1
|
Lu HH, Froidevaux C, Biermann I, Kaňková H, Büchner M, Schubert DW, Salehi S, Boccaccini AR. Printable ADA-GEL-based composite inks containing Zn-doped bioactive inorganic fillers for skeletal muscle biofabrication. BIOMATERIALS ADVANCES 2025; 172:214233. [PMID: 40048902 DOI: 10.1016/j.bioadv.2025.214233] [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: 11/10/2024] [Revised: 01/27/2025] [Accepted: 02/13/2025] [Indexed: 03/17/2025]
Abstract
Bioprinting shows significant promise in advancing medical treatments by offering patient-specific solutions for repairing skeletal muscle tissues. Zinc (Zn) is one of the key ions in the human body involved in the development of myogenic cells. This study investigates the integration of Zn-doped bioactive inorganic fillers (BIFs) into alginate-dialdehyde-gelatin (ADA-GEL) as composite ink for bioprinting applications. BIFs were mesoporous calcium-silicate-based bioactive glass nanoparticles with nominal composition: 80% SiO2-X% CaO-Y% ZnO (X = 20, 18, 15, or 10; Y = 0, 2, 5, or 10; mol%). Ion release profiles confirmed that the addition of Zn ions prevented the burst release of Si and Ca ions. The incorporation of BIFs, particularly at higher dopant concentrations, significantly affected the hydrogel swelling and mechanical properties. With increasing concentration of Zn ions (to 5 mol%), the hydrogels exhibited greater dimensional swelling after 24 h of incubation in aqueous solutions, while all compositions lost weight over time after the initial swelling phase. Indirect cell studies demonstrated that 0.1 wt% BIFs extracts, obtained after 24 h-incubation in a cell culture medium, were cytocompatible with C2C12 cells. Furthermore, bioprinted C2C12 cells encapsulated in the bioinks showed a clear increase in cell number after seven days of culture. In particular, cells in the composite inks containing Zn-BIFs exhibited higher spreading, elongation, and alignment than those in pure ADA-GEL, indicating the biological activity provided by the Zn-BIF particles. This study introduces therefore an effective formulation of ADA-GEL-based composite bioinks enriched with Zn-containing nanoparticles for skeletal muscle tissue engineering applications.
Collapse
Affiliation(s)
- Hsuan-Heng Lu
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - Clara Froidevaux
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - Isabell Biermann
- Department for Functional Materials in Medicine and Dentistry, University of Würzburg, 97070 Würzburg, Germany
| | - Hana Kaňková
- FunGlass - Centre for Functional and Surface Functionalized Glass, Alexander Dubček University of Trenčín, 911 50 Trenčín, Slovakia
| | - Margitta Büchner
- Institute of Polymer Materials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - Dirk W Schubert
- Institute of Polymer Materials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany
| | - Sahar Salehi
- Department of Biomaterials, Faculty of Engineering Science, University of Bayreuth, 95447 Bayreuth, Germany
| | - Aldo R Boccaccini
- Institute of Biomaterials, Department of Materials Science and Engineering, University of Erlangen-Nuremberg, 91058 Erlangen, Germany.
| |
Collapse
|
2
|
Kim JT, Han SW, Youn DH, Jung H, Lee EH, Kang SM, Cho YJ, Jeon JP. Advanced hydrogel mesh platform with neural stem cells and human umbilical vein endothelial cells for enhanced axonal regeneration. APL Bioeng 2025; 9:026101. [PMID: 40181802 PMCID: PMC11964475 DOI: 10.1063/5.0244057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 03/15/2025] [Indexed: 04/05/2025] Open
Abstract
One of the major obstacles to neural recovery following intracerebral hemorrhage (ICH) is the cavity-like lesion that occurs at the site of the hemorrhage, which impedes axonal regeneration. Here, we aim to address this challenge by investigating the migratory mechanisms of neural stem cells (NSCs) within the cavity in vitro using a hydrogel and endothelial cells. Mouse NSCs (mNSCs) isolated from the subventricular and subgranular zones using the 3D hydrogel culture were evaluated for their neurogenic, extracellular matrix (ECM), and adhesion-related mRNA expression compared to microglia (BV2) and secretory factors of human umbilical vein endothelial cells (HUVECs) in vitro and in vivo conditions. A hydrogel mesh combining mNSCs and HUVECs was developed for its therapeutic potential. mNSCs exhibit high stemness, neurogenesis, and ECM remodeling capabilities. mNSCs demonstrated close interaction with HUVECs and the surrounding vascular structures in in vitro and in vivo studies. Furthermore, mNSCs could degrade high concentrations of fibrin to facilitate migration and adhesion. mNSCs and HUVECs formed mesh networks through cell-cell contacts and maintained the structure through Matrigel support, potentially ensuring sufficient survival and regeneration capabilities. Our proposed hydrogel mesh platform with mNSCs and HUVECs demonstrated successful maintenance of cell survival and provision of structural support for the delivered cells by promoting ECM remodeling and neurogenesis, which may aid in axonal regeneration in the cavity lesions following ICH.
Collapse
Affiliation(s)
- Jong-Tae Kim
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea
| | - Sung Woo Han
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea
| | - Dong Hyuk Youn
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea
| | - Harry Jung
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea
| | - Eun-Ho Lee
- Department of Green Chemical Engineering, Sangmyung University, Cheonan 31066, Republic of Korea
| | - Sung-Min Kang
- Department of Green Chemical Engineering, Sangmyung University, Cheonan 31066, Republic of Korea
| | - Yong-Jun Cho
- Department of Neurosurgery, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea
| | - Jin Pyeong Jeon
- Department of Neurosurgery, Hallym University College of Medicine, Chuncheon 24252, Republic of Korea
| |
Collapse
|
3
|
Contessi Negrini N, Pellegrinelli V, Salem V, Celiz A, Vidal-Puig A. Breaking barriers in obesity research: 3D models of dysfunctional adipose tissue. Trends Biotechnol 2025; 43:1079-1093. [PMID: 39443224 DOI: 10.1016/j.tibtech.2024.09.017] [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: 07/27/2024] [Revised: 09/23/2024] [Accepted: 09/25/2024] [Indexed: 10/25/2024]
Abstract
Obesity is a global health crisis characterised by excessive accumulation of adipose tissue (AT). Under obesogenic conditions, this metabolically active tissue undergoes fibrosis and inflammation, leading to obesity-linked comorbidities. Modelling AT is essential for understanding its pathophysiology and developing treatments to protect against metabolic complications. 3D in vitro AT models are promising tools that address the limitations of traditional 2D in vitro models and in vivo animal models, providing enhanced biomimetic and human-relevant platforms. 3D models facilitate the study of AT pathophysiology and therapeutic screening. This review discusses the crucial role of AT in obesity-linked comorbidities, its dynamicity and complexity, and recent advances in engineering 3D scaffold-based in vitro dysfunctional AT models, highlighting potential breakthroughs in metabolic research and beyond.
Collapse
Affiliation(s)
- Nicola Contessi Negrini
- Department of Bioengineering, Imperial College London, London, UK; The Francis Crick Institute, London, UK.
| | | | - Victoria Salem
- Department of Bioengineering, Imperial College London, London, UK
| | - Adam Celiz
- Department of Bioengineering, Imperial College London, London, UK; The Francis Crick Institute, London, UK
| | - Antonio Vidal-Puig
- MRC Institute of Metabolic Science and Medical Research Council, Cambridge, UK; Cambridge University Nanjing Centre of Technology and Innovation, Nanjing, PR China; Centro de Investigacion Principe Felipe (CIPF), Valencia, Spain; Cambridge Heart and Lung Research Institute, Cambridge, UK
| |
Collapse
|
4
|
Zhang M, Zhang B. Extracellular matrix stiffness: mechanisms in tumor progression and therapeutic potential in cancer. Exp Hematol Oncol 2025; 14:54. [PMID: 40211368 PMCID: PMC11984264 DOI: 10.1186/s40164-025-00647-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Accepted: 03/23/2025] [Indexed: 04/14/2025] Open
Abstract
Tumor microenvironment (TME) is a complex ecosystem composed of both cellular and non-cellular components that surround tumor tissue. The extracellular matrix (ECM) is a key component of the TME, performing multiple essential functions by providing mechanical support, shaping the TME, regulating metabolism and signaling, and modulating immune responses, all of which profoundly influence cell behavior. The quantity and cross-linking status of stromal components are primary determinants of tissue stiffness. During tumor development, ECM stiffness not only serves as a barrier to hinder drug delivery but also promotes cancer progression by inducing mechanical stimulation that activates cell membrane receptors and mechanical sensors. Thus, a comprehensive understanding of how ECM stiffness regulates tumor progression is crucial for identifying potential therapeutic targets for cancer. This review examines the effects of ECM stiffness on tumor progression, encompassing proliferation, migration, metastasis, drug resistance, angiogenesis, epithelial-mesenchymal transition (EMT), immune evasion, stemness, metabolic reprogramming, and genomic stability. Finally, we explore therapeutic strategies that target ECM stiffness and their implications for tumor progression.
Collapse
Affiliation(s)
- Meiling Zhang
- School of Basic Medicine, China Three Gorges University, 8 Daxue Road, Yichang, 443002, Hubei, China
- Central Laboratory, The First Affiliated Hospital of Jinan University, No. 613 Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China
| | - Bin Zhang
- School of Basic Medicine, China Three Gorges University, 8 Daxue Road, Yichang, 443002, Hubei, China.
- Central Laboratory, The First Affiliated Hospital of Jinan University, No. 613 Huangpu West Road, Tianhe District, Guangzhou, 510627, Guangdong, China.
| |
Collapse
|
5
|
Li G, Gao F, Yang D, Lin L, Yu W, Tang J, Yang R, Jin M, Gu Y, Wang P, Lu E. ECM-mimicking composite hydrogel for accelerated vascularized bone regeneration. Bioact Mater 2024; 42:241-256. [PMID: 39285909 PMCID: PMC11404060 DOI: 10.1016/j.bioactmat.2024.08.035] [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: 05/15/2024] [Revised: 08/07/2024] [Accepted: 08/27/2024] [Indexed: 09/19/2024] Open
Abstract
Bioactive hydrogel materials have great potential for applications in bone tissue engineering. However, fabrication of functional hydrogels that mimic the natural bone extracellular matrix (ECM) remains a challenge, because they need to provide mechanical support and embody physiological cues for angiogenesis and osteogenesis. Inspired by the features of ECM, we constructed a dual-component composite hydrogel comprising interpenetrating polymer networks of gelatin methacryloyl (GelMA) and deoxyribonucleic acid (DNA). Within the composite hydrogel, the GelMA network serves as the backbone for mechanical and biological stability, whereas the DNA network realizes dynamic capabilities (e.g., stress relaxation), thereby promoting cell proliferation and osteogenic differentiation. Furthermore, functional aptamers (Apt19S and AptV) are readily attached to the DNA network to recruit bone marrow mesenchymal stem cells (BMSCs) and achieve sustained release of loaded vascular endothelial growth factor towards angiogenesis. Our results showed that the composite hydrogel could facilitate the adhesion of BMSCs, promote osteogenic differentiation by activating focal adhesion kinase (FAK)/phosphatidylinositol 3-kinase (PI3K)/protein kinase B (Akt)/β-Catenin signaling pathway, and eventually enhance vascularized bone regeneration. This study shows that the multifunctional composite hydrogel of GelMA and DNA can successfully simulate the biological functions of natural bone ECM and has great potential for repairing bone defects.
Collapse
Affiliation(s)
- Guanglong Li
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Fei Gao
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Donglei Yang
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Lu Lin
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Weijun Yu
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Jiaqi Tang
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Ruhan Yang
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Min Jin
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Yuting Gu
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Pengfei Wang
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| | - Eryi Lu
- Department of Stomatology, Renji Hospital, Shanghai Jiao Tong University School of Medicine, 160 Pujian Road, Shanghai, 200127, China
| |
Collapse
|
6
|
Zhu H, Sun H, Dai J, Hao J, Zhou B. Chitosan-based hydrogels in cancer therapy: Drug and gene delivery, stimuli-responsive carriers, phototherapy and immunotherapy. Int J Biol Macromol 2024; 282:137047. [PMID: 39489261 DOI: 10.1016/j.ijbiomac.2024.137047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/18/2024] [Accepted: 10/28/2024] [Indexed: 11/05/2024]
Abstract
Nanotechnology has transformed the oncology sector by particularly targeting cancer cells and enhancing the efficacy of conventional therapies, not only improving efficacy of conventional therapeutics, but also reducing systemic toxicity. Environmentally friendly materials are the top choice for treating cancer. Chitosan, sourced from chitin, is widely used with its derivatives for the extensive synthesis or modification of nanostructures. Chitosan has been deployed to develop hydrogels, as 3D polymeric networks capable of water absorption with wide biomedical application. The chitosan hydrogels are biocompatible and biodegradable structures that can deliver drugs, genes or a combination of them in cancer therapy. Increased tumor ablation, reducing off-targeting feature and protection of genes against degradation are benefits of using chitosan hydrogels in cancer therapy. The efficacy of cancer immunotherapy can be improved by chitosan hydrogels to prevent emergence of immune evasion. In addition, chitosan hydrogels facilitate photothermal and photodynamic therapy for tumor suppression. Chitosan hydrogels can synergistically integrate chemotherapy, immunotherapy, and phototherapy in cancer treatment. Additionally, chitosan hydrogels that respond to stimuli, specifically thermo-sensitive hydrogels, have been developed for inhibiting tumors.
Collapse
Affiliation(s)
- Hailin Zhu
- Department of Pathology, Ganzhou Cancer Hospital, Ganzhou City, Jiangxi Province, China
| | - Hao Sun
- Faculty of Science, Autonomous University of Madrid, Spainish National Research Council-Consejo Superior de Investigaciones Científicas, (UAM-CSIC), 28049 Madrid, Spain
| | - Jingyuan Dai
- School of Computer Science and Information Systems, Northwest Missouri State University, MO, USA
| | - Junfeng Hao
- Guangdong Provincial Key Laboratory of Autophagy and Major Chronic Non-Communicable Diseases, Key Laboratory of Prevention and Management of Chronic Kidney Disease of Zhanjiang City, Institute of Nephrology, Affiliated Hospital of Guangdong Medical University, Zhanjiang 524001, Guangdong, China; Department of Family Medicine, Shengjing Hospital of China Medical University, Shenyang 110022, Liaoning, China.
| | - Boxuan Zhou
- Department of General Surgery, Breast Disease Center, the First Affiliated Hospital, Jiangxi Medical College, Nanchang University, Nanchang, China.
| |
Collapse
|
7
|
Neves SC, Sousa A, Nascimento DS, Orge ID, Ferreira SA, Mota C, Moroni L, Barrias CC, Granja PL. A hybrid construct with tailored 3D structure for directing pre-vascularization in engineered tissues. Mater Today Bio 2024; 29:101291. [PMID: 39435373 PMCID: PMC11492604 DOI: 10.1016/j.mtbio.2024.101291] [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: 06/09/2024] [Revised: 09/28/2024] [Accepted: 10/03/2024] [Indexed: 10/23/2024] Open
Abstract
Hybrid 3D constructs combining different structural components afford unique opportunities to engineer functional tissues. Creating functional microvascular networks within these constructs is crucial for promoting integration with host vessels and ensuring successful engraftment. Here, we present a hybrid 3D system in which poly (ethylene oxide terephthalate)/poly (butylene terephthalate) fibrous scaffolds are combined with pectin hydrogels to provide internal topography and guide the formation of microvascular beds. The sequence/method of seeding human endothelial cells (EC) and mesenchymal stromal cells (MSC) into the system had a significant impact on microvessel formation. Optimal results were obtained when EC were directly seeded onto the fibrous scaffold, followed by the addition of hydrogel-embedded MSC. This approach facilitated the development of highly oriented microvascular networks along the fibers. These networks were lumenized, supported by a basement membrane, and stabilized by pericyte-like cells, persisting for at least 28 days in vitro. Furthermore, culture under pro-angiogenic and osteoinductive conditions induced MSC osteogenic differentiation without impairing microvessel formation. Upon subcutaneous implantation in mice, the pre-vascularized constructs were infiltrated by host vessels, and human microvessels were still present after 2 weeks. Overall, the proposed hybrid 3D system, combined with an optimized cell-seeding protocol, offers an effective approach for directing the formation of robust and geometrically oriented microvessels, making it promising for tissue engineering applications.
