1
|
Savelyev MS, Kuksin AV, Murashko DT, Otsupko EP, Suchkova VV, Popovich KD, Vasilevsky PN, Vasilevskaya YO, Kurilova UE, Eganova EM, Edelbekova PA, Selishchev SV, Pavlov AA, Gerasimenko AY. Formation of Neurointerfaces Based on Electrically Conductive Biopolymers by Two-Photon Polymerization Method. Polymers (Basel) 2025; 17:1300. [PMID: 40430596 PMCID: PMC12114728 DOI: 10.3390/polym17101300] [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: 03/04/2025] [Revised: 05/07/2025] [Accepted: 05/07/2025] [Indexed: 05/29/2025] Open
Abstract
Preventing false signals of phantom pain after limb amputation is crucial. The development of neurointerfaces capable of bidirectional information exchange between the brain and external devices, along with long-term use, is a key research priority. The main problem with existing devices lies in the potential formation of scar tissue and the death of adjacent neurons. To address this issue, a polymer composite based on new composition: chitosan, bovine serum albumin, single-walled carbon nanotubes, and Eosin Y, which was created for the fabrication of a neurointerface. A polymer composite of the required shape was formed by two-photon polymerization. In studying its nonlinear optical properties, the new effect of phase self-modulation was discovered, which is observed after exposure to laser radiation prior to the formation of the composite. The time of appearance of diffraction rings was measured. This allowed optimization of laser parameters-scanner speed and intensity. The resulting homogeneous composite exhibited a specific conductivity of 20 mS × cm-1, sufficient for electrophysiological signal transmission.
Collapse
Affiliation(s)
- Mikhail S. Savelyev
- Institute of Biomedical Systems, National Research University of Electronic Technology, 124498 Zelenograd, Russia; (A.V.K.); (E.P.O.); (V.V.S.); (K.D.P.); (P.N.V.); (U.E.K.); (S.V.S.); (A.Y.G.)
- Institute for Bionic Technologies and Engineering, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Artem V. Kuksin
- Institute of Biomedical Systems, National Research University of Electronic Technology, 124498 Zelenograd, Russia; (A.V.K.); (E.P.O.); (V.V.S.); (K.D.P.); (P.N.V.); (U.E.K.); (S.V.S.); (A.Y.G.)
| | - Denis T. Murashko
- Institute of Biomedical Systems, National Research University of Electronic Technology, 124498 Zelenograd, Russia; (A.V.K.); (E.P.O.); (V.V.S.); (K.D.P.); (P.N.V.); (U.E.K.); (S.V.S.); (A.Y.G.)
| | - Ekaterina P. Otsupko
- Institute of Biomedical Systems, National Research University of Electronic Technology, 124498 Zelenograd, Russia; (A.V.K.); (E.P.O.); (V.V.S.); (K.D.P.); (P.N.V.); (U.E.K.); (S.V.S.); (A.Y.G.)
| | - Victoria V. Suchkova
- Institute of Biomedical Systems, National Research University of Electronic Technology, 124498 Zelenograd, Russia; (A.V.K.); (E.P.O.); (V.V.S.); (K.D.P.); (P.N.V.); (U.E.K.); (S.V.S.); (A.Y.G.)
- Institute for Bionic Technologies and Engineering, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Kristina D. Popovich
- Institute of Biomedical Systems, National Research University of Electronic Technology, 124498 Zelenograd, Russia; (A.V.K.); (E.P.O.); (V.V.S.); (K.D.P.); (P.N.V.); (U.E.K.); (S.V.S.); (A.Y.G.)
- Institute for Bionic Technologies and Engineering, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Pavel N. Vasilevsky
- Institute of Biomedical Systems, National Research University of Electronic Technology, 124498 Zelenograd, Russia; (A.V.K.); (E.P.O.); (V.V.S.); (K.D.P.); (P.N.V.); (U.E.K.); (S.V.S.); (A.Y.G.)
| | | | - Ulyana E. Kurilova
- Institute of Biomedical Systems, National Research University of Electronic Technology, 124498 Zelenograd, Russia; (A.V.K.); (E.P.O.); (V.V.S.); (K.D.P.); (P.N.V.); (U.E.K.); (S.V.S.); (A.Y.G.)
- Institute for Bionic Technologies and Engineering, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| | - Elena M. Eganova
- Institute of Nanotechnology of Microelectronics of the Russian Academy of Sciences, 119991 Moscow, Russia; (E.M.E.); (P.A.E.); (A.A.P.)
| | - Polina A. Edelbekova
- Institute of Nanotechnology of Microelectronics of the Russian Academy of Sciences, 119991 Moscow, Russia; (E.M.E.); (P.A.E.); (A.A.P.)
| | - Sergey V. Selishchev
- Institute of Biomedical Systems, National Research University of Electronic Technology, 124498 Zelenograd, Russia; (A.V.K.); (E.P.O.); (V.V.S.); (K.D.P.); (P.N.V.); (U.E.K.); (S.V.S.); (A.Y.G.)
| | - Alexander A. Pavlov
- Institute of Nanotechnology of Microelectronics of the Russian Academy of Sciences, 119991 Moscow, Russia; (E.M.E.); (P.A.E.); (A.A.P.)
| | - Alexander Yu. Gerasimenko
- Institute of Biomedical Systems, National Research University of Electronic Technology, 124498 Zelenograd, Russia; (A.V.K.); (E.P.O.); (V.V.S.); (K.D.P.); (P.N.V.); (U.E.K.); (S.V.S.); (A.Y.G.)
- Institute for Bionic Technologies and Engineering, I. M. Sechenov First Moscow State Medical University, 119991 Moscow, Russia
| |
Collapse
|
2
|
De Jesus Morales KJ, Santosa U, Brazhkina O, Rajurkar P, Jo H, Davis ME. A Biomimetic Leaflet Scaffold for Aortic Valve Remodeling. Adv Healthc Mater 2024; 13:e2303972. [PMID: 38692263 DOI: 10.1002/adhm.202303972] [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: 11/13/2023] [Revised: 03/28/2024] [Indexed: 05/03/2024]
Abstract
Heart valve disease poses a significant clinical challenge, especially in pediatric populations, due to the inability of existing valve replacements to grow or respond biologically to their microenvironment. Tissue-engineered heart valves (TEHVs) provide a solution by facilitating patient-specific models for self-repair and remodeling. In this study, a 3D-bioprinted TEHV is designed to emulate the trilayer leaflet structure of an aortic valve. A cell-laden hydrogel scaffold made from gelatin methacrylate and polyethylene glycol diacrylate (GelMA/PEGDA) incorporates valvular interstitial-like (VIC-like) cells, being reinforced with a layer of polycaprolactone (PCL). The composition of the hydrogel scaffold remains stable over 7 days, having increased mechanical strength compared to pure GelMA. The scaffold maintains VIC-like cell function and promotes extracellular matrix (ECM) protein expression up to 14 days under two dynamic culture conditions: shear stress and stretching; replicating heart valve behavior within a more physiological-like setting and suggesting remodeling potential via ECM synthesis. This TEHV offers a promising avenue for valve replacements, closely replicating the structural and functional attributes of a native aortic valve, leading to mechanical and biological integration through biomaterial-cellular interactions.
Collapse
Affiliation(s)
- Kenneth J De Jesus Morales
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine & Georgia Institute of Technology, Atlanta, GA, 30322, USA
| | - Utari Santosa
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine & Georgia Institute of Technology, Atlanta, GA, 30322, USA
| | - Olga Brazhkina
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine & Georgia Institute of Technology, Atlanta, GA, 30322, USA
| | - Pranshu Rajurkar
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine & Georgia Institute of Technology, Atlanta, GA, 30322, USA
| | - Hanjoong Jo
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine & Georgia Institute of Technology, Atlanta, GA, 30322, USA
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
| | - Michael E Davis
- Wallace H. Coulter Department of Biomedical Engineering, Emory University School of Medicine & Georgia Institute of Technology, Atlanta, GA, 30322, USA
- Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA, 30322, USA
- Children's Heart Research & Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, GA, 30322, USA
| |
Collapse
|
3
|
Altin-Yavuzarslan G, Sadaba N, Brooks SM, Alper HS, Nelson A. Engineered Living Material Bioreactors with Tunable Mechanical Properties using Vat Photopolymerization. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2306564. [PMID: 38105580 DOI: 10.1002/smll.202306564] [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: 08/01/2023] [Revised: 11/29/2023] [Indexed: 12/19/2023]
Abstract
3D-printed engineered living materials (ELM) are promising bioproduction platforms for agriculture, biotechnology, sustainable energy, and green technology applications. However, the design of these platforms faces several challenges, such as the processability of these materials into complex form factors and control over their mechanical properties. Herein, ELM are presented as 3D-printed bioreactors with arbitrary shape geometries and tunable mechanical properties (moduli and toughness). Poly(ethylene glycol) diacrylate (PEGDA) is used as the precursor to create polymer networks that encapsulate the microorganisms during the vat photopolymerization process. A major limitation of PEGDA networks is their propensity to swell and fracture when submerged in water. The authors overcame this issue by adding glycerol to the resin formulation to 3D print mechanically tough ELM hydrogels. While polymer concentration affects the modulus and reduces bioproduction, ELM bioreactors still maintain their metabolic activity regardless of polymer concentration. These ELM bioreactors have the potential to be used in different applications for sustainable architecture, food production, and biomedical devices that require different mechanical properties from soft to stiff.
Collapse
Affiliation(s)
- Gokce Altin-Yavuzarslan
- Molecular Engineering and Sciences Institute, University of Washington, Box 351700, Seattle, WA, 98195, USA
- Department of Chemistry, University of Washington, Box 351700, Seattle, WA, 98195, USA
| | - Naroa Sadaba
- Department of Chemistry, University of Washington, Box 351700, Seattle, WA, 98195, USA
| | - Sierra M Brooks
- McKetta Department of Chemical Engineering, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Hal S Alper
- Department of Chemistry, University of Washington, Box 351700, Seattle, WA, 98195, USA
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX, 78712, USA
| | - Alshakim Nelson
- Molecular Engineering and Sciences Institute, University of Washington, Box 351700, Seattle, WA, 98195, USA
- Department of Chemistry, University of Washington, Box 351700, Seattle, WA, 98195, USA
| |
Collapse
|
4
|
Cai Z, Zhu M, Xu L, Wang Y, Xu Y, Yim WY, Cao H, Guo R, Qiu X, He X, Shi J, Qiao W, Dong N. Directed Differentiation of Human Induced Pluripotent Stem Cells to Heart Valve Cells. Circulation 2024; 149:1435-1456. [PMID: 38357822 PMCID: PMC11062615 DOI: 10.1161/circulationaha.123.065143] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/12/2023] [Accepted: 01/19/2024] [Indexed: 02/16/2024]
Abstract
BACKGROUND A main obstacle in current valvular heart disease research is the lack of high-quality homogeneous functional heart valve cells. Human induced pluripotent stem cells (hiPSCs)-derived heart valve cells may help with this dilemma. However, there are no well-established protocols to induce hiPSCs to differentiate into functional heart valve cells, and the networks that mediate the differentiation have not been fully elucidated. METHODS To generate heart valve cells from hiPSCs, we sequentially activated the Wnt, BMP4, VEGF (vascular endothelial growth factor), and NFATc1 signaling pathways using CHIR-99021, BMP4, VEGF-165, and forskolin, respectively. The transcriptional and functional similarity of hiPSC-derived heart valve cells compared with primary heart valve cells were characterized. Longitudinal single-cell RNA sequencing was used to uncover the trajectory, switch genes, pathways, and transcription factors of the differentiation. RESULTS An efficient protocol was developed to induce hiPSCs to differentiate into functional hiPSC-derived valve endothelial-like cells and hiPSC-derived valve interstitial-like cells. After 6-day differentiation and CD144 magnetic bead sorting, ≈70% CD144+ cells and 30% CD144- cells were obtained. On the basis of single-cell RNA sequencing data, the CD144+ cells and CD144- cells were found to be highly similar to primary heart valve endothelial cells and primary heart valve interstitial cells in gene expression profile. Furthermore, CD144+ cells had the typical function of primary heart valve endothelial cells, including tube formation, uptake of low-density lipoprotein, generation of endothelial nitric oxide synthase, and response to shear stress. Meanwhile, CD144- cells could secret collagen and matrix metalloproteinases, and differentiate into osteogenic or adipogenic lineages like primary heart valve interstitial cells. Therefore, we identified CD144+ cells and CD144- cells as hiPSC-derived valve endothelial-like cells and hiPSC-derived valve interstitial-like cells, respectively. Using single-cell RNA sequencing analysis, we demonstrated that the trajectory of heart valve cell differentiation was consistent with embryonic valve development. We identified the main switch genes (NOTCH1, HEY1, and MEF2C), signaling pathways (TGF-β, Wnt, and NOTCH), and transcription factors (MSX1, SP5, and MECOM) that mediated the differentiation. Finally, we found that hiPSC-derived valve interstitial-like cells might derive from hiPSC-derived valve endothelial-like cells undergoing endocardial-mesenchymal transition. CONCLUSIONS In summary, this is the first study to report an efficient strategy to generate functional hiPSC-derived valve endothelial-like cells and hiPSC-derived valve interstitial-like cells from hiPSCs, as well as to elucidate the differentiation trajectory and transcriptional dynamics of hiPSCs differentiated into heart valve cells.
