoptosis, oxidative stress and genotoxicity while highlighting amifostine's cyto/geno protective properties on HUVECs. Amifostine significantly decreased the levels of ROS and LPO while preserving the status of GSH and SOD activity. Furthermore, it inhibited genotoxicity (tail length, %DNA in tail, and tail moment) in the comet assay. Amifostine markedly attenuated methotrexate-induced apoptotic cell death (early and late apoptotic rates). These findings convey that amifostine can operate as a cytoprotectant agent.
Collapse
Affiliation(s)
- Shaghayegh Aghajanshakeri
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Amirhossein Salmanmahiny
- Department of Toxicology and Pharmacology, Student Research Committee, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Shahin Aghajanshakeri
- Biological Oncology (Orchid Pharmed) Department, CinnaGen Pharmaceutical Company, Tehran, Iran
| | - Amirhossein Babaei
- Department of Pharmaceutics, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Farhad Alishahi
- Department of Toxicology and Pharmacology, Student Research Committee, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Erfan Babayani
- Department of Toxicology and Pharmacology, Student Research Committee, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| | - Mohammad Shokrzadeh
- Department of Toxicology and Pharmacology, Faculty of Pharmacy, Mazandaran University of Medical Sciences, Sari, Iran
| |
Collapse
|
17
|
Li M, Wang L, Du J. Clinical observation of liposomal doxorubicin on liver and renal function in patients with breast cancer. Toxicol Res (Camb) 2023; 12:807-813. [PMID: 37915489 PMCID: PMC10615824 DOI: 10.1093/toxres/tfad072] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Revised: 07/18/2023] [Accepted: 08/08/2023] [Indexed: 11/03/2023] Open
Abstract
Background Doxorubicin has become the first-line antitumor drug clinically, but severely limited by multiple side effects, especially cardiotoxicity. Liposomal doxorubicin therefore replaced traditional doxorubicin for low toxicity and high efficiency. Previous studies have suggested liver and kidney may be the main organs affected by liposomal doxorubicin. Due to insufficient clinical evidence, we set out to analyze the effect of liposomal doxorubicin on liver and renal function in breast cancer patients. Materials and Methods Our retrospective analysis included breast cancer patients aged 30-70 years old who were assigned to two groups based on liposomal doxorubicin intake. We evaluated changes in liver and renal function. Multivariate logistic regression model was used to assess the risk factors of liver function damage. Results Ultimately, 631 patients for liver function analysis cohort and 611 cases for renal function analysis cohort. Patients receiving liposomal doxorubicin had significantly higher liver function damage rate compared to control group (52.20% vs 9.82%, p < 0.001), but there was no difference in the incidence of renal damage events between the two groups. Multivariate analysis shows total doses divided by body surface area is a significant, independent risk factor for liver function damage (odds ratio 1.005 [1.002-1.018], p < 0.001). Conclusion Liposomal doxorubicin treatment is associated with higher liver function damage in breast cancer patients, but has no effect on renal function. Together with risk factor analysis, our study underlines the importance to pay attention for patient's age before taking liposomal doxorubicin, alongside liver function after the first and long-term treatments.
Collapse
Affiliation(s)
- Mingliang Li
- Department of Urology, The Third Hospital of Changsha, No. 176, Labor West Road, Tianxin District, Changsha, Hunan 410035, China
| | - Ling Wang
- Department of Pharmacy, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Kaifu District, Changsha, Hunan 410008, China
| | - Jie Du
- Department of Pharmacy, Xiangya Hospital, Central South University, No. 87, Xiangya Road, Kaifu District, Changsha, Hunan 410008, China
| |
Collapse
|
18
|
Harbuz I, Banciu DD, David R, Cercel C, Cotîrță O, Ciurea BM, Radu SM, Dinescu S, Jinga SI, Banciu A. Perspectives on Scaffold Designs with Roles in Liver Cell Asymmetry and Medical and Industrial Applications by Using a New Type of Specialized 3D Bioprinter. Int J Mol Sci 2023; 24:14722. [PMID: 37834167 PMCID: PMC10573170 DOI: 10.3390/ijms241914722] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 09/18/2023] [Accepted: 09/26/2023] [Indexed: 10/15/2023] Open
Abstract
Cellular asymmetry is an important element of efficiency in the compartmentalization of intracellular chemical reactions that ensure efficient tissue function. Improving the current 3D printing methods by using cellular asymmetry is essential in producing complex tissues and organs such as the liver. The use of cell spots containing at least two cells and basement membrane-like bio support materials allows cells to be tethered at two points on the basement membrane and with another cell in order to maintain cell asymmetry. Our model is a new type of 3D bioprinter that uses oriented multicellular complexes with cellular asymmetry. This novel approach is necessary to replace the sequential and slow processes of organogenesis with rapid methods of growth and 3D organ printing. The use of the extracellular matrix in the process of bioprinting with cells allows one to preserve the cellular asymmetry in the 3D printing process and thus preserve the compartmentalization of biological processes and metabolic efficiency.
