1
|
Esmaeili H, Zhang Y, Ravi K, Neff K, Zhu W, Migrino RQ, Park JG, Nikkhah M. Development of an electroconductive Heart-on-a-chip model to investigate cellular and molecular response of human cardiac tissue to gold nanomaterials. Biomaterials 2025; 320:123275. [PMID: 40138961 DOI: 10.1016/j.biomaterials.2025.123275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 02/16/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025]
Abstract
To date, various strategies have been developed to construct biomimetic and functional in vitro cardiac tissue models utilizing human induced pluripotent stem cells (hiPSCs). Among these approaches, microfluidic-based Heart-on-a-chip (HOC) models are promising, as they enable the engineering of miniaturized, physiologically relevant in vitro cardiac tissues with precise control over cellular constituents and tissue architecture. Despite significant advancements, previously reported HOC models often lack the electroconductivity features of the native human myocardium. In this study, we developed a 3D electroconductive HOC (referred to as eHOC) model through the co-culture of isogenic hiPSC-derived cardiomyocytes (hiCMs) and cardiac fibroblasts (hiCFs), embedded within an electroconductive hydrogel scaffold in a microfluidic-based chip system. Functional and gene expression analyses demonstrated that, compared to non-conductive HOC, the eHOC model exhibited enhanced contractile functionality, improved calcium transients, and increased expression of structural and calcium handling genes. The eHOC model was further leveraged to investigate the underlying electroconduction-induced pathway(s) associated with cardiac tissue development through single-cell RNA sequencing (scRNA-seq). Notably, scRNA-seq analyses revealed a significant downregulation of a set of cardiac genes, associated with the fetal stage of heart development, as well as upregulation of sarcomere- and conduction-related genes within the eHOC model. Additionally, upregulation of the cardiac muscle contraction and motor protein pathways were observed in the eHOC model, consistent with enhanced contractile functionality of the engineered cardiac tissues. Comparison of scRNA-seq data from the 3D eHOC model with published datasets of adult human hearts demonstrated a similar expression pattern of fetal- and adult-like cardiac genes. Overall, this study provides a unique eHOC model with improved biomimcry and organotypic features, which could be potentially used for drug testing and discovery, as well as disease modeling applications.
Collapse
Affiliation(s)
- Hamid Esmaeili
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Yining Zhang
- Center for Personalized Diagnostics (CPD), Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA
| | - Kalpana Ravi
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Keagan Neff
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA
| | - Wuqiang Zhu
- Department of Cardiovascular Medicine, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic, Scottsdale, AZ, 85259, USA
| | - Raymond Q Migrino
- Phoenix Veterans Affairs Health Care System, Phoenix, AZ, 85022, USA; University of Arizona College of Medicine, Phoenix, AZ, 85004, USA
| | - Jin G Park
- Center for Personalized Diagnostics (CPD), Biodesign Institute, Arizona State University, Tempe, AZ, 85287, USA
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, 85287, USA; Biodesign Virginia G. Piper Center for Personalized Diagnosis, Arizona State University, Tempe, AZ, 85287, USA.
| |
Collapse
|
2
|
Roshanbinfar K, Evans AD, Samanta S, Kolesnik-Gray M, Fiedler M, Krstic V, Engel FB, Oommen OP. Enhancing biofabrication: Shrink-resistant collagen-hyaluronan composite hydrogel for tissue engineering and 3D bioprinting applications. Biomaterials 2025; 318:123174. [PMID: 39951830 DOI: 10.1016/j.biomaterials.2025.123174] [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: 09/12/2024] [Revised: 01/21/2025] [Accepted: 02/05/2025] [Indexed: 02/16/2025]
Abstract
Biofabrication represents a promising technique for creating tissues for regeneration or as models for drug testing. Collagen-based hydrogels are widely used as suitable matrix owing to their biocompatibility and tunable mechanical properties. However, one major challenge is that the encapsulated cells interact with the collagen matrix causing construct shrinkage. Here, we present a hydrogel with high shape fidelity, mimicking the major components of the extracellular matrix. We engineered a composite hydrogel comprising gallic acid (GA)-functionalized hyaluronic acid (HA), collagen I, and HA-coated multiwall carbon nanotubes (MWCNT). This hydrogel supports cell encapsulation, exhibits shear-thinning properties enhancing injectability and printability, and importantly significantly mitigates shrinkage when loaded with human fibroblasts compared to collagen I hydrogels (∼20 % vs. > 90 %). 3D-bioprinted rings utilizing human fibroblast-loaded inks maintain their shape over 7 days in culture. Furthermore, inclusion of HAGA into collagen I hydrogels increases mechanical stiffness, radical scavenging capability, and tissue adhesiveness. Notably, the here developed hydrogel is also suitable for human induced pluripotent stem cell-derived cardiomyocytes and allows printing of functional heart ventricles responsive to pharmacological treatment. Cardiomyocytes behave similar in the newly developed hydrogels compared to collagen I, based on survival, sarcomere appearance, and calcium handling. Collectively, we developed a novel material to overcome the challenge of post-fabrication matrix shrinkage conferring high shape fidelity.
Collapse
Affiliation(s)
- Kaveh Roshanbinfar
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology and Department of Cardiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Austin Donnelly Evans
- Bioengineering and Nanomedicine Group, Faculty of Medicine and Health Technologies, Tampere University, 33720, Tampere, Finland
| | - Sumanta Samanta
- Bioengineering and Nanomedicine Group, Faculty of Medicine and Health Technologies, Tampere University, 33720, Tampere, Finland
| | - Maria Kolesnik-Gray
- Department of Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Staudtstr. 7, 91058, Erlangen, Germany
| | - Maren Fiedler
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology and Department of Cardiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Vojislav Krstic
- Department of Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Staudtstr. 7, 91058, Erlangen, Germany; Department of Physics, Wake-Forest-University, Winston Salem, NC, 27109, USA
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology and Department of Cardiology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany.
| | - Oommen P Oommen
- School of Pharmacy and Pharmaceutical Sciences, Cardiff University, King Edward VII Avenue, Cardiff, CF10 3NB, UK.
| |
Collapse
|
3
|
Ng XJ, Esser TU, Trossmann VT, Rudisch C, Fiedler M, Roshanbinfar K, Lamberger Z, Stahlhut P, Lang G, Scheibel T, Engel FB. Enhancing Form Stability: Shrink-Resistant Hydrogels Made of Interpenetrating Networks of Recombinant Spider Silk and Collagen-I. Adv Healthc Mater 2025; 14:e2500311. [PMID: 40143764 PMCID: PMC12057611 DOI: 10.1002/adhm.202500311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 03/09/2025] [Indexed: 03/28/2025]
Abstract
Tissue engineering enables the production of tissues and organ-like structures as models for drug testing and mechanistical studies or functional replacements for injured tissues. Available cytocompatible materials are limited in number, suffer from insufficient mechanical properties, and cells interacting with them often cause construct shrinkage. As shape is important for function, identifying cytocompatible, shrink-resistant materials are a major aim. Here, it is shown that hydrogels made of interpenetrating networks of collagen-I and recombinant spider silk protein eADF4(C16)-RGD nanofibrils exhibit synergistic and tunable mechanical properties. Composite hydrogels allow cell adhesion and spreading and are resistant to shrinkage mediated by fibroblasts, C2C12 myoblasts, and human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes. Myoblasts differentiate and fuse into myotubes, and hiPSC-cardiomyocytes can be cultured long-term, show spontaneous contractions, and remain drug responsive. Collectively, a novel composite material is developed to overcome the challenge of post-fabrication matrix shrinkage conferring high shape fidelity suitable for tissue engineering.
