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
|
Mulero-Russe A, Mora-Boza A, Marquez EN, Ziegelski M, Helmrath M, García AJ. Synthetic hydrogel substrate for human induced pluripotent stem cell definitive endoderm differentiation. Biomaterials 2024; 315:122920. [PMID: 39504708 DOI: 10.1016/j.biomaterials.2024.122920] [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: 06/27/2024] [Revised: 10/18/2024] [Accepted: 10/23/2024] [Indexed: 11/08/2024]
Abstract
Human induced pluripotent stem cells (hiPSCs) can give rise to multiple lineages derived from three germ layers, endoderm, mesoderm and ectoderm. Definitive endoderm (DE) cell types and tissues have great potential for regenerative medicine applications. Current hiPSC differentiation protocols focus on the addition of soluble factors; however, extracellular matrix properties are known to also play a role in dictating cell fate. Matrigel™ is the gold standard for DE differentiation, but this xenogeneic, poorly defined basement membrane extract limits the clinical translatability of DE-derived tissues. Here we present a fully defined PEG-based hydrogel substrate to support hiPSC-derived DE differentiation. We screened hydrogel formulations presenting different adhesive peptides and matrix stiffness. Our results demonstrate that presenting a short peptide, cyclic RGD, on the engineered PEG hydrogel supports the transition from undifferentiated hiPSCs to DE using a serum-free, commercially available kit. We show that increasing substrate stiffness (G' = 1.0-4.0 kPa) results in an increased linear response in DE differentiation efficiency. We also include a temporal analysis of the expression of integrin and syndecan receptors as the hiPSCs undergo specification towards DE lineage. Finally, we show that focal adhesion kinase activity regulates hiPSC growth and DE differentiation efficiency. Overall, we present a fully defined matrix as a synthetic alternative for Matrigel™ supporting DE differentiation.
Collapse
Affiliation(s)
- Adriana Mulero-Russe
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Ana Mora-Boza
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Elijah N Marquez
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Morgan Ziegelski
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, USA
| | - Michael Helmrath
- Division of Pediatric Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, 45229, USA
| | - Andrés J García
- Petit Institute for Bioengineering and Bioscience, Georgia Institute of Technology, Atlanta, GA, USA; Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, USA.
| |
Collapse
|
3
|
Dini C, Borges MHR, Malheiros SS, Piazza RD, van den Beucken JJJP, de Avila ED, Souza JGS, Barão VAR. Progress in Designing Therapeutic Antimicrobial Hydrogels Targeting Implant-associated Infections: Paving the Way for a Sustainable Platform Applied to Biomedical Devices. Adv Healthc Mater 2024:e2402926. [PMID: 39440583 DOI: 10.1002/adhm.202402926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/19/2024] [Indexed: 10/25/2024]
Abstract
Implantable biomedical devices have found widespread use in restoring lost functions or structures within the human body, but they face a significant challenge from microbial-related infections, which often lead to implant failure. In this context, antimicrobial hydrogels emerge as a promising strategy for treating implant-associated infections owing to their tunable physicochemical properties. However, the literature lacks a comprehensive analysis of antimicrobial hydrogels, encompassing their development, mechanisms, and effect on implant-associated infections, mainly in light of existing in vitro, in vivo, and clinical evidence. Thus, this review addresses the strategies employed by existing studies to tailor hydrogel properties to meet the specific needs of each application. Furthermore, this comprehensive review critically appraises the development of antimicrobial hydrogels, with a particular focus on solving infections related to metallic orthopedic or dental implants. Then, preclinical and clinical studies centering on providing quantitative microbiological results associated with the application of antimicrobial hydrogels are systematically summarized. Overall, antimicrobial hydrogels benefit from the tunable properties of polymers and hold promise as an effective strategy for the local treatment of implant-associated infections. However, future clinical investigations, grounded on robust evidence from in vitro and preclinical studies, are required to explore and validate new antimicrobial hydrogels for clinical use.
Collapse
Affiliation(s)
- Caroline Dini
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, Universidade Estadual de Campinas (UNICAMP), Piracicaba, São Paulo, 13414-903, Brazil
| | - Maria Helena Rossy Borges
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, Universidade Estadual de Campinas (UNICAMP), Piracicaba, São Paulo, 13414-903, Brazil
| | - Samuel Santana Malheiros
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, Universidade Estadual de Campinas (UNICAMP), Piracicaba, São Paulo, 13414-903, Brazil
| | - Rodolfo Debone Piazza
- Physical Chemistry Department, Institute of Chemistry, São Paulo State University (UNESP), Araraquara, São Paulo, 14800-900, Brazil
| | | | - Erica Dorigatti de Avila
- Department of Dental Materials and Prosthodontics, School of Dentistry at Araçatuba, São Paulo State University (UNESP), Araçatuba, São Paulo, 16015-050, Brazil
| | - João Gabriel S Souza
- Dental Research Division, Guarulhos University (UNG), Guarulhos, São Paulo, 07023-070, Brazil
| | - Valentim A R Barão
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, Universidade Estadual de Campinas (UNICAMP), Piracicaba, São Paulo, 13414-903, Brazil
| |
Collapse
|
4
|
Brien H, Lee JC, Sharma J, Hamann CA, Spetz MR, Lippmann ES, Brunger JM. Templated Pluripotent Stem Cell Differentiation via Substratum-Guided Artificial Signaling. ACS Biomater Sci Eng 2024; 10:6465-6482. [PMID: 39352143 PMCID: PMC11480943 DOI: 10.1021/acsbiomaterials.4c00885] [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: 05/13/2024] [Revised: 09/20/2024] [Accepted: 09/24/2024] [Indexed: 10/15/2024]
Abstract
The emerging field of synthetic morphogenesis implements synthetic biology tools to investigate the minimal cellular processes sufficient for orchestrating key developmental events. As the field continues to grow, there is a need for new tools that enable scientists to uncover nuances in the molecular mechanisms driving cell fate patterning that emerge during morphogenesis. Here, we present a platform that combines cell engineering with biomaterial design to potentiate artificial signaling in pluripotent stem cells (PSCs). This platform, referred to as PSC-MATRIX, extends the use of programmable biomaterials to PSCs competent to activate morphogen production through orthogonal signaling, giving rise to the opportunity to probe developmental events by initiating morphogenetic programs in a spatially constrained manner through non-native signaling channels. We show that the PSC-MATRIX platform enables temporal and spatial control of transgene expression in response to bulk, soluble inputs in synthetic Notch (synNotch)-engineered human PSCs for an extended culture of up to 11 days. Furthermore, we used PSC-MATRIX to regulate multiple differentiation events via material-mediated artificial signaling in engineered PSCs using the orthogonal ligand green fluorescent protein, highlighting the potential of this platform for probing and guiding fate acquisition. Overall, this platform offers a synthetic approach to interrogate the molecular mechanisms driving PSC differentiation that could be applied to a variety of differentiation protocols.
Collapse
Affiliation(s)
- Hannah
J. Brien
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Joanne C. Lee
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Jhanvi Sharma
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Catherine A. Hamann
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Madeline R. Spetz
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Ethan S. Lippmann
- Department
of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Center
for Stem Cell Biology, Vanderbilt University, Nashville, Tennessee 37235, United States
| | - Jonathan M. Brunger
- Department
of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee 37235, United States
- Center
for Stem Cell Biology, Vanderbilt University, Nashville, Tennessee 37235, United States
| |
Collapse
|
5
|
Xin Q, Niu R, Chen Q, Liu D, Xu E. Stable cytoactivity of piscine satellite cells in rice bran-gelatin hydrogel scaffold of cultured meat. Int J Biol Macromol 2024; 277:134242. [PMID: 39084438 DOI: 10.1016/j.ijbiomac.2024.134242] [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: 01/10/2024] [Revised: 07/04/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
In order to achieve high cell adhesion and growth efficiency on scaffolds for cultured meat, animal materials, especially gelatin, are necessary though the disadvantages of weak mechanical properties and poor stability of their hydrogel scaffolds are present during cell cultivation. Here, we use rice bran as a kind of filling and supporting materials to develop a composite scaffold with gelatin for fish cell cultivation, where rice bran is also inexpensive from high yield fibrous agricultural by-product. The rice bran (with a proportion of 1, 3, 5, 7, 10 to 3 of gelatin) could evenly distributed in the three-dimensional network composed of gelatin hydrogel. It contributed to delaying swelling and degradation rates, fixing water and improving elastic modulus. It is important that rice bran-gelatin hydrogel scaffolds (especially the hydrogel with 70 % rice bran, db) promoted piscine satellite cells (PSCs) proliferation effectively compared to the pure gelatin hydrogel, and the former could also support the differentiation of PSCs. Overall, this work showed a positive promotion to explore new source of scaffold materials like agricultural by-product for reducing the cost of cell cultured meat production.
Collapse
Affiliation(s)
- Qipu Xin
- College of Biosystems Engineering and Food Science, National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China
| | - Ruihao Niu
- College of Biosystems Engineering and Food Science, National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China
| | - Qihe Chen
- College of Biosystems Engineering and Food Science, National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314102, China
| | - Donghong Liu
- College of Biosystems Engineering and Food Science, National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314102, China.
| | - Enbo Xu
- College of Biosystems Engineering and Food Science, National Engineering Laboratory of Intelligent Food Technology and Equipment, Zhejiang Key Laboratory for Agro-Food Processing, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China; Innovation Center of Yangtze River Delta, Zhejiang University, Jiaxing 314102, China.
| |
Collapse
|
6
|
Hou S, Yan X, Gao X, Jockusch S, Gibson KM, Shan Z, Bi L. Enhancing Cardiomyocyte Resilience to Ischemia-Reperfusion Injury: The Therapeutic Potential of an Indole-Peptide-Tempo Conjugate (IPTC). ACS OMEGA 2024; 9:39401-39418. [PMID: 39346824 PMCID: PMC11425819 DOI: 10.1021/acsomega.4c02725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 06/18/2024] [Accepted: 06/20/2024] [Indexed: 10/01/2024]
Abstract
Ischemia/reperfusion (I/R) injury leads to apoptosis and extensive cellular and mitochondrial damage, triggered by the early generation and subsequent accumulation of mitochondrial reactive oxygen species (mtROS). This condition not only contributes to the pathology of I/R injury itself but is also implicated in a variety of other diseases, especially within the cardiovascular domain. Addressing mitochondrial oxidative stress thus emerges as a critical therapeutic target. In this context, our study introduces an indole-peptide-tempo conjugate (IPTC), a compound designed with dual functionalities: antioxidative properties and the ability to modulate autophagy. Our findings reveal that IPTC effectively shields H9C2 cardiomyocytes against hypoxia/reoxygenation (H/R) damage, primarily through counteracting mtROS overproduction linked to impaired mitophagy and mitochondrial dysfunction. We propose that IPTC operates by simultaneously reducing mtROS levels and inducing mitophagy, highlighting its potential as a novel therapeutic strategy for mitigating mitochondrial oxidative damage and, by extension, easing I/R injury and potentially other related cardiovascular conditions.
Collapse
Affiliation(s)
- Shanshan Hou
- Department of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Xin Yan
- Department of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Xiang Gao
- Department of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Steffen Jockusch
- Center of Photochemical Sciences, Bowling Green State University, Bowling Green, Ohio 43403, United States
| | - K Michael Gibson
- Department of Pharmacotherapy, College of Pharmacy and Pharmaceutical Sciences, Washington State University, Spokane, Washington 99202, United States
| | - Zhiying Shan
- Department of Kinesiology and Integrative Physiology, Health Research Institute, Michigan Technological University, Houghton, Michigan 49931, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan 49931, United States
| | - Lanrong Bi
- Department of Chemistry, Michigan Technological University, Houghton, Michigan 49931, United States
- Health Research Institute, Michigan Technological University, Houghton, Michigan 49931, United States
| |
Collapse
|
7
|
Lee NK, Jang WB, Seo DS, Goo HG, Lim HJ, Lee EJ, Kwon SM. Development of advanced cardiac progenitor cell culture system through fibronectin and vitronectin derived peptide coated plate. Stem Cell Res 2024; 79:103476. [PMID: 38941882 DOI: 10.1016/j.scr.2024.103476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2023] [Revised: 05/23/2024] [Accepted: 06/17/2024] [Indexed: 06/30/2024] Open
Abstract
Cardiovascular disease remains a global health concern. Stem cell therapy utilizing human cardiac progenitor cells (hCPCs) shows promise in treating cardiac vascular disease. However, limited availability and senescence of hCPCs hinder their widespread use. To address these challenges, researchers are exploring innovative approaches. In this study, a bioengineered cell culture plate was developed to mimic the natural cardiac tissue microenvironment. It was coated with a combination of extracellular matrix (ECM) peptide motifs and mussel adhesive protein (MAP). The selected ECM peptide motifs, derived from fibronectin and vitronectin, play crucial roles in hCPCs. Results revealed that the Fibro-P and Vitro-P coated plates significantly improved hCPC adhesion, proliferation, migration, and differentiation compared to uncoated plates. Additionally, long-term culture on the coated plates delayed cellular senescence and maintained hCPC stemness. These enhancements were attributed to the activation of integrin downstream signaling pathways. The findings suggest that the engineered ECM peptide motif-MAP-coated plates hold potential for enhancing the therapeutic efficacy of stem cell-based therapies in cardiac tissue engineering and regenerative medicine.
Collapse
Affiliation(s)
- Na Kyung Lee
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Korea; Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Korea
| | - Woong Bi Jang
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Korea; Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Korea
| | - Dong Sik Seo
- AMO Lifescience Co., Ltd., Seoul, Seocho-gu, Republic of Korea
| | - Hui-Gwan Goo
- AMO Lifescience Co., Ltd., Seoul, Seocho-gu, Republic of Korea
| | - Hye Ji Lim
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Korea; Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Korea
| | - Eun Ji Lee
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Korea; Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Korea
| | - Sang-Mo Kwon
- Laboratory for Vascular Medicine and Stem Cell Biology, Department of Physiology, Medical Research Institute, School of Medicine, Pusan National University, Yangsan 50612, Korea; Convergence Stem Cell Research Center, Pusan National University, Yangsan 50612, Korea.
| |
Collapse
|
8
|
Kim DH, Han SG, Lim SJ, Hong SJ, Kwon HC, Jung HS, Han SG. Comparison of Soy and Pea Protein for Cultured Meat Scaffolds: Evaluating Gelation, Physical Properties, and Cell Adhesion. Food Sci Anim Resour 2024; 44:1108-1125. [PMID: 39246534 PMCID: PMC11377198 DOI: 10.5851/kosfa.2024.e46] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 04/08/2024] [Accepted: 06/10/2024] [Indexed: 09/10/2024] Open
Abstract
Cultured meat is under investigation as an environmentally sustainable substitute for conventional animal-derived meat. Employing a scaffolding technique is one approach to developing cultured meat products. The objective of this research was to compare soy and pea protein in the production of hydrogel scaffolds intended for cultured meat. We examined the gelation process, physical characteristics, and the ability of scaffolds to facilitate cell adhesion using mesenchymal stem cells derived from porcine adipose tissue (ADSCs). The combination of soy and pea proteins with agarose and agar powders was found to generate solid hydrogels with a porous structure. Soy protein-based scaffolds exhibited a higher water absorption rate, whereas scaffolds containing agarose had a higher compressive strength. Based on Fourier transform infrared spectroscopy analysis, the number of hydrophobic interactions increased between proteins and polysaccharides in the scaffolds containing pea proteins. All scaffolds were nontoxic toward ADSCs, and soy protein-based scaffolds displayed higher cell adhesion and proliferation properties. Overall, the soy protein-agarose scaffold was found to be optimal for cultured meat production.