Collapse
Affiliation(s)
- Sara C. Neves
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade Do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- FEUP - Faculdade de Engenharia da Universidade Do Porto, Departamento de Engenharia Metalúrgica e de Materiais, Rua Dr. Roberto Frias, s/n, 4200-465, Porto, Portugal
- MERLN - Institute for Technology-Inspired Regenerative Medicine, Department of Complex Tissue Regeneration, Maastricht University, Universiteitssingel 40, 6229ER Maastricht, the Netherlands
| | - Aureliana Sousa
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade Do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - Diana S. Nascimento
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade Do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade Do Porto, R. de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Iasmim D. Orge
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade Do Porto, R. de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Sílvia A. Ferreira
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| | - Carlos Mota
- MERLN - Institute for Technology-Inspired Regenerative Medicine, Department of Complex Tissue Regeneration, Maastricht University, Universiteitssingel 40, 6229ER Maastricht, the Netherlands
| | - Lorenzo Moroni
- MERLN - Institute for Technology-Inspired Regenerative Medicine, Department of Complex Tissue Regeneration, Maastricht University, Universiteitssingel 40, 6229ER Maastricht, the Netherlands
| | - Cristina C. Barrias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade Do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- ICBAS - Instituto de Ciências Biomédicas Abel Salazar, Universidade Do Porto, R. de Jorge Viterbo Ferreira, 228, 4050-313, Porto, Portugal
| | - Pedro L. Granja
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade Do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
- INEB - Instituto de Engenharia Biomédica, Universidade Do Porto, Rua Alfredo Allen, 208, 4200-135, Porto, Portugal
| |
Collapse
|
8
|
Semitela A, Marques PAAP, Completo A. Strategies to engineer articular cartilage with biomimetic zonal features: a review. Biomater Sci 2024; 12:5961-6005. [PMID: 39463257 DOI: 10.1039/d4bm00579a] [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: 10/29/2024]
Abstract
Articular cartilage (AC) is a highly specialized tissue with restricted ability for self-regeneration, given its avascular and acellular nature. Although a considerable number of surgical treatments is available for the repair, reconstruction, and regeneration of AC defects, most of them do not prioritize the development of engineered cartilage with zonal stratification derived from biomimetic biochemical, biomechanical and topographic cues. In the absence of these zonal elements, engineered cartilage will exhibit increased susceptibility to failure and will neither be able to withstand the mechanical loading to which AC is subjected nor will it integrate well with the surrounding tissue. In this regard, new breakthroughs in the development of hierarchical stratified engineered cartilage are highly sought after. Initially, this review provides a comprehensive analysis of the composition and zonal organization of AC, aiming to enhance our understanding of the significance of the structure of AC for its function. Next, we direct our attention towards the existing in vitro and in vivo studies that introduce zonal elements in engineered cartilage to elicit appropriate AC regeneration by employing tissue engineering strategies. Finally, the advantages, challenges, and future perspectives of these approaches are presented.
Collapse
Affiliation(s)
- Angela Semitela
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - Paula A A P Marques
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal.
| | - António Completo
- Centre of Mechanical Technology and Automation (TEMA), Department of Mechanical Engineering, University of Aveiro, 3810-193 Aveiro, Portugal.
| |
Collapse
|
9
|
Mamidi N, De Silva FF, Vacas AB, Gutiérrez Gómez JA, Montes Goo NY, Mendoza DR, Reis RL, Kundu SC. Multifaceted Hydrogel Scaffolds: Bridging the Gap between Biomedical Needs and Environmental Sustainability. Adv Healthc Mater 2024; 13:e2401195. [PMID: 38824416 DOI: 10.1002/adhm.202401195] [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: 03/30/2024] [Revised: 05/29/2024] [Indexed: 06/03/2024]
Abstract
Hydrogels are dynamically evolving 3D networks composed of hydrophilic polymer scaffolds with significant applications in the healthcare and environmental sectors. Notably, protein-based hydrogels mimic the extracellular matrix, promoting cell adhesion. Further enhancing cell proliferation within these scaffolds are matrix-metalloproteinase-triggered amino acid motifs. Integration of cell-friendly modules like peptides and proteins expands hydrogel functionality. These exceptional properties position hydrogels for diverse applications, including biomedicine, biosensors, environmental remediation, and the food industry. Despite significant progress, there is ongoing research to optimize hydrogels for biomedical and environmental applications further. Engineering novel hydrogels with favorable characteristics is crucial for regulating tissue architecture and facilitating ecological remediation. This review explores the synthesis, physicochemical properties, and biological implications of various hydrogel types and their extensive applications in biomedicine and environmental sectors. It elaborates on their potential applications, bridging the gap between advancements in the healthcare sector and solutions for environmental issues.
Collapse
Affiliation(s)
- Narsimha Mamidi
- Wisconsin Center for NanoBioSystems, School of Pharmacy, University of Wisconsin-Madison, Madison, WI, 53705, USA
- Department of Chemistry and Nanotechnology, The School of Engineering and Science, Tecnologico de Monterrey, Nuevo Leon, Monterrey, 64849, Mexico
| | - Fátima Franco De Silva
- Department of Chemistry and Nanotechnology, The School of Engineering and Science, Tecnologico de Monterrey, Nuevo Leon, Monterrey, 64849, Mexico
| | - Alejandro Bedón Vacas
- Department of Chemistry and Nanotechnology, The School of Engineering and Science, Tecnologico de Monterrey, Nuevo Leon, Monterrey, 64849, Mexico
| | - Javier Adonay Gutiérrez Gómez
- Department of Chemistry and Nanotechnology, The School of Engineering and Science, Tecnologico de Monterrey, Nuevo Leon, Monterrey, 64849, Mexico
| | - Naomi Yael Montes Goo
- Department of Chemistry and Nanotechnology, The School of Engineering and Science, Tecnologico de Monterrey, Nuevo Leon, Monterrey, 64849, Mexico
| | - Daniela Ruiz Mendoza
- Department of Chemistry and Nanotechnology, The School of Engineering and Science, Tecnologico de Monterrey, Nuevo Leon, Monterrey, 64849, Mexico
| | - Rui L Reis
- 3Bs Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| | - Subhas C Kundu
- 3Bs Research Group, I3Bs-Research Institute on Biomaterials, Biodegradables and Biomimetics, Headquarters of the European Institute of Excellence on Tissue Engineering and Regenerative Medicine, University of Minho, Barco, Guimarães, 4805-017, Portugal
- ICVS/3B's-PT Government Associate Laboratory, Braga, Guimarães, Portugal
| |
Collapse
|
10
|
Lu P, Ruan D, Huang M, Tian M, Zhu K, Gan Z, Xiao Z. Harnessing the potential of hydrogels for advanced therapeutic applications: current achievements and future directions. Signal Transduct Target Ther 2024; 9:166. [PMID: 38945949 PMCID: PMC11214942 DOI: 10.1038/s41392-024-01852-x] [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: 10/19/2023] [Revised: 04/02/2024] [Accepted: 04/28/2024] [Indexed: 07/02/2024] Open
Abstract
The applications of hydrogels have expanded significantly due to their versatile, highly tunable properties and breakthroughs in biomaterial technologies. In this review, we cover the major achievements and the potential of hydrogels in therapeutic applications, focusing primarily on two areas: emerging cell-based therapies and promising non-cell therapeutic modalities. Within the context of cell therapy, we discuss the capacity of hydrogels to overcome the existing translational challenges faced by mainstream cell therapy paradigms, provide a detailed discussion on the advantages and principal design considerations of hydrogels for boosting the efficacy of cell therapy, as well as list specific examples of their applications in different disease scenarios. We then explore the potential of hydrogels in drug delivery, physical intervention therapies, and other non-cell therapeutic areas (e.g., bioadhesives, artificial tissues, and biosensors), emphasizing their utility beyond mere delivery vehicles. Additionally, we complement our discussion on the latest progress and challenges in the clinical application of hydrogels and outline future research directions, particularly in terms of integration with advanced biomanufacturing technologies. This review aims to present a comprehensive view and critical insights into the design and selection of hydrogels for both cell therapy and non-cell therapies, tailored to meet the therapeutic requirements of diverse diseases and situations.
Collapse
Affiliation(s)
- Peilin Lu
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, PR China
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Dongxue Ruan
- State Key Laboratory of Respiratory Disease, National Clinical Research Center for Respiratory Disease, National Center for Respiratory Medicine, Department of Respiratory and Critical Care Medicine, Guangzhou Institute for Respiratory Health, The First Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510120, PR China
| | - Meiqi Huang
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China
| | - Mi Tian
- Department of Stomatology, Chengdu Second People's Hospital, Chengdu, 610021, PR China
| | - Kangshun Zhu
- Department of Minimally Invasive Interventional Radiology, and Laboratory of Interventional Radiology, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510260, PR China.
| | - Ziqi Gan
- Hospital of Stomatology, Guangdong Provincial Key Laboratory of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China.
| | - Zecong Xiao
- Nanomedicine Research Center, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, 510630, PR China.
| |
Collapse
|
11
|
Pasini C, Re F, Trenta F, Russo D, Sartore L. Gelatin-Based Scaffolds with Carrageenan and Chitosan for Soft Tissue Regeneration. Gels 2024; 10:426. [PMID: 39057449 PMCID: PMC11276450 DOI: 10.3390/gels10070426] [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: 06/04/2024] [Revised: 06/21/2024] [Accepted: 06/26/2024] [Indexed: 07/28/2024] Open
Abstract
Motivated by the enormous potential of hydrogels in regenerative medicine, new biocompatible gelatin-based hybrid hydrogels were developed through a green process using poly(ethylene glycol) diglycidyl ether as a cross-linking agent, adding carrageenan and chitosan polysaccharides to the network to better mimic the hybrid composition of native extracellular matrix. Overall, the hydrogels show suitable structural stability, high porosity and pore interconnectivity, good swellability, and finally, biocompatibility. Their mechanical behavior, investigated by tensile and compression tests, appears to be characterized by nonlinear elasticity with high compliance values, fast stress-relaxation, and good strain reversibility with no sign of mechanical failure for compressive loading-unloading cycles at relatively high deformation levels of 50%. Degradation tests confirm the hydrogel bioresorbability by gradual hydrolysis, during which the structural integrity of both materials is maintained, while their mechanical behavior becomes more and more compliant. Human Umbilical Cord-derived Mesenchymal Stem Cells (hUC-MSCs) were used to test the hydrogels as potential carriers for cell delivery in tissue engineering. hUC-MSCs cultured inside the hydrogels show a homogenous distribution and maintain their growth and viability for at least 21 days of culture, with an increasing proliferation trend. Hence, this study contributes to a further understanding of the potential use of hybrid hydrogels and hUC-MSCs for a wide range of biomedical applications, particularly in soft tissue engineering.
Collapse
Affiliation(s)
- Chiara Pasini
- Department of Mechanical and Industrial Engineering, University of Brescia, 25123 Brescia, Italy;
| | - Federica Re
- Blood Diseases and Cell Therapies Unit, Bone Marrow Transplant Unit, “ASST-Spedali Civili” Hospital of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (F.R.); (F.T.); (D.R.)
- Centro di Ricerca Emato-Oncologico AIL (CREA), “ASST-Spedali Civili” Hospital, 25123 Brescia, Italy
| | - Federica Trenta
- Blood Diseases and Cell Therapies Unit, Bone Marrow Transplant Unit, “ASST-Spedali Civili” Hospital of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (F.R.); (F.T.); (D.R.)
- Centro di Ricerca Emato-Oncologico AIL (CREA), “ASST-Spedali Civili” Hospital, 25123 Brescia, Italy
| | - Domenico Russo
- Blood Diseases and Cell Therapies Unit, Bone Marrow Transplant Unit, “ASST-Spedali Civili” Hospital of Brescia, Department of Clinical and Experimental Sciences, University of Brescia, 25123 Brescia, Italy; (F.R.); (F.T.); (D.R.)
| | - Luciana Sartore
- Department of Mechanical and Industrial Engineering, University of Brescia, 25123 Brescia, Italy;
| |
Collapse
|
12
|
Wu S, Gai T, Chen J, Chen X, Chen W. Smart responsive in situ hydrogel systems applied in bone tissue engineering. Front Bioeng Biotechnol 2024; 12:1389733. [PMID: 38863497 PMCID: PMC11165218 DOI: 10.3389/fbioe.2024.1389733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2024] [Accepted: 04/15/2024] [Indexed: 06/13/2024] Open
Abstract
The repair of irregular bone tissue suffers severe clinical problems due to the scarcity of an appropriate therapeutic carrier that can match dynamic and complex bone damage. Fortunately, stimuli-responsive in situ hydrogel systems that are triggered by a special microenvironment could be an ideal method of regenerating bone tissue because of the injectability, in situ gelatin, and spatiotemporally tunable drug release. Herein, we introduce the two main stimulus-response approaches, exogenous and endogenous, to forming in situ hydrogels in bone tissue engineering. First, we summarize specific and distinct responses to an extensive range of external stimuli (e.g., ultraviolet, near-infrared, ultrasound, etc.) to form in situ hydrogels created from biocompatible materials modified by various functional groups or hybrid functional nanoparticles. Furthermore, "smart" hydrogels, which respond to endogenous physiological or environmental stimuli (e.g., temperature, pH, enzyme, etc.), can achieve in situ gelation by one injection in vivo without additional intervention. Moreover, the mild chemistry response-mediated in situ hydrogel systems also offer fascinating prospects in bone tissue engineering, such as a Diels-Alder, Michael addition, thiol-Michael addition, and Schiff reactions, etc. The recent developments and challenges of various smart in situ hydrogels and their application to drug administration and bone tissue engineering are discussed in this review. It is anticipated that advanced strategies and innovative ideas of in situ hydrogels will be exploited in the clinical field and increase the quality of life for patients with bone damage.
