1
|
Sun B, Zhao Y, Yang S, Li X, Li N, Wang Y, Han Q, Liu X, Tu Q, Zheng J, Zhang X. Celecoxib as a potential treatment for hepatocellular carcinoma in populations exposed to high PFAS levels. JOURNAL OF HAZARDOUS MATERIALS 2025; 489:137613. [PMID: 39955994 DOI: 10.1016/j.jhazmat.2025.137613] [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: 09/17/2024] [Revised: 02/12/2025] [Accepted: 02/12/2025] [Indexed: 02/18/2025]
Abstract
Per- and polyfluoroalkyl substances (PFAS), including perfluorooctane sulfonate and perfluorooctanoic acid, are associated with adverse human effects. However, few studies have assessed the effects of PFAS mixtures on hepatocellular carcinoma (HCC). In this study, we systematically investigated the effects and underlying mechanisms of PFAS mixtures on the proliferation, migration, and invasion of HCC cells (JHH-7 and Li-7) in vitro using a combination of biological techniques and high-coverage untargeted metabolomics. A six day exposure to a 5 μM PFAS mixture significantly enhanced the malignant progression of HCC in vitro. Metabolomic analysis identified the upregulation of prostaglandin E2 (PGE2) as a key factor associated with these effects. This hypothesis was further validated using celecoxib, a PGE2 inhibitor, which reduced PGE2 levels in HCC cells, consequently slowing their migration and invasion. Additionally, mice treated with celecoxib exhibited reduced tumor volumes compared with those treated with PFAS alone. These results suggest that PFAS exposure enhances HCC malignancy through the PI3K/AKT signaling pathway via increased PGE2 production. In conclusion, a 5 μM PFAS mixture accelerates HCC proliferation and invasion; moreover, celecoxib demonstrates potential as a therapeutic agent that inhibits these effects.
Collapse
Affiliation(s)
- Boshi Sun
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, No. 148 BaoJian-ro, Harbin, Heilongjiang Province 150086, China
| | - Yuqiao Zhao
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, No. 148 BaoJian-ro, Harbin, Heilongjiang Province 150086, China
| | - Shifeng Yang
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, No. 148 BaoJian-ro, Harbin, Heilongjiang Province 150086, China
| | - Xiaodong Li
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, No. 148 BaoJian-ro, Harbin, Heilongjiang Province 150086, China
| | - Nana Li
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, No. 148 BaoJian-ro, Harbin, Heilongjiang Province 150086, China
| | - Yujie Wang
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, No. 148 BaoJian-ro, Harbin, Heilongjiang Province 150086, China
| | - Qixiang Han
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, No. 148 BaoJian-ro, Harbin, Heilongjiang Province 150086, China
| | - Xuyun Liu
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, No. 148 BaoJian-ro, Harbin, Heilongjiang Province 150086, China
| | - Qiushi Tu
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, No. 148 BaoJian-ro, Harbin, Heilongjiang Province 150086, China
| | - Jie Zheng
- Department of Environmental Health Sciences, Yale School of Public Health, Yale University, New Haven, CT 06510, United States.
| | - Xinyu Zhang
- Department of General Surgery, The 2nd Affiliated Hospital of Harbin Medical University, No. 148 BaoJian-ro, Harbin, Heilongjiang Province 150086, China.