Collapse
Affiliation(s)
- Do Hyun Kim
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Korea
| | - Seo Gu Han
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Korea
| | - Su Jin Lim
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Korea
| | - Seong Joon Hong
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Korea
| | - Hyuk Cheol Kwon
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Korea
| | - Hyun Su Jung
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Korea
| | - Sung Gu Han
- Department of Food Science and Biotechnology of Animal Resources, Konkuk University, Seoul 05029, Korea
| |
Collapse
|
9
|
Li J, Qiao W, Liu Y, Lei H, Wang S, Xu Y, Zhou Y, Wen S, Yang Z, Wan W, Shi J, Dong N, Wu Y. Facile engineering of interactive double network hydrogels for heart valve regeneration. Nat Commun 2024; 15:7462. [PMID: 39198477 PMCID: PMC11358442 DOI: 10.1038/s41467-024-51773-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 08/16/2024] [Indexed: 09/01/2024] Open
Abstract
Regenerative heart valve prostheses are essential for treating valvular heart disease, which requested interactive materials that can adapt to the tissue remodeling process. Such materials typically involves intricate designs with multiple active components, limiting their translational potential. This study introduces a facile method to engineer interactive materials for heart valve regeneration using 1,1'-thiocarbonyldiimidazole (TCDI) chemistry. TCDI crosslinking forms cleavable thiourea and thiocarbamate linkages which could gradually release H2S during degradation, therefore regulates the immune microenvironment and accelerates tissue remodeling. By employing this approach, a double network hydrogel was formed on decellularized heart valves (DHVs), showcasing robust anti-calcification and anti-thrombosis properties post fatigue testing. Post-implantation, the DHVs could adaptively degrade during recellularization, releasing H2S to further support tissue regeneration. Therefore, the comprehensive endothelial cell coverage and notable extracellular matrix remodeling could be clearly observed. This accessible and integrated strategy effectively overcomes various limitations of bioprosthetic valves, showing promise as an attractive approach for immune modulation of biomaterials.
Collapse
Affiliation(s)
- Jinsheng Li
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, China
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, State Key Laboratory of Materials Processing and Die & Mould Technology, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), 1037 Luoyu Road, Wuhan, China
| | - Weihua Qiao
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, China
| | - Yuqi Liu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, China
| | - Huiling Lei
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, State Key Laboratory of Materials Processing and Die & Mould Technology, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), 1037 Luoyu Road, Wuhan, China
| | - Shuangshuang Wang
- School of Life Science and Chemistry, Wuhan Donghu University, Wuhan, P. R. China
| | - Yin Xu
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, China
| | - Ying Zhou
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, China
| | - Shuyu Wen
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, China
| | - Zhuoran Yang
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, State Key Laboratory of Materials Processing and Die & Mould Technology, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), 1037 Luoyu Road, Wuhan, China
| | - Wenyi Wan
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, China
| | - Jiawei Shi
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, China.
| | - Nianguo Dong
- Department of Cardiovascular Surgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, China.
| | - Yuzhou Wu
- Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, Hubei Key Laboratory of Bioinorganic Chemistry & Materia Medica, State Key Laboratory of Materials Processing and Die & Mould Technology, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology (HUST), 1037 Luoyu Road, Wuhan, China.
| |
Collapse
|
10
|
Yuan G, Yu C, Du X, Li D, Dou H, Lu P, Wu T, Hao C, Wang Y. Injectable GelMA Hydrogel Microspheres with Sustained Release of Platelet-Rich Plasma for the Treatment of Thin Endometrium. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2024:e2403890. [PMID: 39206600 DOI: 10.1002/smll.202403890] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 08/02/2024] [Indexed: 09/04/2024]
Abstract
Platelet-rich plasma (PRP) intrauterine infusion has been demonstrated to be effective in treating thin endometrium and achieving pregnancy. However, the rapid release of growth factors limits its effectiveness in clinical applications, and thus, multiple intrauterine infusions are often required to achieve therapeutic efficacy. In this study, a GelMA hydrogel microsphere biomaterial is developed using droplet microfluidics to modify the delivery mode of PRP and thus prolong its duration of action. Its biocompatibility is confirmed through both in vivo and in vitro studies. Cell experiments show that PRP-loaded microspheres significantly enhance cell proliferation, migration, and angiogenesis. In vivo experiments show that the effects of PRP-loaded microspheres on repairing the endometrium and restoring fertility in mice could achieve the impact of triple PRP intrauterine infusions. Further mechanistic investigations reveal that PRP could facilitate endometrial repair by regulating the expression of E2Fs, a group of transcription factors. This study demonstrates that hydrogel microspheres could modify the delivery of PRP and prolong its duration of action, enabling endometrial repair and functional reconstruction. This design avoids repeated intrauterine injections of PRP in the clinic, reduces the number of patient visits, and provides a new avenue for clinical treatment of thin endometrium.
Collapse
Affiliation(s)
- Guanghui Yuan
- Centre for Reproductive Medicine, Women and Children's Hospital, Qingdao University, Qingdao, 266011, China
- Branch of Shandong Provincial Clinical Research Center for Reproductive Health, Qingdao, 266011, China
| | - Chenghao Yu
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266000, China
| | - Xin Du
- Centre for Reproductive Medicine, Women and Children's Hospital, Qingdao University, Qingdao, 266011, China
- Branch of Shandong Provincial Clinical Research Center for Reproductive Health, Qingdao, 266011, China
| | - Duan Li
- Centre for Reproductive Medicine, Women and Children's Hospital, Qingdao University, Qingdao, 266011, China
- Branch of Shandong Provincial Clinical Research Center for Reproductive Health, Qingdao, 266011, China
| | - Huaiqian Dou
- Centre for Reproductive Medicine, Women and Children's Hospital, Qingdao University, Qingdao, 266011, China
- Branch of Shandong Provincial Clinical Research Center for Reproductive Health, Qingdao, 266011, China
| | - Panpan Lu
- Centre for Reproductive Medicine, Women and Children's Hospital, Qingdao University, Qingdao, 266011, China
- Branch of Shandong Provincial Clinical Research Center for Reproductive Health, Qingdao, 266011, China
| | - Tong Wu
- Medical Research Center, The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao, 266000, China
- Shandong Key Laboratory of Medical and Health Textile Materials, College of Textile & Clothing, Qingdao University, Qingdao, 266071, China
| | - Cuifang Hao
- Centre for Reproductive Medicine, Women and Children's Hospital, Qingdao University, Qingdao, 266011, China
- Branch of Shandong Provincial Clinical Research Center for Reproductive Health, Qingdao, 266011, China
| | - Yuanfei Wang
- Qingdao Stomatological Hospital Affiliated to Qingdao University, Qingdao, 266001, China
| |
Collapse
|
11
|
Shpun G, Markus A, Farah N, Zalevsky Z, Mandel Y. Adhesion of retinal cells to gold surfaces by biomimetic molecules. Front Cell Dev Biol 2024; 12:1438716. [PMID: 39263323 PMCID: PMC11387177 DOI: 10.3389/fcell.2024.1438716] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Accepted: 08/15/2024] [Indexed: 09/13/2024] Open
Abstract
Background Neural cell-electrode coupling is crucial for effective neural and retinal prostheses. Enhancing this coupling can be achieved through surface modification and geometrical design to increase neuron-electrode proximity. In the current research, we focused on designing and studying various biomolecules as a method to elicit neural cell-electrode adhesion via cell-specific integrin mechanisms. Methods We designed extracellular matrix biomimetic molecules with different head sequences (RGD or YIGSR), structures (linear or cyclic), and spacer lengths (short or long). These molecules, anchored by a thiol (SH) group, were deposited onto gold surfaces at various concentrations. We assessed the modifications using contact angle measurements, fluorescence imaging, and X-ray Photoelectron Spectroscopy (XPS). We then analyzed the adhesion of retinal cells and HEK293 cells to the modified surfaces by measuring cell density, surface area, and focal adhesion spots, and examined changes in adhesion-related gene and integrin expression. Results Results showed that YIGSR biomolecules significantly enhanced retinal cell adhesion, regardless of spacer length. For HEK293 cells, RGD biomolecules were more effective, especially with cyclic RGD and long spacers. Both cell types showed increased expression of specific adhesion integrins and proteins like vinculin and PTK2; these results were in agreement with the adhesion studies, confirming the cell-specific interactions with modified surfaces. Conclusion This study highlights the importance of tailored biomolecules for improving neural cell adhesion to electrodes. By customizing biomolecules to foster specific and effective interactions with adhesion integrins, our study provides valuable insights for enhancing the integration and functionality of retinal prostheses and other neural implants.
Collapse
Affiliation(s)
- Gal Shpun
- The Alexander Kofkin Faculty of Engineering, Bar Ilan University, Ramat Gan, Israel
- School of Optometry and Visual Science, Faculty of Life Sciences, Bar Ilan University, Ramat Gan, Israel
- Bar Ilan Institute for Nanotechnology and Advanced Materials (BINA), Bar Ilan University, Ramat Gan, Israel
| | - Amos Markus
- School of Optometry and Visual Science, Faculty of Life Sciences, Bar Ilan University, Ramat Gan, Israel
- Bar Ilan Institute for Nanotechnology and Advanced Materials (BINA), Bar Ilan University, Ramat Gan, Israel
| | - Nairouz Farah
- School of Optometry and Visual Science, Faculty of Life Sciences, Bar Ilan University, Ramat Gan, Israel
- Bar Ilan Institute for Nanotechnology and Advanced Materials (BINA), Bar Ilan University, Ramat Gan, Israel
| | - Zeev Zalevsky
- The Alexander Kofkin Faculty of Engineering, Bar Ilan University, Ramat Gan, Israel
- Bar Ilan Institute for Nanotechnology and Advanced Materials (BINA), Bar Ilan University, Ramat Gan, Israel
| | - Yossi Mandel
- School of Optometry and Visual Science, Faculty of Life Sciences, Bar Ilan University, Ramat Gan, Israel
- Bar Ilan Institute for Nanotechnology and Advanced Materials (BINA), Bar Ilan University, Ramat Gan, Israel
- The Gonda Multidisciplinary Brain Research Centre, Bar-Ilan University, Ramat Gan, Israel
| |
Collapse
|
12
|
Lubitz LJ, Haffner MP, Rieger H, Leneweit G. Increased Cellular Uptake of ApoE3- or c(RGD)-Modified Liposomes for Glioblastoma Therapy Depending on the Target Cells. Pharmaceutics 2024; 16:1112. [PMID: 39339149 PMCID: PMC11434700 DOI: 10.3390/pharmaceutics16091112] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/15/2024] [Accepted: 08/21/2024] [Indexed: 09/30/2024] Open
Abstract
As effective treatment of glioblastoma is still an unmet need, targeted delivery systems for efficient treatment are of utmost interest. Therefore, in this paper, surface modifications with a small peptide c(RGD) or physiological protein (ApoE3) were investigated. Cellular uptake in murine endothelial cells (bEnd.3) and different glioma cells (human U-87 MG, rat F98) was tested to elucidate possible differences and to correlate the uptake to the receptor expression. Different liposomal formulations were measured at 1 and 3 h for three lipid incubation concentrations. We calculated the liposomal uptake saturation S and the saturation half-time t1/2. An up to 9.6-fold increased uptake for ApoE3-modified liposomes, primarily in tumor cells, was found. Contrarily, c(RGD) liposomes showed a stronger increase in uptake in endothelial cells (up to 40.5-fold). The uptake of modified liposomes revealed enormous differences in S and t1/2 when comparing different tumor cell lines. However, for ApoE3-modified liposomes, we proved comparable saturation values (~25,000) for F98 cells and U-87 MG cells despite a 6-fold lower expression of LRP1 in F98 cells and a 5-fold slower uptake rate. Our findings suggest that cellular uptake of surface-modified liposomes depends more on the target structure than the ligand type, with significant differences between cell types of different origins.
Collapse
Affiliation(s)
- Larissa J. Lubitz
- ABNOBA GmbH, 75223 Niefern-Öschelbronn, Germany
- Carl Gustav Carus-Institute, 75223 Niefern-Öschelbronn, Germany
- Department of Chemical and Process Engineering, Institute of Mechanical Process Engineering and Mechanics, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany
| | | | - Harden Rieger
- ABNOBA GmbH, 75223 Niefern-Öschelbronn, Germany
- Carl Gustav Carus-Institute, 75223 Niefern-Öschelbronn, Germany
| | - Gero Leneweit
- ABNOBA GmbH, 75223 Niefern-Öschelbronn, Germany
- Carl Gustav Carus-Institute, 75223 Niefern-Öschelbronn, Germany
- Department of Chemical and Process Engineering, Institute of Mechanical Process Engineering and Mechanics, Karlsruhe Institute of Technology, 76131 Karlsruhe, Germany
| |
Collapse
|
13
|
Liu Y, Li L, He M, Xu Y, Wu Z, Xu X, Luo K, Lv H. Self-assembled peptide hydrogel loaded with functional peptide Dentonin accelerates vascularized bone tissue regeneration in critical-size bone defects. Regen Biomater 2024; 11:rbae106. [PMID: 39263324 PMCID: PMC11387769 DOI: 10.1093/rb/rbae106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 07/18/2024] [Accepted: 08/18/2024] [Indexed: 09/13/2024] Open
Abstract
Regeneration of oral craniofacial bone defects is a complex process, and reconstruction of large bone defects without the use of exogenous cells or bioactive substances remains a major challenge. Hydrogels are highly hydrophilic polymer networks with the potential to promote bone tissue regeneration. In this study, functional peptide Dentonin was loaded onto self-assembled peptide hydrogels (RAD) to constitute functionally self-assembling peptide RAD/Dentonin hydrogel scaffolds with a view that RAD/Dentonin hydrogel could facilitate vascularized bone regeneration in critical-size calvarial defects. The functionalized peptide RAD/Dentonin forms highly ordered β-sheet supramolecular structures via non-covalent interactions like hydrogen bonding, ultimately assembling into nano-fiber network. RAD/Dentonin hydrogels exhibited desirable porosity and swelling properties, and appropriate biodegradability. RAD/Dentonin hydrogel supported the adhesion, proliferation and three-dimensional migration of bone marrow mesenchymal stem cells (BMSCs) and has the potential to induce differentiation of BMSCs towards osteogenesis through activation of the Wnt/β-catenin pathway. Moreover, RAD/Dentonin hydrogel modulated paracrine secretion of BMSCs and increased the migration, tube formation and angiogenic gene expression of human umbilical vein endothelial cells (HUVECs), which boosted the angiogenic capacity of HUVECs. In vivo, RAD/Dentonin hydrogel significantly strengthened vascularized bone formation in rat calvarial defect. Taken together, these results indicated that the functionalized self-assembling peptide RAD/Dentonin hydrogel effectively enhance osteogenic differentiation of BMSCs, indirectly induce angiogenic effects in HUVECs, and facilitate vascularized bone regeneration in vivo. Thus, it is a promising bioactive material for oral and maxillofacial regeneration.
Collapse
Affiliation(s)
- Yijuan Liu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| | - Li Li
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| | - Mengjiao He
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| | - Yanmei Xu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| | - Zekai Wu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| | - Xiongcheng Xu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| | - Kai Luo
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| | - Hongbing Lv
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou 350002, People's Republic of China
| |
Collapse
|
14
|
Zhou Y, He X, Zhang W, Zhang W, Zhao H, Zhou X, Gu Q, Shen H, Yang H, Liu X, Huang L, Shi Q. Cell-recruited microspheres for OA treatment by dual-modulating inflammatory and chondrocyte metabolism. Mater Today Bio 2024; 27:101127. [PMID: 38979128 PMCID: PMC11228804 DOI: 10.1016/j.mtbio.2024.101127] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2024] [Revised: 06/06/2024] [Accepted: 06/12/2024] [Indexed: 07/10/2024] Open
Abstract
Osteoarthritis (OA) is a degenerative disease potentially exacerbated due to inflammation, cartilage degeneration, and increased friction. Both mesenchymal stem cells (MSCs) and pro-inflammatory macrophages play important roles in OA. A promising approach to treating OA is to modify multi-functional hydrogel microspheres to target the OA microenvironment and structure. Arginyl-glycyl-aspartic acid (RGD) is a peptide widely used in bioengineering owing to its cell adhesion properties, which can recruit BMSCs and macrophages. We developed TLC-R, a microsphere loaded with TGF-β1-containing liposomes. The recruitment effect of TLC-R on macrophages and BMSCs was verified by in vitro experiments, along with its function of promoting chondrogenic differentiation of BMSCs. And we evaluated the effect of TLC-R in balancing OA metabolism in vitro and in vivo. When TLC-R was co-cultured with BMSCs and lipopolysaccharide (LPS)-treated macrophages, it showed the ability to recruit both cells in substantial numbers. As the microspheres degraded, TGF-β1 and chondroitin sulfate (ChS) were released to promote chondrogenic differentiation of the recruited BMSCs, modulate chondrocyte metabolism and inhibit inflammation induced by the macrophages. Furthermore, in vivo analysis showed that TLC-R restored the narrowed space, reduced osteophyte volume, and improved cartilage metabolic homeostasis in OA rats. Altogether, TLC-R provides a comprehensive and novel solution for OA treatment by dual-modulating inflammatory and chondrocyte metabolism.
