1
|
Chu Z, Liu X, Zhao T, Jiang D, Zhao J, Dong X, Yeung KWK, Liu X, Liao Y, Ouyang L. Self-healing Ppy-hydrogel promotes diabetic skin wound healing through enhanced sterilization and macrophage orchestration triggered by NIR. Biomaterials 2025; 315:122964. [PMID: 39550986 DOI: 10.1016/j.biomaterials.2024.122964] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 11/10/2024] [Accepted: 11/11/2024] [Indexed: 11/19/2024]
Abstract
Non-healing diabetic foot ulcers are the knotty public health issue due to the uncontrolled bacterial infection, prolonged inflammation, and inferior vessel remodeling. In this work, polypyrrole (Ppy) was added into the hybrid hydrogel containing polyvinyl alcohol (PVA), polyethylene glycol (PEG), and hyaluronan (HA) to acquire superior mechanism and photothermal ability. The Ppy composited hybrid hydrogel could effectively kill bacteria through accumulating heat on the hydrogel surface. RNA-Seq analysis shows that the heat accumulation could enhance phagosome of macrophage and M1 activation, which further accelerate bacteria clearance. Benefitting from the bacteria clearance, macrophage could transform its phenotype to M2 in Ppy composited hybrid hydrogel group with near infrared light (NIR) stimulation. The related genes expression in keratinization, keratinocyte differentiation, and establishment of the skin barrier in the skin were up-regulated and collagen and vascular endothelial growth factor (VEGF) expression level are also enhanced. In summary, Ppy composited hybrid hydrogel could effectively solve the issues of infection and poor wound healing in diabetic foot ulcers, making it an ideal candidate dressing for the treatment of chronic wounds.
Collapse
Affiliation(s)
- Zhuangzhuang Chu
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China; Department of Dental Implantology, Linyi People's Hospital, Linyi, 276003, China
| | - Xingdan Liu
- Department of Pharmacy, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China; Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China; State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China
| | - Tong Zhao
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China; Department of Oral and Maxillo-facial Implantology, Shanghai Ninth People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200011, China
| | - Dongya Jiang
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Jing Zhao
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Xiaohua Dong
- Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Kelvin W K Yeung
- Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, Guangdong Engineering Technology Research Center for Orthopaedic Trauma Repair, Department of Orthopaedics and Traumatology, The University of Hong Kong Shenzhen Hospital, Shenzhen, China
| | - Xuanyong Liu
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure, Shanghai Institute of Ceramics, Chinese Academy of Sciences, Shanghai, 200050, China.
| | - Yun Liao
- Department of Pharmacy, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China.
| | - Liping Ouyang
- Department of Pharmacy, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200336, China.
| |
Collapse
|
2
|
Zhang H, Faber J, Budday S, Gao Q, Kuth S, Zheng K, Boccaccini AR. Monophasic hyaluronic acid-silica hybrid hydrogels for articular cartilage applications. BIOMATERIALS ADVANCES 2025; 167:214089. [PMID: 39546842 DOI: 10.1016/j.bioadv.2024.214089] [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: 05/03/2024] [Revised: 09/21/2024] [Accepted: 10/25/2024] [Indexed: 11/17/2024]
Abstract
Hyaluronic acid (HA), an FDA-approved natural polymer and important component of the extracellular matrix (ECM), has been widely used to develop hydrogels for cartilage regeneration. However, HA hydrogels often exhibit poor mechanical properties and unsuitable degradability, limiting their capability to support cell growth in cartilage. To overcome these challenges, this study modifies HA with a silica precursor and the coupling agent (3-Glycidyloxypropyl) trimethoxysilane (GPTMS) to develop a monophasic organic-inorganic hybrid HA-silica hydrogel. In this system, the inorganic silicate and organic HA components interpenetrate and bond covalently at the molecular level. The HA-silica hybrid hydrogel achieves a compressive modulus of 143 kPa at the highest GPTMS/HA molar ratio of 400. Additionally, in vitro cell studies show that these hybrid hydrogels have no cytotoxicity against MC3T3-E1 and ATDC-5 cells. Cell viability and morphology tests further confirm excellent cell adhesion on the hybrid scaffold. These results indicate that the developed HA-silica hybrid hydrogel is a suitable candidate for cartilage regeneration applications.
Collapse
Affiliation(s)
- Huijun Zhang
- Institute of Biomaterials, Friedrich-Alexander-Universität Erlangen-Nuremberg, Erlangen, Germany
| | - Jessica Faber
- Institute of Continuum Mechanics and Biomechanics, Friedrich-Alexander-Universität Erlangen-Nuremberg, Erlangen, Germany
| | - Silvia Budday
- Institute of Continuum Mechanics and Biomechanics, Friedrich-Alexander-Universität Erlangen-Nuremberg, Erlangen, Germany
| | - Qingsen Gao
- Institute of Polymer Materials, Friedrich-Alexander-Universität Erlangen-Nuremberg, Erlangen, Germany
| | - Sonja Kuth
- Institute of Biomaterials, Friedrich-Alexander-Universität Erlangen-Nuremberg, Erlangen, Germany
| | - Kai Zheng
- Jiangsu Engineering Research Center of Stomatological Translational Medicine, Nanjing Medical University, Nanjing 210029, China; Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, Nanjing 210029, China.
