1
|
Liu Y, Gilchrist AE, Heilshorn SC. Engineered Protein Hydrogels as Biomimetic Cellular Scaffolds. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2407794. [PMID: 39233559 DOI: 10.1002/adma.202407794] [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: 05/31/2024] [Revised: 08/01/2024] [Indexed: 09/06/2024]
Abstract
The biochemical and biophysical properties of the extracellular matrix (ECM) play a pivotal role in regulating cellular behaviors such as proliferation, migration, and differentiation. Engineered protein-based hydrogels, with highly tunable multifunctional properties, have the potential to replicate key features of the native ECM. Formed by self-assembly or crosslinking, engineered protein-based hydrogels can induce a range of cell behaviors through bioactive and functional domains incorporated into the polymer backbone. Using recombinant techniques, the amino acid sequence of the protein backbone can be designed with precise control over the chain-length, folded structure, and cell-interaction sites. In this review, the modular design of engineered protein-based hydrogels from both a molecular- and network-level perspective are discussed, and summarize recent progress and case studies to highlight the diverse strategies used to construct biomimetic scaffolds. This review focuses on amino acid sequences that form structural blocks, bioactive blocks, and stimuli-responsive blocks designed into the protein backbone for highly precise and tunable control of scaffold properties. Both physical and chemical methods to stabilize dynamic protein networks with defined structure and bioactivity for cell culture applications are discussed. Finally, a discussion of future directions of engineered protein-based hydrogels as biomimetic cellular scaffolds is concluded.
Collapse
Affiliation(s)
- Yueming Liu
- Department of Materials Science & Engineering, Stanford University, Stanford, CA, 94305, USA
| | - Aidan E Gilchrist
- Department of Biomedical Engineering, University of California, Davis 451 Health Sciences Dr, GBSF 3315, Davis, CA, 95616, USA
| | - Sarah C Heilshorn
- Department of Materials Science & Engineering, 476 Lomita Mall, McCullough Room 246, Stanford, CA, 94305, USA
| |
Collapse
|
2
|
Zhang X, Gao X, Zhang X, Yao X, Kang X. Revolutionizing Intervertebral Disc Regeneration: Advances and Future Directions in Three-Dimensional Bioprinting of Hydrogel Scaffolds. Int J Nanomedicine 2024; 19:10661-10684. [PMID: 39464675 PMCID: PMC11505483 DOI: 10.2147/ijn.s469302] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 08/10/2024] [Indexed: 10/29/2024] Open
Abstract
Hydrogels are multifunctional platforms. Through reasonable structure and function design, they use material engineering to adjust their physical and chemical properties, such as pore size, microstructure, degradability, stimulus-response characteristics, etc. and have a variety of biomedical applications. Hydrogel three-dimensional (3D) printing has emerged as a promising technique for the precise deposition of cell-laden biomaterials, enabling the fabrication of intricate 3D structures such as artificial vertebrae and intervertebral discs (IVDs). Despite being in the early stages, 3D printing techniques have shown great potential in the field of regenerative medicine for the fabrication of various transplantable tissues within the human body. Currently, the utilization of engineered hydrogels as carriers or scaffolds for treating intervertebral disc degeneration (IVDD) presents numerous challenges. However, it remains an indispensable multifunctional manufacturing technology that is imperative in addressing the escalating issue of IVDD. Moreover, it holds the potential to serve as a micron-scale platform for a diverse range of applications. This review primarily concentrates on emerging treatment strategies for IVDD, providing an in-depth analysis of their merits and drawbacks, as well as the challenges that need to be addressed. Furthermore, it extensively explores the biological properties of hydrogels and various nanoscale biomaterial inks, compares different prevalent manufacturing processes utilized in 3D printing, and thoroughly examines the potential clinical applications and prospects of integrating 3D printing technology with hydrogels.
Collapse
Affiliation(s)
- Xiaobo Zhang
- Department of Spine Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’An, Shaanxi, P.R. China
| | - Xidan Gao
- Department of Spine Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’An, Shaanxi, P.R. China
| | - Xuefang Zhang
- Department of Spine Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’An, Shaanxi, P.R. China
| | - Xin Yao
- Department of Spine Surgery, Honghui Hospital, Xi’an Jiaotong University, Xi’An, Shaanxi, P.R. China
| | - Xin Kang
- Department of Sports Medicine, Honghui Hospital, Xi’an Jiao Tong University, Xi’An, Shaanxi, P.R. China
| |
Collapse
|
3
|
Camal Ruggieri IN, Aimone M, Juanes-Gusano D, Ibáñez-Fonseca A, Santiago O, Stur M, Mardegan Issa JP, Missana LR, Alonso M, Rodríguez-Cabello JC, Feldman S. Biocompatibility and bone regeneration with elastin-like recombinamer-based catalyst-free click gels. Sci Rep 2024; 14:20223. [PMID: 39215050 PMCID: PMC11364658 DOI: 10.1038/s41598-024-69658-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 08/07/2024] [Indexed: 09/04/2024] Open
Abstract
Large bone defects are a significant health problem today with various origins, including extensive trauma, tumours, or congenital musculoskeletal disorders. Tissue engineering, and in particular bone tissue engineering, aims to respond to this demand. As such, we propose a specific model based on Elastin-Like Recombinamers-based click-chemistry hydrogels given their high biocompatibility and their potent on bone regeneration effect conferred by different bioactive sequences. In this work we demonstrate, using biochemistry, histology, histomorphometry and imaging techniques, the biocompatibility of our matrix and its potent effect on bone regeneration in a model of bone parietal lesion in female New Zealand rabbits.
Collapse
Affiliation(s)
- I N Camal Ruggieri
- LABOATEM. Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory, School of Medicine, National Rosario University, Rosario, Argentina.
| | - M Aimone
- LABOATEM. Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory, School of Medicine, National Rosario University, Rosario, Argentina
| | - D Juanes-Gusano
- BIOFORGE Lab, University of Valladolid, CIBER-BBN, Valladolid, Spain
| | - A Ibáñez-Fonseca
- BIOFORGE Lab, University of Valladolid, CIBER-BBN, Valladolid, Spain
| | - O Santiago
- LABOATEM. Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory, School of Medicine, National Rosario University, Rosario, Argentina
| | - M Stur
- LABOATEM. Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory, School of Medicine, National Rosario University, Rosario, Argentina
- Diagnostic Imaging and Radiology, School of Medicine, National Rosario University, Rosario, Argentina
| | - J P Mardegan Issa
- Ribeirão Preto School of Dentistry, São Paulo University, São Paulo, Brazil
| | - L R Missana
- Experimental Pathology and Tissue Engineering Laboratory, School of Dentistry, National Tucumán University, Tucumán, Argentina
- Tissues Laboratory, IMMCA-CONICET, Tucumán, Argentina
| | - M Alonso
- BIOFORGE Lab, University of Valladolid, CIBER-BBN, Valladolid, Spain.
| | | | - S Feldman
- LABOATEM. Osteoarticular Biology, Tissue Engineering and Emerging Therapies Laboratory, School of Medicine, National Rosario University, Rosario, Argentina.
- Research Council of the National Rosario University (CIUNR) and CONICET, Rosario, Argentina.
| |
Collapse
|
4
|
Wang B, Hu S, Teng Y, Chen J, Wang H, Xu Y, Wang K, Xu J, Cheng Y, Gao X. Current advance of nanotechnology in diagnosis and treatment for malignant tumors. Signal Transduct Target Ther 2024; 9:200. [PMID: 39128942 PMCID: PMC11323968 DOI: 10.1038/s41392-024-01889-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 05/04/2024] [Accepted: 06/02/2024] [Indexed: 08/13/2024] Open
Abstract
Cancer remains a significant risk to human health. Nanomedicine is a new multidisciplinary field that is garnering a lot of interest and investigation. Nanomedicine shows great potential for cancer diagnosis and treatment. Specifically engineered nanoparticles can be employed as contrast agents in cancer diagnostics to enable high sensitivity and high-resolution tumor detection by imaging examinations. Novel approaches for tumor labeling and detection are also made possible by the use of nanoprobes and nanobiosensors. The achievement of targeted medication delivery in cancer therapy can be accomplished through the rational design and manufacture of nanodrug carriers. Nanoparticles have the capability to effectively transport medications or gene fragments to tumor tissues via passive or active targeting processes, thus enhancing treatment outcomes while minimizing harm to healthy tissues. Simultaneously, nanoparticles can be employed in the context of radiation sensitization and photothermal therapy to enhance the therapeutic efficacy of malignant tumors. This review presents a literature overview and summary of how nanotechnology is used in the diagnosis and treatment of malignant tumors. According to oncological diseases originating from different systems of the body and combining the pathophysiological features of cancers at different sites, we review the most recent developments in nanotechnology applications. Finally, we briefly discuss the prospects and challenges of nanotechnology in cancer.
Collapse
Affiliation(s)
- Bilan Wang
- Department of Pharmacy, Evidence-based Pharmacy Center, Children's Medicine Key Laboratory of Sichuan Province, West China Second University Hospital, Sichuan University, Chengdu, 610041, China
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Shiqi Hu
- Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
- Department of Gynecology and Obstetrics, Development and Related Diseases of Women and Children Key Laboratory of Sichuan Province, Key Laboratory of Birth Defects and Related Diseases of Women and Children, Ministry of Education, West China Second University Hospital, Sichuan University, Chengdu, 610041, P.R. China
| | - Yan Teng
- Institute of Laboratory Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610072, P.R. China
| | - Junli Chen
- West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu, 610041, China
| | - Haoyuan Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yezhen Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Kaiyu Wang
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Jianguo Xu
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China
| | - Yongzhong Cheng
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| | - Xiang Gao
- Department of Neurosurgery and Institute of Neurosurgery, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, West China Medical School, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, 610041, China.
| |
Collapse
|
5
|
Yu C, Qiu Y, Yao F, Wang C, Li J. Chemically Programmed Hydrogels for Spatiotemporal Modulation of the Cardiac Pathological Microenvironment. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2404264. [PMID: 38830198 DOI: 10.1002/adma.202404264] [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: 03/24/2024] [Revised: 05/20/2024] [Indexed: 06/05/2024]
Abstract
After myocardial infarction (MI), sustained ischemic events induce pathological microenvironments characterized by ischemia-hypoxia, oxidative stress, inflammatory responses, matrix remodeling, and fibrous scarring. Conventional clinical therapies lack spatially targeted and temporally responsive modulation of the infarct microenvironment, leading to limited myocardial repair. Engineered hydrogels have a chemically programmed toolbox for minimally invasive localization of the pathological microenvironment and personalized responsive modulation over different pathological periods. Chemically programmed strategies for crosslinking interactions, interfacial binding, and topological microstructures in hydrogels enable minimally invasive implantation and in situ integration tailored to the myocardium. This enhances substance exchange and signal interactions within the infarcted microenvironment. Programmed responsive polymer networks, intelligent micro/nanoplatforms, and biological therapeutic cues contribute to the formation of microenvironment-modulated hydrogels with precise targeting, spatiotemporal control, and on-demand feedback. Therefore, this review summarizes the features of the MI microenvironment and chemically programmed schemes for hydrogels to conform, integrate, and modulate the cardiac pathological microenvironment. Chemically programmed strategies for oxygen-generating, antioxidant, anti-inflammatory, provascular, and electrointegrated hydrogels to stimulate iterative and translational cardiac tissue engineering are discussed.
Collapse
Affiliation(s)
- Chaojie Yu
- School of Chemical Engineering and Technology, Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering, Tianjin University, Tianjin, 300350, China
| | - Yuwei Qiu
- School of Chemical Engineering and Technology, Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering, Tianjin University, Tianjin, 300350, China
| | - Fanglian Yao
- School of Chemical Engineering and Technology, Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering, Tianjin University, Tianjin, 300350, China
| | - Changyong Wang
- Tissue Engineering Research Center, Beijing Institute of Basic Medical Sciences, Beijing, 100850, China
| | - Junjie Li
- School of Chemical Engineering and Technology, Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering, Tianjin University, Tianjin, 300350, China
| |
Collapse
|
6
|
Krukiewicz K, Contessotto P, Nedjari S, Martino MM, Redenski I, Gabet Y, Speranza G, O'Brien T, Altankov G, Awaja F. Clinical potential of plasma-functionalized graphene oxide ultrathin sheets for bone and blood vessel regeneration: Insights from cellular and animal models. BIOMATERIALS ADVANCES 2024; 161:213867. [PMID: 38669824 DOI: 10.1016/j.bioadv.2024.213867] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/29/2023] [Revised: 04/01/2024] [Accepted: 04/18/2024] [Indexed: 04/28/2024]
Abstract
Graphene and graphene oxide (GO), due to their unique chemical and physical properties, possess biochemical characteristics that can trigger intercellular signals promoting tissue regeneration. Clinical applications of thin GO-derived sheets have inspired the development of various tissue regeneration and repair approaches. In this study, we demonstrate that ultrathin sheets of plasma-functionalized and reduced GO, with the oxygen content ranging from 3.2 % to 22 % and the nitrogen content from 0 % to 8.3 %, retain their essential mechanical and molecular integrity, and exhibit robust potential for regenerating bone tissue and blood vessels across multiple cellular and animal models. Initially, we observed the growth of blood vessels and bone tissue in vitro using these functionalized GO sheets on human adipose-derived mesenchymal stem cells and umbilical vein endothelial cells. Remarkably, our study indicates a 2.5-fold increase in mineralization and two-fold increase in tubule formation even in media lacking osteogenic and angiogenic supplements. Subsequently, we observed the initiation, conduction, and formation of bone and blood vessels in a rat tibial osteotomy model, evident from a marked 4-fold increase in the volume of low radio-opacity bone tissue and a significant elevation in connectivity density, all without the use of stem cells or growth factors. Finally, we validated these findings in a mouse critical-size calvarial defect model (33 % higher healing rate) and a rat skin lesion model (up to 2.5-fold increase in the number of blood vessels, and 35 % increase in blood vessels diameter). This study elucidates the pro-osteogenic and pro-angiogenic properties of both pristine and plasma-treated GO ultrathin films. These properties suggest their significant potential for clinical applications, and as valuable biomaterials for investigating fundamental aspects of bone and blood vessel regeneration.