Collapse
Affiliation(s)
- Yun Zhou
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Medical College of Soochow University, Orthopedic Institute of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, PR China
| | - Xu He
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Medical College of Soochow University, Orthopedic Institute of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, PR China
| | - Wen Zhang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Medical College of Soochow University, Orthopedic Institute of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, PR China
| | - Weiguo Zhang
- Department of Radiology, Dushu Lake Hospital Affiliated to Soochow University, Medical Center of Soochow University, 9 Chongwen Road, Suzhou, Jiangsu, 215123, PR China
| | - Huan Zhao
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Medical College of Soochow University, Orthopedic Institute of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, PR China
| | - Xichao Zhou
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Medical College of Soochow University, Orthopedic Institute of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, PR China
| | - Qiaoli Gu
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Medical College of Soochow University, Orthopedic Institute of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, PR China
| | - Hao Shen
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Medical College of Soochow University, Orthopedic Institute of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, PR China
| | - Huilin Yang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Medical College of Soochow University, Orthopedic Institute of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, PR China
| | - Xingzhi Liu
- CAS Key Laboratory of Nano-Bio Interface, Suzhou Institute of Nano-Tech and Nano-Bionics, Chinese Academy of Sciences, 398 Ruoshui Road, Suzhou, Jiangsu, 215123, PR China
| | - Lixin Huang
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Medical College of Soochow University, Orthopedic Institute of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, PR China
| | - Qin Shi
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Medical College of Soochow University, Orthopedic Institute of Soochow University, 899 Pinghai Road, Suzhou, Jiangsu, 215031, PR China
| |
Collapse
|
15
|
Ishikawa S, Kamata H, Sakai T. Enhancing cell adhesion in synthetic hydrogels via physical confinement of peptide-functionalized polymer clusters. J Mater Chem B 2024; 12:7103-7112. [PMID: 38919129 DOI: 10.1039/d4tb00761a] [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: 06/27/2024]
Abstract
Artificially synthesized poly(ethylene glycol) (PEG)-based hydrogels are extensively utilized as biomaterials for tissue scaffolds and cell culture matrices due to their non-protein adsorbing properties. Although these hydrogels are inherently non-cell-adhesive, advancements in modifying polymer networks with functional peptides have led to PEG hydrogels with diverse functionalities, such as cell adhesion and angiogenesis. However, traditional methods of incorporating additives into hydrogel networks often result in the capping of crosslinking points with heterogeneous substances, potentially impairing mechanical properties and obscuring the causal relationships of biological functions. This study introduces polymer additives designed to resist prolonged elution from hydrogels, providing a novel approach to facilitate cell culture on non-adhesive surfaces. By clustering tetra-branched PEG to form ultra-high molecular weight hyper-branched structures and functionalizing their termini with cell-adhesive peptides, we successfully entrapped these clusters within the hydrogel matrix without compromising mechanical strength. This method has enabled successful cell culture on inherently non-adhesive PEG hydrogel surfaces at high peptide densities, a feat challenging to achieve with conventional means. The approach proposed in this study not only paves the way for new possibilities with polymer additives but also serves as a new design paradigm for cell culturing on non-cell-adhesive hydrogels.
Collapse
Affiliation(s)
- Shohei Ishikawa
- Department of Chemistry & Biotechnology, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
| | - Hiroyuki Kamata
- Department of Chemistry & Biotechnology, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
| | - Takamasa Sakai
- Department of Chemistry & Biotechnology, School of Engineering, The University of Tokyo, Bunkyo-ku, Tokyo, Japan.
| |
Collapse
|
16
|
Wu DJ, Rutten MGTA, Huang J, Schotman MJG, van Sprang JF, Tiemeijer BM, ter Huurne GM, Wijnands SPW, Diba M, Dankers PYW. Tuning Structural Organization via Molecular Design and Hierarchical Assembly to Develop Supramolecular Thermoresponsive Hydrogels. Macromolecules 2024; 57:6606-6615. [PMID: 39071041 PMCID: PMC11270986 DOI: 10.1021/acs.macromol.4c00567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Revised: 05/14/2024] [Accepted: 06/03/2024] [Indexed: 07/30/2024]
Abstract
The cellular microenvironment is composed of a dynamic hierarchical fibrillar architecture providing a variety of physical and bioactive signals to the surrounding cells. This dynamicity, although common in biology, is a challenge to control in synthetic matrices. Here, responsive synthetic supramolecular monomers were designed that are able to assemble into hierarchical fibrous structures, combining supramolecular fiber formation via hydrogen bonding interactions, with a temperature-responsive hydrophobic collapse, resulting in cross-linking and hydrogel formation. Therefore, amphiphilic molecules were synthesized, composed of a hydrogen bonding ureido-pyrimidinone (UPy) unit, a hydrophobic alkyl spacer, and a hydrophilic oligo(ethylene glycol) tail. The temperature responsive behavior was introduced by functionalizing these supramolecular amphiphiles with a relatively short poly(N-isopropylacrylamide) (PNIPAM) chain (M n ∼ 2.5 or 5.5 kg/mol). To precisely control the assembly of these monomers, the length of the alkyl spacer between the UPy moiety and PNIPAM was varied in length. A robust sol-gel transition, with the dodecyl UPy-PNIPAM molecule, was obtained, with a network elasticity enhancing over 2000 times upon heating above room temperature. The UPy-PNIPAM compounds with shorter alkyl spacers were already hydrogels at room temperature. The sol-gel transition of the dodecyl UPy-PNIPAM hydrogelator could be tuned by the incorporation of different UPy-functionalized monomers. Furthermore, we demonstrated the suitability of this system for microfluidic cell encapsulation through a convenient temperature sol-gel transition. Our results indicate that this novel thermoresponsive supramolecular system offers a modular platform to study and guide single-cell behavior.
Collapse
Affiliation(s)
- Dan Jing Wu
- Laboratory
for Cell and Tissue Engineering, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, PO Box 513 ,Eindhoven MB 5600, The Netherlands
- Institute
for Complex Molecular Systems, Eindhoven
University of Technology, PO Box 513 ,Eindhoven MB 5600, The Netherlands
- Laboratory
of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513 ,Eindhoven MB 5600, The Netherlands
| | - Martin G. T. A. Rutten
- Institute
for Complex Molecular Systems, Eindhoven
University of Technology, PO Box 513 ,Eindhoven MB 5600, The Netherlands
- Laboratory
of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513 ,Eindhoven MB 5600, The Netherlands
| | - Jingyi Huang
- Laboratory
for Cell and Tissue Engineering, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, PO Box 513 ,Eindhoven MB 5600, The Netherlands
- Institute
for Complex Molecular Systems, Eindhoven
University of Technology, PO Box 513 ,Eindhoven MB 5600, The Netherlands
- Laboratory
of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513 ,Eindhoven MB 5600, The Netherlands
| | - Maaike J. G. Schotman
- Institute
for Complex Molecular Systems, Eindhoven
University of Technology, PO Box 513 ,Eindhoven MB 5600, The Netherlands
- Laboratory
of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513 ,Eindhoven MB 5600, The Netherlands
| | - Johnick F. van Sprang
- Laboratory
for Cell and Tissue Engineering, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, PO Box 513 ,Eindhoven MB 5600, The Netherlands
- Institute
for Complex Molecular Systems, Eindhoven
University of Technology, PO Box 513 ,Eindhoven MB 5600, The Netherlands
- Laboratory
of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513 ,Eindhoven MB 5600, The Netherlands
| | - Bart M. Tiemeijer
- Institute
for Complex Molecular Systems, Eindhoven
University of Technology, PO Box 513 ,Eindhoven MB 5600, The Netherlands
- Laboratory
of Immunoengineering, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven MB 5600, The Netherlands
| | - Gijs M. ter Huurne
- Institute
for Complex Molecular Systems, Eindhoven
University of Technology, PO Box 513 ,Eindhoven MB 5600, The Netherlands
- Laboratory
of Macromolecular and Organic Chemistry, Department of Chemical Engineering
and Chemistry, Eindhoven University of Technology, PO Box 513 ,Eindhoven MB 5600, The Netherlands
| | - Sjors P. W. Wijnands
- Institute
for Complex Molecular Systems, Eindhoven
University of Technology, PO Box 513 ,Eindhoven MB 5600, The Netherlands
- Laboratory
of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513 ,Eindhoven MB 5600, The Netherlands
| | - Mani Diba
- Institute
for Complex Molecular Systems, Eindhoven
University of Technology, PO Box 513 ,Eindhoven MB 5600, The Netherlands
- Laboratory
of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513 ,Eindhoven MB 5600, The Netherlands
- Department
of Dentistry-Regenerative Biomaterials, Research Institute for Medical
Innovation, Radboud University Medical Center, 6525EX ,Nijmegen 6500 HB, The Netherlands
| | - Patricia Y. W. Dankers
- Laboratory
for Cell and Tissue Engineering, Department of Biomedical Engineering, Eindhoven University of Technology, Eindhoven, PO Box 513 ,Eindhoven MB 5600, The Netherlands
- Institute
for Complex Molecular Systems, Eindhoven
University of Technology, PO Box 513 ,Eindhoven MB 5600, The Netherlands
- Laboratory
of Chemical Biology, Department of Biomedical Engineering, Eindhoven University of Technology, PO Box 513 ,Eindhoven MB 5600, The Netherlands
| |
Collapse
|
17
|
Wei Y, Lv J, Zhu S, Wang S, Su J, Xu C. Enzyme-responsive liposomes for controlled drug release. Drug Discov Today 2024; 29:104014. [PMID: 38705509 DOI: 10.1016/j.drudis.2024.104014] [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: 12/16/2023] [Revised: 04/19/2024] [Accepted: 04/29/2024] [Indexed: 05/07/2024]
Abstract
Compared to other nanovectors, liposomes exhibit unique advantages, such as good biosafety and high drug-loading capacity. However, slow drug release from conventional liposomes makes most payloads unavailable, restricting the therapeutic efficacy. Therefore, in the last ∼20 years, enzyme-responsive liposomes have been extensively investigated, which liberate drugs under the stimulation of enzymes overexpressed at disease sites. In this review, we elaborate on the research progress on enzyme-responsive liposomes. The involved enzymes mainly include phospholipases, particularly phospholipase A2, matrix metalloproteinases, cathepsins, and esterases. These enzymes can cleave ester bonds or specific peptide sequences incorporated in the liposomes for controlled drug release by disrupting the primary structure of liposomes, detaching protective polyethylene glycol shells, or activating liposome-associated prodrugs. Despite decades of efforts, there are still a lack marketed products of enzyme-responsive liposomes. Therefore, more efforts should be made to improve the safety and effectiveness of enzyme-responsive liposomes and address the issues associated with production scale-up.
Collapse
Affiliation(s)
- Yan Wei
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China; Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai 200941, China.
| | - Jiajing Lv
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China
| | - Shiyu Zhu
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China
| | - Sicheng Wang
- Department of Orthopedics, Shanghai Zhongye Hospital, Shanghai 200941, China.
| | - Jiacan Su
- Institute of Translational Medicine, Shanghai University, Shanghai 200444, China; Organoid Research Center, Shanghai University, Shanghai 200444, China; Department of Orthopedics, Xinhua Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200092, China.
| | - Can Xu
- Department of Gastroenterology, Changhai Hospital, Shanghai 200433, China.
| |
Collapse
|
18
|
Ghorbani S, Sutherland DS. Building better habitats: Spatiotemporal signaling cues in 3D biointerfaces for tailored cellular functionality. Biointerphases 2024; 19:048501. [PMID: 38975887 DOI: 10.1116/6.0003685] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2024] [Accepted: 06/11/2024] [Indexed: 07/09/2024] Open
Abstract
A promising research direction in the field of biological engineering is the design and functional programming of three-dimensional (3D) biointerfaces designed to support living cell functionality and growth in vitro, offering a route to precisely regulate cellular behaviors and phenotypes for addressing therapeutic challenges. While traditional two-dimensional (2D) biointerfaces have provided valuable insights, incorporating specific signaling cues into a 3D biointeractive microenvironment at the right locations and time is now recognized as crucial for accurately programming cellular decision-making and communication processes. This approach aims to engineer cell-centric microenvironments with the potential to recapitulate complex biological functions into a finite set of growing cellular organizations. Additionally, they provide insights into the hierarchical logic governing the relationship between molecular components and higher-order multicellular functionality. The functional live cell-based microenvironment engineered through such innovative biointerfaces has the potential to be used as an in vitro model system for expanding our understanding of cellular behaviors or as a therapeutic habitat where cellular functions can be reprogrammed.
Collapse
Affiliation(s)
- Sadegh Ghorbani
- Department of Materials Science and Engineering, Stanford University, Stanford, California 94305
- Department of Health Technology, Technical University of Denmark, 2800 Kgs Lyngby, Denmark
| | - Duncan S Sutherland
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, Aarhus C 8000, Denmark
- The Centre for Cellular Signal Patterns (CELLPAT), Aarhus University, Aarhus C 8000, Denmark
| |
Collapse
|
19
|
Liao Y, Yang Y, Zhou G, Chen L, Yang Y, Guo S, Zuo Q, Zou J. Anoikis and SPP1 in idiopathic pulmonary fibrosis: integrating bioinformatics, cell, and animal studies to explore prognostic biomarkers and PI3K/AKT signaling regulation. Expert Rev Clin Immunol 2024; 20:679-693. [PMID: 38318669 DOI: 10.1080/1744666x.2024.2315218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2023] [Accepted: 02/01/2024] [Indexed: 02/07/2024]
Abstract
OBJECTIVE This study aims to explore the relevance of anoikis in idiopathic pulmonary fibrosis (IPF) and identify associated biomarkers and signaling pathways. METHOD Unsupervised consensus cluster analysis was employed to categorize IPF patients into subtypes. We utilized Weighted Gene Co-Expression Network Analysis (WGCNA) and Protein-Protein Interaction network construction to identify anoikis-related modules and key genes. A prognostic signature was developed using Lasso and multivariate Cox regression analysis. Single-cell sequencing assessed hub gene expression in various cell types, and both cell and animal experiments confirmed IPF-related pathways. RESULTS We identified two distinct anoikis-associated subtypes with differing prognoses. WGCNA revealed essential hub genes, with SPP1 being prominent in the anoikis-related signature. The anoikis-related signature is effective in determining the prognosis of patients with IPF. Single-cell sequencing highlighted significant differences in SPP1 expression, notably elevated in fibroblasts derived from IPF patients. In vivo and in vitro experiments demonstrated that SPP1 enhances fibrosis in mouse lung fibroblasts by regulating p27 through the PI3K/Akt pathway. CONCLUSION Our research demonstrates a robust prognostic signature associated with anoikis and highlights SPP1 as a pivotal regulator of the PI3K/AKT signaling pathway in pulmonary fibrosis.