Collapse
Affiliation(s)
- Shunli Wu
- College of Marine Life Sciences, Ocean University of China, Qingdao, China
- Hangzhou Singclean Medical Products Co., Ltd, Hangzhou, China
| | - Tingting Gai
- School of Medicine, Shanghai University, Shanghai, China
| | - Jie Chen
- Jiaxing Vocational Technical College, Department of Student Affairs, Jiaxing, China
| | - Xiguang Chen
- College of Marine Life Science, Ocean University of China, Qingdao, China
- Laoshan Laboratory, Qingdao, China
| | - Weikai Chen
- Department of Orthopedics, The Second Affiliated Hospital and Yuying Children’s Hospital of Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
13
|
Asl SK, Rahimzadegan M, Asl AK. Progress in cardiac tissue engineering and regeneration: Implications of gelatin-based hybrid scaffolds. Int J Biol Macromol 2024; 261:129924. [PMID: 38311143 DOI: 10.1016/j.ijbiomac.2024.129924] [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: 11/08/2023] [Revised: 01/06/2024] [Accepted: 01/31/2024] [Indexed: 02/09/2024]
Abstract
Cardiovascular diseases, particularly myocardial infarction (MI), remain a leading cause of morbidity and mortality worldwide. Current treatments for MI, more palliative than curative, have limitations in reversing the disease completely. Tissue engineering (TE) has emerged as a promising strategy to address this challenge and may lead to improved therapeutic approaches for MI. Gelatin-based scaffolds, including gelatin and its derivative, gelatin methacrylate (GelMA), have attracted significant attention in cardiac tissue engineering (CTE) due to their optimal physical and biochemical properties and capacity to mimic the native extracellular matrix (ECM). CTE mainly recruits two classes of gelatin/GelMA-based scaffolds: hydrogels and nanofibrous. This article reviews state-of-the-art gelatin/GelMA-based hybrid scaffolds currently applied for CTE and regenerative therapy. Hybrid scaffolds, fabricated by combining gelatin/GelMA hydrogel or nanofibrous scaffolds with other materials such as natural/synthetic polymers, nanoparticles, protein-based biomaterials, etc., are explored for enhanced cardiac tissue regeneration functionality. The engraftment of stem/cardiac cells, bioactive molecules, or drugs into these hybrid systems shows great promise in cardiac tissue repair and regeneration. Finally, the role of gelatin/GelMA scaffolds combined with the 3D bioprinting strategy in CTE will also be briefly highlighted.
Collapse
Affiliation(s)
- Siamak Kazemi Asl
- Deputy of Education, Ministry of Health and Medical Education, Tehran, Iran.
| | - Milad Rahimzadegan
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Kazemi Asl
- Functional Neurosurgery Research Center, Shohada Tajrish Comprehensive Neurosurgical Center of Excellence, Shahid Beheshti University of Medical Sciences, Tehran, Iran
| |
Collapse
|
14
|
Gao H, Liu S, Qin S, Yang J, Yue T, Ye B, Tang Y, Feng J, Hou J, Danzeng D. Injectable hydrogel-based combination therapy for myocardial infarction: a systematic review and Meta-analysis of preclinical trials. BMC Cardiovasc Disord 2024; 24:119. [PMID: 38383333 PMCID: PMC10882925 DOI: 10.1186/s12872-024-03742-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2023] [Accepted: 01/19/2024] [Indexed: 02/23/2024] Open
Abstract
INTRODUCTION This study evaluates the effectiveness of a combined regimen involving injectable hydrogels for the treatment of experimental myocardial infarction. PATIENT CONCERNS Myocardial infarction is an acute illness that negatively affects quality of life and increases mortality rates. Experimental models of myocardial infarction can aid in disease research by allowing for the development of therapies that effectively manage disease progression and promote tissue repair. DIAGNOSIS Experimental animal models of myocardial infarction were established using the ligation method on the anterior descending branch of the left coronary artery (LAD). INTERVENTIONS The efficacy of intracardiac injection of hydrogels, combined with cells, drugs, cytokines, extracellular vesicles, or nucleic acid therapies, was evaluated to assess the functional and morphological improvements in the post-infarction heart achieved through the combined hydrogel regimen. OUTCOMES A literature review was conducted using PubMed, Web of Science, Scopus, and Cochrane databases. A total of 83 papers, including studies on 1332 experimental animals (rats, mice, rabbits, sheep, and pigs), were included in the meta-analysis based on the inclusion and exclusion criteria. The overall effect size observed in the group receiving combined hydrogel therapy, compared to the group receiving hydrogel treatment alone, resulted in an ejection fraction (EF) improvement of 8.87% [95% confidence interval (CI): 7.53, 10.21] and a fractional shortening (FS) improvement of 6.31% [95% CI: 5.94, 6.67] in rat models, while in mice models, the improvements were 16.45% [95% CI: 11.29, 21.61] for EF and 5.68% [95% CI: 5.15, 6.22] for FS. The most significant improvements in EF (rats: MD = 9.63% [95% CI: 4.02, 15.23]; mice: MD = 23.93% [95% CI: 17.52, 30.84]) and FS (rats: MD = 8.55% [95% CI: 2.54, 14.56]; mice: MD = 5.68% [95% CI: 5.15, 6.22]) were observed when extracellular vesicle therapy was used. Although there have been significant results in large animal experiments, the number of studies conducted in this area is limited. CONCLUSION The present study demonstrates that combining hydrogel with other therapies effectively improves heart function and morphology. Further preclinical research using large animal models is necessary for additional study and validation.
Collapse
Affiliation(s)
- Han Gao
- School of Medicine, Tibet University, Lhasa, Tibet, China
| | - Song Liu
- School of Medicine, Tibet University, Lhasa, Tibet, China
| | - Shanshan Qin
- School of Medicine, Tibet University, Lhasa, Tibet, China
| | - Jiali Yang
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Tian Yue
- School of Life Science and Engineering, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Bengui Ye
- West China School of Pharmacy, Sichuan University, Chengdu, Sichuan, China
| | - Yue Tang
- School of Pharmacy, North Sichuan Medical College, Nanchong, Sichuan, China
| | - Jie Feng
- School of Medicine, Southwest Jiaotong University, Chengdu, Sichuan, China
| | - Jun Hou
- Department of Cardiology, Chengdu Third People's Hospital, Chengdu, Sichuan, China.
| | - Dunzhu Danzeng
- School of Medicine, Tibet University, Lhasa, Tibet, China.
| |
Collapse
|
15
|
Pasini C, Pandini S, Ramorino G, Sartore L. Tailoring the properties of composite scaffolds with a 3D-Printed lattice core and a bioactive hydrogel shell for tissue engineering. J Mech Behav Biomed Mater 2024; 150:106305. [PMID: 38096608 DOI: 10.1016/j.jmbbm.2023.106305] [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: 03/01/2023] [Revised: 10/04/2023] [Accepted: 12/02/2023] [Indexed: 01/09/2024]
Abstract
The optimal performance of scaffolds for tissue engineering relies on a proper combination of their constituent biomaterials and on the design of their structure. In this work, composite scaffolds with a core-shell architecture are realized by grafting a gelatin-chitosan hydrogel onto a 3D-printed polylactic acid (PLA) core, aiming in particular at bone regeneration. This hydrogel was recently found to sustain osteogenic differentiation of mesenchymal stromal cells, leading to new bone tissue formation. Here, the integration with rigid PLA lattice structures provides improved mechanical support and finer control of strength and stiffness. The core is prepared by fused deposition modeling with the specific aim to study several lattice structures and thereby better tune the scaffold mechanical properties. In fact, the core architecture dictates the scaffold strength and stiffness, which are seen to match those of different types of bone tissue. For all lattice types, the hydrogel is found to penetrate throughout the entire core and to present highly interconnected pores for cell colonization. By varying the void volume fraction in the core it is possible to significantly change the bioactive shell content, as well as the mechanical properties, over a wide range of values. Looking for design guidelines, relationships between stiffness/strength and density are here outlined for scaffolds featuring different lattice parameters. Moreover, by acting on the core strut arrangement, scaffolds are reinforced along specific directions, as evaluated under compressive and bending loading conditions.
Collapse
Affiliation(s)
- C Pasini
- Department of Mechanical and Industrial Engineering, University of Brescia, Via Branze, 38, 25123, Brescia, Italy
| | - S Pandini
- Department of Mechanical and Industrial Engineering, University of Brescia, Via Branze, 38, 25123, Brescia, Italy.
| | - G Ramorino
- Department of Mechanical and Industrial Engineering, University of Brescia, Via Branze, 38, 25123, Brescia, Italy
| | - L Sartore
- Department of Mechanical and Industrial Engineering, University of Brescia, Via Branze, 38, 25123, Brescia, Italy
| |
Collapse
|
16
|
Mohanty S, Swarup J, Priya S, Jain R, Singhvi G. Exploring the potential of polysaccharide-based hybrid hydrogel systems for their biomedical and therapeutic applications: A review. Int J Biol Macromol 2024; 256:128348. [PMID: 38007021 DOI: 10.1016/j.ijbiomac.2023.128348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2023] [Revised: 11/18/2023] [Accepted: 11/20/2023] [Indexed: 11/27/2023]
Abstract
Hydrogels are a versatile category of biomaterials that have been widely applied in the fields of biomedicine for the last several decades. The three-dimensional polymeric crosslinked hydrophilic structures of the hydrogel can proficiently hold drugs, nanoparticles, and cells, making them a potential delivery system. However, disadvantages like low mechanical strength, poor biocompatibility, and unusual in-vivo biodegradation are associated with conventional hydrogels. To overcome these hurdles, hybrid hydrogels are designed using two or more structurally different polymeric units. Polysaccharides, characterized by their innate biocompatibility, biodegradability, and abundance, establish an ideal foundation for the development of these hybrid hydrogels. This review aims to discuss the studies that have utilized naturally occurring polysaccharides to prepare hybrid systems, which were aimed for various biomedical applications such as tissue engineering, bone and cartilage regeneration, wound healing, skin cancer treatment, antimicrobial therapy, osteoarthritis treatment, and drug delivery. Furthermore, this review extensively examines the properties of the employed polysaccharides within hydrogel matrices, emphasizing the advantageous characteristics that make them a preferred choice. Furthermore, the challenges associated with the commercial implementation of these systems are explored alongside an assessment of the current patent landscape.
Collapse
Affiliation(s)
- Shambo Mohanty
- Industrial Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science (BITS) - Pilani, Pilani Campus, Rajasthan 333031, India
| | - Jayanti Swarup
- Industrial Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science (BITS) - Pilani, Pilani Campus, Rajasthan 333031, India
| | - Sakshi Priya
- Industrial Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science (BITS) - Pilani, Pilani Campus, Rajasthan 333031, India
| | - Rupesh Jain
- Industrial Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science (BITS) - Pilani, Pilani Campus, Rajasthan 333031, India
| | - Gautam Singhvi
- Industrial Research Laboratory, Department of Pharmacy, Birla Institute of Technology and Science (BITS) - Pilani, Pilani Campus, Rajasthan 333031, India.
| |
Collapse
|
17
|
Yang Y, Yang Y, Hou Z, Wang T, Wu P, Shen L, Li P, Zhang K, Yang L, Sun S. Comprehensive review of materials, applications, and future innovations in biodegradable esophageal stents. Front Bioeng Biotechnol 2023; 11:1327517. [PMID: 38125305 PMCID: PMC10731276 DOI: 10.3389/fbioe.2023.1327517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Accepted: 11/27/2023] [Indexed: 12/23/2023] Open
Abstract
Esophageal stricture (ES) results from benign and malignant conditions, such as uncontrolled gastroesophageal reflux disease (GERD) and esophageal neoplasms. Upper gastrointestinal endoscopy is the preferred diagnostic approach for ES and its underlying causes. Stent insertion using an endoscope is a prevalent method for alleviating or treating ES. Nevertheless, the widely used self-expandable metal stents (SEMS) and self-expandable plastic stents (SEPS) can result in complications such as migration and restenosis. Furthermore, they necessitate secondary extraction in cases of benign esophageal stricture (BES), rendering them unsatisfactory for clinical requirements. Over the past 3 decades, significant attention has been devoted to biodegradable materials, including synthetic polyester polymers and magnesium-based alloys, owing to their exceptional biocompatibility and biodegradability while addressing the challenges associated with recurring procedures after BES resolves. Novel esophageal stents have been developed and are undergoing experimental and clinical trials. Drug-eluting stents (DES) with drug-loading and drug-releasing capabilities are currently a research focal point, offering more efficient and precise ES treatments. Functional innovations have been investigated to optimize stent performance, including unidirectional drug-release and anti-migration features. Emerging manufacturing technologies such as three-dimensional (3D) printing and new biodegradable materials such as hydrogels have also contributed to the innovation of esophageal stents. The ultimate objective of the research and development of these materials is their clinical application in the treatment of ES and other benign conditions and the palliative treatment of malignant esophageal stricture (MES). This review aimed to offer a comprehensive overview of current biodegradable esophageal stent materials and their applications, highlight current research limitations and innovations, and offer insights into future development priorities and directions.
Collapse
Affiliation(s)
- Yaochen Yang
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang, China
- Research Center for Biomedical Materials, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang, China
| | - Yuanyuan Yang
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang, China
| | - Zhipeng Hou
- Research Center for Biomedical Materials, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang, China
| | - Tingting Wang
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang, China
| | - Peng Wu
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang, China
| | - Lufan Shen
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang, China
| | - Peng Li
- Liaoning Research Institute for Eugenic Birth and Fertility, China Medical University, Shenyang, China
| | - Kai Zhang
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang, China
| | - Liqun Yang
- Research Center for Biomedical Materials, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang, China
- Liaoning Research Institute for Eugenic Birth and Fertility, China Medical University, Shenyang, China
| | - Siyu Sun
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang, China
| |
Collapse
|
18
|
Pereira I, Lopez-Martinez MJ, Samitier J. Advances in current in vitro models on neurodegenerative diseases. Front Bioeng Biotechnol 2023; 11:1260397. [PMID: 38026882 PMCID: PMC10658011 DOI: 10.3389/fbioe.2023.1260397] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2023] [Accepted: 10/23/2023] [Indexed: 12/01/2023] Open
Abstract
Many neurodegenerative diseases are identified but their causes and cure are far from being well-known. The problem resides in the complexity of the neural tissue and its location which hinders its easy evaluation. Although necessary in the drug discovery process, in vivo animal models need to be reduced and show relevant differences with the human tissues that guide scientists to inquire about other possible options which lead to in vitro models being explored. From organoids to organ-on-a-chips, 3D models are considered the cutting-edge technology in cell culture. Cell choice is a big parameter to take into consideration when planning an in vitro model and cells capable of mimicking both healthy and diseased tissue, such as induced pluripotent stem cells (iPSC), are recognized as good candidates. Hence, we present a critical review of the latest models used to study neurodegenerative disease, how these models have evolved introducing microfluidics platforms, 3D cell cultures, and the use of induced pluripotent cells to better mimic the neural tissue environment in pathological conditions.