Collapse
Affiliation(s)
- Katarzyna Krukiewicz
- Department of Physical Chemistry and Technology of Polymers, Silesian University of Technology, Gliwice, Poland; Centre for Organic and Nanohybrid Electronics, Silesian University of Technology, Gliwice, Poland.
| | - Paolo Contessotto
- Department of Molecular Medicine, Università degli Studi di Padova, Padua, Italy.
| | - Salima Nedjari
- Molecular Dynamics at Cell-Biomaterial Interface, Institute for Bioengineering of Catalonia, Barcelona, Spain
| | - Mikaël M Martino
- European Molecular Biology Laboratory Australia, Australian Regenerative Medicine Institute, Monash University, Melbourne, VIC, Australia.
| | - Idan Redenski
- Department of Biomedical Engineering, Technion - Israel Institute of Technology, Haifa 32000, Israel
| | - Yankel Gabet
- Department of Anatomy and Anthropology, Faculty of Medical and Health Sciences, Tel-Aviv University, Tel-Aviv, Israel.
| | | | - Timothy O'Brien
- Centre for Research in Medical Devices, University of Galway, Galway, Ireland.
| | - George Altankov
- ICREA & Institute for Bioengineering of Catalonia, Barcelona, Spain; Medical University Pleven, Bulgaria
| | - Firas Awaja
- Department of Medicine, University of Galway, Galway, Ireland; Engmat Ltd., Clybaun Road, Galway, Ireland.
| |
Collapse
|
7
|
Kruczkowska W, Gałęziewska J, Grabowska K, Liese G, Buczek P, Kłosiński KK, Kciuk M, Pasieka Z, Kałuzińska-Kołat Ż, Kołat D. Biomedical Trends in Stimuli-Responsive Hydrogels with Emphasis on Chitosan-Based Formulations. Gels 2024; 10:295. [PMID: 38786212 PMCID: PMC11121652 DOI: 10.3390/gels10050295] [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: 03/21/2024] [Revised: 04/13/2024] [Accepted: 04/23/2024] [Indexed: 05/25/2024] Open
Abstract
Biomedicine is constantly evolving to ensure a significant and positive impact on healthcare, which has resulted in innovative and distinct requisites such as hydrogels. Chitosan-based formulations stand out for their versatile utilization in drug encapsulation, transport, and controlled release, which is complemented by their biocompatibility, biodegradability, and non-immunogenic nature. Stimuli-responsive hydrogels, also known as smart hydrogels, have strictly regulated release patterns since they respond and adapt based on various external stimuli. Moreover, they can imitate the intrinsic tissues' mechanical, biological, and physicochemical properties. These characteristics allow stimuli-responsive hydrogels to provide cutting-edge, effective, and safe treatment. Constant progress in the field necessitates an up-to-date summary of current trends and breakthroughs in the biomedical application of stimuli-responsive chitosan-based hydrogels, which was the aim of this review. General data about hydrogels sensitive to ions, pH, redox potential, light, electric field, temperature, and magnetic field are recapitulated. Additionally, formulations responsive to multiple stimuli are mentioned. Focusing on chitosan-based smart hydrogels, their multifaceted utilization was thoroughly described. The vast application spectrum encompasses neurological disorders, tumors, wound healing, and dermal infections. Available data on smart chitosan hydrogels strongly support the idea that current approaches and developing novel solutions are worth improving. The present paper constitutes a valuable resource for researchers and practitioners in the currently evolving field.
Collapse
Affiliation(s)
- Weronika Kruczkowska
- Department of Biomedicine and Experimental Surgery, Faculty of Medicine, Medical University of Lodz, Narutowicza 60, 90-136 Lodz, Poland; (W.K.); (J.G.); (K.G.); (G.L.); (P.B.); (K.K.K.); (Z.P.); (Ż.K.-K.)
| | - Julia Gałęziewska
- Department of Biomedicine and Experimental Surgery, Faculty of Medicine, Medical University of Lodz, Narutowicza 60, 90-136 Lodz, Poland; (W.K.); (J.G.); (K.G.); (G.L.); (P.B.); (K.K.K.); (Z.P.); (Ż.K.-K.)
| | - Katarzyna Grabowska
- Department of Biomedicine and Experimental Surgery, Faculty of Medicine, Medical University of Lodz, Narutowicza 60, 90-136 Lodz, Poland; (W.K.); (J.G.); (K.G.); (G.L.); (P.B.); (K.K.K.); (Z.P.); (Ż.K.-K.)
| | - Gabriela Liese
- Department of Biomedicine and Experimental Surgery, Faculty of Medicine, Medical University of Lodz, Narutowicza 60, 90-136 Lodz, Poland; (W.K.); (J.G.); (K.G.); (G.L.); (P.B.); (K.K.K.); (Z.P.); (Ż.K.-K.)
| | - Paulina Buczek
- Department of Biomedicine and Experimental Surgery, Faculty of Medicine, Medical University of Lodz, Narutowicza 60, 90-136 Lodz, Poland; (W.K.); (J.G.); (K.G.); (G.L.); (P.B.); (K.K.K.); (Z.P.); (Ż.K.-K.)
| | - Karol Kamil Kłosiński
- Department of Biomedicine and Experimental Surgery, Faculty of Medicine, Medical University of Lodz, Narutowicza 60, 90-136 Lodz, Poland; (W.K.); (J.G.); (K.G.); (G.L.); (P.B.); (K.K.K.); (Z.P.); (Ż.K.-K.)
| | - Mateusz Kciuk
- Department of Molecular Biotechnology and Genetics, University of Lodz, Banacha 12/16, 90-237 Lodz, Poland;
| | - Zbigniew Pasieka
- Department of Biomedicine and Experimental Surgery, Faculty of Medicine, Medical University of Lodz, Narutowicza 60, 90-136 Lodz, Poland; (W.K.); (J.G.); (K.G.); (G.L.); (P.B.); (K.K.K.); (Z.P.); (Ż.K.-K.)
| | - Żaneta Kałuzińska-Kołat
- Department of Biomedicine and Experimental Surgery, Faculty of Medicine, Medical University of Lodz, Narutowicza 60, 90-136 Lodz, Poland; (W.K.); (J.G.); (K.G.); (G.L.); (P.B.); (K.K.K.); (Z.P.); (Ż.K.-K.)
- Department of Functional Genomics, Faculty of Medicine, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland
| | - Damian Kołat
- Department of Biomedicine and Experimental Surgery, Faculty of Medicine, Medical University of Lodz, Narutowicza 60, 90-136 Lodz, Poland; (W.K.); (J.G.); (K.G.); (G.L.); (P.B.); (K.K.K.); (Z.P.); (Ż.K.-K.)
- Department of Functional Genomics, Faculty of Medicine, Medical University of Lodz, Zeligowskiego 7/9, 90-752 Lodz, Poland
| |
Collapse
|
8
|
Puertas-Bartolomé M, Venegas-Bustos D, Acosta S, Rodríguez-Cabello JC. Contribution of the ELRs to the development of advanced in vitro models. Front Bioeng Biotechnol 2024; 12:1363865. [PMID: 38650751 PMCID: PMC11033926 DOI: 10.3389/fbioe.2024.1363865] [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: 12/31/2023] [Accepted: 03/18/2024] [Indexed: 04/25/2024] Open
Abstract
Developing in vitro models that accurately mimic the microenvironment of biological structures or processes holds substantial promise for gaining insights into specific biological functions. In the field of tissue engineering and regenerative medicine, in vitro models able to capture the precise structural, topographical, and functional complexity of living tissues, prove to be valuable tools for comprehending disease mechanisms, assessing drug responses, and serving as alternatives or complements to animal testing. The choice of the right biomaterial and fabrication technique for the development of these in vitro models plays an important role in their functionality. In this sense, elastin-like recombinamers (ELRs) have emerged as an important tool for the fabrication of in vitro models overcoming the challenges encountered in natural and synthetic materials due to their intrinsic properties, such as phase transition behavior, tunable biological properties, viscoelasticity, and easy processability. In this review article, we will delve into the use of ELRs for molecular models of intrinsically disordered proteins (IDPs), as well as for the development of in vitro 3D models for regenerative medicine. The easy processability of the ELRs and their rational design has allowed their use for the development of spheroids and organoids, or bioinks for 3D bioprinting. Thus, incorporating ELRs into the toolkit of biomaterials used for the fabrication of in vitro models, represents a transformative step forward in improving the accuracy, efficiency, and functionality of these models, and opening up a wide range of possibilities in combination with advanced biofabrication techniques that remains to be explored.
Collapse
Affiliation(s)
- María Puertas-Bartolomé
- Technical Proteins Nanobiotechnology, S.L. (TPNBT), Valladolid, Spain
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER's Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| | - Desiré Venegas-Bustos
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER's Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| | - Sergio Acosta
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER's Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| | - José Carlos Rodríguez-Cabello
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER's Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| |
Collapse
|
9
|
Zhao Y, Dong H, Xia Q, Wang Y, Zhu L, Hu Z, Xia J, Mao Q, Weng Z, Yi J, Feng S, Jiang Y, Liao W, Xin Z. A new strategy for intervertebral disc regeneration: The synergistic potential of mesenchymal stem cells and their extracellular vesicles with hydrogel scaffolds. Biomed Pharmacother 2024; 172:116238. [PMID: 38308965 DOI: 10.1016/j.biopha.2024.116238] [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: 12/07/2023] [Revised: 01/28/2024] [Accepted: 01/30/2024] [Indexed: 02/05/2024] Open
Abstract
Intervertebral disc degeneration (IDD) is a disease that severely affects spinal health and is prevalent worldwide. Mesenchymal stem cells (MSCs) and their derived extracellular vesicles (EVs) have regenerative potential and have emerged as promising therapeutic tools for treating degenerative discs. However, challenges such as the harsh microenvironment of degenerated intervertebral discs and EVs' limited stability and efficacy have hindered their clinical application. In recent years, hydrogels have attracted much attention in the field of IDD therapy because they can mimic the physiologic microenvironment of the disc and provide a potential solution by providing a suitable growth environment for MSCs and EVs. This review introduced the biological properties of MSCs and their derived EVs, summarized the research on the application of MSCs and EVs in IDD, summarized the current clinical trial studies of MSCs and EVs, and also explored the mechanism of action of MSCs and EVs in intervertebral discs. In addition, plenty of research elaborated on the mechanism of action of different classified hydrogels in tissue engineering, the synergistic effect of MSCs and EVs in promoting intervertebral disc regeneration, and their wide application in treating IDD. Finally, the challenges and problems still faced by hydrogel-loaded MSCs and EVs in the treatment of IDD are summarized, and potential solutions are proposed. This paper outlines the synergistic effects of MSCs and EVs in treating IDD in combination with hydrogels and aims to provide theoretical references for future related studies.
Collapse
Affiliation(s)
- Yan Zhao
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Huaize Dong
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Qiuqiu Xia
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Yanyang Wang
- Department of Cell Engineering Laboratory, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, China
| | - Lu Zhu
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Zongyue Hu
- Department of Pain Rehabilitation, Affiliated Sinopharm Gezhouba Central Hospital, Third Clinical Medical College of Three Gorges University, Yichang 443003, Hubei, China
| | - Jiyue Xia
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Qiming Mao
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Zijing Weng
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Jiangbi Yi
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Shuai Feng
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Youhong Jiang
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Wenbo Liao
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, China
| | - Zhijun Xin
- Department of Orthopedic Surgery, Affiliated Hospital of Zunyi Medical University, Zunyi 563000, Guizhou, China; Institut Curie, PSL Research University, CNRS UMR3244, Dynamics of Genetic Information, Sorbonne Université, 75005 Paris, France.
| |
Collapse
|
10
|
Xiao N, Xiong S, Zhou Z, Zhong M, Bai H, Li Q, Tang Y, Xie J. Recent progress in biomaterials-driven ferroptosis for cancer therapy. Biomater Sci 2024; 12:288-307. [PMID: 38189655 DOI: 10.1039/d3bm01832f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Ferroptosis, first suggested in 2012, is a type of non-apoptotic programmed cell death caused by the buildup of lipid peroxidation and marked by an overabundance of oxidized poly unsaturated fatty acids. During the last decade, researchers have uncovered the formation of ferroptosis and created multiple drugs aimed at it, but due to poor selectivity and pharmacokinetics, clinical application has been hindered. In recent years, biomedical discoveries and developments in nanotechnology have spurred the investigation of ferroptosis nanomaterials, providing new opportunities for the ferroptosis driven tumours treatment. Additionally, hydrogels have been widely studied in ferroptosis because of their unique 3D structure and excellent controllability. By using these biomaterials, it is possible to achieve controlled release and targeted delivery of drugs, thus increasing the potency of the drugs and minimizing adverse effects. Therefore, summarizing the biomedical nanomaterials, including hydrogels, used in ferroptosis for cancer therapy is a must. This article provides an overview of ferroptosis, detailing its properties and underlying mechanisms. It also categorizes and reviews the use of various nanomaterials in ferroptosis, along with relevant explanations and illustrations. In addition, we discuss the opportunities and challenges facing the application of nanomaterials in ferroptosis. Finally, the development prospects of this field are prospected. This review is intended to provide a foundation for the development and application of biomedical nanomaterials in ferroptosis.
Collapse
Affiliation(s)
- Nianting Xiao
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
| | - Su Xiong
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
| | - Ziwei Zhou
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
| | - Min Zhong
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
| | - Huayang Bai
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
| | - Qiyu Li
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
| | - Yaqin Tang
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
- College of Pharmaceutical Sciences, Southwest University, Chongqing 400715, China.
| | - Jing Xie
- Chongqing Key Laboratory of Medicinal Chemistry and Molecular Pharmacology, Chongqing University of Technology, Chongqing 400054, China
| |
Collapse
|
11
|
Zhang Q, Kuang G, Wang L, Duan P, Sun W, Ye F. Designing Bioorthogonal Reactions for Biomedical Applications. RESEARCH (WASHINGTON, D.C.) 2023; 6:0251. [PMID: 38107023 PMCID: PMC10723801 DOI: 10.34133/research.0251] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Accepted: 09/25/2023] [Indexed: 12/19/2023]
Abstract
Bioorthogonal reactions are a class of chemical reactions that can be carried out in living organisms without interfering with other reactions, possessing high yield, high selectivity, and high efficiency. Since the first proposal of the conception by Professor Carolyn Bertozzi in 2003, bioorthogonal chemistry has attracted great attention and has been quickly developed. As an important chemical biology tool, bioorthogonal reactions have been applied broadly in biomedicine, including bio-labeling, nucleic acid functionalization, drug discovery, drug activation, synthesis of antibody-drug conjugates, and proteolysis-targeting chimeras. Given this, we summarized the basic knowledge, development history, research status, and prospects of bioorthogonal reactions and their biomedical applications. The main purpose of this paper is to furnish an overview of the intriguing bioorthogonal reactions in a variety of biomedical applications and to provide guidance for the design of novel reactions to enrich bioorthogonal chemistry toolkits.
Collapse
Affiliation(s)
- Qingfei Zhang
- Wenzhou Institute,
University of Chinese Academy of Sciences, Wenzhou 325001, China
- Beijing National Laboratory for Condensed Matter Physics, Institute of Physics,
Chinese Academy of Sciences, Beijing 100190, China
| | - Gaizhen Kuang
- Wenzhou Institute,
University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Li Wang
- Wenzhou Institute,
University of Chinese Academy of Sciences, Wenzhou 325001, China
| | - Ping Duan
- Department of Obstetrics and Gynecology, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325027, China
| | - Weijian Sun
- Wenzhou Institute,
University of Chinese Academy of Sciences, Wenzhou 325001, China
- Department of Gastrointestinal Surgery, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, 325027, China
| | - Fangfu Ye
- Wenzhou Institute,
University of Chinese Academy of Sciences, Wenzhou 325001, China
- Beijing National Laboratory for Condensed Matter Physics, Institute of Physics,
Chinese Academy of Sciences, Beijing 100190, China
| |
Collapse
|
12
|
Vetter VC, Bouten CVC, van der Pol A. Hydrogels for Cardiac Restorative Support: Relevance of Gelation Mechanisms for Prospective Clinical Use. Curr Heart Fail Rep 2023; 20:519-529. [PMID: 37812347 PMCID: PMC10746579 DOI: 10.1007/s11897-023-00630-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/20/2023] [Indexed: 10/10/2023]
Abstract
PURPOSE OF REVIEW Cardiac tissue regenerative strategies have gained much traction over the years, in particular those utilizing hydrogels. With our review, and with special focus on supporting post-myocardial infarcted tissue, we aim to provide insights in determining crucial design considerations of a hydrogel and the implications these could have for future clinical use. RECENT FINDINGS To date, two hydrogel delivery strategies are being explored, cardiac injection or patch, to treat myocardial infarction. Recent advances have demonstrated that the mechanism by which a hydrogel is gelated (i.e., physically or chemically cross-linked) not only impacts the biocompatibility, mechanical properties, and chemical structure, but also the route of delivery of the hydrogel and thus its effect on cardiac repair. With regard to cardiac regeneration, various hydrogels have been developed with the ability to function as a delivery system for therapeutic strategies (e.g., drug and stem cells treatments), as well as a scaffold to guide cardiac tissue regeneration following myocardial infarction. However, these developments remain within the experimental and pre-clinical realm and have yet to transition towards the clinical setting.
Collapse
Affiliation(s)
- Valentine C Vetter
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Carlijn V C Bouten
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands
| | - Atze van der Pol
- Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, The Netherlands.