Collapse
Affiliation(s)
- Xuen J. Ng
- Chair of BiomaterialsUniversity of BayreuthProf.‐Rüdiger‐Bormann‐Str. 195447BayreuthGermany
| | - Tilman U. Esser
- Experimental Renal and Cardiovascular ResearchDepartment of NephropathologyInstitute of Pathology and Department of CardiologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)Kussmaulallee 1291054ErlangenGermany
| | - Vanessa T. Trossmann
- Chair of BiomaterialsUniversity of BayreuthProf.‐Rüdiger‐Bormann‐Str. 195447BayreuthGermany
| | - Christoph Rudisch
- Chair of BiomaterialsUniversity of BayreuthProf.‐Rüdiger‐Bormann‐Str. 195447BayreuthGermany
| | - Maren Fiedler
- Experimental Renal and Cardiovascular ResearchDepartment of NephropathologyInstitute of Pathology and Department of CardiologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)Kussmaulallee 1291054ErlangenGermany
| | - Kaveh Roshanbinfar
- Experimental Renal and Cardiovascular ResearchDepartment of NephropathologyInstitute of Pathology and Department of CardiologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)Kussmaulallee 1291054ErlangenGermany
| | - Zan Lamberger
- Department for Functional Materials in Medicine and DentistryUniversity Hospital of WürzburgPleicherwall 2D‐97070WürzburgGermany
| | - Philipp Stahlhut
- Department for Functional Materials in Medicine and DentistryUniversity Hospital of WürzburgPleicherwall 2D‐97070WürzburgGermany
| | - Gregor Lang
- Department for Functional Materials in Medicine and DentistryUniversity Hospital of WürzburgPleicherwall 2D‐97070WürzburgGermany
| | - Thomas Scheibel
- Chair of BiomaterialsUniversity of BayreuthProf.‐Rüdiger‐Bormann‐Str. 195447BayreuthGermany
- Bayreuth Center for Colloids and Interfaces (BZKG)University of Bayreuth95447BayreuthGermany
- Bavarian Polymer Institute (BPI)University of Bayreuth95447BayreuthGermany
- Bayreuth Center for Molecular Biosciences (BZMB)University of Bayreuth95447BayreuthGermany
- Bayreuth Center for Material Science (BayMAT)University of Bayreuth95447BayreuthGermany
- Faculty of MedicineUniversity of Würzburg97080WürzburgGermany
| | - Felix B. Engel
- Experimental Renal and Cardiovascular ResearchDepartment of NephropathologyInstitute of Pathology and Department of CardiologyFriedrich‐Alexander‐Universität Erlangen‐Nürnberg (FAU)Kussmaulallee 1291054ErlangenGermany
| |
Collapse
|
4
|
Kariuki R, Bryant SJ, Shepherd TP, Meftahi N, Bryant G, Conn CE, Christofferson AJ, Elbourne A. Single-particle adsorption of ultra-small gold nanoparticles at the biomembrane phase boundary. Colloids Surf B Biointerfaces 2025; 253:114734. [PMID: 40318394 DOI: 10.1016/j.colsurfb.2025.114734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Revised: 04/12/2025] [Accepted: 04/22/2025] [Indexed: 05/07/2025]
Abstract
Nanomaterials are revolutionizing biomedical research by enabling the development of novel therapies, with applications ranging from drug delivery and diagnostics to the modulation of specific biological processes. Current research focuses on tasks such as enhancing cellular uptake of materials while preserving their functionality. However, the mechanisms governing interactions between nanomaterials and biological systems-particularly cellular membranes-remain challenging to elucidate due to the complex, dynamic nature of the lipid bilayer environment. This complexity arises from factors such as coexisting lipid domains (conserved regions of lipids) or lipid rafts, as well as cellular behaviors that induce state changes. The heterogeneous membrane landscape may offer unique adsorption properties and other functional effects, making it crucial to understand these interactions for greater biological control in nanotherapeutics. In this work, we systematically expose a phase-separated phospholipid-supported lipid bilayer (SLB)-specifically, a fluid-gel DOPC:DPPC bilayer-to low concentrations of citrate-capped 5 nm gold nanoparticles (AuNPs) to observe the adsorption process of individual AuNPs at the molecular scale. Using atomic force microscopy (AFM), we experimentally detect the adsorption of some AuNPs at the phase boundary. Complementary molecular dynamics (MD) simulations further elucidate the mechanism of single AuNP adsorption at lipid phase boundaries. Our findings indicate that the AuNP preferentially incorporates into the fluid-phase DOPC lipids while maintaining partial association with the gel-phase DPPC lipids due to diffusion effects. During adsorption, the AuNP disrupts lipid organization by increasing lateral lipid mixing across the phase boundary. This disruption to lipid molecular ordering is further evident upon AuNP incorporation into the bilayer. The ability to modulate the spatial organization and structure of lipid molecules has significant implications for therapeutics that leverage lipid diffusion pathways for alternative drug delivery mechanisms or to induce specific lipid behaviors.
Collapse
Affiliation(s)
- Rashad Kariuki
- School of Science, STEM College, RMIT University, Melbourne, VIC 3001, Australia
| | - Saffron J Bryant
- School of Science, STEM College, RMIT University, Melbourne, VIC 3001, Australia
| | - Tilly P Shepherd
- School of Science, STEM College, RMIT University, Melbourne, VIC 3001, Australia
| | - Nastaran Meftahi
- Department of Civil and Construction Engineering, Swinburne University of Technology, Melbourne, VIC Australia
| | - Gary Bryant
- School of Science, STEM College, RMIT University, Melbourne, VIC 3001, Australia
| | - Charlotte E Conn
- School of Science, STEM College, RMIT University, Melbourne, VIC 3001, Australia
| | - Andrew J Christofferson
- School of Science, STEM College, RMIT University, Melbourne, VIC 3001, Australia; ARC Centre of Excellence in Exciton Science, School of Science, RMIT University, Melbourne, VIC 3001, Australia.
| | - Aaron Elbourne
- School of Science, STEM College, RMIT University, Melbourne, VIC 3001, Australia.
| |
Collapse
|
5
|
Xu Y, Yu Y, Guo Z. Hydrogels in cardiac tissue engineering: application and challenges. Mol Cell Biochem 2025; 480:2201-2222. [PMID: 39495368 DOI: 10.1007/s11010-024-05145-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 10/18/2024] [Indexed: 11/05/2024]
Abstract
Cardiovascular disease remains the leading cause of global mortality. Current stem cell therapy and heart transplant therapy have limited long-term stability in cardiac function. Cardiac tissue engineering may be one of the key methods for regenerating damaged myocardial tissue. As an ideal scaffold material, hydrogel has become a viable tissue engineering therapy for the heart. Hydrogel can not only provide mechanical support for infarcted myocardium but also serve as a carrier for various drugs, bioactive factors, and cells to increase myocardial contractility and improve the cell microenvironment in the infarcted area, thereby improving cardiac function. This paper reviews the applications of hydrogels and biomedical mechanisms in cardiac tissue engineering and discusses the challenge of clinical transformation of hydrogel in cardiac tissue engineering, providing new strategies for treating cardiovascular diseases.
Collapse
Affiliation(s)
- Yaping Xu
- Henan Key Laboratory of Cardiac Remodeling and Transplantation, Zhengzhou Seventh People's Hospital, Henan, 450016, Zhengzhou, People's Republic of China
| | - Yuexin Yu
- Henan Key Laboratory of Cardiac Remodeling and Transplantation, Zhengzhou Seventh People's Hospital, Henan, 450016, Zhengzhou, People's Republic of China
| | - Zhikun Guo
- Henan Key Laboratory of Cardiac Remodeling and Transplantation, Zhengzhou Seventh People's Hospital, Henan, 450016, Zhengzhou, People's Republic of China.
