1
|
Wijerathna-Yapa A, Isaac KS, Combe M, Hume S, Sokolenko S. Re-imagining human cell culture media: Challenges, innovations, and future directions. Biotechnol Adv 2025; 81:108564. [PMID: 40101881 DOI: 10.1016/j.biotechadv.2025.108564] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/19/2025] [Accepted: 03/15/2025] [Indexed: 03/20/2025]
Abstract
The development of optimized culture media is pivotal to advancements in human cell culture, underpinning progress in regenerative medicine, cell therapies, and personalized medicine. While foundational formulations like Eagle's Minimum Essential Medium (MEM) and Dulbecco's Modified Eagle Medium (DMEM) have historically enabled significant biological research, these media were primarily designed for non-human cells and do not adequately address the unique metabolic and functional requirements of human cells. This review examines the evolution of cell culture media, identifying persistent challenges in reproducibility, scalability, and ethical concerns, particularly regarding the reliance on animal-derived components such as fetal bovine serum (FBS). We highlight innovations in serum-free and chemically defined media that offer promising alternatives by enhancing consistency, aligning with Good Manufacturing Practices, and addressing ethical concerns. Emerging approaches, including omics-based profiling, high-throughput screening, and artificial intelligence (AI)-driven media design, are reshaping media optimization by enabling precise tailoring to the needs of specific human cell types and patient-derived cells. Furthermore, we discuss economic and regulatory challenges, emphasizing the need for cost-effective and scalable solutions to facilitate clinical translation. Looking forward, integrating advanced biotechnological tools such as 3D bioprinting, organ-on-a-chip systems, and personalized media formulations presents a transformative opportunity for human cell culture. These innovations, aligned with ethical and clinical standards, can drive the development of human-specific media systems that ensure reproducibility, scalability, and enhanced therapeutic potential, thereby advancing both research and clinical applications.
Collapse
Affiliation(s)
- Akila Wijerathna-Yapa
- Department of Process Engineering and Applied Science, Faculty of Engineering, Dalhousie University, PO Box 15000, Halifax, Nova Scotia B3H 4R2, Canada
| | - Kathy Sharon Isaac
- Department of Process Engineering and Applied Science, Faculty of Engineering, Dalhousie University, PO Box 15000, Halifax, Nova Scotia B3H 4R2, Canada
| | - Michelle Combe
- Department of Process Engineering and Applied Science, Faculty of Engineering, Dalhousie University, PO Box 15000, Halifax, Nova Scotia B3H 4R2, Canada
| | - Samuel Hume
- Medical Sciences Division, University of Oxford, Oxford OX3 9DU, UK
| | - Stanislav Sokolenko
- Department of Process Engineering and Applied Science, Faculty of Engineering, Dalhousie University, PO Box 15000, Halifax, Nova Scotia B3H 4R2, Canada.
| |
Collapse
|
2
|
Stealey S, Dharmesh E, Gaharwar AK, Rudra JS, Zustiak SP. Protein structure and bioactivity upon adsorption and desorption from nanosilicate sustained release delivery devices. NANOSCALE 2025. [PMID: 40387592 DOI: 10.1039/d5nr00861a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Sustained local delivery of biologics via hydrogel carriers is a promising approach to enhance protein safety and efficacy. The addition of nanoparticles to polymeric hydrogels has been shown to further improve the retention and delivery kinetics of biologics. Specifically, nanoparticles with high surface area, such as nanosilicates, have shown potential for complexing with biologics to enable highly tunable release profiles. Here, LAPONITE® XLG nanosilicate (NS) was examined due to its platelet-like structure with negatively charged faces and positively charged edges. Our previous results have shown NS to greatly slow the release of model proteins from poly(ethylene glycol) (PEG) hydrogels due to NS-protein complexation. This work aims to determine the structure and stability of several NS-protein complexes, as well as protein activity and structure upon complexation. Binding affinity assays revealed a strong correlation between affinity and protein charge, with positively charged proteins being more attracted to NS. Proteins were shown to unfold in the presence of NS in solution, leading to a partial loss in bioactivity. However, this unfolding was determined to be temporary, as proteins released from PEG-NS hydrogels recovered secondary structure and bioactivity. Binding to NS also provided some protection against protein denaturant guanidine thiocyanate. Through understanding the interactions between proteins and NS, this study paves the way for the application of these NS-protein complexes as tunable, sustained-release delivery devices.
Collapse
Affiliation(s)
- Samuel Stealey
- Department of Biomedical Engineering, Saint Louis University, Saint Louis, MO, USA.
| | - Ether Dharmesh
- Department of Biomedical Engineering, Saint Louis University, Saint Louis, MO, USA.
| | - Akhilesh K Gaharwar
- Department of Biomedical Engineering, Texas A&M University, College Station, TX, USA
| | - Jai S Rudra
- Department of Biomedical Engineering, Washington University in Saint Louis, Saint Louis, MO, USA
| | - Silviya P Zustiak
- Department of Biomedical Engineering, Saint Louis University, Saint Louis, MO, USA.
| |
Collapse
|
3
|
Ghosh S, Mohol SS, Datta A, Pandey PM, Kulshreshtha R, Nandan B, Hakkarainen M, Srivastava RK. Leveraging the Shape Fidelity of 3D Printed Bone Scaffolds Through Architectural Tailoring of an Emulsion Ink: A Combined Experimental and Computational Analysis. Adv Healthc Mater 2025; 14:e2404866. [PMID: 40159818 DOI: 10.1002/adhm.202404866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Revised: 03/09/2025] [Indexed: 04/02/2025]
Abstract
Hierarchical porous, bioactive, and biocompatible scaffolds with customizable multi-functionality are promising alternatives for autografts and allografts in bone tissue engineering. Combining high internal phase emulsion (HIPE) templating with additive manufacturing provides possibilities to produce such multiscale porous scaffolds. 3D printing of HIPE remains a challenging task due to the intense phase separation under high shear extrusion and reported printability (Pr) of either less than or greater than 1. Tuning viscoelastic properties of emulsion is therefore required to achieve a Pr ≈1. This study addresses these issues by preparing Pickering HIPEs using dual networks with synergistic viscous and elastic properties, stabilized by Cloisite 30B interphase. This configuration enhances viscoelasticity and achieves Pr values close to 1 (0.98-1.02). The printed scaffolds exhibit trabecular bone-like, hierarchical interconnected porosity (77%-86%). Computational simulations accurately predict the mechanical, biological, and degradation behavior. Functionalization with Cissus quadrangularis bioactivates the scaffolds, demonstrates in vivo biocompatibility, promotes MC3T3-E1 adhesion, and proliferation, accelerates osteogenesis, and reduces oxidative stress compared to neat PCL scaffolds. This work introduces a facile strategy for "engineering printability" to produce regenerative materials with hierarchical design and holds the potential for developing optimized bone tissue engineering scaffolds.
Collapse
Affiliation(s)
- Sagnik Ghosh
- Department of Textile and Fibre Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Shubham Shankar Mohol
- Department of Mechanical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Anupama Datta
- Institute of Nuclear Medicine and Allied Sciences, Defence Research and Development Organization, Brig S. K. Mazumdar Marg, Delhi, 110054, India
| | - Pulak Mohan Pandey
- Department of Mechanical Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Ritu Kulshreshtha
- Department of Biochemical Engineering and Biotechnology, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Bhanu Nandan
- Department of Textile and Fibre Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| | - Minna Hakkarainen
- KTH Royal Institute of Technology, Department of Fibre and Polymer Technology, Teknikringen 58, Stockholm, 100 44, Sweden
| | - Rajiv K Srivastava
- Department of Textile and Fibre Engineering, Indian Institute of Technology Delhi, Hauz Khas, New Delhi, 110016, India
| |
Collapse
|
4
|
Hu Y, Wang Y, Liu Y, Dai J, Wang J, Ju C. Injectable laponite nanocomposite hydrogel with synergistic antibacterial and odontogenic activity for endodontic regeneration. Colloids Surf B Biointerfaces 2025; 253:114745. [PMID: 40318397 DOI: 10.1016/j.colsurfb.2025.114745] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/23/2025] [Accepted: 04/26/2025] [Indexed: 05/07/2025]
Abstract
Persistent pulpitis often leads to irreversible dentin defects and pulp necrosis, posing significant challenges for functional dental restoration. To address the critical limitation of insufficient odontogenic differentiation in dental pulp stem cells (DPSCs), we develop an injectable photo-crosslinking methacrylated hyaluronic acid hydrogel (PLSr2+@HAMA) containing laponite-based ternary nanocomposites which self-assemble by laponite, strontium ions (Sr2+), and antibacterial peptide P-113. We demonstrate that PLSr2+@HAMA features three distinctive advantages: (1) Unique injectability enabling minimally invasive delivery and UV-triggered in situ gelation for anatomical adaptation; (2) Sustained co-release of antimicrobial peptide P-113 and bioactive strontium ions through laponite-mediated orchestrated delivery; (3) Multi-ion and P-113 synergistic action achieving simultaneous infection control and dentin regeneration in several in vitro and in vivo models, promoting endodontic regeneration. By integrating antibacterial action with DPSCs modulation in a single minimally invasive platform, this strategy pioneers a new paradigm for comprehensive endodontic regeneration, demonstrating significant translational potential for inflammatory dental pulp therapy.
Collapse
Affiliation(s)
- Yijie Hu
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210038, China
| | - Yongbin Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210038, China
| | - Yuqing Liu
- Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, 210029, China; State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, 210029, China
| | - Jiayun Dai
- Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, 210029, China; State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, 210029, China
| | - Juan Wang
- Department of Endodontics, The Affiliated Stomatological Hospital of Nanjing Medical University, 210029, China; State Key Laboratory Cultivation Base of Research, Prevention and Treatment for Oral Diseases, 210029, China.
| | - Caoyun Ju
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, 210038, China.
| |
Collapse
|
5
|
Zheng M, Tan Y, Yuan Y, Wang H, Yang H. Regulatory Mechanisms of Cytotoxicity and Hemocompatibility Induced by Phase Transformation of Kaolinite Nanocarrier. J Phys Chem Lett 2025; 16:3945-3953. [PMID: 40211690 DOI: 10.1021/acs.jpclett.5c00793] [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: 04/25/2025]
Abstract
Revealing the regulatory mechanism underlying the cytotoxicity and hemocompatibility of nanocarriers is crucial for their biofunctionalized design and practical application in nanotherapeutics. However, the microstructural and physicochemical properties of nanocarriers inevitably change during the modification process, and the impact of these changes on biosafety remains unclear. Herein, we investigate the effects of phase transformation of kaolinite (Kaol) nanoclay on its biosafety. Experimental results indicate that the adjoint dehydration, dehydroxylation, and disaggregation during phase transformation of Kaol could alter the mode of interaction at the cell interface and mitigate damage to cell membranes. Furthermore, the heat-treated Kaol exhibits reduced hemolysis while maintaining red blood cell adhesion and pro-coagulant functions, without affecting the structure of plasma proteins. Collectively, this study could provide a novel insight into the correlation between Kaol microstructure and biosafety.
Collapse
Affiliation(s)
- Meng Zheng
- School of Earth Sciences, China University of Geosciences, Wuhan 430074, China
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, China University of Geosciences, Wuhan 430074, China
- Laboratory of Advanced Mineral Materials, China University of Geosciences, Wuhan 430074, China
| | - Ya Tan
- Hunan Key Laboratory of Mineral Materials and Application, School of Minerals Processing and Bioengineering, Central South University, Changsha 410083, China
| | - Yiting Yuan
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, China University of Geosciences, Wuhan 430074, China
- Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
- Laboratory of Advanced Mineral Materials, China University of Geosciences, Wuhan 430074, China
| | - Hao Wang
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, China University of Geosciences, Wuhan 430074, China
- Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
- Laboratory of Advanced Mineral Materials, China University of Geosciences, Wuhan 430074, China
| | - Huaming Yang
- School of Earth Sciences, China University of Geosciences, Wuhan 430074, China
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, China University of Geosciences, Wuhan 430074, China
- Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan 430074, China
- Laboratory of Advanced Mineral Materials, China University of Geosciences, Wuhan 430074, China
- Hunan Key Laboratory of Mineral Materials and Application, School of Minerals Processing and Bioengineering, Central South University, Changsha 410083, China
| |
Collapse
|
6
|
Hua W, Zhang C, Cui H, Mitchell K, Hensley DK, Chen J, Do C, Raymond L, Coulter R, Bandala E, Rubbi F, Chai G, Zhang Z, Liao Y, Zhao D, Wang Y, Gaharwar AK, Jin Y. High-Speed Embedded Ink Writing of Anatomic-Size Organ Constructs. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2405980. [PMID: 39932855 PMCID: PMC11967790 DOI: 10.1002/advs.202405980] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/02/2024] [Revised: 07/29/2024] [Indexed: 02/13/2025]
Abstract
Embedded ink writing (EIW) is an emerging 3D printing technique that fabricates complex 3D structures from various biomaterial inks but is limited to a printing speed of ∼10 mm s-1 due to suboptimal rheological properties of particulate-dominated yield-stress fluids when used as liquid baths. In this work, a particle-hydrogel interactive system to design advanced baths with enhanced yield stress and extended thixotropic response time for realizing high-speed EIW is developed. In this system, the interactions between particle additive and three representative polymeric hydrogels enable the resulting nanocomposites to demonstrate different rheological behaviors. Accordingly, the interaction models for the nanocomposites are established, which are subsequently validated by macroscale rheological measurements and advanced microstructure characterization techniques. Filament formation mechanisms in the particle-hydrogel interactive baths are comprehensively investigated at high printing speeds. To demonstrate the effectiveness of the proposed high-speed EIW method, an anatomic-size human kidney construct is successfully printed at 110 mm s-1, which only takes ∼4 h. This work breaks the printing speed barrier in current EIW and propels the maximum printing speed by at least 10 times, providing an efficient and promising solution for organ reconstruction in the future.
Collapse
Affiliation(s)
- Weijian Hua
- Mechanical Engineering DepartmentUniversity of Nevada RenoRenoNevada89557USA
| | - Cheng Zhang
- Mechanical Engineering DepartmentUniversity of Nevada RenoRenoNevada89557USA
- State Key Laboratory of High‐Performance Precision ManufacturingDalian University of TechnologyDalianLiaoning116024China
| | - Haoran Cui
- Mechanical Engineering DepartmentUniversity of Nevada RenoRenoNevada89557USA
| | - Kellen Mitchell
- Mechanical Engineering DepartmentUniversity of Nevada RenoRenoNevada89557USA
| | - Dale K. Hensley
- Center for Nanophase Materials SciencesOak Ridge National LaboratoryOak RidgeTennessee37830USA
| | - Jihua Chen
- Center for Nanophase Materials SciencesOak Ridge National LaboratoryOak RidgeTennessee37830USA
| | - Changwoo Do
- Neutron Scattering DivisionOak Ridge National LaboratoryOak RidgeTennessee37831USA
| | - Lily Raymond
- Mechanical Engineering DepartmentUniversity of Nevada RenoRenoNevada89557USA
| | - Ryan Coulter
- Mechanical Engineering DepartmentUniversity of Nevada RenoRenoNevada89557USA
| | - Erick Bandala
- Mechanical Engineering DepartmentUniversity of Nevada RenoRenoNevada89557USA
| | - Fazlay Rubbi
- Department of Industrial and Manufacturing Systems EngineeringIowa State UniversityAmesIowa50011USA
| | - Guangrui Chai
- Department of OphthalmologyShengjing Hospital of China Medical UniversityShenyangLiaoning110004China
| | - Zhengyi Zhang
- School of Naval Architecture and Ocean EngineeringHuazhong University of Science and TechnologyWuhanHubei430074China
| | - Yiliang Liao
- Department of Industrial and Manufacturing Systems EngineeringIowa State UniversityAmesIowa50011USA
| | - Danyang Zhao
- State Key Laboratory of High‐Performance Precision ManufacturingDalian University of TechnologyDalianLiaoning116024China
| | - Yan Wang
- Mechanical Engineering DepartmentUniversity of Nevada RenoRenoNevada89557USA
| | - Akhilesh K. Gaharwar
- Department of Biomedical EngineeringTexas A&M University, College StationTexas77843USA
| | - Yifei Jin
- Mechanical Engineering DepartmentUniversity of Nevada RenoRenoNevada89557USA
| |
Collapse
|
7
|
Wu F, He W, Song D, Wu Z, Dai P, Zheng X, Wang H, Xie C. Ropivacaine and celecoxib-loaded injectable composite hydrogel for improved chronic pain-exacerbated myocardial ischemia-reperfusion injury. J Control Release 2025; 379:266-284. [PMID: 39800238 DOI: 10.1016/j.jconrel.2025.01.028] [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: 10/01/2024] [Revised: 12/26/2024] [Accepted: 01/09/2025] [Indexed: 01/15/2025]
Abstract
Chronic pain is a prevalent condition affecting a significant portion of the global population and is known to be associated with an increased risk of cardiovascular diseases. Despite the clinical relevance, the mechanisms underlying the link between chronic pain and myocardial ischemia-reperfusion (MI/R) injury remain poorly understood. This study aimed to investigate the role of the superior cervical ganglion (SCG) in mediating the effects of chronic pain on MI/R injury and to develop a novel therapeutic strategy. We identified that chronic pain upregulated TNF-α expression and induced hyperactivity in SCG sympathetic neurons, exacerbating MI/R injury. To address this, we engineered an injectable Pluronic/alginate-based composite hydrogel loaded with celecoxib and ropivacaine (celecoxib@Laponite-dopamine-alginate-Pluronic F-127@ropivacaine, CLDAFR). This hydrogel was designed to target the SCG, providing a localized and sustained release of the therapeutic agents, thereby mitigating neuronal inflammation and inhibiting neuronal hyperactivity. The CLDAFR hydrogel demonstrated excellent biocompatibility, heat-sensitive gelation properties, and controlled drug release in vitro. In vivo studies showed that applying CLDAFR effectively reduced MI/R injury in a chronic pain model by suppressing TNF-α expression and SCG neuronal activity. In conclusion, the CLDAFR hydrogel represents a promising therapeutic material for treating chronic pain-exacerbated MI/R injury by precisely targeting the SCG and providing a sustained anti-inflammatory and analgesic effect.