| |
Collapse
|
2
|
Jeong T, Lee MS, Jeon J, Park JH, Chung Y, Yang HS. Advanced stem cell therapy using both cell spheroids transplant and paracrine factor release hydrogel patches for myocardial infarction. Colloids Surf B Biointerfaces 2025; 253:114772. [PMID: 40378458 DOI: 10.1016/j.colsurfb.2025.114772] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2025] [Revised: 04/25/2025] [Accepted: 05/06/2025] [Indexed: 05/19/2025]
Abstract
Conventional micro-concave systems have been proposed as effective methods for facile cell spheroid formation, culture. However, these systems face challenges in terms of ease of cell transplantation and a low cell survival rate in ischemic disease. We present a novel open/close type hydrogel micro-concave patch (OC) designed for in situ 3D cell spheroid formation, culture, and a transplantable system utilizing a 3D printed mold. Open-type patches were fabricated with a rigid hydrogel, while closed-type patches were prepared with a combination of swellable soft hydrogel and rigid hydrogel. The open-type concave was intended for cell spheroid formation and subsequent transplantation into the ischemic region. Conversely, the close-type concave allowed released cytokines from cell spheroids, which were located inside the concave, to promote survival of transplanted cell spheroid. We hypothesized that transplant of open-type cell spheroids, combined with the release of paracrine factors from close-type cell spheroids, could enhance therapeutic outcomes in ischemic regions. The OC was prepared using different concentration ratios of swellable polyacrylamide (PAAM) hydrogel through 3D printed micropillar mold. Additionally, PAAM was characterized to enhance the compactness of close-type 3D cell spheroids. Transplantation of OC improved the therapeutic effect in a rat cardiac infarction model compared to open-type patches.
Collapse
Affiliation(s)
- Taekgwang Jeong
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Min Suk Lee
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea; Richard and Loan Hill Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Jin Jeon
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea; Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul 02792, Republic of Korea
| | - Jin Hee Park
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Youngdoo Chung
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea
| | - Hee Seok Yang
- Department of Nanobiomedical Science & BK21 FOUR NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan 31116, Republic of Korea; Department of Biomedical Science & Engineering, Dankook University, Cheonan 31116, Republic of Korea; Center for Bio-Medical Engineering Core-Facility, Dankook University, Cheonan 31116, Republic of Korea.
| |
Collapse
|
3
|
Zhai Y, Fu J, Yang J, Zhou Y. Quercetin in Shengxian Decoction exhibits anti-ferroptosis protective roles in a myocardial infarction model via targeting DPP4/ HMOX1, based on network pharmacology and molecular docking. Front Pharmacol 2025; 16:1583509. [PMID: 40365322 PMCID: PMC12069271 DOI: 10.3389/fphar.2025.1583509] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 03/31/2025] [Indexed: 05/15/2025] Open
Abstract
Background Myocardial infarction (MI) is characterized by high morbidity. In this study, we aimed to elucidate potential targets of Shengxian Decoction (SXD) against MI. Methods Pairing of SXD active ingredients and MI targets was conducted using the Chinese Medicine System Pharmacological Database, Gene Expression Omnibus (GEO), and STRING databases. The effects of SXD on MI were validated in vitro. Molecular docking was verified using cellular thermal shift assay (CETSA). Results A total of 40 active ingredients and 28 MI-related targets were obtained. Cross-analysis on 28 targets and cell death-related genes identified two crucial ferroptosis-related targets, namely, dipeptidyl peptidase 4 (DPP4) and heme oxygenase 1 (HMOX1). In cobalt chloride (CoCl2)-induced hypoxic H9c2 cells, SXD could remarkably improve cell viability and inhibit cell death. Meanwhile, SXD treatment significantly affected the ferroptosis-related markers in hypoxic H9c2 cells. Molecular docking and CETSA results showed that quercetin had good binding activity with DPP4 and HMOX1. Conclusion Important active ingredient quercetin in SXD could exert anti-ferroptosis protective roles on MI through targeting ferroptosis-related genes (DPP4/HMOX1), thereby contributing to the protective role of SXD on MI.