Collapse
Affiliation(s)
- Inês Pereira
- Nanobioengineering Group, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Maria J. Lopez-Martinez
- Nanobioengineering Group, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro Investigación Biomédica en Red: Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Barcelona, Spain
| | - Josep Samitier
- Nanobioengineering Group, Institute for Bioengineering of Catalonia, Barcelona Institute of Science and Technology, Barcelona, Spain
- Centro Investigación Biomédica en Red: Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Instituto de Salud Carlos III (ISCIII), Madrid, Spain
- Department of Electronics and Biomedical Engineering, University of Barcelona, Barcelona, Spain
| |
Collapse
|
19
|
Azizi P, Drobek C, Budday S, Seitz H. Simulating the mechanical stimulation of cells on a porous hydrogel scaffold using an FSI model to predict cell differentiation. Front Bioeng Biotechnol 2023; 11:1249867. [PMID: 37799813 PMCID: PMC10549991 DOI: 10.3389/fbioe.2023.1249867] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Accepted: 09/07/2023] [Indexed: 10/07/2023] Open
Abstract
3D-structured hydrogel scaffolds are frequently used in tissue engineering applications as they can provide a supportive and biocompatible environment for the growth and regeneration of new tissue. Hydrogel scaffolds seeded with human mesenchymal stem cells (MSCs) can be mechanically stimulated in bioreactors to promote the formation of cartilage or bone tissue. Although in vitro and in vivo experiments are necessary to understand the biological response of cells and tissues to mechanical stimulation, in silico methods are cost-effective and powerful approaches that can support these experimental investigations. In this study, we simulated the fluid-structure interaction (FSI) to predict cell differentiation on the entire surface of a 3D-structured hydrogel scaffold seeded with cells due to dynamic compressive load stimulation. The computational FSI model made it possible to simultaneously investigate the influence of both mechanical deformation and flow of the culture medium on the cells on the scaffold surface during stimulation. The transient one-way FSI model thus opens up significantly more possibilities for predicting cell differentiation in mechanically stimulated scaffolds than previous static microscale computational approaches used in mechanobiology. In a first parameter study, the impact of the amplitude of a sinusoidal compression ranging from 1% to 10% on the phenotype of cells seeded on a porous hydrogel scaffold was analyzed. The simulation results show that the number of cells differentiating into bone tissue gradually decreases with increasing compression amplitude, while differentiation into cartilage cells initially multiplied with increasing compression amplitude in the range of 2% up to 7% and then decreased. Fibrous cell differentiation was predicted from a compression of 5% and increased moderately up to a compression of 10%. At high compression amplitudes of 9% and 10%, negligible areas on the scaffold surface experienced high stimuli where no cell differentiation could occur. In summary, this study shows that simulation of the FSI system is a versatile approach in computational mechanobiology that can be used to study the effects of, for example, different scaffold designs and stimulation parameters on cell differentiation in mechanically stimulated 3D-structured scaffolds.
Collapse
Affiliation(s)
- Pedram Azizi
- Chair of Microfluidics, Faculty of Mechanical Engineering and Marine Technology, University of Rostock, Rostock, Germany
| | - Christoph Drobek
- Chair of Microfluidics, Faculty of Mechanical Engineering and Marine Technology, University of Rostock, Rostock, Germany
| | - Silvia Budday
- Department of Mechanical Engineering, Institute of Applied Mechanics, Friedrich-Alexander-University Erlangen-Nürnberg, Erlangen, Germany
| | - Hermann Seitz
- Chair of Microfluidics, Faculty of Mechanical Engineering and Marine Technology, University of Rostock, Rostock, Germany
| |
Collapse
|
20
|
Tang Y, Xu H, Wang X, Dong S, Guo L, Zhang S, Yang X, Liu C, Jiang X, Kan M, Wu S, Zhang J, Xu C. Advances in preparation and application of antibacterial hydrogels. J Nanobiotechnology 2023; 21:300. [PMID: 37633883 PMCID: PMC10463510 DOI: 10.1186/s12951-023-02025-8] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Accepted: 07/24/2023] [Indexed: 08/28/2023] Open
Abstract
Bacterial infections, especially those caused by drug-resistant bacteria, have seriously threatened human life and health. There is urgent to develop new antibacterial agents to reduce the problem of antibiotics. Biomedical materials with good antimicrobial properties have been widely used in antibacterial applications. Among them, hydrogels have become the focus of research in the field of biomedical materials due to their unique three-dimensional network structure, high hydrophilicity, and good biocompatibility. In this review, the latest research progresses about hydrogels in recent years were summarized, mainly including the preparation methods of hydrogels and their antibacterial applications. According to their different antibacterial mechanisms, several representative antibacterial hydrogels were introduced, such as antibiotics loaded hydrogels, antibiotic-free hydrogels including metal-based hydrogels, antibacterial peptide and antibacterial polymers, stimuli-responsive smart hydrogels, and light-mediated hydrogels. In addition, we also discussed the applications and challenges of antibacterial hydrogels in biomedicine, which are expected to provide new directions and ideas for the application of hydrogels in clinical antibacterial therapy.
Collapse
Affiliation(s)
- Yixin Tang
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun, 130021 Jilin China
| | - Huiqing Xu
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun, 130021 Jilin China
| | - Xue Wang
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun, 130021 Jilin China
| | - Shuhan Dong
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun, 130021 Jilin China
- Department of Preventive Medicine, School of Public Health, Jilin University, Changchun, 130021 Jilin China
| | - Lei Guo
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun, 130021 Jilin China
| | - Shichen Zhang
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun, 130021 Jilin China
- Department of Oral and Maxillofacial Surgery, Hospital of Stomatology, Jilin University, Changchun, 130021 Jilin China
| | - Xi Yang
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun, 130021 Jilin China
| | - Chang Liu
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun, 130021 Jilin China
| | - Xin Jiang
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun, 130021 Jilin China
| | - Mujie Kan
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun, 130021 Jilin China
| | - Shanli Wu
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun, 130021 Jilin China
| | - Jizhou Zhang
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun, 130021 Jilin China
| | - Caina Xu
- Department of Biochemistry, College of Basic Medical Sciences, Jilin University, Changchun, 130021 Jilin China
| |
Collapse
|
21
|
Fernández‐Colino A, Kiessling F, Slabu I, De Laporte L, Akhyari P, Nagel SK, Stingl J, Reese S, Jockenhoevel S. Lifelike Transformative Materials for Biohybrid Implants: Inspired by Nature, Driven by Technology. Adv Healthc Mater 2023; 12:e2300991. [PMID: 37290055 PMCID: PMC11469152 DOI: 10.1002/adhm.202300991] [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: 03/28/2023] [Revised: 05/25/2023] [Indexed: 06/10/2023]
Abstract
Today's living world is enriched with a myriad of natural biological designs, shaped by billions of years of evolution. Unraveling the construction rules of living organisms offers the potential to create new materials and systems for biomedicine. From the close examination of living organisms, several concepts emerge: hierarchy, pattern repetition, adaptation, and irreducible complexity. All these aspects must be tackled to develop transformative materials with lifelike behavior. This perspective article highlights recent progress in the development of transformative biohybrid systems for applications in the fields of tissue regeneration and biomedicine. Advances in computational simulations and data-driven predictions are also discussed. These tools enable the virtual high-throughput screening of implant design and performance before committing to fabrication, thus reducing the development time and cost of biomimetic and biohybrid constructs. The ongoing progress of imaging methods also constitutes an essential part of this matter in order to validate the computation models and enable longitudinal monitoring. Finally, the current challenges of lifelike biohybrid materials, including reproducibility, ethical considerations, and translation, are discussed. Advances in the development of lifelike materials will open new biomedical horizons, where perhaps what is currently envisioned as science fiction will become a science-driven reality in the future.
Collapse
Affiliation(s)
- Alicia Fernández‐Colino
- Department of Biohybrid & Medical Textiles (BioTex)AME‐Institute of Applied Medical EngineeringHelmholtz InstituteRWTH Aachen UniversityForckenbeckstraße 5552074AachenGermany
| | - Fabian Kiessling
- Institute for Experimental Molecular ImagingFaculty of MedicineRWTH Aachen UniversityForckenbeckstraße 5552074AachenGermany
| | - Ioana Slabu
- Institute of Applied Medical EngineeringHelmholtz InstituteMedical FacultyRWTH Aachen UniversityPauwelsstraße 2052074AachenGermany
| | - Laura De Laporte
- DWI – Leibniz‐Institute for Interactive MaterialsForckenbeckstraße 5052074AachenGermany
- Institute of Technical and Macromolecular Chemistry (ITMC)RWTH Aachen UniversityWorringerweg 252074AachenGermany
- Advanced Materials for Biomedicine (AMB)Institute of Applied Medical Engineering (AME)University Hospital RWTH AachenCenter for Biohybrid Medical Systems (CMBS)Forckenbeckstraße 5552074AachenGermany
| | - Payam Akhyari
- Clinic for Cardiac SurgeryUniversity Hospital RWTH AachenPauwelsstraße 3052074AachenGermany
| | - Saskia K. Nagel
- Applied Ethics GroupRWTH Aachen UniversityTheaterplatz 1452062AachenGermany
| | - Julia Stingl
- Institute of Clinical PharmacologyUniversity Hospital RWTH AachenWendlingweg 252074AachenGermany
| | - Stefanie Reese
- Institute of Applied MechanicsRWTH Aachen UniversityMies‐van‐der‐Rohe‐Str. 152074AachenGermany
| | - Stefan Jockenhoevel
- Department of Biohybrid & Medical Textiles (BioTex)AME‐Institute of Applied Medical EngineeringHelmholtz InstituteRWTH Aachen UniversityForckenbeckstraße 5552074AachenGermany
| |
Collapse
|
22
|
Kim JT, Cho SM, Youn DH, Hong EP, Park CH, Lee Y, Jung H, Jeon JP. Therapeutic Effect of a Hydrogel-based Neural Stem Cell Delivery Sheet for Mild Traumatic Brain Injury. Acta Biomater 2023:S1742-7061(23)00351-3. [PMID: 37356785 DOI: 10.1016/j.actbio.2023.06.027] [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: 01/02/2023] [Revised: 06/18/2023] [Accepted: 06/20/2023] [Indexed: 06/27/2023]
Abstract
OBJECTIVE There are no effective clinically applicable treatments for neuronal dysfunction after mild traumatic brain injury (TBI). Here, we evaluated the therapeutic effect of a new delivery method of mouse neural stem cell (mNSC) spheroids using a hydrogel, in terms of improvement in damaged cortical lesions and cognitive impairment after mild TBI. METHODS mNSCs were isolated from the subventricular zone and subgranular zone by a hydrogel-based culture system. GFP-transduced mNSCs were generated into spheroids and wrapped into a sheet for transplantation. Male C57BL/6J mice were randomly divided into four groups: sham operation, TBI, TBI with mNSC spheroids, and TBI with mNSC spheroid sheet transplantation covering the damaged cortex. Histopathological and immunohistochemical features and cognitive function were evaluated 7, 14, and 28 days after transplantation following TBI. RESULTS Hydrogel-based culture systems and mNSC isolation were successfully established from the adult mice. Essential transcription factors for NSCs, such as SOX2, PAX6, Olig2, nestin, and doublecortin (DCX), were highly expressed in the mNSCs. A transplanted hydrogel-based mNSC spheroid sheet showed good engraftment and survival ability, differentiated into TUJ1-positive neurons, promoted angiogenesis, and reduced neuronal degeneration. Also, TBI mice treated with mNSC spheroid sheet transplantation exhibited a significantly increased preference for a new object, suggesting improved cognitive function compared to the mNSC spheroids or no treatment groups. CONCLUSION Transplantation with a hydrogel-based mNSC spheroid sheet showed engraftment, migration, and stability of delivered cells in a hostile microenvironment after TBI, resulting in improved cognitive function via reconstruction of the damaged cortex. STATEMENT OF SIGNIFICANCE This study presents the therapeutic effect of a new delivery method of mouse neural stem cells spheroids using a hydrogel, in terms of improvement in damaged cortical lesions and cognitive impairment after traumatic brain injury. Collagen/fibrin hydrogel allowed long-term survival and migratory ability of NSCs spheroids. Furthermore, transplanted hydrogel-based mNSCs spheroids sheet showed good engraftment, migration, and stability of delivered cells in a hostile microenvironment, resulting in reconstruction of the damaged cortex and improved cognitive function after TBI. Therefore, we suggest that a hydrogel-based mNSCs spheroids sheet could help to improve cognitive impairment after TBI.
Collapse
Affiliation(s)
- Jong-Tae Kim
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon, Korea
| | - Sung Min Cho
- Department of Neurosurgery, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Dong Hyuk Youn
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon, Korea
| | - Eun Pyo Hong
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon, Korea
| | - Chan Hum Park
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon, Korea
| | - Younghyurk Lee
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon, Korea
| | - Harry Jung
- Institute of New Frontier Research, Hallym University College of Medicine, Chuncheon, Korea
| | - Jin Pyeong Jeon
- Department of Neurosurgery, Hallym University College of Medicine, Chuncheon, Korea.
| |
Collapse
|
23
|
Hu Y, Lyu C, Teng L, Wu A, Zhu Z, He Y, Lu J. Glycopolypeptide hydrogels with adjustable enzyme-triggered degradation: A novel proteoglycans analogue to repair articular-cartilage defects. Mater Today Bio 2023; 20:100659. [PMID: 37229212 PMCID: PMC10205498 DOI: 10.1016/j.mtbio.2023.100659] [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: 01/17/2023] [Revised: 04/28/2023] [Accepted: 05/04/2023] [Indexed: 05/27/2023] Open
Abstract
Proteoglycans (PGs), also known as a viscous lubricant, is the main component of the cartilage extracellular matrix (ECM). The loss of PGs is accompanied by the chronic degeneration of cartilage tissue, which is an irreversible degeneration process that eventually develops into osteoarthritis (OA). Unfortunately, there is still no substitute for PGs in clinical treatments. Herein, we propose a new PGs analogue. The Glycopolypeptide hydrogels in the experimental groups with different concentrations were prepared by Schiff base reaction (Gel-1, Gel-2, Gel-3, Gel-4, Gel-5 and Gel-6). They have good biocompatibility and adjustable enzyme-triggered degradability. The hydrogels have a loose and porous structure suitable for the proliferation, adhesion, and migration of chondrocytes, good anti-swelling, and reduce the reactive oxygen species (ROS) in chondrocytes. In vitro experiments confirmed that the glycopolypeptide hydrogels significantly promoted ECM deposition and up-regulated the expression of cartilage-specific genes, such as type-II collagen, aggrecan, and glycosaminoglycans (sGAG). In vivo, the New Zealand rabbit knee articular cartilage defect model was established and the hydrogels were implanted to repair it, the results showed good cartilage regeneration potential. It is worth noting that the Gel-3 group, with a pore size of 122 ± 12 μm, was particularly prominent in the above experiments, and provides a theoretical reference for the design of cartilage-tissue regeneration materials in the future.