- Institute for Complex Molecular Systems, Eindhoven University of Technology, Eindhoven, The Netherlands.
| |
Collapse
|
13
|
Guo Y, Liu S, Jing D, Liu N, Luo X. The construction of elastin-like polypeptides and their applications in drug delivery system and tissue repair. J Nanobiotechnology 2023; 21:418. [PMID: 37951928 PMCID: PMC10638729 DOI: 10.1186/s12951-023-02184-8] [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: 03/29/2023] [Accepted: 11/02/2023] [Indexed: 11/14/2023] Open
Abstract
Elastin-like polypeptides (ELPs) are thermally responsive biopolymers derived from natural elastin. These peptides have a low critical solution temperature phase behavior and can be used to prepare stimuli-responsive biomaterials. Through genetic engineering, biomaterials prepared from ELPs can have unique and customizable properties. By adjusting the amino acid sequence and length of ELPs, nanostructures, such as micelles and nanofibers, can be formed. Correspondingly, ELPs have been used for improving the stability and prolonging drug-release time. Furthermore, ELPs have widespread use in tissue repair due to their biocompatibility and biodegradability. Here, this review summarizes the basic property composition of ELPs and the methods for modulating their phase transition properties, discusses the application of drug delivery system and tissue repair and clarifies the current challenges and future directions of ELPs in applications.
Collapse
Affiliation(s)
- Yingshu Guo
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China.
| | - Shiwei Liu
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Dan Jing
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Nianzu Liu
- School of Chemistry and Chemical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan, 250353, China
| | - Xiliang Luo
- Key Laboratory of Optic-Electric Sensing and Analytical Chemistry for Life Science, MOE, College of Chemistry and Molecular Engineering, Qingdao University of Science and Technology, Qingdao, 266042, China.
| |
Collapse
|
14
|
Blanco-Fernandez B, Ibañez-Fonseca A, Orbanic D, Ximenes-Carballo C, Perez-Amodio S, Rodríguez-Cabello JC, Engel E. Elastin-like Recombinamer Hydrogels as Platforms for Breast Cancer Modeling. Biomacromolecules 2023; 24:4408-4418. [PMID: 36597885 PMCID: PMC10565832 DOI: 10.1021/acs.biomac.2c01080] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 12/07/2022] [Indexed: 01/05/2023]
Abstract
The involvement of the extracellular matrix (ECM) in tumor progression has motivated the development of biomaterials mimicking the tumor ECM to develop more predictive cancer models. Particularly, polypeptides based on elastin could be an interesting approach to mimic the ECM due to their tunable properties. Here, we demonstrated that elastin-like recombinamer (ELR) hydrogels can be suitable biomaterials to develop breast cancer models. This hydrogel was formed by two ELR polypeptides, one containing sequences biodegradable by matrix metalloproteinase and cyclooctyne and the other carrying arginylglycylaspartic acid and azide groups to allow cell adhesion, biodegradability, and suitable stiffness through "click-chemistry" cross-linking. Our findings show that breast cancer or nontumorigenic breast cells showed high viability and cell proliferation for up to 7 days. MCF7 and MCF10A formed spheroids whereas MDA-MB-231 formed cell networks, with the expression of ECM and high drug resistance in all cases, evidencing that ELR hydrogels are a promising biomaterial for breast cancer modeling.
Collapse
Affiliation(s)
- Barbara Blanco-Fernandez
- Institute
for Bioengineering of Catalonia (IBEC), The Barcelona Institute of
Science and Technology (BIST), Baldiri Reixac 10-12, Barcelona 08028, Spain
- CIBER
en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid 28029, Spain
| | - Arturo Ibañez-Fonseca
- BIOFORGE
Lab, CIBER-BBN, University of Valladolid, Paseo de Belén 19, 47011 Valladolid, Spain
| | - Doriana Orbanic
- BIOFORGE
Lab, CIBER-BBN, University of Valladolid, Paseo de Belén 19, 47011 Valladolid, Spain
| | - Celia Ximenes-Carballo
- Institute
for Bioengineering of Catalonia (IBEC), The Barcelona Institute of
Science and Technology (BIST), Baldiri Reixac 10-12, Barcelona 08028, Spain
| | - Soledad Perez-Amodio
- Institute
for Bioengineering of Catalonia (IBEC), The Barcelona Institute of
Science and Technology (BIST), Baldiri Reixac 10-12, Barcelona 08028, Spain
| | | | - Elisabeth Engel
- Institute
for Bioengineering of Catalonia (IBEC), The Barcelona Institute of
Science and Technology (BIST), Baldiri Reixac 10-12, Barcelona 08028, Spain
- CIBER
en Bioingeniería, Biomateriales y Nanomedicina, CIBER-BBN, Madrid 28029, Spain
- IMEM-BRT
Group, Department of Materials Science and Engineering, EEBE, Technical University of Catalonia (UPC), Barcelona 08019, Spain
| |
Collapse
|
15
|
Yu C, Shi M, He S, Yao M, Sun H, Yue Z, Qiu Y, Liu B, Liang L, Zhao Z, Yao F, Zhang H, Li J. Chronological adhesive cardiac patch for synchronous mechanophysiological monitoring and electrocoupling therapy. Nat Commun 2023; 14:6226. [PMID: 37803005 PMCID: PMC10558550 DOI: 10.1038/s41467-023-42008-9] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2023] [Accepted: 09/22/2023] [Indexed: 10/08/2023] Open
Abstract
With advances in tissue engineering and bioelectronics, flexible electronic hydrogels that allow conformal tissue integration, online precision diagnosis, and simultaneous tissue regeneration are expected to be the next-generation platform for the treatment of myocardial infarction. Here, we report a functionalized polyaniline-based chronological adhesive hydrogel patch (CAHP) that achieves spatiotemporally selective and conformal embedded integration with a moist and dynamic epicardium surface. Significantly, CAHP has high adhesion toughness, rapid self-healing ability, and enhanced electrochemical performance, facilitating sensitive sensing of cardiac mechanophysiology-mediated microdeformations and simultaneous improvement of myocardial fibrosis-induced electrophysiology. As a result, the flexible CAHP platform monitors diastolic-systolic amplitude and rhythm in the infarcted myocardium online while effectively inhibiting ventricular remodeling, promoting vascular regeneration, and improving electrophysiological function through electrocoupling therapy. Therefore, this diagnostic and therapeutic integration provides a promising monitorable treatment protocol for cardiac disease.
Collapse
Affiliation(s)
- Chaojie Yu
- School of Chemical Engineering and Technology, Tianjin University, 300350, Tianjin, China
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering, Ministry of Education, Tianjin University, 300350, Tianjin, China
| | - Mingyue Shi
- School of Chemical Engineering and Technology, Tianjin University, 300350, Tianjin, China
- School of Chemical Science and Engineering, Tongji University, 200092, Shanghai, China
| | - Shaoshuai He
- School of Chemical Engineering and Technology, Tianjin University, 300350, Tianjin, China
- Thrust of Sustainable Energy and Environment, The Hong Kong University of Science and Technology (Guangzhou), 511400, Guangzhou, China
| | - Mengmeng Yao
- School of Chemical Engineering and Technology, Tianjin University, 300350, Tianjin, China
| | - Hong Sun
- School of Basic Medical Sciences, North China University of Science and Technology, 063210, Tangshan, China.
| | - Zhiwei Yue
- School of Basic Medical Sciences, North China University of Science and Technology, 063210, Tangshan, China
| | - Yuwei Qiu
- School of Chemical Engineering and Technology, Tianjin University, 300350, Tianjin, China
| | - Baijun Liu
- School of Chemical Engineering and Technology, Tianjin University, 300350, Tianjin, China
| | - Lei Liang
- School of Chemical Engineering and Technology, Tianjin University, 300350, Tianjin, China
| | - Zhongming Zhao
- School of Chemical Engineering and Technology, Tianjin University, 300350, Tianjin, China
| | - Fanglian Yao
- School of Chemical Engineering and Technology, Tianjin University, 300350, Tianjin, China.
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering, Ministry of Education, Tianjin University, 300350, Tianjin, China.
| | - Hong Zhang
- School of Chemical Engineering and Technology, Tianjin University, 300350, Tianjin, China.
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering, Ministry of Education, Tianjin University, 300350, Tianjin, China.
| | - Junjie Li
- School of Chemical Engineering and Technology, Tianjin University, 300350, Tianjin, China.
- Frontiers Science Center for Synthetic Biology and Key Laboratory of Systems Bioengineering, Ministry of Education, Tianjin University, 300350, Tianjin, China.
| |
Collapse
|
16
|
Wang Z, Yang N, Hou Y, Li Y, Yin C, Yang E, Cao H, Hu G, Xue J, Yang J, Liao Z, Wang W, Sun D, Fan C, Zheng L. L-Arginine-Loaded Gold Nanocages Ameliorate Myocardial Ischemia/Reperfusion Injury by Promoting Nitric Oxide Production and Maintaining Mitochondrial Function. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2302123. [PMID: 37449329 PMCID: PMC10502842 DOI: 10.1002/advs.202302123] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/02/2023] [Revised: 06/01/2023] [Indexed: 07/18/2023]
Abstract
Cardiovascular disease is the leading cause of death worldwide. Reperfusion therapy is vital to patient survival after a heart attack but can cause myocardial ischemia/reperfusion injury (MI/RI). Nitric oxide (NO) can ameliorate MI/RI and is a key molecule for drug development. However, reactive oxygen species (ROS) can easily oxidize NO to peroxynitrite, which causes secondary cardiomyocyte damage. Herein, L-arginine-loaded selenium-coated gold nanocages (AAS) are designed, synthesized, and modified with PCM (WLSEAGPVVTVRALRGTGSW) to obtain AASP, which targets cardiomyocytes, exhibits increased cellular uptake, and improves photoacoustic imaging in vitro and in vivo. AASP significantly inhibits oxygen glucose deprivation/reoxygenation (OGD/R)-induced H9C2 cell cytotoxicity and apoptosis. Mechanistic investigation revealed that AASP improves mitochondrial membrane potential (MMP), restores ATP synthase activity, blocks ROS generation, and prevents NO oxidation, and NO blocks ROS release by regulating the closing of the mitochondrial permeability transition pore (mPTP). AASP administration in vivo improves myocardial function, inhibits myocardial apoptosis and fibrosis, and ultimately attenuates MI/RI in rats by maintaining mitochondrial function and regulating NO signaling. AASP shows good safety and biocompatibility in vivo. This findings confirm the rational design of AASP, which can provide effective treatment for MI/RI.
Collapse
Affiliation(s)
- Zekun Wang
- School of Life SciencesAnhui Agricultural UniversityHefeiAnhui230036China
| | - Nana Yang
- School of Bioscience and TechnologyWeifang Key Laboratory of Animal Model Research on Cardiovascular and Cerebrovascular DiseasesWeifang Medical UniversityWeifang261053China
| | - Yajun Hou
- Department of NeurologySecond Affiliated HospitalShandong First Medical University & Shandong Academy of Medical SciencesTaianShandong271000China
| | - Yuqing Li
- School of Life SciencesAnhui Agricultural UniversityHefeiAnhui230036China
| | - Chenyang Yin
- School of Life SciencesAnhui Agricultural UniversityHefeiAnhui230036China
| | - Endong Yang
- School of Life SciencesAnhui Agricultural UniversityHefeiAnhui230036China
| | - Huanhuan Cao
- The Institute of Cardiovascular Sciences and Institute of Systems BiomedicineSchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingNHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory PeptidesBeijing Key Laboratory of Cardiovascular Receptors ResearchHealth Science CenterPeking UniversityBeijing100191China
| | - Gaofei Hu
- The Institute of Cardiovascular Sciences and Institute of Systems BiomedicineSchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingNHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory PeptidesBeijing Key Laboratory of Cardiovascular Receptors ResearchHealth Science CenterPeking UniversityBeijing100191China
| | - Jing Xue
- Department of NeurologyChina National Clinical Research Center for Neurological DiseasesBeijing Tiantan HospitalCapital Medical UniversityBeijing100070China
| | - Jialei Yang
- Department of NeurologyChina National Clinical Research Center for Neurological DiseasesBeijing Tiantan HospitalCapital Medical UniversityBeijing100070China
| | - Ziyu Liao
- School of Life SciencesAnhui Agricultural UniversityHefeiAnhui230036China
| | - Weiyun Wang
- School of Life SciencesAnhui Agricultural UniversityHefeiAnhui230036China
| | - Dongdong Sun
- School of Life SciencesAnhui Agricultural UniversityHefeiAnhui230036China
| | - Cundong Fan
- Department of NeurologySecond Affiliated HospitalShandong First Medical University & Shandong Academy of Medical SciencesTaianShandong271000China
| | - Lemin Zheng
- The Institute of Cardiovascular Sciences and Institute of Systems BiomedicineSchool of Basic Medical SciencesState Key Laboratory of Vascular Homeostasis and RemodelingNHC Key Laboratory of Cardiovascular Molecular Biology and Regulatory PeptidesBeijing Key Laboratory of Cardiovascular Receptors ResearchHealth Science CenterPeking UniversityBeijing100191China
- Department of NeurologyChina National Clinical Research Center for Neurological DiseasesBeijing Tiantan HospitalCapital Medical UniversityBeijing100070China
| |
Collapse
|
17
|
Malektaj H, Nour S, Imani R, Siadati MH. Angiogenesis induction as a key step in cardiac tissue Regeneration: From angiogenic agents to biomaterials. Int J Pharm 2023; 643:123233. [PMID: 37460050 DOI: 10.1016/j.ijpharm.2023.123233] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2023] [Revised: 07/02/2023] [Accepted: 07/14/2023] [Indexed: 07/23/2023]
Abstract
Cardiovascular diseases are the leading cause of death worldwide. After myocardial infarction, the vascular supply of the heart is damaged or blocked, leading to the formation of scar tissue, followed by several cardiac dysfunctions or even death. In this regard, induction of angiogenesis is considered as a vital process for supplying nutrients and oxygen to the cells in cardiac tissue engineering. The current review aims to summarize different approaches of angiogenesis induction for effective cardiac tissue repair. Accordingly, a comprehensive classification of induction of pro-angiogenic signaling pathways through using engineered biomaterials, drugs, angiogenic factors, as well as combinatorial approaches is introduced as a potential platform for cardiac regeneration application. The angiogenic induction for cardiac repair can enhance patient treatment outcomes and generate economic prospects for the biomedical industry. The development and commercialization of angiogenesis methods often involves collaboration between academic institutions, research organizations, and biomedical companies.