- Henan Key Laboratory of Medical Tissue Regeneration, Xinxiang Medical University, Xinxiang, 453003, Henan, People's Republic of China.
| |
Collapse
|
6
|
Akter MZ, Tufail F, Ahmad A, Oh YW, Kim JM, Kim S, Hasan MM, Li L, Lee DW, Kim YS, Lee SJ, Kim HS, Ahn Y, Choi YJ, Yi HG. Harnessing native blueprints for designing bioinks to bioprint functional cardiac tissue. iScience 2025; 28:111882. [PMID: 40177403 PMCID: PMC11964760 DOI: 10.1016/j.isci.2025.111882] [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] [Indexed: 04/05/2025] Open
Abstract
Cardiac tissue lacks regenerative capacity, making heart transplantation the primary treatment for end-stage heart failure. Engineered cardiac tissues developed through three-dimensional bioprinting (3DBP) offer a promising alternative. However, reproducing the native structure, cellular diversity, and functionality of cardiac tissue requires advanced cardiac bioinks. Major obstacles in CTE (cardiac tissue engineering) include accurately characterizing bioink properties, replicating the cardiac microenvironment, and achieving precise spatial organization. Optimizing bioink properties to closely mimic the extracellular matrix (ECM) is essential, as deviations may result in pathological effects. This review encompasses the rheological and electromechanical properties of bioinks and the function of the cardiac microenvironment in the design of functional cardiac constructs. Furthermore, it focuses on improving the rheological characteristics, printability, and functionality of bioinks, offering valuable perspectives for developing new bioinks especially designed for CTE.
Collapse
Affiliation(s)
- Mst Zobaida Akter
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju 61186, Republic of Korea
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Fatima Tufail
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju 61186, Republic of Korea
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Ashfaq Ahmad
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju 61186, Republic of Korea
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Yoon Wha Oh
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Jung Min Kim
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Seoyeon Kim
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Md Mehedee Hasan
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Longlong Li
- Department of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Dong-Weon Lee
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
- Department of Mechanical Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
- Center for Next-Generation Sensor Research and Development, Chonnam National University, Gwangju 61186, Republic of Korea
| | - Yong Sook Kim
- Biomedical Research Institute, Chonnam National University Hospital, Gwangju 61469, Republic of Korea
| | - Su-jin Lee
- Biomedical Research Institute, Chonnam National University Hospital, Gwangju 61469, Republic of Korea
- Department of Forensic Medicine, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Hyung-Seok Kim
- Department of Forensic Medicine, Chonnam National University Medical School, Gwangju 61469, Republic of Korea
| | - Youngkeun Ahn
- Division of Cardiology, Department of Internal Medicine, Chonnam National University Hospital, Chonnam National University Medical School, Gwangju, 61469, Republic of Korea
| | - Yeong-Jin Choi
- Advanced Bio and Healthcare Materials Research Division, Korea Institute of Materials Science (KIMS), Changwon 51508, Republic of Korea
- Advanced Materials Engineering, Korea National University of Science and Technology (UST), Changwon, Republic of Korea
| | - Hee-Gyeong Yi
- Department of Convergence Biosystems Engineering, Chonnam National University, Gwangju 61186, Republic of Korea
- Interdisciplinary Program in IT-Bio Convergence System, Chonnam National University, Gwangju 61186, Republic of Korea
- Advanced Medical Device Research Center for Cardiovascular Disease, Chonnam National University, Gwangju 61186, Republic of Korea
| |
Collapse
|
7
|
Baniasadi M, Baghban Salehi M, Baniasadi H. Acrylamide/Alyssum campestre seed gum hydrogels enhanced with titanium carbide: Rheological insights for cardiac tissue engineering. Int J Biol Macromol 2025; 293:139240. [PMID: 39732250 DOI: 10.1016/j.ijbiomac.2024.139240] [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: 09/05/2024] [Revised: 12/13/2024] [Accepted: 12/25/2024] [Indexed: 12/30/2024]
Abstract
This study investigates the use of acrylamide and Alyssum campestre seed gum (ACSG) to create hydrogel composites with enhanced electrical and mechanical properties by incorporating titanium carbide (TiC). The composites were analyzed through techniques such as FTIR, SEM, TEM, TGA, swelling, rheology, tensile, electrical conductivity, antibacterial, and MTT assays. XRD analysis showed that 0.5 % TiC NPs were exfoliated in the hydrogel, while 1 % was intercalated. SEM images showed that ACSG created a semi-interpenetrating polymer network with interconnected cavities averaging 9.1 μm, which reduced to 3.6 μm with 0.5%TiC and 51.8 nm with 1%TiC due to increased crosslinking density. TGA results confirmed hydrogel stability at autoclave temperatures. Rheological testing revealed that the hydrogel exhibited a maximum resistance of 317 kPa. The addition of 1 % TiC enhanced its electrical conductivity to 1.5 × 10-2S/cm, making it suitable for applications in cardiac tissue engineering. MTT assays confirmed the hydrogel's biocompatibility and demonstrated its superior antibacterial activity against Staphylococcus aureus compared to Escherichia coli. The AM/ACSG/TiC hydrogel is a promising material for cardiac tissue engineering because of its adjustable mechanical properties, excellent electrical conductivity, and strong compatibility with cell cultures. The addition of ACSG improves the hydrogel's rheological behavior, which is crucial for promoting effective cell growth.
Collapse
Affiliation(s)
- Mona Baniasadi
- Department of Petroleum Engineering, Chemistry and Chemical Engineering Research Center of Iran, Tehran, Iran; Polymer Synthesis Technology, School of Chemical Engineering, Aalto University, Espoo, Finland
| | - Mahsa Baghban Salehi
- Department of Petroleum Engineering, Chemistry and Chemical Engineering Research Center of Iran, Tehran, Iran; Polymer Synthesis Technology, School of Chemical Engineering, Aalto University, Espoo, Finland.
| | - Hossein Baniasadi
- Department of Petroleum Engineering, Chemistry and Chemical Engineering Research Center of Iran, Tehran, Iran; Polymer Synthesis Technology, School of Chemical Engineering, Aalto University, Espoo, Finland
| |
Collapse
|
8
|
Liao L, Zhang J, Ding J, Xu C, Zhu L, Hou Y, Li S, Zhang J, Wei B, Wang H. Adhesive, Biocompatible, Antibacterial, and Degradable Collagen-Based Conductive Hydrogel as Strain Sensor for Human Motion Monitoring. Molecules 2024; 29:5728. [PMID: 39683887 DOI: 10.3390/molecules29235728] [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: 11/05/2024] [Revised: 11/30/2024] [Accepted: 12/02/2024] [Indexed: 12/18/2024] Open
Abstract
The conductive hydrogels (CHs) are promising for developing flexible energy storage devices, flexible sensors, and electronic skin due to the unique features of excellent flexibility and high conductivity. However, poor biocompatibility and antibacterial properties seriously limit their application in the biomedical field. Collagen, one of the main components of the extracellular Matrix (ECM), is the ideal matrix for constructing hydrogels due to good biocompatibility with human tissue. Here, dopamine-polypyrrole-collagen (DA-PPY-COL) hydrogel was constructed by dopamine-mediated pyrrole in situ polymerization in a collagen matrix. As a strain sensor, it can be affixed to different parts of the human body to monitor large-scale motion movements and fine micro-expressions in real time. The performance was attributed to its good self-adhesion, flexibility, and electrical conductivity. Biological experiments have shown that it has good antimicrobial properties, biocompatibility, and degradability, allowing the hydrogel to safely monitor human motor behavior. This work not only offers a material preparation strategy for constructing biomimetic electronic skin and wearable sensors but also demonstrates the great potential prospect for implantable degradable medical device applications.