Collapse
Affiliation(s)
- Yinghan Hu
- Department of Stomatology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Chengqi Lyu
- Department of Stomatology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Lin Teng
- Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Anqian Wu
- Department of Stomatology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - Zeyu Zhu
- Department of Stomatology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| | - YuShi He
- Shanghai Electrochemical Energy Devices Research Center, School of Chemistry and Chemical Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Jiayu Lu
- Department of Stomatology, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai, 200233, China
| |
Collapse
|
24
|
Baei P, Daemi H, Aramesh F, Baharvand H, Eslaminejad MB. Advances in mechanically robust and biomimetic polysaccharide-based constructs for cartilage tissue engineering. Carbohydr Polym 2023; 308:120650. [PMID: 36813342 DOI: 10.1016/j.carbpol.2023.120650] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/28/2023] [Accepted: 01/30/2023] [Indexed: 02/05/2023]
Abstract
The purpose of cartilage tissue engineering is to provide artificial constructs with biological functions and mechanical features that resemble native tissue to improve tissue regeneration. Biochemical characteristics of the cartilage extracellular matrix (ECM) microenvironment provide a platform for researchers to develop biomimetic materials for optimal tissue repair. Due to the structural similarity of polysaccharides into physicochemical characteristics of cartilage ECM, these natural polymers capture special attention for developing biomimetic materials. The mechanical properties of constructs play a crucial influence in load-bearing cartilage tissues. Moreover, the addition of appropriate bioactive molecules to these constructs can promote chondrogenesis. Here, we discuss polysaccharide-based constructs that can be used to create substitutes for cartilage regeneration. We intend to focus on newly developed bioinspired materials, fine-tuning the mechanical properties of constructs, the design of carriers loaded by chondroinductive agents, and development of appropriate bioinks as a bioprinting approach for cartilage regeneration.
Collapse
Affiliation(s)
- Payam Baei
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Tissue Engineering, School of Advanced Technologies in Medicine, Royan Institute, Tehran, Iran
| | - Hamed Daemi
- Department of Cell Engineering, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Tissue Engineering, School of Advanced Technologies in Medicine, Royan Institute, Tehran, Iran.
| | - Fatemeh Aramesh
- Department of Surgery and Radiology, Faculty of Veterinary Medicine, University ofTehran, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Developmental Biology, School of Basic Sciences and Advanced Technologies in Biology, University of Science and Culture, Tehran, Iran
| | - Mohamadreza Baghaban Eslaminejad
- Department of Tissue Engineering, School of Advanced Technologies in Medicine, Royan Institute, Tehran, Iran; Department of Stem Cells and Developmental Biology, Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran.
| |
Collapse
|
25
|
Horvath-Pereira BDO, Almeida GHDR, da Silva Júnior LN, do Nascimento PG, Horvath Pereira BDO, Fireman JVBT, Pereira MLDRF, Carreira ACO, Miglino MA. Biomaterials for Testicular Bioengineering: How far have we come and where do we have to go? Front Endocrinol (Lausanne) 2023; 14:1085872. [PMID: 37008920 PMCID: PMC10060902 DOI: 10.3389/fendo.2023.1085872] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2022] [Accepted: 02/24/2023] [Indexed: 03/18/2023] Open
Abstract
Traditional therapeutic interventions aim to restore male fertile potential or preserve sperm viability in severe cases, such as semen cryopreservation, testicular tissue, germ cell transplantation and testicular graft. However, these techniques demonstrate several methodological, clinical, and biological limitations, that impact in their results. In this scenario, reproductive medicine has sought biotechnological alternatives applied for infertility treatment, or to improve gamete preservation and thus increase reproductive rates in vitro and in vivo. One of the main approaches employed is the biomimetic testicular tissue reconstruction, which uses tissue-engineering principles and methodologies. This strategy pursues to mimic the testicular microenvironment, simulating physiological conditions. Such approach allows male gametes maintenance in culture or produce viable grafts that can be transplanted and restore reproductive functions. In this context, the application of several biomaterials have been proposed to be used in artificial biological systems. From synthetic polymers to decellularized matrixes, each biomaterial has advantages and disadvantages regarding its application in cell culture and tissue reconstruction. Therefore, the present review aims to list the progress that has been made and the continued challenges facing testicular regenerative medicine and the preservation of male reproductive capacity, based on the development of tissue bioengineering approaches for testicular tissue microenvironment reconstruction.
Collapse
Affiliation(s)
| | | | | | - Pedro Gabriel do Nascimento
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| | | | | | | | - Ana Claudia Oliveira Carreira
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
- Centre for Natural and Human Sciences, Federal University of ABC, São Paulo, Brazil
| | - Maria Angelica Miglino
- Department of Surgery, School of Veterinary Medicine and Animal Science, University of São Paulo, São Paulo, Brazil
| |
Collapse
|
26
|
Stie MB, Öblom H, Hansen ACN, Jacobsen J, Chronakis IS, Rantanen J, Nielsen HM, Genina N. Mucoadhesive chitosan- and cellulose derivative-based nanofiber-on-foam-on-film system for non-invasive peptide delivery. Carbohydr Polym 2023; 303:120429. [PMID: 36657829 DOI: 10.1016/j.carbpol.2022.120429] [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: 09/13/2022] [Revised: 11/18/2022] [Accepted: 11/30/2022] [Indexed: 12/05/2022]
Abstract
Oromucosal administration is an attractive non-invasive route. However, drug absorption is challenged by salivary flow and the mucosa being a significant permeability barrier. The aim of this study was to design and investigate a multi-layered nanofiber-on-foam-on-film (NFF) drug delivery system with unique properties and based on polysaccharides combined as i) mucoadhesive chitosan-based nanofibers, ii) a peptide loaded hydroxypropyl methylcellulose foam, and iii) a saliva-repelling backing film based on ethylcellulose. NFF displays optimal mechanical properties shown by dynamic mechanical analysis, and biocompatibility demonstrated after exposure to a TR146 cell monolayer. Chitosan-based nanofibers provided the NFF with improved mucoadhesion compared to that of the foam alone. After 1 h, >80 % of the peptide desmopressin was released from the NFF. Ex vivo permeation studies across porcine buccal mucosa indicated that NFF improved the permeation of desmopressin compared to a commercial freeze-dried tablet. The findings demonstrate the potential of the NFF as a biocompatible drug delivery system.
Collapse
Affiliation(s)
- Mai Bay Stie
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark; Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Heidi Öblom
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark; Pharmaceutical Sciences Laboratory, Åbo Akademi University, Artillerigatan 6A, 20520 Åbo, Finland
| | | | - Jette Jacobsen
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Ioannis S Chronakis
- DTU-Food, Technical University of Denmark, B202, Kemitorvet, 2800 Kgs. Lyngby, Denmark
| | - Jukka Rantanen
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| | - Hanne Mørck Nielsen
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark; Center for Biopharmaceuticals and Biobarriers in Drug Delivery (BioDelivery), Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark.
| | - Natalja Genina
- Department of Pharmacy, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| |
Collapse
|
27
|
Rahnamaee SY, Dehnavi SM, Bagheri R, Barjasteh M, Golizadeh M, Zamani H, Karimi A. Boosting bone cell growth using nanofibrous carboxymethylated cellulose and chitosan on titanium dioxide nanotube array with dual surface charges as a novel multifunctional bioimplant surface. Int J Biol Macromol 2023; 228:570-581. [PMID: 36563824 DOI: 10.1016/j.ijbiomac.2022.12.159] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2022] [Revised: 12/02/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022]
Abstract
One of the most vital aspects of the orthopedic implant field has been the development of multifunctional coatings that improve bone-implant contact while simultaneously preventing bacterial infection. The present study investigates the fabrication and characterization of multifunctional polysaccharides, including carboxymethyl cellulose (CMCn) and carboxymethyl chitosan nanofibers (CMCHn), as a novel implant coating on titania nanotube arrays (T). Field emission scanning electron microscopy (FESEM) images revealed a nanofibrous morphology with a narrow diameter for CMCn and CMCHn, similar to extracellular matrix nanostructures. Compared to the T surface, the roughness of CMCn and CMCHn samples increased by over 250 %. An improved cell proliferation rate was observed on CMCHn nanofibers with a positively charged surface caused by the amino groups. Furthermore, in an antibacterial experiment, CMCn and CMCHn inhibited bacterial colony formation by 80 % and 73 %, respectively. According to the results, constructed modified CMCn and CMCHn increased osteoblast cell survival while inhibiting bacterial biofilm formation owing to their surface charge and bioinspired physicochemical properties.
Collapse
Affiliation(s)
- Seyed Yahya Rahnamaee
- Polymeric Materials Research Group (PMRG), Department of Materials Science and Engineering, Sharif University of Technology, Tehran, Iran
| | - Seyed Mohsen Dehnavi
- Department of Life Science and Biotechnology, Shahid Beheshti University, Tehran, Iran.
| | - Reza Bagheri
- Polymeric Materials Research Group (PMRG), Department of Materials Science and Engineering, Sharif University of Technology, Tehran, Iran.
| | - Mahdi Barjasteh
- Institute for Nanoscience & Nanotechnology (INST), Sharif University of Technology, Tehran, Iran; BioTex Innovation Factory, Sharif Development of Health and Biotechnology Institute, Tehran, Iran
| | - Mortaza Golizadeh
- BioTex Innovation Factory, Sharif Development of Health and Biotechnology Institute, Tehran, Iran
| | - Hedyeh Zamani
- Department of Life Science and Biotechnology, Shahid Beheshti University, Tehran, Iran
| | - Afzal Karimi
- Department of Medical Biotechnology, School of Advanced Technologies in Medicine, Iran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
28
|
Loi G, Stucchi G, Scocozza F, Cansolino L, Cadamuro F, Delgrosso E, Riva F, Ferrari C, Russo L, Conti M. Characterization of a Bioink Combining Extracellular Matrix-like Hydrogel with Osteosarcoma Cells: Preliminary Results. Gels 2023; 9:gels9020129. [PMID: 36826299 PMCID: PMC9957231 DOI: 10.3390/gels9020129] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 01/27/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023] Open
Abstract
Three-dimensional (3D) bioprinting allows the production of artificial 3D cellular microenvironments thanks to the controlled spatial deposition of bioinks. Proper bioink characterization is required to achieve the essential characteristics of printability and biocompatibility for 3D bioprinting. In this work, a protocol to standardize the experimental characterization of a new bioink is proposed. A functionalized hydrogel based on gelatin and chitosan was used. The protocol was divided into three steps: pre-printing, 3D bioprinting, and post-printing. For the pre-printing step, the hydrogel formulation and its repeatability were evaluated. For the 3D-bioprinting step, the hydrogel-printability performance was assessed through qualitative and quantitative tests. Finally, for the post-printing step, the hydrogel biocompatibility was investigated using UMR-106 cells. The hydrogel was suitable for printing grids with good resolution from 4 h after the cross-linker addition. To guarantee a constant printing pressure, it was necessary to set the extruder to 37 °C. Furthermore, the hydrogel was shown to be a valid biomaterial for the UMR-106 cells' growth. However, fragmentation of the constructs appeared after 14 days, probably due to the negative osteosarcoma-cell interference. The protocol that we describe here denotes a strong approach to bioink characterization to improve standardization for future biomaterial screening and development.
Collapse
Affiliation(s)
- Giada Loi
- Department of Civil Engineering and Architecture, University of Pavia, Via Adolfo Ferrata 3, 27100 Pavia, Italy
- Correspondence:
| | - Gaia Stucchi
- Department of Clinical Surgical Sciences, University of Pavia, Via Adolfo Ferrata 5, 27100 Pavia, Italy
| | - Franca Scocozza
- Department of Civil Engineering and Architecture, University of Pavia, Via Adolfo Ferrata 3, 27100 Pavia, Italy
| | - Laura Cansolino
- Department of Clinical Surgical Sciences, University of Pavia, Via Adolfo Ferrata 5, 27100 Pavia, Italy
| | - Francesca Cadamuro
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
| | - Elena Delgrosso
- Department of Clinical Surgical Sciences, University of Pavia, Via Adolfo Ferrata 5, 27100 Pavia, Italy
| | - Federica Riva
- Department of Public Health, Experimental and Forensic Medicine, Histology and Embryology Unit, University of Pavia, Via Forlanini 2, 27100 Pavia, Italy
| | - Cinzia Ferrari
- Department of Clinical Surgical Sciences, University of Pavia, Via Adolfo Ferrata 5, 27100 Pavia, Italy
- Animal Welfare and Radiobiology Service Center, University of Pavia, Via Adolfo Ferrata 5, 27100 Pavia, Italy
| | - Laura Russo
- Department of Biotechnology and Biosciences, University of Milano-Bicocca, Piazza della Scienza 2, 20126 Milan, Italy
- CÚRAM SFI Research Centre for Medical Devices, National University of Ireland Galway, H92 W2TY Galway, Ireland
| | - Michele Conti
- Department of Civil Engineering and Architecture, University of Pavia, Via Adolfo Ferrata 3, 27100 Pavia, Italy
| |
Collapse
|
29
|
Ultralong hydroxyapatite nanowires-incorporated dipeptide hydrogel with enhanced mechanical strength and superior in vivo osteogenesis activity. Colloids Surf A Physicochem Eng Asp 2023. [DOI: 10.1016/j.colsurfa.2023.131153] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/18/2023]
|
30
|
Chen L, Li L, Mo Q, Zhang X, Chen C, Wu Y, Zeng X, Deng K, Liu N, Zhu P, Liu M, Xiao Y. An injectable gelatin/sericin hydrogel loaded with human umbilical cord mesenchymal stem cells for the treatment of uterine injury. Bioeng Transl Med 2023; 8:e10328. [PMID: 36684066 PMCID: PMC9842051 DOI: 10.1002/btm2.10328] [Citation(s) in RCA: 22] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2022] [Revised: 03/17/2022] [Accepted: 04/04/2022] [Indexed: 01/25/2023] Open
Abstract
Abnormal endometrial receptivity is a major cause of the failure of embryo transplantation, which may lead to infertility, adverse pregnancy, and neonatal outcomes. While hormonal treatment has dramatically improved the fertility outcomes in women with endometriosis, a substantial unmet need persists in the treatment. In this study, methacrylate gelatin (GelMA) and methacrylate sericin (SerMA) hydrogel with human umbilical cord mesenchymal stem cells (HUMSC) encapsulation was designed for facilitating endometrial regeneration and fertility restoration through in situ injection. The presented GelMA/10%SerMA hydrogel showed appropriate swelling ratio, good mechanical properties, and degradation stability. In vitro cell experiments showed that the prepared hydrogels had excellent biocompatibility and cell encapsulation ability of HUMSC. Further in vivo experiments demonstrated that GelMA/SerMA@HUMSC hydrogel could increase the thickness of endometrium and improve the endometrial interstitial fibrosis. Moreover, regenerated endometrial tissue was more receptive to transfer embryos. Summary, we believed that GelMA/SerMA@HUMSC hydrogel will hold tremendous promise to repair or regenerate damaged endometrium.