Collapse
Affiliation(s)
- Yuming Zhai
- Graduate School, Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Jiamei Fu
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Jianfei Yang
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| | - Yabin Zhou
- Department of Cardiovascular Medicine, The First Affiliated Hospital of Heilongjiang University of Chinese Medicine, Harbin, Heilongjiang, China
| |
Collapse
|
4
|
Kim YJ, Kim H, Lee DH, Kim YH, Park JH, Sim WS, Kim JJ, Ban K, Um SH, Park HJ, Davis ME, Park HJ, Bhang SH. Reinforcing Stromal Cell Spheroid Through Red-Light Preconditioning for Advanced Vascularization. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2500788. [PMID: 40278796 DOI: 10.1002/advs.202500788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 03/30/2025] [Indexed: 04/26/2025]
Abstract
Despite the promising potential of stromal cell therapy in treating myocardial infarction (MI), its effectiveness is limited by poor cell retention and engraftment in ischemic environments. This study introduces a novel strategy that combines the preconditioning of human adipose-derived stromal cells (hADSCs) using OLED-based photobiomodulation (OPBM) and culturing these cells into 3D spheroids. The preconditioned 3D spheroids (APCS group) exhibit significantly enhanced angiogenic, arterialized, and tissue remodeling capabilities compared with those of traditional 2D cultures and non-preconditioned spheroids. In vivo transplantation of these spheroids into the border zone of infarcted area significantly improve cardiac function and reduce adverse remodeling by enhancing anti-fibrosis and angiogenesis including arterialization. The combined strategy with OPBM preconditioning and 3D spheroid culture system can enhance therapeutic potential of hADSCs with multiple paracrine effects for cardiac repair. This novel approach provides next generation of cell therapeutics to overcome the limitation of adult stromal cell therapy in patients with post-MI heart failure.
Collapse
Affiliation(s)
- Yu-Jin Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Hyeok Kim
- Center for Biomaterials, Biomedical Research Institute, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Dong-Hyun Lee
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Yeong Hwan Kim
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Jae-Hyun Park
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Woo-Sup Sim
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Jin-Ju Kim
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Kiwon Ban
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong, 999077, China
| | - Soong Ho Um
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| | - Hyun Ji Park
- Department of Molecular Science and Technology, Ajou University, Suwon, 16499, Republic of Korea
| | - Michael E Davis
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University, Atlanta, GA, 30332, USA
- Children's Heart Research & Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, GA, 30322, USA
| | - Hun-Jun Park
- Department of Biomedicine & Health Sciences, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Division of Cardiology, Department of Internal Medicine, Uijeongbu St. Mary's Hospital, The Catholic University of Korea, Seoul, 06591, Republic of Korea
- Cell Death Disease Research Center, College of Medicine, The Catholic University of Korea, Seoul, 06591, Republic of Korea
| | - Suk Ho Bhang
- School of Chemical Engineering, Sungkyunkwan University, Suwon, 16419, Republic of Korea
| |
Collapse
|
5
|
Abdurakhmanova МM, Leonteva AA, Vasilieva NS, Kuligina EV, Nushtaeva AA. 3D cell culture models: how to obtain and characterize the main models. Vavilovskii Zhurnal Genet Selektsii 2025; 29:175-188. [PMID: 40264808 PMCID: PMC12011624 DOI: 10.18699/vjgb-25-21] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 11/28/2024] [Accepted: 11/28/2024] [Indexed: 04/24/2025] Open
Abstract
For many years, the gold standard in the study of malignant tumors has been the in vitro culture of tumor cells, in vivo xenografts or genetically modified animal models. Meanwhile, three-dimensional cell models (3D cultures) have been added to the arsenal of modern biomedical research. 3D cultures reproduce tissue-specific features of tissue topology. This makes them relevant tissue models in terms of cell differentiation, metabolism and the development of drug resistance. Such models are already being used by many research groups for both basic and translational research, and may substantially reduce the number of animal studies, for example in the field of oncological research. In the current literature, 3D cultures are classified according to the technique of their formation (with or without a scaffold), cultivation conditions (static or dynamic), as well as their cellular organization and function. In terms of cellular organization, 3D cultures are divided into "spheroid models", "organoids", "organs-on-a-chip" and "microtissues". Each of these models has its own unique features, which should be taken into account when using a particular model in an experiment. The simplest 3D cultures are spheroid models which are floating spherical cell aggregates. An organoid is a more complex 3D model, in which a self-organizing 3D structure is formed from stem cells (SCs) capable of self-renewal and differentiation within the model. Organ-on-a-chip models are chips of microfluidic systems that simulate dynamic physical and biological processes found in organs and tissues in vitro. By combining different cell types into a single structure, spheroids and organoids can act as a basis for the formation of a microtissue - a hybrid 3D model imitating a specific tissue phenotype and containing tissue-specific extracellular matrix (ECM) components. This review presents a brief history of 3D cell culture. It describes the main characteristics and perspectives of the use of "spheroid models", "organoids", "organ-on-a-chip" models and "microtissues" in immune oncology research of solid tumors.