Collapse
Affiliation(s)
- Yi Liao
- Department of Respiratory and Critical Care Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yan Yang
- Department of Respiratory and Critical Care Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Guanghong Zhou
- Department of Respiratory and Critical Care Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Lijuan Chen
- Department of Respiratory and Critical Care Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Yang Yang
- Department of Respiratory and Critical Care Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Shujin Guo
- Department of Health Management & Institute of Health Management, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Qiunan Zuo
- Department of Geriatric Respiratory, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| | - Jun Zou
- Department of Respiratory and Critical Care Medicine, Sichuan Provincial People's Hospital, School of Medicine, University of Electronic Science and Technology of China, Chengdu, China
| |
Collapse
|
20
|
Bonde S, Osmani RAM, Trivedi R, Patravale V, Angolkar M, Prasad AG, Ravikumar AA. Harnessing DNA origami's therapeutic potential for revolutionizing cardiovascular disease treatment: A comprehensive review. Int J Biol Macromol 2024; 270:132246. [PMID: 38735608 DOI: 10.1016/j.ijbiomac.2024.132246] [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: 12/05/2023] [Revised: 03/25/2024] [Accepted: 05/07/2024] [Indexed: 05/14/2024]
Abstract
DNA origami is a cutting-edge nanotechnology approach that creates precise and detailed 2D and 3D nanostructures. The crucial feature of DNA origami is how it is created, which enables precise control over its size and shape. Biocompatibility, targetability, programmability, and stability are further advantages that make it a potentially beneficial technique for a variety of applications. The preclinical studies of sophisticated programmable nanomedicines and nanodevices that can precisely respond to particular disease-associated triggers and microenvironments have been made possible by recent developments in DNA origami. These stimuli, which are endogenous to the targeted disorders, include protein upregulation, pH, redox status, and small chemicals. Oncology has traditionally been the focus of the majority of past and current research on this subject. Therefore, in this comprehensive review, we delve into the intricate world of DNA origami, exploring its defining features and capabilities. This review covers the fundamental characteristics of DNA origami, targeting DNA origami to cells, cellular uptake, and subcellular localization. Throughout the review, we emphasised on elucidating the imperative for such a therapeutic platform, especially in addressing the complexities of cardiovascular disease (CVD). Moreover, we explore the vast potential inherent in DNA origami technology, envisioning its promising role in the realm of CVD treatment and beyond.
Collapse
Affiliation(s)
- Smita Bonde
- Department of Pharmaceutics, SSR College of Pharmacy, Silvassa 396230, UT of Dadra and Nagar Haveli, India.
| | - Riyaz Ali M Osmani
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSS AHER), Mysuru 570015, Karnataka, India.
| | - Rashmi Trivedi
- Department of Pharmaceutics, Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur 441002, Maharashtra, India.
| | - Vandana Patravale
- Department of Pharmaceutical Sciences and Technology, Institute of Chemical Technology, Nathalal Parekh Marg, Matunga (E), Mumbai 400019, Maharashtra, India.
| | - Mohit Angolkar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSS AHER), Mysuru 570015, Karnataka, India.
| | - Aprameya Ganesh Prasad
- Department of Chemical and Biomolecular Engineering, Johns Hopkins University, Baltimore, MD 21218, USA.
| | - Akhila Akkihebbal Ravikumar
- Department of Pharmaceutics, JSS College of Pharmacy, JSS Academy of Higher Education and Research (JSS AHER), Mysuru 570015, Karnataka, India.
| |
Collapse
|
21
|
Hu Q, Williams SL, Palladino A, Ecker M. Screening of MMP-13 Inhibitors Using a GelMA-Alginate Interpenetrating Network Hydrogel-Based Model Mimicking Cytokine-Induced Key Features of Osteoarthritis In Vitro. Polymers (Basel) 2024; 16:1572. [PMID: 38891518 PMCID: PMC11174780 DOI: 10.3390/polym16111572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Revised: 05/26/2024] [Accepted: 05/27/2024] [Indexed: 06/21/2024] Open
Abstract
Osteoarthritis (OA) is a chronic joint disease characterized by irreversible cartilage degradation. Current clinical treatment options lack effective pharmaceutical interventions targeting the disease's root causes. MMP (matrix metalloproteinase) inhibitors represent a new approach to slowing OA progression by addressing cartilage degradation mechanisms. However, very few drugs within this class are in preclinical or clinical trial phases. Hydrogel-based 3D in vitro models have shown promise as preclinical testing platforms due to their resemblance to native extracellular matrix (ECM), abundant availability, and ease of use. Metalloproteinase-13 (MMP-13) is thought to be a major contributor to the degradation of articular cartilage in OA by aggressively breaking down type II collagen. This study focused on testing MMP-13 inhibitors using a GelMA-alginate hydrogel-based OA model induced by cytokines interleukin-1 beta (IL-1β) and tumor necrosis factor alpha (TNF-α). The results demonstrate a significant inhibition of type II collagen breakdown by measuring C2C concentration using ELISA after treatment with MMP-13 inhibitors. However, inconsistencies in human cartilage explant samples led to inconclusive results. Nonetheless, the study highlights the GelMA-alginate hydrogel-based OA model as an alternative to human-sourced cartilage explants for in vitro drug screening.
Collapse
Affiliation(s)
- Qichan Hu
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76203, USA
| | - Steven L. Williams
- Department of Biological Sciences, University of North Texas, Denton, TX 76203, USA
| | - Alessandra Palladino
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76203, USA
| | - Melanie Ecker
- Department of Biomedical Engineering, University of North Texas, Denton, TX 76203, USA
| |
Collapse
|
22
|
Tutoni GG, McDonald SM, Zhong R, Lu A, Huang TJ, Becker ML. Microfluidic Assembly of Degradable, Stereocomplexed Hydrogel Microparticles. J Am Chem Soc 2024; 146:14705-14714. [PMID: 38749060 DOI: 10.1021/jacs.4c02317] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
Hydrogel microparticles (HMPs) have been investigated widely for their use in tissue engineering and drug delivery applications. However, translation of these highly tunable systems has been hindered by covalent cross-linking methods within microparticles. Stereocomplexation, a stereospecific form of physical cross-linking, provides a robust yet degradable alternative for creating translationally relevant HMPs. Herein, 4-arm polyethylene glycol (PEG) stars were used as macromolecular initiators from which oligomeric poly(l-lactic acid) (PLLA) was polymerized with a degree of polymerization (DPn) of 20 on each arm. Similarly, complementary propargyl-containing ABA cross-linkers with enantiomeric poly(d-lactic acid) (PDLA) segments (DPn = 20) on each arm. Droplets of these gel precursors were formed via a microfluidic organic-in-oil-in-water system where microparticles self-assembled via stereocomplexation and were stabilized after precipitation in deionized water. By varying the flow rate of the dispersed phase, well-defined microparticles with diameters of 33.7 ± 0.5, 62.4 ± 0.6, and 105.7 ± 0.8 μm were fabricated. Gelation due to stereocomplexation was confirmed via wide-angle X-ray scattering in which HMPs exhibited the signature diffraction pattern of stereocomplexed PLA at 2θ = 12.2, 21.2, 24.2°. Differential scanning calorimetry also confirmed stereocomplexation by the appearance of a crystallization exotherm (Tc = 37 °C) and a high-temperature endotherm (Tm = 159 °C) that does not appear in the homocrystallization of PLLA or the hydrogel precursors. Additionally, the propargyl handle present on the cross-linker allows for pre- or post-assembly thiol-yne "click" functionalization as demonstrated by the addition of thiol-containing fluorophores to the HMPs.
Collapse
Affiliation(s)
- Gianna G Tutoni
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Samantha M McDonald
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Ruoyu Zhong
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina 27708, United States
| | - Annette Lu
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
| | - Tony Jun Huang
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina 27708, United States
| | - Matthew L Becker
- Department of Chemistry, Duke University, Durham, North Carolina 27708, United States
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina 27708, United States
- Department of Biomedical Engineering, Duke University, Durham, North Carolina 27708, United States
- Department of Orthopedic Surgery, Duke University, Durham, North Carolina 27708, United States
| |
Collapse
|
23
|
Cecuda-Adamczewska V, Romanik-Chruścielewska A, Kosowska K, Sokołowska I, Łukasiewicz N, Korycka P, Florys-Jankowska K, Zakrzewska A, Wszoła M, Klak M. Elasticity Modification of Biomaterials Used in 3D Printing with an Elastin-Silk-like Recombinant Protein. J Funct Biomater 2024; 15:141. [PMID: 38921515 PMCID: PMC11204424 DOI: 10.3390/jfb15060141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Revised: 05/10/2024] [Accepted: 05/22/2024] [Indexed: 06/27/2024] Open
Abstract
The recombinant structural protein described in this study was designed based on sequences derived from elastin and silk. Silk-elastin hybrid copolymers are characterized by high solubility while maintaining high product flexibility. The phase transition temperature from aqueous solution to hydrogel, as well as other physicochemical and mechanical properties of such particles, can differ significantly depending on the number of sequence repeats. We present a preliminary characterization of the EJ17zipR protein obtained in high yield in a prokaryotic expression system and efficiently purified via a multistep process. Its addition significantly improves biomaterial's rheological and mechanical properties, especially elasticity. As a result, EJ17zipR appears to be a promising component for bioinks designed to print spatially complex structures that positively influence both shape retention and the internal transport of body fluids. The results of biological studies indicate that the addition of the studied protein creates a favorable microenvironment for cell adhesion, growth, and migration.
Collapse
Affiliation(s)
- Violetta Cecuda-Adamczewska
- Foundation of Research and Science Development, 01-424 Warsaw, Poland; (A.R.-C.); (K.K.); (I.S.); (N.Ł.); (P.K.); (K.F.-J.)
| | | | - Katarzyna Kosowska
- Foundation of Research and Science Development, 01-424 Warsaw, Poland; (A.R.-C.); (K.K.); (I.S.); (N.Ł.); (P.K.); (K.F.-J.)
| | - Iwona Sokołowska
- Foundation of Research and Science Development, 01-424 Warsaw, Poland; (A.R.-C.); (K.K.); (I.S.); (N.Ł.); (P.K.); (K.F.-J.)
| | - Natalia Łukasiewicz
- Foundation of Research and Science Development, 01-424 Warsaw, Poland; (A.R.-C.); (K.K.); (I.S.); (N.Ł.); (P.K.); (K.F.-J.)
| | - Paulina Korycka
- Foundation of Research and Science Development, 01-424 Warsaw, Poland; (A.R.-C.); (K.K.); (I.S.); (N.Ł.); (P.K.); (K.F.-J.)
| | - Katarzyna Florys-Jankowska
- Foundation of Research and Science Development, 01-424 Warsaw, Poland; (A.R.-C.); (K.K.); (I.S.); (N.Ł.); (P.K.); (K.F.-J.)
| | | | - Michał Wszoła
- Polbionica Ltd., 01-424 Warsaw, Poland; (A.Z.); (M.W.)
| | - Marta Klak
- Polbionica Ltd., 01-424 Warsaw, Poland; (A.Z.); (M.W.)
| |
Collapse
|
24
|
Pascouau C, Schweitzer M, Besenius P. Supramolecular Assembly and Thermogelation Strategies Using Peptide-Polymer Conjugates. Biomacromolecules 2024; 25:2659-2678. [PMID: 38663862 PMCID: PMC11095398 DOI: 10.1021/acs.biomac.4c00031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 04/04/2024] [Accepted: 04/05/2024] [Indexed: 05/14/2024]
Abstract
Peptide-polymer conjugates (PPCs) are of particular interest in the development of responsive, adaptive, and interactive materials due to the benefits offered by combining both building blocks and components. This review presents pioneering work as well as recent advances in the design of peptide-polymer conjugates, with a specific focus on their thermoresponsive behavior. This unique class of materials has shown great promise in the development of supramolecular structures with physicochemical properties that are modulated using soft and biorthogonal external stimuli. The temperature-induced self-assembly of PPCs into various supramolecular architectures, gelation processes, and tuning of accessible processing parameters to biologically relevant temperature windows are described. The discussion covers the chemical design of the conjugates, the supramolecular driving forces involved, and the mutual influence of the polymer and peptide segments. Additionally, some selected examples for potential biomedical applications of thermoresponsive PPCs in tissue engineering, delivery systems, tumor therapy, and biosensing are highlighted, as well as perspectives on future challenges.
Collapse
Affiliation(s)
- Chloé Pascouau
- Department of Chemistry, Johannes Gutenberg-University Mainz, Duesbergweg 1014, D-55128 Mainz, Germany
| | - Maren Schweitzer
- Department of Chemistry, Johannes Gutenberg-University Mainz, Duesbergweg 1014, D-55128 Mainz, Germany
| | - Pol Besenius
- Department of Chemistry, Johannes Gutenberg-University Mainz, Duesbergweg 1014, D-55128 Mainz, Germany
| |
Collapse
|
25
|
Song H, Hao D, Zhou J, Farmer D, Wang A. Development of pro-angiogenic skin substitutes for wound healing. Wound Repair Regen 2024; 32:208-216. [PMID: 38308588 DOI: 10.1111/wrr.13154] [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: 08/21/2023] [Revised: 11/13/2023] [Accepted: 12/12/2023] [Indexed: 02/05/2024]
Abstract
Wounds pose significant challenges to public health, primarily due to the loss of the mechanical integrity and barrier function of the skin and impaired angiogenesis, causing physical morbidities and psychological trauma to affect patients. Reconstructing the vasculature of the wound bed is crucial for promoting wound healing, reducing scar formation and enhancing the quality of life for patients. The development of pro-angiogenic skin substitutes has emerged as a promising strategy to facilitate vascularization and expedite the healing process of burn wounds. This review provides an overview of the various types of skin substitutes employed in wound healing, explicitly emphasising those designed to enhance angiogenesis. Synthetic scaffolds, biological matrices and tissue-engineered constructs incorporating stem cells and primary cells, cell-derived extracellular vesicles (EVs), pro-angiogenic growth factors and peptides, as well as gene therapy-based skin substitutes are thoroughly examined. The review summarises the existing challenges, future directions and potential innovations in pro-angiogenic dressing for skin substitutes. It highlights the need for continued research to develop new technologies and combine multiple strategies and factors, and to overcome obstacles and advance the field, ultimately leading to improved outcomes for wound patients.
Collapse
Affiliation(s)
- Hengyue Song
- Center for Surgical Bioengineering, Department of Surgery, UC Davis Health, Sacramento, California, USA
- Department of Burns and Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California, USA
| | - Dake Hao
- Center for Surgical Bioengineering, Department of Surgery, UC Davis Health, Sacramento, California, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California, USA
| | - Jianda Zhou
- Department of Burns and Plastic Surgery, The Third Xiangya Hospital of Central South University, Changsha, Hunan, People's Republic of China
| | - Diana Farmer
- Center for Surgical Bioengineering, Department of Surgery, UC Davis Health, Sacramento, California, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California, USA
| | - Aijun Wang
- Center for Surgical Bioengineering, Department of Surgery, UC Davis Health, Sacramento, California, USA
- Institute for Pediatric Regenerative Medicine, Shriners Hospitals for Children, Sacramento, California, USA
- Department of Biomedical Engineering, UC Davis, Davis, California, USA
| |
Collapse
|
26
|
Karakaya E, Gleichauf L, Schöbel L, Hassan A, Soufivand AA, Tessmar J, Budday S, Boccaccini AR, Detsch R. Engineering peptide-modified alginate-based bioinks with cell-adhesive properties for biofabrication. RSC Adv 2024; 14:13769-13786. [PMID: 38681843 PMCID: PMC11046382 DOI: 10.1039/d3ra08394b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2023] [Accepted: 03/16/2024] [Indexed: 05/01/2024] Open
Abstract
Alginate (ALG) and its oxidised form alginate-dialdehyde (ADA) are highly attractive materials for hydrogels used in 3D bioprinting as well as drop-on-demand (DoD) approaches. However, both polymers need to be modified using cell-adhesive peptide sequences, to obtain bioinks exhibiting promising cell-material interactions. Our study explores the modification of ALG- and ADA-based bioinks with the adhesive peptides YIGSR (derived from laminin), RRETEWA (derived from fibronectin) and IKVAV (derived from laminin) for 3D bioprinting. Two coupling methods, carbodiimide and Schiff base reactions, were employed to modify the polymers with peptides. Analytical techniques, including FTIR and NMR were used to assess the chemical composition, revealing challenges in confirming the presence of peptides. The modified bioinks exhibited decreased stability, viscosity, and stiffness, particularly-ADA-based bioinks in contrast to ALG. Sterile hydrogel capsules or droplets were produced using a manual manufacturing process and DoD printing. NIH/3T3 cell spreading analysis showed enhanced cell spreading in carbodiimide-modified ADA, Schiff base-modified ADA, and PEG-Mal-modified ADA. The carbodiimide coupling of peptides worked for ADA, however the same was not observed for ALG. Finally, a novel mixture of all used peptides was evaluated regarding synergistic effects on cell spreading which was found to be effective, showing higher aspect ratios compared to the single peptide coupled hydrogels in all cases. The study suggests potential applications of these modified bioinks in 3D bioprinting approaches and highlights the importance of peptide selection as well as their combination for improved cell-material interactions.