Collapse
Affiliation(s)
- Lixuan Chen
- Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Jinshazhou Hospital of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Ling Li
- Jiangmen Maternity and Child Health Care HospitalJiangmenGuangdongChina
| | - Qinglin Mo
- Translational Medicine CenterThe Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Xiaomin Zhang
- Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Jinshazhou Hospital of Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
| | - Chaolin Chen
- Translational Medicine CenterThe Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Yingnan Wu
- Translational Medicine CenterThe Second Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Xiaoli Zeng
- National Seed Cell Bank of South China for Tissue EngineeringGuangzhouGuangdongChina
| | - Kaixian Deng
- Department of Gynecology, Shunde HospitalSouthern Medical University (The First People's Hospital of Shunde)FoshanGuangdongChina
| | - Nanbo Liu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
| | - Ping Zhu
- Guangdong Cardiovascular Institute, Guangdong Provincial People's HospitalGuangdong Academy of Medical SciencesGuangzhouChina
| | - Mingxing Liu
- Department of Obstetrics and Gynecology, Key Laboratory for Major Obstetric Diseases of Guangdong ProvinceThe Third Affiliated Hospital of Guangzhou Medical UniversityGuangzhouGuangdongChina
| | - Yang Xiao
- Guangzhou University of Chinese MedicineGuangzhouGuangdongChina
- Shenzhen Qianhai Shekou Pilot Free Trade Zone Hospital, ShekouShenzhenGuangdongChina
| |
Collapse
|
31
|
Schuiringa GH, Pastrama M, Ito K, van Donkelaar CC. Towards a load bearing hydrogel: A proof of principle in the use of osmotic pressure for biomimetic cartilage constructs. J Mech Behav Biomed Mater 2023; 137:105552. [PMID: 36371992 DOI: 10.1016/j.jmbbm.2022.105552] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/27/2022] [Accepted: 10/31/2022] [Indexed: 11/09/2022]
Abstract
Cartilage defects occur frequently and can lead to osteoarthritis. Hydrogels are a promising regenerative strategy for treating such defects, using their ability of mimicking the native extracellular matrix. However, commonly used hydrogels for tissue regeneration are too soft to resist load-bearing in the joint. To overcome this, an implant is being developed in which the mechanical loadbearing function originates from the osmotic pressure generated by the swelling potential of a charged hydrogel, which is restricted from swelling by a textile spacer fabric. This study aims to quantify the relationship between the swelling potential of the hydrogel and the compressive stiffness of the implant.
Collapse
Affiliation(s)
- Gerke H Schuiringa
- Orthopaedic Biomechanics, Dept. Biomedical Engineering, Eindhoven University of Technology, the Netherlands
| | - Maria Pastrama
- Orthopaedic Biomechanics, Dept. Biomedical Engineering, Eindhoven University of Technology, the Netherlands
| | - Keita Ito
- Orthopaedic Biomechanics, Dept. Biomedical Engineering, Eindhoven University of Technology, the Netherlands
| | - Corrinus C van Donkelaar
- Orthopaedic Biomechanics, Dept. Biomedical Engineering, Eindhoven University of Technology, the Netherlands.
| |
Collapse
|
32
|
Sithole MN, Mndlovu H, du Toit LC, Choonara YE. Advances in Stimuli-responsive Hydrogels for Tissue Engineering and Regenerative Medicine Applications: A Review Towards Improving Structural Design for 3D Printing. Curr Pharm Des 2023; 29:3187-3205. [PMID: 37779402 DOI: 10.2174/0113816128246888230920060802] [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] [Revised: 06/17/2023] [Accepted: 07/14/2023] [Indexed: 10/03/2023]
Abstract
The physicochemical properties of polymeric hydrogels render them attractive for the development of 3D printed prototypes for tissue engineering in regenerative medicine. Significant effort has been made to design hydrogels with desirable attributes that facilitate 3D printability. In addition, there is significant interest in exploring stimuli-responsive hydrogels to support automated 3D printing into more structurally organised prototypes such as customizable bio-scaffolds for regenerative medicine applications. Synthesizing stimuli-responsive hydrogels is dependent on the type of design and modulation of various polymeric materials to open novel opportunities for applications in biomedicine and bio-engineering. In this review, the salient advances made in the design of stimuli-responsive polymeric hydrogels for 3D printing in tissue engineering are discussed with a specific focus on the different methods of manipulation to develop 3D printed stimuli-responsive polymeric hydrogels. Polymeric functionalisation, nano-enabling and crosslinking are amongst the most common manipulative attributes that affect the assembly and structure of 3D printed bio-scaffolds and their stimuli- responsiveness. The review also provides a concise incursion into the various applications of stimuli to enhance the automated production of structurally organized 3D printed medical prototypes.
Collapse
Affiliation(s)
- Mduduzi Nkosinathi Sithole
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, Gauteng, 2193, South Africa
| | - Hillary Mndlovu
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, Gauteng, 2193, South Africa
| | - Lisa C du Toit
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, Gauteng, 2193, South Africa
| | - Yahya Essop Choonara
- Wits Advanced Drug Delivery Platform Research Unit, Department of Pharmacy and Pharmacology, School of Therapeutic Sciences, Faculty of Health Sciences, University of the Witwatersrand, 7 York Road, Parktown, Johannesburg, Gauteng, 2193, South Africa
| |
Collapse
|
33
|
Xu Z, Wu L, Tang Y, Xi K, Tang J, Xu Y, Xu J, Lu J, Guo K, Gu Y, Chen L. Spatiotemporal Regulation of the Bone Immune Microenvironment via Dam-Like Biphasic Bionic Periosteum for Bone Regeneration. Adv Healthc Mater 2023; 12:e2201661. [PMID: 36189833 PMCID: PMC11469314 DOI: 10.1002/adhm.202201661] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Revised: 09/24/2022] [Indexed: 02/03/2023]
Abstract
The bone immune microenvironment (BIM) regulates bone regeneration and affects the prognosis of fractures. However, there is currently no effective strategy that can precisely modulate macrophage polarization to improve BIM for bone regeneration. Herein, a hybridized biphasic bionic periosteum, inspired by the BIM and functional structure of the natural periosteum, is presented. The gel phase is composed of genipin-crosslinked carboxymethyl chitosan and collagen self-assembled hybrid hydrogels, which act as the "dam" to intercept IL-4 released during the initial burst from the bionic periosteum fiber phase, thus maintaining the moderate inflammatory response of M1 macrophages for mesenchymal stem cell recruitment and vascular sprouting at the acute fracture. With the degradation of the gel phase, released IL-4 cooperates with collagen to promote the polarization towards M2 macrophages, which reconfigure the local microenvironment by secreting PDGF-BB and BMP-2 to improve vascular maturation and osteogenesis twofold. In rat cranial defect models, the controlled regulation of the BIM is validated with the temporal transition of the inflammatory/anti-inflammatory process to achieve faster and better bone defect repair. This strategy provides a drug delivery system that constructs a coordinated BIM, so as to break through the predicament of the contradiction between immune response and bone tissue regeneration.
Collapse
Affiliation(s)
- Zonghan Xu
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| | - Liang Wu
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| | - Yu Tang
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| | - Kun Xi
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| | - Jincheng Tang
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| | - Yichang Xu
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| | - Jingzhi Xu
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| | - Jian Lu
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| | - Kaijin Guo
- Department of Orthopedicsthe Affiliated Hospital of Xuzhou Medical University99 Huaihai West RoadXuzhouJiangsu221000P. R. China
| | - Yong Gu
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| | - Liang Chen
- Department of Orthopedicsthe First Affiliated Hospital of Soochow UniversityOrthopedic InstituteSoochow University188 Shizi RoadSuzhouJiangsu215006P. R. China
| |
Collapse
|
34
|
Zhou Y, Yang Y, Liu R, Zhou Q, Lu H, Zhang W. Research Progress of Polydopamine Hydrogel in the Prevention and Treatment of Oral Diseases. Int J Nanomedicine 2023; 18:2623-2645. [PMID: 37213351 PMCID: PMC10199686 DOI: 10.2147/ijn.s407044] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2023] [Accepted: 04/12/2023] [Indexed: 05/23/2023] Open
Abstract
Oral diseases represent one of the most prevalent diseases globally and are associated with serious health and economic burdens, greatly altering the quality of life of affected individuals. Various biomaterials play important roles in the treatment of oral diseases. To some extent, the development of biomaterials has promoted progress in clinically available oral medicines. Hydrogels have unique tunable advantages that make them useful in the next generation of regenerative strategies and have been widely applied in both oral soft and hard tissues repair. However, most hydrogels lack self-adhesive properties, which may result in low repair efficacy. Polydopamine (PDA), the primary adhesive component, has attracted increasing attention in recent years. PDA-modified hydrogels exhibit reliable and suitable adherence to tissues and easily integrate into tissues to promote repair efficiency. This paper reviews the latest research progress on PDA hydrogels and elaborates on the mechanism of the reaction between PDA functional groups and hydrogels, and summarizes the biological properties and the applications of PDA hydrogels in the prevention and treatment of the field of oral diseases. It is also proposed that in future research we should simulate the complex microenvironment of the oral cavity as much as possible, coordinate and plan various biological events rationally, and realize the translation from scientific research to clinical practice.
Collapse
Affiliation(s)
- Yuqi Zhou
- Department of Prosthodontics, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Yuanmeng Yang
- Department of Preventive Dentistry, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Rongpu Liu
- Department of Prosthodontics, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Qin Zhou
- Department of Oral Surgery, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
| | - Haixia Lu
- Department of Preventive Dentistry, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Haixia Lu, Department of Preventive Dentistry, Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, People’s Republic of China, Email
| | - Wenjie Zhang
- Department of Prosthodontics, Ninth People’s Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, People’s Republic of China
- Correspondence: Wenjie Zhang, Department of Prosthodontics, Ninth People’s Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, 200011, People’s Republic of China, Email
| |
Collapse
|
35
|
Blache U, Ford EM, Ha B, Rijns L, Chaudhuri O, Dankers PY, Kloxin AM, Snedeker JG, Gentleman E. Engineered hydrogels for mechanobiology. NATURE REVIEWS. METHODS PRIMERS 2022; 2:98. [PMID: 37461429 PMCID: PMC7614763 DOI: 10.1038/s43586-022-00179-7] [Citation(s) in RCA: 58] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Accepted: 10/17/2022] [Indexed: 07/20/2023]
Abstract
Cells' local mechanical environment can be as important in guiding cellular responses as many well-characterized biochemical cues. Hydrogels that mimic the native extracellular matrix can provide these mechanical cues to encapsulated cells, allowing for the study of their impact on cellular behaviours. Moreover, by harnessing cellular responses to mechanical cues, hydrogels can be used to create tissues in vitro for regenerative medicine applications and for disease modelling. This Primer outlines the importance and challenges of creating hydrogels that mimic the mechanical and biological properties of the native extracellular matrix. The design of hydrogels for mechanobiology studies is discussed, including appropriate choice of cross-linking chemistry and strategies to tailor hydrogel mechanical cues. Techniques for characterizing hydrogels are explained, highlighting methods used to analyze cell behaviour. Example applications for studying fundamental mechanobiological processes and regenerative therapies are provided, along with a discussion of the limitations of hydrogels as mimetics of the native extracellular matrix. The article ends with an outlook for the field, focusing on emerging technologies that will enable new insights into mechanobiology and its role in tissue homeostasis and disease.
Collapse
Affiliation(s)
- Ulrich Blache
- Fraunhofer Institute for Cell Therapy and Immunology and Fraunhofer Cluster of Excellence for Immune-Mediated Disease, Leipzig, Germany
| | - Eden M. Ford
- Department of Chemical and Biomolecular Engineering, University of Delaware, DE, USA
| | - Byunghang Ha
- Department of Mechanical Engineering, Stanford University, CA, USA
| | - Laura Rijns
- Institute for Complex Molecular Systems, Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
| | - Ovijit Chaudhuri
- Department of Mechanical Engineering, Stanford University, CA, USA
| | - Patricia Y.W. Dankers
- Institute for Complex Molecular Systems, Department of Biomedical Engineering, Laboratory of Chemical Biology, Eindhoven University of Technology, PO Box 513, 5600 MB Eindhoven, The Netherlands
| | - April M. Kloxin
- Department of Chemical and Biomolecular Engineering, University of Delaware, DE, USA
- Department of Material Science and Engineering, University of Delaware, DE, USA
| | - Jess G. Snedeker
- University Hospital Balgrist and ETH Zurich, Zurich, Switzerland
| | - Eileen Gentleman
- Centre for Craniofacial and Regenerative Biology, King’s College London, London SE1 9RT, UK
| |
Collapse
|
36
|
Sacramento MMA, Borges J, Correia FJS, Calado R, Rodrigues JMM, Patrício SG, Mano JF. Green approaches for extraction, chemical modification and processing of marine polysaccharides for biomedical applications. Front Bioeng Biotechnol 2022; 10:1041102. [PMID: 36568299 PMCID: PMC9773402 DOI: 10.3389/fbioe.2022.1041102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2022] [Accepted: 11/24/2022] [Indexed: 12/13/2022] Open
Abstract
Over the past few decades, natural-origin polysaccharides have received increasing attention across different fields of application, including biomedicine and biotechnology, because of their specific physicochemical and biological properties that have afforded the fabrication of a plethora of multifunctional devices for healthcare applications. More recently, marine raw materials from fisheries and aquaculture have emerged as a highly sustainable approach to convert marine biomass into added-value polysaccharides for human benefit. Nowadays, significant efforts have been made to combine such circular bio-based approach with cost-effective and environmentally-friendly technologies that enable the isolation of marine-origin polysaccharides up to the final construction of a biomedical device, thus developing an entirely sustainable pipeline. In this regard, the present review intends to provide an up-to-date outlook on the current green extraction methodologies of marine-origin polysaccharides and their molecular engineering toolbox for designing a multitude of biomaterial platforms for healthcare. Furthermore, we discuss how to foster circular bio-based approaches to pursue the further development of added-value biomedical devices, while preserving the marine ecosystem.
Collapse
Affiliation(s)
| | - João Borges
- CICECO–Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Fernando J. S. Correia
- Laboratory of Scientific Illustration, Department of Biology, University of Aveiro, Aveiro, Portugal
| | - Ricardo Calado
- Centre for Environmental and Marine Studies (CESAM), Department of Biology, University of Aveiro, Aveiro, Portugal
| | - João M. M. Rodrigues
- CICECO–Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - Sónia G. Patrício
- CICECO–Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| | - João F. Mano
- CICECO–Aveiro Institute of Materials, Department of Chemistry, University of Aveiro, Aveiro, Portugal
| |
Collapse
|
37
|
Li X, Xiong Y. Application of "Click" Chemistry in Biomedical Hydrogels. ACS OMEGA 2022; 7:36918-36928. [PMID: 36312409 PMCID: PMC9608400 DOI: 10.1021/acsomega.2c03931] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/23/2022] [Accepted: 09/21/2022] [Indexed: 06/12/2023]
Abstract
Since "click" chemistry was first reported in 2001, it has remained a popular research topic in the field of chemistry due to its high yield without byproducts, fast reaction rate, simple reaction, and biocompatibility. It has achieved good applications in various fields, especially for the preparation of hydrogels. The development of biomedicine presents new challenges and opportunities for hydrogels, and "click" chemistry provides a library of chemical tools for the preparation of various innovative hydrogels, including cell culture, 3D bioprinting, and drug release. This article summarizes several common "click" reactions, including copper-catalyzed azide-alkyne cycloaddition reactions, strain-promoted azide-alkyne cycloaddition (SPAAC) reaction, thiol-ene reaction, the Diels-Alder reaction, and the inverse electron demand Diels-Alder (IEDDA) reaction. We introduce the "click" reaction in the nucleic acid field to expand the concept of "click" chemistry. This article focuses on the application of "click" chemistry for preparing various types of biomedical hydrogels and highlights the advantages of "click" reactions for cross-linking to obtain hydrogels. This review also discusses applications of "click" chemistry outside the field of hydrogels, such as drug synthesis, targeted delivery, and surface modification, hydrogels have great application potential in these fields in the future and hopefully inspire other applications of hydrogels.