Collapse
Affiliation(s)
- М M Abdurakhmanova
- Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - A A Leonteva
- Sirius University of Science and Technology, Sirius Federal Territory, Krasnodar Region, Russia Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - N S Vasilieva
- Sirius University of Science and Technology, Sirius Federal Territory, Krasnodar Region, Russia Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - E V Kuligina
- Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| | - A A Nushtaeva
- Sirius University of Science and Technology, Sirius Federal Territory, Krasnodar Region, Russia Institute of Chemical Biology and Fundamental Medicine of the Siberian Branch of the Russian Academy of Sciences, Novosibirsk, Russia
| |
Collapse
|
6
|
Krungkraipetch L, Supajitgulchai D, Assawaboonyadech A, Puranawit W. Efficacy and safety of mesenchymal stem cell therapies in retinitis pigmentosa: a systematic review and meta-analysis. Int Ophthalmol 2025; 45:85. [PMID: 40072800 DOI: 10.1007/s10792-025-03478-6] [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: 12/11/2024] [Accepted: 02/22/2025] [Indexed: 03/14/2025]
Abstract
BACKGROUND Retinitis pigmentosa (RP) is a retinal dystrophy and genetically heterogeneous group that causes vision loss and necessitates innovative therapeutic strategies, and mesenchymal stem cell (MSC) therapy has shown potential due to its regenerative and immunomodulatory properties. This meta-analysis aims to evaluate the efficacy and safety of MSC therapies in improving visual outcomes, focusing on the impact of various MSC types, administration methods, and duration of benefits. METHODS A systematic search of peer-reviewed studies was conducted to identify clinical trials and observational studies investigating MSC therapies for retinal conditions. Outcomes of interest included best-corrected visual acuity (BCVA), central macular thickness, retinal sensitivity, quality of life, and safety profiles. Data were synthesized and analyzed using random-effects meta-analysis to calculate pooled effect sizes and heterogeneity. PROSPERO CRD42024618158. RESULTS Eleven studies involving 355 RP patients were included. Umbilical cord-derived MSCs and bone marrow-derived MSCs demonstrated significant short-term improvements in BCVA and retinal function. Subretinal and suprachoroidal delivery methods were associated with better outcomes compared to systemic infusion. Adverse effects were minimal, with transient inflammation being the most reported. The duration of benefits varied, with most studies reporting sustained improvements up to 12 months, while long-term efficacy beyond this period was less conclusive. CONCLUSIONS MSC therapies show promise in improving visual function and retinal health, with safety profiles supporting their clinical feasibility. However, differences in administration methods and MSC types influence outcomes. Further large-scale, long-term randomized controlled trials are needed to optimize treatment protocols and validate sustained benefits.