Collapse
Affiliation(s)
- Emine Karakaya
- Department of Material Science and Engineering, Institute for Biomaterials, Friedrich-Alexander University Erlangen-Nuremberg Germany
| | - Luisa Gleichauf
- Department of Material Science and Engineering, Institute for Biomaterials, Friedrich-Alexander University Erlangen-Nuremberg Germany
| | - Lisa Schöbel
- Department of Material Science and Engineering, Institute for Biomaterials, Friedrich-Alexander University Erlangen-Nuremberg Germany
| | - Ahmed Hassan
- Department of Material Science and Engineering, Institute for Biomaterials, Friedrich-Alexander University Erlangen-Nuremberg Germany
| | - Anahita Ahmadi Soufivand
- Department of Mechanical Engineering, Institute of Continuum Mechanics and Biomechanics, Friedrich-Alexander-University Erlangen-Nuremberg Germany
| | - Joerg Tessmar
- Department for Functional Materials in Medicine and Dentistry, University of Würzburg Germany
| | - Silvia Budday
- Department of Mechanical Engineering, Institute of Continuum Mechanics and Biomechanics, Friedrich-Alexander-University Erlangen-Nuremberg Germany
| | - Aldo R Boccaccini
- Department of Material Science and Engineering, Institute for Biomaterials, Friedrich-Alexander University Erlangen-Nuremberg Germany
| | - Rainer Detsch
- Department of Material Science and Engineering, Institute for Biomaterials, Friedrich-Alexander University Erlangen-Nuremberg Germany
| |
Collapse
|
27
|
Li L, Griebel ME, Uroz M, Bubli SY, Gagnon KA, Trappmann B, Baker BM, Eyckmans J, Chen CS. A Protein-Adsorbent Hydrogel with Tunable Stiffness for Tissue Culture Demonstrates Matrix-Dependent Stiffness Responses. ADVANCED FUNCTIONAL MATERIALS 2024; 34:2309567. [PMID: 38693998 PMCID: PMC11060701 DOI: 10.1002/adfm.202309567] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Indexed: 05/03/2024]
Abstract
Although tissue culture plastic has been widely employed for cell culture, the rigidity of plastic is not physiologic. Softer hydrogels used to culture cells have not been widely adopted in part because coupling chemistries are required to covalently capture extracellular matrix (ECM) proteins and support cell adhesion. To create an in vitro system with tunable stiffnesses that readily adsorbs ECM proteins for cell culture, we present a novel hydrophobic hydrogel system via chemically converting hydroxyl residues on the dextran backbone to methacrylate groups, thereby transforming non-protein adhesive, hydrophilic dextran to highly protein adsorbent substrates. Increasing methacrylate functionality increases the hydrophobicity in the resulting hydrogels and enhances ECM protein adsorption without additional chemical reactions. These hydrophobic hydrogels permit facile and tunable modulation of substrate stiffness independent of hydrophobicity or ECM coatings. Using this approach, we show that substrate stiffness and ECM adsorption work together to affect cell morphology and proliferation, but the strengths of these effects vary in different cell types. Furthermore, we reveal that stiffness mediated differentiation of dermal fibroblasts into myofibroblasts is modulated by the substrate ECM. Our material system demonstrates remarkable simplicity and flexibility to tune ECM coatings and substrate stiffness and study their effects on cell function.
Collapse
Affiliation(s)
- Linqing Li
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA, 02215, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, 02115, United States
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, New Hampshire, 03824, United States
| | - Megan E Griebel
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA, 02215, United States
| | - Marina Uroz
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA, 02215, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, 02115, United States
| | - Saniya Yesmin Bubli
- Department of Chemical Engineering and Bioengineering, University of New Hampshire, Durham, New Hampshire, 03824, United States
| | - Keith A Gagnon
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA, 02215, United States
| | - Britta Trappmann
- Bioactive Materials Laboratory, Max Planck Institute for Molecular Biomedicine, Röntgenstraße 20, Münster, 48149 Germany
| | - Brendon M Baker
- Engineered Microenvironments and Mechanobiology Lab, Department of Biomedical Engineering, University of Michigan, Ann Arbor, MI, 48109 United States
| | - Jeroen Eyckmans
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA, 02215, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, 02115, United States
| | - Christopher S Chen
- Department of Biomedical Engineering, Biological Design Center, Boston University, Boston, MA, 02215, United States
- Wyss Institute for Biologically Inspired Engineering, Harvard University, Boston, 02115, United States
| |
Collapse
|
28
|
Lienig P, Banerjee S, Kenneweg F, Thum T, Kirschning A, Dräger G. Dextrans, Pullulan and Lentinan, New Scaffold Materials for Use as Hydrogels in Tissue Engineering. Chemistry 2024; 30:e202303843. [PMID: 38217885 DOI: 10.1002/chem.202303843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2023] [Revised: 01/10/2024] [Accepted: 01/11/2024] [Indexed: 01/15/2024]
Abstract
The development of hydrogels based on dextrans, pullulan and lentinan to be used in biomedical applications including tissue engineering is reported. Despite the fact that selected polysaccharides such as hyaluronic acid are well established, little is known, how these polysaccharides can be chemically modified to create hydrogels under controlled conditions. In this study we present a small library of chemically modified polysaccharides which are used for a divergent approach to achieve biomedical relevant hydrogels. In this case the crosslinking is based on thio ether formation between thiol modified donor and vinylsulfone or maleimide modified acceptor components. Successful synthesis of the linker systems and coupling at the polysaccharides, hydrogel formation takes place under physiological conditions. We extended the study by coupling small molecules like adhesion factors for increasing cell compatibility as well as a dye for further studies. The different hydrogels were studied to their rheological properties, water uptake, their permeability, biodegrability and their cytotoxicity.
Collapse
Affiliation(s)
- Pascal Lienig
- Institute of Organic Chemistry, Leibniz University Hannover, Schneiderberg 1B, 30167, Hannover, Germany
| | - Samhita Banerjee
- Institute of Organic Chemistry, Leibniz University Hannover, Schneiderberg 1B, 30167, Hannover, Germany
| | - Franziska Kenneweg
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Thomas Thum
- Institute of Molecular and Translational Therapeutic Strategies (IMTTS), Hannover Medical School, Carl-Neuberg-Straße 1, 30625, Hannover, Germany
| | - Andreas Kirschning
- Institute of Organic Chemistry, Leibniz University Hannover, Schneiderberg 1B, 30167, Hannover, Germany
| | - Gerald Dräger
- Institute of Organic Chemistry, Leibniz University Hannover, Schneiderberg 1B, 30167, Hannover, Germany
| |
Collapse
|
29
|
MacPherson DS, Dave D, Kassem S, Doganata S, Zeglis BM, Ulijn RV. Tuning Supramolecular Chirality in Iodinated Amphiphilic Peptides Through Tripeptide Linker Editing. Biomacromolecules 2024; 25:2277-2285. [PMID: 38445833 DOI: 10.1021/acs.biomac.3c01120] [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: 03/07/2024]
Abstract
Protease-cleavable supramolecular oligopeptide nanofilaments are promising materials for targeted therapeutics and diagnostics. In these systems, single amino acid substitutions can have profound effects on the supramolecular structure and consequent proteolytic degradation, which are critical parameters for their intended applications. Herein, we describe changes to the self-assembly and proteolytic cleavage of iodine containing sequences for future translation into matrix metalloprotease (MMP-9)-activated supramolecular radio-imaging probes. We use a systematic single amino acid exchange in the tripeptide linker region of these peptide amphiphiles to provide insights into the role of each residue in the supramolecular assemblies. These modifications resulted in dramatic changes in the nature of the assembled structures formed, including an unexpected chiral inversion. By using circular dichroism, atomic force microscopy, Fourier transform infrared spectroscopy, and molecular dynamics simulations, we found that the GD loop, a common motif in β-turn elements, induced a reversal of the chiral orientation of the assembled nanofibers. In addition to the impact on peptide packing and chirality, MMP-9-catalyzed hydrolysis was evaluated for the four peptides, with the β-sheet content found to be a stronger determinant of enzymatic hydrolysis than supramolecular chirality. These observations provide fundamental insights into the sequence design in protease cleavable amphiphilic peptides with the potential for radio-labeling and selective biomedical applications.
Collapse
Affiliation(s)
- Douglas S MacPherson
- Advanced Science Research Center (ASRC) at The Graduate Center, City University of New York, 85 Saint Nicholas Terrace, New York, New York 10031, United States
- Department of Chemistry, Hunter College of the City University of New York, New York, New York 10028, United States
- Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, New York 10016, United States
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
| | - Dhwanit Dave
- Advanced Science Research Center (ASRC) at The Graduate Center, City University of New York, 85 Saint Nicholas Terrace, New York, New York 10031, United States
- Department of Chemistry, Hunter College of the City University of New York, New York, New York 10028, United States
- Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, New York 10016, United States
| | - Salma Kassem
- Advanced Science Research Center (ASRC) at The Graduate Center, City University of New York, 85 Saint Nicholas Terrace, New York, New York 10031, United States
| | - Selma Doganata
- Advanced Science Research Center (ASRC) at The Graduate Center, City University of New York, 85 Saint Nicholas Terrace, New York, New York 10031, United States
- Macaulay Honors College, City University of New York, New York, New York 10031, United States
| | - Brian M Zeglis
- Department of Chemistry, Hunter College of the City University of New York, New York, New York 10028, United States
- Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, New York 10016, United States
- Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, New York 10016, United States
- Department of Radiology, Memorial Sloan Kettering Cancer Center, New York, New York 10065, United States
- Department of Radiology, Weill Cornell Medical College, New York, New York 10065, United States
| | - Rein V Ulijn
- Advanced Science Research Center (ASRC) at The Graduate Center, City University of New York, 85 Saint Nicholas Terrace, New York, New York 10031, United States
- Department of Chemistry, Hunter College of the City University of New York, New York, New York 10028, United States
- Ph.D. Program in Biochemistry, The Graduate Center of the City University of New York, New York, New York 10016, United States
- Ph.D. Program in Chemistry, The Graduate Center of the City University of New York, New York, New York 10016, United States
| |
Collapse
|
30
|
Lin M, Xu X, Zhou X, Feng H, Wang R, Yang Y, Li J, Fan N, Jiang Y, Li X, Guan F, Tan Z. Sialylation on vesicular integrin β1 determined endocytic entry of small extracellular vesicles into recipient cells. Cell Mol Biol Lett 2024; 29:46. [PMID: 38561669 PMCID: PMC10983696 DOI: 10.1186/s11658-024-00562-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2023] [Accepted: 03/20/2024] [Indexed: 04/04/2024] Open
Abstract
BACKGROUND Small extracellular vesicles (sEV) are closely associated with the development and metastasis of many types of mammalian cancer. Glycoconjugates are highly expressed on sEV and play important roles in sEV biogenesis and their interaction with other cells. However, the study on vesicular glycoconjugates are far behind proteins and nucleic acids. Especially, the functions of sialic acids which are the terminal components of glycoconjugates, are poorly understood in sEV. METHODS Sialic acid levels on sEV from plasma and bladder cancer cells were determined by ELISA and lectin blotting. Effects of sialylation on sEV uptake were determined by flow cytometry. Vesicular glycoproteins bearing sialic acids responsible for sEV uptake was identified by proteomics and density gradient centrifugation, and their site-specific sialylation functions were assayed by N-glycosylation site mutation. Effects of integrin β1 bearing sialic acids on the pro-metastatic function of sEV in vivo were explored using Balb/c nu/nu mice. RESULTS (1) Increased sialic acid levels were observed in sEV from malignant bladder cancer cells. (2) Elimination of sialic acids on sEV impaired sEV uptake by recipient cells. (3) Vesicular integrin β1 bearing sialic acids was identified to play a key role in sEV uptake. (4) Desialylation of the hybrid domain of vesicular integrin β1 inhibited its binding to matrix fibronectin, and reduced sEV entry into recipient cells. (5) Sialylation on integrin β1 affected pro-metastatic function of sEV in Balb/c nu/nu mice. CONCLUSIONS Taken together, our findings indicate important functional roles of sialic acids in sEV uptake and reprogramming plasticity of surrounding normal epithelial cells.
Collapse
Affiliation(s)
- Meixuan Lin
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, China
| | - Xiaoqiang Xu
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, China
| | - Xiaoman Zhou
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, China
| | - Hui Feng
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, China
| | - Ruili Wang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, China
| | - Yunyun Yang
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, China
| | - Jing Li
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, China
| | - Ning Fan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, China
| | - Yazhuo Jiang
- Department of Urology, Provincial People's Hospital, Xi'an, China
| | - Xiang Li
- Institute of Hematology, School of Medicine, Northwest University, Xi'an, China
| | - Feng Guan
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education, Provincial Key Laboratory of Biotechnology, College of Life Sciences, Northwest University, Xi'an, China.
| | - Zengqi Tan
- Institute of Hematology, School of Medicine, Northwest University, Xi'an, China.
| |
Collapse
|
31
|
Mayer K, Ruhoff A, Chan NJ, Waterhouse A, O'Connor AJ, Scheibel T, Heath DE. REDV-Functionalized Recombinant Spider Silk for Next-Generation Coronary Artery Stent Coatings: Hemocompatible, Drug-Eluting, and Endothelial Cell-Specific Materials. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 38470984 DOI: 10.1021/acsami.3c17861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/14/2024]
Abstract
Coronary artery stents are life-saving devices, and millions of these devices are implanted annually to treat coronary heart disease. The current gold standard in treatment is drug-eluting stents, which are coated with a biodegradable polymer layer that elutes antiproliferative drugs to prevent restenosis due to neointimal hyperplasia. Stenting is commonly paired with systemic antiplatelet therapy to prevent stent thrombosis. Despite their clinical success, current stents have significant limitations including inducing local inflammation that drives hyperplasia; a lack of hemocompatibility that promotes thrombosis, increasing need for antiplatelet therapy; and limited endothelialization, which is a critical step in the healing process. In this research, we designed a novel material for use as a next-generation coating for drug-eluting stents that addresses the limitations described above. Specifically, we developed a recombinant spider silk material that is functionalized with an REDV cell-adhesive ligand, a peptide motif that promotes specific adhesion of endothelial cells in the cardiovascular environment. We illustrated that this REDV-modified spider silk variant [eADF4(C16)-REDV] is an endothelial-cell-specific material that can promote the formation of a near-confluent endothelium. We additionally performed hemocompatibility assays using human whole blood and demonstrated that spider silk materials exhibit excellent hemocompatibility under both static and flow conditions. Furthermore, we showed that the material displayed slow enzyme-mediated degradation. Finally, we illustrated the ability to load and release the clinically relevant drug everolimus from recombinant spider silk coatings in a quantity and at a rate similar to that of commercial devices. These results support the use of REDV-functionalized recombinant spider silk as a coating for drug-eluting stents.