Collapse
Affiliation(s)
- Ziwen Cai
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (Z.C., L.X., Y.X., W.Y.Y., H.C., R.G., X.Q, J.S., W.Q., N.D.)
- Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Fuzhou, China (Z.C.)
| | - Miaomiao Zhu
- Department of Cardiovascular Surgery, Union Hospital, Fujian Medical University, Fuzhou, China (Z.C.)
- Institute of Maternal and Children Health, Wuhan Children’s Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji medical College, Huazhong University of Science & Technology, Hubei, China (M.Z.)
| | - Li Xu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (Z.C., L.X., Y.X., W.Y.Y., H.C., R.G., X.Q, J.S., W.Q., N.D.)
| | - Yue Wang
- Department of Anesthesiology, Union Hospital, Fujian Medical University, Fuzhou, China (Y.W.)
| | - Yin Xu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (Z.C., L.X., Y.X., W.Y.Y., H.C., R.G., X.Q, J.S., W.Q., N.D.)
| | - Wai Yen Yim
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (Z.C., L.X., Y.X., W.Y.Y., H.C., R.G., X.Q, J.S., W.Q., N.D.)
| | - Hong Cao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (Z.C., L.X., Y.X., W.Y.Y., H.C., R.G., X.Q, J.S., W.Q., N.D.)
| | - Ruikang Guo
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (Z.C., L.X., Y.X., W.Y.Y., H.C., R.G., X.Q, J.S., W.Q., N.D.)
| | - Xiang Qiu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (Z.C., L.X., Y.X., W.Y.Y., H.C., R.G., X.Q, J.S., W.Q., N.D.)
| | - Ximiao He
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (M.Z., X.H.)
| | - Jiawei Shi
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (Z.C., L.X., Y.X., W.Y.Y., H.C., R.G., X.Q, J.S., W.Q., N.D.)
| | - Weihua Qiao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (Z.C., L.X., Y.X., W.Y.Y., H.C., R.G., X.Q, J.S., W.Q., N.D.)
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, China (Z.C., L.X., Y.X., W.Y.Y., H.C., R.G., X.Q, J.S., W.Q., N.D.)
| |
Collapse
|
5
|
Li M, Jin M, Yang H. Remodelers of the vascular microenvironment: The effect of biopolymeric hydrogels on vascular diseases. Int J Biol Macromol 2024; 264:130764. [PMID: 38462100 DOI: 10.1016/j.ijbiomac.2024.130764] [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: 12/19/2023] [Revised: 01/31/2024] [Accepted: 03/07/2024] [Indexed: 03/12/2024]
Abstract
Vascular disease is the leading health problem worldwide. Vascular microenvironment encompasses diverse cell types, including those within the vascular wall, blood cells, stromal cells, and immune cells. Initiation of the inflammatory state of the vascular microenvironment and changes in its mechanics can profoundly affect vascular homeostasis. Biomedical materials play a crucial role in modern medicine, hydrogels, characterized by their high-water content, have been increasingly utilized as a three-dimensional interaction network. In recent times, the remarkable progress in utilizing hydrogels and understanding vascular microenvironment have enabled the treatment of vascular diseases. In this review, we give an emphasis on the utilization of hydrogels and their advantages in the various vascular diseases including atherosclerosis, aneurysm, vascular ulcers of the lower limbs and myocardial infarction. Further, we highlight the importance and advantages of hydrogels as artificial microenvironments.
Collapse
Affiliation(s)
- Minhao Li
- School of Intelligent Medicine, China Medical University, No.77, Puhe Road, Shenyang 110122, Liaoning Province, China
| | - Meiqi Jin
- School of Intelligent Medicine, China Medical University, No.77, Puhe Road, Shenyang 110122, Liaoning Province, China
| | - Huazhe Yang
- School of Intelligent Medicine, China Medical University, No.77, Puhe Road, Shenyang 110122, Liaoning Province, China.
| |
Collapse
|
6
|
Zhu Z, Liu Z, Zhang D, Li L, Pei J, Cai L. Models for calcific aortic valve disease in vivo and in vitro. CELL REGENERATION (LONDON, ENGLAND) 2024; 13:6. [PMID: 38424219 PMCID: PMC10904700 DOI: 10.1186/s13619-024-00189-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 02/06/2024] [Indexed: 03/02/2024]
Abstract
Calcific Aortic Valve Disease (CAVD) is prevalent among the elderly as the most common valvular heart disease. Currently, no pharmaceutical interventions can effectively reverse or prevent CAVD, making valve replacement the primary therapeutic recourse. Extensive research spanning decades has contributed to the establishment of animal and in vitro cell models, which facilitates a deeper understanding of the pathophysiological progression and underlying mechanisms of CAVD. In this review, we provide a comprehensive summary and analysis of the strengths and limitations associated with commonly employed models for the study of valve calcification. We specifically emphasize the advancements in three-dimensional culture technologies, which replicate the structural complexity of the valve. Furthermore, we delve into prospective recommendations for advancing in vivo and in vitro model studies of CAVD.
Collapse
Affiliation(s)
- Zijin Zhu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, National & Local Joint Engineering Research Center of High-Throughput Drug Screening Technology, Hubei University, Wuhan, 430062, China
| | - Zhirong Liu
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, National & Local Joint Engineering Research Center of High-Throughput Drug Screening Technology, Hubei University, Wuhan, 430062, China
| | - Donghui Zhang
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, National & Local Joint Engineering Research Center of High-Throughput Drug Screening Technology, Hubei University, Wuhan, 430062, China
| | - Li Li
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, National & Local Joint Engineering Research Center of High-Throughput Drug Screening Technology, Hubei University, Wuhan, 430062, China.
| | - Jianqiu Pei
- State Key Laboratory of Cardiovascular Disease, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China.
- Department of Biochemistry and Molecular Biology, School of Basic Medical Sciences, Beijing Key Laboratory of Metabolic Disorders Related Cardiovascular Disease, Capital Medical University, Beijing, 100069, China.
| | - Lin Cai
- State Key Laboratory of Biocatalysis and Enzyme Engineering, School of Life Science, National & Local Joint Engineering Research Center of High-Throughput Drug Screening Technology, Hubei University, Wuhan, 430062, China.
| |
Collapse
|
7
|
Brodeur A, Roy V, Touzel-Deschênes L, Bianco S, Droit A, Fradette J, Ruel J, Gros-Louis F. Transcriptomic Analysis of Mineralized Adipose-Derived Stem Cell Tissues for Calcific Valve Disease Modelling. Int J Mol Sci 2024; 25:2291. [PMID: 38396969 PMCID: PMC10889332 DOI: 10.3390/ijms25042291] [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: 12/21/2023] [Revised: 02/07/2024] [Accepted: 02/12/2024] [Indexed: 02/25/2024] Open
Abstract
Calcific aortic valve disease (CAVD) is characterized by the fibrosis and mineralization of the aortic valve, which leads to aortic stenosis and heart failure. At the cellular level, this is due to the osteoblastic-like differentiation of valve interstitial cells (VICs), resulting in the calcification of the tissue. Unfortunately, human VICs are not readily available to study CAVD pathogenesis and the implicated mechanisms in vitro; however, adipose-derived stromal/stem cells (ASCs), carrying the patient's specific genomic features, have emerged as a promising cell source to model cardiovascular diseases due to their multipotent nature, availability, and patient-specific characteristics. In this study, we describe a comprehensive transcriptomic analysis of tissue-engineered, scaffold-free, ASC-embedded mineralized tissue sheets using bulk RNA sequencing. Bioinformatic and gene set enrichment analyses revealed the up-regulation of genes associated with the organization of the extracellular matrix (ECM), suggesting that the ECM could play a vital role in the enhanced mineralization observed in these tissue-engineered ASC-embedded sheets. Upon comparison with publicly available gene expression datasets from CAVD patients, striking similarities emerged regarding cardiovascular diseases and ECM functions, suggesting a potential link between ECM gene expression and CAVDs pathogenesis. A matrisome-related sub-analysis revealed the ECM microenvironment promotes the transcriptional activation of the master gene runt-related transcription factor 2 (RUNX2), which is essential in CAVD development. Tissue-engineered ASC-embedded sheets with enhanced mineralization could be a valuable tool for research and a promising avenue for the identification of more effective aortic valve replacement therapies.
Collapse
Affiliation(s)
- Alyssa Brodeur
- Department of Surgery, Faculty of Medicine, Université Laval, Quebec City, QC G1V 5C3, Canada; (A.B.); (V.R.); (L.T.-D.); (J.F.)
- Division of Regenerative Medicine, CHU de Quebec Université Laval Research Centre, Quebec City, QC G1J 5B3, Canada;
| | - Vincent Roy
- Department of Surgery, Faculty of Medicine, Université Laval, Quebec City, QC G1V 5C3, Canada; (A.B.); (V.R.); (L.T.-D.); (J.F.)
- Division of Regenerative Medicine, CHU de Quebec Université Laval Research Centre, Quebec City, QC G1J 5B3, Canada;
| | - Lydia Touzel-Deschênes
- Department of Surgery, Faculty of Medicine, Université Laval, Quebec City, QC G1V 5C3, Canada; (A.B.); (V.R.); (L.T.-D.); (J.F.)
- Division of Regenerative Medicine, CHU de Quebec Université Laval Research Centre, Quebec City, QC G1J 5B3, Canada;
| | - Stéphanie Bianco
- Department of Molecular Medicine, CHU de Quebec Université Laval Research Centre, Quebec City, QC G1V 5C3, Canada; (S.B.); (A.D.)
- Computational Biology Laboratory, CHU de Quebec Université Laval Research Centre, Quebec City, QC G1V 4G2, Canada
| | - Arnaud Droit
- Department of Molecular Medicine, CHU de Quebec Université Laval Research Centre, Quebec City, QC G1V 5C3, Canada; (S.B.); (A.D.)
- Computational Biology Laboratory, CHU de Quebec Université Laval Research Centre, Quebec City, QC G1V 4G2, Canada
| | - Julie Fradette
- Department of Surgery, Faculty of Medicine, Université Laval, Quebec City, QC G1V 5C3, Canada; (A.B.); (V.R.); (L.T.-D.); (J.F.)
- Division of Regenerative Medicine, CHU de Quebec Université Laval Research Centre, Quebec City, QC G1J 5B3, Canada;
| | - Jean Ruel
- Division of Regenerative Medicine, CHU de Quebec Université Laval Research Centre, Quebec City, QC G1J 5B3, Canada;
- Department of Mechanical Engineering, Faculty of Sciences and Engineering, Laval University, Quebec City, QC G1V 0A6, Canada
| | - François Gros-Louis
- Department of Surgery, Faculty of Medicine, Université Laval, Quebec City, QC G1V 5C3, Canada; (A.B.); (V.R.); (L.T.-D.); (J.F.)
- Division of Regenerative Medicine, CHU de Quebec Université Laval Research Centre, Quebec City, QC G1J 5B3, Canada;
| |
Collapse
|
8
|
Ma L, Wu Q, Tam PKH. The Current Proceedings of PSC-Based Liver Fibrosis Therapy. Stem Cell Rev Rep 2023; 19:2155-2165. [PMID: 37490204 DOI: 10.1007/s12015-023-10592-4] [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] [Accepted: 07/12/2023] [Indexed: 07/26/2023]
Abstract
Liver fibrosis was initially considered to be an irreversible process which will eventually lead to the occurrence of liver cancer. So far there has been no effective therapeutic approach to treat liver fibrosis although scientists have put tremendous efforts into the underlying mechanisms of this disease. Therefore, in-depth research on novel and safe treatments of liver fibrosis is of great significance to human health. Pluripotent stem cells (PSCs) play important roles in the study of liver fibrosis due to their unique features in self-renewal ability, pluripotency, and paracrine function. This article mainly reviews the applications of PSCs in the study of liver fibrosis in recent years. We discuss the role of PSC-derived liver organoids in the study of liver fibrosis, and the latest research advances on the differentiation of PSCs into hepatocytes or macrophages. We also highlight the importance of exosomes of PSCs for the treatment of liver fibrosis.