Collapse
Affiliation(s)
- Lixia Liao
- School of Chemistry and Environmental Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Jiyuan Zhang
- School of Chemistry and Environmental Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Jiaqi Ding
- School of Chemistry and Environmental Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Chengzhi Xu
- School of Chemistry and Environmental Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Lian Zhu
- School of Chemistry and Environmental Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Yuanjing Hou
- School of Chemistry and Environmental Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Sheng Li
- School of Chemistry and Environmental Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Juntao Zhang
- School of Chemistry and Environmental Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Benmei Wei
- School of Chemistry and Environmental Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Haibo Wang
- School of Chemistry and Environmental Engineering, Wuhan Polytechnic University, Wuhan 430023, China
- Hubei Key Laboratory of Quality Control of Characteristic Fruits and Vegetables, College of Life Science and Technology, Hubei Engineering University, Xiaogan 432000, China
| |
Collapse
|
9
|
Kong C, Guo Z, Teng T, Yao Q, Yu J, Wang M, Ma Y, Wang P, Tang Q. Electroactive Nanomaterials for the Prevention and Treatment of Heart Failure: From Materials and Mechanisms to Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024; 20:e2406206. [PMID: 39268781 DOI: 10.1002/smll.202406206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 09/02/2024] [Indexed: 09/15/2024]
Abstract
Heart failure (HF) represents a cardiovascular disease that significantly threatens global well-being and quality of life. Electroactive nanomaterials, characterized by their distinctive physical and chemical properties, emerge as promising candidates for HF prevention and management. This review comprehensively examines electroactive nanomaterials and their applications in HF intervention. It presents the definition, classification, and intrinsic characteristics of conductive, piezoelectric, and triboelectric nanomaterials, emphasizing their mechanical robustness, electrical conductivity, and piezoelectric coefficients. The review elucidates their applications and mechanisms: 1) early detection and diagnosis, employing nanomaterial-based sensors for real-time cardiac health monitoring; 2) cardiac tissue repair and regeneration, providing mechanical, chemical, and electrical stimuli for tissue restoration; 3) localized administration of bioactive biomolecules, genes, or pharmacotherapeutic agents, using nanomaterials as advanced drug delivery systems; and 4) electrical stimulation therapies, leveraging their properties for innovative pacemaker and neurostimulation technologies. Challenges in clinical translation, such as biocompatibility, stability, and scalability, are discussed, along with future prospects and potential innovations, including multifunctional and stimuli-responsive nanomaterials for precise HF therapies. This review encapsulates current research and future directions concerning the use of electroactive nanomaterials in HF prevention and management, highlighting their potential to innovating in cardiovascular medicine.
Collapse
Affiliation(s)
- Chunyan Kong
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Zhen Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Teng Teng
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Qi Yao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Jiabin Yu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Mingyu Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Yulan Ma
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Pan Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| | - Qizhu Tang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, 430060, P. R. China
- Hubei Key Laboratory of Metabolic and Chronic Diseases, Wuhan, 430060, P. R. China
| |
Collapse
|
10
|
Zuo X, Xiao Y, Yang J, He Y, He Y, Liu K, Chen X, Guo J. Engineering collagen-based biomaterials for cardiovascular medicine. COLLAGEN AND LEATHER 2024; 6:33. [DOI: 10.1186/s42825-024-00174-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Revised: 07/25/2024] [Accepted: 08/21/2024] [Indexed: 01/06/2025]
Abstract
AbstractCardiovascular diseases have been the leading cause of global mortality and disability. In addition to traditional drug and surgical treatment, more and more studies investigate tissue engineering therapeutic strategies in cardiovascular medicine. Collagen interweaves in the form of trimeric chains to form the physiological network framework of the extracellular matrix of cardiac and vascular cells, possessing excellent biological properties (such as low immunogenicity and good biocompatibility) and adjustable mechanical properties, which renders it a vital tissue engineering biomaterial for the treatment of cardiovascular diseases. In recent years, promising advances have been made in the application of collagen materials in blood vessel prostheses, injectable cardiac hydrogels, cardiac patches, and hemostatic materials, although their clinical translation still faces some obstacles. Thus, we reviewed these findings and systematically summarizes the application progress as well as problems of clinical translation of collagen biomaterials in the cardiovascular field. The present review contributes to a comprehensive understanding of the application of collagen biomaterials in cardiovascular medicine.
Graphical abstract
Collapse
|
11
|
Kim S, Shin Y, Han J, Kim HJ, Sunwoo SH. Introductory Review of Soft Implantable Bioelectronics Using Conductive and Functional Hydrogels and Hydrogel Nanocomposites. Gels 2024; 10:614. [PMID: 39451267 PMCID: PMC11506957 DOI: 10.3390/gels10100614] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 09/20/2024] [Accepted: 09/21/2024] [Indexed: 10/26/2024] Open
Abstract
Interfaces between implantable bioelectrodes and tissues provide critical insights into the biological and pathological conditions of targeted organs, aiding diagnosis and treatment. While conventional bioelectronics, made from rigid materials like metals and silicon, have been essential for recording signals and delivering electric stimulation, they face limitations due to the mechanical mismatch between rigid devices and soft tissues. Recently, focus has shifted toward soft conductive materials, such as conductive hydrogels and hydrogel nanocomposites, known for their tissue-like softness, biocompatibility, and potential for functionalization. This review introduces these materials and provides an overview of recent advances in soft hydrogel nanocomposites for implantable electronics. It covers material strategies for conductive hydrogels, including both intrinsically conductive hydrogels and hydrogel nanocomposites, and explores key functionalization techniques like biodegradation, bioadhesiveness, injectability, and self-healing. Practical applications of these materials in implantable electronics are also highlighted, showcasing their effectiveness in real-world scenarios. Finally, we discuss emerging technologies and future needs for chronically implantable bioelectronics, offering insights into the evolving landscape of this field.
Collapse
Affiliation(s)
- San Kim
- Department of Chemical Engineering, Kumoh National Institute of Technology, Gumi 39177, Republic of Korea
| | - Yumin Shin
- Department of Chemical Engineering, Kumoh National Institute of Technology, Gumi 39177, Republic of Korea
| | - Jaewon Han
- Division of Biomedical Engineering, Yonsei University, Wonju 26493, Republic of Korea
| | - Hye Jin Kim
- Division of Biomedical Engineering, Yonsei University, Wonju 26493, Republic of Korea
| | - Sung-Hyuk Sunwoo
- Department of Chemical Engineering, Kumoh National Institute of Technology, Gumi 39177, Republic of Korea
- Andrew and Peggy Cherng Department of Medical Engineering, California Institute of Technology, Pasadena, CA 91106, USA
| |
Collapse
|
12
|
Roshanbinfar K, Schiffer M, Carls E, Angeloni M, Koleśnik-Gray M, Schruefer S, Schubert DW, Ferrazzi F, Krstić V, Fleischmann BK, Roell W, Engel FB. Electrically Conductive Collagen-PEDOT:PSS Hydrogel Prevents Post-Infarct Cardiac Arrhythmia and Supports hiPSC-Cardiomyocyte Function. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2403642. [PMID: 38653478 DOI: 10.1002/adma.202403642] [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: 03/11/2024] [Indexed: 04/25/2024]
Abstract
Myocardial infarction (MI) causes cell death, disrupts electrical activity, triggers arrhythmia, and results in heart failure, whereby 50-60% of MI-associated deaths manifest as sudden cardiac deaths (SCD). The most effective therapy for SCD prevention is implantable cardioverter defibrillators (ICDs). However, ICDs contribute to adverse remodeling and disease progression and do not prevent arrhythmia. This work develops an injectable collagen-PEDOT:PSS (poly(3,4-ethylenedioxythiophene) polystyrene sulfonate) hydrogel that protects infarcted hearts against ventricular tachycardia (VT) and can be combined with human induced pluripotent stem cell (hiPSC)-cardiomyocytes to promote partial cardiac remuscularization. PEDOT:PSS improves collagen gel formation, micromorphology, and conductivity. hiPSC-cardiomyocytes in collagen-PEDOT:PSS hydrogels exhibit near-adult sarcomeric length, improved contractility, enhanced calcium handling, and conduction velocity. RNA-sequencing data indicate enhanced maturation and improved cell-matrix interactions. Injecting collagen-PEDOT:PSS hydrogels in infarcted mouse hearts decreases VT to the levels of healthy hearts. Collectively, collagen-PEDOT:PSS hydrogels offer a versatile platform for treating cardiac injuries.