Collapse
Affiliation(s)
- Haniyeh Malektaj
- Department of Materials and Production, Aalborg University, Fibigerstraede 16, Aalborg 9220, Denmark
| | - Shirin Nour
- Department of Biomedical Engineering, Graeme Clark Institute, The University of Melbourne, VIC 3010, Australia; Department of Chemical Engineering, The University of Melbourne, VIC 3010, Australia
| | - Rana Imani
- Department of Biomedical Engineering, Amirkabir University of Technology, Tehran, Iran.
| | - Mohammad H Siadati
- Materials Science and Engineering Faculty, K. N. Toosi University of Technology, Tehran, Iran
| |
Collapse
|
18
|
Sarisoy A, Acosta S, Rodríguez-Cabello JC, Czichowski P, Kopp A, Jockenhoevel S, Fernández-Colino A. Bioglues Based on an Elastin-Like Recombinamer: Effect of Tannic Acid as an Additive on Tissue Adhesion and Cytocompatibility. Int J Mol Sci 2023; 24:ijms24076776. [PMID: 37047749 PMCID: PMC10095112 DOI: 10.3390/ijms24076776] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/23/2023] [Revised: 03/24/2023] [Accepted: 04/03/2023] [Indexed: 04/09/2023] Open
Abstract
More than 260 million surgical procedures are performed worldwide each year. Although sutures and staples are widely used to reconnect tissues, they can cause further damage and increase the risk of infection. Bioadhesives have been proposed as an alternative to reconnect tissues. However, clinical adhesives that combine strong adhesion with cytocompatibility have yet to be developed. In this study, we explored the production of adhesives based on protein-engineered polymers bioinspired by the sequence of elastin (i.e., elastin-like recombinamers, ELRs). We hypothesized that the combination of polyphenols (i.e., tannic acid, TA) and ELRs would produce an adhesive coacervate (ELR+TA), as reported for other protein polymers such as silk fibroin (SF). Notably, the adhesion of ELR alone surpassed that of ELR+TA. Indeed, ELR alone achieved adhesive strengths of 88.8 ± 33.2 kPa and 17.0 ± 2.0 kPa on porcine bone and skin tissues, respectively. This surprising result led us to explore a multicomponent bioadhesive to encompass the complementary roles of elastin (mimicked here by ELR) and silk fibroin (SF), and subsequently mirror more closely the multicomponent nature of the extracellular matrix. Tensile testing showed that ELR+SF achieved an adhesive strength of 123.3 ± 60.2 kPa on porcine bone and excellent cytocompatibility. To express this in a more visual and intuitive way, a small surface of only 2.5 cm2 was able to lift at least 2 kg of weight. This opens the door for further studies focusing on the ability of protein-engineered polymers to adhere to biological tissues without further chemical modification for applications in tissue engineering.
Collapse
Affiliation(s)
- Alp Sarisoy
- Department of Biohybrid & Medical Textiles (BioTex), AME–Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, D-52074 Aachen, Germany
| | - Sergio Acosta
- Department of Biohybrid & Medical Textiles (BioTex), AME–Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, D-52074 Aachen, Germany
| | - José Carlos Rodríguez-Cabello
- Bioforge Lab, Group for Advanced Materials and Nanobiotechnology, Biomedical Networking Research Center of Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Edificio LUCIA, Universidad de Valladolid, 47011 Valladolid, Spain
| | | | | | - Stefan Jockenhoevel
- Department of Biohybrid & Medical Textiles (BioTex), AME–Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, D-52074 Aachen, Germany
- AMIBM-Aachen-Maastricht-Institute for Biobased Materials, Faculty of Science and Engineering, Brightlands Chemelot Campus, Maastricht University, 6167 RD Geleen, The Netherlands
| | - Alicia Fernández-Colino
- Department of Biohybrid & Medical Textiles (BioTex), AME–Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, D-52074 Aachen, Germany
| |
Collapse
|
19
|
Chen W, Wang C, Liu W, Zhao B, Zeng Z, Long F, Wang C, Li S, Lin N, Zhou J. A Matrix-Metalloproteinase-Responsive Hydrogel System for Modulating the Immune Microenvironment in Myocardial Infarction. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2209041. [PMID: 36754377 DOI: 10.1002/adma.202209041] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Revised: 02/02/2023] [Indexed: 06/18/2023]
Abstract
Injectable hydrogels carrying therapeutic factors to modulate the infarct immune microenvironment show great potential in the treatment of myocardial infarction (MI). However, conventional injectable hydrogels release therapeutic factors in an uncontrolled manner, which leads to poor treatment efficacy and acute side effects on normal tissues. In this work, a matrix metalloproteinase (MMP)2/9-responsive hydrogel system (MPGC4) is developed, considering the characteristics of the post-MI microenvironment. MPGC4 consists of tetra-poly(ethylene glycol) (PEG) hydrogels and a composite gene nanocarrier (CTL4) that is composed of carbon dots (CDots) coupled with interleukin-4 plasmid DNA via electrostatic interactions. MPGC4 can be automatically triggered to release CTL4 on demand after MI to regulate the infarct immune microenvironment. In addition, due to the photoluminescence properties of CDots, a large amount of viscoelastic MPGC4 is found to be retained in situ after injection into the infarct region without leakage. The in vitro results demonstrate that CTL4 promotes proinflammatory M1 macrophage polarization to the anti-inflammatory M2 subtype and contributes to cardiomyocyte survival through macrophage transition. In a rat model of MI, MPGC4 clears MMPs and precisely targets CTL4 to the infarcted region. In particular, MPGC4 improves cardiac function by modulating macrophage transition to reduce early inflammatory responses and proangiogenic activity.
Collapse
Affiliation(s)
- Wei Chen
- Beijing Institute of Basic Medical Sciences, 27 Taiping Rd, Beijing, 100850, P. R. China
- The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province, Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, 422 Siming Nan Road, Xiamen, 361005, P. R. China
| | - Changyong Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Rd, Beijing, 100850, P. R. China
| | - Wei Liu
- Beijing Institute of Basic Medical Sciences, 27 Taiping Rd, Beijing, 100850, P. R. China
| | - Bicheng Zhao
- The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province, Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, 422 Siming Nan Road, Xiamen, 361005, P. R. China
| | - Zhicheng Zeng
- The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province, Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, 422 Siming Nan Road, Xiamen, 361005, P. R. China
| | - Fen Long
- The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province, Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, 422 Siming Nan Road, Xiamen, 361005, P. R. China
| | - Chunlan Wang
- Beijing Institute of Basic Medical Sciences, 27 Taiping Rd, Beijing, 100850, P. R. China
| | - Siwei Li
- Beijing Institute of Basic Medical Sciences, 27 Taiping Rd, Beijing, 100850, P. R. China
| | - Naibo Lin
- The Higher Educational Key Laboratory for Biomedical Engineering of Fujian Province, Research Center of Biomedical Engineering of Xiamen, Department of Biomaterials, College of Materials, Xiamen University, 422 Siming Nan Road, Xiamen, 361005, P. R. China
| | - Jin Zhou
- Beijing Institute of Basic Medical Sciences, 27 Taiping Rd, Beijing, 100850, P. R. China
| |
Collapse
|
20
|
Scafa Udriște A, Niculescu AG, Iliuță L, Bajeu T, Georgescu A, Grumezescu AM, Bădilă E. Progress in Biomaterials for Cardiac Tissue Engineering and Regeneration. Polymers (Basel) 2023; 15:polym15051177. [PMID: 36904419 PMCID: PMC10007484 DOI: 10.3390/polym15051177] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/16/2023] [Accepted: 02/22/2023] [Indexed: 03/02/2023] Open
Abstract
Cardiovascular diseases are one of the leading global causes of morbidity and mortality, posing considerable health and economic burden on patients and medical systems worldwide. This phenomenon is attributed to two main motives: poor regeneration capacity of adult cardiac tissues and insufficient therapeutic options. Thus, the context calls for upgrading treatments to deliver better outcomes. In this respect, recent research has approached the topic from an interdisciplinary perspective. Combining the advances encountered in chemistry, biology, material science, medicine, and nanotechnology, performant biomaterial-based structures have been created to carry different cells and bioactive molecules for repairing and restoring heart tissues. In this regard, this paper aims to present the advantages of biomaterial-based approaches for cardiac tissue engineering and regeneration, focusing on four main strategies: cardiac patches, injectable hydrogels, extracellular vesicles, and scaffolds and reviewing the most recent developments in these fields.
Collapse
Affiliation(s)
- Alexandru Scafa Udriște
- Department 4 Cardio-Thoracic Pathology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Adelina-Gabriela Niculescu
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050657 Bucharest, Romania
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Politehnica University of Bucharest, 011061 Bucharest, Romania
| | - Luminița Iliuță
- Department 4 Cardio-Thoracic Pathology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Teodor Bajeu
- Department 4 Cardio-Thoracic Pathology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
| | - Adriana Georgescu
- Pathophysiology and Pharmacology Department, Institute of Cellular Biology and Pathology “Nicolae Simionescu” of the Romanian Academy, 050568 Bucharest, Romania
| | - Alexandru Mihai Grumezescu
- Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050657 Bucharest, Romania
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Politehnica University of Bucharest, 011061 Bucharest, Romania
- Academy of Romanian Scientists, Ilfov No. 3, 050044 Bucharest, Romania
- Correspondence:
| | - Elisabeta Bădilă
- Department 4 Cardio-Thoracic Pathology, “Carol Davila” University of Medicine and Pharmacy, 050474 Bucharest, Romania
- Cardiology Department, Colentina Clinical Hospital, 020125 Bucharest, Romania
| |
Collapse
|
21
|
Ciarleglio G, Toto E, Santonicola MG. Conductive and Thermo-Responsive Composite Hydrogels with Poly(N-isopropylacrylamide) and Carbon Nanotubes Fabricated by Two-Step Photopolymerization. Polymers (Basel) 2023; 15:polym15041022. [PMID: 36850305 PMCID: PMC9962410 DOI: 10.3390/polym15041022] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2023] [Revised: 02/04/2023] [Accepted: 02/07/2023] [Indexed: 02/22/2023] Open
Abstract
Biocompatible and conductive polymer hydrogels are the subject of intensive research in the bioengineering field because of their use in bioelectronic devices and for the fabrication of electro-responsive tissues and drug delivery systems. In this study, we report the synthesis of conductive composite hydrogels consisting of a poly(N-isopropylacrylamide) (PNIPAM) matrix embedding carboxyl-functionalized multi-walled carbon nanotubes (MWCNT-COOH) using a two-step photopolymerization method. Thermo-responsive hydrogels with controlled hydrophilicity and conductivity were prepared by varying the carbon nanotube concentration in the range 0.5-3 wt%. The thermal response of the PNIPAM-based composite hydrogels was measured by differential scanning calorimetry with both ultrapure water and PBS solution as swelling liquid. Results show that the endothermic peak associated with the temperature-induced volume phase transition (VPT) shifts to higher temperatures upon increasing the concentration of the nanotubes, indicating that more energy is required to dissociate the hydrogen bonds of the polymer/filler network. In PBS solution, the swelling ratios and the VPT temperatures of the composite hydrogels are reduced because of salt-induced screening of the oppositely charged polymer/filler assembly, and the electrical resistivity decreases by a factor of 10 with respect to the water-swollen hydrogels.
Collapse
|
22
|
Liu W, Xu B, Zhao S, Han S, Quan R, Liu W, Ji C, Chen B, Xiao Z, Yin M, Yin Y, Dai J, Zhao Y. Spinal cord tissue engineering via covalent interaction between biomaterials and cells. SCIENCE ADVANCES 2023; 9:eade8829. [PMID: 36753555 PMCID: PMC9908024 DOI: 10.1126/sciadv.ade8829] [Citation(s) in RCA: 22] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/14/2022] [Accepted: 01/10/2023] [Indexed: 05/29/2023]
Abstract
Noncovalent interactions between cells and environmental cues have been recognized as fundamental physiological interactions that regulate cell behavior. However, the effects of the covalent interactions between cells and biomaterials on cell behavior have not been examined. Here, we demonstrate a combined strategy based on covalent conjugation between biomaterials (collagen fibers/lipid nanoparticles) and various cells (exogenous neural progenitor cells/astrocytes/endogenous tissue-resident cells) to promote neural regeneration after spinal cord injury (SCI). We found that metabolic azido-labeled human neural progenitor cells conjugated on dibenzocyclooctyne-modified collagen fibers significantly promoted cell adhesion, spreading, and differentiation compared with noncovalent adhesion. In addition, dibenzocyclooctyne-modified lipid nanoparticles containing edaravone, a well-known ROS scavenger, could target azide-labeled spinal cord tissues or transplanted azide-modified astrocytes to improve the SCI microenvironment. The combined application of these covalent conjugation strategies in a rat SCI model boosted neural regeneration, suggesting that the covalent interactions between cells and biomaterials have great potential for tissue regeneration.
Collapse
Affiliation(s)
- Weiyuan Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
- University of Chinese Academy of Sciences, Beijing 100190, China
| | - Bai Xu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
| | - Shuaijing Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
- University of Chinese Academy of Sciences, Beijing 100190, China
| | - Shuyu Han
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
- University of Chinese Academy of Sciences, Beijing 100190, China
| | - Rui Quan
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
- University of Chinese Academy of Sciences, Beijing 100190, China
| | - Wenbin Liu
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
| | - Chunnan Ji
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
| | - Bing Chen
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
| | - Zhifeng Xiao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
| | - Man Yin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
- University of Chinese Academy of Sciences, Beijing 100190, China
| | - Yanyun Yin
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
| | - Jianwu Dai
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
- University of Chinese Academy of Sciences, Beijing 100190, China
| | - Yannan Zhao
- State Key Laboratory of Molecular Developmental Biology, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Beijing 100080, China
| |
Collapse
|
23
|
Wang Y, Yang M, Zhang J, Ren J, Liu N, Liu B, Lu L, Yang B. S-Doped carbonized polymer dots inhibit early myocardial fibrosis by regulating mitochondrial function. Biomater Sci 2023; 11:894-907. [PMID: 36524407 DOI: 10.1039/d2bm00578f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Myocardial fibrosis (MF) is a critical pathological lesion in the progression of various acute and chronic cardiovascular diseases. However, there is still a lack of clinically effective drugs and treatments for MF therapies. Herein, for the first time, we developed fluorescent sulfur-doped carbonized polymer dots (S-CPDs) as new nano-antioxidants to reduce the cardiomyocyte damage caused by reactive oxygen species (ROS) in the early stage of fibrotic lesions. In vitro results suggested that the pre-protection of S-CPDs significantly increased the survival rate of H9c2 cells under severe oxidative stress, inhibited the isoproterenol (ISO)-induced hypertrophy of myocardial cells through improving the content of mitochondria related proteins and adenosine triphosphate (ATP) in cells. Moreover, S-CPD administration could effectively decrease cardiac hypertrophy and promote heart function in MF rat models. The rapid internalization, high biocompatibility and fluorescence imaging potential of S-CPDs revealed their promising application prospects in the diagnoses and treatments of cardiovascular diseases.
Collapse
Affiliation(s)
- Yiran Wang
- Department of Cardiology, The Second Hospital of Jilin University, Changchun 130021, P.R. China.
| | - Mingxi Yang
- Department of Hand Surgery, The First Hospital of Jilin University, Changchun 130021, P.R. China. .,State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P.R. China.
| | - Jiayi Zhang
- Department of Hand Surgery, The First Hospital of Jilin University, Changchun 130021, P.R. China.
| | - Jingyan Ren
- Department of Hand Surgery, The First Hospital of Jilin University, Changchun 130021, P.R. China.
| | - Ning Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun 130021, P.R. China.
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun 130021, P.R. China.
| | - Laijin Lu
- Department of Hand Surgery, The First Hospital of Jilin University, Changchun 130021, P.R. China.
| | - Bai Yang
- State Key Laboratory of Supramolecular Structure and Materials, College of Chemistry, Jilin University, Changchun 130012, P.R. China.
| |
Collapse
|
24
|
Li P, Hu J, Wang J, Zhang J, Wang L, Zhang C. The Role of Hydrogel in Cardiac Repair and Regeneration for Myocardial Infarction: Recent Advances and Future Perspectives. Bioengineering (Basel) 2023; 10:bioengineering10020165. [PMID: 36829659 PMCID: PMC9952459 DOI: 10.3390/bioengineering10020165] [Citation(s) in RCA: 13] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 01/19/2023] [Accepted: 01/24/2023] [Indexed: 01/31/2023] Open
Abstract
A myocardial infarction (MI) is the leading cause of morbidity and mortality, seriously threatens human health, and becomes a major health burden of our society. It is urgent to pursue effective therapeutic strategies for the regeneration and restore myocardial function after MI. This review discusses the role of hydrogel in cardiac repair and regeneration for MI. Hydrogel-based cardiac patches and injectable hydrogels are the most commonly used applications in cardiac regeneration medicine. With injectable hydrogels, bioactive compounds and cells can be delivered in situ, promoting in situ repair and regeneration, while hydrogel-based cardiac patches reduce myocardial wall stress, which passively inhibits ventricular expansion. Hydrogel-based cardiac patches work as mechanically supportive biomaterials. In cardiac regeneration medicine, clinical trials and commercial products are limited. Biomaterials, biochemistry, and biological actives, such as intelligent hydrogels and hydrogel-based exosome patches, which may serve as an effective treatment for MI in the future, are still under development. Further investigation of clinical feasibility is warranted. We can anticipate hydrogels having immense translational potential for cardiac regeneration in the near future.