Collapse
Affiliation(s)
- Li Ma
- The State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, China
| | - Qiang Wu
- The State Key Laboratory of Quality Research in Chinese Medicine, Macau University of Science and Technology, Taipa, China.
| | - Paul Kwong-Hang Tam
- Faculty of Medicine, Macau University of Science and Technology, Taipa, China.
| |
Collapse
|
9
|
Salerno N, Panuccio G, Sabatino J, Leo I, Torella M, Sorrentino S, De Rosa S, Torella D. Cellular and Molecular Mechanisms Underlying Tricuspid Valve Development and Disease. J Clin Med 2023; 12:jcm12103454. [PMID: 37240563 DOI: 10.3390/jcm12103454] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2023] [Revised: 05/01/2023] [Accepted: 05/11/2023] [Indexed: 05/28/2023] Open
Abstract
Tricuspid valve (TV) disease is highly prevalent in the general population. For ages considered "the forgotten valve" because of the predominant interest in left-side valve disease, the TV has now received significant attention in recent years, with significant improvement both in diagnosis and in management of tricuspid disease. TV is characterized by complex anatomy, physiology, and pathophysiology, in which the right ventricle plays a fundamental role. Comprehensive knowledge of molecular and cellular mechanisms underlying TV development, TV disease, and tricuspid regurgitation-related right-ventricle cardiomyopathy is necessary to enhance TV disease understanding to improve the ability to risk stratify TR patients, while also predicting valve dysfunction and/or response to tricuspid regurgitation treatment. Scientific efforts are still needed to eventually decipher the complete picture describing the etiopathogenesis of TV and TV-associated cardiomyopathy, and future advances to this aim may be achieved by combining emerging diagnostic imaging modalities with molecular and cellular studies. Overall, basic science studies could help to streamline a new coherent hypothesis underlying both the development of TV during embryogenesis and TV-associated disease and its complications in adult life, providing the conceptual basis for the ultimate and innovative field of valve repair and regeneration using tissue-engineered heart valves.
Collapse
Affiliation(s)
- Nadia Salerno
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Giuseppe Panuccio
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Jolanda Sabatino
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Isabella Leo
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| | - Michele Torella
- Department of Translational Medical Sciences, University of Campania Luigi Vanvitelli, 80138 Naples, Italy
| | - Sabato Sorrentino
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Salvatore De Rosa
- Department of Medical and Surgical Sciences, Magna Graecia University, 88100 Catanzaro, Italy
| | - Daniele Torella
- Department of Experimental and Clinical Medicine, Magna Graecia University, 88100 Catanzaro, Italy
| |
Collapse
|
10
|
Zhang J, Dong H, Jing X, Wang X, Shi Y, He C, Ma B, Nie J, Zhang J, Ma G. Injectable In Situ Photocrosslinked Hydrogel Dressing for Infected Wound Healing. ACS APPLIED BIO MATERIALS 2023; 6:1992-2002. [PMID: 37126576 DOI: 10.1021/acsabm.3c00205] [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: 05/03/2023]
Abstract
A traditional injectable photocrosslinked hydrogel had disadvantages of the residual photoinitiator and toxic crosslinker, slow in situ curing, and a complex preparation process. At the same time, hydrogels cannot act as artificial skin to restore skin sensory function during the wound healing cycle. In this work, an injectable photocrosslinked hydrogel was prepared which can be quickly in situ cured without photoinitiator. Oxidized sodium alginate was used as a natural macromolecular crosslinking agent to form an injectable hydrogel framework with the photosensitive polymer polyvinyl alcohol bearing styrylpyridinium group (PVA-SBQ). In addition, the hydrogel was endowed with photothermal therapy property after the introduction of biomass-like polydopamine particles. When used as a wound dressing, the hydrogel exhibited an excellent antibacterial property, with an antibacterial rate of 99.56% Escherichia coli and 97.96% Staphylococcus aureus. As a result, the hydrogel could significantly accelerate the repair of infected wounds, with a wound healing rate of 96.45% after 14 days. Moreover, the hydrogel exhibited a sensitive and stable sensing property, making it promising to reconstitute the sensory function of damaged skin during treatment. This work provides an idea for the development of injectable photocrosslinked hydrogel dressing.
Collapse
Affiliation(s)
- Jiaxu Zhang
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Huifeng Dong
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Xizhuo Jing
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Xiaoyue Wang
- Department of Gastroenterology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Yunchang Shi
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Chunju He
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Donghua University, Shanghai 201620, P. R. China
| | - Bomou Ma
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials, Donghua University, Shanghai 201620, P. R. China
| | - Jun Nie
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| | - Jie Zhang
- Department of Gastroenterology, Beijing Anzhen Hospital, Capital Medical University, Beijing 100029, China
| | - Guiping Ma
- Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, P. R. China
| |
Collapse
|
11
|
Prakash N, Kim J, Jeon J, Kim S, Arai Y, Bello AB, Park H, Lee SH. Progress and emerging techniques for biomaterial-based derivation of mesenchymal stem cells (MSCs) from pluripotent stem cells (PSCs). Biomater Res 2023; 27:31. [PMID: 37072836 PMCID: PMC10114339 DOI: 10.1186/s40824-023-00371-0] [Citation(s) in RCA: 31] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2023] [Accepted: 03/26/2023] [Indexed: 04/20/2023] Open
Abstract
The use of mesenchymal stem cells (MSCs) for clinical purposes has skyrocketed in the past decade. Their multilineage differentiation potentials and immunomodulatory properties have facilitated the discovery of therapies for various illnesses. MSCs can be isolated from infant and adult tissue sources, which means they are easily available. However, this raises concerns because of the heterogeneity among the various MSC sources, which limits their effective use. Variabilities arise from donor- and tissue-specific differences, such as age, sex, and tissue source. Moreover, adult-sourced MSCs have limited proliferation potentials, which hinders their long-term therapeutic efficacy. These limitations of adult MSCs have prompted researchers to develop a new method for generating MSCs. Pluripotent stem cells (PSCs), such as embryonic stem cells and induced PSCs (iPSCs), can differentiate into various types of cells. Herein, a thorough review of the characteristics, functions, and clinical importance of MSCs is presented. The existing sources of MSCs, including adult- and infant-based sources, are compared. The most recent techniques for deriving MSCs from iPSCs, with a focus on biomaterial-assisted methods in both two- and three-dimensional culture systems, are listed and elaborated. Finally, several opportunities to develop improved methods for efficiently producing MSCs with the aim of advancing their various clinical applications are described.
Collapse
Affiliation(s)
- Nityanand Prakash
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Jiseong Kim
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Jieun Jeon
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Siyeon Kim
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Yoshie Arai
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea
| | - Alvin Bacero Bello
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea.
| | - Hansoo Park
- School of Integrative Engineering, Chung-Ang University, Seoul, 06911, Korea.
| | - Soo-Hong Lee
- Department of Biomedical Engineering, Dongguk University, Seoul, 04620, Korea.
| |
Collapse
|
12
|
Small Extracellular Vesicles Derived from Induced Pluripotent Stem Cells in the Treatment of Myocardial Injury. Int J Mol Sci 2023; 24:ijms24054577. [PMID: 36902008 PMCID: PMC10003569 DOI: 10.3390/ijms24054577] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2023] [Revised: 02/19/2023] [Accepted: 02/21/2023] [Indexed: 03/02/2023] Open
Abstract
Induced pluripotent stem cell (iPSC) therapy brings great hope to the treatment of myocardial injuries, while extracellular vesicles may be one of the main mechanisms of its action. iPSC-derived small extracellular vesicles (iPSCs-sEVs) can carry genetic and proteinaceous substances and mediate the interaction between iPSCs and target cells. In recent years, more and more studies have focused on the therapeutic effect of iPSCs-sEVs in myocardial injury. IPSCs-sEVs may be a new cell-free-based treatment for myocardial injury, including myocardial infarction, myocardial ischemia-reperfusion injury, coronary heart disease, and heart failure. In the current research on myocardial injury, the extraction of sEVs from mesenchymal stem cells induced by iPSCs was widely used. Isolation methods of iPSCs-sEVs for the treatment of myocardial injury include ultracentrifugation, isodensity gradient centrifugation, and size exclusion chromatography. Tail vein injection and intraductal administration are the most widely used routes of iPSCs-sEV administration. The characteristics of sEVs derived from iPSCs which were induced from different species and organs, including fibroblasts and bone marrow, were further compared. In addition, the beneficial genes of iPSC can be regulated through CRISPR/Cas9 to change the composition of sEVs and improve the abundance and expression diversity of them. This review focused on the strategies and mechanisms of iPSCs-sEVs in the treatment of myocardial injury, which provides a reference for future research and the application of iPSCs-sEVs.
Collapse
|
13
|
Albert BJ, Butcher JT. Future prospects in the tissue engineering of heart valves: a focus on the role of stem cells. Expert Opin Biol Ther 2023; 23:553-564. [PMID: 37171790 PMCID: PMC10461076 DOI: 10.1080/14712598.2023.2214313] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2023] [Accepted: 05/11/2023] [Indexed: 05/13/2023]
Abstract
INTRODUCTION Heart valve disease is a growing burden on the healthcare system. Current solutions are insufficient for young patients and do not offer relief from reintervention. Tissue engineered heart valves (TEHVs) offer a solution that grows and responds to the native environment in a similar way to a healthy valve. Stem cells hold potential to populate these valves as a malleable source that can adapt to environmental cues. AREAS COVERED This review covers current methods of recapitulating features of native heart valves with tissue engineering through use of stem cell populations with in situ and in vitro methods. EXPERT OPINION In the field of TEHVs, we see a variety of approaches in cell source, biomaterial, and maturation methods. Choosing appropriate cell populations may be very patient specific; consistency and predictability will be key to long-term success. In situ methods are closer to translation but struggle with consistent cellularization. In vitro culture requires specialized methods but may recapitulate native valve cell populations with higher fidelity. Understanding how cell populations react to valve conditions and immune response is vital for success. Detrimental valve pathologies have proven to be difficult to avoid in early translation attempts.
Collapse
Affiliation(s)
- Benjamin J Albert
- Cornell University, Meinig School of Biomedical Engineering, Ithaca, NY, USA
| | - Jonathan T Butcher
- Cornell University, Meinig School of Biomedical Engineering, Ithaca, NY, USA
| |
Collapse
|
14
|
Geevarghese R, Sajjadi SS, Hudecki A, Sajjadi S, Jalal NR, Madrakian T, Ahmadi M, Włodarczyk-Biegun MK, Ghavami S, Likus W, Siemianowicz K, Łos MJ. Biodegradable and Non-Biodegradable Biomaterials and Their Effect on Cell Differentiation. Int J Mol Sci 2022; 23:ijms232416185. [PMID: 36555829 PMCID: PMC9785373 DOI: 10.3390/ijms232416185] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 12/05/2022] [Accepted: 12/09/2022] [Indexed: 12/23/2022] Open
Abstract
Biomaterials for tissue scaffolds are key components in modern tissue engineering and regenerative medicine. Targeted reconstructive therapies require a proper choice of biomaterial and an adequate choice of cells to be seeded on it. The introduction of stem cells, and the transdifferentiation procedures, into regenerative medicine opened a new era and created new challenges for modern biomaterials. They must not only fulfill the mechanical functions of a scaffold for implanted cells and represent the expected mechanical strength of the artificial tissue, but furthermore, they should also assure their survival and, if possible, affect their desired way of differentiation. This paper aims to review how modern biomaterials, including synthetic (i.e., polylactic acid, polyurethane, polyvinyl alcohol, polyethylene terephthalate, ceramics) and natural (i.e., silk fibroin, decellularized scaffolds), both non-biodegradable and biodegradable, could influence (tissue) stem cells fate, regulate and direct their differentiation into desired target somatic cells.