Collapse
Affiliation(s)
- Kai Mayer
- Department of Biomedical Engineering, Graeme Clark Institute, University of Melbourne, Melbourne, VIC 3010, Australia
- Chair for Biomaterials, Faculty of Engineering Science, University of Bayreuth, Prof. Rüdiger-Bormann-Straße 1, 95447 Bayreuth, Germany
| | - Alexander Ruhoff
- Heart Research Institute, The University of Sydney, Newtown, NSW 2042, Australia
| | - Nicholas J Chan
- Department of Biomedical Engineering, Graeme Clark Institute, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Anna Waterhouse
- School of Medical Sciences, Faculty of Medicine and Health, The University of Sydney, Sydney, NSW 2006, Australia
| | - Andrea J O'Connor
- Department of Biomedical Engineering, Graeme Clark Institute, University of Melbourne, Melbourne, VIC 3010, Australia
| | - Thomas Scheibel
- Chair for Biomaterials, Faculty of Engineering Science, University of Bayreuth, Prof. Rüdiger-Bormann-Straße 1, 95447 Bayreuth, Germany
- Bayreuther Zentrum für Kolloide und Grenzflächen (BZKG), Universität Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
- Bayreuther Materialzentrum (BayMat), Universität Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
- Bayreuther Zentrum für Molekulare Biowissenschaften (BZMB), Universität Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
- Bayrisches Polymerinstitut (BPI), Universität Bayreuth, Universitätsstraße 30, 95440 Bayreuth, Germany
| | - Daniel E Heath
- Department of Biomedical Engineering, Graeme Clark Institute, University of Melbourne, Melbourne, VIC 3010, Australia
| |
Collapse
|
32
|
Shi J, Yao H, Wang B, Yang J, Liu D, Shang X, Chong H, Fei W, Wang DA. Construction of a Decellularized Multicomponent Extracellular Matrix Interpenetrating Network Scaffold by Gelatin Microporous Hydrogel 3D Cell Culture System. Macromol Rapid Commun 2024; 45:e2300508. [PMID: 38049086 DOI: 10.1002/marc.202300508] [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: 08/25/2023] [Revised: 11/25/2023] [Indexed: 12/06/2023]
Abstract
Interface tissue repair requires the construction of biomaterials with integrated structures of multiple protein types. Hydrogels that modulate internal porous structures provide a 3D microenvironment for encapsulated cells, making them promise for interface tissue repair. Currently, reduction of intrinsic immunogenicity and increase of bioactive extracellular matrix (ECM) secretion are issues to be considered in these materials. In this study, gelatin methacrylate (GelMA) hydrogel is used to encapsulate chondrocytes and construct a phase transition 3D cell culture system (PTCC) by utilizing the thermosensitivity of gelatin microspheres to create micropores within the hydrogel. The types of bioactive extracellular matrix protein formation by chondrocytes encapsulated in hydrogels are investigated in vitro. After 28 days of culture, GelMA PTCC forms an extracellular matrix predominantly composed of collagen type II, collagen type I, and fibronectin. After decellularization, the protein types and mechanical properties are well preserved, fabricating a decellularized tissue-engineered extracellular matrix and GelMA hydrogel interpenetrating network hydrogel (dECM-GelMA IPN) consisting of GelMA hydrogel as the first-level network and the ECM secreted by chondrocytes as the second-level network. This material has the potential to mediate the repair and regeneration of tendon-bone interface tissues with multiple protein types.
Collapse
Affiliation(s)
- Junli Shi
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Hang Yao
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Bowen Wang
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Jian Yang
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People's Hospital, Yangzhou, 225001, P. R. China
- Clinical Medical College, Yangzhou University, Yangzhou, 225001, P. R. China
| | - Dianwei Liu
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People's Hospital, Yangzhou, 225001, P. R. China
| | - Xianfeng Shang
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Hui Chong
- School of Chemistry and Chemical Engineering, Yangzhou University, Yangzhou, 225009, P. R. China
| | - Wenyong Fei
- Department of Orthopedics and Sports Medicine, Northern Jiangsu People's Hospital, Yangzhou, 225001, P. R. China
- Clinical Medical College, Yangzhou University, Yangzhou, 225001, P. R. China
| | - Dong-An Wang
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong S.A.R
- Shenzhen Research Institute, City University of Hong Kong, Shenzhen, 518057, P. R. China
| |
Collapse
|
33
|
Mulero-Russe A, García AJ. Engineered Synthetic Matrices for Human Intestinal Organoid Culture and Therapeutic Delivery. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2307678. [PMID: 37987171 PMCID: PMC10922691 DOI: 10.1002/adma.202307678] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 11/02/2023] [Indexed: 11/22/2023]
Abstract
Human intestinal organoids (HIOs) derived from pluripotent stem cells or adult stem cell biopsies represent a powerful platform to study human development, drug testing, and disease modeling in vitro, and serve as a cell source for tissue regeneration and therapeutic advances in vivo. Synthetic hydrogels can be engineered to serve as analogs of the extracellular matrix to support HIO growth and differentiation. These hydrogels allow for tuning the mechanical and biochemical properties of the matrix, offering an advantage over biologically derived hydrogels such as Matrigel. Human intestinal organoids have been used for repopulating transplantable intestinal grafts and for in vivo delivery to an injured intestinal site. The use of synthetic hydrogels for in vitro culture and for in vivo delivery is expected to significantly increase the relevance of human intestinal organoids for drug screening, disease modeling, and therapeutic applications.
Collapse
Affiliation(s)
- Adriana Mulero-Russe
- School of Chemical and Biomolecular Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| | - Andrés J García
- Parker H. Petit Institute for Bioengineering and Biosciences, Georgia Institute of Technology, Atlanta, GA, 30332, USA
- George W. Woodruff School of Mechanical Engineering, Georgia Institute of Technology, Atlanta, GA, 30332, USA
| |
Collapse
|
34
|
Liu X, Li J, Liu S, Long Y, Kang C, Zhao C, Wei L, Huang S, Luo Y, Dai B, Zhu X. Fabrication of a 3D bioprinting model for posterior capsule opacification using GelMA and PLMA hydrogel-coated resin. Regen Biomater 2024; 11:rbae020. [PMID: 38529352 PMCID: PMC10963077 DOI: 10.1093/rb/rbae020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2023] [Revised: 02/14/2024] [Accepted: 02/18/2024] [Indexed: 03/27/2024] Open
Abstract
Posterior capsule opacification (PCO) remains the predominant complication following cataract surgery, significantly impairing visual function restoration. In this study, we developed a PCO model that closely mimics the anatomical structure of the crystalline lens capsule post-surgery. The model incorporated a threaded structure for accurate positioning and observation, allowing for opening and closing. Utilizing 3D printing technology, a stable external support system was created using resin material consisting of a rigid, hollow base and cover. To replicate the lens capsule structure, a thin hydrogel coating was applied to the resin scaffold. The biocompatibility and impact on cellular functionality of various hydrogel compositions were assessed through an array of staining techniques, including calcein-AM/PI staining, rhodamine staining, BODIPY-C11 staining and EdU staining in conjunction with transwell assays. Additionally, the PCO model was utilized to investigate the effects of eight drugs with anti-inflammatory and anti-proliferative properties, including 5-aminoimidazole-4-carboxamide ribonucleotide (AICAR), THZ1, sorbinil, 4-octyl itaconate (4-OI), xanthohumol, zebularine, rapamycin and caffeic acid phenethyl ester, on human lens epithelial cells (HLECs). Confocal microscopy facilitated comprehensive imaging of the PCO model. The results demonstrated that the GelMA 60 5% + PLMA 2% composite hydrogel exhibited superior biocompatibility and minimal lipid peroxidation levels among the tested hydrogels. Moreover, compared to using hydrogel as the material for 3D printing the entire model, applying surface hydrogel spin coating with parameters of 2000 rpm × 2 on the resin-based 3D printed base yielded a more uniform cell distribution and reduced apoptosis. Furthermore, rapamycin, 4-OI and AICAR demonstrated potent antiproliferative effects in the drug intervention study. Confocal microscopy imaging revealed a uniform distribution of HLECs along the anatomical structure of the crystalline lens capsule within the PCO model, showcasing robust cell viability and regular morphology. In conclusion, the PCO model provides a valuable experimental platform for studying PCO pathogenesis and exploring potential therapeutic interventions.
Collapse
Affiliation(s)
- Xin Liu
- Cataract and Lens Refractive Surgery Group, Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai 200031, People’s Republic of China
- NHC Key Laboratory of Myopia, Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Fudan University, Shanghai 200031, People’s Republic of China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, People’s Republic of China
| | - Jiale Li
- Engineering Research Center of Optical Instrument and System, the Ministry of Education, Shanghai Key Laboratory of Modern Optical System, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Shuyu Liu
- Cataract and Lens Refractive Surgery Group, Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai 200031, People’s Republic of China
- NHC Key Laboratory of Myopia, Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Fudan University, Shanghai 200031, People’s Republic of China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, People’s Republic of China
| | - Yan Long
- Engineering Research Center of Optical Instrument and System, the Ministry of Education, Shanghai Key Laboratory of Modern Optical System, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Ching Kang
- Cataract and Lens Refractive Surgery Group, Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai 200031, People’s Republic of China
- NHC Key Laboratory of Myopia, Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Fudan University, Shanghai 200031, People’s Republic of China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, People’s Republic of China
| | - Chen Zhao
- Cataract and Lens Refractive Surgery Group, Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai 200031, People’s Republic of China
- NHC Key Laboratory of Myopia, Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Fudan University, Shanghai 200031, People’s Republic of China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, People’s Republic of China
| | - Ling Wei
- Cataract and Lens Refractive Surgery Group, Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai 200031, People’s Republic of China
- NHC Key Laboratory of Myopia, Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Fudan University, Shanghai 200031, People’s Republic of China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, People’s Republic of China
| | - Shaoqi Huang
- Engineering Research Center of Optical Instrument and System, the Ministry of Education, Shanghai Key Laboratory of Modern Optical System, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Yi Luo
- Cataract and Lens Refractive Surgery Group, Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai 200031, People’s Republic of China
- NHC Key Laboratory of Myopia, Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Fudan University, Shanghai 200031, People’s Republic of China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, People’s Republic of China
| | - Bo Dai
- Engineering Research Center of Optical Instrument and System, the Ministry of Education, Shanghai Key Laboratory of Modern Optical System, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Xiangjia Zhu
- Cataract and Lens Refractive Surgery Group, Department of Ophthalmology, Eye & ENT Hospital of Fudan University, Shanghai 200031, People’s Republic of China
- NHC Key Laboratory of Myopia, Key Laboratory of Myopia, Chinese Academy of Medical Sciences, Fudan University, Shanghai 200031, People’s Republic of China
- Shanghai Key Laboratory of Visual Impairment and Restoration, Shanghai 200031, People’s Republic of China
| |
Collapse
|
35
|
Podhorská B, Chylíková-Krumbholcová E, Dvořáková J, Šlouf M, Kobera L, Pop-Georgievski O, Frejková M, Proks V, Janoušková O, Filipová M, Chytil P. Soft Hydrogels with Double Porosity Modified with RGDS for Tissue Engineering. Macromol Biosci 2024; 24:e2300266. [PMID: 37821117 DOI: 10.1002/mabi.202300266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Revised: 09/27/2023] [Indexed: 10/13/2023]
Abstract
This study develops and characterizes novel biodegradable soft hydrogels with dual porosity based on N-(2-hydroxypropyl)methacrylamide (HPMA) copolymers cross-linked by hydrolytically degradable linkers. The structure and properties of the hydrogels are designed as scaffolds for tissue engineering and they are tested in vitro with model mesenchymal stem cells (rMSCs). Detailed morphological characterization confirms dual porosity suitable for cell growth and nutrient transport. The dual porosity of hydrogels slightly improves rMSCs proliferation compared to the hydrogel with uniform pores. In addition, the laminin coating supports the adhesion of rMSCs to the hydrogel surface. However, hydrogels modified by heptapeptide RGDSGGY significantly stimulate cell adhesion and growth. Moreover, the RGDS-modified hydrogels also affect the topology of proliferating rMSCs, ranging from single-cell to multicellular clusters. The 3D reconstruction of the hydrogels with cells obtained by laser scanning confocal microscopy (LSCM) confirms cell penetration into the inner structure of the hydrogel and its corresponding microstructure. The prepared biodegradable oligopeptide-modified hydrogels with dual porosity are suitable candidates for further in vivo evaluation in soft tissue regeneration.
Collapse
Affiliation(s)
- Bohumila Podhorská
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského náměstí 2, Prague 6, 162 06, Czech Republic
| | - Eva Chylíková-Krumbholcová
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského náměstí 2, Prague 6, 162 06, Czech Republic
| | - Jana Dvořáková
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského náměstí 2, Prague 6, 162 06, Czech Republic
| | - Miroslav Šlouf
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského náměstí 2, Prague 6, 162 06, Czech Republic
| | - Libor Kobera
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského náměstí 2, Prague 6, 162 06, Czech Republic
| | - Ognen Pop-Georgievski
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského náměstí 2, Prague 6, 162 06, Czech Republic
| | - Markéta Frejková
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského náměstí 2, Prague 6, 162 06, Czech Republic
| | - Vladimír Proks
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského náměstí 2, Prague 6, 162 06, Czech Republic
| | - Olga Janoušková
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského náměstí 2, Prague 6, 162 06, Czech Republic
| | - Marcela Filipová
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského náměstí 2, Prague 6, 162 06, Czech Republic
| | - Petr Chytil
- Institute of Macromolecular Chemistry of the Czech Academy of Sciences, Heyrovského náměstí 2, Prague 6, 162 06, Czech Republic
| |
Collapse
|
36
|
Browne S, Petit N, Quondamatteo F. Functionalised biomaterials as synthetic extracellular matrices to promote vascularisation and healing of diabetic wounds. Cell Tissue Res 2024; 395:133-145. [PMID: 38051351 DOI: 10.1007/s00441-023-03849-4] [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: 03/08/2023] [Accepted: 11/24/2023] [Indexed: 12/07/2023]
Abstract
Diabetic foot ulcers (DFU) are a type of chronic wound that constitute one of the most serious and debilitating complications associated with diabetes. The lack of clinically efficacious treatments to treat these recalcitrant wounds can lead to amputations for those worst affected. Biomaterial-based approaches offer great hope in this regard, as they provide a template for cell infiltration and tissue repair. However, there is an additional need to treat the underlying pathophysiology of DFUs, in particular insufficient vascularization of the wound which significantly hampers healing. Thus, the addition of pro-angiogenic moieties to biomaterials is a promising strategy to promote the healing of DFUs and other chronic wounds. In this review, we discuss the potential of biomaterials as treatments for DFU and the approaches that can be taken to functionalise these biomaterials such that they promote vascularisation and wound healing in pre-clinical models.
Collapse
Affiliation(s)
- Shane Browne
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Dublin, Ireland.
- CÚRAM, Centre for Research in Medical Devices, University of Galway, H91 W2TY, Galway, Ireland.
- Trinity Centre for Biomedical Engineering, Trinity College Dublin, Dublin 2, Ireland.
| | - Noémie Petit
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Dublin, Ireland
| | - Fabio Quondamatteo
- Tissue Engineering Research Group, Department of Anatomy and Regenerative Medicine, Royal College of Surgeons in Ireland, 123, St Stephen's Green, Dublin 2, Dublin, Ireland.
| |
Collapse
|
37
|
Fumasi FM, MacCulloch T, Bernal-Chanchavac J, Stephanopoulos N, Holloway JL. Using dynamic biomaterials to study the temporal role of bioactive peptides during osteogenesis. BIOMATERIALS ADVANCES 2024; 157:213726. [PMID: 38096646 PMCID: PMC10842892 DOI: 10.1016/j.bioadv.2023.213726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Revised: 11/23/2023] [Accepted: 12/05/2023] [Indexed: 12/26/2023]
Abstract
The extracellular matrix is a highly dynamic environment, and the precise temporal presentation of biochemical signals is critical for regulating cell behavior during development, healing, and disease progression. To mimic this behavior, we developed a modular DNA-based hydrogel platform to enable independent and reversible control over the immobilization of multiple biomolecules during in vitro cell culture. We combined reversible DNA handles with a norbornene-modified hyaluronic acid hydrogel to orthogonally add and remove multiple biomolecule-DNA conjugates at user-defined timepoints. We demonstrated that the persistent presentation of the cell adhesion peptide RGD was required to maintain cell spreading on hyaluronic acid hydrogels. Further, we discovered the delayed presentation of osteogenic growth peptide (OGP) increased alkaline phosphatase activity compared to other temporal variations. This finding is critically important when considering the design of OGP delivery approaches for bone repair. More broadly, this platform provides a unique approach to tease apart the temporal role of multiple biomolecules during development, regeneration, and disease progression.