Collapse
Affiliation(s)
- Fancan Wu
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, Guangdong, PR China
| | - Wanyou He
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, Guangdong, PR China
| | - Da Song
- Department of Biomedical Engineering, Southern University of Science and Technology, Shenzhen, Guangdong, PR China
| | - Zhen Wu
- Department of Critical Care Medicine, Guangdong Second Provincial General Hospital, Jinan University, Guangzhou, Guangdong, PR China
| | - Peng Dai
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, Guangdong, PR China
| | - Xueqin Zheng
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, Guangdong, PR China.
| | - Hanbing Wang
- Department of Anesthesiology, The First People's Hospital of Foshan, Foshan, Guangdong, PR China.
| | - Chao Xie
- Department of Joint and Orthopedics, Zhujiang Hospital, Southern Medical University, Guangzhou, Guangdong, PR China.
| |
Collapse
|
8
|
Li XL, Zhao YQ, Miao L, An YX, Wu F, Han JY, Han JY, Tay FR, Mu Z, Jiao Y, Wang J. Strategies for promoting neurovascularization in bone regeneration. Mil Med Res 2025; 12:9. [PMID: 40025573 PMCID: PMC11874146 DOI: 10.1186/s40779-025-00596-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2024] [Accepted: 01/26/2025] [Indexed: 03/04/2025] Open
Abstract
Bone tissue relies on the intricate interplay between blood vessels and nerve fibers, both are essential for many physiological and pathological processes of the skeletal system. Blood vessels provide the necessary oxygen and nutrients to nerve and bone tissues, and remove metabolic waste. Concomitantly, nerve fibers precede blood vessels during growth, promote vascularization, and influence bone cells by secreting neurotransmitters to stimulate osteogenesis. Despite the critical roles of both components, current biomaterials generally focus on enhancing intraosseous blood vessel repair, while often neglecting the contribution of nerves. Understanding the distribution and main functions of blood vessels and nerve fibers in bone is crucial for developing effective biomaterials for bone tissue engineering. This review first explores the anatomy of intraosseous blood vessels and nerve fibers, highlighting their vital roles in bone embryonic development, metabolism, and repair. It covers innovative bone regeneration strategies directed at accelerating the intrabony neurovascular system over the past 10 years. The issues covered included material properties (stiffness, surface topography, pore structures, conductivity, and piezoelectricity) and acellular biological factors [neurotrophins, peptides, ribonucleic acids (RNAs), inorganic ions, and exosomes]. Major challenges encountered by neurovascularized materials during their clinical translation have also been highlighted. Furthermore, the review discusses future research directions and potential developments aimed at producing bone repair materials that more accurately mimic the natural healing processes of bone tissue. This review will serve as a valuable reference for researchers and clinicians in developing novel neurovascularized biomaterials and accelerating their translation into clinical practice. By bridging the gap between experimental research and practical application, these advancements have the potential to transform the treatment of bone defects and significantly improve the quality of life for patients with bone-related conditions.
Collapse
Affiliation(s)
- Xin-Ling Li
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yu-Qing Zhao
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Li Miao
- Department of Stomatology, The Seventh Medical Center of PLA General Hospital, Beijing, 100700, China
| | - Yan-Xin An
- Department of General Surgery, The First Affiliated Hospital of Xi'an Medical University, Xi'an, 710077, China
| | - Fan Wu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Jin-Yu Han
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Jing-Yuan Han
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China
| | - Franklin R Tay
- Graduate School of Augusta University, Augusta, GA, 30912, USA
| | - Zhao Mu
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Yang Jiao
- Department of Stomatology, The Seventh Medical Center of PLA General Hospital, Beijing, 100700, China.
| | - Jing Wang
- State Key Laboratory of Oral & Maxillofacial Reconstruction and Regeneration, National Clinical Research Center for Oral Diseases, Shaanxi Engineering Research Center for Dental Materials and Advanced Manufacture, Department of Oral Implants, School of Stomatology, The Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
9
|
Sahare P, Ruiz-Manriquez LM, Anguiano B, Banerjee A, Pathak S, Duttaroy AK, Luna-Bárcenas G, Paul S. Recent advances in nanomedicine for the diagnosis and therapy of thyroid disorders. 3 Biotech 2025; 15:67. [PMID: 40012722 PMCID: PMC11850684 DOI: 10.1007/s13205-025-04234-4] [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: 10/09/2023] [Accepted: 02/09/2025] [Indexed: 02/28/2025] Open
Abstract
Thyroid diseases, including hypo- or hyperthyroidism and thyroid cancer, are among the most prevalent endocrine disorders that remain challenging to diagnose accurately, and conventional treatments have low therapeutic efficacy. Recently, a wide variety of innovations in the area of nanomedicine, including nanotechnology-based quick and portable diagnostic tools and targeted delivery of pharmaceutics for thyroid disorders, are looking extremely promising. Hence, this review elaborates on the pathogenesis of thyroid disorders and the unique composition, structural attributes, synthesis methodologies, and surface design of nanoplatforms, focusing on the three principal application domains of nanomedicine in thyroid disorders: nanodiagnosis, nanotherapy, and nanotheranostics. Subsequently, cutting-edge studies featuring a series of notable and noteworthy accomplishments are comprehensively summarized, highlighting the intrinsic interaction between nanomedicines and thyroid dysfunctions; particular emphasis is placed on the biosafety of nanomedicines, which seeks to facilitate the future clinical translation of this emerging discipline. This comprehensive review concludes by identifying potential challenges and future perspectives of nanomedicine for the diagnosis and therapy of thyroid disorders.
Collapse
Affiliation(s)
- Padmavati Sahare
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM 3001, 76230 Juriquilla, QRO Mexico
- Institute of Advanced Materials for Sustainable Manufacturing, Tecnologico de Monterrey, Campus Querétaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130 Querétaro, Mexico
| | - Luis M. Ruiz-Manriquez
- School of Engineering and Sciences, Tecnologico de Monterrey, Campus Querétaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130 Querétaro, Mexico
- School of Medicine and Health Science, Tecnologico de Monterrey, Monterrey, Mexico
| | - Brenda Anguiano
- Instituto de Neurobiología, Universidad Nacional Autónoma de México, Campus UNAM 3001, 76230 Juriquilla, QRO Mexico
| | - Antara Banerjee
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Surajit Pathak
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Chennai, India
| | - Asim K. Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, Oslo, Norway
| | - Gabriel Luna-Bárcenas
- Institute of Advanced Materials for Sustainable Manufacturing, Tecnologico de Monterrey, Campus Querétaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130 Querétaro, Mexico
| | - Sujay Paul
- School of Engineering and Sciences, Tecnologico de Monterrey, Campus Querétaro, Av. Epigmenio Gonzalez, No. 500 Fracc. San Pablo, 76130 Querétaro, Mexico
| |
Collapse
|
10
|
Zhu J, Ma H, Du J, Fang H, Cheng YY, Xu J, Pan B, Song K. A coaxial 3D bioprinted hybrid vascular scaffold based on decellularized extracellular matrix/nano clay/sodium alginate bioink. Int J Biol Macromol 2025; 290:139056. [PMID: 39710031 DOI: 10.1016/j.ijbiomac.2024.139056] [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: 08/09/2024] [Revised: 11/28/2024] [Accepted: 12/19/2024] [Indexed: 12/24/2024]
Abstract
Currently, vascular grafting is the preferred option to replace or bypass the defective vascular segments, but finding materials with good biocompatibility and diversity alternative for practical clinical applications are still the challenge. The construction of tissue engineered blood vessels (TEBVs) with complex structures will be realized using 3D bioprinting technology, which provides a new idea for vascular transplantation. In this paper, the decellularized extracellular matrix (dECM)/nano clay (NC)/sodium alginate (SA) hybrid bioink was prepared to construct tubular scaffolds in vitro by coaxial 3D bioprinting. The physical properties of the tubular scaffolds showed that there were plenty of pores, of which the size was ranged from 5 μm to 100 μm. Among them, the 2d/NC/SA scaffold not only has good hydrophilicity (>300 %), good biomechanical properties (tensile strength: 0.99 ± 0.01 MPa, Young's modulus: 0.61 ± 0.02 MPa) and low hemolysis ratio (0.3 %), but also can effectively enhance cell adhesion and proliferation. Cell experiment also showed that the cell density and cell colonies were large and more on the coaxial printed tubular scaffolds compare to those on the 3D printed lamellar scaffolds, and the 2d/NC/SA tubular scaffold has the best bioactivity of tunica intima model. Overall, the advanced dECM/NC/SA tubular scaffold has a considerable potential to be applied in vascular tissue engineering.
Collapse
Affiliation(s)
- Jingjing Zhu
- Cancer Hospital of Dalian University of Technology, State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Hailin Ma
- Cancer Hospital of Dalian University of Technology, State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Jing Du
- Cancer Hospital of Dalian University of Technology, State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Huan Fang
- Cancer Hospital of Dalian University of Technology, State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China
| | - Yuen Yee Cheng
- Institute for Biomedical Materials and Devices, Faculty of Science, University of Technology Sydney, NSW 2007, Australia
| | - Jie Xu
- Cancer Hospital of Dalian University of Technology, State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China.
| | - Bo Pan
- Department of Breast Surgery, The Second Hospital of Dalian Medical University, 467 Zhongshan Road, Shahekou District, Dalian 116023, China.
| | - Kedong Song
- Cancer Hospital of Dalian University of Technology, State Key Laboratory of Fine Chemicals, Dalian R&D Center for Stem Cell and Tissue Engineering, Dalian University of Technology, Dalian 116024, China.
| |
Collapse
|
11
|
Furuichi T, Hirai H, Kitahara T, Bun M, Ikuta M, Ukon Y, Furuya M, Oreffo RO, Janeczek AA, Dawson JI, Okada S, Kaito T. Nanoclay gels attenuate BMP2-associated inflammation and promote chondrogenesis to enhance BMP2-spinal fusion. Bioact Mater 2025; 44:474-487. [PMID: 39559426 PMCID: PMC11570687 DOI: 10.1016/j.bioactmat.2024.10.027] [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: 05/18/2024] [Revised: 10/30/2024] [Accepted: 10/30/2024] [Indexed: 11/20/2024] Open
Abstract
Bone morphogenetic protein 2 (BMP2) is clinically applied for treating intractable fractures and promoting spinal fusion because of its osteogenic potency. However, adverse effects following the release of supraphysiological doses of BMP2 from collagen carriers are widely reported. Nanoclay gel (NC) is attracting attention as a biomaterial, given the potential for localized efficacy of administered agents. However, the efficacy and mechanism of action of NC/BMP2 remain unclear. This study explored the efficacy of NC as a BMP2 carrier in bone regeneration and the enhancement mechanism. Subfascial implantation of NC containing BMP2 elicited superior bone formation compared with collagen sponge (CS). Cartilage was uniformly formed inside the NC, whereas CS formed cartilage only on the perimeter. Additionally, CS induced a dose-dependent inflammatory response around the implantation site, whereas NC induced a minor response, and inflammatory cells were observed inside the NC. In a rat spinal fusion model, NC promoted high-quality bony fusion compared to CS. In vitro, NC enhanced chondrogenic and osteogenic differentiation of hBMSCs and ATDC5 cells while inhibiting osteoclastogenesis. Overall, NC/BMP2 facilitates spatially controlled, high-quality endochondral bone formation without BMP2-induced inflammation and promotes high-density new bone, functioning as a next-generation BMP2 carrier.
Collapse
Affiliation(s)
- Takuya Furuichi
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Hiromasa Hirai
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Takayuki Kitahara
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masayuki Bun
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masato Ikuta
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Yuichiro Ukon
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Masayuki Furuya
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Richard O.C. Oreffo
- Bone & Joint Research Group, Centre for Human Development, Stem Cells & Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, SO16 6YD, United Kingdom
| | - Agnieszka A. Janeczek
- Renovos Biologics Limited, 2 Venture Road, University of Southampton Science Park, Southampton, SO16 7NP, United Kingdom
| | - Jonathan I. Dawson
- Bone & Joint Research Group, Centre for Human Development, Stem Cells & Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton, SO16 6YD, United Kingdom
| | - Seiji Okada
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| | - Takashi Kaito
- Department of Orthopedic Surgery, Osaka University Graduate School of Medicine, 2-2 Yamadaoka, Suita, Osaka, 565-0871, Japan
| |
Collapse
|
12
|
Li T, Zhang X, Ma L, Qi X, Wang H, Zhou Q, Sun X, Wang F, Zhao L, Shi W. 3D printing of stiff, tough, and ROS-scavenging nanocomposite hydrogel scaffold for in situ corneal repair. Acta Biomater 2025; 192:189-205. [PMID: 39643222 DOI: 10.1016/j.actbio.2024.12.005] [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: 07/31/2024] [Revised: 11/19/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
Despite significant advancements in hydrogels in recent years, their application in corneal repair remains limited by several challenges, including unfitted curvatures, inferior mechanical properties, and insufficient reactive oxygen species (ROS)-scavenging activities. To address these issues, this study introduces a 3D-printed corneal scaffold with nanocomposite hydrogel consisting of gelatin methacrylate (GelMA), poly (ethylene glycol) diacrylate (PEGDA), Laponite, and dopamine. GelMA and PEGDA act as matrix materials with photo-crosslinking abilities. As a two-dimensional nanoclay, Laponite enhances the rheological properties of the hydrogel, making it suitable for 3D printing. Dopamine self-polymerizes into polydopamine (PDA), providing the hydrogel with ROS-scavenging activity. The incorporation of Laponite and the synergistic effect of PDA endow the hydrogel with good mechanical properties. In vitro investigations demonstrated the cytocompatibility of GelMA-PEGDA-Laponite-dopamine (GPLD) hydrogel and its ROS-scavenging activity. Furthermore, in vivo experiments using a rabbit model of lamellar keratoplasty showed accelerated corneal re-epithelialization and complete stromal repair after the implantation of the 3D-printed scaffold. Overall, due to its high bioactivity and simple preparation, the 3D-printed scaffold using GPLD hydrogel offers an alternative for corneal repair with potential for clinical translation. STATEMENT OF SIGNIFICANCE: The clinical application of hydrogel corneal scaffolds has been constrained by their inadequate mechanical properties and the complex microenvironment created by elevated levels of ROS post-transplantation. In this study, we developed a kind of nanocomposite hydrogel by integrating Laponite and dopamine into GelMA and PEGDA. This advanced hydrogel was utilized to 3D print a corneal scaffold with high mechanical strength and ROS-scavenging abilities. When applied to a rabbit model of lamellar keratoplasty, the 3D-printed scaffold enabled complete re-epithelialization of the cornea within one week. Three months after surgery, the corneal stroma was fully repaired, and regeneration of corneal nerve fibers was also observed. This 3D-printed scaffold demonstrated exceptional efficacy in repairing corneal defects with potential for clinical translation.