Collapse
Affiliation(s)
- Rency Geevarghese
- Biotechnology Center, Silesian University of Technology, 44-100 Gliwice, Poland
| | - Seyedeh Sara Sajjadi
- School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1971653313, Iran
| | - Andrzej Hudecki
- Łukasiewicz Network-Institute of Non-Ferrous Metals, 44-121 Gliwice, Poland
| | - Samad Sajjadi
- School of Allied Medical Sciences, Shahid Beheshti University of Medical Sciences, Tehran 1971653313, Iran
| | | | - Tayyebeh Madrakian
- Faculty of Chemistry, Bu-Ali Sina University, Hamedan 6516738695, Iran
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| | - Mazaher Ahmadi
- Faculty of Chemistry, Bu-Ali Sina University, Hamedan 6516738695, Iran
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
| | - Małgorzata K. Włodarczyk-Biegun
- Biotechnology Center, Silesian University of Technology, 44-100 Gliwice, Poland
- Polymer Science, Zernike Institute for Advanced Materials, University of Groningen, Nijenborgh 4, 9747 AG Groningen, The Netherlands
| | - Saeid Ghavami
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Department of Human Anatomy and Cell Science, University of Manitoba College of Medicine, Winnipeg, MB R3E 0V9, Canada
- Research Institutes of Oncology and Hematology, Cancer Care Manitoba-University of Manitoba, Winnipeg, MB R3E 0V9, Canada
- Biology of Breathing Theme, Children Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 0V9, Canada
- Faculty of Medicine in Zabrze, University of Technology in Katowice, 41-800 Zabrze, Poland
| | - Wirginia Likus
- Department of Anatomy, Faculty of Health Sciences in Katowice, Medical University of Silesia, 40-752 Katowice, Poland
| | - Krzysztof Siemianowicz
- Department of Biochemistry, Faculty of Medicine in Katowice, Medical University of Silesia, 40-752 Katowice, Poland
- Correspondence: (K.S.); (M.J.Ł.); Tel.: +48-32-237-2913 (M.J.Ł.)
| | - Marek J. Łos
- Biotechnology Center, Silesian University of Technology, 44-100 Gliwice, Poland
- Autophagy Research Center, Shiraz University of Medical Sciences, Shiraz 7134845794, Iran
- Correspondence: (K.S.); (M.J.Ł.); Tel.: +48-32-237-2913 (M.J.Ł.)
| |
Collapse
|
15
|
Mechanical stretching of 3D hydrogels for neural stem cell differentiation. Biodes Manuf 2022. [DOI: 10.1007/s42242-022-00209-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
|
16
|
Walker CJ, Batan D, Bishop CT, Ramirez D, Aguado BA, Schroeder ME, Crocini C, Schwisow J, Moulton K, Macdougall L, Weiss RM, Allen MA, Dowell R, Leinwand LA, Anseth KS. Extracellular matrix stiffness controls cardiac valve myofibroblast activation through epigenetic remodeling. Bioeng Transl Med 2022; 7:e10394. [PMID: 36176599 PMCID: PMC9472021 DOI: 10.1002/btm2.10394] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2021] [Revised: 07/11/2022] [Accepted: 07/27/2022] [Indexed: 11/29/2022] Open
Abstract
Aortic valve stenosis (AVS) is a progressive fibrotic disease that is caused by thickening and stiffening of valve leaflets. At the cellular level, quiescent valve interstitial cells (qVICs) activate to myofibroblasts (aVICs) that persist within the valve tissue. Given the persistence of myofibroblasts in AVS, epigenetic mechanisms have been implicated. Here, we studied changes that occur in VICs during myofibroblast activation by using a hydrogel matrix to recapitulate different stiffnesses in the valve leaflet during fibrosis. We first compared the chromatin landscape of qVICs cultured on soft hydrogels and aVICs cultured on stiff hydrogels, representing the native and diseased phenotypes respectively. Using assay for transposase-accessible chromatin sequencing (ATAC-Seq), we found that open chromatin regions in aVICs were enriched for transcription factor binding motifs associated with mechanosensing pathways compared to qVICs. Next, we used RNA-Seq to show that the open chromatin regions in aVICs correlated with pro-fibrotic gene expression, as aVICs expressed higher levels of contractile fiber genes, including myofibroblast markers such as alpha smooth muscle actin (αSMA), compared to qVICs. In contrast, chromatin remodeling genes were downregulated in aVICs compared to qVICs, indicating qVICs may be protected from myofibroblast activation through epigenetic mechanisms. Small molecule inhibition of one of these remodelers, CREB Binding Protein (CREBBP), prevented qVICs from activating to aVICs. Notably, CREBBP is more abundant in valves from healthy patients compared to fibrotic valves. Our findings reveal the role of mechanical regulation in chromatin remodeling during VIC activation and quiescence and highlight one potential therapeutic target for treating AVS.
Collapse
Affiliation(s)
- Cierra J. Walker
- Materials Science and Engineering ProgramUniversity of Colorado BoulderBoulderColoradoUSA
- BioFrontiers Institute, University of Colorado BoulderBoulderColoradoUSA
| | - Dilara Batan
- BioFrontiers Institute, University of Colorado BoulderBoulderColoradoUSA
- Biochemistry DepartmentUniversity of Colorado BoulderBoulderColoradoUSA
| | - Carrie T. Bishop
- Chemical and Biological Engineering DepartmentUniversity of Colorado BoulderBoulderColoradoUSA
| | - Daniel Ramirez
- BioFrontiers Institute, University of Colorado BoulderBoulderColoradoUSA
- Molecular, Cellular, and Developmental Biology DepartmentUniversity of Colorado BoulderBoulderColoradoUSA
| | - Brian A. Aguado
- BioFrontiers Institute, University of Colorado BoulderBoulderColoradoUSA
- Chemical and Biological Engineering DepartmentUniversity of Colorado BoulderBoulderColoradoUSA
| | - Megan E. Schroeder
- BioFrontiers Institute, University of Colorado BoulderBoulderColoradoUSA
- Chemical and Biological Engineering DepartmentUniversity of Colorado BoulderBoulderColoradoUSA
| | - Claudia Crocini
- BioFrontiers Institute, University of Colorado BoulderBoulderColoradoUSA
- Molecular, Cellular, and Developmental Biology DepartmentUniversity of Colorado BoulderBoulderColoradoUSA
| | - Jessica Schwisow
- Division of CardiologyUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Karen Moulton
- Division of CardiologyUniversity of Colorado Anschutz Medical CampusAuroraColoradoUSA
| | - Laura Macdougall
- BioFrontiers Institute, University of Colorado BoulderBoulderColoradoUSA
- Chemical and Biological Engineering DepartmentUniversity of Colorado BoulderBoulderColoradoUSA
| | - Robert M. Weiss
- Department of Internal MedicineUniversity of IowaIowa CityIowaUSA
| | - Mary A. Allen
- BioFrontiers Institute, University of Colorado BoulderBoulderColoradoUSA
- Molecular, Cellular, and Developmental Biology DepartmentUniversity of Colorado BoulderBoulderColoradoUSA
| | - Robin Dowell
- BioFrontiers Institute, University of Colorado BoulderBoulderColoradoUSA
- Molecular, Cellular, and Developmental Biology DepartmentUniversity of Colorado BoulderBoulderColoradoUSA
| | - Leslie A. Leinwand
- BioFrontiers Institute, University of Colorado BoulderBoulderColoradoUSA
- Molecular, Cellular, and Developmental Biology DepartmentUniversity of Colorado BoulderBoulderColoradoUSA
| | - Kristi S. Anseth
- BioFrontiers Institute, University of Colorado BoulderBoulderColoradoUSA
- Chemical and Biological Engineering DepartmentUniversity of Colorado BoulderBoulderColoradoUSA
| |
Collapse
|
17
|
Chalard AE, Dixon AW, Taberner AJ, Malmström J. Visible-Light Stiffness Patterning of GelMA Hydrogels Towards In Vitro Scar Tissue Models. Front Cell Dev Biol 2022; 10:946754. [PMID: 35865624 PMCID: PMC9294371 DOI: 10.3389/fcell.2022.946754] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Accepted: 06/20/2022] [Indexed: 11/30/2022] Open
Abstract
Variations in mechanical properties of the extracellular matrix occurs in various processes, such as tissue fibrosis. The impact of changes in tissue stiffness on cell behaviour are studied in vitro using various types of biomaterials and methods. Stiffness patterning of hydrogel scaffolds, through the use of stiffness gradients for instance, allows the modelling and studying of cellular responses to fibrotic mechanisms. Gelatine methacryloyl (GelMA) has been used extensively in tissue engineering for its inherent biocompatibility and the ability to precisely tune its mechanical properties. Visible light is now increasingly employed for crosslinking GelMA hydrogels as it enables improved cell survival when performing cell encapsulation. We report here, the photopatterning of mechanical properties of GelMA hydrogels with visible light and eosin Y as the photoinitiator using physical photomasks and projection with a digital micromirror device. Using both methods, binary hydrogels with areas of different stiffnesses and hydrogels with stiffness gradients were fabricated. Their mechanical properties were characterised using force indentation with atomic force microscopy, which showed the efficiency of both methods to spatially pattern the elastic modulus of GelMA according to the photomask or the projected pattern. Crosslinking through projection was also used to build constructs with complex shapes. Overall, this work shows the feasibility of patterning the stiffness of GelMA scaffolds, in the range from healthy to pathological stiffness, with visible light. Consequently, this method could be used to build in vitro models of healthy and fibrotic tissue and study the cellular behaviours involved at the interface between the two.
Collapse
Affiliation(s)
- Anaïs E. Chalard
- Department of Chemical and Materials Engineering, Faculty of Engineering, The University of Auckland, Auckland, New Zealand
- The MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington, New Zealand
- *Correspondence: Anaïs E. Chalard, ; Jenny Malmström,
| | - Alexander W. Dixon
- The Auckland Bioengineering Institute (ABI), The University of Auckland, Auckland, New Zealand
| | - Andrew J. Taberner
- The Auckland Bioengineering Institute (ABI), The University of Auckland, Auckland, New Zealand
- Department of Engineering Science, Faculty of Engineering, The University of Auckland, Auckland, New Zealand
| | - Jenny Malmström
- Department of Chemical and Materials Engineering, Faculty of Engineering, The University of Auckland, Auckland, New Zealand
- The MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington, New Zealand
- *Correspondence: Anaïs E. Chalard, ; Jenny Malmström,
| |
Collapse
|
18
|
Zhang L, Ma XJN, Fei YY, Han HT, Xu J, Cheng L, Li X. Stem cell therapy in liver regeneration: Focus on mesenchymal stem cells and induced pluripotent stem cells. Pharmacol Ther 2022; 232:108004. [PMID: 34597754 DOI: 10.1016/j.pharmthera.2021.108004] [Citation(s) in RCA: 36] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2021] [Revised: 08/11/2021] [Accepted: 09/23/2021] [Indexed: 02/07/2023]
Abstract
The liver has the ability to repair itself after injury; however, a variety of pathological changes in the liver can affect its ability to regenerate, and this could lead to liver failure. Mesenchymal stem cells (MSCs) are considered a good source of cells for regenerative medicine, as they regulate liver regeneration through different mechanisms, and their efficacy has been demonstrated by many animal experiments and clinical studies. Induced pluripotent stem cells, another good source of MSCs, have also made great progress in the establishment of organoids, such as liver disease models, and in drug screening. Owing to the recent developments in MSCs and induced pluripotent stem cells, combined with emerging technologies including graphene, nano-biomaterials, and gene editing, precision medicine and individualized clinical treatment may be realized in the near future.
Collapse
Affiliation(s)
- Lu Zhang
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, PR China; Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, PR China; The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China
| | - Xiao-Jing-Nan Ma
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China
| | - Yuan-Yuan Fei
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, PR China; Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, PR China
| | - Heng-Tong Han
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China
| | - Jun Xu
- The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China
| | - Lu Cheng
- Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, PR China
| | - Xun Li
- Department of General Surgery, The First Hospital of Lanzhou University, Lanzhou 730000, PR China; Key Laboratory Biotherapy and Regenerative Medicine of Gansu Province, Lanzhou 730000, PR China; Medical Frontier Innovation Research Center, The First Hospital of Lanzhou University, Lanzhou 730000, PR China; Hepatopancreatobiliary Surgery Institute of Gansu Province, Lanzhou 730000, PR China; The First School of Clinical Medicine, Lanzhou University, Lanzhou 730000, PR China.
| |
Collapse
|
19
|
Rekowska N, Huling J, Brietzke A, Arbeiter D, Eickner T, Konasch J, Riess A, Mau R, Seitz H, Grabow N, Teske M. Thermal, Mechanical and Biocompatibility Analyses of Photochemically Polymerized PEGDA 250 for Photopolymerization-Based Manufacturing Processes. Pharmaceutics 2022; 14:628. [PMID: 35336002 PMCID: PMC8951438 DOI: 10.3390/pharmaceutics14030628] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2022] [Revised: 03/03/2022] [Accepted: 03/08/2022] [Indexed: 01/06/2023] Open
Abstract
Novel fabrication techniques based on photopolymerization enable the preparation of complex multi-material constructs for biomedical applications. This requires an understanding of the influence of the used reaction components on the properties of the generated copolymers. The identification of fundamental characteristics of these copolymers is necessary to evaluate their potential for biomaterial applications. Additionally, knowledge of the properties of the starting materials enables subsequent tailoring of the biomaterials to meet individual implantation needs. In our study, we have analyzed the biological, chemical, mechanical and thermal properties of photopolymerized poly(ethyleneglycol) diacrylate (PEGDA) and specific copolymers with different photoinitiator (PI) concentrations before and after applying a post treatment washing process. As comonomers, 1,3-butanediol diacrylate, pentaerythritol triacrylate and pentaerythritol tetraacrylate were used. The in vitro studies confirm the biocompatibility of all investigated copolymers. Uniaxial tensile tests show significantly lower tensile strength (82% decrease) and elongation at break (76% decrease) values for washed samples. Altered tensile strength is also observed for different PI concentrations: on average, 6.2 MPa for 1.25% PI and 3.1 MPa for 0.5% PI. The addition of comonomers lowers elongation at break on average by 45%. Moreover, our observations show glass transition temperatures (Tg) ranging from 27 °C to 56 °C, which significantly increase with higher comonomer content. These results confirm the ability to generate biocompatible PEGDA copolymers with specific thermal and mechanical properties. These can be considered as resins for various additive manufacturing-based applications to obtain personalized medical devices, such as drug delivery systems (DDS). Therefore, our study has advanced the understanding of PEGDA multi-materials and will contribute to the future development of tools ensuring safe and effective individual therapy for patients.