Collapse
Affiliation(s)
- Kaveh Roshanbinfar
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Miriam Schiffer
- Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany
| | - Esther Carls
- Department of Cardiac Surgery, UKB, University of Bonn, Germany
| | - Miriam Angeloni
- Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| | - Maria Koleśnik-Gray
- Department of Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Staudtstr. 7, 91058, Erlangen, Germany
| | - Stefan Schruefer
- Institute of Polymer Materials, Department of Materials Science and Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Martensstr. 7, 91058, Erlangen, Germany
| | - Dirk W Schubert
- Institute of Polymer Materials, Department of Materials Science and Engineering, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Martensstr. 7, 91058, Erlangen, Germany
| | - Fulvia Ferrazzi
- Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
- Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Muscle Research Center Erlangen (MURCE), 91054, Erlangen, Germany
| | - Vojislav Krstić
- Department of Physics, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Staudtstr. 7, 91058, Erlangen, Germany
| | - Bernd K Fleischmann
- Institute of Physiology I, Life and Brain Center, Medical Faculty, University of Bonn, Germany
| | - Wilhelm Roell
- Department of Cardiac Surgery, UKB, University of Bonn, Germany
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), 91054, Erlangen, Germany
| |
Collapse
|
13
|
Lai H, Huang R, Weng X, Huang B, Yao J, Pian Y. Classification and applications of nanomaterials in vitro diagnosis. Heliyon 2024; 10:e32314. [PMID: 38868029 PMCID: PMC11168482 DOI: 10.1016/j.heliyon.2024.e32314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2023] [Revised: 05/19/2024] [Accepted: 05/31/2024] [Indexed: 06/14/2024] Open
Abstract
With the rapid development of clinical diagnosis and treatment, many traditional and conventional in vitro diagnosis technologies are unable to meet the demands of clinical medicine development. In this situation, nanomaterials are rapidly developing and widely used in the field of in vitro diagnosis. Nanomaterials have distinct size-dependent physical or chemical properties, and their optical, magnetic, electrical, thermal, and biological properties can be modulated at the nanoscale by changing their size, shape, chemical composition, and surface functional groups, particularly because they have a larger specific surface area than macromaterials. They provide an amount of space to modify different molecules on their surface, allowing them to detect small substances, nucleic acids, proteins, and microorganisms. Combining nanomaterials with in vitro diagnosis is expected to result in lower detection limits, higher sensitivity, and stronger selectivity. In this review, we will discuss the classfication and properties of some common nanomaterials, as well as their applications in protein, nucleic acids, and other aspect detection and analysis for in vitro diagnosis, especially on aging-related nanodiagnostics. Finally, it is summarized with guidelines for in vitro diagnosis.
Collapse
Affiliation(s)
- Huiying Lai
- Department of Laboratory Medicine, Beijing Hospital, National Center of Gerontology, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing, PR China
| | - Rongfu Huang
- The Second Affiliated Hospital, Fujian Medical University, Quanzhou, PR China
| | - Xin Weng
- The Second Affiliated Hospital, Fujian Medical University, Quanzhou, PR China
| | - Baoshan Huang
- The Second Affiliated Hospital, Fujian Medical University, Quanzhou, PR China
| | - Jianfeng Yao
- Quanzhou Maternity and Child Healthcare Hospital, Quanzhou, PR China
| | - Yaya Pian
- National Center for Clinical Laboratories, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology, Beijing, PR China
| |
Collapse
|
14
|
Esmaeili H, Patino-Guerrero A, Nelson RA, Karamanova N, M Fisher T, Zhu W, Perreault F, Migrino RQ, Nikkhah M. Engineered Gold and Silica Nanoparticle-Incorporated Hydrogel Scaffolds for Human Stem Cell-Derived Cardiac Tissue Engineering. ACS Biomater Sci Eng 2024; 10:2351-2366. [PMID: 38323834 PMCID: PMC11075803 DOI: 10.1021/acsbiomaterials.3c01256] [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] [Indexed: 02/08/2024]
Abstract
Electrically conductive biomaterials and nanomaterials have demonstrated great potential in the development of functional and mature cardiac tissues. In particular, gold nanomaterials have emerged as promising candidates due to their biocompatibility and ease of fabrication for cardiac tissue engineering utilizing rat- or stem cell-derived cardiomyocytes (CMs). However, despite significant advancements, it is still not clear whether the enhancement in cardiac tissue function is primarily due to the electroconductivity features of gold nanoparticles or the structural changes of the scaffold resulting from the addition of these nanoparticles. To address this question, we developed nanoengineered hydrogel scaffolds comprising gelatin methacrylate (GelMA) embedded with either electrically conductive gold nanorods (GNRs) or nonconductive silica nanoparticles (SNPs). This enabled us to simultaneously assess the roles of electrically conductive and nonconductive nanomaterials in the functionality and fate of human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs). Our studies revealed that both GNR- and SNP-incorporated hydrogel scaffolds exhibited excellent biocompatibility and similar cardiac cell attachment. Although the expression of sarcomere alpha-actinin did not significantly differ among the conditions, a more organized sarcomere structure was observed within the GNR-embedded hydrogels compared to the nonconductive nanoengineered scaffolds. Furthermore, electrical coupling was notably improved in GNR-embedded scaffolds, as evidenced by the synchronous calcium flux and enhanced calcium transient intensity. While we did not observe a significant difference in the gene expression profile of human cardiac tissues formed on the conductive GNR- and nonconductive SNP-incorporated hydrogels, we noticed marginal improvements in the expression of some calcium and structural genes in the nanomaterial-embedded hydrogel groups as compared to the control condition. Given that the cardiac tissues formed atop the nonconductive SNP-based scaffolds (used as the control for conductivity) also displayed similar levels of gene expression as compared to the conductive hydrogels, it suggests that the electrical conductivity of nanomaterials (i.e., GNRs) may not be the sole factor influencing the function and fate of hiPSC-derived cardiac tissues when cells are cultured atop the scaffolds. Overall, our findings provide additional insights into the role of electrically conductive gold nanoparticles in regulating the functionalities of hiPSC-CMs.
Collapse
Affiliation(s)
- Hamid Esmaeili
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, United States
| | - Alejandra Patino-Guerrero
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, United States
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic, Scottsdale, Arizona 85259, United States
| | - Ronald A Nelson
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, United States
| | - Nina Karamanova
- Phoenix Veterans Affairs Health Care System, Phoenix, Arizona 85022, United States
| | - Taylor M Fisher
- School of Sustainable Engineering and the Built Environment, Arizona State University, Tempe, Arizona 85287, United States
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Center for Regenerative Medicine, Mayo Clinic, Scottsdale, Arizona 85259, United States
| | - François Perreault
- School of Sustainable Engineering and the Built Environment, Arizona State University, Tempe, Arizona 85287, United States
| | - Raymond Q Migrino
- Phoenix Veterans Affairs Health Care System, Phoenix, Arizona 85022, United States
- University of Arizona College of Medicine, Phoenix, Arizona 85004, United States
| | - Mehdi Nikkhah
- School of Biological and Health Systems Engineering, Arizona State University, Tempe, Arizona 85287, United States
- Biodesign Virginia G. Piper Center for Personalized Diagnosis, Arizona State University, Tempe, Arizona 85287, United States
| |
Collapse
|
15
|
Jo HJ, Kang MS, Heo HJ, Jang HJ, Park R, Hong SW, Kim YH, Han DW. Skeletal muscle regeneration with 3D bioprinted hyaluronate/gelatin hydrogels incorporating MXene nanoparticles. Int J Biol Macromol 2024; 265:130696. [PMID: 38458288 DOI: 10.1016/j.ijbiomac.2024.130696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2023] [Revised: 01/31/2024] [Accepted: 03/05/2024] [Indexed: 03/10/2024]
Abstract
There has been significant progress in the field of three-dimensional (3D) bioprinting technology, leading to active research on creating bioinks capable of producing structurally and functionally tissue-mimetic constructs. Ti3C2Tx MXene nanoparticles (NPs), promising two-dimensional nanomaterials, are being investigated for their potential in muscle regeneration due to their unique physicochemical properties. In this study, we integrated MXene NPs into composite hydrogels made of gelatin methacryloyl (GelMA) and hyaluronic acid methacryloyl (HAMA) to develop bioinks (namely, GHM bioink) that promote myogenesis. The prepared GHM bioinks were found to offer excellent printability with structural integrity, cytocompatibility, and microporosity. Additionally, MXene NPs within the 3D bioprinted constructs encouraged the differentiation of C2C12 cells into skeletal muscle cells without additional support of myogenic agents. Genetic analysis indicated that representative myogenic markers both for early and late myogenesis were significantly up-regulated. Moreover, animal studies demonstrated that GHM bioinks contributed to enhanced regeneration of skeletal muscle while reducing immune responses in mice models with volumetric muscle loss (VML). Our results suggest that the GHM hydrogel can be exploited to craft a range of strategies for the development of a novel bioink to facilitate skeletal muscle regeneration because these MXene-incorporated composite materials have the potential to promote myogenesis.