Collapse
Affiliation(s)
- Tan Li
- State Key Laboratory Cultivation Base, Shandong Key Laboratory of Eye Diseases, Eye Institute of Shandong First Medical University, Qingdao 266071, China
| | - Xiaoyu Zhang
- State Key Laboratory Cultivation Base, Shandong Key Laboratory of Eye Diseases, Eye Institute of Shandong First Medical University, Qingdao 266071, China; Eye Hospital of Shandong First Medical University (Shandong Eye Hospital), Jinan 250021, China
| | - Li Ma
- State Key Laboratory Cultivation Base, Shandong Key Laboratory of Eye Diseases, Eye Institute of Shandong First Medical University, Qingdao 266071, China
| | - Xia Qi
- State Key Laboratory Cultivation Base, Shandong Key Laboratory of Eye Diseases, Eye Institute of Shandong First Medical University, Qingdao 266071, China
| | - Hongwei Wang
- State Key Laboratory Cultivation Base, Shandong Key Laboratory of Eye Diseases, Eye Institute of Shandong First Medical University, Qingdao 266071, China
| | - Qingjun Zhou
- State Key Laboratory Cultivation Base, Shandong Key Laboratory of Eye Diseases, Eye Institute of Shandong First Medical University, Qingdao 266071, China
| | - Xiuli Sun
- State Key Laboratory Cultivation Base, Shandong Key Laboratory of Eye Diseases, Eye Institute of Shandong First Medical University, Qingdao 266071, China
| | - Fuyan Wang
- State Key Laboratory Cultivation Base, Shandong Key Laboratory of Eye Diseases, Eye Institute of Shandong First Medical University, Qingdao 266071, China
| | - Long Zhao
- State Key Laboratory Cultivation Base, Shandong Key Laboratory of Eye Diseases, Eye Institute of Shandong First Medical University, Qingdao 266071, China
| | - Weiyun Shi
- State Key Laboratory Cultivation Base, Shandong Key Laboratory of Eye Diseases, Eye Institute of Shandong First Medical University, Qingdao 266071, China; Eye Hospital of Shandong First Medical University (Shandong Eye Hospital), Jinan 250021, China.
| |
Collapse
|
13
|
Li B, Li C, Yan Z, Yang X, Xiao W, Zhang D, Liu Z, Liao X. A review of self-healing hydrogels for bone repair and regeneration: Materials, mechanisms, and applications. Int J Biol Macromol 2025; 287:138323. [PMID: 39645113 DOI: 10.1016/j.ijbiomac.2024.138323] [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: 06/21/2024] [Revised: 11/27/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
Bone defects, which arise from various factors such as trauma, tumor resection, and infection, present a significant clinical challenge. There is an urgent need to develop new biomaterials capable of repairing a wide array of damage and defects in bone tissue. Self-healing hydrogels, a groundbreaking advancement in the field of biomaterials, displaying remarkable ability to regenerate damaged connections after partial severing, thus offering a promising solution for bone defect repair. This review first presents a comprehensive overview of the progress made in the design and preparation of these hydrogels, focusing on the self-healing mechanisms based on physical non-covalent interactions and dynamic chemical covalent bonds. Subsequently, the applications of self-healing hydrogels including natural polymers, synthetic polymers, and nano-hybrid materials, are discussed in detail, emphasizing their mechanisms in promoting bone tissue regeneration. Finally, the review addresses current challenges as well as future prospects for the use of hydrogels in bone repair and regeneration, identifying osteogenic properties, mechanical performance, and long-term biocompatibility as key areas for further improvement. In summary, this paper provides an in-depth analysis of recent advances in self-healing hydrogels for bone repair and regeneration, underscoring their immense potential for clinical application.
Collapse
Affiliation(s)
- Bo Li
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, Chongqing University of Science and Technology, Chongqing 401331, China
| | - Chenchen Li
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, Chongqing University of Science and Technology, Chongqing 401331, China
| | - Ziyi Yan
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, Chongqing University of Science and Technology, Chongqing 401331, China
| | - Xiaoling Yang
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, Chongqing University of Science and Technology, Chongqing 401331, China
| | - Wenqian Xiao
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, Chongqing University of Science and Technology, Chongqing 401331, China.
| | - Dawei Zhang
- Department of Orthopedics, The 960th Hospital of the PLA Joint Logistice Support Force, Jinan 250031, China.
| | - Zhongning Liu
- Department of Prosthodontics, Peking University School and Hospital of Stomatology & National Center for Stomatology & National Clinical Research Center for Oral Diseases & National Engineering Research Center of Oral Biomaterials and Digital Medical Devices, Beijing 100081, China.
| | - Xiaoling Liao
- Chongqing Key Laboratory of Nano/Micro Composite Materials and Devices, Chongqing Engineering Laboratory of Nano/Micro Biomedical Detection Technology, Chongqing University of Science and Technology, Chongqing 401331, China
| |
Collapse
|
14
|
Zhou L, Zhang C, Shi T, Wu D, Chen H, Han J, Chen D, Lin J, Liu W. Functionalized 3D-printed GelMA/Laponite hydrogel scaffold promotes BMSCs recruitment through osteoimmunomodulatory enhance osteogenic via AMPK/mTOR signaling pathway. Mater Today Bio 2024; 29:101261. [PMID: 39381262 PMCID: PMC11460517 DOI: 10.1016/j.mtbio.2024.101261] [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: 05/30/2024] [Revised: 09/04/2024] [Accepted: 09/19/2024] [Indexed: 10/10/2024] Open
Abstract
The migration and differentiation of bone marrow mesenchymal stem cells (BMSCs) play crucial roles in bone repair processes. However, conventional scaffolds often lack of effectively inducing and recruiting BMSCs. In our study, we present a novel approach by introducing a 3D-bioprinted scaffold composed of hydrogels, with the addition of laponite to the GelMA solution, aimed at enhancing scaffold performance. Both in vivo and in vitro experiments have confirmed the outstanding biocompatibility of the scaffold. Furthermore, for the first time, Apt19s has been chemically modified onto the surface of the hydrogel scaffold, resulting in a remarkable enhancement in the migration and adhesion of BMSCs. Moreover, the scaffold has demonstrated robust osteogenic differentiation capability in both in vivo and in vitro environments. Additionally, the hydrogel scaffold has shown the ability to induce the polarization of macrophages from M1 to M2, thereby facilitating the osteogenic differentiation of BMSCs via the bone immune pathway. Through RNA-seq analysis, it has been revealed that macrophages regulate the osteogenic differentiation of BMSCs through the AMPK/mTOR signaling pathway. In summary, the functionalized GelMA/Laponite scaffold offers a cost-effective approach for tailored in situ bone regeneration.
Collapse
Affiliation(s)
- Linquan Zhou
- Fujian Medical University Union Hospital, Fuzhou, 350000, China
| | - Chengcheng Zhang
- The School of Health, Fujian Medical University, Fuzhou, 350000, China
| | - Tengbin Shi
- Fujian Medical University Union Hospital, Fuzhou, 350000, China
| | - Dingwei Wu
- Fujian Medical University Union Hospital, Fuzhou, 350000, China
| | - Huina Chen
- The School of Health, Fujian Medical University, Fuzhou, 350000, China
| | - Jiaxin Han
- The School of Health, Fujian Medical University, Fuzhou, 350000, China
| | - Dehui Chen
- Fujian Medical University Union Hospital, Fuzhou, 350000, China
| | - Jinxin Lin
- Key Laboratory of Optoelectronic Materials Chemistry and Physics, Fujian Institute of Research on the Structure of Matter, Chinese Academy of Sciences, Fuzhou, 350000, China
| | - Wenge Liu
- Fujian Medical University Union Hospital, Fuzhou, 350000, China
| |
Collapse
|
15
|
Wu N, Li J, Li X, Wang R, Zhang L, Liu Z, Jiao T. 3D printed biopolymer/black phosphorus nanoscaffolds for bone implants: A review. Int J Biol Macromol 2024; 279:135227. [PMID: 39218178 DOI: 10.1016/j.ijbiomac.2024.135227] [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: 07/03/2024] [Revised: 08/20/2024] [Accepted: 08/29/2024] [Indexed: 09/04/2024]
Abstract
Bone implantation is one of the recognized and effective means of treating bone defects, but osteoporosis and bone tumor-related bone abnormalities have a series of problems such as susceptibility to infection, difficulty in healing, and poor therapeutic effect, which poses a great challenge to clinical medicine. Three-dimensional things may be printed using 3D printing. Researchers can feed materials through the printer layer by layer to create the desired shape for a 3D structure. It is widely employed in the healing of bone defects, and it is an improved form of additive manufacturing technology with prospective future applications. This review's objective is to provide an overview of the findings reports pertaining to 3D printing biopolymers in recent years, provide an overview of biopolymer materials and their composites with black phosphorus for 3D printing bone implants, and the characterization methods of composite materials are also summarized. In addition, summarizes 3D printing methods based on ink printing and laser printing, pointing out their special features and advantages, and provide a combination strategy of photothermal therapy and bone regeneration materials for black phosphorus-based materials. Finally, the associations between bone implant materials and immune cells, the bio-environment, as well as the 3D printing bone implants prospects are outlined.
Collapse
Affiliation(s)
- Nannan Wu
- State Key Laboratory of Metastable Materials Science and Technology, Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao 066004, China
| | - Jinghong Li
- State Key Laboratory of Metastable Materials Science and Technology, Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao 066004, China.
| | - Xinyu Li
- State Key Laboratory of Metastable Materials Science and Technology, Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao 066004, China
| | - Ran Wang
- State Key Laboratory of Metastable Materials Science and Technology, Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao 066004, China
| | - Lexin Zhang
- State Key Laboratory of Metastable Materials Science and Technology, Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao 066004, China
| | - Zhiwei Liu
- State Key Laboratory of Metastable Materials Science and Technology, Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao 066004, China
| | - Tifeng Jiao
- State Key Laboratory of Metastable Materials Science and Technology, Hebei Key Laboratory of Applied Chemistry, Hebei Key Laboratory of Nanobiotechnology, Hebei Key Laboratory of Heavy Metal Deep-Remediation in Water and Resource Reuse, Yanshan University, Qinhuangdao 066004, China.
| |
Collapse
|
16
|
Marshall KM, McLaren JS, Wojciechowski JP, Callens SJP, Echalier C, Kanczler JM, Rose FRAJ, Stevens MM, Dawson JI, Oreffo ROC. Bioactive coatings on 3D printed scaffolds for bone regeneration: Use of Laponite® to deliver BMP-2 in an ovine femoral condyle defect model. BIOMATERIALS ADVANCES 2024; 164:213959. [PMID: 39083876 DOI: 10.1016/j.bioadv.2024.213959] [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: 05/05/2024] [Revised: 07/07/2024] [Accepted: 07/14/2024] [Indexed: 08/02/2024]
Abstract
Biomaterial-based approaches for bone regeneration seek to explore alternative strategies to repair non-healing fractures and critical-sized bone defects. Fracture non-union occurs due to a number of factors resulting in the formation of bone defects. Rigorous evaluation of the biomaterials in relevant models and assessment of their potential to translate towards clinical use is vital. Large animal experimentation can be used to model fracture non-union while scaling-up materials for clinical use. Growth factors modulate cell phenotype, behaviour and initiate signalling pathways leading to changes in matrix deposition and tissue formation. Bone morphogenetic protein-2 (BMP-2) is a potent osteogenic growth factor, with a rapid clearance time in vivo necessitating clinical use at a high dose, with potential deleterious side-effects. The current studies have examined the potential for Laponite® nanoclay coated poly(caprolactone) trimethacrylate (PCL-TMA900) scaffolds to bind BMP-2 for enhanced osteoinduction in a large animal critical-sized bone defect. An ovine femoral condyle defect model confirmed PCL-TMA900 scaffolds coated with Laponite®/BMP-2 produced significant bone formation compared to the uncoated PCL-TMA 900 scaffold in vivo, assessed by micro-computed tomography (μCT) and histology. This indicated the ability of Laponite® to deliver the bioactive BMP-2 on the PCL-TMA900 scaffold. Bone formed around the Laponite®/BMP-2 coated PCL-TMA900 scaffold, with no erroneous bone formation observed away from the scaffold material confirming localisation of BMP-2 delivery. The current studies demonstrate the ability of a nanoclay to localise and deliver bioactive BMP-2 within a tailored octet-truss scaffold for efficacious bone defect repair in a large animal model with significant implications for translation to the clinic.
Collapse
Affiliation(s)
- Karen M Marshall
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton SO16 6YD, UK.
| | - Jane S McLaren
- School of Pharmacy, Faculty of Science, University of Nottingham, Nottingham NG7 2RD, UK
| | - Jonathan P Wojciechowski
- Department of Materials, Department of Bioengineering, and Institute for Biomedical Engineering, Imperial College London, London SW7 2AZ, UK; Department of Physiology, Anatomy and Genetics, Department of Engineering Science, and Kavli Institute for Nanoscience Discovery, University of Oxford, OX1 3QU Oxford, UK
| | - Sebastien J P Callens
- Department of Materials, Department of Bioengineering, and Institute for Biomedical Engineering, Imperial College London, London SW7 2AZ, UK; Department of Physiology, Anatomy and Genetics, Department of Engineering Science, and Kavli Institute for Nanoscience Discovery, University of Oxford, OX1 3QU Oxford, UK
| | - Cécile Echalier
- Department of Materials, Department of Bioengineering, and Institute for Biomedical Engineering, Imperial College London, London SW7 2AZ, UK
| | - Janos M Kanczler
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton SO16 6YD, UK
| | - Felicity R A J Rose
- School of Pharmacy, Faculty of Science, University of Nottingham, Nottingham NG7 2RD, UK
| | - Molly M Stevens
- Department of Materials, Department of Bioengineering, and Institute for Biomedical Engineering, Imperial College London, London SW7 2AZ, UK; Department of Physiology, Anatomy and Genetics, Department of Engineering Science, and Kavli Institute for Nanoscience Discovery, University of Oxford, OX1 3QU Oxford, UK
| | - Jonathan I Dawson
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton SO16 6YD, UK
| | - Richard O C Oreffo
- Bone and Joint Research Group, Centre for Human Development, Stem Cells and Regeneration, Institute of Developmental Sciences, University of Southampton, Southampton SO16 6YD, UK.
| |
Collapse
|
17
|
Bhattacharyya A, Heo J, Priyajanani J, Kim SH, Khatun MR, Nagarajan R, Noh I. Simultaneous processing of both handheld biomixing and biowriting of kombucha cultured pre-crosslinked nanocellulose bioink for regeneration of irregular and multi-layered tissue defects. Int J Biol Macromol 2024; 282:136966. [PMID: 39490478 DOI: 10.1016/j.ijbiomac.2024.136966] [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: 04/29/2024] [Revised: 10/09/2024] [Accepted: 10/25/2024] [Indexed: 11/05/2024]
Abstract
The nanocellulosic pellicle derived from the symbiotic culture of bacteria and yeast (Kombucha SCOBY) is an important biomaterial for 3D bioprinting in tissue engineering. However, this nanocellulosic hydrogel has a highly entangled gel network. This needs to be partially modified to improve its processability and extrusion ability for its applications in the 3D bioprinting area. To control its mechanical and biological properties for direct 3D bioprinting applications, uniform reinforcement of nanocellulose-interacting polymers and nanoparticles in such a prefabricated gel network is essential. In this study, the hydrogel network is partially hydrolyzed with organic acid and subsequently transformed into a 3D bioprintable polyelectrolyte complex with chitosan and kaolin nanoparticles without any chemical crosslinker using a handheld 3D bioprinter. This handheld bioprinter ensures homogeneity in both biomixing and bioprinting of chitosan and kaolin within the modified nanocellulose network for multi-layered bioprinted scaffolds through an extensional shear mechanism. The biomixing simulation, mechanical (static, dynamic, and cyclic), 3D bioprinting, and cellular studies confirm the homogeneous biomixing of kaolin nanoparticles and live cells in this nanocellulose-chitosan polyelectrolyte hydrogel. The combination of SCOBY-derived nanocellulose-chitosan bioink with kaolin nanoparticles and a screw-driven handheld extrusion bioprinter demonstrates a promising platform for layer-by-layer regeneration of complex tissues with homogeneous cell/particle distribution with high cell viability.