Collapse
Affiliation(s)
| | | | | | - Warisanan Puranawit
- Burapha University Hospital, Burapha University, Saen Suk, Chonburi, Thailand
| |
Collapse
|
7
|
Voloshin N, Tyurin-Kuzmin P, Karagyaur M, Akopyan Z, Kulebyakin K. Practical Use of Immortalized Cells in Medicine: Current Advances and Future Perspectives. Int J Mol Sci 2023; 24:12716. [PMID: 37628897 PMCID: PMC10454025 DOI: 10.3390/ijms241612716] [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: 06/21/2023] [Revised: 07/23/2023] [Accepted: 08/09/2023] [Indexed: 08/27/2023] Open
Abstract
In modern science, immortalized cells are not only a convenient tool in fundamental research, but they are also increasingly used in practical medicine. This happens due to their advantages compared to the primary cells, such as the possibility to produce larger amounts of cells and to use them for longer periods of time, the convenience of genetic modification, the absence of donor-to-donor variability when comparing the results of different experiments, etc. On the other hand, immortalization comes with drawbacks: possibilities of malignant transformation and/or major phenotype change due to genetic modification itself or upon long-term cultivation appear. At first glance, such issues are huge hurdles in the way of immortalized cells translation into medicine. However, there are certain ways to overcome such barriers that we describe in this review. We determined four major areas of usage of immortalized cells for practical medicinal purposes, and each has its own means to negate the drawbacks associated with immortalization. Moreover, here we describe specific fields of application of immortalized cells in which these problems are of much lesser concern, for example, in some cases where the possibility of malignant growth is not there at all. In general, we can conclude that immortalized cells have their niches in certain areas of practical medicine where they can successfully compete with other therapeutic approaches, and more preclinical and clinical trials with them should be expected.
Collapse
Affiliation(s)
- Nikita Voloshin
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia; (N.V.); (P.T.-K.); (M.K.)
| | - Pyotr Tyurin-Kuzmin
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia; (N.V.); (P.T.-K.); (M.K.)
| | - Maxim Karagyaur
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia; (N.V.); (P.T.-K.); (M.K.)
| | - Zhanna Akopyan
- Medical Research and Education Center, Lomonosov Moscow State University, 119234 Moscow, Russia;
| | - Konstantin Kulebyakin
- Faculty of Medicine, Lomonosov Moscow State University, 119991 Moscow, Russia; (N.V.); (P.T.-K.); (M.K.)
- Medical Research and Education Center, Lomonosov Moscow State University, 119234 Moscow, Russia;
| |
Collapse
|
8
|
Wu K, Wang Y, Yang H, Chen Y, Lu K, Wu Y, Liu C, Zhang H, Meng H, Yu Q, Zhang Y, Shen Z. Injectable Decellularized Extracellular Matrix Hydrogel Containing Stromal Cell-Derived Factor 1 Promotes Transplanted Cardiomyocyte Engraftment and Functional Regeneration after Myocardial Infarction. ACS APPLIED MATERIALS & INTERFACES 2023; 15:2578-2589. [PMID: 36598791 DOI: 10.1021/acsami.2c16682] [Citation(s) in RCA: 20] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/17/2023]
Abstract
Transplantation of exogenous cardiomyocytes (CMs) is a hopeful method to treat myocardial infarction (MI). However, its clinical application still remains challenging due to low retention and survival rates of the transplanted cells. Herein, a stromal cell-derived factor 1 (SDF-1)-loaded injectable hydrogel based on a decellularized porcine extracellular matrix (dECM) is developed to encapsulate and deliver CMs locally to the infarct area of the heart. The soluble porcine cardiac dECM is composed of similar components such as the human cardiac ECM, which could be self-assembled into a nanofibrous hydrogel at physiological temperature to improve the retention of transplanted CMs. Furthermore, the chemokine SDF-1 could recruit endogenous cells to promote angiogenesis, mitigating the ischemic microenvironment and improving the survival of CMs. The results in vitro show that this composite hydrogel exhibits good biocompatibility, anti-apoptosis property, and chemotactic effects for mesenchymal stromal cells and endothelial cells through SDF-1-CXCR4 axis. Moreover, intramyocardial injection of this composite hydrogel to the infarcted area leads to the promotion of angiogenesis and inhibition of fibrosis, reducing the infarction size and improving the cardiac function. The combination of natural biomaterials, exogenous cells, and bioactive factors shows potential for MI treatment in the clinical application.