Collapse
Affiliation(s)
- Hyo Jung Jo
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Republic of Korea
| | - Moon Sung Kang
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Republic of Korea
| | - Hye Jin Heo
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea
| | - Hee Jeong Jang
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Republic of Korea
| | - Rowoon Park
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Republic of Korea
| | - Suck Won Hong
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Republic of Korea; Engineering Research Center for Color-Modulated Extra-Sensory Perception Technology, Pusan National University, Busan 46241, Republic of Korea
| | - Yun Hak Kim
- Department of Anatomy, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; Department of Biomedical Informatics, School of Medicine, Pusan National University, Yangsan 50612, Republic of Korea; Periodontal Disease Signaling Network Research Center & Dental and Life Science Institute, School of Dentistry, Pusan National University, Yangsan 50612, Republic of Korea.
| | - Dong-Wook Han
- Department of Cogno-Mechatronics Engineering, College of Nanoscience and Nanotechnology, Pusan National University, Busan 46241, Republic of Korea; BIO-IT Fusion Technology Research Institute, Pusan National University, Busan 46241, Republic of Korea.
| |
Collapse
|
16
|
Liu B, Alimi OA, Wang Y, Kong Y, Kuss M, Krishnan MA, Hu G, Xiao Y, Dong J, DiMaio DJ, Duan B. Differentiated mesenchymal stem cells-derived exosomes immobilized in decellularized sciatic nerve hydrogels for peripheral nerve repair. J Control Release 2024; 368:24-41. [PMID: 38367864 PMCID: PMC11411504 DOI: 10.1016/j.jconrel.2024.02.019] [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: 11/23/2023] [Revised: 01/31/2024] [Accepted: 02/12/2024] [Indexed: 02/19/2024]
Abstract
Peripheral nerve injury (PNI) and the limitations of current treatments often result in incomplete sensory and motor function recovery, which significantly impact the patient's quality of life. While exosomes (Exo) derived from stem cells and Schwann cells have shown promise on promoting PNI repair following systemic administration or intraneural injection, achieving effective local and sustained Exo delivery holds promise to treat local PNI and remains challenging. In this study, we developed Exo-loaded decellularized porcine nerve hydrogels (DNH) for PNI repair. We successfully isolated Exo from differentiated human adipose-derived mesenchymal stem cells (hADMSC) with a Schwann cell-like phenotype (denoted as dExo). These dExo were further combined with polyethylenimine (PEI), and DNH to create polyplex hydrogels (dExo-loaded pDNH). At a PEI content of 0.1%, pDNH showed cytocompatibility for hADMSCs and supported neurite outgrowth of dorsal root ganglions. The sustained release of dExos from dExo-loaded pDNH persisted for at least 21 days both in vitro and in vivo. When applied around injured nerves in a mouse sciatic nerve crush injury model, the dExo-loaded pDNH group significantly improved sensory and motor function recovery and enhanced remyelination compared to dExo and pDNH only groups, highlighting the synergistic regenerative effects. Interestingly, we observed a negative correlation between the number of colony-stimulating factor-1 receptor (CSF-1R) positive cells and the extent of PNI regeneration at the 21-day post-surgery stage. Subsequent in vitro experiments demonstrated the potential involvement of the CSF-1/CSF-1R axis in Schwann cells and macrophage interaction, with dExo effectively downregulating CSF-1/CSF-1R signaling.
Collapse
Affiliation(s)
- Bo Liu
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Olawale A Alimi
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yanfei Wang
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; College of Osteopathic Medicine, Lake Erie College of Osteopathic Medicine, Erie, PA 16509, USA
| | - Yunfan Kong
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Mitchell Kuss
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Mena Asha Krishnan
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Guoku Hu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Yi Xiao
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Jixin Dong
- Eppley Institute for Research in Cancer and Allied Diseases, Fred & Pamela Buffett Cancer Center, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Dominick J DiMaio
- Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE 68198, USA
| | - Bin Duan
- Mary & Dick Holland Regenerative Medicine Program and Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE, USA; Division of Cardiology, Department of Internal Medicine, University of Nebraska Medical Center, Omaha, NE 68198, USA; Department of Mechanical and Materials Engineering, University of Nebraska-Lincoln, Lincoln, NE 68588, USA.
| |
Collapse
|
17
|
Yue C, Ding C, Hu M, Zhang R, Cheng B. Collagen/functionalized cellulose nanofibril composite aerogels with pH-responsive characteristics for drug delivery system. Int J Biol Macromol 2024; 261:129650. [PMID: 38286379 DOI: 10.1016/j.ijbiomac.2024.129650] [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: 09/09/2023] [Revised: 01/10/2024] [Accepted: 01/18/2024] [Indexed: 01/31/2024]
Abstract
In this work, carboxylated and amination modified cellulose nanofibrils (CNFs) were fabricated via the TEMPO catalytic oxidation system and diethylenetriamine, and collagen composite aerogels were fabricated through a simple self-assembly pretreatment and directional freeze-drying technology. Morphology analysis showed that the collagen composite aerogels had distinct layered-oriented double network structures after the self-assembly pretreatment. The intermolecular interactions between the collagen fibrils and functionalized CNFs (fCNFs) on the structures and properties of the composite aerogels were also examined through various characterization techniques. Water contact angle tests demonstrated the pH-responsive characteristics of the collagen/fCNF composite aerogels. Using 5-fluorouracil as the model drug, the pH-response mechanism was revealed. These results indicated that the collagen/fCNF composite aerogels exhibited excellent pH-responsive drug release capacities. Therefore, these pH-responsive collagen composite aerogels might have potential applications in industrial production in the biomedical, drug delivery, and tissue engineering fields.