Collapse
Affiliation(s)
- Amitava Bhattacharyya
- Department of Chemical and Biomolecular Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea; Convergence Institute of Biomedical Engineering and Biomaterials, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
| | - Junwon Heo
- Department of Chemical and Biomolecular Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
| | - J Priyajanani
- Functional, Innovative and Smart Textiles, PSG Institute of Advanced Studies, Coimbatore 641004, India
| | - Seon Ho Kim
- Department of Chemical and Biomolecular Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
| | - Mst Rita Khatun
- Department of Chemical and Biomolecular Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea
| | - R Nagarajan
- Functional, Innovative and Smart Textiles, PSG Institute of Advanced Studies, Coimbatore 641004, India
| | - Insup Noh
- Department of Chemical and Biomolecular Engineering, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea; Convergence Institute of Biomedical Engineering and Biomaterials, Seoul National University of Science and Technology, Seoul 01811, Republic of Korea.
| |
Collapse
|
18
|
Gaihre B, Camilleri E, Tilton M, Astudillo Potes MD, Liu X, Lucien F, Lu L. LAPONITE® nano-silicates potentiate the angiogenic effects of FG-4592 and osteogenic effects of BMP-2. Biomater Sci 2024; 12:5610-5619. [PMID: 39359127 PMCID: PMC11822916 DOI: 10.1039/d4bm00636d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/04/2024]
Abstract
LAPONITE®-based drug delivery systems offer many advantages due to the unique ionic and physical properties of LAPONITE®. The high ionicity and large surface area of LAPONITE® nanoparticles enable the intercalation and dissolution of biomolecules. In this study, we explored the potential of LAPONITE® as a carrier for FG-4592 to support angiogenesis and as a carrier for bone morphogenic protein-2 (BMP-2) to support osteogenesis. Interestingly, we found that LAPONITE® promoted the FG-4592 induced upregulation of vascular endothelial growth factor (VEGF) gene expression of human umbilical cord endothelial cells (HUVECs). Additionally, we observed that LAPONITE® could provide a sustained release of BMP-2 and significantly potentiate the osteogenic effects of BMP-2 on adipose derived mesenchymal stem cells (AMSCs). Overall, current findings on the LAPONITE®-drug/protein model system provide a unique way to potentiate the angiogenic activities of FG-4592 on HUVECs and osteogenic effects of BMP-2 on AMSCs for tissue engineering application. Future studies will be directed towards gaining a deeper understanding of these effects on a co-culture system of HUVECs and AMSCs.
Collapse
Affiliation(s)
- Bipin Gaihre
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Emily Camilleri
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Maryam Tilton
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Maria D Astudillo Potes
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Xifeng Liu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| | - Fabrice Lucien
- Department of Urology, Mayo Clinic, Rochester, MN, 55905, USA
- Department of Immunology, Mayo Clinic, Rochester, MN, 55905, USA
| | - Lichun Lu
- Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, 55905, USA.
- Department of Orthopedic Surgery, Mayo Clinic, Rochester, MN, 55905, USA
| |
Collapse
|
19
|
Gonzalez-Pujana A, Igartua M, Hernandez RM, Santos-Vizcaino E. Laponite nanoclays for the sustained delivery of therapeutic proteins. Eur J Pharm Sci 2024; 201:106858. [PMID: 39033884 DOI: 10.1016/j.ejps.2024.106858] [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: 04/30/2024] [Revised: 06/22/2024] [Accepted: 07/17/2024] [Indexed: 07/23/2024]
Abstract
Protein therapeutics hold immense promise for treating a wide array of diseases. However, their efficacy is often compromised by rapid degradation and clearance. The synthetic smectite clay Laponite emerges as a promising candidate for their sustained delivery. Despite its unique properties allow to load and release proteins mitigating burst release and extending their effects, precise control over Laponite-protein interactions remains challenging since it depends on a complex interplay of factors whose implication is not fully understood yet. The aim of this review article is to shed light on this issue, providing a comprehensive discussion of the factors influencing protein loading and release, including the physicochemical properties of the nanoclay and proteins, pH, dispersion buffer, clay/protein concentration and Laponite degradation. Furthermore, we thoroughly revise the array of bioactive proteins that have been delivered from formulations containing the nanoclay, highlighting Laponite-polymer nanocomposite hydrogels, a promising avenue currently under extensive investigation.
Collapse
Affiliation(s)
- Ainhoa Gonzalez-Pujana
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, Vitoria-Gasteiz 01006, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Madrid, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Manoli Igartua
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, Vitoria-Gasteiz 01006, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Madrid, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain
| | - Rosa Maria Hernandez
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, Vitoria-Gasteiz 01006, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Madrid, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.
| | - Edorta Santos-Vizcaino
- NanoBioCel Research Group, Laboratory of Pharmaceutics, School of Pharmacy, University of the Basque Country (UPV/EHU), Paseo de la Universidad 7, Vitoria-Gasteiz 01006, Spain; Biomedical Research Networking Centre in Bioengineering, Biomaterials and Nanomedicine (CIBER-BBN), Institute of Health Carlos III, Madrid, Spain; Bioaraba, NanoBioCel Research Group, Vitoria-Gasteiz, Spain.
| |
Collapse
|
20
|
Pansuriya R, Patel T, Singh K, Al Ghamdi A, Kasoju N, Kumar A, Kailasa SK, Malek NI. Self-healable, stimuli-responsive bio-ionic liquid and sodium alginate conjugated hydrogel with tunable Injectability and mechanical properties for the treatment of breast cancer. Int J Biol Macromol 2024; 277:134112. [PMID: 39048011 DOI: 10.1016/j.ijbiomac.2024.134112] [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: 05/09/2024] [Revised: 07/13/2024] [Accepted: 07/21/2024] [Indexed: 07/27/2024]
Abstract
Designing stimuli-responsive drug delivery vehicles with higher drug loading capacity, sustained and targeted release of anti-cancer drugs and able to mitigate the shortcomings of traditional systems is need of hour. Herein, we designed stimuli-responsive, self-healable, and adhesive hydrogel through synergetic interaction between [Cho][Gly] (Choline-Glycine) and sodium alginate (SA). The hydrogel was formed as a result of non-covalent interaction between the components of the mixture forming the fibre kind morphology; confirmed through FTIR/computational analysis and SEM/AFM images. The hydrogel exhibited excellent mechanical strength, self-healing ability, adhesive character and most importantly; adjustable injectability. In vitro biocompatibility of the hydrogel was tested on HaCaT and MCF-7 cells, showing >92 % cell viability after 48 h. The hemolysis ratio (<4 %) of the hydrogel confirmed the blood compatibility of the hydrogel. When tested for drug-loading capacity, the hydrogel show 1500 times drug loading for the 5-fluorouracil (5-FU) against the SA based hydrogel. In vitro release data indicated that 5-FU have more preference towards the cancerous cell condition, i.e. acidic pH (>85 %), whereas the drug-loaded hydrogel successfully killed the MCF-7 and HeLa cell with a
Collapse
Affiliation(s)
- Raviraj Pansuriya
- Ionic Liquids Research Laboratory, Department of Chemistry, Sardar Vallabhbhai National Institute of Technology, Surat 395007, Gujarat, India
| | - Tapas Patel
- Ionic Liquids Research Laboratory, Department of Chemistry, Sardar Vallabhbhai National Institute of Technology, Surat 395007, Gujarat, India
| | - Kuldeep Singh
- Salt and Marine Chemicals Division, CSIR-Central Salt and Marine Chemicals Research Institute, Council of Scientific and Industrial Research, G. B. Marg, Bhavnagar 364002, India
| | - Azza Al Ghamdi
- Department of Chemistry, College of Science, Imam Abdul Rahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia; Basic & Applied Scientific Research Center (BASRC), Water Treatment Unit, Imam Abdulrahman Bin Faisal University, P.O. Box 1982, Dammam 31441, Saudi Arabia
| | - Naresh Kasoju
- Sree Chitra Tirunal Institute for Medical Sciences and Technology, Trivandrum 695011, Kerala, India
| | - Arvind Kumar
- Salt and Marine Chemicals Division, CSIR-Central Salt and Marine Chemicals Research Institute, Council of Scientific and Industrial Research, G. B. Marg, Bhavnagar 364002, India
| | - Suresh Kumar Kailasa
- Ionic Liquids Research Laboratory, Department of Chemistry, Sardar Vallabhbhai National Institute of Technology, Surat 395007, Gujarat, India
| | - Naved I Malek
- Ionic Liquids Research Laboratory, Department of Chemistry, Sardar Vallabhbhai National Institute of Technology, Surat 395007, Gujarat, India.
| |
Collapse
|
21
|
Agarwal P, Mathur V, Kasturi M, Srinivasan V, Seetharam RN, S Vasanthan K. A Futuristic Development in 3D Printing Technique Using Nanomaterials with a Step Toward 4D Printing. ACS OMEGA 2024; 9:37445-37458. [PMID: 39281933 PMCID: PMC11391532 DOI: 10.1021/acsomega.4c04123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/30/2024] [Revised: 07/27/2024] [Accepted: 08/06/2024] [Indexed: 09/18/2024]
Abstract
3D bioprinting has shown great promise in tissue engineering and regenerative medicine for creating patient-specific tissue scaffolds and medicinal devices. The quickness, accurate imaging, and design targeting of this emerging technology have excited biomedical engineers and translational medicine researchers. Recently, scaffolds made from 3D bioprinted tissue have become more clinically effective due to nanomaterials and nanotechnology. Because of quantum confinement effects and high surface area/volume ratios, nanomaterials and nanotechnological techniques have unique physical, chemical, and biological features. The use of nanomaterials and 3D bioprinting has led to scaffolds with improved physicochemical and biological properties. Nanotechnology and nanomaterials affect 3D bioprinted tissue engineered scaffolds for regenerative medicine and tissue engineering. Biomaterials and cells that respond to stimuli change the structural shape in 4D bioprinting. With such dynamic designs, tissue architecture can change morphologically. New 4D bioprinting techniques will aid in bioactuation, biorobotics, and biosensing. The potential of 4D bioprinting in biomedical technologies is also discussed in this article.
Collapse
Affiliation(s)
- Prachi Agarwal
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal 576104, India
| | - Vidhi Mathur
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal 576104, India
| | - Meghana Kasturi
- Department of Mechanical Engineering, University of Michigan, Dearborn, Michigan 48128, United States
| | - Varadharajan Srinivasan
- Manipal Institute of Technology, Manipal Academy of Higher Education, Karnataka, Manipal 576104, India
| | - Raviraja N Seetharam
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal 576104, India
| | - Kirthanashri S Vasanthan
- Manipal Centre for Biotherapeutics Research, Manipal Academy of Higher Education, Karnataka, Manipal 576104, India
| |
Collapse
|
22
|
Chen Z, Xiao N, Luo L, Zhang L, Yin F, Hu W, Wu Z, Chen Y, Luo K, Xu X. Nanosilicates facilitate periodontal regeneration potential by activating the PI3K-AKT signaling pathway in periodontal ligament cells. J Nanobiotechnology 2024; 22:532. [PMID: 39223550 PMCID: PMC11370094 DOI: 10.1186/s12951-024-02798-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2024] [Accepted: 08/22/2024] [Indexed: 09/04/2024] Open
Abstract
The recent development of nanobiomaterials has shed some light on the field of periodontal tissue regeneration. Laponite (LAP), an artificially synthesized two-dimensional (2D) disk-shaped nanosilicate, has garnered substantial attention in regenerative biomedical applications owing to its distinctive structure, exceptional biocompatibility and bioactivity. This study endeavors to comprehensively evaluate the influence of LAP on periodontal regeneration. The effects of LAP on periodontal ligament cells (PDLCs) on osteogenesis, cementogenesis and angiogenesis were systematically assessed, and the potential mechanism was explored through RNA sequencing. The results indicated that LAP improved osteogenic and cementogenic differentiation of PDLCs, the regulatory effects of LAP on PDLCs were closely correlated with activation of PI3K-AKT signaling pathway. Moreover, LAP enhanced angiogenesis indirectly via manipulating paracrine of PDLCs. Then, LAP was implanted into rat periodontal defect to confirm its regenerative potential. Both micro-CT and histological analysis indicated that LAP could facilitate periodontal tissue regeneration in vivo. These findings provide insights into the bioactivity and underlying mechanism of LAP on PDLCs, highlighting it might be a potential therapeutic option in periodontal therapy.
Collapse
Affiliation(s)
- Ziqin Chen
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Nianqi Xiao
- Gannan Health Vocational College, Ganzhou, Jiangxi, 341000, P.R. China
| | - Lan Luo
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Lu Zhang
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Fan Yin
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Weiqiang Hu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Zekai Wu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Yuling Chen
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China
| | - Kai Luo
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China.
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China.
| | - Xiongcheng Xu
- Fujian Key Laboratory of Oral Diseases & Fujian Provincial Engineering Research Center of Oral Biomaterial & Stomatological Key Laboratory of Fujian College and University, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, P.R. China.
- Institute of Stomatology & Laboratory of Oral Tissue Engineering, School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, P.R. China.
| |
Collapse
|
23
|
Zhang J, Fu C, Luo Q, Qin X, Batur S, Xie Q, Kong L, Yang C, Zhang Z. A laponite-based immunologically active gel delivery system for long-acting tumor vaccine. J Control Release 2024; 373:201-215. [PMID: 39004104 DOI: 10.1016/j.jconrel.2024.07.030] [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: 04/02/2024] [Revised: 07/01/2024] [Accepted: 07/11/2024] [Indexed: 07/16/2024]
Abstract
Traditional bolus vaccines typically require multiple doses, which complicates the vaccination process and may cause missed shots, leading to sub-optimal immunity and reduced vaccine effectiveness. Herein, a gel-based long-acting vaccine system with self-adjuvant properties based on laponite was constructed to simplify vaccination procedures and improve vaccine effectiveness. Firstly, the gel system could recruit multiple types of immune cells to form immune niches. Secondly, it could achieve sustained delivery of antigens to lymph nodes by active transport and passive drainage. Then, the gel system triggered the formation of a large number of germinal centers, which elicited enhanced and durable humoral immune responses, as well as strong cellular immune responses. As a result, it eventually showed good prophylactic and therapeutic effects in a variety of tumor models including melanoma, colorectal cancer and peritoneal metastasis models. By further combining the immunoadjuvant CpG ODN and cytokine IL-12, the effect of the gel-vaccine could be further enhanced. In a murine peritoneal metastasis model of colorectal carcinoma, a single administration of the gel-vaccine resulted in complete tumor eradication in 8/9 mice. In summary, this study developed an immunologically active gel-vaccine system. And as a robust and versatile vaccine platform, by loading different antigens and adjuvants, this gel-vaccine system is expected to realize its better therapeutic potential.