Collapse
Affiliation(s)
- Kui Wu
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215007, P. R. China
- Department of Cardiovascular Surgery, The Affiliated Hospital of Guizhou Medical University, Guiyang 550004, P. R. China
| | - Yuanyuan Wang
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215007, P. R. China
| | - Hong Yang
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215007, P. R. China
| | - Yihuan Chen
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215007, P. R. China
| | - Kunyan Lu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, P. R. China
| | - Yong Wu
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215007, P. R. China
| | - Chunxia Liu
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215007, P. R. China
| | - Haixin Zhang
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, P. R. China
| | - Hanyu Meng
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215007, P. R. China
| | - Qian Yu
- State and Local Joint Engineering Laboratory for Novel Functional Polymeric Materials, College of Chemistry, Chemical Engineering and Materials Science, Soochow University, Suzhou 215123, P. R. China
| | - Yanxia Zhang
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215007, P. R. China
| | - Zhenya Shen
- Department of Cardiovascular Surgery of the First Affiliated Hospital and Institute for Cardiovascular Science, Suzhou Medical College of Soochow University, Soochow University, Suzhou 215007, P. R. China
| |
Collapse
|
9
|
Kim W, Gwon Y, Park S, Kim H, Kim J. Therapeutic strategies of three-dimensional stem cell spheroids and organoids for tissue repair and regeneration. Bioact Mater 2023; 19:50-74. [PMID: 35441116 PMCID: PMC8987319 DOI: 10.1016/j.bioactmat.2022.03.039] [Citation(s) in RCA: 86] [Impact Index Per Article: 43.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 03/16/2022] [Accepted: 03/25/2022] [Indexed: 02/07/2023] Open
Abstract
Three-dimensional (3D) stem cell culture systems have attracted considerable attention as a way to better mimic the complex interactions between individual cells and the extracellular matrix (ECM) that occur in vivo. Moreover, 3D cell culture systems have unique properties that help guide specific functions, growth, and processes of stem cells (e.g., embryogenesis, morphogenesis, and organogenesis). Thus, 3D stem cell culture systems that mimic in vivo environments enable basic research about various tissues and organs. In this review, we focus on the advanced therapeutic applications of stem cell-based 3D culture systems generated using different engineering techniques. Specifically, we summarize the historical advancements of 3D cell culture systems and discuss the therapeutic applications of stem cell-based spheroids and organoids, including engineering techniques for tissue repair and regeneration.
Collapse
Affiliation(s)
- Woochan Kim
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Yonghyun Gwon
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Sunho Park
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hyoseong Kim
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Jangho Kim
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Department of Rural and Biosystems Engineering, Chonnam National University, Gwangju, 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju, 61186, Republic of Korea
| |
Collapse
|
10
|
Sung K, Patel NR, Ashammakhi N, Nguyen KL. 3-Dimensional Bioprinting of Cardiovascular Tissues: Emerging Technology. JACC Basic Transl Sci 2021; 6:467-482. [PMID: 34095635 PMCID: PMC8165127 DOI: 10.1016/j.jacbts.2020.12.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 11/16/2020] [Accepted: 12/02/2020] [Indexed: 12/19/2022]
Abstract
Three-dimensional (3D) bioprinting may overcome challenges in tissue engineering. Unlike conventional tissue engineering approaches, 3D bioprinting has a proven ability to support vascularization of larger scale constructs and has been used for several cardiovascular applications. An overview of 3D bioprinting techniques, in vivo translation, and challenges are described.