Collapse
Affiliation(s)
- Chengfei Yue
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, School of Textile Science and Engineering, Wuhan Textile University, Wuhan, 430200, China; Tianjin Key Laboratory of Advanced Fibers and Energy Storage, School of Materials Science and Engineering, Tiangong University, Tianjin, 300387, China
| | - Changkun Ding
- Tianjin Key Laboratory of Advanced Fibers and Energy Storage, School of Materials Science and Engineering, Tiangong University, Tianjin, 300387, China.
| | - Min Hu
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, School of Textile Science and Engineering, Wuhan Textile University, Wuhan, 430200, China
| | - Ruquan Zhang
- State Key Laboratory of New Textile Materials and Advanced Processing Technologies, School of Textile Science and Engineering, Wuhan Textile University, Wuhan, 430200, China.
| | - Bowen Cheng
- Tianjin Key Laboratory of Pulp and Paper, Tianjin University of Science & Technology, Tianjin 300457, China
| |
Collapse
|
18
|
Rosales RM, Mountris KA, Oliván-Viguera A, Pérez-Zabalza M, Cedillo-Servin G, Iglesias-García O, Hrynevich A, Castilho M, Malda J, Prósper F, Doblaré M, Mazo MM, Pueyo E. Experimentally-guided in silico design of engineered heart tissues to improve cardiac electrical function after myocardial infarction. Comput Biol Med 2024; 171:108044. [PMID: 38335818 DOI: 10.1016/j.compbiomed.2024.108044] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2023] [Revised: 12/23/2023] [Accepted: 01/26/2024] [Indexed: 02/12/2024]
Abstract
Engineered heart tissues (EHTs) built from human induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) showed promising results for cardiac function restoration following myocardial infarction. Nevertheless, human iPSC-CMs have longer action potential and lower cell-to-cell coupling than adult-like CMs. These immature electrophysiological properties favor arrhythmias due to the generation of electrophysiological gradients when hiPSC-CMs are injected in the cardiac tissue. Culturing hiPSC-CMs on three-dimensional (3D) scaffolds can promote their maturation and influence their alignment. However, it is still uncertain how on-scaffold culturing influences the overall electrophysiology of the in vitro and implanted EHTs, as it requires expensive and time consuming experimentation. Here, we computationally investigated the impact of the scaffold design on the EHT electrical depolarization and repolarization before and after engraftment on infarcted tissue. We first acquired and processed electrical recordings from in vitro EHTs, which we used to calibrate the modeling and simulation of in silico EHTs to replicate experimental outcomes. Next, we built in silico EHT models for a range of scaffold pore sizes, shapes (square, rectangular, auxetic, hexagonal) and thicknesses. In this setup, we found that scaffolds made of small (0.2 mm2), elongated (30° half-angle) hexagons led to faster EHT activation and better mimicked the cardiac anisotropy. The scaffold thickness had a marginal role on the not engrafted EHT electrophysiology. Moreover, EHT engraftment on infarcted tissue showed that the EHT conductivity should be at least 5% of that in healthy tissue for bidirectional EHT-myocardium electrical propagation. For conductivities above such threshold, the scaffold made of small elongated hexagons led to the lowest activation time (AT) in the coupled EHT-myocardium. If the EHT conductivity was further increased and the hiPSC-CMs were uniformly oriented parallel to the epicardial cells, the total AT and the repolarization time gradient decreased substantially, thus minimizing the likelihood for arrhythmias after EHT transplantation.
Collapse
Affiliation(s)
- Ricardo M Rosales
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Zaragoza, Aragón, Spain; CIBER-BBN, ISCIII, Madrid, Spain; Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Aragón, Spain.
| | | | - Aida Oliván-Viguera
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Zaragoza, Aragón, Spain; CIBER-BBN, ISCIII, Madrid, Spain; Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Aragón, Spain.
| | - María Pérez-Zabalza
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Zaragoza, Aragón, Spain; CIBER-BBN, ISCIII, Madrid, Spain; Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Aragón, Spain; Defense University Centre (CUD), Zaragoza, Spain.
| | - Gerardo Cedillo-Servin
- Regenerative Medicine Center, Utrecht, The Netherlands; Department of Orthopedics, University Medical Center, Utrecht, The Netherlands.
| | - Olalla Iglesias-García
- Regenerative Medicine Program, CIMA Universidad de Navarra, Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Spain.
| | - Andrei Hrynevich
- Regenerative Medicine Center, Utrecht, The Netherlands; Department of Orthopedics, University Medical Center, Utrecht, The Netherlands.
| | - Miguel Castilho
- Department of Orthopedics, University Medical Center, Utrecht, The Netherlands; Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.
| | - Jos Malda
- Regenerative Medicine Center, Utrecht, The Netherlands; Department of Orthopedics, University Medical Center, Utrecht, The Netherlands; Department of Equine Sciences, Faculty of Veterinary Medicine, Utrecht University, Utrecht, The Netherlands.
| | - Felipe Prósper
- Regenerative Medicine Program, CIMA Universidad de Navarra, Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Spain; Hematology and Cell Therapy, Clínica Universidad de Navarra, Pamplona, Spain; CIBER de Cáncer (CIBERONC, team CB16/12/00489), Pamplona, Spain.
| | - Manuel Doblaré
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Zaragoza, Aragón, Spain; CIBER-BBN, ISCIII, Madrid, Spain; Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Aragón, Spain.
| | - Manuel M Mazo
- Regenerative Medicine Program, CIMA Universidad de Navarra, Pamplona, Spain; Instituto de Investigación Sanitaria de Navarra (IdiSNA), Spain; Hematology and Cell Therapy, Clínica Universidad de Navarra, Pamplona, Spain.
| | - Esther Pueyo
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Zaragoza, Aragón, Spain; CIBER-BBN, ISCIII, Madrid, Spain; Aragón Institute of Engineering Research (I3A), University of Zaragoza, Zaragoza, Aragón, Spain.
| |
Collapse
|
19
|
Cao L, Zhang Z, Yuan D, Yu M, Min J. Tissue engineering applications of recombinant human collagen: a review of recent progress. Front Bioeng Biotechnol 2024; 12:1358246. [PMID: 38419725 PMCID: PMC10900516 DOI: 10.3389/fbioe.2024.1358246] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2023] [Accepted: 01/31/2024] [Indexed: 03/02/2024] Open
Abstract
With the rapid development of synthetic biology, recombinant human collagen has emerged as a cutting-edge biological material globally. Its innovative applications in the fields of material science and medicine have opened new horizons in biomedical research. Recombinant human collagen stands out as a highly promising biomaterial, playing a pivotal role in crucial areas such as wound healing, stroma regeneration, and orthopedics. However, realizing its full potential by efficiently delivering it for optimal therapeutic outcomes remains a formidable challenge. This review provides a comprehensive overview of the applications of recombinant human collagen in biomedical systems, focusing on resolving this crucial issue. Additionally, it encompasses the exploration of 3D printing technologies incorporating recombinant collagen to address some urgent clinical challenges in regenerative repair in the future. The primary aim of this review also is to spotlight the advancements in the realm of biomaterials utilizing recombinant collagen, with the intention of fostering additional innovation and making significant contributions to the enhancement of regenerative biomaterials, therapeutic methodologies, and overall patient outcomes.
Collapse
Affiliation(s)
- Lili Cao
- Department of Plastic Surgery, Zhejiang Rongjun Hospital, Jiaxing, Zhejiang, China
| | - Zhongfeng Zhang
- Department of Plastic Surgery, Zhejiang Rongjun Hospital, Jiaxing, Zhejiang, China
| | - Dan Yuan
- Department of Plastic Surgery, Zhejiang Rongjun Hospital, Jiaxing, Zhejiang, China
| | - Meiping Yu
- Department of Plastic Surgery, Zhejiang Rongjun Hospital, Jiaxing, Zhejiang, China
| | - Jie Min
- General Surgery Department, Jiaxing No.1 Hospital, Jiaxing, Zhejiang, China
| |
Collapse
|
20
|
Sesena-Rubfiaro A, Prajapati NJ, Lou L, Ghimire G, Agarwal A, He J. Improving the development of human engineered cardiac tissue by gold nanorods embedded extracellular matrix for long-term viability. NANOSCALE 2024; 16:2983-2992. [PMID: 38259163 DOI: 10.1039/d3nr05422e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/24/2024]
Abstract
A myocardial infarction (MI), commonly called a heart attack, results in the death of cardiomyocytes (CMs) in the heart. Tissue engineering provides a promising strategy for the treatment of MI, but the maturation of human engineered cardiac tissue (hECT) still requires improvement. Conductive polymers and nanomaterials have been incorporated into the extracellular matrix to enhance the mechanical and electrical coupling between cardiac cells. Here we report a simple approach to incorporate gold nanorods (GNRs) into the fibrin hydrogel to form a GNR-fibrin matrix, which is used as the major component of the extracellular matrix for forming a 3D hECT construct suspended between two flexible posts. The hECTs made with GNR-fibrin hydrogel showed markers of maturation such as higher twitch force, synchronous beating activity, sarcomere maturation and alignment, t-tubule network development, and calcium handling improvement. Most importantly, the GNR-hECTs can survive over 9 months. We envision that the hECT with GNRs holds the potential to restore the functionality of the infarcted heart.