Collapse
Affiliation(s)
- Jiao Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Chuansheng Fu
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qian Luo
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Xianya Qin
- Department of Pharmacy, Maternal and Child Health Hospital of Hubei Province, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430070, China
| | - Samira Batur
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Qi Xie
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Li Kong
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Conglian Yang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China
| | - Zhiping Zhang
- Tongji School of Pharmacy, Huazhong University of Science and Technology, Wuhan 430030, China; National Engineering Research Center for Nanomedicine, Huazhong University of Science and Technology, Wuhan 430030, China; Hubei Engineering Research Center for Novel Drug Delivery System, Huazhong University of Science and Technology, Wuhan 430030, China.
| |
Collapse
|
24
|
Zheng Y, Li Z, Li S, Zhao P, Wang X, Lu S, Shi Y, Chang H. Separable nanocomposite hydrogel microneedles for intradermal and sustained delivery of antigens to enhance adaptive immune responses. Acta Biomater 2024; 185:203-214. [PMID: 39053817 DOI: 10.1016/j.actbio.2024.07.031] [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: 05/02/2024] [Revised: 07/03/2024] [Accepted: 07/18/2024] [Indexed: 07/27/2024]
Abstract
Vaccines play a critical role in combating infectious diseases and cancers, yet improving their efficacy remains challenging. Here, we introduce a separable nanocomposite hydrogel microneedle (NHMN) patch designed for intradermal and sustained delivery of ovalbumin (OVA), a model antigen, to enhance adaptive immune responses. The NHMN patch consists of an array of OVA-loaded microneedles made from photo-cross-linked methacrylated hyaluronic acid and laponite (LAP), supported by a hyaluronic acid backing. The incorporation of LAP not only enhances the mechanical strength of the pure hydrogel microneedles but also significantly prolongs OVA release. Furthermore, in vitro cell experiments demonstrate that NHMNs effectively activate dendritic cells without compromising cell viability. Upon skin penetration, NHMNs detach from the backing as the hyaluronic acid rapidly dissolves upon contact with the skin interstitial fluid, thereby acting as antigen reservoirs to release antigens to abundant skin dendritic cells. NHMNs containing 0.5% w/v LAP achieved a 15-day OVA release in vivo. Immunization studies demonstrate that the intradermal and sustained release of OVA via NHMNs elicited stronger and longer-lasting adaptive immune responses compared to conventional bolus injection. Given its easy to use, painless and minimally invasive features, the NHMN patch shows promise in improving vaccination accessibility and efficacy against a range of diseases. STATEMENT OF SIGNIFICANCE: The study introduces a separable nanocomposite hydrogel microneedle (NHMN) patch. This patch consists of an array of ovalbumin (OVA, a model antigen)-loaded microneedles made from photo-cross-linked methacrylated hyaluronic acid and laponite, with a hyaluronic acid backing, designed for intradermal and sustained delivery of antigens. This patch addresses several key challenges in traditional vaccination methods, including poor antigen uptake and presentation, and rapid systematic clearance. The incorporation of laponite enhances mechanical strength of microneedles, promotes dendritic cell activation, and significantly slows down antigen release. NHMN-based vaccination elicits stronger and longer-lasting adaptive immune responses compared to conventional bolus injection. This NHMN patch holds great potential for improving the efficacy, accessibility, and patient comfort of vaccinations against a range of diseases.
Collapse
Affiliation(s)
- Yanting Zheng
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, Zhejiang, China; College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, Zhejiang, China
| | - Zhiming Li
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, Zhejiang, China
| | - Shaohua Li
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, Zhejiang, China; College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou 310014, Zhejiang, China
| | - Puxuan Zhao
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, Zhejiang, China; College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Xin Wang
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, Zhejiang, China; College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Shaojie Lu
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, Zhejiang, China; College of Materials Science and Engineering, Zhejiang University of Technology, Hangzhou, Zhejiang, 310014, China
| | - Yanan Shi
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, Zhejiang, China; Medical School, Faculty of Medicine, Tianjin University, Tianjin 300072, China
| | - Hao Chang
- Hangzhou Institute of Medicine, Chinese Academy of Sciences, Hangzhou 310018, Zhejiang, China.
| |
Collapse
|
25
|
Wu X, Yang Z, Dong Y, Teng L, Li D, Han H, Zhu S, Sun X, Zeng Z, Zeng X, Zheng Q. A Self-Powered, Skin Adhesive, and Flexible Human-Machine Interface Based on Triboelectric Nanogenerator. NANOMATERIALS (BASEL, SWITZERLAND) 2024; 14:1365. [PMID: 39195403 DOI: 10.3390/nano14161365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 08/16/2024] [Accepted: 08/19/2024] [Indexed: 08/29/2024]
Abstract
Human-machine interactions (HMIs) have penetrated into various academic and industrial fields, such as robotics, virtual reality, and wearable electronics. However, the practical application of most human-machine interfaces faces notable obstacles due to their complex structure and materials, high power consumption, limited effective skin adhesion, and high cost. Herein, we report a self-powered, skin adhesive, and flexible human-machine interface based on a triboelectric nanogenerator (SSFHMI). Characterized by its simple structure and low cost, the SSFHMI can easily convert touch stimuli into a stable electrical signal at the trigger pressure from a finger touch, without requiring an external power supply. A skeleton spacer has been specially designed in order to increase the stability and homogeneity of the output signals of each TENG unit and prevent crosstalk between them. Moreover, we constructed a hydrogel adhesive interface with skin-adhesive properties to adapt to easy wear on complex human body surfaces. By integrating the SSFHMI with a microcontroller, a programmable touch operation platform has been constructed that is capable of multiple interactions. These include medical calling, music media playback, security unlocking, and electronic piano playing. This self-powered, cost-effective SSFHMI holds potential relevance for the next generation of highly integrated and sustainable portable smart electronic products and applications.
Collapse
Affiliation(s)
- Xujie Wu
- Engineering Research Center of Intelligent Materials and Advanced Medical Devices, School of Biology and Engineering, Guizhou Medical University, Guian New District, Guiyang 561113, China
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guian New District, Guiyang 561113, China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guian New District, Guiyang 561113, China
| | - Ziyi Yang
- Engineering Research Center of Intelligent Materials and Advanced Medical Devices, School of Biology and Engineering, Guizhou Medical University, Guian New District, Guiyang 561113, China
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guian New District, Guiyang 561113, China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guian New District, Guiyang 561113, China
| | - Yu Dong
- Engineering Research Center of Intelligent Materials and Advanced Medical Devices, School of Biology and Engineering, Guizhou Medical University, Guian New District, Guiyang 561113, China
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guian New District, Guiyang 561113, China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guian New District, Guiyang 561113, China
| | - Lijing Teng
- Engineering Research Center of Intelligent Materials and Advanced Medical Devices, School of Biology and Engineering, Guizhou Medical University, Guian New District, Guiyang 561113, China
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guian New District, Guiyang 561113, China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guian New District, Guiyang 561113, China
| | - Dan Li
- Engineering Research Center of Intelligent Materials and Advanced Medical Devices, School of Biology and Engineering, Guizhou Medical University, Guian New District, Guiyang 561113, China
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guian New District, Guiyang 561113, China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guian New District, Guiyang 561113, China
| | - Hang Han
- Engineering Research Center of Intelligent Materials and Advanced Medical Devices, School of Biology and Engineering, Guizhou Medical University, Guian New District, Guiyang 561113, China
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guian New District, Guiyang 561113, China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guian New District, Guiyang 561113, China
| | - Simian Zhu
- Engineering Research Center of Intelligent Materials and Advanced Medical Devices, School of Biology and Engineering, Guizhou Medical University, Guian New District, Guiyang 561113, China
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guian New District, Guiyang 561113, China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guian New District, Guiyang 561113, China
| | - Xiaomin Sun
- Engineering Research Center of Intelligent Materials and Advanced Medical Devices, School of Biology and Engineering, Guizhou Medical University, Guian New District, Guiyang 561113, China
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guian New District, Guiyang 561113, China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guian New District, Guiyang 561113, China
| | - Zhu Zeng
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guian New District, Guiyang 561113, China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guian New District, Guiyang 561113, China
| | - Xiangyu Zeng
- Engineering Research Center of Intelligent Materials and Advanced Medical Devices, School of Biology and Engineering, Guizhou Medical University, Guian New District, Guiyang 561113, China
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guian New District, Guiyang 561113, China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guian New District, Guiyang 561113, China
| | - Qiang Zheng
- Engineering Research Center of Intelligent Materials and Advanced Medical Devices, School of Biology and Engineering, Guizhou Medical University, Guian New District, Guiyang 561113, China
- Key Laboratory of Infectious Immune and Antibody Engineering of Guizhou Province, Engineering Research Center of Cellular Immunotherapy of Guizhou Province, School of Biology and Engineering/School of Basic Medical Sciences, Guizhou Medical University, Guian New District, Guiyang 561113, China
- Immune Cells and Antibody Engineering Research Center of Guizhou Province, Key Laboratory of Biology and Medical Engineering, Guizhou Medical University, Guian New District, Guiyang 561113, China
| |
Collapse
|
26
|
Hwang HS, Lee CS. Nanoclay-Composite Hydrogels for Bone Tissue Engineering. Gels 2024; 10:513. [PMID: 39195042 DOI: 10.3390/gels10080513] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 07/30/2024] [Accepted: 08/01/2024] [Indexed: 08/29/2024] Open
Abstract
Nanoclay-composite hydrogels represent a promising avenue for advancing bone tissue engineering. Traditional hydrogels face challenges in providing mechanical strength, biocompatibility, and bioactivity necessary for successful bone regeneration. The incorporation of nanoclay into hydrogel matrices offers a potential unique solution to these challenges. This review provides a comprehensive overview of the fabrication, physico-chemical/biological performance, and applications of nanoclay-composite hydrogels in bone tissue engineering. Various fabrication techniques, including in situ polymerization, physical blending, and 3D printing, are discussed. In vitro and in vivo studies evaluating biocompatibility and bioactivity have demonstrated the potential of these hydrogels for promoting cell adhesion, proliferation, and differentiation. Their applications in bone defect repair, osteochondral tissue engineering and drug delivery are also explored. Despite their potential in bone tissue engineering, nanoclay-composite hydrogels face challenges such as optimal dispersion, scalability, biocompatibility, long-term stability, regulatory approval, and integration with emerging technologies to achieve clinical application. Future research directions need to focus on refining fabrication techniques, enhancing understanding of biological interactions, and advancing towards clinical translation and commercialization. Overall, nanoclay-composite hydrogels offer exciting opportunities for improving bone regeneration strategies.
Collapse
Affiliation(s)
- Hee Sook Hwang
- Department of Pharmaceutical Engineering, Dankook University, Cheonan 31116, Republic of Korea
| | - Chung-Sung Lee
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan 31538, Republic of Korea
| |
Collapse
|
27
|
Mina M, Wu KY, Kalevar A, Tran SD. In Situ Bioprinting. 3D BIOPRINTING FROM LAB TO INDUSTRY 2024:347-390. [DOI: 10.1002/9781119894407.ch12] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
28
|
Murali A, Brokesh AM, Cross LM, Kersey AL, Jaiswal MK, Singh I, Gaharwar A. Inorganic Biomaterials Shape the Transcriptome Profile to Induce Endochondral Differentiation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402468. [PMID: 38738803 PMCID: PMC11304299 DOI: 10.1002/advs.202402468] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Revised: 03/27/2024] [Indexed: 05/14/2024]
Abstract
Minerals play a vital role, working synergistically with enzymes and other cofactors to regulate physiological functions including tissue healing and regeneration. The bioactive characteristics of mineral-based nanomaterials can be harnessed to facilitate in situ tissue regeneration by attracting endogenous progenitor and stem cells and subsequently directing tissue-specific differentiation. Here, cellular responses of human mesenchymal stem/stromal cells to traditional bioactive mineral-based nanomaterials, such as hydroxyapatite, whitlockite, silicon-dioxide, and the emerging synthetic 2D nanosilicates are investigated. Transcriptome sequencing is utilized to probe the cellular response and determine the significantly affected signaling pathways due to exposure to these inorganic nanomaterials. Transcriptome profiles of stem cells treated with nanosilicates reveals a stabilized skeletal progenitor state suggestive of endochondral differentiation. This observation is bolstered by enhanced deposition of matrix mineralization in nanosilicate treated stem cells compared to control or other treatments. Specifically, use of 2D nanosilicates directs osteogenic differentiation of stem cells via activation of bone morphogenetic proteins and hypoxia-inducible factor 1-alpha signaling pathway. This study provides insight into impact of nanomaterials on cellular gene expression profile and predicts downstream effects of nanomaterial induction of endochondral differentiation.
Collapse
Affiliation(s)
- Aparna Murali
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
| | - Anna M. Brokesh
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
| | - Lauren M. Cross
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
| | - Anna L. Kersey
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
| | - Manish K. Jaiswal
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
| | - Irtisha Singh
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
- Department of Cell Biology and GeneticsCollege of MedicineTexas A&M UniversityBryanTX77807‐3260USA
- Interdisciplinary Program in Genetics and GenomicsTexas A&M UniversityCollege StationTX77843USA
| | - Akhilesh Gaharwar
- Department of Biomedical EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
- Interdisciplinary Program in Genetics and GenomicsTexas A&M UniversityCollege StationTX77843USA
- Department of Material Science and EngineeringCollege of EngineeringTexas A&M UniversityCollege StationTX77843USA
| |
Collapse
|
29
|
Zhou R, Zhang W, Zhang Y, Wu X, Huang J, Bo R, Liu M, Yu J, Li J. Laponite/lactoferrin hydrogel loaded with eugenol for methicillin-resistant Staphylococcus aureus-infected chronic skin wound healing. J Tissue Viability 2024; 33:487-503. [PMID: 38769034 DOI: 10.1016/j.jtv.2024.05.006] [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: 10/10/2023] [Revised: 05/01/2024] [Accepted: 05/15/2024] [Indexed: 05/22/2024]
Abstract
Severe bacterial infections can give rise to protracted wound healing processes, thereby posing a significant risk to a patient's well-being. Consequently, the development of a versatile hydrogel dressing possessing robust bioactivity becomes imperative, as it holds the potential to expedite wound healing and yield enhanced clinical therapeutic outcomes. In this context, the present study involves the formulation of an injectable multifunctional hydrogel utilizing laponite (LAP) and lactoferrin (LF) as foundational components and loaded with eugenol (EG). This hydrogel is fabricated employing a straightforward one-pot mixing approach that leverages the principle of electrostatic interaction. The resulting LAP/LF/EG2% composite hydrogel can be conveniently injected to address irregular wound geometries effectively. Once administered, the hydrogel continually releases lactoferrin and eugenol, mitigating unwarranted oxidative stress and eradicating bacterial infections. This orchestrated action culminates in the acceleration of wound healing specifically in the context of MRSA-infected wounds. Importantly, the LAP/LF/EG2% hydrogel exhibits commendable qualities including exceptional injectability, potent antioxidant attributes, and proficient hemostatic functionality. Furthermore, the hydrogel composition notably encourages cellular migration while maintaining favorable cytocompatibility. Additionally, the hydrogel manifests noteworthy bactericidal efficacy against the formidable multidrug-resistant MRSA bacterium. Most significantly, this hydrogel formulation distinctly expedites the healing of MRSA-infected wounds by promptly inducing hemostasis, curbing bacterial proliferation, and fostering angiogenesis, collagen deposition, and re-epithelialization processes. As such, the innovative hydrogel material introduced in this investigation emerges as a promising dressing for the facilitation of bacterial-infected wound healing and consequent tissue regeneration.