Collapse
Affiliation(s)
- Xin Li
- Department of Polymer Materials
and Engineering, Guizhou University, Guiyang 550025, P. R. China
| | - Yuzhu Xiong
- Department of Polymer Materials
and Engineering, Guizhou University, Guiyang 550025, P. R. China
| |
Collapse
|
38
|
González-Pérez F, Acosta S, Rütten S, Emonts C, Kopp A, Henke HW, Bruners P, Gries T, Rodríguez-Cabello JC, Jockenhoevel S, Fernández-Colino A. Biohybrid elastin-like venous valve with potential for in situ tissue engineering. Front Bioeng Biotechnol 2022; 10:988533. [PMID: 36213079 PMCID: PMC9532864 DOI: 10.3389/fbioe.2022.988533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2022] [Accepted: 08/22/2022] [Indexed: 11/15/2022] Open
Abstract
Chronic venous insufficiency (CVI) is a leading vascular disease whose clinical manifestations include varicose veins, edemas, venous ulcers, and venous hypertension, among others. Therapies targeting this medical issue are scarce, and so far, no single venous valve prosthesis is clinically available. Herein, we have designed a bi-leaflet transcatheter venous valve that consists of (i) elastin-like recombinamers, (ii) a textile mesh reinforcement, and (iii) a bioabsorbable magnesium stent structure. Mechanical characterization of the resulting biohybrid elastin-like venous valves (EVV) showed an anisotropic behavior equivalent to the native bovine saphenous vein valves and mechanical strength suitable for vascular implantation. The EVV also featured minimal hemolysis and platelet adhesion, besides actively supporting endothelialization in vitro, thus setting the basis for its application as an in situ tissue engineering implant. In addition, the hydrodynamic testing in a pulsatile bioreactor demonstrated excellent hemodynamic valve performance, with minimal regurgitation (<10%) and pressure drop (<5 mmHg). No stagnation points were detected and an in vitro simulated transcatheter delivery showed the ability of the venous valve to withstand the implantation procedure. These results present a promising concept of a biohybrid transcatheter venous valve as an off-the-shelf implant, with great potential to provide clinical solutions for CVI treatment.
Collapse
Affiliation(s)
- Fernando González-Pérez
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER-BBN, Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| | - Sergio Acosta
- Department of Biohybrid and Medical Textiles (BioTex), AME–Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
| | - Stephan Rütten
- Electron Microscopy Facility, Uniklinik RWTH Aachen, Aachen, Germany
| | - Caroline Emonts
- Institut für Textiltechnik Aachen (ITA), RWTH Aachen University, Aachen, Germany
| | | | | | - Philipp Bruners
- Klinik für Diagnostische and Interventionelle Radiologie, Universitätsklinikum Aachen, Aachen, Germany
| | - Thomas Gries
- Institut für Textiltechnik Aachen (ITA), RWTH Aachen University, Aachen, Germany
| | - J. Carlos Rodríguez-Cabello
- Bioforge Lab (Group for Advanced Materials and Nanobiotechnology), CIBER-BBN, Edificio LUCIA, Universidad de Valladolid, Valladolid, Spain
| | - Stefan Jockenhoevel
- Department of Biohybrid and Medical Textiles (BioTex), AME–Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
- AMIBM-Aachen-Maastricht-Institute for Biobased Materials, Maastricht University, Maastricht, Netherlands
- *Correspondence: Stefan Jockenhoevel, ; Alicia Fernández-Colino,
| | - Alicia Fernández-Colino
- Department of Biohybrid and Medical Textiles (BioTex), AME–Institute of Applied Medical Engineering, Helmholtz Institute, RWTH Aachen University, Aachen, Germany
- *Correspondence: Stefan Jockenhoevel, ; Alicia Fernández-Colino,
| |
Collapse
|
39
|
Sánchez-Cid P, Jiménez-Rosado M, Romero A, Pérez-Puyana V. Novel Trends in Hydrogel Development for Biomedical Applications: A Review. Polymers (Basel) 2022; 14:polym14153023. [PMID: 35893984 PMCID: PMC9370620 DOI: 10.3390/polym14153023] [Citation(s) in RCA: 114] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Revised: 07/21/2022] [Accepted: 07/22/2022] [Indexed: 12/11/2022] Open
Abstract
Nowadays, there are still numerous challenges for well-known biomedical applications, such as tissue engineering (TE), wound healing and controlled drug delivery, which must be faced and solved. Hydrogels have been proposed as excellent candidates for these applications, as they have promising properties for the mentioned applications, including biocompatibility, biodegradability, great absorption capacity and tunable mechanical properties. However, depending on the material or the manufacturing method, the resulting hydrogel may not be up to the specific task for which it is designed, thus there are different approaches proposed to enhance hydrogel performance for the requirements of the application in question. The main purpose of this review article was to summarize the most recent trends of hydrogel technology, going through the most used polymeric materials and the most popular hydrogel synthesis methods in recent years, including different strategies of enhancing hydrogels’ properties, such as cross-linking and the manufacture of composite hydrogels. In addition, the secondary objective of this review was to briefly discuss other novel applications of hydrogels that have been proposed in the past few years which have drawn a lot of attention.
Collapse
Affiliation(s)
| | | | - Alberto Romero
- Correspondence: (P.S.-C.); (A.R.); Tel.: +34-954557179 (A.R.)
| | | |
Collapse
|
40
|
Emerging Delivery Strategies of Platelet-Rich Plasma with Hydrogels for Wound Healing. ADVANCES IN POLYMER TECHNOLOGY 2022. [DOI: 10.1155/2022/5446291] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Platelet-rich plasma (PRP), a platelet-rich plasma concentrate obtained from whole blood, has been widely used to treat wounds due to its high contents of growth factors that can not only play a role in the hemostasis, repair, and anti-infection of wounds but also promote cell proliferation, maturation, and angiogenesis. However, after PRP activation, its clinical effect was limited because of burst and uncontrolled release of growth factors and poor mechanical properties of PRP gels. In recent years, increasing attention has been moved to the loading and sustained release of growth factors in PRP by polymeric carriers. Hydrogels, as an interesting carrier, enable controlled delivery of growth factors by structural designs. Moreover, using hydrogels to encapsulate PRP is favorable to controlling the mechanical properties and water maintenance of PRP gels, which can provide a stable and moist wound repair environment to promote coordinated operations of skin tissue cells and cytokines as well as wound healing. In this review, the state of the art of hydrogels that have been used to load PRP for wound treatments is introduced, and further prospects in the research area are proposed.
Collapse
|
41
|
Schuiringa GH, Mihajlovic M, van Donkelaar CC, Vermonden T, Ito K. Creating a Functional Biomimetic Cartilage Implant Using Hydrogels Based on Methacrylated Chondroitin Sulfate and Hyaluronic Acid. Gels 2022; 8:gels8070457. [PMID: 35877542 PMCID: PMC9315485 DOI: 10.3390/gels8070457] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2022] [Revised: 07/19/2022] [Accepted: 07/20/2022] [Indexed: 02/04/2023] Open
Abstract
The load-bearing function of articular cartilage tissue contrasts with the poor load-bearing capacity of most soft hydrogels used for its regeneration. The present study explores whether a hydrogel based on the methacrylated natural polymers chondroitin sulfate (CSMA) and hyaluronic acid (HAMA), injected into warp-knitted spacer fabrics, could be used to create a biomimetic construct with cartilage-like mechanical properties. The swelling ratio of the combined CSMA/HAMA hydrogels in the first 20 days was higher for hydrogels with a higher CSMA concentration, and these hydrogels also degraded quicker, whereas those with a 1.33 wt% of HAMA were stable for more than 120 days. When confined by a polyamide 6 (PA6) spacer fabric, the volumetric swelling of the combined CSMA/HAMA gels (10 wt%, 6.5 × CSMA:HAMA ratio) was reduced by ~53%. Both the apparent peak and the equilibrium modulus significantly increased in the PA6-restricted constructs compared to the free-swelling hydrogels after 28 days of swelling, and no significant differences in the moduli and time constant compared to native bovine cartilage were observed. Moreover, the cell viability in the CSMA/HAMA PA6 constructs was comparable to that in gelatin–methacrylamide (GelMA) PA6 constructs at one day after polymerization. These results suggest that using a HydroSpacer construct with an extracellular matrix (ECM)-like biopolymer-based hydrogel is a promising approach for mimicking the load-bearing properties of native cartilage.
Collapse
Affiliation(s)
- Gerke H. Schuiringa
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Gem-Z 1.106, P.O. Box 513, 5600 MB Eindhoven, The Netherlands; (G.H.S.); (M.M.); (K.I.)
| | - Marko Mihajlovic
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Gem-Z 1.106, P.O. Box 513, 5600 MB Eindhoven, The Netherlands; (G.H.S.); (M.M.); (K.I.)
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Science for Life, Utrecht University, 3508 TB Utrecht, The Netherlands;
| | - Corrinus C. van Donkelaar
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Gem-Z 1.106, P.O. Box 513, 5600 MB Eindhoven, The Netherlands; (G.H.S.); (M.M.); (K.I.)
- Correspondence:
| | - Tina Vermonden
- Department of Pharmaceutics, Utrecht Institute for Pharmaceutical Sciences (UIPS), Science for Life, Utrecht University, 3508 TB Utrecht, The Netherlands;
| | - Keita Ito
- Orthopaedic Biomechanics, Department of Biomedical Engineering, Eindhoven University of Technology, Gem-Z 1.106, P.O. Box 513, 5600 MB Eindhoven, The Netherlands; (G.H.S.); (M.M.); (K.I.)
| |
Collapse
|
42
|
Aparici Herraiz I, Caires HR, Castillo-Fernández Ó, Sima N, Méndez-Mora L, Risueño RM, Sattabongkot J, Roobsoong W, Hernández-Machado A, Fernandez-Becerra C, Barrias CC, del Portillo HA. Advancing Key Gaps in the Knowledge of Plasmodium vivax Cryptic Infections Using Humanized Mouse Models and Organs-on-Chips. Front Cell Infect Microbiol 2022; 12:920204. [PMID: 35873153 PMCID: PMC9302440 DOI: 10.3389/fcimb.2022.920204] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Accepted: 06/06/2022] [Indexed: 11/13/2022] Open
Abstract
Plasmodium vivax is the most widely distributed human malaria parasite representing 36.3% of disease burden in the South-East Asia region and the most predominant species in the region of the Americas. Recent estimates indicate that 3.3 billion of people are under risk of infection with circa 7 million clinical cases reported each year. This burden is certainly underestimated as the vast majority of chronic infections are asymptomatic. For centuries, it has been widely accepted that the only source of cryptic parasites is the liver dormant stages known as hypnozoites. However, recent evidence indicates that niches outside the liver, in particular in the spleen and the bone marrow, can represent a major source of cryptic chronic erythrocytic infections. The origin of such chronic infections is highly controversial as many key knowledge gaps remain unanswered. Yet, as parasites in these niches seem to be sheltered from immune response and antimalarial drugs, research on this area should be reinforced if elimination of malaria is to be achieved. Due to ethical and technical considerations, working with the liver, bone marrow and spleen from natural infections is very difficult. Recent advances in the development of humanized mouse models and organs-on-a-chip models, offer novel technological frontiers to study human diseases, vaccine validation and drug discovery. Here, we review current data of these frontier technologies in malaria, highlighting major challenges ahead to study P. vivax cryptic niches, which perpetuate transmission and burden.
Collapse
Affiliation(s)
- Iris Aparici Herraiz
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Hugo R. Caires
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Óscar Castillo-Fernández
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
| | - Núria Sima
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Lourdes Méndez-Mora
- Department of Condensed Matter Physics, University of Barcelona (UB), Barcelona, Spain
| | - Ruth M. Risueño
- Josep Carreras Leukaemia Research Institute (IJC), Barcelona, Spain
| | - Jetsumon Sattabongkot
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Wanlapa Roobsoong
- Mahidol Vivax Research Unit, Faculty of Tropical Medicine, Mahidol University, Bangkok, Thailand
| | - Aurora Hernández-Machado
- Institute of Nanoscience and Nanotechnology (IN2UB), University of Barcelona, Barcelona, Spain
- Department of Condensed Matter Physics, University of Barcelona (UB), Barcelona, Spain
- Centre de Recerca Matemàtica (CRM), Barcelona, Spain
| | - Carmen Fernandez-Becerra
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
| | - Cristina C. Barrias
- i3S - Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
- INEB – Instituto de Engenharia Biomédica, Universidade do Porto, Porto, Portugal
- ICBAS – Instituto de Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
| | - Hernando A. del Portillo
- Barcelona Institute for Global Health (ISGlobal), Hospital Clínic - Universitat de Barcelona, Barcelona, Spain
- Institut d’Investigació en Ciències de la Salut Germans Trias i Pujol, Badalona, Spain
- Institució Catalana de Recerca i Estudis Avançats (ICREA), Barcelona, Spain
- *Correspondence: Hernando A. del Portillo,
| |
Collapse
|
43
|
Manferdini C, Trucco D, Saleh Y, Gabusi E, Dolzani P, Lenzi E, Vannozzi L, Ricotti L, Lisignoli G. RGD-Functionalized Hydrogel Supports the Chondrogenic Commitment of Adipose Mesenchymal Stromal Cells. Gels 2022; 8:382. [PMID: 35735726 PMCID: PMC9222613 DOI: 10.3390/gels8060382] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 06/09/2022] [Accepted: 06/13/2022] [Indexed: 02/01/2023] Open
Abstract
Articular cartilage is known to have limited intrinsic self-healing capacity when a defect or a degeneration process occurs. Hydrogels represent promising biomaterials for cell encapsulation and injection in cartilage defects by creating an environment that mimics the cartilage extracellular matrix. The aim of this study is the analysis of two different concentrations (1:1 and 1:2) of VitroGel® (VG) hydrogels without (VG-3D) and with arginine-glycine-aspartic acid (RGD) motifs, (VG-RGD), verifying their ability to support chondrogenic differentiation of encapsulated human adipose mesenchymal stromal cells (hASCs). We analyzed the hydrogel properties in terms of rheometric measurements, cell viability, cytotoxicity, and the expression of chondrogenic markers using gene expression, histology, and immunohistochemical tests. We highlighted a shear-thinning behavior of both hydrogels, which showed good injectability. We demonstrated a good morphology and high viability of hASCs in both hydrogels. VG-RGD 1:2 hydrogels were the most effective, both at the gene and protein levels, to support the expression of the typical chondrogenic markers, including collagen type 2, SOX9, aggrecan, glycosaminoglycan, and cartilage oligomeric matrix protein and to decrease the proliferation marker MKI67 and the fibrotic marker collagen type 1. This study demonstrated that both hydrogels, at different concentrations, and the presence of RGD motifs, significantly contributed to the chondrogenic commitment of the laden hASCs.
Collapse
Affiliation(s)
- Cristina Manferdini
- IRCCS Istituto Ortopedico Rizzoli, SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, 40136 Bologna, Italy; (C.M.); (D.T.); (Y.S.); (E.G.); (P.D.); (E.L.)
| | - Diego Trucco
- IRCCS Istituto Ortopedico Rizzoli, SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, 40136 Bologna, Italy; (C.M.); (D.T.); (Y.S.); (E.G.); (P.D.); (E.L.)