Collapse
Affiliation(s)
- Fallon M Fumasi
- Chemical Engineering, School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ, United States of America
| | - Tara MacCulloch
- Biodesign Center for Molecular Design and Biomimetics, Arizona State University, Tempe, AZ, United States of America; School of Molecular Sciences, Arizona State University, Tempe, AZ, United States of America
| | - Julio Bernal-Chanchavac
- Biodesign Center for Molecular Design and Biomimetics, Arizona State University, Tempe, AZ, United States of America; School of Molecular Sciences, Arizona State University, Tempe, AZ, United States of America.
| | - Nicholas Stephanopoulos
- Biodesign Center for Molecular Design and Biomimetics, Arizona State University, Tempe, AZ, United States of America; School of Molecular Sciences, Arizona State University, Tempe, AZ, United States of America.
| | - Julianne L Holloway
- Chemical Engineering, School for Engineering of Matter, Transport and Energy, Arizona State University, Tempe, AZ, United States of America; Biodesign Center for Molecular Design and Biomimetics, Arizona State University, Tempe, AZ, United States of America.
| |
Collapse
|
38
|
Cho Y, Choi Y, Seong H. Nanoscale surface coatings and topographies for neural interfaces. Acta Biomater 2024; 175:55-75. [PMID: 38141934 DOI: 10.1016/j.actbio.2023.12.025] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Revised: 11/28/2023] [Accepted: 12/14/2023] [Indexed: 12/25/2023]
Abstract
With the lack of minimally invasive tools for probing neuronal systems across spatiotemporal scales, understanding the working mechanism of the nervous system and limited assessments available are imperative to prevent or treat neurological disorders. In particular, nanoengineered neural interfaces can provide a solution to this technological barrier. This review covers recent surface engineering approaches, including nanoscale surface coatings, and a range of topographies from the microscale to the nanoscale, primarily focusing on neural-interfaced biosystems. Specifically, the immobilization of bioactive molecules to fertilize the neural cell lineage, topographical engineering to induce mechanotransduction in neural cells, and enhanced cell-chip coupling using three-dimensional structured surfaces are highlighted. Advances in neural interface design will help us understand the nervous system, thereby achieving the effective treatments for neurological disorders. STATEMENT OF SIGNIFICANCE: • This review focuses on designing bioactive neural interface with a nanoscale chemical modification and topographical engineering at multiscale perspective. • Versatile nanoscale surface coatings and topographies for neural interface are summarized. • Recent advances in bioactive materials applicable for neural cell culture, electrophysiological sensing, and neural implants are reviewed.
Collapse
Affiliation(s)
- Younghak Cho
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea
| | - Yunyoung Choi
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea; Department of Chemical and Biomolecular Engineering, Korea Advanced Institute of Science and Technology (KAIST), Daejeon, Republic of Korea
| | - Hyejeong Seong
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, Republic of Korea; Division of Bio-Medical Science and Technology, KIST School, Korea University of Science and Technology (UST), Seoul, Republic of Korea.
| |
Collapse
|
39
|
Li J, Wang Y, Tang M, Zhang C, Fei Y, Li M, Li M, Gui S, Guo J. New insights into nanotherapeutics for periodontitis: a triple concerto of antimicrobial activity, immunomodulation and periodontium regeneration. J Nanobiotechnology 2024; 22:19. [PMID: 38178140 PMCID: PMC10768271 DOI: 10.1186/s12951-023-02261-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: 09/03/2023] [Accepted: 12/12/2023] [Indexed: 01/06/2024] Open
Abstract
Periodontitis is a chronic inflammatory disease caused by the local microbiome and the host immune response, resulting in periodontal structure damage and even tooth loss. Scaling and root planning combined with antibiotics are the conventional means of nonsurgical treatment of periodontitis, but they are insufficient to fully heal periodontitis due to intractable bacterial attachment and drug resistance. Novel and effective therapeutic options in clinical drug therapy remain scarce. Nanotherapeutics achieve stable cell targeting, oral retention and smart release by great flexibility in changing the chemical composition or physical characteristics of nanoparticles. Meanwhile, the protectiveness and high surface area to volume ratio of nanoparticles enable high drug loading, ensuring a remarkable therapeutic efficacy. Currently, the combination of advanced nanoparticles and novel therapeutic strategies is the most active research area in periodontitis treatment. In this review, we first introduce the pathogenesis of periodontitis, and then summarize the state-of-the-art nanotherapeutic strategies based on the triple concerto of antibacterial activity, immunomodulation and periodontium regeneration, particularly focusing on the therapeutic mechanism and ingenious design of nanomedicines. Finally, the challenges and prospects of nano therapy for periodontitis are discussed from the perspective of current treatment problems and future development trends.
Collapse
Affiliation(s)
- Jiaxin Li
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Yuxiao Wang
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Maomao Tang
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Chengdong Zhang
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Yachen Fei
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Meng Li
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China
| | - Mengjie Li
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China.
| | - Shuangying Gui
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China.
- Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui, China.
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, 230012, Anhui, China.
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department, Hefei, 230012, Anhui, China.
| | - Jian Guo
- Department of Pharmacy, Anhui University of Chinese Medicine, Hefei, 230012, Anhui, China.
- Institute of Pharmaceutics, Anhui Academy of Chinese Medicine, Hefei, 230012, Anhui, China.
- Anhui Province Key Laboratory of Pharmaceutical Preparation Technology and Application, Hefei, 230012, Anhui, China.
- Engineering Technology Research Center of Modernized Pharmaceutics, Anhui Education Department, Hefei, 230012, Anhui, China.
| |
Collapse
|
40
|
Galindo JM, San-Millán MI, Castillo-Sarmiento CA, Ballesteros-Yáñez I, Vázquez E, Merino S, Herrero MA. Optimization of 3D Synthetic Scaffolds for Neuronal Tissue Engineering Applications. Chemistry 2024; 30:e202302481. [PMID: 37823243 DOI: 10.1002/chem.202302481] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 10/05/2023] [Accepted: 10/07/2023] [Indexed: 10/13/2023]
Abstract
The increasing prevalence of neurodegenerative diseases has spurred researchers to develop advanced 3D models that accurately mimic neural tissues. Hydrogels stand out as ideal candidates as their properties closely resemble those of the extracellular matrix. A critical challenge in this regard is to comprehend the influence of the scaffold's mechanical properties on cell growth and differentiation, thus enabling targeted modifications. In light of this, a synthesis and comprehensive analysis of acrylamide-based hydrogels incorporating a peptide has been conducted. Adequate cell adhesion and development is achieved due to their bioactive nature and specific interactions with cellular receptors. The integration of a precisely controlled physicochemical hydrogel matrix and inclusion of the arginine-glycine-aspartic acid peptide sequence has endowed this system with an optimal structure, thus providing a unique ability to interact effectively with biomolecules. The analysis fully examined essential properties governing cell behavior, including pore size, mechanical characteristics, and swelling ability. Cell-viability experiments were performed to assess the hydrogel's biocompatibility, while the incorporation of grow factors aimed to promote the differentiation of neuroblastoma cells. The results underscore the hydrogel's ability to stimulate cell viability and differentiation in the presence of the peptide within the matrix.
Collapse
Affiliation(s)
- Josué M Galindo
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071, Ciudad Real, Spain
| | - Ms Irene San-Millán
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071, Ciudad Real, Spain
| | | | | | - Ester Vázquez
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071, Ciudad Real, Spain
| | - Sonia Merino
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071, Ciudad Real, Spain
| | - M Antonia Herrero
- Instituto Regional de Investigación Científica Aplicada (IRICA) and Facultad de Ciencias y Tecnologías Químicas, Universidad de Castilla-La Mancha, 13071, Ciudad Real, Spain
| |
Collapse
|
41
|
Carvalho ED, Morais MRG, Pêgo AP, Barrias CC, Araújo M. The interplay between chemical conjugation and biologic performance in the development of alginate-based 3D matrices to mimic neural microenvironments. Carbohydr Polym 2024; 323:121412. [PMID: 37940293 DOI: 10.1016/j.carbpol.2023.121412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2023] [Revised: 09/09/2023] [Accepted: 09/16/2023] [Indexed: 11/10/2023]
Abstract
Biofunctionalization of polysaccharides is a widely used strategy for obtaining extracellular matrix (ECM)-mimicking biomaterials. Still, commonly employed chemistries present low reaction yields and the selection of the most adequate bioconjugation route can be challenging. Herein, we compared the performance of carbodiimide and reductive amination chemistries for the synthesis of tailored peptide-alginate hybrid hydrogels as neural tissue mimics. Reductive amination dramatically improved the peptide grafting efficiency, with yields of 50 % vs. 20 %, allowing 1.5 to 3-fold higher incorporation of cell-adhesive and matrix-metalloproteinases (MMP)-sensitive peptides, respectively. The conjugation of dual-end reactive MMP-sensitive peptides promoted a partial crosslinking, allowing adjusting gelation, stiffness, and degradability of hydrogels. Such parameters depended on the glycosidic position where the bioactive peptide binds, determined by the adopted chemical strategy, and this significantly impacted the biological response. Reductive amination provided softer (50-210 Pa) and fully degradable (60-100 % weight loss) hydrogels, depending on the amount of peptide in formulation, contrasting with the stiffer (400 Pa) and less degradable (40 % weight loss) carbodiimide-based hydrogels. Due to their opened polymer chain and increased peptide availability to cells, such hydrogels better supported the 3D culture of primary astrocytes, which present high complexity and process branching, allowing the development of improved brain ECM-mimicking systems.
Collapse
Affiliation(s)
- Eva D Carvalho
- i3S - Instituto de Inovação e Investigação em Saúde, Universidade do Porto, Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal; FEUP - Faculdade de Engenharia, Universidade do Porto, Porto, Portugal
| | - Miguel R G Morais
- i3S - Instituto de Inovação e Investigação em Saúde, Universidade do Porto, Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Ana P Pêgo
- i3S - Instituto de Inovação e Investigação em Saúde, Universidade do Porto, Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal.
| | - Cristina C Barrias
- i3S - Instituto de Inovação e Investigação em Saúde, Universidade do Porto, Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal; ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade do Porto, Porto, Portugal.
| | - Marco Araújo
- i3S - Instituto de Inovação e Investigação em Saúde, Universidade do Porto, Porto, Portugal; INEB - Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
42
|
Yamada Y. Characterization of Novel Cell-Adhesive Peptides for Biomaterial Development. Biol Pharm Bull 2024; 47:1072-1078. [PMID: 38825460 DOI: 10.1248/bpb.b24-00030] [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] [Indexed: 06/04/2024]
Abstract
In previous studies, my group developed cell-adhesive peptide-polysaccharide complexes as biomaterials for tissue engineering. Having a wide variety of cell-adhesive peptides is important as the biological functions of peptide-polysaccharide complexes are highly dependent on the biological activity of peptides. This paper reviews the biological activities of two types of recently characterized cell-adhesive peptides. The first is peptides rich in basic amino acids originating from octaarginine. We analyzed the relationships between the amino acid composition of basic peptides and cell adhesion, elongation, and proliferation and identified the most suitable peptide for cell culture. The second was arginine-glycine-aspartic acid (RGD)-containing peptides that promote the adhesion of induced pluripotent stem cells (iPSCs). We identified the RGD-surrounding sequences necessary for iPSC adhesion, clarified the underlying mechanism, and improved cell adhesion by modifying the structure-activity relationships. The novel cell-adhesive peptides identified in our previous studies may aid in the development of novel peptide-based biomaterials.
Collapse
Affiliation(s)
- Yuji Yamada
- Department of Clinical Biochemistry, School of Pharmacy, Tokyo University of Pharmacy and Life Sciences
| |
Collapse
|
43
|
Rose SC, Larsen M, Xie Y, Sharfstein ST. Salivary Gland Bioengineering. Bioengineering (Basel) 2023; 11:28. [PMID: 38247905 PMCID: PMC10813147 DOI: 10.3390/bioengineering11010028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2023] [Revised: 11/19/2023] [Accepted: 11/30/2023] [Indexed: 01/23/2024] Open
Abstract
Salivary gland dysfunction affects millions globally, and tissue engineering may provide a promising therapeutic avenue. This review delves into the current state of salivary gland tissue engineering research, starting with a study of normal salivary gland development and function. It discusses the impact of fibrosis and cellular senescence on salivary gland pathologies. A diverse range of cells suitable for tissue engineering including cell lines, primary salivary gland cells, and stem cells are examined. Moreover, the paper explores various supportive biomaterials and scaffold fabrication methodologies that enhance salivary gland cell survival, differentiation, and engraftment. Innovative engineering strategies for the improvement of vascularization, innervation, and engraftment of engineered salivary gland tissue, including bioprinting, microfluidic hydrogels, mesh electronics, and nanoparticles, are also evaluated. This review underscores the promising potential of this research field for the treatment of salivary gland dysfunction and suggests directions for future exploration.
Collapse
Affiliation(s)
- Stephen C. Rose
- Department of Nanoscale Science and Engineering, College of Nanotechnology, Science, and Engineering, University at Albany, SUNY, 257 Fuller Road, Albany, NY 12203, USA (Y.X.)
| | - Melinda Larsen
- Department of Biological Sciences and The RNA Institute, University at Albany, SUNY, 1400 Washington Ave., Albany, NY 12222, USA;
| | - Yubing Xie
- Department of Nanoscale Science and Engineering, College of Nanotechnology, Science, and Engineering, University at Albany, SUNY, 257 Fuller Road, Albany, NY 12203, USA (Y.X.)
| | - Susan T. Sharfstein
- Department of Nanoscale Science and Engineering, College of Nanotechnology, Science, and Engineering, University at Albany, SUNY, 257 Fuller Road, Albany, NY 12203, USA (Y.X.)
| |
Collapse
|
44
|
Chen Y, Zhou Y, Hu Z, Lu W, Li Z, Gao N, Liu N, Li Y, He J, Gao Q, Xie Z, Li J, He Y. Gelatin-Based Metamaterial Hydrogel Films with High Conformality for Ultra-Soft Tissue Monitoring. NANO-MICRO LETTERS 2023; 16:34. [PMID: 38019305 PMCID: PMC10686972 DOI: 10.1007/s40820-023-01225-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/25/2023] [Accepted: 09/24/2023] [Indexed: 11/30/2023]
Abstract
Implantable hydrogel-based bioelectronics (IHB) can precisely monitor human health and diagnose diseases. However, achieving biodegradability, biocompatibility, and high conformality with soft tissues poses significant challenges for IHB. Gelatin is the most suitable candidate for IHB since it is a collagen hydrolysate and a substantial part of the extracellular matrix found naturally in most tissues. This study used 3D printing ultrafine fiber networks with metamaterial design to embed into ultra-low elastic modulus hydrogel to create a novel gelatin-based conductive film (GCF) with mechanical programmability. The regulation of GCF nearly covers soft tissue mechanics, an elastic modulus from 20 to 420 kPa, and a Poisson's ratio from - 0.25 to 0.52. The negative Poisson's ratio promotes conformality with soft tissues to improve the efficiency of biological interfaces. The GCF can monitor heartbeat signals and respiratory rate by determining cardiac deformation due to its high conformability. Notably, the gelatin characteristics of the biodegradable GCF enable the sensor to monitor and support tissue restoration. The GCF metamaterial design offers a unique idea for bioelectronics to develop implantable sensors that integrate monitoring and tissue repair and a customized method for endowing implanted sensors to be highly conformal with soft tissues.