Collapse
Affiliation(s)
- Kevin Sung
- Division of Cardiology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California, USA
- Division of Cardiology, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, USA
| | - Nisha R. Patel
- Division of Cardiology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California, USA
- Division of Cardiology, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, USA
- Stritch School of Medicine, Loyola University of Chicago, Maywood, Illinois, USA
| | - Nureddin Ashammakhi
- Department of Biomedical Engineering, Henry Samueli School of Engineering, University of California-Los Angeles, Los Angeles, California, USA
- Department of Radiology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California, USA
| | - Kim-Lien Nguyen
- Division of Cardiology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California, USA
- Division of Cardiology, Veterans Affairs Greater Los Angeles Healthcare System, Los Angeles, California, USA
- Department of Radiology, David Geffen School of Medicine, University of California-Los Angeles, Los Angeles, California, USA
- Physics and Biology in Medicine Graduate Program, University of California-Los Angeles, Los Angeles, California, USA
| |
Collapse
|
11
|
Liu Chung Ming C, Sesperez K, Ben-Sefer E, Arpon D, McGrath K, McClements L, Gentile C. Considerations to Model Heart Disease in Women with Preeclampsia and Cardiovascular Disease. Cells 2021; 10:899. [PMID: 33919808 PMCID: PMC8070848 DOI: 10.3390/cells10040899] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2021] [Revised: 04/11/2021] [Accepted: 04/12/2021] [Indexed: 12/12/2022] Open
Abstract
Preeclampsia is a multifactorial cardiovascular disorder diagnosed after 20 weeks of gestation, and is the leading cause of death for both mothers and babies in pregnancy. The pathophysiology remains poorly understood due to the variability and unpredictability of disease manifestation when studied in animal models. After preeclampsia, both mothers and offspring have a higher risk of cardiovascular disease (CVD), including myocardial infarction or heart attack and heart failure (HF). Myocardial infarction is an acute myocardial damage that can be treated through reperfusion; however, this therapeutic approach leads to ischemic/reperfusion injury (IRI), often leading to HF. In this review, we compared the current in vivo, in vitro and ex vivo model systems used to study preeclampsia, IRI and HF. Future studies aiming at evaluating CVD in preeclampsia patients could benefit from novel models that better mimic the complex scenario described in this article.
Collapse
Affiliation(s)
- Clara Liu Chung Ming
- School of Biomedical Engineering/FEIT, University of Technology Sydney, Sydney, NSW 2007, Australia; (C.L.C.M.); (E.B.-S.); (D.A.)
| | - Kimberly Sesperez
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (K.S.); (K.M.); (L.M.)
| | - Eitan Ben-Sefer
- School of Biomedical Engineering/FEIT, University of Technology Sydney, Sydney, NSW 2007, Australia; (C.L.C.M.); (E.B.-S.); (D.A.)
| | - David Arpon
- School of Biomedical Engineering/FEIT, University of Technology Sydney, Sydney, NSW 2007, Australia; (C.L.C.M.); (E.B.-S.); (D.A.)
| | - Kristine McGrath
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (K.S.); (K.M.); (L.M.)
| | - Lana McClements
- School of Life Sciences, Faculty of Science, University of Technology Sydney, Sydney, NSW 2007, Australia; (K.S.); (K.M.); (L.M.)
| | - Carmine Gentile
- School of Biomedical Engineering/FEIT, University of Technology Sydney, Sydney, NSW 2007, Australia; (C.L.C.M.); (E.B.-S.); (D.A.)
- Sydney Medical School, The University of Sydney, Sydney, NSW 2000, Australia
- Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
12
|
Huang RY, Liu ZH, Weng WH, Chang CW. Magnetic nanocomplexes for gene delivery applications. J Mater Chem B 2021; 9:4267-4286. [PMID: 33942822 DOI: 10.1039/d0tb02713h] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Gene delivery is an indispensable technique for various biomedical applications such as gene therapy, stem cell engineering and gene editing. Recently, magnetic nanoparticles (MNPs) have received increasing attention for their use in promoting gene delivery efficiency. Under magnetic attraction, gene delivery efficiency using viral or nonviral gene carriers could be universally enhanced. Besides, magnetic nanoparticles could be utilized in magnetic resonance imaging or magnetic hyperthermia therapy, providing extra theranostic opportunities. In this review, recent research integrating MNPs with a viral or nonviral gene vector is summarized from both technical and application perspectives. Applications of MNPs in cutting-edge research technologies, such as biomimetic cell membrane nano-gene carriers, exosome-based gene delivery, cell-based drug delivery systems or CRISPR/Cas9 gene editing, are also discussed.