Collapse
Affiliation(s)
- Natalia Rekowska
- Institute for Biomedical Engineering, University Medical Center Rostock, Friedrich-Barnewitz-Straße 4, 18119 Rostock, Germany; (J.H.); (A.B.); (D.A.); (T.E.); (N.G.); (M.T.)
| | - Jennifer Huling
- Institute for Biomedical Engineering, University Medical Center Rostock, Friedrich-Barnewitz-Straße 4, 18119 Rostock, Germany; (J.H.); (A.B.); (D.A.); (T.E.); (N.G.); (M.T.)
| | - Andreas Brietzke
- Institute for Biomedical Engineering, University Medical Center Rostock, Friedrich-Barnewitz-Straße 4, 18119 Rostock, Germany; (J.H.); (A.B.); (D.A.); (T.E.); (N.G.); (M.T.)
| | - Daniela Arbeiter
- Institute for Biomedical Engineering, University Medical Center Rostock, Friedrich-Barnewitz-Straße 4, 18119 Rostock, Germany; (J.H.); (A.B.); (D.A.); (T.E.); (N.G.); (M.T.)
| | - Thomas Eickner
- Institute for Biomedical Engineering, University Medical Center Rostock, Friedrich-Barnewitz-Straße 4, 18119 Rostock, Germany; (J.H.); (A.B.); (D.A.); (T.E.); (N.G.); (M.T.)
| | - Jan Konasch
- Microfluidics, Faculty of Mechanical Engineering and Marine Technology, University of Rostock, Justus-von-Liebig Weg 6, 18059 Rostock, Germany; (J.K.); (A.R.); (R.M.); (H.S.)
| | - Alexander Riess
- Microfluidics, Faculty of Mechanical Engineering and Marine Technology, University of Rostock, Justus-von-Liebig Weg 6, 18059 Rostock, Germany; (J.K.); (A.R.); (R.M.); (H.S.)
| | - Robert Mau
- Microfluidics, Faculty of Mechanical Engineering and Marine Technology, University of Rostock, Justus-von-Liebig Weg 6, 18059 Rostock, Germany; (J.K.); (A.R.); (R.M.); (H.S.)
| | - Hermann Seitz
- Microfluidics, Faculty of Mechanical Engineering and Marine Technology, University of Rostock, Justus-von-Liebig Weg 6, 18059 Rostock, Germany; (J.K.); (A.R.); (R.M.); (H.S.)
- Department LL&M, Interdisciplinary Faculty, University of Rostock, Albert-Einstein-Str. 25, 18059 Rostock, Germany
| | - Niels Grabow
- Institute for Biomedical Engineering, University Medical Center Rostock, Friedrich-Barnewitz-Straße 4, 18119 Rostock, Germany; (J.H.); (A.B.); (D.A.); (T.E.); (N.G.); (M.T.)
- Department LL&M, Interdisciplinary Faculty, University of Rostock, Albert-Einstein-Str. 25, 18059 Rostock, Germany
| | - Michael Teske
- Institute for Biomedical Engineering, University Medical Center Rostock, Friedrich-Barnewitz-Straße 4, 18119 Rostock, Germany; (J.H.); (A.B.); (D.A.); (T.E.); (N.G.); (M.T.)
| |
Collapse
|
20
|
Human Induced Pluripotent Stem Cell as a Disease Modeling and Drug Development Platform-A Cardiac Perspective. Cells 2021; 10:cells10123483. [PMID: 34943991 PMCID: PMC8699880 DOI: 10.3390/cells10123483] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2021] [Revised: 12/03/2021] [Accepted: 12/06/2021] [Indexed: 02/07/2023] Open
Abstract
A comprehensive understanding of the pathophysiology and cellular responses to drugs in human heart disease is limited by species differences between humans and experimental animals. In addition, isolation of human cardiomyocytes (CMs) is complicated because cells obtained by biopsy do not proliferate to provide sufficient numbers of cells for preclinical studies in vitro. Interestingly, the discovery of human-induced pluripotent stem cell (hiPSC) has opened up the possibility of generating and studying heart disease in a culture dish. The combination of reprogramming and genome editing technologies to generate a broad spectrum of human heart diseases in vitro offers a great opportunity to elucidate gene function and mechanisms. However, to exploit the potential applications of hiPSC-derived-CMs for drug testing and studying adult-onset cardiac disease, a full functional characterization of maturation and metabolic traits is required. In this review, we focus on methods to reprogram somatic cells into hiPSC and the solutions for overcome immaturity of the hiPSC-derived-CMs to mimic the structure and physiological properties of the adult human CMs to accurately model disease and test drug safety. Finally, we discuss how to improve the culture, differentiation, and purification of CMs to obtain sufficient numbers of desired types of hiPSC-derived-CMs for disease modeling and drug development platform.
Collapse
|
21
|
Parisi C, Qin K, Fernandes FM. Colonization versus encapsulation in cell-laden materials design: porosity and process biocompatibility determine cellularization pathways. PHILOSOPHICAL TRANSACTIONS. SERIES A, MATHEMATICAL, PHYSICAL, AND ENGINEERING SCIENCES 2021; 379:20200344. [PMID: 34334019 DOI: 10.1098/rsta.2020.0344] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/28/2021] [Indexed: 06/13/2023]
Abstract
Seeding materials with living cells has been-and still is-one of the most promising approaches to reproduce the complexity and the functionality of living matter. The strategies to associate living cells with materials are limited to cell encapsulation and colonization, however, the requirements for these two approaches have been seldom discussed systematically. Here we propose a simple two-dimensional map based on materials' pore size and the cytocompatibility of their fabrication process to draw, for the first time, a guide to building cellularized materials. We believe this approach may serve as a straightforward guideline to design new, more relevant materials, able to seize the complexity and the function of biological materials. This article is part of the theme issue 'Bio-derived and bioinspired sustainable advanced materials for emerging technologies (part 1)'.
Collapse
Affiliation(s)
- Cleo Parisi
- Laboratoire de Chimie de la Matière Condensée de Paris, Sorbonne Université, UMR7574, 4 Place Jussieu, 75005 Paris, France
| | - Kankan Qin
- Laboratoire de Chimie de la Matière Condensée de Paris, Sorbonne Université, UMR7574, 4 Place Jussieu, 75005 Paris, France
| | - Francisco M Fernandes
- Laboratoire de Chimie de la Matière Condensée de Paris, Sorbonne Université, UMR7574, 4 Place Jussieu, 75005 Paris, France
| |
Collapse
|
22
|
Tayler IM, Stowers RS. Engineering hydrogels for personalized disease modeling and regenerative medicine. Acta Biomater 2021; 132:4-22. [PMID: 33882354 DOI: 10.1016/j.actbio.2021.04.020] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2020] [Revised: 03/26/2021] [Accepted: 04/12/2021] [Indexed: 02/06/2023]
Abstract
Technological innovations and advances in scientific understanding have created an environment where data can be collected, analyzed, and interpreted at scale, ushering in the era of personalized medicine. The ability to isolate cells from individual patients offers tremendous promise if those cells can be used to generate functional tissue replacements or used in disease modeling to determine optimal treatment strategies. Here, we review recent progress in the use of hydrogels to create artificial cellular microenvironments for personalized tissue engineering and regenerative medicine applications, as well as to develop personalized disease models. We highlight engineering strategies to control stem cell fate through hydrogel design, and the use of hydrogels in combination with organoids, advanced imaging methods, and novel bioprinting techniques to generate functional tissues. We also discuss the use of hydrogels to study molecular mechanisms underlying diseases and to create personalized in vitro disease models to complement existing pre-clinical models. Continued progress in the development of engineered hydrogels, in combination with other emerging technologies, will be essential to realize the immense potential of personalized medicine. STATEMENT OF SIGNIFICANCE: In this review, we cover recent advances in hydrogel engineering strategies with applications in personalized medicine. Specifically, we focus on material systems to expand or control differentiation of patient-derived stem cells, and hydrogels to reprogram somatic cells to pluripotent states. We then review applications of hydrogels in developing personalized engineered tissues. We also highlight the use of hydrogel systems as personalized disease models, focusing on specific examples in fibrosis and cancer, and more broadly on drug screening strategies using patient-derived cells and hydrogels. We believe this review will be a valuable contribution to the Special Issue and the readership of Acta Biomaterialia will appreciate the comprehensive overview of the utility of hydrogels in the developing field of personalized medicine.
Collapse
|
23
|
Dupuis V, Oltra E. Methods to produce induced pluripotent stem cell-derived mesenchymal stem cells: Mesenchymal stem cells from induced pluripotent stem cells. World J Stem Cells 2021; 13:1094-1111. [PMID: 34567428 PMCID: PMC8422924 DOI: 10.4252/wjsc.v13.i8.1094] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Revised: 05/03/2021] [Accepted: 07/14/2021] [Indexed: 02/06/2023] Open
Abstract
Mesenchymal stem cells (MSCs) have received significant attention in recent years due to their large potential for cell therapy. Indeed, they secrete a wide variety of immunomodulatory factors of interest for the treatment of immune-related disorders and inflammatory diseases. MSCs can be extracted from multiple tissues of the human body. However, several factors may restrict their use for clinical applications: the requirement of invasive procedures for their isolation, their limited numbers, and their heterogeneity according to the tissue of origin or donor. In addition, MSCs often present early signs of replicative senescence limiting their expansion in vitro, and their therapeutic capacity in vivo. Due to the clinical potential of MSCs, a considerable number of methods to differentiate induced pluripotent stem cells (iPSCs) into MSCs have emerged. iPSCs represent a new reliable, unlimited source to generate MSCs (MSCs derived from iPSC, iMSCs) from homogeneous and well-characterized cell lines, which would relieve many of the above mentioned technical and biological limitations. Additionally, the use of iPSCs prevents some of the ethical concerns surrounding the use of human embryonic stem cells. In this review, we analyze the main current protocols used to differentiate human iPSCs into MSCs, which we classify into five different categories: MSC Switch, Embryoid Body Formation, Specific Differentiation, Pathway Inhibitor, and Platelet Lysate. We also evaluate common and method-specific culture components and provide a list of positive and negative markers for MSC characterization. Further guidance on material requirements to produce iMSCs with these methods and on the phenotypic features of the iMSCs obtained is added. The information may help researchers identify protocol options to design and/or refine standardized procedures for large-scale production of iMSCs fitting clinical demands.
Collapse
Affiliation(s)
- Victoria Dupuis
- Faculté des Sciences et d’Ingénierie, Sorbonne Université, Paris 75252, France
| | - Elisa Oltra
- Department of Pathology, Universidad Católica de Valencia San Vicente Mártir, Valencia 46001, Spain
- Centro de Investigación Traslacional San Alberto Magno, Universidad Católica de Valencia San Vicente Mártir, Valencia 46001, Spain.
| |
Collapse
|
24
|
Hamilton M, Harrington S, Dhar P, Stehno-Bittel L. Hyaluronic Acid Hydrogel Microspheres for Slow Release Stem Cell Delivery. ACS Biomater Sci Eng 2021; 7:3754-3763. [PMID: 34323078 DOI: 10.1021/acsbiomaterials.1c00658] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Cell therapies are hampered by a lack of available delivery systems, resulting in inconsistent outcomes in animal studies and human clinical trials. Hydrogel encapsulants offer a broad range of tunable characteristics in the design of cell delivery vehicles. The focus of the hydrogel field has been on durable encapsulants that provide long-term paracrine function of the cells. However, some cell therapies require cell-to-cell contact in order to elicit their effect. Controlled release microencapsulants would be beneficial in these situations, but appropriate polymers have not been adaptable to microsphere manufacturing because they harden too slowly. We developed and tested a novel microencapsulant formulation (acrylated hyaluronic acid: AHA) with degradation characteristics as a controlled release cell delivery vehicle. The properties of AHA microspheres were evaluated and compared to those of poly(ethylene glycol) diacrylate (PEGDA), a durable hydrogel. AHA microspheres possessed a higher swelling ratio, lower diffusion barrier, faster degradation rate, a lower storage modulus, and a larger average diameter than microspheres composed of PEGDA. Additionally, in vitro cell viability and release and short-term in vivo biocompatibility in immune competent Sprague-Dawley rats was assessed for each microsphere type. Compared to PEGDA, microspheres composed of AHA resulted in significantly less foreign body response in vivo as measured by a lack of cellularity or fibrotic ring in the surrounding tissue and no cellular infiltration into the microsphere. This study illustrates the potential of AHA microspheres as a degradable cell delivery system with superior encapsulated cell viability and biocompatibility with the surrounding tissue.