Collapse
Affiliation(s)
- Iuliana Harbuz
- Department of Biomaterials and Medical Devices, Faculty of Medical Engineering, Politehnica University of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania; (I.H.); (O.C.); (B.M.C.); (S.I.J.)
| | - Daniel Dumitru Banciu
- Department of Biomaterials and Medical Devices, Faculty of Medical Engineering, Politehnica University of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania; (I.H.); (O.C.); (B.M.C.); (S.I.J.)
| | - Rodica David
- Institute for Research on the Quality of Society and the Sciences of Education, University Constantin Brancusi of Targu Jiu, Republicii 1, 210185 Targu Jiu, Romania;
- Department of Mechanical Industrial and Transportation Engineering, University of Petrosani, 332006 Petrosani, Romania; (S.M.R.); (S.D.)
| | - Cristina Cercel
- University of Medicine and Pharmacy “Carol Davila” Bucharest, 37 Dionisie Lupu Street, 020021 Bucharest, Romania;
| | - Octavian Cotîrță
- Department of Biomaterials and Medical Devices, Faculty of Medical Engineering, Politehnica University of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania; (I.H.); (O.C.); (B.M.C.); (S.I.J.)
| | - Bogdan Marius Ciurea
- Department of Biomaterials and Medical Devices, Faculty of Medical Engineering, Politehnica University of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania; (I.H.); (O.C.); (B.M.C.); (S.I.J.)
| | - Sorin Mihai Radu
- Department of Mechanical Industrial and Transportation Engineering, University of Petrosani, 332006 Petrosani, Romania; (S.M.R.); (S.D.)
| | - Stela Dinescu
- Department of Mechanical Industrial and Transportation Engineering, University of Petrosani, 332006 Petrosani, Romania; (S.M.R.); (S.D.)
| | - Sorin Ion Jinga
- Department of Biomaterials and Medical Devices, Faculty of Medical Engineering, Politehnica University of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania; (I.H.); (O.C.); (B.M.C.); (S.I.J.)
| | - Adela Banciu
- Department of Biomaterials and Medical Devices, Faculty of Medical Engineering, Politehnica University of Bucharest, 1-7 Gh. Polizu Street, 011061 Bucharest, Romania; (I.H.); (O.C.); (B.M.C.); (S.I.J.)
| |
Collapse
|
19
|
Quintás G, Castell JV, Moreno-Torres M. The assessment of the potential hepatotoxicity of new drugs by in vitro metabolomics. Front Pharmacol 2023; 14:1155271. [PMID: 37214440 PMCID: PMC10196061 DOI: 10.3389/fphar.2023.1155271] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 04/19/2023] [Indexed: 05/24/2023] Open
Abstract
Drug hepatotoxicity assessment is a relevant issue both in the course of drug development as well as in the post marketing phase. The use of human relevant in vitro models in combination with powerful analytical methods (metabolomic analysis) is a promising approach to anticipate, as well as to understand and investigate the effects and mechanisms of drug hepatotoxicity in man. The metabolic profile analysis of biological liver models treated with hepatotoxins, as compared to that of those treated with non-hepatotoxic compounds, provides useful information for identifying disturbed cellular metabolic reactions, pathways, and networks. This can later be used to anticipate, as well to assess, the potential hepatotoxicity of new compounds. However, the applicability of the metabolomic analysis to assess the hepatotoxicity of drugs is complex and requires careful and systematic work, precise controls, wise data preprocessing and appropriate biological interpretation to make meaningful interpretations and/or predictions of drug hepatotoxicity. This review provides an updated look at recent in vitro studies which used principally mass spectrometry-based metabolomics to evaluate the hepatotoxicity of drugs. It also analyzes the principal drawbacks that still limit its general applicability in safety assessment screenings. We discuss the analytical workflow, essential factors that need to be considered and suggestions to overcome these drawbacks, as well as recent advancements made in this rapidly growing field of research.