- The BioRobotics Institute, Scuola Superiore Sant’Anna, 56025 Pisa, Italy; (L.V.); (L.R.)
- Department of Excellence in Robotics & AI, Scuola Superiore Sant’Anna, 56025 Pisa, Italy
| | - Yasmin Saleh
- IRCCS Istituto Ortopedico Rizzoli, SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, 40136 Bologna, Italy; (C.M.); (D.T.); (Y.S.); (E.G.); (P.D.); (E.L.)
| | - Elena Gabusi
- IRCCS Istituto Ortopedico Rizzoli, SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, 40136 Bologna, Italy; (C.M.); (D.T.); (Y.S.); (E.G.); (P.D.); (E.L.)
| | - Paolo Dolzani
- IRCCS Istituto Ortopedico Rizzoli, SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, 40136 Bologna, Italy; (C.M.); (D.T.); (Y.S.); (E.G.); (P.D.); (E.L.)
| | - Enrico Lenzi
- IRCCS Istituto Ortopedico Rizzoli, SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, 40136 Bologna, Italy; (C.M.); (D.T.); (Y.S.); (E.G.); (P.D.); (E.L.)
| | - Lorenzo Vannozzi
- The BioRobotics Institute, Scuola Superiore Sant’Anna, 56025 Pisa, Italy; (L.V.); (L.R.)
- Department of Excellence in Robotics & AI, Scuola Superiore Sant’Anna, 56025 Pisa, Italy
| | - Leonardo Ricotti
- The BioRobotics Institute, Scuola Superiore Sant’Anna, 56025 Pisa, Italy; (L.V.); (L.R.)
- Department of Excellence in Robotics & AI, Scuola Superiore Sant’Anna, 56025 Pisa, Italy
| | - Gina Lisignoli
- IRCCS Istituto Ortopedico Rizzoli, SC Laboratorio di Immunoreumatologia e Rigenerazione Tissutale, 40136 Bologna, Italy; (C.M.); (D.T.); (Y.S.); (E.G.); (P.D.); (E.L.)
| |
Collapse
|
44
|
Demirci G, Niedźwiedź MJ, Kantor-Malujdy N, El Fray M. Elastomer-Hydrogel Systems: From Bio-Inspired Interfaces to Medical Applications. Polymers (Basel) 2022; 14:1822. [PMID: 35566990 PMCID: PMC9104885 DOI: 10.3390/polym14091822] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 04/13/2022] [Accepted: 04/27/2022] [Indexed: 12/10/2022] Open
Abstract
Novel advanced biomaterials have recently gained great attention, especially in minimally invasive surgical techniques. By applying sophisticated design and engineering methods, various elastomer-hydrogel systems (EHS) with outstanding performance have been developed in the last decades. These systems composed of elastomers and hydrogels are very attractive due to their high biocompatibility, injectability, controlled porosity and often antimicrobial properties. Moreover, their elastomeric properties and bioadhesiveness are making them suitable for soft tissue engineering. Herein, we present the advances in the current state-of-the-art design principles and strategies for strong interface formation inspired by nature (bio-inspiration), the diverse properties and applications of elastomer-hydrogel systems in different medical fields, in particular, in tissue engineering. The functionalities of these systems, including adhesive properties, injectability, antimicrobial properties and degradability, applicable to tissue engineering will be discussed in a context of future efforts towards the development of advanced biomaterials.
Collapse
Affiliation(s)
| | | | | | - Miroslawa El Fray
- Department of Polymer and Biomaterials Science, Faculty of Chemical Technology and Engineering, West Pomeranian University of Technology in Szczecin, Al. Piastów 45, 70-311 Szczecin, Poland; (G.D.); (M.J.N.); (N.K.-M.)
| |
Collapse
|
45
|
Hybrid Core-Shell Polymer Scaffold for Bone Tissue Regeneration. Int J Mol Sci 2022; 23:ijms23094533. [PMID: 35562923 PMCID: PMC9101363 DOI: 10.3390/ijms23094533] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2022] [Revised: 04/13/2022] [Accepted: 04/17/2022] [Indexed: 12/17/2022] Open
Abstract
A great promise for tissue engineering is represented by scaffolds that host stem cells during proliferation and differentiation and simultaneously replace damaged tissue while maintaining the main vital functions. In this paper, a novel process was adopted to develop composite scaffolds with a core-shell structure for bone tissue regeneration, in which the core has the main function of temporary mechanical support, and the shell enhances biocompatibility and provides bioactive properties. An interconnected porous core was safely obtained, avoiding solvents or other chemical issues, by blending poly(lactic acid), poly(ε-caprolactone) and leachable superabsorbent polymer particles. After particle leaching in water, the core was grafted with a gelatin/chitosan hydrogel shell to create a cell-friendly bioactive environment within its pores. The physicochemical, morphological, and mechanical characterization of the hybrid structure and of its component materials was carried out by means of infrared spectroscopy, thermogravimetric analysis, scanning electron microscopy, and mechanical testing under different loading conditions. These hybrid polymer devices were found to closely mimic both the morphology and the stiffness of bones. In addition, in vitro studies showed that the core-shell scaffolds are efficiently seeded by human mesenchymal stromal cells, which remain viable, proliferate, and are capable of differentiating towards the osteogenic phenotype if adequately stimulated.
Collapse
|
46
|
Swelling, Protein Adsorption, and Biocompatibility In Vitro of Gel Beads Prepared from Pectin of Hogweed Heracleum sosnówskyi Manden in Comparison with Gel Beads from Apple Pectin. Int J Mol Sci 2022; 23:ijms23063388. [PMID: 35328806 PMCID: PMC8954847 DOI: 10.3390/ijms23063388] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2022] [Revised: 03/16/2022] [Accepted: 03/19/2022] [Indexed: 02/04/2023] Open
Abstract
The study aims to develop gel beads with improved functional properties and biocompatibility from hogweed (HS) pectin. HS4 and AP4 gel beads were prepared from the HS pectin and apple pectin (AP) using gelling with calcium ions. HS4 and AP4 gel beads swelled in PBS in dependence on pH. The swelling degree of HS4 and AP4 gel beads was 191 and 136%, respectively, in PBS at pH 7.4. The hardness of HS4 and AP4 gel beads reduced 8.2 and 60 times, respectively, compared with the initial value after 24 h incubation. Both pectin gel beads swelled less in Hanks’ solution than in PBS and swelled less in Hanks’ solution containing peritoneal macrophages than in cell-free Hanks’ solution. Serum protein adsorption by HS4 and AP4 gel beads was 118 ± 44 and 196 ± 68 μg/cm2 after 24 h of incubation. Both pectin gel beads demonstrated low rates of hemolysis and complement activation. However, HS4 gel beads inhibited the LPS-stimulated secretion of TNF-α and the expression of TLR4 and NF-κB by macrophages, whereas AP4 gel beads stimulated the inflammatory response of macrophages. HS4 gel beads adsorbed 1.3 times more LPS and adhered to 1.6 times more macrophages than AP4 gel beads. Thus, HS pectin gel has advantages over AP gel concerning swelling behavior, protein adsorption, and biocompatibility.
Collapse
|
47
|
Zou M, Chi J, Jiang Z, Zhang W, Hu H, Ju R, Liu C, Xu T, Wang S, Feng Z, Liu W, Han B. Functional thermosensitive hydrogels based on chitin as RIN-m5F cell carrier for the treatment of diabetes. Int J Biol Macromol 2022; 206:453-466. [PMID: 35247418 DOI: 10.1016/j.ijbiomac.2022.02.175] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 02/15/2022] [Accepted: 02/27/2022] [Indexed: 12/19/2022]
Abstract
Herein, the thermosensitive hydroxypropyl chitin (HPCT) hydrogel was prepared and the chemical structures, microstructures, rheological properties and degradation in vitro were investigated. The HPCT hydrogel possessed satisfactory biocompatibility in mouse fibroblast cells and Sprague Dawley rats. On the other hand, N-acetylglucosamine (NAG) and carboxymethyl chitosan (CMCS) provided favorable capacity for promoting cell proliferation, delaying cell apoptosis, and facilitating the insulin secretion of rat pancreatic beta cells (RIN-m5F) in three-dimensional culture. Most importantly, the effects of HPCT/NAG and HPCT/CMCS thermosensitive hydrogels as RIN-m5F cells carriers were evaluated via injection into different areas of diabetic rats. Our results demonstrated that HPCT/NAG and HPCT/CMCS hydrogels loaded RIN-m5F cells could keep cells survival, maintain insulin secretion and reduce blood glucose for one week. Overall, the functional thermosensitive hydrogels based on HPCT were effective cell carriers for RIN-m5F cells and might provide novel strategy for the treatment of diabetes via cell engineering.
Collapse
Affiliation(s)
- Mingyu Zou
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Jinhua Chi
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Zhiwen Jiang
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, PR China
| | - Wei Zhang
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Huiwen Hu
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Ruibao Ju
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Chenqi Liu
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Tianjiao Xu
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Shuo Wang
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Zhilong Feng
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Wanshun Liu
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China
| | - Baoqin Han
- Laboratory of Biochemistry and Biomedical Materials, College of Marine Life Sciences, Ocean University of China, Qingdao 266003, PR China; Laboratory for Marine Drugs and Bioproducts of Qingdao National Laboratory for Marine Science and Technology, Qingdao 266235, PR China.
| |
Collapse
|
48
|
Radulescu DM, Neacsu IA, Grumezescu AM, Andronescu E. New Insights of Scaffolds Based on Hydrogels in Tissue Engineering. Polymers (Basel) 2022; 14:799. [PMID: 35215710 PMCID: PMC8875010 DOI: 10.3390/polym14040799] [Citation(s) in RCA: 78] [Impact Index Per Article: 26.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/11/2022] [Accepted: 02/16/2022] [Indexed: 02/04/2023] Open
Abstract
In recent years, biomaterials development and characterization for new applications in regenerative medicine or controlled release represent one of the biggest challenges. Tissue engineering is one of the most intensively studied domain where hydrogels are considered optimum applications in the biomedical field. The delicate nature of hydrogels and their low mechanical strength limit their exploitation in tissue engineering. Hence, developing new, stronger, and more stable hydrogels with increased biocompatibility, is essential. However, both natural and synthetic polymers possess many limitations. Hydrogels based on natural polymers offer particularly high biocompatibility and biodegradability, low immunogenicity, excellent cytocompatibility, variable, and controllable solubility. At the same time, they have poor mechanical properties, high production costs, and low reproducibility. Synthetic polymers come to their aid through superior mechanical strength, high reproducibility, reduced costs, and the ability to regulate their composition to improve processes such as hydrolysis or biodegradation over variable periods. The development of hydrogels based on mixtures of synthetic and natural polymers can lead to the optimization of their properties to obtain ideal scaffolds. Also, incorporating different nanoparticles can improve the hydrogel's stability and obtain several biological effects. In this regard, essential oils and drug molecules facilitate the desired biological effect or even produce a synergistic effect. This study's main purpose is to establish the main properties needed to develop sustainable polymeric scaffolds. These scaffolds can be applied in tissue engineering to improve the tissue regeneration process without producing other side effects to the environment.
Collapse
Affiliation(s)
- Denisa-Maria Radulescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 011061 Bucharest, Romania; (D.-M.R.); (A.-M.G.); (E.A.)
| | - Ionela Andreea Neacsu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 011061 Bucharest, Romania; (D.-M.R.); (A.-M.G.); (E.A.)
- Academy of Romanian Scientists, 54 Independentei, 050094 Bucharest, Romania
- National Research Center for Micro and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 060042 Bucharest, Romania
| | - Alexandru-Mihai Grumezescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 011061 Bucharest, Romania; (D.-M.R.); (A.-M.G.); (E.A.)
- Academy of Romanian Scientists, 54 Independentei, 050094 Bucharest, Romania
- Research Institute of the University of Bucharest (ICUB), University of Bucharest, 050657 Bucharest, Romania
| | - Ecaterina Andronescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 011061 Bucharest, Romania; (D.-M.R.); (A.-M.G.); (E.A.)
- Academy of Romanian Scientists, 54 Independentei, 050094 Bucharest, Romania
- National Research Center for Micro and Nanomaterials, Faculty of Applied Chemistry and Materials Science, University Politehnica of Bucharest, 060042 Bucharest, Romania
| |
Collapse
|
49
|
Polysaccharide hydrogels: Functionalization, construction and served as scaffold for tissue engineering. Carbohydr Polym 2022; 278:118952. [PMID: 34973769 DOI: 10.1016/j.carbpol.2021.118952] [Citation(s) in RCA: 112] [Impact Index Per Article: 37.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2021] [Revised: 11/07/2021] [Accepted: 11/26/2021] [Indexed: 02/07/2023]
Abstract
Polysaccharide hydrogels have been widely utilized in tissue engineering. They interact with the organismal environments, modulating the cargos release and realizing of long-term survival and activations of living cells. In this review, the potential strategies for modification of polysaccharides were introduced firstly. It is not only used to functionalize the polysaccharides for the consequent formation of hydrogels, but also used to introduce versatile side groups for the regulation of cell behavior. Then, techniques and underlying mechanisms in inducing the formation of hydrogels by polysaccharides or their derivatives are briefly summarized. Finally, the applications of polysaccharide hydrogels in vivo, mainly focus on the performance for alleviation of foreign-body response (FBR) and as cell scaffolds for tissue regeneration, are exemplified. In addition, the perspectives and challenges for further research are addressed. It aims to provide a comprehensive framework about the potentials and challenges that the polysaccharide hydrogels confronting in tissue engineering.
Collapse
|
50
|
Characterization and Biocompatibility Properties In Vitro of Gel Beads Based on the Pectin and κ-Carrageenan. Mar Drugs 2022; 20:md20020094. [PMID: 35200624 PMCID: PMC8878971 DOI: 10.3390/md20020094] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2021] [Revised: 01/19/2022] [Accepted: 01/20/2022] [Indexed: 12/23/2022] Open
Abstract
This study aimed to investigate the influence of kappa (κ)-carrageenan on the initial stages of the foreign body response against pectin gel. Pectin-carrageenan (P-Car) gel beads were prepared from the apple pectin and κ-carrageenan using gelling with calcium ions. The inclusion of 0.5% κ-carrageenan (Car0.5) in the 1.5 (P1.5) and 2% pectin (P2) gel formulations decreased the gel strength by 2.5 times. Car0.5 was found to increase the swelling of P2 gel beads in the cell culture medium. P2 gel beads adsorbed 30–42 mg/g of bovine serum albumin (BSA) depending on pH. P2-Car0.2, P2-Car0.5, and P1.5-Car0.5 beads reduced BSA adsorption by 3.1, 5.2, and 4.0 times compared to P2 beads, respectively, at pH 7. The P1.5-Car0.5 beads activated complement and induced the haemolysis less than gel beads of pure pectin. Moreover, P1.5-Car0.5 gel beads allowed less adhesion of mouse peritoneal macrophages, TNF-α production, and NF-κB activation than the pure pectin gel beads. There were no differences in TLR4 and ICAM-1 levels in macrophages treated with P and P-Car gel beads. P2-Car0.5 hydrogel demonstrated lower adhesion to serous membrane than P2 hydrogel. Thus, the data obtained indicate that the inclusion of κ-carrageenan in the apple pectin gel improves its biocompatibility.
Collapse
|