Collapse
Affiliation(s)
- Ping Li
- Department of Obstetrics, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jiajia Hu
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Jian Wang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Junjie Zhang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Lu Wang
- Department of Anesthesiology, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Chengliang Zhang
- Department of Cardiovascular Surgery, Xiangya Hospital, Central South University, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
- Correspondence:
| |
Collapse
|
25
|
Chen X, Zhu L, Wang X, Xiao J. Insight into Heart-Tailored Architectures of Hydrogel to Restore Cardiac Functions after Myocardial Infarction. Mol Pharm 2023; 20:57-81. [PMID: 36413809 DOI: 10.1021/acs.molpharmaceut.2c00650] [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: 11/23/2022]
Abstract
With permanent heart muscle injury or death, myocardial infarction (MI) is complicated by inflammatory, proliferation and remodeling phases from both the early ischemic period and subsequent infarct expansion. Though in situ re-establishment of blood flow to the infarct zone and delays of the ventricular remodeling process are current treatment options of MI, they fail to address massive loss of viable cardiomyocytes while transplanting stem cells to regenerate heart is hindered by their poor retention in the infarct bed. Equipped with heart-specific mimicry and extracellular matrix (ECM)-like functionality on the network structure, hydrogels leveraging tissue-matching biomechanics and biocompatibility can mechanically constrain the infarct and act as localized transport of bioactive ingredients to refresh the dysfunctional heart under the constant cyclic stress. Given diverse characteristics of hydrogel including conductivity, anisotropy, adhesiveness, biodegradability, self-healing and mechanical properties driving local cardiac repair, we aim to investigate and conclude the dynamic balance between ordered architectures of hydrogels and the post-MI pathological milieu. Additionally, our review summarizes advantages of heart-tailored architectures of hydrogels in cardiac repair following MI. Finally, we propose challenges and prospects in clinical translation of hydrogels to draw theoretical guidance on cardiac repair and regeneration after MI.
Collapse
Affiliation(s)
- Xuerui Chen
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Liyun Zhu
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| | - Xu Wang
- Hangzhou Medical College, Binjiang Higher Education Park, Binwen Road 481, Hangzhou 310053, China
| | - Junjie Xiao
- Institute of Geriatrics (Shanghai University), Affiliated Nantong Hospital of Shanghai University (The Sixth People's Hospital of Nantong), School of Medicine, Shanghai University, Nantong 226011, China.,Cardiac Regeneration and Ageing Lab, Institute of Cardiovascular Sciences, Shanghai Engineering Research Center of Organ Repair, School of Life Science, Shanghai University, Shanghai 200444, China
| |
Collapse
|
26
|
Bayaraa O, Dashnyam K, Singh RK, Mandakhbayar N, Lee JH, Park JT, Lee JH, Kim HW. Nanoceria-GO-intercalated multicellular spheroids revascularize and salvage critical ischemic limbs through anti-apoptotic and pro-angiogenic functions. Biomaterials 2023; 292:121914. [PMID: 36436306 DOI: 10.1016/j.biomaterials.2022.121914] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2022] [Revised: 11/07/2022] [Accepted: 11/11/2022] [Indexed: 11/15/2022]
Abstract
Critical limb ischemia (CLI) is a serious form of peripheral arterial disease that involves severe blockage of blood flow in lower extremities, often leading to foot necrosis and limb loss. Lack of blood flow and high pro-inflammation with overproduced reactive oxygen species (ROS) in CLI aggravate the degenerative events. Among other therapies, cell delivery is considered potential for restoring regenerative capacity, and preservation of cell survival under high oxidative stress has been challenging and prerequisite to harness cellular functions. Here, we introduce a multicellular delivery system that is intercalated with nanoceria-decorated graphene oxide (CeGO), which is considered to have high ROS scavenging ability while providing cell-matrix interaction signals. The CeGO nano-microsheets (8-nm-nanoceria/0.9-μm-GO) incorporated in HUVEC/MSC (7/3) could form cell-material hybrid spheroids mediated by cellular contraction. Under in vitro oxidative-stress-challenge with H2O2, the CeGO-intercalation enhanced the survival and anti-apoptotic capacity of cellular spheroids. Pro-angiogenic events of cellular spheroids, including cell sprouting and expression of angiogenic markers (HIF1α, VEGF, FGF2, eNOS) were significantly enhanced by the CeGO-intercalation. Proteomics analysis also confirmed substantial up-regulation of a series of angiogenesis-related secretome molecules. Such pro-angiogenic events with CeGO-intercalation were proven to be mediated by the APE/Ref-1 signaling pathway. When delivered to ischemic hindlimb in mice, the CeGO-cell spheroids could inhibit the accumulation of in vivo ROS rapidly, preserving high cell survival rate (cells were more proliferative and less apoptotic vs. those in cell-only spheroids), and up-regulated angiogenic molecular expressions. Monitoring over 28 days revealed significantly enhanced blood reperfusion and tissue recovery, and an ultimate limb salvage with the CeGO-cell delivery (∼60% salvaged vs. ∼29% in cell-only delivery vs. 0% in ischemia control). Together, the CeGO intercalated in HUVEC/MSC delivery is considered a potential nano-microplatform for CLI treatment, by scavenging excessive ROS and enhancing transplanted cell survival, while stimulating angiogenic events, which collectively help revascularization and tissue recovery, salvaging critical ischemic limbs.
Collapse
Affiliation(s)
- Oyunchimeg Bayaraa
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea; Drug Research Institute, Mongolian University of Pharmaceutical Science, 14250, Mongolia
| | - Khandmaa Dashnyam
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Drug Research Institute, Mongolian University of Pharmaceutical Science, 14250, Mongolia
| | - Rajendra K Singh
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Nandin Mandakhbayar
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jun Hee Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea; Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea; Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jong-Tae Park
- Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea; Department of Oral Anatomy, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea
| | - Jung-Hwan Lee
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea; Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea; Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea; Cell & Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea; UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea
| | - Hae-Won Kim
- Institute of Tissue Regeneration Engineering (ITREN), Dankook University, Cheonan, 31116, Republic of Korea; Department of Nanobiomedical Science and BK21 NBM Global Research Center for Regenerative Medicine, Dankook University, Cheonan, 31116, Republic of Korea; Department of Biomaterials Science, College of Dentistry, Dankook University, Cheonan, 31116, Republic of Korea; Mechanobiology Dental Medicine Research Center, Dankook University, Cheonan, 31116, Republic of Korea; Cell & Matter Institute, Dankook University, Cheonan, 31116, Republic of Korea; UCL Eastman-Korea Dental Medicine Innovation Centre, Dankook University, Cheonan, 31116, Republic of Korea.
| |
Collapse
|
27
|
Spelat R, Ferro F, Contessotto P, Aljaabary A, Martin-Saldaña S, Jin C, Karlsson NG, Grealy M, Hilscher MM, Magni F, Chinello C, Kilcoyne M, Pandit A. Metabolic reprogramming and membrane glycan remodeling as potential drivers of zebrafish heart regeneration. Commun Biol 2022; 5:1365. [PMID: 36509839 PMCID: PMC9744865 DOI: 10.1038/s42003-022-04328-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Accepted: 12/01/2022] [Indexed: 12/15/2022] Open
Abstract
The ability of the zebrafish heart to regenerate following injury makes it a valuable model to deduce why this capability in mammals is limited to early neonatal stages. Although metabolic reprogramming and glycosylation remodeling have emerged as key aspects in many biological processes, how they may trigger a cardiac regenerative response in zebrafish is still a crucial question. Here, by using an up-to-date panel of transcriptomic, proteomic and glycomic approaches, we identify a metabolic switch from mitochondrial oxidative phosphorylation to glycolysis associated with membrane glycosylation remodeling during heart regeneration. Importantly, we establish the N- and O-linked glycan structural repertoire of the regenerating zebrafish heart, and link alterations in both sialylation and high mannose structures across the phases of regeneration. Our results show that metabolic reprogramming and glycan structural remodeling are potential drivers of tissue regeneration after cardiac injury, providing the biological rationale to develop novel therapeutics to elicit heart regeneration in mammals.
Collapse
Affiliation(s)
- Renza Spelat
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland ,grid.5970.b0000 0004 1762 9868Neurobiology Sector, International School for Advanced Studies (SISSA), Trieste, Italy
| | - Federico Ferro
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland ,grid.5133.40000 0001 1941 4308Department of Medical Surgery and Health Science, University of Trieste, Trieste, Italy
| | - Paolo Contessotto
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland ,grid.5608.b0000 0004 1757 3470Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Amal Aljaabary
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Sergio Martin-Saldaña
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| | - Chunsheng Jin
- grid.8761.80000 0000 9919 9582Department of Medical Biochemistry, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Niclas G. Karlsson
- grid.8761.80000 0000 9919 9582Department of Medical Biochemistry, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden
| | - Maura Grealy
- Pharmacology and Therapeutics, School of Medicine, University of Galway, Galway, Ireland
| | - Markus M. Hilscher
- grid.10548.380000 0004 1936 9377Science for Life Laboratory, Department of Biochemistry and Biophysics, Stockholm University, Stockholm, Sweden
| | - Fulvio Magni
- grid.7563.70000 0001 2174 1754Clinical Proteomics and Metabolomics Unit, School of Medicine and Surgery, University of Milano-Bicocca, Vedano al Lambro, Italy
| | - Clizia Chinello
- grid.7563.70000 0001 2174 1754Clinical Proteomics and Metabolomics Unit, School of Medicine and Surgery, University of Milano-Bicocca, Vedano al Lambro, Italy
| | - Michelle Kilcoyne
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland ,Carbohydrate Signalling Group, Microbiology, School of Natural Sciences, University of Galway, Galway, Ireland
| | - Abhay Pandit
- CÚRAM, SFI Research Centre for Medical Devices, University of Galway, Galway, Ireland
| |
Collapse
|
28
|
Söderlund Z, Ibáñez-Fonseca A, Hajizadeh S, Rodríguez-Cabello JC, Liu J, Ye L, Tykesson E, Elowsson L, Westergren-Thorsson G. Controlled release of growth factors using synthetic glycosaminoglycans in a modular macroporous scaffold for tissue regeneration. Commun Biol 2022; 5:1349. [PMID: 36482075 PMCID: PMC9732287 DOI: 10.1038/s42003-022-04305-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022] Open
Abstract
Healthy regeneration of tissue relies on a well-orchestrated release of growth factors. Herein, we show the use of synthetic glycosaminoglycans for controlled binding and release of growth factors to induce a desired cellular response. First, we screened glycosaminoglycans with growth factors of interest to determine kon (association rate constant), koff (dissociation rate constant), and Kd (equilibrium rate constant). As proof-of-concept, we functionalized an elastin-like recombinamer (ELR) hydrogel with a synthetic glycosaminoglycan and immobilized fibroblast growth factor 2 (FGF2), demonstrating that human umbilical vein endothelial cells cultured on top of ELR hydrogel differentiated into tube-like structures. Taking this concept further, we developed a tunable macroporous ELR cryogel material, containing a synthetic glycosaminoglycan and FGF2 that showed increased blood vessel formation and reduced immune response compared to control when implanted in a subcutaneous mouse model. These results demonstrated the possibility for specific release of desired growth factors in/from a modular 3D scaffold in vitro and in vivo.
Collapse
Affiliation(s)
- Z Söderlund
- Lung Biology, Department of Experimental Medical Science, Lund University, Lund, Sweden.
| | - A Ibáñez-Fonseca
- Lung Biology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - S Hajizadeh
- Division Pure and Applied Biochemistry, Lund University, Lund, Sweden
| | | | - J Liu
- Division of Chemical Biology and Medicinal Chemistry, Eshelman School of Phamarcy, University of North Carolina, Chapel Hill, NC, USA
| | - L Ye
- Division Pure and Applied Biochemistry, Lund University, Lund, Sweden
| | - E Tykesson
- Lung Biology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - L Elowsson
- Lung Biology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| | - G Westergren-Thorsson
- Lung Biology, Department of Experimental Medical Science, Lund University, Lund, Sweden
| |
Collapse
|
29
|
González‐Pérez F, Alonso M, González de Torre I, Santos M, Rodríguez‐Cabello JC. Protease-Sensitive, VEGF-Mimetic Peptide, and IKVAV Laminin-Derived Peptide Sequences within Elastin-Like Recombinamer Scaffolds Provide Spatiotemporally Synchronized Guidance of Angiogenesis and Neurogenesis. Adv Healthc Mater 2022; 11:e2201646. [PMID: 36099430 PMCID: PMC11468767 DOI: 10.1002/adhm.202201646] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 08/29/2022] [Indexed: 01/28/2023]
Abstract
Spatiotemporal control of vascularization and innervation is a desired hallmark in advanced tissue regeneration. For this purpose, we design a 3D model scaffold, based on elastin-like recombinamer (ELR) hydrogels. This contains two interior and well-defined areas, small cylinders, with differentiated bioactivities with respect to the bulk. Both are constructed on a protease sensitive ELR with a fast-proteolyzed domain, but one bears a VEGF-mimetic peptide (QK) and the other a laminin-derived pentapeptide (IKVAV), to promote angiogenesis and neurogenesis, respectively. The outer bulk is based on a slow proteolytic sequence and RGD cell adhesion domains. In vitro studies show the effect of QK and IKVAV peptides on the promotion of endothelial cell and axon spreading, respectively. The subcutaneous implantation of the final 3D scaffold demonstrates the ability to spatiotemporally control angiogenesis and neurogenesis in vivo. Specifically, the inner small cylinder containing the QK peptide promotes fast endothelialization, whereas the one with IKVAV peptide promotes fast neurogenesis. Both, vascularization and innervation take place in advance of the bulk scaffold infiltration. This scaffold shows that it is possible to induce vascularization and innervation in predetermined areas of the scaffold well ahead to the bulk infiltration. That significantly increases the efficiency of the regenerative activity.
Collapse
Affiliation(s)
- Fernando González‐Pérez
- G.I.R. BIOFORGE (Group for Advanced Materials and Nanobiotechnology)CIBER‐BBNEdificio LUCIAUniversidad de ValladolidPaseo Belén 19Valladolid47011Spain
| | - Matilde Alonso
- G.I.R. BIOFORGE (Group for Advanced Materials and Nanobiotechnology)CIBER‐BBNEdificio LUCIAUniversidad de ValladolidPaseo Belén 19Valladolid47011Spain
| | - Israel González de Torre
- G.I.R. BIOFORGE (Group for Advanced Materials and Nanobiotechnology)CIBER‐BBNEdificio LUCIAUniversidad de ValladolidPaseo Belén 19Valladolid47011Spain
| | - Mercedes Santos
- G.I.R. BIOFORGE (Group for Advanced Materials and Nanobiotechnology)CIBER‐BBNEdificio LUCIAUniversidad de ValladolidPaseo Belén 19Valladolid47011Spain
| | - José Carlos Rodríguez‐Cabello
- G.I.R. BIOFORGE (Group for Advanced Materials and Nanobiotechnology)CIBER‐BBNEdificio LUCIAUniversidad de ValladolidPaseo Belén 19Valladolid47011Spain
| |
Collapse
|
30
|
Hu S, Zhu D, Li Z, Cheng K. Detachable Microneedle Patches Deliver Mesenchymal Stromal Cell Factor-Loaded Nanoparticles for Cardiac Repair. ACS NANO 2022; 16:15935-15945. [PMID: 36148975 DOI: 10.1021/acsnano.2c03060] [Citation(s) in RCA: 25] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/16/2023]
Abstract
Intramyocardial injection is a direct and efficient approach to deliver therapeutics to the heart. However, the injected volume must be very limited, and there is injury to the injection site and leakage issues during heart beating. Herein, we developed a detachable therapeutic microneedle (MN) patch, which is comprised of mesenchymal stromal cell-secreted factors (MSCF)-loaded poly(lactic-co-glycolic acid) nanoparticles (NP) in MN tips made of elastin-like polypeptide gel, with a resolvable non-cross-linked hyaluronic acid (HA) gel as the MN base. The tips can be firmly inserted into the infarcted myocardium after base removal, and no suture is needed. In isolated neonatal rat cardiac cells, we found that the cellular uptake of MSCF-NP in the cardiomyocytes was higher than in cardiac fibroblasts. MSCF-NP promoted the proliferation of injured cardiomyocytes. In a rat model of myocardial infarction, MN-MSCF-NP treatment reduced cardiomyocyte apoptosis, restored myocardium volume, and reduced fibrosis during the cardiac remodeling process. Our work demonstrated the therapeutic potential of MN to deliver MSCF directly into the myocardium and provides a promising treatment approach for cardiac diseases.