Collapse
Affiliation(s)
- Megan Hamilton
- University of Kansas Bioengineering Program, 1132 Learned Hall, 1530 West 15th Street, Lawrence, Kansas 66045, United States
| | - Stephen Harrington
- Likarda LLC, 10330 Hickman Mills Drive, Suite B, Kansas City, Missouri 64137, United States
| | - Prajnaparamita Dhar
- University of Kansas Bioengineering Program, 1132 Learned Hall, 1530 West 15th Street, Lawrence, Kansas 66045, United States.,Department of Chemical and Petroleum Engineering, The University of Kansas, 4132 Learned Hall, 1530 West 15th Street, Lawrence, Kansas 66045, United States
| | - Lisa Stehno-Bittel
- Likarda LLC, 10330 Hickman Mills Drive, Suite B, Kansas City, Missouri 64137, United States
| |
Collapse
|
25
|
Abstract
Calcific aortic valve disease sits at the confluence of multiple world-wide epidemics of aging, obesity, diabetes, and renal dysfunction, and its prevalence is expected to nearly triple over the next 3 decades. This is of particularly dire clinical relevance, as calcific aortic valve disease can progress rapidly to aortic stenosis, heart failure, and eventually premature death. Unlike in atherosclerosis, and despite the heavy clinical toll, to date, no pharmacotherapy has proven effective to halt calcific aortic valve disease progression, with invasive and costly aortic valve replacement representing the only treatment option currently available. This substantial gap in care is largely because of our still-limited understanding of both normal aortic valve biology and the key regulatory mechanisms that drive disease initiation and progression. Drug discovery is further hampered by the inherent intricacy of the valvular microenvironment: a unique anatomic structure, a complex mixture of dynamic biomechanical forces, and diverse and multipotent cell populations collectively contributing to this currently intractable problem. One promising and rapidly evolving tactic is the application of multiomics approaches to fully define disease pathogenesis. Herein, we summarize the application of (epi)genomics, transcriptomics, proteomics, and metabolomics to the study of valvular heart disease. We also discuss recent forays toward the omics-based characterization of valvular (patho)biology at single-cell resolution; these efforts promise to shed new light on cellular heterogeneity in healthy and diseased valvular tissues and represent the potential to efficaciously target and treat key cell subpopulations. Last, we discuss systems biology- and network medicine-based strategies to extract meaning, mechanisms, and prioritized drug targets from multiomics datasets.
Collapse
Affiliation(s)
- Mark C. Blaser
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| | - Simon Kraler
- Center for Molecular Cardiology, University of Zurich, Schlieren, CH
| | - Thomas F. Lüscher
- Center for Molecular Cardiology, University of Zurich, Schlieren, CH
- Heart Division, Royal Brompton & Harefield Hospitals, London, UK
- National Heart and Lung Institute, Imperial College, London, UK
| | - Elena Aikawa
- Center for Interdisciplinary Cardiovascular Sciences, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
- Center for Excellence in Vascular Biology, Cardiovascular Division, Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, USA
| |
Collapse
|
26
|
Augustine R, Dan P, Hasan A, Khalaf IM, Prasad P, Ghosal K, Gentile C, McClements L, Maureira P. Stem cell-based approaches in cardiac tissue engineering: controlling the microenvironment for autologous cells. Biomed Pharmacother 2021; 138:111425. [PMID: 33756154 DOI: 10.1016/j.biopha.2021.111425] [Citation(s) in RCA: 41] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2020] [Revised: 02/08/2021] [Accepted: 02/21/2021] [Indexed: 02/07/2023] Open
Abstract
Cardiovascular disease is one of the leading causes of mortality worldwide. Cardiac tissue engineering strategies focusing on biomaterial scaffolds incorporating cells and growth factors are emerging as highly promising for cardiac repair and regeneration. The use of stem cells within cardiac microengineered tissue constructs present an inherent ability to differentiate into cell types of the human heart. Stem cells derived from various tissues including bone marrow, dental pulp, adipose tissue and umbilical cord can be used for this purpose. Approaches ranging from stem cell injections, stem cell spheroids, cell encapsulation in a suitable hydrogel, use of prefabricated scaffold and bioprinting technology are at the forefront in the field of cardiac tissue engineering. The stem cell microenvironment plays a key role in the maintenance of stemness and/or differentiation into cardiac specific lineages. This review provides a detailed overview of the recent advances in microengineering of autologous stem cell-based tissue engineering platforms for the repair of damaged cardiac tissue. A particular emphasis is given to the roles played by the extracellular matrix (ECM) in regulating the physiological response of stem cells within cardiac tissue engineering platforms.
Collapse
Affiliation(s)
- Robin Augustine
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713, Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713, Doha, Qatar.
| | - Pan Dan
- Department of Cardiovascular and Transplantation Surgery, Regional Central Hospital of Nancy, Lorraine University, Nancy 54500, France; Department of Thoracic and Cardiovascular Surgery, Zhongnan Hospital of Wuhan University, Wuhan 430071, China
| | - Anwarul Hasan
- Department of Mechanical and Industrial Engineering, College of Engineering, Qatar University, 2713, Doha, Qatar; Biomedical Research Center (BRC), Qatar University, PO Box 2713, Doha, Qatar.
| | | | - Parvathy Prasad
- International and Inter University Center for Nanoscience and Nanotechnology, Mahatma Gandhi University, Kottayam, Kerala 686560, India
| | - Kajal Ghosal
- Dr. B. C. Roy College of Pharmacy and AHS, Durgapur 713206, India
| | - Carmine Gentile
- School of Biomedical Engineering, Faculty of Engineering and IT, University of Technology Sydney, NSW 2007, Australia; School of Medicine, Faculty of Medicine and Health, University of Sydney, NSW 2000, Australia; Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA
| | - Lana McClements
- School of Life Sciences, Faculty of Science, University of Technology Sydney, NSW 2007, Australia
| | - Pablo Maureira
- Department of Cardiovascular and Transplantation Surgery, Regional Central Hospital of Nancy, Lorraine University, Nancy 54500, France
| |
Collapse
|
27
|
Teixeira MO, Antunes JC, Felgueiras HP. Recent Advances in Fiber-Hydrogel Composites for Wound Healing and Drug Delivery Systems. Antibiotics (Basel) 2021; 10:248. [PMID: 33801438 PMCID: PMC8001440 DOI: 10.3390/antibiotics10030248] [Citation(s) in RCA: 43] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2021] [Revised: 02/23/2021] [Accepted: 02/25/2021] [Indexed: 12/19/2022] Open
Abstract
In the last decades, much research has been done to fasten wound healing and target-direct drug delivery. Hydrogel-based scaffolds have been a recurrent solution in both cases, with some reaching already the market, even though their mechanical stability remains a challenge. To overcome this limitation, reinforcement of hydrogels with fibers has been explored. The structural resemblance of fiber-hydrogel composites to natural tissues has been a driving force for the optimization and exploration of these systems in biomedicine. Indeed, the combination of hydrogel-forming techniques and fiber spinning approaches has been crucial in the development of scaffolding systems with improved mechanical strength and medicinal properties. In this review, a comprehensive overview of the recently developed fiber-hydrogel composite strategies for wound healing and drug delivery is provided. The methodologies employed in fiber and hydrogel formation are also highlighted, together with the most compatible polymer combinations, as well as drug incorporation approaches creating stimuli-sensitive and triggered drug release towards an enhanced host response.
Collapse
Affiliation(s)
| | | | - Helena P. Felgueiras
- Centre for Textile Science and Technology (2C2T), Department of Textile Engineering, University of Minho, Campus of Azurém, 4800-058 Guimarães, Portugal; (M.O.T.); (J.C.A.)
| |
Collapse
|
28
|
Blacutt J, Lan Z, Cosgriff-Hernandez EM, Gordon VD. Quantitative confocal microscopy and calibration for measuring differences in cyclic-di-GMP signalling by bacteria on biomedical hydrogels. ROYAL SOCIETY OPEN SCIENCE 2021; 8:201453. [PMID: 33614081 PMCID: PMC7890475 DOI: 10.1098/rsos.201453] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/14/2020] [Accepted: 11/30/2020] [Indexed: 06/12/2023]
Abstract
The growth of bacterial biofilms on implanted medical devices causes harmful infections and device failure. Biofilm development initiates when bacteria attach to and sense a surface. For the common nosocomial pathogen Pseudomonas aeruginosa and many others, the transition to the biofilm phenotype is controlled by the intracellular signal and second messenger cyclic-di-GMP (c-di-GMP). It is not known how biomedical materials might be adjusted to impede c-di-GMP signalling, and there are few extant methods for conducting such studies. Here, we develop such a method. We allowed P. aeruginosa to attach to the surfaces of poly(ethylene glycol) diacrylate (PEGDA) hydrogels. These bacteria contained a plasmid for a green fluorescent protein (GFP) reporter for c-di-GMP. We used laser-scanning confocal microscopy to measure the dynamics of the GFP reporter for 3 h, beginning 1 h after introducing bacteria to the hydrogel. We controlled for the effects of changes in bacterial metabolism using a promoterless plasmid for GFP, and for the effects of light passing through different hydrogels being differently attenuated by using fluorescent plastic beads as 'standard candles' for calibration. We demonstrate that this method can measure statistically significant differences in c-di-GMP signalling associated with different PEGDA gel types and with the surface-exposed protein PilY1.
Collapse
Affiliation(s)
- Jacob Blacutt
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX, USA
- Center for Nonlinear Dynamics, The University of Texas at Austin, Austin, TX, USA
| | - Ziyang Lan
- Department of Biomedical Engineering, The University of Texas at Austin, Austin, TX, USA
| | | | - Vernita D. Gordon
- Institute for Cellular and Molecular Biology, The University of Texas at Austin, Austin, TX, USA
- Center for Nonlinear Dynamics, The University of Texas at Austin, Austin, TX, USA
- Department of Physics, The University of Texas at Austin, Austin, TX, USA
- LaMontagne Center for Infectious Disease, The University of Texas at Austin, Austin, TX, USA
| |
Collapse
|
29
|
Weeratunga P, Shahsavari A, Fennis E, Wolvetang EJ, Ovchinnikov DA, Whitworth DJ. Induced Pluripotent Stem Cell-Derived Mesenchymal Stem Cells from the Tasmanian Devil ( Sarcophilus harrisii) Express Immunomodulatory Factors and a Tropism Toward Devil Facial Tumor Cells. Stem Cells Dev 2020; 29:25-37. [PMID: 31709909 DOI: 10.1089/scd.2019.0203] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
Marsupials have long attracted scientific interest because of their unique biological features and their position in mammalian evolution. Mesenchymal stem cells (MSCs) are of considerable research interest in translational medicine due to their immunomodulatory, anti-inflammatory, and regenerative properties. MSCs have been harvested from various tissues in numerous eutherian species; however, there are no descriptions of MSCs derived from a marsupial. In this study, we have generated Tasmanian devil (Sarcophilus harrisii) MSCs from devil induced pluripotent stem cells (iPSCs), thus providing an unlimited source of devil MSCs and circumventing the need to harvest tissues from live animals. Devil iPSCs were differentiated into MSCs (iMSCs) through both embryoid body formation assays (EB-iMSCs) and through inhibition of the transforming growth factor beta/activin signaling pathway (SB-iMSCs). Both EB-iMSCs and SB-iMSCs are highly proliferative and express the MSC-specific surface proteins CD73, CD90, and CD105, in addition to the pluripotency transcription factors OCT4/POU5F1, SOX2, and NANOG. Expression of the marsupial pluripotency factor POU5F3, a paralogue of OCT4/POU5F1, is significantly reduced in association with the transition from pluripotency to multipotency. Devil iMSCs readily differentiate along the adipogenic, osteogenic, and chondrogenic pathways in vitro, confirming their trilineage differentiation potential. Importantly, in vitro teratoma assays confirmed their multipotency, rather than pluripotency, since the iMSCs only formed derivatives of the mesodermal germ layer. Devil iMSCs show a tropism toward medium conditioned by devil facial tumor cells and express a range of immunomodulatory and anti-inflammatory factors. Therefore, devil iMSCs will be a valuable tool for further studies on marsupial biology and may facilitate the development of an MSC-based treatment strategy against Devil Facial Tumor Disease.