Collapse
Affiliation(s)
- Guillermo Quintás
- Metabolomics and Bioanalysis, Health and Biomedicine, Leitat Technological Center, Barcelona, Spain
- Analytical Unit, Health Research Institute La Fe, Valencia, Spain
| | - José V. Castell
- Unidad Mixta de Hepatología Experimental, Instituto de Investigación Sanitaria del Hospital La Fe (IIS La Fe), Valencia, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
- CIBEREHD, Instituto de Salud Carlos III, Madrid, Spain
| | - Marta Moreno-Torres
- Unidad Mixta de Hepatología Experimental, Instituto de Investigación Sanitaria del Hospital La Fe (IIS La Fe), Valencia, Spain
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad de Valencia, Valencia, Spain
- CIBEREHD, Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
20
|
Boggio E, Gigliotti CL, Stoppa I, Pantham D, Sacchetti S, Rolla R, Grattarola M, Monge C, Pizzimenti S, Dianzani U, Dianzani C, Battaglia L. Exploiting Nanomedicine for Cancer Polychemotherapy: Recent Advances and Clinical Applications. Pharmaceutics 2023; 15:937. [PMID: 36986798 PMCID: PMC10057931 DOI: 10.3390/pharmaceutics15030937] [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: 01/20/2023] [Revised: 03/07/2023] [Accepted: 03/10/2023] [Indexed: 03/18/2023] Open
Abstract
The most important limitations of chemotherapeutic agents are severe side effects and the development of multi-drug resistance. Recently, the clinical successes achieved with immunotherapy have revolutionized the treatment of several advanced-stage malignancies, but most patients do not respond and many of them develop immune-related adverse events. Loading synergistic combinations of different anti-tumor drugs in nanocarriers may enhance their efficacy and reduce life-threatening toxicities. Thereafter, nanomedicines may synergize with pharmacological, immunological, and physical combined treatments, and should be increasingly integrated in multimodal combination therapy regimens. The goal of this manuscript is to provide better understanding and key considerations for developing new combined nanomedicines and nanotheranostics. We will clarify the potential of combined nanomedicine strategies that are designed to target different steps of the cancer growth as well as its microenvironment and immunity interactions. Moreover, we will describe relevant experiments in animal models and discuss issues raised by translation in the human setting.
Collapse
Affiliation(s)
- Elena Boggio
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Casimiro Luca Gigliotti
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Ian Stoppa
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Deepika Pantham
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale, 28100 Novara, Italy
| | - Sara Sacchetti
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale, 28100 Novara, Italy
- Ospedale Universitario Maggiore della Carità, 28100 Novara, Italy
| | - Roberta Rolla
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale, 28100 Novara, Italy
- Ospedale Universitario Maggiore della Carità, 28100 Novara, Italy
| | - Margherita Grattarola
- Dipartimento di Scienze Cliniche e Biologiche, Università degli Studi di Torino, Corso Raffaello 30, 10125 Torino, Italy
| | - Chiara Monge
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, 10125 Torino, Italy
| | - Stefania Pizzimenti
- Dipartimento di Scienze Cliniche e Biologiche, Università degli Studi di Torino, Corso Raffaello 30, 10125 Torino, Italy
| | - Umberto Dianzani
- Dipartimento di Scienze della Salute, Università del Piemonte Orientale, 28100 Novara, Italy
- Ospedale Universitario Maggiore della Carità, 28100 Novara, Italy
| | - Chiara Dianzani
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, 10125 Torino, Italy
| | - Luigi Battaglia
- Dipartimento di Scienza e Tecnologia del Farmaco, Università degli Studi di Torino, 10125 Torino, Italy
- Centro Interdipartimentale Nanostructured Interfaces and Surfaces (NIS) Interdepartmental Centre, Università degli Studi di Torino, 10124 Torino, Italy
| |
Collapse
|
21
|
Shnayder NA, Grechkina VV, Khasanova AK, Bochanova EN, Dontceva EA, Petrova MM, Asadullin AR, Shipulin GA, Altynbekov KS, Al-Zamil M, Nasyrova RF. Therapeutic and Toxic Effects of Valproic Acid Metabolites. Metabolites 2023; 13:metabo13010134. [PMID: 36677060 PMCID: PMC9862929 DOI: 10.3390/metabo13010134] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/26/2022] [Revised: 01/11/2023] [Accepted: 01/12/2023] [Indexed: 01/18/2023] Open
Abstract
Valproic acid (VPA) and its salts are psychotropic drugs that are widely used in neurological diseases (epilepsy, neuropathic pain, migraine, etc.) and psychiatric disorders (schizophrenia, bipolar affective disorder, addiction diseases, etc.). In addition, the indications for the appointment of valproate have been expanding in recent years in connection with the study of new mechanisms of action of therapeutic and toxic metabolites of VPA in the human body. Thus, VPA is considered a component of disease-modifying therapy for multiple tumors, neurodegenerative diseases (Huntington's disease, Parkinson's disease, Duchenne progressive dystrophy, etc.), and human immunodeficiency syndrome. The metabolism of VPA is complex and continues to be studied. Known pathways of VPA metabolism include: β-oxidation in the tricarboxylic acid cycle (acetylation); oxidation with the participation of cytochrome P-450 isoenzymes (P-oxidation); and glucuronidation. The complex metabolism of VPA explains the diversity of its active and inactive metabolites, which have therapeutic, neutral, or toxic effects. It is known that some active metabolites of VPA may have a stronger clinical effect than VPA itself. These reasons explain the relevance of this narrative review, which summarizes the results of studies of blood (serum, plasma) and urinary metabolites of VPA from the standpoint of the pharmacogenomics and pharmacometabolomics. In addition, a new personalized approach to assessing the cumulative risk of developing VPA-induced adverse reactions is presented and ways for their correction are proposed depending on the patient's pharmacogenetic profile and the level of therapeutic and toxic VPA metabolites in the human body fluids (blood, urine).