Collapse
Affiliation(s)
- Shiqi Hu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States, and North Carolina State University, Raleigh, North Carolina 27606, United States
| | - Dashuai Zhu
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States, and North Carolina State University, Raleigh, North Carolina 27606, United States
| | - Zhenhua Li
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States, and North Carolina State University, Raleigh, North Carolina 27606, United States
| | - Ke Cheng
- Department of Molecular Biomedical Sciences and Comparative Medicine Institute, North Carolina State University, Raleigh, North Carolina 27607, United States
- Joint Department of Biomedical Engineering, University of North Carolina at Chapel Hill, Chapel Hill, North Carolina 27599, United States, and North Carolina State University, Raleigh, North Carolina 27606, United States
| |
Collapse
|
31
|
González-Pérez F, Acosta S, Rütten S, Emonts C, Kopp A, Henke HW, Bruners P, Gries T, Rodríguez-Cabello JC, Jockenhoevel S, Fernández-Colino A. Biohybrid elastin-like venous valve with potential for in situ tissue engineering. Front Bioeng Biotechnol 2022; 10:988533. [PMID: 36213079 PMCID: PMC9532864 DOI: 10.3389/fbioe.2022.988533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/22/2022] [Indexed: 11/15/2022] Open
Abstract
Chronic venous insufficiency (CVI) is a leading vascular disease whose clinical manifestations include varicose veins, edemas, venous ulcers, and venous hypertension, among others. Therapies targeting this medical issue are scarce, and so far, no single venous valve prosthesis is clinically available. Herein, we have designed a bi-leaflet transcatheter venous valve that consists of (i) elastin-like recombinamers, (ii) a textile mesh reinforcement, and (iii) a bioabsorbable magnesium stent structure. Mechanical characterization of the resulting biohybrid elastin-like venous valves (EVV) showed an anisotropic behavior equivalent to the native bovine saphenous vein valves and mechanical strength suitable for vascular implantation. The EVV also featured minimal hemolysis and platelet adhesion, besides actively supporting endothelialization in vitro, thus setting the basis for its application as an in situ tissue engineering implant. In addition, the hydrodynamic testing in a pulsatile bioreactor demonstrated excellent hemodynamic valve performance, with minimal regurgitation (<10%) and pressure drop (<5 mmHg). No stagnation points were detected and an in vitro simulated transcatheter delivery showed the ability of the venous valve to withstand the implantation procedure. These results present a promising concept of a biohybrid transcatheter venous valve as an off-the-shelf implant, with great potential to provide clinical solutions for CVI treatment.
Collapse
Affiliation(s)
- Fernando González-Pérez
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER-BBN, Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| | - Sergio Acosta
- Department of Biohybrid and Medical Textiles (BioTex), AME–Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
| | - Stephan Rütten
- Electron Microscopy Facility, Uniklinik RWTH Aachen, Aachen, Germany
| | - Caroline Emonts
- Institut für Textiltechnik Aachen (ITA), RWTH Aachen University, Aachen, Germany
| | | | | | - Philipp Bruners
- Klinik für Diagnostische and Interventionelle Radiologie, Universitätsklinikum Aachen, Aachen, Germany
| | - Thomas Gries
- Institut für Textiltechnik Aachen (ITA), RWTH Aachen University, Aachen, Germany
| | - J. Carlos Rodríguez-Cabello
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER-BBN, Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| | - Stefan Jockenhoevel
- Department of Biohybrid and Medical Textiles (BioTex), AME–Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
- AMIBM-Aachen-Maastricht-Institute for Biobased Materials, Maastricht University, Maastricht, Netherlands
- *Correspondence: Stefan Jockenhoevel, ; Alicia Fernández-Colino,
| | - Alicia Fernández-Colino
- Department of Biohybrid and Medical Textiles (BioTex), AME–Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
- *Correspondence: Stefan Jockenhoevel, ; Alicia Fernández-Colino,
| |
Collapse
|
32
|
Basara G, Bahcecioglu G, Ozcebe SG, Ellis BW, Ronan G, Zorlutuna P. Myocardial infarction from a tissue engineering and regenerative medicine point of view: A comprehensive review on models and treatments. BIOPHYSICS REVIEWS 2022; 3:031305. [PMID: 36091931 PMCID: PMC9447372 DOI: 10.1063/5.0093399] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 08/08/2022] [Indexed: 05/12/2023]
Abstract
In the modern world, myocardial infarction is one of the most common cardiovascular diseases, which are responsible for around 18 million deaths every year or almost 32% of all deaths. Due to the detrimental effects of COVID-19 on the cardiovascular system, this rate is expected to increase in the coming years. Although there has been some progress in myocardial infarction treatment, translating pre-clinical findings to the clinic remains a major challenge. One reason for this is the lack of reliable and human representative healthy and fibrotic cardiac tissue models that can be used to understand the fundamentals of ischemic/reperfusion injury caused by myocardial infarction and to test new drugs and therapeutic strategies. In this review, we first present an overview of the anatomy of the heart and the pathophysiology of myocardial infarction, and then discuss the recent developments on pre-clinical infarct models, focusing mainly on the engineered three-dimensional cardiac ischemic/reperfusion injury and fibrosis models developed using different engineering methods such as organoids, microfluidic devices, and bioprinted constructs. We also present the benefits and limitations of emerging and promising regenerative therapy treatments for myocardial infarction such as cell therapies, extracellular vesicles, and cardiac patches. This review aims to overview recent advances in three-dimensional engineered infarct models and current regenerative therapeutic options, which can be used as a guide for developing new models and treatment strategies.
Collapse
Affiliation(s)
- Gozde Basara
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Gokhan Bahcecioglu
- Department of Aerospace and Mechanical Engineering, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - S. Gulberk Ozcebe
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Bradley W Ellis
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - George Ronan
- Bioengineering Graduate Program, University of Notre Dame, Notre Dame, Indiana 46556, USA
| | - Pinar Zorlutuna
- Present address: 143 Multidisciplinary Research Building, University of Notre Dame, Notre Dame, IN 46556. Author to whom correspondence should be addressed:. Tel.: +1 574 631 8543. Fax: +1 574 631 8341
| |
Collapse
|
33
|
Tapeinos C, Gao H, Bauleth-Ramos T, Santos HA. Progress in Stimuli-Responsive Biomaterials for Treating Cardiovascular and Cerebrovascular Diseases. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2022; 18:e2200291. [PMID: 35306751 DOI: 10.1002/smll.202200291] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/15/2022] [Indexed: 06/14/2023]
Abstract
Cardiovascular and cerebrovascular diseases (CCVDs) describe abnormal vascular system conditions affecting the brain and heart. Among these, ischemic heart disease and ischemic stroke are the leading causes of death worldwide, resulting in 16% and 11% of deaths globally. Although several therapeutic approaches are presented over the years, the continuously increasing mortality rates suggest the need for more advanced strategies for their treatment. One of these strategies lies in the use of stimuli-responsive biomaterials. These "smart" biomaterials can specifically target the diseased tissue, and after "reading" the altered environmental cues, they can respond by altering their physicochemical properties and/or their morphology. In this review, the progress in the field of stimuli-responsive biomaterials for CCVDs in the last five years, aiming at highlighting their potential as early-stage therapeutics in the preclinical scenery, is described.
Collapse
Affiliation(s)
- Christos Tapeinos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
| | - Han Gao
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
- Department of Biomedical Engineeringand and W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Tomás Bauleth-Ramos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
- Department of Biomedical Engineeringand and W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| | - Hélder A Santos
- Drug Research Program, Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, Helsinki, FI-00014, Finland
- Department of Biomedical Engineeringand and W.J. Kolff Institute for Biomedical Engineering and Materials Science, University Medical Center Groningen, University of Groningen, Ant. Deusinglaan 1, Groningen, 9713 AV, The Netherlands
| |
Collapse
|
34
|
Ferro F, Spelat R, Valente C, Contessotto P. Understanding How Heart Metabolic Derangement Shows Differential Stage Specificity for Heart Failure with Preserved and Reduced Ejection Fraction. Biomolecules 2022; 12:biom12070969. [PMID: 35883525 PMCID: PMC9312956 DOI: 10.3390/biom12070969] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2022] [Revised: 06/30/2022] [Accepted: 07/06/2022] [Indexed: 12/12/2022] Open
Abstract
Heart failure (HF) is a clinical condition defined by structural and functional abnormalities in the heart that gradually result in reduced cardiac output (HFrEF) and/or increased cardiac pressures at rest and under stress (HFpEF). The presence of asymptomatic individuals hampers HF identification, resulting in delays in recognizing patients until heart dysfunction is manifested, thus increasing the chance of poor prognosis. Given the recent advances in metabolomics, in this review we dissect the main alterations occurring in the metabolic pathways behind the decrease in cardiac function caused by HF. Indeed, relevant preclinical and clinical research has been conducted on the metabolite connections and differences between HFpEF and HFrEF. Despite these promising results, it is crucial to note that, in addition to identifying single markers and reliable threshold levels within the healthy population, the introduction of composite panels would strongly help in the identification of those individuals with an increased HF risk. That said, additional research in the field is required to overcome the current drawbacks and shed light on the pathophysiological changes that lead to HF. Finally, greater collaborative data sharing, as well as standardization of procedures and approaches, would enhance this research field to fulfil its potential.
Collapse
Affiliation(s)
- Federico Ferro
- Department of Medical, Surgery and Health Sciences, University of Trieste, 34125 Trieste, Italy
- Correspondence:
| | - Renza Spelat
- Neurobiology Sector, International School for Advanced Studies (SISSA), 34136 Trieste, Italy;
| | - Camilla Valente
- Department of Molecular Medicine, University of Padova, 35122 Padova, Italy; (C.V.); (P.C.)
| | - Paolo Contessotto
- Department of Molecular Medicine, University of Padova, 35122 Padova, Italy; (C.V.); (P.C.)
| |
Collapse
|
35
|
Hu W, Yang C, Guo X, Wu Y, Loh XJ, Li Z, Wu YL, Wu C. Research Advances of Injectable Functional Hydrogel Materials in the Treatment of Myocardial Infarction. Gels 2022; 8:423. [PMID: 35877508 PMCID: PMC9316750 DOI: 10.3390/gels8070423] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 06/30/2022] [Accepted: 07/03/2022] [Indexed: 12/10/2022] Open
Abstract
Myocardial infarction (MI) has become one of the serious diseases threatening human life and health. However, traditional treatment methods for MI have some limitations, such as irreversible myocardial necrosis and cardiac dysfunction. Fortunately, recent endeavors have shown that hydrogel materials can effectively prevent negative remodeling of the heart and improve the heart function and long-term prognosis of patients with MI due to their good biocompatibility, mechanical properties, and electrical conductivity. Therefore, this review aims to summarize the research progress of injectable hydrogel in the treatment of MI in recent years and to introduce the rational design of injectable hydrogels in myocardial repair. Finally, the potential challenges and perspectives of injectable hydrogel in this field will be discussed, in order to provide theoretical guidance for the development of new and effective treatment strategies for MI.
Collapse
Affiliation(s)
- Wei Hu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China; (W.H.); (X.G.); (Y.W.)
| | - Cui Yang
- School of Medicine, Xiamen University, Xiamen 361003, China;
| | - Xiaodan Guo
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China; (W.H.); (X.G.); (Y.W.)
| | - Yihong Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China; (W.H.); (X.G.); (Y.W.)
| | - Xian Jun Loh
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), Singapore 138634, Singapore;
| | - Zibiao Li
- Institute of Materials Research and Engineering (IMRE), Agency for Science, Technology and Research (A*STAR), Singapore 138634, Singapore;
- Institute of Sustainability for Chemicals, Energy and Environment (ISCE) Agency for Science, Technology and Research (A*STAR), Singapore 138634, Singapore
- Department of Materials Science and Engineering, National University of Singapore, 9 Engineering Drive 1, Singapore 117576, Singapore
| | - Yun-Long Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China; (W.H.); (X.G.); (Y.W.)
| | - Caisheng Wu
- Fujian Provincial Key Laboratory of Innovative Drug Target Research and State Key Laboratory of Cellular Stress Biology, School of Pharmaceutical Sciences, Xiamen University, Xiamen 361102, China; (W.H.); (X.G.); (Y.W.)
| |
Collapse
|
36
|
Zhu S, Yu C, Liu N, Zhao M, Chen Z, Liu J, Li G, Huang H, Guo H, Sun T, Chen J, Zhuang J, Zhu P. Injectable conductive gelatin methacrylate / oxidized dextran hydrogel encapsulating umbilical cord mesenchymal stem cells for myocardial infarction treatment. Bioact Mater 2022; 13:119-134. [PMID: 35224296 PMCID: PMC8844712 DOI: 10.1016/j.bioactmat.2021.11.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 11/04/2021] [Accepted: 11/07/2021] [Indexed: 02/03/2023] Open
Abstract
Umbilical cord mesenchymal stem cells (UCMSCs) transplantation has been proposed as a promising treatment modality for myocardial infarction (MI), but the low retention rate remains a considerable challenge. Injectable natural polymer hydrogels with conductivity ability are highly desirable as cell delivery vehicles to repair infarct myocardium and restore the cardiac function. In this work, we developed a hydrogel system based on gelatin methacrylate (GelMA) and oxidized dextran (ODEX) as cell delivery vehicles for MI. And dopamine could be used as a reductant of graphene oxide (GO) to form reductive GO (rGO). By adjusting the amount of rGO, the conductivity of hydrogels with 0.5 mg/mL rGO concentration (≈10−4 S/cm) was similar to that of natural heart tissue. In vitro cell experiments showed that the prepared hydrogels had excellent biocompatibility and cell delivery ability of UCMSCs. More importantly, GelMA-O5/rGO hydrogel could promote UCMSCs growth and proliferation, improve the myocardial differentiation ability of UCMSCs, and up-regulate the expression of cTnI and Cx43. Further in vivo experiments demonstrated that GelMA-O5/rGO/UCMSCs Hydrogel could significantly improve the ejection fraction (EF) of rats and significantly reduce myocardial infarct area compared to PBS group, promote the survival of UCMSCs, enhance the expression level of cTnI and Cx43, and decrease the expression level of caspase-3. The findings of this study suggested that the injectable conductive GelMA-O5/rGO hydrogel encapsulating UCMSCs could improve damaged myocardial tissue and reconstruct myocardial function, which will be a promising therapeutic strategy for cardiac repair. Conducting interpenetrating polymer network (IPN) hydrogels were synthesized for myocardial infarction treatment. The conductivity of hydrogel with 0.5 mg/mL rGO concentration (≈10−4 S/cm) was similar to that of natural heart tissue. The hydrogel could promote the growth and proliferation of UCMSCs, and improve the myocardial differentiation ability of UCMSCs. The hydrogel could reduce infarct size and cardiac fibrosis in the infarct zone, increase ventricular ejection fraction. The hydrogel could promote the survival of UCMSCs, up-regulate the expression level of cTnI and Cx43, down-regulate the expression level of caspase-3.