Collapse
Affiliation(s)
- Rih-Yang Huang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| | - Zhuo-Hao Liu
- Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Chang Gung Medical College and University, Taiwan.
| | - Wei-Han Weng
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| | - Chien-Wen Chang
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, Taiwan.
| |
Collapse
|
13
|
Mancuso T, Barone A, Salatino A, Molinaro C, Marino F, Scalise M, Torella M, De Angelis A, Urbanek K, Torella D, Cianflone E. Unravelling the Biology of Adult Cardiac Stem Cell-Derived Exosomes to Foster Endogenous Cardiac Regeneration and Repair. Int J Mol Sci 2020; 21:E3725. [PMID: 32466282 PMCID: PMC7279257 DOI: 10.3390/ijms21103725] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2020] [Revised: 05/15/2020] [Accepted: 05/20/2020] [Indexed: 12/11/2022] Open
Abstract
Cardiac remuscularization has been the stated goal of the field of regenerative cardiology since its inception. Along with the refreshment of lost and dysfunctional cardiac muscle cells, the field of cell therapy has expanded in scope encompassing also the potential of the injected cells as cardioprotective and cardio-reparative agents for cardiovascular diseases. The latter has been the result of the findings that cell therapies so far tested in clinical trials exert their beneficial effects through paracrine mechanisms acting on the endogenous myocardial reparative/regenerative potential. The endogenous regenerative potential of the adult heart is still highly debated. While it has been widely accepted that adult cardiomyocytes (CMs) are renewed throughout life either in response to wear and tear and after injury, the rate and origin of this phenomenon are yet to be clarified. The adult heart harbors resident cardiac/stem progenitor cells (CSCs/CPCs), whose discovery and characterization were initially sufficient to explain CM renewal in response to physiological and pathological stresses, when also considering that adult CMs are terminally differentiated cells. The role of CSCs in CM formation in the adult heart has been however questioned by some recent genetic fate map studies, which have been proved to have serious limitations. Nevertheless, uncontested evidence shows that clonal CSCs are effective transplantable regenerative agents either for their direct myogenic differentiation and for their paracrine effects in the allogeneic setting. In particular, the paracrine potential of CSCs has been the focus of the recent investigation, whereby CSC-derived exosomes appear to harbor relevant regenerative and reparative signals underlying the beneficial effects of CSC transplantation. This review focuses on recent advances in our knowledge about the biological role of exosomes in heart tissue homeostasis and repair with the idea to use them as tools for new therapeutic biotechnologies for "cell-less" effective cardiac regeneration approaches.
Collapse
Affiliation(s)
- Teresa Mancuso
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Antonella Barone
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Alessandro Salatino
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Claudia Molinaro
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Fabiola Marino
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Mariangela Scalise
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Michele Torella
- Department of Translational Medical Sciences, AORN dei Colli/Monaldi Hospital, University of Campania “L. Vanvitelli”, Via Leonardo Bianchi, 80131 Naples, Italy;
| | - Antonella De Angelis
- Department of Experimental Medicine, Section of Pharmacology, University of Campania “L.Vanvitelli”, 80121 Naples, Italy;
| | - Konrad Urbanek
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Daniele Torella
- Molecular and Cellular Cardiology, Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy; (T.M.); (A.B.); (A.S.); (C.M.); (F.M.); (M.S.); (K.U.)
| | - Eleonora Cianflone
- Molecular and Cellular Cardiology, Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy;
| |
Collapse
|