Collapse
Affiliation(s)
- Natalia A. Shnayder
- Institute of Personalized Psychiatry and Neurology, Shared Core Facilities, V.M. Bekhterev National Medical Research Centre for Psychiatry and Neurology, 192019 Saint Petersburg, Russia
- Shared Core Facilities “Molecular and Cell Technologies”, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
- Correspondence: (N.A.S.); (R.F.N.); Tel.: +7-(812)-620-0222 (N.A.S. & R.F.N.)
| | - Violetta V. Grechkina
- Institute of Personalized Psychiatry and Neurology, Shared Core Facilities, V.M. Bekhterev National Medical Research Centre for Psychiatry and Neurology, 192019 Saint Petersburg, Russia
| | - Aiperi K. Khasanova
- Department of Psychiatry, Russian Medical Academy for Continual Professional Education, 125993 Moscow, Russia
| | - Elena N. Bochanova
- Shared Core Facilities “Molecular and Cell Technologies”, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
| | - Evgenia A. Dontceva
- Shared Core Facilities “Molecular and Cell Technologies”, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
| | - Marina M. Petrova
- Shared Core Facilities “Molecular and Cell Technologies”, V.F. Voino-Yasenetsky Krasnoyarsk State Medical University, 660022 Krasnoyarsk, Russia
| | - Azat R. Asadullin
- Department of Psychiatry and Addiction, Bashkir State Medical University, 45000 Ufa, Russia
| | - German A. Shipulin
- Centre for Strategic Planning and Management of Biomedical Health Risks, 119121 Moscow, Russia
| | - Kuanysh S. Altynbekov
- Republican Scientific and Practical Center of Mental Health, Almaty 050022, Kazakhstan
- Department of Psychiatry and Narcology, S.D. Asfendiarov Kazakh National Medical University, Almaty 050022, Kazakhstan
| | - Mustafa Al-Zamil
- Department of Physiotherapy, Faculty of Continuing Medical Education, Peoples’ Friendship University of Russia, 11798 Moscow, Russia
| | - Regina F. Nasyrova
- Institute of Personalized Psychiatry and Neurology, Shared Core Facilities, V.M. Bekhterev National Medical Research Centre for Psychiatry and Neurology, 192019 Saint Petersburg, Russia
- Correspondence: (N.A.S.); (R.F.N.); Tel.: +7-(812)-620-0222 (N.A.S. & R.F.N.)
| |
Collapse
|
22
|
Sreedharan S, Zouganelis G, Drake SJ, Tripathi G, Kermanizadeh A. Nanomaterial-induced toxicity in pathophysiological models representative of individuals with pre-existing medical conditions. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2023; 26:1-27. [PMID: 36474307 DOI: 10.1080/10937404.2022.2153456] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
The integration of nanomaterials (NMs) into an ever-expanding number of daily used products has proven to be highly desirable in numerous industries and applications. Unfortunately, the same "nano" specific physicochemical properties, which make these materials attractive, may also contribute to hazards for individuals exposed to these materials. In 2021, it was estimated that 7 out of 10 deaths globally were accredited to chronic diseases, such as chronic liver disease, asthma, and cardiovascular-related illnesses. Crucially, it is also understood that a significant proportion of global populace numbering in the billions are currently living with a range of chronic undiagnosed health conditions. Due to the significant number of individuals affected, it is important that people suffering from chronic disease also be considered and incorporated in NM hazard assessment strategies. This review examined and analyzed the literature that focused on NM-induced adverse health effects in models which are representative of individuals exhibiting pre-existing medical conditions with focus on the pulmonary, cardiovascular, hepatic, gastrointestinal, and central nervous systems. The overall objective of this review was to outline available data, highlighting the important role of pre-existing disease in NM-induced toxicity with the aim of establishing a weight of evidence approach to inform the public on the potential hazards posed by NMs in both healthy and compromised persons in general population.