Collapse
Affiliation(s)
- Prasanna Weeratunga
- School of Veterinary Science, The University of Queensland, Gatton, Australia
| | - Arash Shahsavari
- School of Veterinary Science, The University of Queensland, Gatton, Australia
| | - Evelien Fennis
- Faculty of Veterinary Medicine, Utrecht University, Utrecht, Netherlands
| | - Ernst J Wolvetang
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Australia
| | - Dmitry A Ovchinnikov
- Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Australia.,StemCore, The University of Queensland, St. Lucia, Australia
| | - Deanne J Whitworth
- School of Veterinary Science, The University of Queensland, Gatton, Australia.,Australian Institute for Bioengineering and Nanotechnology, The University of Queensland, St. Lucia, Australia
| |
Collapse
|
30
|
Bartoli-Leonard F, Aikawa E. Heart Valve Disease: Challenges and New Opportunities. Front Cardiovasc Med 2020; 7:602271. [PMID: 33195488 PMCID: PMC7642276 DOI: 10.3389/fcvm.2020.602271] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2020] [Accepted: 09/09/2020] [Indexed: 01/23/2023] Open
Affiliation(s)
- Francesca Bartoli-Leonard
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States
| | - Elena Aikawa
- Division of Cardiovascular Medicine, Department of Medicine, Center for Interdisciplinary Cardiovascular Sciences, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States.,Division of Cardiovascular Medicine, Department of Medicine, Center for Excellence in Vascular Biology, Harvard Medical School, Brigham and Women's Hospital, Boston, MA, United States.,Department of Human Pathology, Sechenov First Moscow State Medical University, Moscow, Russia
| |
Collapse
|
31
|
Tomal W, Ortyl J. Water-Soluble Photoinitiators in Biomedical Applications. Polymers (Basel) 2020; 12:E1073. [PMID: 32392892 PMCID: PMC7285382 DOI: 10.3390/polym12051073] [Citation(s) in RCA: 105] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2020] [Revised: 05/02/2020] [Accepted: 05/03/2020] [Indexed: 12/25/2022] Open
Abstract
Light-initiated polymerization processes are currently an important tool in various industrial fields. The advancement of technology has resulted in the use of photopolymerization in various biomedical applications, such as the production of 3D hydrogel structures, the encapsulation of cells, and in drug delivery systems. The use of photopolymerization processes requires an appropriate initiating system that, in biomedical applications, must meet additional criteria such as high water solubility, non-toxicity to cells, and compatibility with visible low-power light sources. This article is a literature review on those compounds that act as photoinitiators of photopolymerization processes in biomedical applications. The division of initiators according to the method of photoinitiation was described and the related mechanisms were discussed. Examples from each group of photoinitiators are presented, and their benefits, limitations, and applications are outlined.
Collapse
Affiliation(s)
- Wiktoria Tomal
- Faculty of Chemical Engineering and Technology, Krakow University of Technology, Warszawska 24, 31-155 Krakow, Poland;
| | - Joanna Ortyl
- Faculty of Chemical Engineering and Technology, Krakow University of Technology, Warszawska 24, 31-155 Krakow, Poland;
- Photo HiTech Ltd., Bobrzyńskiego 14, 30-348 Krakow, Poland
| |
Collapse
|
32
|
Han WT, Jang T, Chen S, Chong LSH, Jung HD, Song J. Improved cell viability for large-scale biofabrication with photo-crosslinkable hydrogel systems through a dual-photoinitiator approach. Biomater Sci 2020; 8:450-461. [PMID: 31748767 DOI: 10.1039/c9bm01347d] [Citation(s) in RCA: 39] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Biofabrication with various hydrogel systems allows the production of tissue or organ constructs in vitro to address various challenges in healthcare and medicine. In particular, photocrosslinkable hydrogels have great advantages such as excellent spatial and temporal selectivity and low processing cost and energy requirements. However, inefficient polymerization kinetics of commercialized photoinitiators upon exposure to UV-A radiation or visible light increase processing time, often compromising cell viability. In this study, we developed a hydrogel crosslinking system which exhibited efficient crosslinking properties and desired mechanical properties with high cell viability, through a dual-photoinitiator approach. Through the co-existence of Irgacure 2959 and VA-086, the overall crosslinking process was completed with a minimal UV dosage during a significantly reduced crosslinking time, producing mechanically robust hydrogel constructs, while most encapsulated cells within the hydrogel constructs remained viable. Moreover, we fabricated a large PEGDA hydrogel construct with a single microchannel as a proof of concept for hydrogels with vasculature to demonstrate the versatility of the system. Our dual-photoinitiator approach allowed the production of this photocrosslinkable hydrogel system with microchannels, significantly improving cell viability and processing efficiency, yet maintaining good mechanical stability. Taken together, we envision the concurrent use of photoinitiators, Irgacure 2959 and VA-086, opening potential avenues for the utilization of various photocrosslinkable hydrogel systems in perfusable large artificial tissue for in vivo and ex vivo applications with improved processing efficiency and cell viability.
Collapse
Affiliation(s)
- Win Tun Han
- School of Chemical and Biomedical Engineering, Nanyang Technological University, 70 Nanyang Drive, 637457, Singapore.
| | | | | | | | | | | |
Collapse
|
33
|
Nachlas ALY, Li S, Streeter BW, De Jesus Morales KJ, Sulejmani F, Madukauwa-David DI, Bejleri D, Sun W, Yoganathan AP, Davis ME. A multilayered valve leaflet promotes cell-laden collagen type I production and aortic valve hemodynamics. Biomaterials 2020; 240:119838. [PMID: 32092591 DOI: 10.1016/j.biomaterials.2020.119838] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 01/27/2020] [Accepted: 01/31/2020] [Indexed: 12/18/2022]
Abstract
Patients with aortic heart valve disease are limited to valve replacements that lack the ability to grow and remodel. This presents a major challenge for pediatric patients who require a valve capable of somatic growth and at a smaller size. A patient-specific heart valve capable of growth and remodeling while maintaining proper valve function would address this major issue. Here, we recreate the native valve leaflet structure composed of poly-ε-caprolactone (PCL) and cell-laden gelatin-methacrylate/poly (ethylene glycol) diacrylate (GelMA/PEGDA) hydrogels using 3D printing and molding, and then evaluate the ability of the multilayered scaffold to produce collagen matrix under physiological shear stress conditions. We also characterized the valve hemodynamics under aortic physiological flow conditions. The valve's fibrosa layer was replicated by 3D printing PCL in a circumferential direction similar to collagen alignment in the native leaflet, and GelMA/PEGDA sustained and promoted cell viability in the spongiosa/ventricularis layers. We found that collagen type I production can be increased in the multilayered scaffold when it is exposed to pulsatile shear stress conditions over static conditions. When the PCL component was mounted onto a valve ring and tested under physiological aortic valve conditions, the hemodynamics were comparable to commercially available valves. Our results demonstrate that a structurally representative valve leaflet can be generated using 3D printing and that the PCL layer of the leaflet can sustain proper valve function under physiological aortic valve conditions.
Collapse
Affiliation(s)
- Aline L Y Nachlas
- Wallace H Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Siyi Li
- Wallace H Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Benjamin W Streeter
- Wallace H Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Kenneth J De Jesus Morales
- Wallace H Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Fatiesa Sulejmani
- Wallace H Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - David Immanuel Madukauwa-David
- Bioengineering Graduate Program, Georgia Institute of Technology, Atlanta, GA, USA; George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Donald Bejleri
- Wallace H Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Wei Sun
- Wallace H Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Ajit P Yoganathan
- Wallace H Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA
| | - Michael E Davis
- Wallace H Coulter Department of Biomedical Engineering, Emory University and Georgia Institute of Technology, Atlanta, GA, USA; Children's Heart Research & Outcomes (HeRO) Center, Children's Healthcare of Atlanta & Emory University, Atlanta, GA, USA.
| |
Collapse
|
34
|
Paik DT, Chandy M, Wu JC. Patient and Disease-Specific Induced Pluripotent Stem Cells for Discovery of Personalized Cardiovascular Drugs and Therapeutics. Pharmacol Rev 2020; 72:320-342. [PMID: 31871214 PMCID: PMC6934989 DOI: 10.1124/pr.116.013003] [Citation(s) in RCA: 131] [Impact Index Per Article: 26.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Human induced pluripotent stem cells (iPSCs) have emerged as an effective platform for regenerative therapy, disease modeling, and drug discovery. iPSCs allow for the production of limitless supply of patient-specific somatic cells that enable advancement in cardiovascular precision medicine. Over the past decade, researchers have developed protocols to differentiate iPSCs to multiple cardiovascular lineages, as well as to enhance the maturity and functionality of these cells. Despite significant advances, drug therapy and discovery for cardiovascular disease have lagged behind other fields such as oncology. We speculate that this paucity of drug discovery is due to a previous lack of efficient, reproducible, and translational model systems. Notably, existing drug discovery and testing platforms rely on animal studies and clinical trials, but investigations in animal models have inherent limitations due to interspecies differences. Moreover, clinical trials are inherently flawed by assuming that all individuals with a disease will respond identically to a therapy, ignoring the genetic and epigenomic variations that define our individuality. With ever-improving differentiation and phenotyping methods, patient-specific iPSC-derived cardiovascular cells allow unprecedented opportunities to discover new drug targets and screen compounds for cardiovascular disease. Imbued with the genetic information of an individual, iPSCs will vastly improve our ability to test drugs efficiently, as well as tailor and titrate drug therapy for each patient.
Collapse
Affiliation(s)
- David T Paik
- Stanford Cardiovascular Institute, Stanford University, Stanford, California
| | - Mark Chandy
- Stanford Cardiovascular Institute, Stanford University, Stanford, California
| | - Joseph C Wu
- Stanford Cardiovascular Institute, Stanford University, Stanford, California
| |
Collapse
|
35
|
Simon LR, Masters KS. Disease-inspired tissue engineering: Investigation of cardiovascular pathologies. ACS Biomater Sci Eng 2019; 6:2518-2532. [PMID: 32974421 DOI: 10.1021/acsbiomaterials.9b01067] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Once focused exclusively on the creation of tissues to repair or replace diseased or damaged organs, the field of tissue engineering has undergone an important evolution in recent years. Namely, tissue engineering techniques are increasingly being applied to intentionally generate pathological conditions. Motivated in part by the wide gap between 2D cultures and animal models in the current disease modeling continuum, disease-inspired tissue-engineered platforms have numerous potential applications, and may serve to advance our understanding and clinical treatment of various diseases. This review will focus on recent progress toward generating tissue-engineered models of cardiovascular diseases, including cardiac hypertrophy, fibrosis, and ischemia reperfusion injury, atherosclerosis, and calcific aortic valve disease, with an emphasis on how these disease-inspired platforms can be used to decipher disease etiology. Each pathology is discussed in the context of generating both disease-specific cells as well as disease-specific extracellular environments, with an eye toward future opportunities to integrate different tools to yield more complex and physiologically relevant culture platforms. Ultimately, the development of effective disease treatments relies upon our ability to develop appropriate experimental models; as cardiovascular diseases are the leading cause of death worldwide, the insights yielded by improved in vitro disease modeling could have substantial ramifications for public health and clinical care.