Collapse
Affiliation(s)
- Ruigang Zhou
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Wenhai Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Yufei Zhang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Xiqian Wu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Junjie Huang
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Ruonan Bo
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Mingjiang Liu
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China
| | - Jie Yu
- The Affiliated Suqian First People's Hospital of Nanjing Medical University, Suzhi Road 120, Suqian 223800, PR China.
| | - Jingui Li
- College of Veterinary Medicine, Yangzhou University, Yangzhou, 225009, China; Jiangsu Co-innovation Center for Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou, 225009, China.
| |
Collapse
|
30
|
Chen X, Wu T, Bu Y, Yan H, Lin Q. Fabrication and Biomedical Application of Alginate Composite Hydrogels in Bone Tissue Engineering: A Review. Int J Mol Sci 2024; 25:7810. [PMID: 39063052 PMCID: PMC11277200 DOI: 10.3390/ijms25147810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 07/15/2024] [Accepted: 07/16/2024] [Indexed: 07/28/2024] Open
Abstract
Nowadays, as a result of the frequent occurrence of accidental injuries and traumas such as bone damage, the number of people causing bone injuries or fractures is increasing around the world. The design and fabrication of ideal bone tissue engineering (BTE) materials have become a research hotspot in the scientific community, and thus provide a novel path for the treatment of bone diseases. Among the materials used to construct scaffolds in BTE, including metals, bioceramics, bioglasses, biomacromolecules, synthetic organic polymers, etc., natural biopolymers have more advantages against them because they can interact with cells well, causing natural polymers to be widely studied and applied in the field of BTE. In particular, alginate has the advantages of excellent biocompatibility, good biodegradability, non-immunogenicity, non-toxicity, wide sources, low price, and easy gelation, enabling itself to be widely used as a biomaterial. However, pure alginate hydrogel as a BTE scaffold material still has many shortcomings, such as insufficient mechanical properties, easy disintegration of materials in physiological environments, and lack of cell-specific recognition sites, which severely limits its clinical application in BTE. In order to overcome the defects of single alginate hydrogels, researchers prepared alginate composite hydrogels by adding one or more materials to the alginate matrix in a certain proportion to improve their bioapplicability. For this reason, this review will introduce in detail the methods for constructing alginate composite hydrogels, including alginate/polymer composite hydrogels, alginate/bioprotein or polypeptide composite hydrogels, alginate/bioceramic composite hydrogels, alginate/bioceramic composite hydrogels, and alginate/nanoclay composite hydrogels, as well as their biological application trends in BTE scaffold materials, and look forward to their future research direction. These alginate composite hydrogel scaffolds exhibit both unexceptionable mechanical and biochemical properties, which exhibit their high application value in bone tissue repair and regeneration, thus providing a theoretical basis for the development and sustainable application of alginate-based functional biomedical materials.
Collapse
Affiliation(s)
- Xiuqiong Chen
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China; (X.C.); (T.W.); (Y.B.); (Q.L.)
- Key Laboratory of Water Pollution Treatment & Resource Reuse of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
- Key Laboratory of Natural Polymer Functional Material of Haikou City, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
| | - Ting Wu
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China; (X.C.); (T.W.); (Y.B.); (Q.L.)
- Key Laboratory of Water Pollution Treatment & Resource Reuse of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
- Key Laboratory of Natural Polymer Functional Material of Haikou City, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
| | - Yanan Bu
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China; (X.C.); (T.W.); (Y.B.); (Q.L.)
- Key Laboratory of Water Pollution Treatment & Resource Reuse of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
- Key Laboratory of Natural Polymer Functional Material of Haikou City, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
| | - Huiqiong Yan
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China; (X.C.); (T.W.); (Y.B.); (Q.L.)
- Key Laboratory of Water Pollution Treatment & Resource Reuse of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
- Key Laboratory of Natural Polymer Functional Material of Haikou City, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
| | - Qiang Lin
- Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China; (X.C.); (T.W.); (Y.B.); (Q.L.)
- Key Laboratory of Water Pollution Treatment & Resource Reuse of Hainan Province, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
- Key Laboratory of Natural Polymer Functional Material of Haikou City, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou 571158, China
| |
Collapse
|
31
|
Arab M, Behboodi P, Malek Khachatourian A, Nemati A. Enhanced mechanical properties and biocompatibility of hydroxyapatite scaffolds by magnesium and titanium oxides for bone tissue applications. Heliyon 2024; 10:e33847. [PMID: 39027606 PMCID: PMC11255589 DOI: 10.1016/j.heliyon.2024.e33847] [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: 03/21/2024] [Revised: 06/07/2024] [Accepted: 06/27/2024] [Indexed: 07/20/2024] Open
Abstract
Significant attention has been devoted to bioactive implants for bone tissue applications, particularly composite scaffolds based on hydroxyapatite (HaP). This study explores the effects of Magnesium and Titanium oxides on the characteristics of HaP-based composite (HMT) scaffolds. The ceramic nanopowders were synthesized using in situ sol-gel, and then the scaffolds were fabricated by gel-casting technique, followed by heat treatment at 1200 °C. The thermal, microstructural, and structural properties of the samples were investigated by different characterization techniques. It was observed that the formation of the MgTiO3 phase in the composite scaffold was likely the key factor contributing to the improved mechanical properties. Finally, to evaluate bioactivity and biodegradability, scaffolds were immersed in simulated body fluid (SBF) buffer and analyzed by Field Emission Scanning Electron Microscopy (FESEM), and the viability of human fibroblast cells was assessed using the MTT assay. The composite scaffolds containing the MgTiO3 phase showed greater HaP layer formation on the scaffold surface, indicating enhanced biocompatibility.
Collapse
Affiliation(s)
- Mehdi Arab
- Department of Materials Science and Engineering, Sharif University of Technology, Tehran, Iran
| | - Panteha Behboodi
- Department of Materials Science and Engineering, Sharif University of Technology, Tehran, Iran
| | | | - Ali Nemati
- Department of Materials Science and Engineering, Sharif University of Technology, Tehran, Iran
| |
Collapse
|
32
|
Zhang X, Ning F, Chen Y, Dong CM. All-in-one polysaccharide hydrogel with resistant vascular burst pressure and cooperative wound microenvironment regulation for fatal arterial hemorrhage and diabetic wound healing. Int J Biol Macromol 2024; 272:132736. [PMID: 38830494 DOI: 10.1016/j.ijbiomac.2024.132736] [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: 02/29/2024] [Revised: 05/14/2024] [Accepted: 05/27/2024] [Indexed: 06/05/2024]
Abstract
Fatal massive hemorrhage and diabetic wound healing are world widely challenging in surgical managements, and uncontrolled bleeding, chronic inflammation and damaged remodeling heavily hinder the whole healing processes. Considering hemostasis, inflammation and wound microenvironment cooperatively affect the healing progression, we design all-in-one beta-glucan (BG) hybrid hydrogels reinforced with laponite nanoclay that demonstrate tunable tissue adhesion, resistant vascular burst pressure and cooperative wound microenvironment regulation for arterial hemostasis and diabetic wound prohealing. Those hydrogels had honeycomb-like porous microstructure with average pore size of 7-19 μm, tissue adhesion strength of 18-46 kPa, and vascular burst pressure of 58-174 mmHg to achieve superior hemostasis in rat liver and femoral artery models. They could effectively scavenge reactive oxygen species, transform macrophages from proinflammatory M1 into prohealing M2, and shorten the inflammation duration via synergistic actions of BG and nitric oxide (NO). Single treatment of NO-releasing BG hybrid hydrogels attained complete closure of diabetic wounds within 14 days, orchestrated to accelerate the epithelization and dermis growth, and restored normal vascularization, achieving high performance healing with optimal collagen deposition and hair follicle regeneration. Consequently, this work opens up a new avenue to design all-in-one polysaccharide hydrogels for applications in massive bleeding hemostats and diabetic wound dressings.
Collapse
Affiliation(s)
- Xueliang Zhang
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Fangrui Ning
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Yanzheng Chen
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, Shanghai Jiao Tong University, Shanghai 200240, PR China
| | - Chang-Ming Dong
- School of Chemistry and Chemical Engineering, Frontiers Science Center for Transformative Molecules, Shanghai Key Laboratory of Electrical Insulation and Thermal Aging, Shanghai Jiao Tong University, Shanghai 200240, PR China.
| |
Collapse
|
33
|
Gangrade A, Zehtabi F, Rashad A, Haghniaz R, Falcone N, Mandal K, Khosravi S, Deka S, Yamauchi A, Voskanian L, Kim HJ, Ermis M, Khademhosseini A, de Barros NR. Nanobioactive Blood-Derived Shear-Thinning Biomaterial for Tissue Engineering Applications. APPLIED MATERIALS TODAY 2024; 38:102250. [PMID: 39006868 PMCID: PMC11242922 DOI: 10.1016/j.apmt.2024.102250] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
The conventional technique for successful bone grafts, involving the use of a patienťs own tissue (autografts), is challenged by limited availability and donor site morbidity. While allografts and xenografts offer alternatives, they come with the risk of rejection. This underscores the pressing need for tailor-made artificial bone graft materials. In this context, injectable hydrogels are emerging as a promising solution for bone regeneration, especially in complex maxillofacial reconstruction cases. These hydrogels can seamlessly adapt to irregular shapes and conservatively fill defects. Our study introduces a shear-thinning biomaterial by blending silicate nanoplatelets (SNs) enriched with human blood-derived plasma rich in growth factors (PRGF) for personalized applications. Notably, our investigations unveil that injectable hydrogel formulations comprising 7.5% PRGF yield sustained protein and growth factor release, affording precise control over critical growth factors essential for tissue regeneration. Moreover, our hydrogel exhibits exceptional biocompatibility in vitro and in vivo and demonstrates hemostatic properties. The hydrogel also presents a robust angiogenic potential and an inherent capacity to promote bone differentiation, proven through Alizarin Red staining, gene expression, and immunostaining assessments of bone-related biomarkers. Given these impressive attributes, our hydrogel stands out as a leading candidate for maxillofacial bone regeneration application. Beyond this, our findings hold immense potential in revolutionizing the field of regenerative medicine, offering an influential platform for crafting precise and effective therapeutic strategies.
Collapse
Affiliation(s)
- Ankit Gangrade
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Fatemeh Zehtabi
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Ahmad Rashad
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Reihaneh Haghniaz
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Natashya Falcone
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Kalpana Mandal
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Safoora Khosravi
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Sangeeta Deka
- Indian Institute of Technology Guwahati, Assam, India, Pin-781039
| | - Alana Yamauchi
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Leon Voskanian
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Han-Jun Kim
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
- College of Pharmacy, Korea University, 30019, Republic of Korea
- Vellore Institute of Technology (VIT), Vellore, 632014, India
| | - Menekse Ermis
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| | - Natan Roberto de Barros
- Terasaki Institute for Biomedical Innovation (TIBI), 1018 Westwood Blvd, Los Angeles, California, USA
| |
Collapse
|
34
|
Yang J, Chen Z, Gao C, Liu J, Liu K, Wang X, Pan X, Wang G, Sang H, Pan H, Liu W, Ruan C. A mechanical-assisted post-bioprinting strategy for challenging bone defects repair. Nat Commun 2024; 15:3565. [PMID: 38670999 PMCID: PMC11053166 DOI: 10.1038/s41467-024-48023-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Accepted: 04/17/2024] [Indexed: 04/28/2024] Open
Abstract
Bioprinting that can synchronously deposit cells and biomaterials has lent fresh impetus to the field of tissue regeneration. However, the unavoidable occurrence of cell damage during fabrication process and intrinsically poor mechanical stability of bioprinted cell-laden scaffolds severely restrict their utilization. As such, on basis of heart-inspired hollow hydrogel-based scaffolds (HHSs), a mechanical-assisted post-bioprinting strategy is proposed to load cells into HHSs in a rapid, uniform, precise and friendly manner. HHSs show mechanical responsiveness to load cells within 4 s, a 13-fold increase in cell number, and partitioned loading of two types of cells compared with those under static conditions. As a proof of concept, HHSs with the loading cells show an enhanced regenerative capability in repair of the critical-sized segmental and osteoporotic bone defects in vivo. We expect that this post-bioprinting strategy can provide a universal, efficient, and promising way to promote cell-based regenerative therapy.
Collapse
Affiliation(s)
- Jirong Yang
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Zhigang Chen
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chongjian Gao
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Juan Liu
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Kaizheng Liu
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Xiao Wang
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, China
| | - Xiaoling Pan
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, China
| | - Guocheng Wang
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hongxun Sang
- Shenzhen Hospital, Southern Medical University, Shenzhen, 518000, China
| | - Haobo Pan
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
- The Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, 518055, China
| | - Wenguang Liu
- School of Materials Science and Engineering, Tianjin Key Laboratory of Composite and Functional Materials, Tianjin University, Tianjin, 300350, China
| | - Changshun Ruan
- Research Center for Human Tissue and Organ Degeneration, Institute of Biomedicine and Biotechnology, Shenzhen Institute of Advanced Technology, Chinese Academy of Sciences, Shenzhen, 518055, China.
- University of Chinese Academy of Sciences, Beijing, 100049, China.
- The Key Laboratory of Biomedical Imaging Science and System, Chinese Academy of Sciences, Shenzhen, 518055, China.
| |
Collapse
|
35
|
Wang R, He X, Chen Z, Su S, Bai J, Liu H, Zhou F. A nanoparticle reinforced microporous methacrylated silk fibroin hydrogel to promote bone regeneration. Biomater Sci 2024; 12:2121-2135. [PMID: 38456326 DOI: 10.1039/d3bm01901b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/09/2024]
Abstract
Natural polymer-based hydrogels have been widely applied in bone tissue engineering due to their excellent biocompatibility and outstanding ability of drug encapsulation. However, they have relatively weak mechanical properties and lack bioactivity. Hence, we developed a bioactive nanoparticle composite hydrogel by incorporating LAPONITE®, which is an osteo-inductive inorganic nanoparticle. The incorporation of the nanoparticle significantly enhanced its mechanical properties. In vitro evaluation indicated that the nanocomposite hydrogel could exhibit good biocompatibility. Besides, the nanocomposite hydrogel was proved to have excellent osteogenic ability with up-regulated expression of osteogenic markers such as type I collagen (COL-I), runt-related transcription factor-2 (Runx-2) and osteocalcin (OCN). Furthermore, the in vivo study confirmed that the composite nanocomposite hydrogel could significantly promote new bone formation, providing a prospective strategy for bone tissue regeneration.
Collapse
Affiliation(s)
- Ruideng Wang
- Department of Orthopedics, Peking University Third Hospital, Beijing, China.
- Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China
| | - Xi He
- Key Laboratory for Biomechanics and Mechanobiology (Beihang University) of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China.
| | - Zhengyang Chen
- Department of Orthopedics, Peking University Third Hospital, Beijing, China.
- Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China
| | - Shilong Su
- Department of Orthopedics, Peking University Third Hospital, Beijing, China.
- Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China
| | - Jinwu Bai
- Department of Orthopedics, Peking University Third Hospital, Beijing, China.
- Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China
| | - Haifeng Liu
- Key Laboratory for Biomechanics and Mechanobiology (Beihang University) of Ministry of Education, Beijing Advanced Innovation Center for Biomedical Engineering, School of Biological Science and Medical Engineering, Beihang University, Beijing, China.
| | - Fang Zhou
- Department of Orthopedics, Peking University Third Hospital, Beijing, China.
- Engineering Research Center of Bone and Joint Precision Medicine, Beijing, China
| |
Collapse
|
36
|
Abdollahi Boraei SB, Bakhshandeh B, Mohammadzadeh F, Haghighi DM, Mohammadpour Z. Clay-reinforced PVC composites and nanocomposites. Heliyon 2024; 10:e29196. [PMID: 38633642 PMCID: PMC11021979 DOI: 10.1016/j.heliyon.2024.e29196] [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: 01/06/2024] [Revised: 03/12/2024] [Accepted: 04/02/2024] [Indexed: 04/19/2024] Open
Abstract
Clay-reinforced polyvinyl chloride (PVC) composites and nanocomposites are one of the newest and most important compounds studied and used in various applications, including the biomedical, automotive industry, water treatment, packaging, fire retarding, and construction. The most important clays used in the synthesis of these composites are Bentonite, Montmorillonite, Kaolinite, and Illite. The addition of these nanoclays to the PVC matrix improves mechanical properties, thermal stability, and yellowness index properties. In this chapter, a detailed study of PVC and its properties, types of nanoclays and their properties, modification of nanoclays, production methods of composites, and nanocomposites of PVC/clay, their characterization, and applications have been performed. Herein, the types, properties, and applications of PVC/clay nanocomposites, as well as their challenges and future remarks, are reviewed.