Collapse
|
23
|
Tutty MA, Prina-Mello A. Three-Dimensional Spheroids for Cancer Research. Methods Mol Biol 2023; 2645:65-103. [PMID: 37202612 DOI: 10.1007/978-1-0716-3056-3_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/20/2023]
Abstract
In vitro cell culture is one of the most widely used tools used today for increasing our understanding of various things such as protein production, mechanisms of drug action, tissue engineering, and overall cellular biology. For the past decades, however, cancer researchers have relied heavily on conventional two-dimensional (2D) monolayer culture techniques to test a variety of aspects of cancer research ranging from the cytotoxic effects of antitumor drugs to the toxicity of diagnostic dyes and contact tracers. However, many promising cancer therapies have either weak or no efficacy in real-life conditions, therefore delaying or stopping altogether their translating to the clinic. This is, in part, due to the reductionist 2D cultures used to test these materials, which lack appropriate cell-cell contacts, have altered signaling, do not represent the natural tumor microenvironment, and have different drug responses, due to their reduced malignant phenotype when compared to real in vivo tumors. With the most recent advances, cancer research has moved into 3D biological investigation. Three-dimensional (3D) cultures of cancer cells not only recapitulate the in vivo environment better than their 2D counterparts, but they have, in recent years, emerged as a relatively low-cost and scientifically accurate methodology for studying cancer. In this chapter, we highlight the importance of 3D culture, specifically 3D spheroid culture, reviewing some key methodologies for forming 3D spheroids, discussing the experimental tools that can be used in conjunction with 3D spheroids and finally their applications in cancer research.
Collapse
Affiliation(s)
- Melissa Anne Tutty
- Laboratory for Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, Dublin, Ireland.
| | - Adriele Prina-Mello
- Laboratory for Biological Characterization of Advanced Materials (LBCAM), Trinity Translational Medicine Institute, Trinity Centre for Health Sciences, Trinity College Dublin, Dublin, Ireland
- Nanomedicine and Molecular Imaging Group, Trinity Translational Medicine Institute, (TTMI), School of Medicine, Trinity College Dublin, Dublin, Ireland
- Trinity St. James's Cancer Institute, St. James's Hospital, Trinity College Dublin, Dublin, Ireland
- Advanced Materials and Bioengineering Research (AMBER) Centre, CRANN Institute, Trinity College Dublin, Dublin, Ireland
| |
Collapse
|
24
|
Hyper-Branched Cationic Cyclodextrin Polymers for Improving Plasmid Transfection in 2D and 3D Spheroid Cells. Pharmaceutics 2022; 14:pharmaceutics14122690. [PMID: 36559184 PMCID: PMC9785855 DOI: 10.3390/pharmaceutics14122690] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2022] [Revised: 11/25/2022] [Accepted: 11/29/2022] [Indexed: 12/04/2022] Open
Abstract
In this article, we used monolayer two dimensional (2D) and 3D multicellular spheroid models to improve our understanding of the gene delivery process of a new modified cationic hyper-branched cyclodextrin-based polymer (Ppoly)-loaded plasmid encoding Enhanced Green Fluorescent Protein (EGFP). A comparison between the cytotoxicity effect and transfection efficiency of the plasmid DNA (pDNA)-loaded Ppoly system in 2D and 3D spheroid cells determined that the transfection efficiency and cytotoxicity of Ppoly-pDNA nanocomplexes were lower in 3D spheroids than in 2D monolayer cells. Furthermore, histopathology visualization of Ppoly-pDNA complex cellular uptake in 3D spheroids demonstrated that Ppoly penetrated into the inner layers. This study indicated that the Ppoly, as a non-viral gene delivery system in complex with pDNA, is hemocompatible, non-toxic, high in encapsulation efficiency, and has good transfection efficiency in both 2D and 3D cell cultures compared to free pDNA and lipofectamine (as the control).
Collapse
|
25
|
REFINE special issue. Drug Deliv Transl Res 2022; 12:2039-2041. [PMID: 35882743 PMCID: PMC9360152 DOI: 10.1007/s13346-022-01209-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/30/2022] [Indexed: 11/30/2022]
|