Collapse
|
37
|
Ibáñez-Fonseca A, Rico A, Preciado S, González-Pérez F, Muntión S, García-Briñón J, García-Macías MC, Rodríguez-Cabello JC, Pericacho M, Alonso M, Sánchez-Guijo F. Mesenchymal Stromal Cells Combined With Elastin-Like Recombinamers Increase Angiogenesis In Vivo After Hindlimb Ischemia. Front Bioeng Biotechnol 2022; 10:918602. [PMID: 35814011 PMCID: PMC9260019 DOI: 10.3389/fbioe.2022.918602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2022] [Accepted: 05/16/2022] [Indexed: 12/03/2022] Open
Abstract
Hindlimb ischemia is an unmet medical need, especially for those patients unable to undergo vascular surgery. Cellular therapy, mainly through mesenchymal stromal cell (MSC) administration, may be a potentially attractive approach in this setting. In the current work, we aimed to assess the potential of the combination of MSCs with a proangiogenic elastin-like recombinamer (ELR)–based hydrogel in a hindlimb ischemia murine model. Human bone marrow MSCs were isolated from four healthy donors, while ELR biomaterials were genetically engineered. Hindlimb ischemia was induced through ligation of the right femoral artery, and mice were intramuscularly injected with ELR biomaterial, 0.5 × 106 MSCs or the combination, and also compared to untreated animals. Tissue perfusion was monitored using laser Doppler perfusion imaging. Histological analysis of hindlimbs was performed after hematoxylin and eosin staining. Immunofluorescence with anti–human mitochondria antibody was used for human MSC detection, and the biomaterial was detected by elastin staining. To analyze the capillary density, immunostaining with an anti–CD31 antibody was performed. Our results show that the injection of MSCs significantly improves tissue reperfusion from day 7 (p = 0.0044) to day 21 (p = 0.0216), similar to the infusion of MSC + ELR (p = 0.0038, p = 0.0014), without significant differences between both groups. After histological evaluation, ELR hydrogels induced minimal inflammation in the injection sites, showing biocompatibility. MSCs persisted with the biomaterial after 21 days, both in vitro and in vivo. Finally, we observed a higher blood vessel density when mice were treated with MSCs compared to control (p<0.0001), but this effect was maximized and significantly different to the remaining experimental conditions when mice were treated with the combination of MSCs and the ELR biomaterial (p < 0.0001). In summary, the combination of an ELR-based hydrogel with MSCs may improve the angiogenic effects of both strategies on revascularization of ischemic tissues.
Collapse
Affiliation(s)
| | - Ana Rico
- Cell Therapy Unit, Hematology Department, University Hospital of Salamanca, Salamanca, Spain
| | - Silvia Preciado
- Cell Therapy Unit, Hematology Department, University Hospital of Salamanca, Salamanca, Spain
- RICORS TERAV, ISCIII, Madrid, Spain
- Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Department of Medicine and Cancer Research Center, University of Salamanca, Salamanca, Spain
- *Correspondence: Silvia Preciado,
| | | | - Sandra Muntión
- Cell Therapy Unit, Hematology Department, University Hospital of Salamanca, Salamanca, Spain
- RICORS TERAV, ISCIII, Madrid, Spain
- Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Department of Medicine and Cancer Research Center, University of Salamanca, Salamanca, Spain
| | - Jesús García-Briñón
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Departamento de Biología Celular y Patología, Facultad de Medicina, Salamanca, Spain
| | | | - José Carlos Rodríguez-Cabello
- BIOFORGE Lab, University of Valladolid, CIBER-BBN, Valladolid, Spain
- Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Salamanca, Spain
| | - Miguel Pericacho
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Renal and Cardiovascular Research Unit, Department of Physiology and Pharmacology, University of Salamanca, Salamanca, Spain
| | - Matilde Alonso
- BIOFORGE Lab, University of Valladolid, CIBER-BBN, Valladolid, Spain
- Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Salamanca, Spain
| | - Fermín Sánchez-Guijo
- Cell Therapy Unit, Hematology Department, University Hospital of Salamanca, Salamanca, Spain
- RICORS TERAV, ISCIII, Madrid, Spain
- Centro en Red de Medicina Regenerativa y Terapia Celular de Castilla y León, Salamanca, Spain
- Instituto de Investigación Biomédica de Salamanca (IBSAL), Salamanca, Spain
- Department of Medicine and Cancer Research Center, University of Salamanca, Salamanca, Spain
| |
Collapse
|
38
|
ABSTRACTS (BY NUMBER). Tissue Eng Part A 2022. [DOI: 10.1089/ten.tea.2022.29025.abstracts] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
|
39
|
Song Y, Sharipol A, Uchida H, Ingalls MH, Piraino L, Mereness JA, Moyston T, DeLouise LA, Ovitt CE, Benoit DS. Encapsulation of Primary Salivary Gland Acinar Cell Clusters and Intercalated Ducts (AIDUCs) within Matrix Metalloproteinase (MMP)-Degradable Hydrogels to Maintain Tissue Structure and Function. Adv Healthc Mater 2022; 11:e2101948. [PMID: 34994104 PMCID: PMC8986612 DOI: 10.1002/adhm.202101948] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 12/08/2021] [Indexed: 12/13/2022]
Abstract
Progress in the development of salivary gland regenerative strategies is limited by poor maintenance of the secretory function of salivary gland cells (SGCs) in vitro. To reduce the precipitous loss of secretory function, a modified approach to isolate intact acinar cell clusters and intercalated ducts (AIDUCs), rather than commonly used single cell suspension, is investigated. This isolation approach yields AIDUCs that maintain many of the cell-cell and cell-matrix interactions of intact glands. Encapsulation of AIDUCs in matrix metalloproteinase (MMP)-degradable PEG hydrogels promotes self-assembly into salivary gland mimetics (SGm) with acinar-like structure. Expression of Mist1, a transcription factor associated with secretory function, is detectable throughout the in vitro culture period up to 14 days. Immunohistochemistry also confirms expression of acinar cell markers (NKCC1, PIP and AQP5), duct cell markers (K7 and K5), and myoepithelial cell markers (SMA). Robust carbachol and ATP-stimulated calcium flux is observed within the SGm for up to 14 days after encapsulation, indicating that secretory function is maintained. Though some acinar-to-ductal metaplasia is observed within SGm, it is reduced compared to previous reports. In conclusion, cell-cell interactions maintained within AIDUCs together with the hydrogel microenvironment may be a promising platform for salivary gland regenerative strategies.
Collapse
Affiliation(s)
- Yuanhui Song
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Oral Biology, University of Rochester, Rochester, NY, USA
| | - Azmeer Sharipol
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Oral Biology, University of Rochester, Rochester, NY, USA
| | - Hitoshi Uchida
- Center for Oral Biology, University of Rochester, Rochester, NY, USA
- Department of Biomedical Genetics, University of Rochester, Rochester, NY, USA
| | - Matthew H. Ingalls
- Center for Oral Biology, University of Rochester, Rochester, NY, USA
- Department of Biomedical Genetics, University of Rochester, Rochester, NY, USA
| | - Lindsay Piraino
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Department of Dermatology, University of Rochester, Rochester, NY, USA
| | - Jared A. Mereness
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Oral Biology, University of Rochester, Rochester, NY, USA
- Department of Environmental Medicine, University of Rochester, Rochester, NY, USA
| | - Tracey Moyston
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
| | - Lisa A. DeLouise
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Department of Dermatology, University of Rochester, Rochester, NY, USA
- Materials Science Program, University of Rochester, Rochester, NY, USA
| | - Catherine E. Ovitt
- Center for Oral Biology, University of Rochester, Rochester, NY, USA
- Department of Biomedical Genetics, University of Rochester, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester, Rochester, NY, USA
| | - Danielle S.W. Benoit
- Department of Biomedical Engineering, University of Rochester, Rochester, NY, USA
- Center for Oral Biology, University of Rochester, Rochester, NY, USA
- Department of Biomedical Genetics, University of Rochester, Rochester, NY, USA
- Department of Environmental Medicine, University of Rochester, Rochester, NY, USA
- Wilmot Cancer Institute, University of Rochester, Rochester, NY, USA
- Materials Science Program, University of Rochester, Rochester, NY, USA
- Department of Chemical Engineering, University of Rochester, Rochester, NY, USA
- Center for Musculoskeletal Research, University of Rochester, Rochester, NY, USA
| |
Collapse
|
40
|
Yoon HY, Lee D, Lim DK, Koo H, Kim K. Copper-Free Click Chemistry: Applications in Drug Delivery, Cell Tracking, and Tissue Engineering. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2022; 34:e2107192. [PMID: 34752658 DOI: 10.1002/adma.202107192] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/10/2021] [Revised: 10/27/2021] [Indexed: 06/13/2023]
Abstract
Traditionally, organic chemical reactions require organic solvents, toxic catalysts, heat, or high pressure. However, copper-free click chemistry has been shown to have favorable reaction rates and orthogonality in water, buffer solutions, and physiological conditions without toxic catalysts. Strain-promoted azide-alkyne cycloaddition and inverse electron-demand Diels-Alder reactions are representative of copper-free click chemistry. Artificial chemical reactions via click chemistry can also be used outside of the laboratory in a controllable manner on live cell surfaces, in the cytosol, and in living bodies. Consequently, copper-free click chemistry has many features that are of interest in biomedical research, and various new materials and strategies for its use have been proposed. Herein, recent remarkable trials that have used copper-free click chemistry are described, focusing on their applications in molecular imaging and therapy. The research is categorized as nanoparticles for drug delivery, imaging agents for cell tracking, and hydrogels for tissue engineering, which are rapidly advancing fields based on click chemistry. The content is based primarily on the experience with click chemistry-based biomaterials over the last 10 years.
Collapse
Affiliation(s)
- Hong Yeol Yoon
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, South Korea
| | - Donghyun Lee
- Department of Medical Life Sciences, Department of Biomedicine & Health Sciences, and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Dong-Kwon Lim
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145, Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| | - Heebeom Koo
- Department of Medical Life Sciences, Department of Biomedicine & Health Sciences, and Catholic Photomedicine Research Institute, College of Medicine, The Catholic University of Korea, 222 Banpo-daero, Seocho-gu, Seoul, 06591, Republic of Korea
| | - Kwangmeyung Kim
- Center for Theragnosis, Biomedical Research Institute, Korea Institute of Science and Technology (KIST), Hwarang-ro 14-gil 5, Seongbuk-gu, Seoul, 02792, South Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, 145, Anam-ro, Seongbuk-gu, Seoul, 02841, Republic of Korea
| |
Collapse
|
41
|
Recombinant Proteins-Based Strategies in Bone Tissue Engineering. Biomolecules 2021; 12:biom12010003. [PMID: 35053152 PMCID: PMC8773742 DOI: 10.3390/biom12010003] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/08/2021] [Accepted: 12/11/2021] [Indexed: 11/29/2022] Open
Abstract
The increase in fracture rates and/or problems associated with missing bones due to accidents or various pathologies generates socio-health problems with a very high impact. Tissue engineering aims to offer some kind of strategy to promote the repair of damaged tissue or its restoration as close as possible to the original tissue. Among the alternatives proposed by this specialty, the development of scaffolds obtained from recombinant proteins is of special importance. Furthermore, science and technology have advanced to obtain recombinant chimera’s proteins. This review aims to offer a synthetic description of the latest and most outstanding advances made with these types of scaffolds, particularly emphasizing the main recombinant proteins that can be used to construct scaffolds in their own right, i.e., not only to impregnate them, but also to make scaffolds from their complex structure, with the purpose of being considered in bone regenerative medicine in the near future.
Collapse
|
42
|
Cao H, Duan L, Zhang Y, Cao J, Zhang K. Current hydrogel advances in physicochemical and biological response-driven biomedical application diversity. Signal Transduct Target Ther 2021; 6:426. [PMID: 34916490 PMCID: PMC8674418 DOI: 10.1038/s41392-021-00830-x] [Citation(s) in RCA: 294] [Impact Index Per Article: 98.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2021] [Revised: 11/10/2021] [Accepted: 11/11/2021] [Indexed: 02/05/2023] Open
Abstract
Hydrogel is a type of versatile platform with various biomedical applications after rational structure and functional design that leverages on material engineering to modulate its physicochemical properties (e.g., stiffness, pore size, viscoelasticity, microarchitecture, degradability, ligand presentation, stimulus-responsive properties, etc.) and influence cell signaling cascades and fate. In the past few decades, a plethora of pioneering studies have been implemented to explore the cell-hydrogel matrix interactions and figure out the underlying mechanisms, paving the way to the lab-to-clinic translation of hydrogel-based therapies. In this review, we first introduced the physicochemical properties of hydrogels and their fabrication approaches concisely. Subsequently, the comprehensive description and deep discussion were elucidated, wherein the influences of different hydrogels properties on cell behaviors and cellular signaling events were highlighted. These behaviors or events included integrin clustering, focal adhesion (FA) complex accumulation and activation, cytoskeleton rearrangement, protein cyto-nuclei shuttling and activation (e.g., Yes-associated protein (YAP), catenin, etc.), cellular compartment reorganization, gene expression, and further cell biology modulation (e.g., spreading, migration, proliferation, lineage commitment, etc.). Based on them, current in vitro and in vivo hydrogel applications that mainly covered diseases models, various cell delivery protocols for tissue regeneration and disease therapy, smart drug carrier, bioimaging, biosensor, and conductive wearable/implantable biodevices, etc. were further summarized and discussed. More significantly, the clinical translation potential and trials of hydrogels were presented, accompanied with which the remaining challenges and future perspectives in this field were emphasized. Collectively, the comprehensive and deep insights in this review will shed light on the design principles of new biomedical hydrogels to understand and modulate cellular processes, which are available for providing significant indications for future hydrogel design and serving for a broad range of biomedical applications.
Collapse
Affiliation(s)
- Huan Cao
- Department of Nuclear Medicine, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, 610064, Chengdu, P. R. China
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China
- School of Materials Science and Engineering, Nanyang Technological University, 50 Nanyang Avenue, Singapore, 639798, Singapore
| | - Lixia Duan
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China
| | - Yan Zhang
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China
| | - Jun Cao
- Department of Nuclear Medicine, West China Hospital, and National Engineering Research Center for Biomaterials, Sichuan University, 610064, Chengdu, P. R. China.
| | - Kun Zhang
- Department of Medical Ultrasound and Central Laboratory, Shanghai Tenth People's Hospital, Tongji University School of Medicine, No. 301 Yan-chang-zhong Road, 200072, Shanghai, People's Republic of China.
| |
Collapse
|
43
|
Vasu S, Zhou J, Chen J, Johnston PV, Kim DH. Biomaterials-based Approaches for Cardiac Regeneration. Korean Circ J 2021; 51:943-960. [PMID: 34854577 PMCID: PMC8636758 DOI: 10.4070/kcj.2021.0291] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2021] [Accepted: 09/08/2021] [Indexed: 12/29/2022] Open
Abstract
Cardiovascular disease is a prevalent cause of mortality and morbidity, largely due to the limited ability of cardiomyocytes to proliferate. Existing therapies for cardiac regeneration include cell-based therapies and bioactive molecules. However, delivery remains one of the major challenges impeding such therapies from having significant clinical impact. Recent advancements in biomaterials-based approaches for cardiac regeneration have shown promise in improving cardiac function, promoting angiogenesis, and reducing adverse immune response in both human clinical trials and animal studies. These advances in therapeutic delivery via extracellular vesicles, cardiac patches, and hydrogels have the potential to enable clinical impact of cardiac regeneration therapies. The limited ability of cardiomyocytes to proliferate is a major cause of mortality and morbidity in cardiovascular diseases. There exist therapies for cardiac regeneration that are cell-based as well as that involve bioactive molecules. However, delivery remains one of the major challenges impeding such therapies from having clinical impact. Recent advancements in biomaterials-based approaches for cardiac regeneration have shown promise in clinical trials and animal studies in improving cardiac function, promoting angiogenesis, and reducing adverse immune response. This review will focus on current clinical studies of three contemporary biomaterials-based approaches for cardiac regeneration (extracellular vesicles, injectable hydrogels, and cardiac patches), remaining challenges and shortcomings to be overcome, and future directions for the use of biomaterials to promote cardiac regeneration.