Collapse
Affiliation(s)
- Seyyed Behnam Abdollahi Boraei
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
- Biomaterials and Tissue Engineering Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, 1517964311, Iran
| | - Behnaz Bakhshandeh
- Department of Biotechnology, College of Science, University of Tehran, Tehran, Iran
| | - Fatemeh Mohammadzadeh
- Department of Polymer Engineering and Color Technology, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Dorrin Mohtadi Haghighi
- Department of Pharmaceutics, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Zahra Mohammadpour
- Biomaterials and Tissue Engineering Department, Breast Cancer Research Center, Motamed Cancer Institute, ACECR, Tehran, 1517964311, Iran
| |
Collapse
|
37
|
Roncoroni M, Martinelli G, Farris S, Marzorati S, Sugni M. Sea Urchin Food Waste into Bioactives: Collagen and Polyhydroxynaphtoquinones from P. lividus and S. granularis. Mar Drugs 2024; 22:163. [PMID: 38667780 PMCID: PMC11051063 DOI: 10.3390/md22040163] [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: 03/04/2024] [Revised: 03/25/2024] [Accepted: 03/27/2024] [Indexed: 04/28/2024] Open
Abstract
Approximately 75,000 tons of different sea urchin species are globally harvested for their edible gonads. Applying a circular economy approach, we have recently demonstrated that non-edible parts of the Mediterranean Sea urchin Paracentrotus lividus can be fully valorized into high-value products: antioxidant pigments (polyhydroxynaphthoquinones-PHNQs) and fibrillar collagen can be extracted to produce innovative biomaterials for biomedical applications. Can waste from other edible sea urchin species (e.g., Sphaerechinus granularis) be similarly valorised? A comparative study on PHNQs and collagen extraction was conducted. PHNQ extraction yields were compared, pigments were quantified and identified, and antioxidant activities were assessed (by ABTS assay) and correlated to specific PHNQ presence (i.e., spinochrome E). Similarly, collagen extraction yields were evaluated, and the resulting collagen-based biomaterials were compared in terms of their ultrastructure, degradation kinetics, and resistance to compression. Results showed a partially similar PHNQ profile in both species, with significantly higher yield in P. lividus, while S. granularis exhibited better antioxidant activity. P. lividus samples showed higher collagen extraction yield, but S. granularis scaffolds showed higher stability. In conclusion, waste from different species can be successfully valorised through PHNQ and collagen extraction, offering diverse applications in the biomedical field, according to specific technical requirements.
Collapse
Affiliation(s)
- Margherita Roncoroni
- Department of Environmental Science and Policy, University of Milan, Via Celoria 2, 20133 Milan, Italy; (M.R.); (G.M.); (M.S.)
| | - Giordana Martinelli
- Department of Environmental Science and Policy, University of Milan, Via Celoria 2, 20133 Milan, Italy; (M.R.); (G.M.); (M.S.)
| | - Stefano Farris
- Department of Food, Environmental and Nutritional Sciences, University of Milan, Via Celoria 2, 20133 Milan, Italy;
| | - Stefania Marzorati
- Department of Environmental Science and Policy, University of Milan, Via Celoria 2, 20133 Milan, Italy; (M.R.); (G.M.); (M.S.)
| | - Michela Sugni
- Department of Environmental Science and Policy, University of Milan, Via Celoria 2, 20133 Milan, Italy; (M.R.); (G.M.); (M.S.)
| |
Collapse
|
38
|
Alva-Ensastegui J, Morales-Avila E, de la Luz AP, Bernad-Bernad M. Determination of pKa values and deprotonation order of methotrexate using a combined experimental-theoretical study and binding constants of the methotrexate-Laponite complex at different pH values. J Photochem Photobiol A Chem 2024; 449:115406. [DOI: 10.1016/j.jphotochem.2023.115406] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
39
|
Abbadessa A, Ronca A, Salerno A. Integrating bioprinting, cell therapies and drug delivery towards in vivo regeneration of cartilage, bone and osteochondral tissue. Drug Deliv Transl Res 2024; 14:858-894. [PMID: 37882983 DOI: 10.1007/s13346-023-01437-1] [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] [Accepted: 09/18/2023] [Indexed: 10/27/2023]
Abstract
The biological and biomechanical functions of cartilage, bone and osteochondral tissue are naturally orchestrated by a complex crosstalk between zonally dependent cells and extracellular matrix components. In fact, this crosstalk involves biomechanical signals and the release of biochemical cues that direct cell fate and regulate tissue morphogenesis and remodelling in vivo. Three-dimensional bioprinting introduced a paradigm shift in tissue engineering and regenerative medicine, since it allows to mimic native tissue anisotropy introducing compositional and architectural gradients. Moreover, the growing synergy between bioprinting and drug delivery may enable to replicate cell/extracellular matrix reciprocity and dynamics by the careful control of the spatial and temporal patterning of bioactive cues. Although significant advances have been made in this direction, unmet challenges and open research questions persist. These include, among others, the optimization of scaffold zonality and architectural features; the preservation of the bioactivity of loaded active molecules, as well as their spatio-temporal release; the in vitro scaffold maturation prior to implantation; the pros and cons of each animal model and the graft-defect mismatch; and the in vivo non-invasive monitoring of new tissue formation. This work critically reviews these aspects and reveals the state of the art of using three-dimensional bioprinting, and its synergy with drug delivery technologies, to pattern the distribution of cells and/or active molecules in cartilage, bone and osteochondral engineered tissues. Most notably, this work focuses on approaches, technologies and biomaterials that are currently under in vivo investigations, as these give important insights on scaffold performance at the implantation site and its interaction/integration with surrounding tissues.
Collapse
Affiliation(s)
- Anna Abbadessa
- Center for Research in Molecular Medicine and Chronic Diseases (CiMUS), IDIS Research Institute, Universidade de Santiago de Compostela, 15782, Santiago de Compostela, Spain.
- Department of Pharmacology, Pharmacy and Pharmaceutical Technology, School of Pharmacy, Universidade de Santiago de Compostela, Campus Vida, Santiago de Compostela, Spain.
| | - Alfredo Ronca
- Institute of Polymers, Composites and Biomaterials, National Research Council, 80125, Naples, Italy.
| | - Aurelio Salerno
- Department of Chemical, Materials and Production Engineering, University of Naples Federico II, 80125, Naples, Italy.
| |
Collapse
|
40
|
Wang H, Hao Y, Guo K, Liu L, Xia B, Gao X, Zheng X, Huang J. Quantitative Biofabrication Platform for Collagen-Based Peripheral Nerve Grafts with Structural and Chemical Guidance. Adv Healthc Mater 2024; 13:e2303505. [PMID: 37988388 DOI: 10.1002/adhm.202303505] [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: 10/12/2023] [Revised: 11/14/2023] [Indexed: 11/23/2023]
Abstract
Owing to its crucial role in the human body, collagen has immense potential as a material for the biofabrication of tissues and organs. However, highly refined fabrication using collagen remains difficult, primarily because of its notably soft properties. A quantitative biofabrication platform to construct collagen-based peripheral nerve grafts, incorporating bionic structural and chemical guidance cues, is introduced. A viscoelastic model for collagen, which facilitates simulating material relaxation and fabricating collagen-based neural grafts, achieving a maximum channel density similar to that of the native nerve structure of longitudinal microchannel arrays, is established. For axonal regeneration over considerable distances, a gradient printing control model and quantitative method are developed to realize the high-precision gradient distribution of nerve growth factor required to obtain nerve grafts through one-step bioprinting. Experiments verify that the bioprinted graft effectively guides linear axonal growth in vitro and in vivo. This study should advance biofabrication methods for a variety of human tissue-engineering applications requiring tailored cues.
Collapse
Affiliation(s)
- Heran Wang
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, 110016, China
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang, 110169, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yiming Hao
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Kai Guo
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, 110016, China
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang, 110169, China
| | - Lianqing Liu
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, 110016, China
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang, 110169, China
| | - Bing Xia
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Xue Gao
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Xiongfei Zheng
- State Key Laboratory of Robotics, Shenyang Institute of Automation, Chinese Academy of Sciences, Shenyang, 110016, China
- Institutes for Robotics and Intelligent Manufacturing, Chinese Academy of Sciences, Shenyang, 110169, China
| | - Jinghui Huang
- Department of Orthopaedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| |
Collapse
|
41
|
Chen C, Li Z, Xu C, Kang M, Lee CS, Aghaloo T, Lee M. Self-Assembled Nanocomposite Hydrogels as Carriers for Demineralized Bone Matrix Particles and Enhanced Bone Repair. Adv Healthc Mater 2024; 13:e2303592. [PMID: 38275216 PMCID: PMC11023793 DOI: 10.1002/adhm.202303592] [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: 10/18/2023] [Revised: 01/17/2024] [Indexed: 01/27/2024]
Abstract
Demineralized bone matrix (DBM) has been widely used as an allogeneic alternative to autologous bone graft for bone repair. However, more extensive use of DBM is limited due to its particulate nature after demineralization and rapid particle dispersion following irrigation, resulting in unpredictable osteoinductivity. Here, a new design of injectable hydrogel carriers for DBM that combine self-healing ability and osteogenic properties based on the self-assembly of guanidinylated hyaluronic acid and silica-rich nanoclays is reported. The nanoclays serve as reversible linkages to form a dynamic hydrogel network with the guanidine moieties on the polymer chains. Gelation kinetics and mechanical properties can be controlled by altering nanoclay content in the hydrogel. The resulting hydrogel exerts self-healing ability due to its dynamic crosslinks and well retains its overall performance with high DBM loading. The hydrogel exhibits great cytocompatibility and osteogenic effects mediated by the nanoclays. In vivo delivery of DBM using the nanocomposite hydrogel further demonstrates robust bone regeneration in a mouse calvarial defect model in comparison to DBM delivered with aqueous HA. This work suggests a promising hydrogel platform for many applications including therapeutic delivery and tissue engineering.
Collapse
Affiliation(s)
- Chen Chen
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - Zhi Li
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - Changlu Xu
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - Minjee Kang
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - Chung-Sung Lee
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, CA 90095, USA
- Department of Pharmaceutical Engineering, Soonchunhyang University, Asan 31538, Republic of Korea
| | - Tara Aghaloo
- Division of Diagnostic and Surgical Sciences, School of Dentistry, University of California, Los Angeles, CA 90095, USA
| | - Min Lee
- Division of Oral and Systemic Health Sciences, School of Dentistry, University of California, Los Angeles, CA 90095, USA
- Department of Bioengineering, University of California, Los Angeles, CA 90095, USA
| |
Collapse
|
42
|
Chen YF, Lu MC, Lee CJ, Chiu CW. Flexible nanohybrid substrates utilizing gold nanocubes/nano mica platelets with 3D lightning-rod effect for highly efficient bacterial biosensors based on surface-enhanced Raman scattering. J Mater Chem B 2024; 12:3226-3239. [PMID: 38451239 DOI: 10.1039/d3tb02897f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/08/2024]
Abstract
In this study, gold nanocubes (AuNCs) were quickly synthesized using the seed-mediated growth method and reduced onto the surface of two-dimensional (2D) delaminated nano mica platelets (NMPs), enabling the development of AuNCs/NMPs nanohybrids with a 3D lightning-rod effect. First, the growth-solution amount can be changed to easily adjust the AuNCs average-particle size within a range of 30-70 nm. The use of the cationic surfactant cetyltrimethylammonium chloride as a protective agent allowed the surface of AuNCs and nanohybrids to be positively charged. Positively charged nanohybrid surfaces presented a good adsorption effect for detecting molecules with negative charges on the surface. Additionally, the NMP surfaces were rich in ionic charges and provided a large specific surface area for stabilizing the growth of AuNCs. Delaminated AuNCs/NMPs nanohybrids can generate a 3D hotspot effect through self-assembly to enhance the Raman signal. Surface-enhanced Raman scattering (SERS) is highly sensitive in detecting adenine biomolecules. Its limit of detection (LOD) and Raman enhancement factor reached 10-9 M and 3.6 × 108, respectively. Excellent reproducibility was obtained owing to the relatively regular arrangement of AuNC particles, and the relative standard deviation (RSD) was 10.7%. Finally, the surface of NMPs was modified by adding the hydrophilic poly(oxyethylene)-diamine (POE2000) and amphiphilic PIB-POE-PIB copolymer at different weight ratios. The adjustment of the surface hydrophilicity and hydrophobicity of AuNCs/NMPs nanohybrids led to better adsorption and selectivity for bacteria. AuNCs/POE/NMPs and AuNCs/PIB-POE-PIB/NMPs were further applied to the SERS detection of hydrophilic Staphylococcus aureus and hydrophobic Escherichia coli, respectively. The SERS-detection results suggest that the LOD of hydrophilic Staphylococcus aureus and hydrophobic Escherichia coli reached 92 CFU mL-1 and 1.6 × 102 CFU mL-1, respectively. The AuNCs/POE/NMPs and AuNCs/PIB-POE-PIB/NMPs nanohybrids had different hydrophilic-hydrophobic affinities, which greatly improved the selectivity and sensitivity for detecting bacteria with different hydrophilicity and hydrophobicity. Therefore, fast, highly selective, and highly sensitive SERS biological-detection results were obtained.
Collapse
Affiliation(s)
- Yan-Feng Chen
- Department of Materials Science and Engineering, National Taiwan University of Science and Technology, Taipei 10607, Taiwan.
| | - Ming-Chang Lu
- Department of Materials Science and Engineering, National Taiwan University of Science and Technology, Taipei 10607, Taiwan.
| | - Chia-Jung Lee
- Ph.D. Program in Clinical Drug Development of Herbal Medicine, College of Pharmacy, Taipei Medical University, Taipei 11031, Taiwan
| | - Chih-Wei Chiu
- Department of Materials Science and Engineering, National Taiwan University of Science and Technology, Taipei 10607, Taiwan.
| |
Collapse
|
43
|
Kafili G, Tamjid E, Niknejad H, Simchi A. Development of bioinspired nanocomposite bioinks based on decellularized amniotic membrane and hydroxyethyl cellulose for skin tissue engineering. CELLULOSE 2024; 31:2989-3013. [DOI: 10.1007/s10570-024-05797-w] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/30/2023] [Accepted: 02/08/2024] [Indexed: 01/06/2025]
|
44
|
Wu T, Li Y, Wu Z, Wang Z, Li Y, Jian K, He C, Zhang C, Shi L, Dai J. Enzyme-immobilized nanoclay hydrogel simultaneously reduces inflammation and scar deposition to treat spinal cord injury. CHEMICAL ENGINEERING JOURNAL 2024; 484:149642. [DOI: 10.1016/j.cej.2024.149642] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
45
|
Lyu J, Chen H, Luo J, Lin S, Yang G, Zhou M, Tao J. Shape memory and hemostatic silk-laponite scaffold for alveolar bone regeneration after tooth extraction trauma. Int J Biol Macromol 2024; 260:129454. [PMID: 38237836 DOI: 10.1016/j.ijbiomac.2024.129454] [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: 11/13/2023] [Revised: 01/08/2024] [Accepted: 01/10/2024] [Indexed: 01/28/2024]
Abstract
Persistent bleeding and the absence of alveolar bone stress following tooth loss can hinder socket healing, complicating future dental implant procedures, and potentially leading to neighboring tooth instability. Therefore, developing materials that promote alveolar bone regeneration and possess both hemostatic and osteogenic properties is crucial for preserving the extraction sites. This study introduces a silk-based laponite composite scaffold material with proficient hemostatic and osteogenic functions, and excellent shape-memory properties for efficient extraction- site filling. In vitro studies research demonstrated that the scaffold's inherent negative charge of the scaffold significantly enhanced blood coagulation and thrombin generation. Moreover, its porous structure and slightly rough inner surface promoted blood cell adhesion and, improved the hemostatic performance. Furthermore, the scaffold facilitated stem cell osteogenic differentiation by activating the TRPM7 channel through the released of magnesium ions. In vivo tests using rat models confirmed its effectiveness in promoting coagulation and mandibular regeneration. Thus, this study proposes a promising approach for post-extraction alveolar bone regenerative repair. The composite scaffold material, with its hemostatic and osteogenic capabilities and shape-memory features, can potentially enhance dental implant success and overall oral health.