Collapse
Affiliation(s)
| | - Navin J Prajapati
- Department of Physics, Florida International University, Miami, FL 33199, USA.
| | - Lihua Lou
- Department of Mechanical and Materials Engineering, Florida International University, Miami, FL 33174, USA
| | - Govinda Ghimire
- Department of Physics, Florida International University, Miami, FL 33199, USA.
| | - Arvind Agarwal
- Department of Mechanical and Materials Engineering, Florida International University, Miami, FL 33174, USA
| | - Jin He
- Department of Physics, Florida International University, Miami, FL 33199, USA.
- Biomolecular Science Institute, Florida International University, Miami, FL 33199, USA
| |
Collapse
|
21
|
Esser TU, Anspach A, Muenzebrock KA, Kah D, Schrüfer S, Schenk J, Heinze KG, Schubert DW, Fabry B, Engel FB. Direct 3D-Bioprinting of hiPSC-Derived Cardiomyocytes to Generate Functional Cardiac Tissues. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2305911. [PMID: 37655652 DOI: 10.1002/adma.202305911] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/18/2023] [Indexed: 09/02/2023]
Abstract
3D-bioprinting is a promising technology to produce human tissues as drug screening tool or for organ repair. However, direct printing of living cells has proven difficult. Here, a method is presented to directly 3D-bioprint human induced pluripotent stem cell (hiPSC)-derived cardiomyocytes embedded in a collagen-hyaluronic acid ink, generating centimeter-sized functional ring- and ventricle-shaped cardiac tissues in an accurate and reproducible manner. The printed tissues contain hiPSC-derived cardiomyocytes with well-organized sarcomeres and exhibit spontaneous and regular contractions, which persist for several months and are able to contract against passive resistance. Importantly, beating frequencies of the printed cardiac tissues can be modulated by pharmacological stimulation. This approach opens up new possibilities for generating complex functional cardiac tissues as models for advanced drug screening or as tissue grafts for organ repair or replacement.
Collapse
Affiliation(s)
- Tilman U Esser
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Muscle Research Center Erlangen (MURCE), 91054, Erlangen, Germany
| | - Annalise Anspach
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Muscle Research Center Erlangen (MURCE), 91054, Erlangen, Germany
| | - Katrin A Muenzebrock
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Muscle Research Center Erlangen (MURCE), 91054, Erlangen, Germany
| | - Delf Kah
- Department of Physics, University of Erlangen-Nuremberg, 91052, Erlangen, Germany
| | - Stefan Schrüfer
- Institute of Polymer Materials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058, Erlangen, Germany
| | - Joachim Schenk
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg (JMU), 97080, Würzburg, Germany
| | - Katrin G Heinze
- Rudolf Virchow Center, Center for Integrative and Translational Bioimaging, Julius-Maximilians-Universität Würzburg (JMU), 97080, Würzburg, Germany
| | - Dirk W Schubert
- Institute of Polymer Materials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058, Erlangen, Germany
| | - Ben Fabry
- Department of Physics, University of Erlangen-Nuremberg, 91052, Erlangen, Germany
| | - Felix B Engel
- Experimental Renal and Cardiovascular Research, Department of Nephropathology, Institute of Pathology, Friedrich-Alexander-Universität Erlangen-Nürnberg (FAU), Muscle Research Center Erlangen (MURCE), 91054, Erlangen, Germany
| |
Collapse
|
22
|
Yoon J, Han H, Jang J. Nanomaterials-incorporated hydrogels for 3D bioprinting technology. NANO CONVERGENCE 2023; 10:52. [PMID: 37968379 PMCID: PMC10651626 DOI: 10.1186/s40580-023-00402-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 10/24/2023] [Indexed: 11/17/2023]
Abstract
In the field of tissue engineering and regenerative medicine, various hydrogels derived from the extracellular matrix have been utilized for creating engineered tissues and implantable scaffolds. While these hydrogels hold immense promise in the healthcare landscape, conventional bioinks based on ECM hydrogels face several challenges, particularly in terms of lacking the necessary mechanical properties required for 3D bioprinting process. To address these limitations, researchers are actively exploring novel nanomaterial-reinforced ECM hydrogels for both mechanical and functional aspects. In this review, we focused on discussing recent advancements in the fabrication of engineered tissues and monitoring systems using nanobioinks and nanomaterials via 3D bioprinting technology. We highlighted the synergistic benefits of combining numerous nanomaterials into ECM hydrogels and imposing geometrical effects by 3D bioprinting technology. Furthermore, we also elaborated on critical issues remaining at the moment, such as the inhomogeneous dispersion of nanomaterials and consequent technical and practical issues, in the fabrication of complex 3D structures with nanobioinks and nanomaterials. Finally, we elaborated on plausible outlooks for facilitating the use of nanomaterials in biofabrication and advancing the function of engineered tissues.
Collapse
Affiliation(s)
- Jungbin Yoon
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Hohyeon Han
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea
| | - Jinah Jang
- Department of Mechanical Engineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea.
- School of Interdisciplinary Bioscience and Bioengineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea.
- Department of Convergence IT Engineering, Pohang University of Science and Technology (POSTECH), Pohang, South Korea.
- Institute of Convergence Science, Yonsei University, Seoul, South Korea.
| |
Collapse
|
23
|
Merotto E, Pavan PG, Piccoli M. Three-Dimensional Bioprinting of Naturally Derived Hydrogels for the Production of Biomimetic Living Tissues: Benefits and Challenges. Biomedicines 2023; 11:1742. [PMID: 37371837 DOI: 10.3390/biomedicines11061742] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 06/07/2023] [Accepted: 06/15/2023] [Indexed: 06/29/2023] Open
Abstract
Three-dimensional bioprinting is the process of manipulating cell-laden bioinks to fabricate living structures. Three-dimensional bioprinting techniques have brought considerable innovation in biomedicine, especially in the field of tissue engineering, allowing the production of 3D organ and tissue models for in vivo transplantation purposes or for in-depth and precise in vitro analyses. Naturally derived hydrogels, especially those obtained from the decellularization of biological tissues, are promising bioinks for 3D printing purposes, as they present the best biocompatibility characteristics. Despite this, many natural hydrogels do not possess the necessary mechanical properties to allow a simple and immediate application in the 3D printing process. In this review, we focus on the bioactive and mechanical characteristics that natural hydrogels may possess to allow efficient production of organs and tissues for biomedical applications, emphasizing the reinforcement techniques to improve their biomechanical properties.
Collapse
Affiliation(s)
- Elena Merotto
- Tissue Engineering Lab, Istituto di Ricerca Pediatrica Città della Speranza, Corso Statu Uniti 4, 35127 Padova, Italy
- Department of Industrial Engineering, University of Padova, Via Gradenigo 6a, 35129 Padova, Italy
| | - Piero G Pavan
- Tissue Engineering Lab, Istituto di Ricerca Pediatrica Città della Speranza, Corso Statu Uniti 4, 35127 Padova, Italy
- Department of Industrial Engineering, University of Padova, Via Gradenigo 6a, 35129 Padova, Italy
| | - Martina Piccoli
- Tissue Engineering Lab, Istituto di Ricerca Pediatrica Città della Speranza, Corso Statu Uniti 4, 35127 Padova, Italy
| |
Collapse
|