Collapse
Affiliation(s)
- Samhita Vasu
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Justin Zhou
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Jeffrey Chen
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA
| | - Peter V Johnston
- Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA
| | - Deok-Ho Kim
- Department of Biomedical Engineering, Johns Hopkins University, Baltimore, MD, USA.,Division of Cardiology, Department of Medicine, Johns Hopkins School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
44
|
Yu D, Wang X, Ye L. Cardiac Tissue Engineering for the Treatment of Myocardial Infarction. J Cardiovasc Dev Dis 2021; 8:jcdd8110153. [PMID: 34821706 PMCID: PMC8617685 DOI: 10.3390/jcdd8110153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2021] [Accepted: 11/04/2021] [Indexed: 11/26/2022] Open
Abstract
Poor cell engraftment rate is one of the primary factors limiting the effectiveness of cell transfer therapy for cardiac repair. Recent studies have shown that the combination of cell-based therapy and tissue engineering technology can improve stem cell engraftment and promote the therapeutic effects of the treatment for myocardial infarction. This mini-review summarizes the recent progress in cardiac tissue engineering of cardiovascular cells from differentiated human pluripotent stem cells (PSCs), highlights their therapeutic applications for the treatment of myocardial infarction, and discusses the present challenges of cardiac tissue engineering and possible future directions from a clinical perspective.
Collapse
Affiliation(s)
- Dongmin Yu
- Department of Cardiovascular Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China;
| | - Xiaowei Wang
- Department of Cardiovascular Surgery, The First Affiliated Hospital with Nanjing Medical University, Nanjing 210029, China;
- Correspondence: (X.W.); (L.Y.); Tel.: +86-02568303105 (X.W.); +65-67042193 2 (L.Y.)
| | - Lei Ye
- National Heart Research Institute Singapore, National Heart Centre Singapore, 5 Hospital Drive, Singapore 169609, Singapore
- Correspondence: (X.W.); (L.Y.); Tel.: +86-02568303105 (X.W.); +65-67042193 2 (L.Y.)
| |
Collapse
|
45
|
Alvi SB, Ahmed S, Sridharan D, Naseer Z, Pracha N, Wang H, Boudoulas KD, Zhu W, Sayed N, Khan M. De novo Drug Delivery Modalities for Treating Damaged Hearts: Current Challenges and Emerging Solutions. Front Cardiovasc Med 2021; 8:742315. [PMID: 34651028 PMCID: PMC8505729 DOI: 10.3389/fcvm.2021.742315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Accepted: 08/31/2021] [Indexed: 11/13/2022] Open
Abstract
Cardiovascular disease (CVD) is the leading cause of mortality, resulting in approximately one-third of deaths worldwide. Among CVD, acute myocardial infarctions (MI) is the leading cause of death. Current treatment modalities for treating CVD have improved over the years, but the demand for new and innovative therapies has been on the rise. The field of nanomedicine and nanotechnology has opened a new paradigm for treating damaged hearts by providing improved drug delivery methods, specifically targeting injured areas of the myocardium. With the advent of innovative biomaterials, newer therapeutics such as growth factors, stem cells, and exosomes have been successfully delivered to the injured myocardial tissue, promoting improvement in cardiac function. This review focuses on three major drug delivery modalities: nanoparticles, microspheres, and hydrogels, and their potential for treating damaged hearts following an MI.
Collapse
Affiliation(s)
- Syed Baseeruddin Alvi
- Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, College of Medicine, Columbus, OH, United States
| | - Salmman Ahmed
- Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, College of Medicine, Columbus, OH, United States
| | - Divya Sridharan
- Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, College of Medicine, Columbus, OH, United States
| | - Zahra Naseer
- Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, College of Medicine, Columbus, OH, United States
| | - Nooruddin Pracha
- Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, College of Medicine, Columbus, OH, United States
| | - Henry Wang
- Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, College of Medicine, Columbus, OH, United States
| | - Konstantinos Dean Boudoulas
- Division of Cardiovascular Medicine, Department of Medicine, The Ohio State University Wexner Medical Center, Columbus, OH, United States
| | - Wuqiang Zhu
- Department of Cardiovascular Diseases, Physiology and Biomedical Engineering, Mayo Clinic, Phoenix, AZ, United States
| | - Nazish Sayed
- Division of Vascular Surgery, Department of Surgery, The Stanford Cardiovascular Institute, Stanford University, Stanford, CA, United States
| | - Mahmood Khan
- Department of Emergency Medicine, The Ohio State University Wexner Medical Center, Davis Heart and Lung Research Institute, College of Medicine, Columbus, OH, United States
| |
Collapse
|
46
|
Juanes-Gusano D, Santos M, Reboto V, Alonso M, Rodríguez-Cabello JC. Self-assembling systems comprising intrinsically disordered protein polymers like elastin-like recombinamers. J Pept Sci 2021; 28:e3362. [PMID: 34545666 DOI: 10.1002/psc.3362] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 07/02/2021] [Accepted: 07/13/2021] [Indexed: 12/19/2022]
Abstract
Despite lacking cooperatively folded structures under native conditions, numerous intrinsically disordered proteins (IDPs) nevertheless have great functional importance. These IDPs are hybrids containing both ordered and intrinsically disordered protein regions (IDPRs), the structure of which is highly flexible in this unfolded state. The conformational flexibility of these disordered systems favors transitions between disordered and ordered states triggered by intrinsic and extrinsic factors, folding into different dynamic molecular assemblies to enable proper protein functions. Indeed, prokaryotic enzymes present less disorder than eukaryotic enzymes, thus showing that this disorder is related to functional and structural complexity. Protein-based polymers that mimic these IDPs include the so-called elastin-like polypeptides (ELPs), which are inspired by the composition of natural elastin. Elastin-like recombinamers (ELRs) are ELPs produced using recombinant techniques and which can therefore be tailored for a specific application. One of the most widely used and studied characteristic structures in this field is the pentapeptide (VPGXG)n . The structural disorder in ELRs probably arises due to the high content of proline and glycine in the ELR backbone, because both these amino acids help to keep the polypeptide structure of elastomers disordered and hydrated. Moreover, the recombinant nature of these systems means that different sequences can be designed, including bioactive domains, to obtain specific structures for each application. Some of these structures, along with their applications as IDPs that self-assemble into functional vesicles or micelles from diblock copolymer ELRs, will be studied in the following sections. The incorporation of additional order- and disorder-promoting peptide/protein domains, such as α-helical coils or β-strands, in the ELR sequence, and their influence on self-assembly, will also be reviewed. In addition, chemically cross-linked systems with controllable order-disorder balance, and their role in biomineralization, will be discussed. Finally, we will review different multivalent IDPs-based coatings and films for different biomedical applications, such as spatially controlled cell adhesion, osseointegration, or biomaterial-associated infection (BAI).
Collapse
Affiliation(s)
- Diana Juanes-Gusano
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology) CIBER-BBN, Edificio Lucía, University of Valladolid, Valladolid, Spain
| | - Mercedes Santos
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology) CIBER-BBN, Edificio Lucía, University of Valladolid, Valladolid, Spain
| | - Virginia Reboto
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology) CIBER-BBN, Edificio Lucía, University of Valladolid, Valladolid, Spain
| | - Matilde Alonso
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology) CIBER-BBN, Edificio Lucía, University of Valladolid, Valladolid, Spain
| | - José Carlos Rodríguez-Cabello
- BIOFORGE (Group for Advanced Materials and Nanobiotechnology) CIBER-BBN, Edificio Lucía, University of Valladolid, Valladolid, Spain
| |
Collapse
|
47
|
Wang B, Patkar SS, Kiick KL. Application of Thermoresponsive Intrinsically Disordered Protein Polymers in Nanostructured and Microstructured Materials. Macromol Biosci 2021; 21:e2100129. [PMID: 34145967 PMCID: PMC8449816 DOI: 10.1002/mabi.202100129] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 05/25/2021] [Indexed: 01/15/2023]
Abstract
Modulation of inter- and intramolecular interactions between bioinspired designer molecules can be harnessed for developing functional structures that mimic the complex hierarchical organization of multicomponent assemblies observed in nature. Furthermore, such multistimuli-responsive molecules offer orthogonal tunability for generating versatile multifunctional platforms via independent biochemical and biophysical cues. In this review, the remarkable physicochemical and mechanical properties of genetically engineered protein polymers derived from intrinsically disordered proteins, specifically elastin and resilin, are discussed. This review highlights emerging technologies which use them as building blocks in the fabrication of highly programmable structured biomaterials for applications in delivery of biotherapeutic cargo and regenerative medicine.
Collapse
Affiliation(s)
- Bin Wang
- Department of Materials Science and Engineering, University of Delaware, 201 DuPont Hall, Newark, DE, 19716, USA
| | - Sai S Patkar
- Department of Materials Science and Engineering, University of Delaware, 201 DuPont Hall, Newark, DE, 19716, USA
| | - Kristi L Kiick
- Department of Materials Science and Engineering, University of Delaware, 201 DuPont Hall, Newark, DE, 19716, USA
- Department of Biomedical Engineering, University of Delaware, 161 Colburn Laboratory, Newark, DE, 19716, USA
- Delaware Biotechnology Institute, Ammon Pinizzotto Biopharmaceutical Innovation Center, 590 Avenue 1743, Newark, DE, 19713, USA
| |
Collapse
|
48
|
Therapies to prevent post-infarction remodelling: From repair to regeneration. Biomaterials 2021; 275:120906. [PMID: 34139506 DOI: 10.1016/j.biomaterials.2021.120906] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2020] [Revised: 05/02/2021] [Accepted: 05/20/2021] [Indexed: 12/15/2022]
Abstract
Myocardial infarction is the first cause of worldwide mortality, with an increasing incidence also reported in developing countries. Over the past decades, preclinical research and clinical trials continually tested the efficacy of cellular and acellular-based treatments. However, none of them resulted in a drug or device currently used in combination with either percutaneous coronary intervention or coronary artery bypass graft. Inflammatory, proliferation and remodelling phases follow the ischaemic event in the myocardial tissue. Only recently, single-cell sequencing analyses provided insights into the specific cell populations which determine the final fibrotic deposition in the affected region. In this review, ischaemia, inflammation, fibrosis, angiogenesis, cellular stress and fundamental cellular and molecular components are evaluated as therapeutic targets. Given the emerging evidence of biomaterial-based systems, the increasing use of injectable hydrogels/scaffolds and epicardial patches is reported both as acellular and cellularised/functionalised treatments. Since several variables influence the outcome of any experimented treatment, we return to the pathological basis with an unbiased view towards any specific process or cellular component. Thus, by evaluating the benefits and limitations of the approaches based on these targets, the reader can weigh the rationale of each of the strategies that reached the clinical trials stage. As recent studies focused on the relevance of the extracellular matrix in modulating ischaemic remodelling and enhancing myocardial regeneration, we aim to portray current trends in the field with this review. Finally, approaches towards feasible translational studies that are as yet unexplored are also suggested.
Collapse
|
49
|
Sazzad F, Kuzemczak M, Loh E, Wu W, Kofidis T. Targeted Myocardial Restoration with Injectable Hydrogels-In Search of The Holy Grail in Regenerating Damaged Heart Tissue. Biomedicines 2021; 9:biomedicines9060595. [PMID: 34073912 PMCID: PMC8225139 DOI: 10.3390/biomedicines9060595] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 05/17/2021] [Accepted: 05/18/2021] [Indexed: 12/20/2022] Open
Abstract
A 3-dimensional, robust, and sustained myocardial restoration by means of tissue engineering remains an experimental approach. Prolific protocols have been developed and tested in small and large animals, but, as clinical cardiac surgeons, we have not arrived at the privilege of utilizing any of them in our clinical practice. The question arises as to why this is. The heart is a unique organ, anatomically and functionally. It is not an easy target to replicate with current techniques, or even to support in its viability and function. Currently, available therapies fail to reverse the loss of functional cardiac tissue, the fundamental pathology remains unaddressed, and heart transplantation is an ultima ratio treatment option. Owing to the equivocal results of cell-based therapies, several strategies have been pursued to overcome the limitations of the current treatment options. Preclinical data, as well as first-in-human studies, conducted to-date have provided important insights into the understanding of injection-based approaches for myocardial restoration. In light of the available data, injectable biomaterials suitable for transcatheter delivery appear to have the highest translational potential. This article presents a current state-of-the-literature review in the field of hydrogel-based myocardial restoration therapy.
Collapse
Affiliation(s)
- Faizus Sazzad
- Myocardial Experimental Laboratory, Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; (F.S.); (E.L.); (W.W.)
| | - Michał Kuzemczak
- Department of Invasive Cardiology, Central Clinical Hospital of the Ministry of Interior and Administration, 02-507 Warsaw, Poland;
- Department of Medical Rescue, Poznan University of Medical Sciences, 60-806 Poznań, Poland
| | - Engracia Loh
- Myocardial Experimental Laboratory, Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; (F.S.); (E.L.); (W.W.)
| | - Wellington Wu
- Myocardial Experimental Laboratory, Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; (F.S.); (E.L.); (W.W.)
| | - Theo Kofidis
- Myocardial Experimental Laboratory, Department of Surgery, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 117599, Singapore; (F.S.); (E.L.); (W.W.)
- Department of Cardiac, Thoracic and Vascular Surgery, National University Health System, Singapore 119228, Singapore
- Correspondence: ; Tel.: +65-6772-6505
| |
Collapse
|
50
|
van der Pol A, Bouten CVC. A Brief History in Cardiac Regeneration, and How the Extra Cellular Matrix May Turn the Tide. Front Cardiovasc Med 2021; 8:682342. [PMID: 34095264 PMCID: PMC8172606 DOI: 10.3389/fcvm.2021.682342] [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: 03/18/2021] [Accepted: 04/27/2021] [Indexed: 11/13/2022] Open
Abstract
Tissue homeostasis is perturbed by stressful events, which can lead to organ dysfunction and failure. This is particularly true for the heart, where injury resulting from myocardial infarction or ischemic heart disease can result in a cascading event ultimately ending with the loss of functional myocardial tissue and heart failure. To help reverse this loss of healthy contractile tissue, researchers have spent decades in the hopes of characterizing a cell source capable of regenerating the injured heart. Unfortunately, these strategies have proven to be ineffective. With the goal of truly understanding cardiac regeneration, researchers have focused on the innate regenerative abilities of zebrafish and neonatal mammals. This has led to the realization that although cells play an important role in the repair of the diseased myocardium, inducing cardiac regeneration may instead lie in the composition of the extra cellular milieu, specifically the extra cellular matrix. In this review we will briefly summarize the current knowledge regarding cell sources used for cardiac regenerative approaches, since these have been extensively reviewed elsewhere. More importantly, by revisiting innate cardiac regeneration observed in zebrafish and neonatal mammals, we will stress the importance the extra cellular matrix has on reactivating this potential in the adult myocardium. Finally, we will address how we can harness the ability of the extra cellular matrix to guide cardiac repair thereby setting the stage of next generation regenerative strategies.
Collapse
Affiliation(s)
- Atze van der Pol
- Eindhoven University of Technology, Department of Biomedical Engineering, Eindhoven, Netherlands
| | | |
Collapse
|