Collapse
Affiliation(s)
- Jiaxuan Lyu
- Department of General Dentistry, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, No. 639 Zhizaoju Road, Shanghai 200011, China
| | - Hongyan Chen
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University; Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, No. 639 Zhizaoju Road, Shanghai 200011, China
| | - Jiaxin Luo
- Department of Dental Implantology, School and Hospital of Stomatology, Guangdong Engineering Research Center of Oral Restoration and Reconstruction, Guangzhou Key Laboratory of Basic and Applied Research of Oral Regenerative Medicine, Guangzhou Medical University, No. 195 Dongfengwest Road, Guangzhou 510160, China
| | - Sihan Lin
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University; Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, No. 639 Zhizaoju Road, Shanghai 200011, China
| | - Guangzheng Yang
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University; Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, No. 639 Zhizaoju Road, Shanghai 200011, China
| | - Mingliang Zhou
- Department of Prosthodontics, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University; Shanghai Engineering Research Center of Advanced Dental Technology and Materials, Shanghai Key Laboratory of Stomatology & Shanghai Research Institute of Stomatology, National Clinical Research Center for Oral Diseases, No. 639 Zhizaoju Road, Shanghai 200011, China.
| | - Jiang Tao
- Department of General Dentistry, Shanghai Ninth People's Hospital, Shanghai Jiao Tong University School of Medicine, College of Stomatology, Shanghai Jiao Tong University; National Center for Stomatology, National Clinical Research Center for Oral Diseases, Shanghai Key Laboratory of Stomatology, Shanghai Research Institute of Stomatology, Research Unit of Oral and Maxillofacial Regenerative Medicine, Chinese Academy of Medical Sciences, No. 639 Zhizaoju Road, Shanghai 200011, China.
| |
Collapse
|
46
|
Chen L, Zhang S, Duan Y, Song X, Chang M, Feng W, Chen Y. Silicon-containing nanomedicine and biomaterials: materials chemistry, multi-dimensional design, and biomedical application. Chem Soc Rev 2024; 53:1167-1315. [PMID: 38168612 DOI: 10.1039/d1cs01022k] [Citation(s) in RCA: 12] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2024]
Abstract
The invention of silica-based bioactive glass in the late 1960s has sparked significant interest in exploring a wide range of silicon-containing biomaterials from the macroscale to the nanoscale. Over the past few decades, these biomaterials have been extensively explored for their potential in diverse biomedical applications, considering their remarkable bioactivity, excellent biocompatibility, facile surface functionalization, controllable synthesis, etc. However, to expedite the clinical translation and the unexpected utilization of silicon-composed nanomedicine and biomaterials, it is highly desirable to achieve a thorough comprehension of their characteristics and biological effects from an overall perspective. In this review, we provide a comprehensive discussion on the state-of-the-art progress of silicon-composed biomaterials, including their classification, characteristics, fabrication methods, and versatile biomedical applications. Additionally, we highlight the multi-dimensional design of both pure and hybrid silicon-composed nanomedicine and biomaterials and their intrinsic biological effects and interactions with biological systems. Their extensive biomedical applications span from drug delivery and bioimaging to therapeutic interventions and regenerative medicine, showcasing the significance of their rational design and fabrication to meet specific requirements and optimize their theranostic performance. Additionally, we offer insights into the future prospects and potential challenges regarding silicon-composed nanomedicine and biomaterials. By shedding light on these exciting research advances, we aspire to foster further progress in the biomedical field and drive the development of innovative silicon-composed nanomedicine and biomaterials with transformative applications in biomedicine.
Collapse
Affiliation(s)
- Liang Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Shanshan Zhang
- Department of Ultrasound Ruijin Hospital, Shanghai Jiaotong University School of Medicine, Shanghai 200025, P. R. China
| | - Yanqiu Duan
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China.
| | - Xinran Song
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Meiqi Chang
- Laboratory Center, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, 200071, P. R. China.
| | - Wei Feng
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| | - Yu Chen
- Materdicine Lab, School of Life Sciences, Shanghai University, Shanghai, 200444, P. R. China.
| |
Collapse
|
47
|
Li S, Li X, Xu Y, Fan C, Li ZA, Zheng L, Luo B, Li ZP, Lin B, Zha ZG, Zhang HT, Wang X. Collagen fibril-like injectable hydrogels from self-assembled nanoparticles for promoting wound healing. Bioact Mater 2024; 32:149-163. [PMID: 37822915 PMCID: PMC10563012 DOI: 10.1016/j.bioactmat.2023.09.012] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Revised: 08/24/2023] [Accepted: 09/18/2023] [Indexed: 10/13/2023] Open
Abstract
Soft hydrogels are excellent candidate materials for repairing various tissue defects, yet the mechanical strength, anti-swelling properties, and biocompatibility of many soft hydrogels need to be improved. Herein, inspired by the nanostructure of collagen fibrils, we developed a strategy toward achieving a soft but tough, anti-swelling nanofibrillar hydrogel by combining the self-assembly and chemical crosslinking of nanoparticles. Specifically, the collagen fibril-like injectable hydrogel was subtly designed and fabricated by self-assembling methylacrylyl hydroxypropyl chitosan (HM) with laponite (LAP) to form nanoparticles, followed by the inter-nanoparticle bonding through photo-crosslinking. The assembly mechanism of nanoparticles was elucidated by both experimental and simulation techniques. Due to the unique structure of the crosslinked nanoparticles, the nanocomposite hydrogels exhibited low stiffness (G'< 2 kPa), high compressive strength (709 kPa), and anti-swelling (swelling ratio of 1.07 in PBS) properties. Additionally, by harnessing the photo-crosslinking ability of the nanoparticles, the nanocomposite hydrogels were processed as microgels, which can be three-dimensionally (3D) printed into complex shapes. Furthermore, we demonstrated that these nanocomposite hydrogels are highly biocompatible, biodegradability, and can effectively promote fibroblast migration and accelerate blood vessel formation during wound healing. This work presents a promising approach to develop biomimetic, nanofibrillar soft hydrogels for regenerative medicine applications.
Collapse
Affiliation(s)
- Shanshan Li
- State Key Laboratory of Pulp & Paper Engineering, South China University of Technology, 381 Wushan Road, Tianhe District, Guangzhou, 510640, China
| | - Xiaoyun Li
- State Key Laboratory of Pulp & Paper Engineering, South China University of Technology, 381 Wushan Road, Tianhe District, Guangzhou, 510640, China
| | - Yidi Xu
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Chaoran Fan
- State Key Laboratory of Pulp & Paper Engineering, South China University of Technology, 381 Wushan Road, Tianhe District, Guangzhou, 510640, China
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning, 530004, China
| | - Zhong Alan Li
- Department of Biomedical Engineering, The Chinese University of Hong Kong, Shatin, Hong Kong SAR, China
| | - Lu Zheng
- State Key Laboratory of Pulp & Paper Engineering, South China University of Technology, 381 Wushan Road, Tianhe District, Guangzhou, 510640, China
| | - Bichong Luo
- State Key Laboratory of Pulp & Paper Engineering, South China University of Technology, 381 Wushan Road, Tianhe District, Guangzhou, 510640, China
| | - Zhi-Peng Li
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Baofeng Lin
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning, 530004, China
| | - Zhen-Gang Zha
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Huan-Tian Zhang
- Department of Bone and Joint Surgery, The First Affiliated Hospital of Jinan University, Guangzhou, 510632, China
| | - Xiaoying Wang
- State Key Laboratory of Pulp & Paper Engineering, South China University of Technology, 381 Wushan Road, Tianhe District, Guangzhou, 510640, China
| |
Collapse
|
48
|
Rodrigo MJ, Cardiel MJ, Fraile JM, Mayoral JA, Pablo LE, Garcia-Martin E. Laponite for biomedical applications: An ophthalmological perspective. Mater Today Bio 2024; 24:100935. [PMID: 38239894 PMCID: PMC10794930 DOI: 10.1016/j.mtbio.2023.100935] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 12/20/2023] [Accepted: 12/27/2023] [Indexed: 01/22/2024] Open
Abstract
Clay minerals have been applied in biomedicine for thousands of years. Laponite is a nanostructured synthetic clay with the capacity to retain and progressively release drugs. In recent years there has been a resurgence of interest in Laponite application in various biomedical areas. This is the first paper to review the potential biomedical applications of Laponite in ophthalmology. The introduction briefly covers the physical, chemical, rheological, and biocompatibility features of different routes of administration. After that, emphasis is placed on 1) drug delivery for antibiotics, anti-inflammatories, growth factors, other proteins, and cancer treatment; 2) bleeding prevention or treatment; and 3) tissue engineering through regenerative medicine using scaffolds in intraocular and extraocular tissue. Although most scientific research is not performed on the eye, both the findings and the new treatments resulting from that research are potentially applicable in ophthalmology since many of the drugs used are the same, the tissue evaluated in vitro or in vivo is also present in the eye, and the pathologies treated also occur in the eye. Finally, future prospects for this emerging field are discussed.
Collapse
Affiliation(s)
- Maria J. Rodrigo
- Department of Ophthalmology, Miguel Servet University Hospital, Zaragoza, Spain
- Aragon Institute for Health Research (IIS Aragon), GIMSO Research Group, University of Zaragoza (Spain), Avda. San Juan Bosco 13, E-50009 Zaragoza, Spain
| | - Maria J. Cardiel
- Aragon Institute for Health Research (IIS Aragon), GIMSO Research Group, University of Zaragoza (Spain), Avda. San Juan Bosco 13, E-50009 Zaragoza, Spain
- Department of Pathology, Lozano Blesa University Hospital, Zaragoza, Spain
| | - Jose M. Fraile
- Institute for Chemical Synthesis and Homogeneous Catalysis (ISQCH), Faculty of Sciences, University of Zaragoza–CSIC, C/Pedro Cerbuna 12, 50009 Zaragoza, Spain
| | - Jose A. Mayoral
- Institute for Chemical Synthesis and Homogeneous Catalysis (ISQCH), Faculty of Sciences, University of Zaragoza–CSIC, C/Pedro Cerbuna 12, 50009 Zaragoza, Spain
| | - Luis E. Pablo
- Department of Ophthalmology, Miguel Servet University Hospital, Zaragoza, Spain
- Aragon Institute for Health Research (IIS Aragon), GIMSO Research Group, University of Zaragoza (Spain), Avda. San Juan Bosco 13, E-50009 Zaragoza, Spain
- Biotech Vision SLP (spin-off Company), University of Zaragoza, Spain
| | - Elena Garcia-Martin
- Department of Ophthalmology, Miguel Servet University Hospital, Zaragoza, Spain
- Aragon Institute for Health Research (IIS Aragon), GIMSO Research Group, University of Zaragoza (Spain), Avda. San Juan Bosco 13, E-50009 Zaragoza, Spain
| |
Collapse
|
49
|
Wei W, Wang M, Liu Z, Zheng W, Tremblay PL, Zhang T. An antibacterial nanoclay- and chitosan-based quad composite with controlled drug release for infected skin wound healing. Carbohydr Polym 2024; 324:121507. [PMID: 37985094 DOI: 10.1016/j.carbpol.2023.121507] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 10/07/2023] [Accepted: 10/16/2023] [Indexed: 11/22/2023]
Abstract
Microbial infections of surgical sites and other wounds represent a major impediment for patients. Multifunctional low-cost dressings promoting tissue reparation while preventing infections are of great interest to medical professionals. Here, clay-based laponite nanodiscs (LAP) were loaded with the antibacterial drug kanamycin (KANA) before being embedded into a poly(lactic-co-glycolic acid) (PLGA) membrane and coated with the biopolymer chitosan (CS). Results indicated that these biocompatible materials combined the excellent capacity of LAP for controlled drug release with the mechanical robustness of PLGA and the antibacterial properties of CS as well as its hydrophilicity to form a composite highly suitable as an infection-preventing wound dressing. In vitro, PLGA/LAP/KANA/CS released drugs in a sustainable manner over 30 d, completely inhibited the growth of infectious bacteria, prompted the adhesion fibroblasts, and accelerated their proliferation 1.3 times. In vivo, the composite enabled the fast healing of infected full-thickness skin wounds with a 96.19 % contraction after 14 d. During the healing process, PLGA/LAP/KANA/CS stimulated re-epithelization, reduced inflammation, and promoted both angiogenesis and the formation of dense collagen fibers with an excellent final collagen volume ratio of 89.27 %. Thus, multifunctional PLGA/LAP/KANA/CS made of low-cost components demonstrated its potential for the treatment of infected skin wounds.
Collapse
Affiliation(s)
- Wenlong Wei
- School of Chemistry, Chemical Engineering, and Life Science, Wuhan University of Technology, Wuhan 430070, PR China
| | - Mayue Wang
- School of Chemistry, Chemical Engineering, and Life Science, Wuhan University of Technology, Wuhan 430070, PR China
| | - Ziru Liu
- School of Materials Science and Engineering, Wuhan University of Technology, Wuhan 430070, PR China; Sanya Science and Education Innovation Park, Wuhan University of Technology, Sanya 572024, PR China
| | - Wen Zheng
- School of Chemistry, Chemical Engineering, and Life Science, Wuhan University of Technology, Wuhan 430070, PR China
| | - Pier-Luc Tremblay
- School of Chemistry, Chemical Engineering, and Life Science, Wuhan University of Technology, Wuhan 430070, PR China; Sanya Science and Education Innovation Park, Wuhan University of Technology, Sanya 572024, PR China; Shaoxing Institute for Advanced Research, Wuhan University of Technology, Shaoxing 312300, PR China.
| | - Tian Zhang
- School of Chemistry, Chemical Engineering, and Life Science, Wuhan University of Technology, Wuhan 430070, PR China; School of Materials Science and Engineering, Wuhan University of Technology, Wuhan 430070, PR China; Sanya Science and Education Innovation Park, Wuhan University of Technology, Sanya 572024, PR China; Shaoxing Institute for Advanced Research, Wuhan University of Technology, Shaoxing 312300, PR China.
| |
Collapse
|
50
|
Wang X, Yang X, Sun Z, Guo X, Teng Y, Hou S, Shi J, Lv Q. Progress in injectable hydrogels for the treatment of incompressible bleeding: an update. Front Bioeng Biotechnol 2024; 11:1335211. [PMID: 38264581 PMCID: PMC10803650 DOI: 10.3389/fbioe.2023.1335211] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Accepted: 12/26/2023] [Indexed: 01/25/2024] Open
Abstract
Uncontrollable haemorrhage from deep, noncompressible wounds remains a persistent and intractable challenge, accounting for a very high proportion of deaths in both war and disaster situations. Recently, injectable hydrogels have been increasingly studied as potential haemostatic materials, highlighting their enormous potential for the management of noncompressible haemorrhages. In this review, we summarize haemostatic mechanisms, commonly used clinical haemostatic methods, and the research progress on injectable haemostatic hydrogels. We emphasize the current status of injectable hydrogels as haemostatic materials, including their physical and chemical properties, design strategy, haemostatic mechanisms, and application in various types of wounds. We discuss the advantages and disadvantages of injectable hydrogels as haemostatic materials, as well as the opportunities and challenges involved. Finally, we propose cutting-edge research avenues to address these challenges and opportunities, including the combination of injectable hydrogels with advanced materials and innovative strategies to increase their biocompatibility and tune their degradation profile. Surface modifications for promoting cell adhesion and proliferation, as well as the delivery of growth factors or other biologics for optimal wound healing, are also suggested. We believe that this paper will inform researchers about the current status of the use of injectable haemostatic hydrogels for noncompressible haemorrhage and spark new ideas for those striving to propel this field forward.
Collapse
Affiliation(s)
- Xiudan Wang
- Institution of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou, China
- Key Laboratory for Disaster Medicine Technology, Tianjin, China
| | - Xinran Yang
- Institution of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou, China
- Key Laboratory for Disaster Medicine Technology, Tianjin, China
| | - Zhiguang Sun
- Institution of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou, China
- Key Laboratory for Disaster Medicine Technology, Tianjin, China
| | - Xiaoqin Guo
- Institution of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Key Laboratory for Disaster Medicine Technology, Tianjin, China
| | - Yanjiao Teng
- Institution of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou, China
- Key Laboratory for Disaster Medicine Technology, Tianjin, China
| | - Shike Hou
- Institution of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou, China
- Key Laboratory for Disaster Medicine Technology, Tianjin, China
| | - Jie Shi
- Institution of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou, China
- Key Laboratory for Disaster Medicine Technology, Tianjin, China
| | - Qi Lv
- Institution of Disaster and Emergency Medicine, Tianjin University, Tianjin, China
- Wenzhou Safety (Emergency) Institute of Tianjin University, Wenzhou, China
- Key Laboratory for Disaster Medicine Technology, Tianjin, China
| |
Collapse
|