Collapse
Affiliation(s)
- Yuewei Chen
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, People's Republic of China
- School of Mechanical Engineering, Guizhou University, Guiyang, 550025, People's Republic of China
| | - Yanyan Zhou
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, People's Republic of China
| | - Zihe Hu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, People's Republic of China
| | - Weiying Lu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, People's Republic of China
| | - Zhuang Li
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, People's Republic of China
| | - Ning Gao
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, People's Republic of China
| | - Nian Liu
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, People's Republic of China
| | - Yuanrong Li
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, People's Republic of China
| | - Jing He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, People's Republic of China
| | - Qing Gao
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, People's Republic of China
| | - Zhijian Xie
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Clinical Research Center for Oral Diseases of Zhejiang Province, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Hangzhou, 310006, People's Republic of China.
| | - Jiachun Li
- School of Mechanical Engineering, Guizhou University, Guiyang, 550025, People's Republic of China.
| | - Yong He
- State Key Laboratory of Fluid Power and Mechatronic Systems, School of Mechanical Engineering, Zhejiang University, Hangzhou, 310027, People's Republic of China.
| |
Collapse
|
45
|
Bello SA, Cruz-Lebrón J, Rodríguez-Rivera OA, Nicolau E. Bioactive Scaffolds as a Promising Alternative for Enhancing Critical-Size Bone Defect Regeneration in the Craniomaxillofacial Region. ACS APPLIED BIO MATERIALS 2023; 6:4465-4503. [PMID: 37877225 DOI: 10.1021/acsabm.3c00432] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2023]
Abstract
Reconstruction of critical-size bone defects (CSDs) in the craniomaxillofacial (CMF) region remains challenging. Scaffold-based bone-engineered constructs have been proposed as an alternative to the classical treatments made with autografts and allografts. Scaffolds, a key component of engineered constructs, have been traditionally viewed as biologically passive temporary replacements of deficient bone lacking intrinsic cues to promote osteogenesis. Nowadays, scaffolds are functionalized, giving rise to bioactive scaffolds promoting bone regeneration more effectively than conventional counterparts. This review focuses on the three approaches most used to bioactivate scaffolds: (1) conferring microarchitectural designs or surface nanotopography; (2) loading bioactive molecules; and (3) seeding stem cells on scaffolds, providing relevant examples of in vivo (preclinical and clinical) studies where these methods are employed to enhance CSDs healing in the CMF region. From these, adding bioactive molecules (specifically bone morphogenetic proteins or BMPs) to scaffolds has been the most explored to bioactivate scaffolds. Nevertheless, the downsides of grafting BMP-loaded scaffolds in patients have limited its successful translation into clinics. Despite these drawbacks, scaffolds containing safer, cheaper, and more effective bioactive molecules, combined with stem cells and topographical cues, remain a promising alternative for clinical use to treat CSDs in the CMF complex replacing autografts and allografts.
Collapse
Affiliation(s)
- Samir A Bello
- Department of Chemistry, University of Puerto Rico, Rio Piedras Campus, PO Box 23346, San Juan, Puerto Rico 00931, United States
- Molecular Sciences Research Center, University of Puerto Rico, 1390 Ponce De León Ave, Suite 1-7, San Juan, Puerto Rico 00926, United States
| | - Junellie Cruz-Lebrón
- Department of Chemistry, University of Puerto Rico, Rio Piedras Campus, PO Box 23346, San Juan, Puerto Rico 00931, United States
- Molecular Sciences Research Center, University of Puerto Rico, 1390 Ponce De León Ave, Suite 1-7, San Juan, Puerto Rico 00926, United States
| | - Osvaldo A Rodríguez-Rivera
- Department of Chemistry, University of Puerto Rico, Rio Piedras Campus, PO Box 23346, San Juan, Puerto Rico 00931, United States
- Molecular Sciences Research Center, University of Puerto Rico, 1390 Ponce De León Ave, Suite 1-7, San Juan, Puerto Rico 00926, United States
| | - Eduardo Nicolau
- Department of Chemistry, University of Puerto Rico, Rio Piedras Campus, PO Box 23346, San Juan, Puerto Rico 00931, United States
- Molecular Sciences Research Center, University of Puerto Rico, 1390 Ponce De León Ave, Suite 1-7, San Juan, Puerto Rico 00926, United States
| |
Collapse
|
46
|
Tang J, Wang H, Wu D, Wang Z. LAMA5-inspired adhesive dodecapeptide facilitates efficient dentine regeneration: An in vitro and in vivo study. Int Endod J 2023; 56:1385-1398. [PMID: 37632694 DOI: 10.1111/iej.13967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2023] [Revised: 08/12/2023] [Accepted: 08/13/2023] [Indexed: 08/28/2023]
Abstract
AIM The primary goal of this study was to investigate the potential effects of A5G81 in inducing reparative dentine (RD) formation both in vitro and in vivo. METHODOLOGY Cell adhesion was observed by crystal violet staining and quantified by Sodium Dodecyl Sulphate (SDS) extraction. Cell proliferation was investigated using Cell Counting Kit-8 (CCK-8) assay. Spreading of cytoskeleton was visualized using immunofluorescence staining. Protein expression level of Akt signalling pathway was compared in a human Akt pathway phosphorylation array. Genes that were up or downregulated by A5G81 were identified by RNA sequencing. The mRNA expression of odontoblastic markers was detected by quantitative real-time polymerase chain reaction (qPCR). Moreover, mineralization of human dental pulp cells (hDPCs) was visualized by alizarin red staining and quantified using cetylpyridinium chloride (CPC). A direct pulp-capping model was established in SD rats and the RD formation at 2 weeks after operation was observed using HE staining. RESULTS A5G81 (optimal coating concentration: 0.5 mg/mL) promoted hDPCs adhesion and proliferation to a level that was similar to Type I collagen (COL-1). Meanwhile, A5G81 activated Akt signalling pathway, albeit to a lesser extent than COL-1. An inhibition test indicated that A5G81 induced hDPCs adhesion by activating PI3K pathway. A5G81 induced the expression of ECM remodelling genes and odontoblastic genes, which were demonstrated by RNA-seq and qPCR, respectively. In addition, A5G81 efficiently accelerated the mineralization of hDPCs in both immobilized and soluble forms, a property that makes it more applicable in dental clinic. Finally, the pulp-capping study in rats suggested that use of A5G81 could successfully induce the formation of RD within 2 weeks. CONCLUSION Coating of A5G81 to non-tissue culture-treated polystyrene facilitates spreading, proliferation and differentiation of hDPCs, resulting in rapid RD formation in artificially exposed pulp.
Collapse
Affiliation(s)
- Jia Tang
- School and Hospital of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Tongji University, Shanghai, China
| | - Haicheng Wang
- School and Hospital of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Tongji University, Shanghai, China
| | - Di Wu
- School and Hospital of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Tongji University, Shanghai, China
| | - Zuolin Wang
- School and Hospital of Stomatology, Shanghai Engineering Research Center of Tooth Restoration and Regeneration, Tongji University, Shanghai, China
| |
Collapse
|
47
|
Jurczak P, Lach S. Hydrogels as Scaffolds in Bone-Related Tissue Engineering and Regeneration. Macromol Biosci 2023; 23:e2300152. [PMID: 37276333 DOI: 10.1002/mabi.202300152] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 05/22/2023] [Indexed: 06/07/2023]
Abstract
Several years have passed since the medical and scientific communities leaned toward tissue engineering as the most promising field to aid bone diseases and defects resulting from degenerative conditions or trauma. Owing to their histocompatibility and non-immunogenicity, bone grafts, precisely autografts, have long been the gold standard in bone tissue therapies. However, due to issues associated with grafting, especially the surgical risks and soaring prices of the procedures, alternatives are being extensively sought and researched. Fibrous and non-fibrous materials, synthetic substitutes, or cell-based products are just a few examples of research directions explored as potential solutions. A very promising subgroup of these replacements involves hydrogels. Biomaterials resembling the bone extracellular matrix and therefore acting as 3D scaffolds, providing the appropriate mechanical support and basis for cell growth and tissue regeneration. Additional possibility of using various stimuli in the form of growth factors, cells, etc., within the hydrogel structure, extends their use as bioactive agent delivery platforms and acts in favor of their further directed development. The aim of this review is to bring the reader closer to the fascinating subject of hydrogel scaffolds and present the potential of these materials, applied in bone and cartilage tissue engineering and regeneration.
Collapse
Affiliation(s)
- Przemyslaw Jurczak
- Laboratory of Molecular and Cellular Nephrology, Mossakowski Medical Research Centre Polish Academy of Sciences, Gdansk, 80-308, Poland
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, Gdansk, 80-308, Poland
| | - Slawomir Lach
- Department of Biomedical Chemistry, Faculty of Chemistry, University of Gdansk, Wita Stwosza 63, Gdansk, 80-308, Poland
| |
Collapse
|
48
|
Zengin A, Teixeira FC, Feliciano T, Habibovic P, Mota CD, Baker MB, van Rijt S. Matrix metalloproteinase degradable, in situ photocrosslinked nanocomposite bioinks for bioprinting applications. BIOMATERIALS ADVANCES 2023; 154:213647. [PMID: 37839298 DOI: 10.1016/j.bioadv.2023.213647] [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: 02/16/2023] [Revised: 08/10/2023] [Accepted: 09/29/2023] [Indexed: 10/17/2023]
Abstract
The development of suitable bioinks with high printability, mechanical strength, biodegradability, and biocompatibility is a key challenge for the clinical translation of 3D constructs produced with bioprinting technologies. In this work, we developed a new type of nanocomposite bioinks containing thiolated mesoporous silica nanoparticles (MSN) that act as active fillers within norbornene-functionalized hydrogels. The MSNs could rapidly covalently crosslink the hydrogels upon exposure to UV light. The mechanical properties of the gels could be modulated from 9.3 to 19.7 kPa with increasing concentrations of MSN. The ability of the MSN to covalently crosslink polymeric networks was, however, significantly influenced by polymer architecture and the number of functional groups. Modification of the outer surface of MSNs with matrix metalloproteinase (MMP) sensitive peptides (MSN-MMPs) resulted in proteinase K and MMP-9 enzyme responsive biodegradable bioinks. Additional cysteine modified RGD peptide incorporation enhanced cell-matrix interactions and reduced the gelation time for bioprinting. The nanocomposite bioinks could be printed by using extrusion-based bioprinting. Our nanocomposite bioinks preserved their shape during in vitro studies and encapsulated MG63 cells preserved their viability and proliferated within the bioinks. As such, our nanocomposite bioinks are promising bioinks for creating bioprinted constructs with tunable mechanical and degradation properties.
Collapse
Affiliation(s)
- Aygul Zengin
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
| | - Filipa Castro Teixeira
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
| | - Tony Feliciano
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
| | - Pamela Habibovic
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
| | - Carlos Domingues Mota
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
| | - Matthew B Baker
- Department of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands
| | - Sabine van Rijt
- Department of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired Regenerative Medicine, Maastricht University, P.O. Box 616, 6200 MD Maastricht, the Netherlands.
| |
Collapse
|
49
|
Palladino S, Schwab A, Copes F, D'Este M, Candiani G, Mantovani D. Development of a hyaluronic acid-collagen bioink for shear-induced fibers and cells alignment. Biomed Mater 2023; 18:065017. [PMID: 37751763 DOI: 10.1088/1748-605x/acfd77] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2023] [Accepted: 09/26/2023] [Indexed: 09/28/2023]
Abstract
Human tissues are characterized by complex composition and cellular and extracellular matrix (ECM) organization at microscopic level. In most of human tissues, cells and ECM show an anisotropic arrangement, which confers them specific properties.In vitro, the ability to closely mimic this complexity is limited. However, in the last years, extrusion bioprinting showed a certain potential for aligning cells and biomolecules, due to the application of shear stress during the bio-fabrication process. In this work, we propose a strategy to combine collagen (col) with tyramine-modified hyaluronic acid (THA) to obtain a printable col-THA bioink for extrusion bioprinting, solely-based on natural-derived components. Collagen fibers formation within the hybrid hydrogel, as well as collagen distribution and spatial organization before and after printing, were studied. For the validation of the biological outcome, fibroblasts were selected as cellular model and embedded in the col-THA matrix. Cell metabolic activity and cell viability, as well as cell distribution and alignment, were studied in the bioink before and after bioprinting. Results demonstrated successful collagen fibers formation within the bioink, as well as collagen anisotropic alignment along the printing direction. Furthermore, results revealed suitable biological properties, with a slightly reduced metabolic activity at day 1, fully recovered within the first 3 d post-cell embedding. Finally, results showed fibroblasts elongation and alignment along the bioprinting direction. Altogether, results validated the potential to obtain collagen-based bioprinted constructs, with both cellular and ECM anisotropy, without detrimental effects of the fabrication process on the biological outcome. This bioink can be potentially used for a wide range of applications in tissue engineering and regenerative medicine in which anisotropy is required.
Collapse
Affiliation(s)
- Sara Palladino
- Laboratory for Biomaterials and Bioengineering, CRC-Tier I, Dept Min-Met-Materials Eng and Regenerative Medicine, CHU de Québec, Laval University, Quebec City, Canada
- genT_LΛB, Department of Chemistry, Materials and Chemical Engineering 'G. Natta', Politecnico di Milano, Milan, Italy
| | | | - Francesco Copes
- Laboratory for Biomaterials and Bioengineering, CRC-Tier I, Dept Min-Met-Materials Eng and Regenerative Medicine, CHU de Québec, Laval University, Quebec City, Canada
| | | | - Gabriele Candiani
- genT_LΛB, Department of Chemistry, Materials and Chemical Engineering 'G. Natta', Politecnico di Milano, Milan, Italy
| | - Diego Mantovani
- Laboratory for Biomaterials and Bioengineering, CRC-Tier I, Dept Min-Met-Materials Eng and Regenerative Medicine, CHU de Québec, Laval University, Quebec City, Canada
| |
Collapse
|
50
|
Gao Y, Zhang X, Zhou H. Biomimetic Hydrogel Applications and Challenges in Bone, Cartilage, and Nerve Repair. Pharmaceutics 2023; 15:2405. [PMID: 37896165 PMCID: PMC10609742 DOI: 10.3390/pharmaceutics15102405] [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: 09/04/2023] [Revised: 09/22/2023] [Accepted: 09/27/2023] [Indexed: 10/29/2023] Open
Abstract
Tissue engineering and regenerative medicine is a highly sought-after field for researchers aiming to compensate and repair defective tissues. However, the design and development of suitable scaffold materials with bioactivity for application in tissue repair and regeneration has been a great challenge. In recent years, biomimetic hydrogels have shown great possibilities for use in tissue engineering, where they can tune mechanical properties and biological properties through functional chemical modifications. Also, biomimetic hydrogels provide three-dimensional (3D) network spatial structures that can imitate normal tissue microenvironments and integrate cells, scaffolds, and bioactive substances for tissue repair and regeneration. Despite the growing interest in various hydrogels for biomedical use in previous decades, there are still many aspects of biomimetic hydrogels that need to be understood for biomedical and clinical trial applications. This review systematically describes the preparation of biomimetic hydrogels and their characteristics, and it details the use of biomimetic hydrogels in bone, cartilage, and nerve tissue repair. In addition, this review outlines the application of biomimetic hydrogels in bone, cartilage, and neural tissues regarding drug delivery. In particular, the advantages and shortcomings of biomimetic hydrogels in biomaterial tissue engineering are highlighted, and future research directions are proposed.
Collapse
Affiliation(s)
- Yanbing Gao
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China;
- Key Laboratory of Bone and Joint Disease Research of Gansu Province, Lanzhou 730030, China
| | - Xiaobo Zhang
- Department of Orthopedics, Honghui Hospital, Xi’an Jiaotong University, Xi’an 710000, China
| | - Haiyu Zhou
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou 730030, China;
- Key Laboratory of Bone and Joint Disease Research of Gansu Province, Lanzhou 730030, China
| |
Collapse
|