Collapse
Affiliation(s)
- LaTonya R Simon
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705
| | - Kristyn S Masters
- Department of Biomedical Engineering, University of Wisconsin-Madison, Madison, WI 53705.,Department of Medicine, University of Wisconsin-Madison, Madison, WI 53705
| |
Collapse
|
36
|
Martín-Martín Y, Fernández-García L, Sanchez-Rebato MH, Marí-Buyé N, Rojo FJ, Pérez-Rigueiro J, Ramos M, Guinea GV, Panetsos F, González-Nieto D. Evaluation of Neurosecretome from Mesenchymal Stem Cells Encapsulated in Silk Fibroin Hydrogels. Sci Rep 2019; 9:8801. [PMID: 31217546 PMCID: PMC6584675 DOI: 10.1038/s41598-019-45238-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2019] [Accepted: 05/31/2019] [Indexed: 12/13/2022] Open
Abstract
Physical and cognitive disabilities are hallmarks of a variety of neurological diseases. Stem cell-based therapies are promising solutions to neuroprotect and repair the injured brain and overcome the limited capacity of the central nervous system to recover from damage. It is widely accepted that most benefits of different exogenously transplanted stem cells rely on the secretion of different factors and biomolecules that modulate inflammation, cell death and repair processes in the damaged host tissue. However, few cells survive in cerebral tissue after transplantation, diminishing the therapeutic efficacy. As general rule, cell encapsulation in natural and artificial polymers increases the in vivo engraftment of the transplanted cells. However, we have ignored the consequences of such encapsulation on the secretory activity of these cells. In this study, we investigated the biological compatibility between silk fibroin hydrogels and stem cells of mesenchymal origin, a cell population that has gained increasing attention and popularity in regenerative medicine. Although the survival of mesenchymal stem cells was not affected inside hydrogels, this biomaterial format caused adhesion and proliferation deficits and impaired secretion of several angiogenic, chemoattractant and neurogenic factors while concurrently potentiating the anti-inflammatory capacity of this cell population through a massive release of TGF-Beta-1. Our results set a milestone for the exploration of engineering polymers to modulate the secretory activity of stem cell-based therapies for neurological disorders.
Collapse
Affiliation(s)
| | | | - Miguel H Sanchez-Rebato
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain
- Neurocomputing and Neurorobotics Research Group: Faculty of Biology and Faculty of Optics, Universidad Complutense de Madrid., Madrid, Spain
- Brain Plasticity Group. Health Research Institute of the Hospital Clínico San Carlos (IdISSC), Madrid, Spain
- GReD, UMR CNRS 6293 - INSERM U1103 - Université Clermont Auvergne, Faculté de Medicine, Clermont-Ferrand, France
| | - Núria Marí-Buyé
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain
- Departamento de Ciencia de Materiales. ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, Madrid, Spain
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Francisco J Rojo
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain
- Departamento de Ciencia de Materiales. ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, Madrid, Spain
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - José Pérez-Rigueiro
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain
- Departamento de Ciencia de Materiales. ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, Madrid, Spain
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Milagros Ramos
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain
- Departamento de Tecnología Fotónica y Bioingeniería. ETSI Telecomunicaciones, Universidad Politécnica de Madrid, Madrid, Spain
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Gustavo V Guinea
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain
- Departamento de Ciencia de Materiales. ETSI Caminos, Canales y Puertos, Universidad Politécnica de Madrid, Madrid, Spain
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain
| | - Fivos Panetsos
- Neurocomputing and Neurorobotics Research Group: Faculty of Biology and Faculty of Optics, Universidad Complutense de Madrid., Madrid, Spain
- Brain Plasticity Group. Health Research Institute of the Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Daniel González-Nieto
- Center for Biomedical Technology, Universidad Politécnica de Madrid, Madrid, Spain.
- Departamento de Tecnología Fotónica y Bioingeniería. ETSI Telecomunicaciones, Universidad Politécnica de Madrid, Madrid, Spain.
- Biomedical Research Networking Center in Bioengineering Biomaterials and Nanomedicine (CIBER-BBN), Madrid, Spain.
| |
Collapse
|
37
|
Electrospun Poly(p-dioxanone)/Poly(ester-urethane)ureas Composite Nanofibers for Potential Heart Valve Tissue Reconstruction. CHINESE JOURNAL OF POLYMER SCIENCE 2019. [DOI: 10.1007/s10118-019-2231-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
38
|
Trac D, Maxwell JT, Brown ME, Xu C, Davis ME. Aggregation of Child Cardiac Progenitor Cells Into Spheres Activates Notch Signaling and Improves Treatment of Right Ventricular Heart Failure. Circ Res 2019; 124:526-538. [PMID: 30590978 PMCID: PMC6375764 DOI: 10.1161/circresaha.118.313845] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
RATIONALE Congenital heart disease can lead to life-threatening right ventricular (RV) heart failure. Results from clinical trials support expanding cardiac progenitor cell (CPC) based therapies. However, our recent data show that CPCs lose function as they age, starting as early as 1 year. OBJECTIVE To determine whether the aggregation of child (1-5-year-old) CPCs into scaffold-free spheres can improve differentiation by enhancing Notch signaling, a known regulator of CPC fate. We hypothesized that aggregated (3-dimensional [3D]) CPCs will repair RV heart failure better than monolayer (2-dimensional [2D]) CPCs. METHODS AND RESULTS Spheres were produced with 1500 CPCs each using a microwell array. CPC aggregation significantly increased gene expression of Notch1 compared with 2D CPCs, accompanied by significant upregulation of cardiogenic transcription factors (GATA4, HAND1, MEF2C, NKX2.5, and TBX5) and endothelial markers (CD31, FLK1, FLT1, VWF). Blocking Notch receptor activation with the γ-secretase inhibitor DAPT (N-[N-(3,5-difluorophenacetyl)-L-alanyl]-S-phenylglycine t-butyl ester) diminished these effects. To evaluate the therapeutic improvements of CPC aggregation, RV heart failure was induced in athymic rats by pulmonary artery banding, and cells were implanted into the RV free wall. Echocardiographic measurements 28 days postimplantation showed significantly improved RV function with 3D compared with 2D CPCs. Tracking implanted CPCs via DiR (1,1'-dioctadecyl-3,3,3',3'-tetramethylindotricarbocyanine iodide)-labeling showed improved retention of 3D CPCs. Transducing 3D CPCs with Notch1-shRNA (short hairpin RNA) did not reduce retention, but significantly reduced RV functional improvements. Histological analyses showed 3D treatment reduced RV fibrosis and increased angiogenesis. Although 3D CPCs formed CD31+ vessel-like cells in vivo, these effects are more likely because of improved 3D CPC exosome function compared with 2D CPC exosomes. CONCLUSIONS Spherical aggregation improves child CPC function in a Notch-dependent manner. The strong reparative ability of CPC spheres warrants further investigation as a treatment for pediatric heart failure, especially in older children where reparative ability may be reduced.
Collapse
Affiliation(s)
- David Trac
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, 30322, USA
| | - Joshua T. Maxwell
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, 30322, USA
| | - Milton E. Brown
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, 30322, USA
| | - Chunhui Xu
- Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, 30322, USA;,Children’s Heart Research & Outcomes (HeRO) Center, Children’s Healthcare of Atlanta & Emory University, Atlanta, Georgia, 30322, USA
| | - Michael E. Davis
- Wallace H. Coulter Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, Georgia, 30322, USA;,Division of Pediatric Cardiology, Department of Pediatrics, Emory University School of Medicine, Atlanta, Georgia, 30322, USA;,Children’s Heart Research & Outcomes (HeRO) Center, Children’s Healthcare of Atlanta & Emory University, Atlanta, Georgia, 30322, USA
| |
Collapse
|
39
|
Jover E, Fagnano M, Angelini G, Madeddu P. Cell Sources for Tissue Engineering Strategies to Treat Calcific Valve Disease. Front Cardiovasc Med 2018; 5:155. [PMID: 30460245 PMCID: PMC6232262 DOI: 10.3389/fcvm.2018.00155] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2018] [Accepted: 10/10/2018] [Indexed: 12/15/2022] Open
Abstract
Cardiovascular calcification is an independent risk factor and an established predictor of adverse cardiovascular events. Despite concomitant factors leading to atherosclerosis and heart valve disease (VHD), the latter has been identified as an independent pathological entity. Calcific aortic valve stenosis is the most common form of VDH resulting of either congenital malformations or senile “degeneration.” About 2% of the population over 65 years is affected by aortic valve stenosis which represents a major cause of morbidity and mortality in the elderly. A multifactorial, complex and active heterotopic bone-like formation process, including extracellular matrix remodeling, osteogenesis and angiogenesis, drives heart valve “degeneration” and calcification, finally causing left ventricle outflow obstruction. Surgical heart valve replacement is the current therapeutic option for those patients diagnosed with severe VHD representing more than 20% of all cardiac surgeries nowadays. Tissue Engineering of Heart Valves (TEHV) is emerging as a valuable alternative for definitive treatment of VHD and promises to overcome either the chronic oral anticoagulation or the time-dependent deterioration and reintervention of current mechanical or biological prosthesis, respectively. Among the plethora of approaches and stablished techniques for TEHV, utilization of different cell sources may confer of additional properties, desirable and not, which need to be considered before moving from the bench to the bedside. This review aims to provide a critical appraisal of current knowledge about calcific VHD and to discuss the pros and cons of the main cell sources tested in studies addressing in vitro TEHV.
Collapse
Affiliation(s)
- Eva Jover
- Bristol Medical School (Translational Health Sciences), Bristol Heart Institute, University of Bristol, Bristol, United Kingdom
| | - Marco Fagnano
- Bristol Medical School (Translational Health Sciences), Bristol Heart Institute, University of Bristol, Bristol, United Kingdom
| | - Gianni Angelini
- Bristol Medical School (Translational Health Sciences), Bristol Heart Institute, University of Bristol, Bristol, United Kingdom
| | - Paolo Madeddu
- Bristol Medical School (Translational Health Sciences), Bristol Heart Institute, University of Bristol, Bristol, United Kingdom
| |
Collapse
|
40
|
Malandraki-Miller S, Lopez CA, Al-Siddiqi H, Carr CA. Changing Metabolism in Differentiating Cardiac Progenitor Cells-Can Stem Cells Become Metabolically Flexible Cardiomyocytes? Front Cardiovasc Med 2018; 5:119. [PMID: 30283788 PMCID: PMC6157401 DOI: 10.3389/fcvm.2018.00119] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 08/10/2018] [Indexed: 12/15/2022] Open
Abstract
The heart is a metabolic omnivore and the adult heart selects the substrate best suited for each circumstance, with fatty acid oxidation preferred in order to fulfill the high energy demand of the contracting myocardium. The fetal heart exists in an hypoxic environment and obtains the bulk of its energy via glycolysis. After birth, the "fetal switch" to oxidative metabolism of glucose and fatty acids has been linked to the loss of the regenerative phenotype. Various stem cell types have been used in differentiation studies, but most are cultured in high glucose media. This does not change in the majority of cardiac differentiation protocols. Despite the fact that metabolic state affects marker expression and cellular function and activity, the substrate composition is currently being overlooked. In this review we discuss changes in cardiac metabolism during development, the various protocols used to differentiate progenitor cells to cardiomyocytes, what is known about stem cell metabolism and how consideration of metabolism can contribute toward maturation of stem cell-derived cardiomyocytes.
Collapse
Affiliation(s)
| | | | | | - Carolyn A. Carr
- Department of Physiology, Anatomy and Genetics, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
41
|
Xue D, Wang Y, Zhang J, Mei D, Wang Y, Chen S. Projection-Based 3D Printing of Cell Patterning Scaffolds with Multiscale Channels. ACS APPLIED MATERIALS & INTERFACES 2018; 10:19428-19435. [PMID: 29782142 DOI: 10.1021/acsami.8b03867] [Citation(s) in RCA: 59] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/08/2023]
Abstract
To fully actualize artificial, cell-laden biological models in tissue engineering, such as 3D organoids and organs-on-a-chip systems, cells need to be patterned such that they can precisely mimic natural microenvironments in vitro. Despite increasing interest in this area, patterning cells at multiscale (∼10 μm to 10 mm) remains a significant challenge in bioengineering. Here, we report a projection-based 3D printing system that achieves rapid and high-resolution fabrication of hydrogel scaffolds featuring intricate channels for multiscale cell patterning. Using this system, we were able to use biocompatible poly(ethylene glycol)diacrylate in fabricating a variety of scaffold architectures, ranging from regular geometries such as serpentine, spiral, and fractal-like to more irregular/intricate geometries, such as biomimetic arborescent and capillary networks. A red food dye solution was able to freely fill all channels in the scaffolds, from the trunk (>1100 μm in width) to the small branch (∼17 μm in width) without an external pump. The dimensions of the printed scaffolds remained stable over 3 days while being immersed in Dulbecco's phosphate-buffered saline at 37 °C, and a penetration analysis revealed that these scaffolds are suitable for metabolic and nutrient transport. Cell patterning experiments showed that red fluorescent protein-transfected A549 human nonsmall lung cancer cells adhered well in the scaffolds' channels, and showed further attachment and penetration during cell culture proliferation.
Collapse
Affiliation(s)
- Dai Xue
- Department of NanoEngineering , University of California , San Diego , California 92093 , United States
| | | | - Jiaxin Zhang
- Department of Toxicology , Fourth Military Medical University , Xi'an 710032 , China
| | | | | | - Shaochen Chen
- Department of NanoEngineering , University of California , San Diego , California 92093 , United States
| |
Collapse
|