| | - Aldo R Boccaccini
- Institute of Biomaterials, Friedrich-Alexander-Universität Erlangen-Nuremberg, Erlangen, Germany.
| |
Collapse
|
3
|
Qin L, Tu J, Zhao J, Zhang Y, Li T, Zhang Y, Zhang P, Ling G, Ji J. Dual-targeted and esterase-responsive cyclodextrin-based host-guest nanocomposites for enhanced antitumor therapy. Colloids Surf B Biointerfaces 2025; 246:114371. [PMID: 39551034 DOI: 10.1016/j.colsurfb.2024.114371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 10/28/2024] [Accepted: 11/07/2024] [Indexed: 11/19/2024]
Abstract
Conventional chemotherapy drugs are difficult to effectively target tumor tissue, leading to poor treatment outcomes and side effects. Actively targeted and stimuli-responsive nanomedicine greatly improves this situation, allowing for more precise drug accumulation at tumor sites. Herein, carboxymethyl-β-cyclodextrin (CMCD) - based host-guest nanocomposites (NPs) encapsulating hydroxycamptothecin (HCPT) were fabricated, which responded to esterase and had the function of targeting CD 44 receptors and the nucleus. PS-CMCD was firstly synthesized through an amide reaction of protamine (PS) and CMCD to enhance the function of penetrating membrane and nuclear localization. PS-CMCD/HCPT/HA NPs were then prepared by the host-guest complexation of PS-CMCD and HCPT and followed by surface modification of hyaluronic acid (HA) with CD44 receptor-targeting properties. The successful inclusion was also validated through computer simulation. The obtained nanocomposites displayed the esterase-responsive release behaviors of HCPT. Moreover, the synthesized PS-CMCD/HCPT/HA NPs enhanced the intracellular drug uptake due to the tumor cell- and nuclear-mediated targeting. In addition, in vivo application exhibited that PS-CMCD/HCPT/HA NPs realized good antitumor effects. These findings suggested its potential for targeted delivery and more effective tumor therapy.
Collapse
Affiliation(s)
- Li Qin
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Jianfei Tu
- College of Medical Devices, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Jiawei Zhao
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Yuanke Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Tiancheng Li
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China
| | - Yuqi Zhang
- College of Medical Devices, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China
| | - Peng Zhang
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China.
| | - Guixia Ling
- Wuya College of Innovation, Shenyang Pharmaceutical University, No. 103, Wenhua Road, Shenyang 110016, China; College of Medical Devices, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, China.
| | - Jiansong Ji
- Zhejiang Provincial Key Laboratory of Imaging Diagnosis and Minimally Invasive Intervention Research, The Fifth Affiliated Hospital of Wenzhou Medical University, Lishui 323000, China.
| |
Collapse
|
4
|
Chen M, Liu Y, Cao Y, Zhao C, Liu Q, Li N, Liu Y, Cui X, Liu P, Liang J, Fan Y, Wang Q, Zhang X. Remodeling the Proinflammatory Microenvironment in Osteoarthritis through Interleukin-1 Beta Tailored Exosome Cargo for Inflammatory Regulation and Cartilage Regeneration. ACS NANO 2025. [PMID: 39848926 DOI: 10.1021/acsnano.4c16785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/25/2025]
Abstract
Osteoarthritis (OA) presents a significant therapeutic challenge, with few options for preserving joint cartilage and repairing associated tissue damage. Inflammation is a pivotal factor in OA-induced cartilage deterioration and synovial inflammation. Recently, exosomes derived from human umbilical cord mesenchymal stem cells (HucMSCs) have gained recognition as a promising noncellular therapeutic modality, but their use is hindered by the challenge of harvesting a sufficient number of exosomes with effective therapeutic efficacy. Given that HucMSCs are highly sensitive to microenvironmental signals, we hypothesized that priming HucMSCs within a proinflammatory environment would increase the number of exosomes secreted with enhanced anti-inflammatory properties. Subsequent miRNA profiling and pathway analysis confirmed that interleukin-1 beta (IL-1β)-induced exosomes (C-Exos) exert positive effects through miRNA regulation and signaling pathway modulation. In vitro experiments revealed that C-Exos enhance chondrocyte functionality and cartilage matrix production, as well as macrophage polarization, thereby enhancing cartilage repair. C-Exos were encapsulated in hyaluronic acid hydrogel microspheres (HMs) to ensure sustained release, leading to substantial improvements in the inflammatory microenvironment and cartilage regeneration in a rat OA model. This study outlines a strategy to tailor exosome cargo for anti-inflammatory and cartilage regenerative purposes, with the functionalized HMs demonstrating potential for OA treatment.
Collapse
Affiliation(s)
- Manyu Chen
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
| | - Yuhan Liu
- The Third Affiliated Hospital of Jinzhou Medical University, Jinzhou, Liaoning 121000, China
| | - Yi Cao
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
| | - Chengkun Zhao
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
| | - Quanying Liu
- Institute of Rocket Force Medicine, State Key Laboratory of Trauma, Burns and Combined Injury, Third Military Medical University (Army Medical University), Chongqing 400038, China
| | - Na Li
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
- Sichuan Testing Center for Biomaterials and Medical Devices, Sichuan University, 29 Wangjiang Road, Chengdu 610000, China
| | - Yuan Liu
- Orthopedics Research Institute, Department of Orthopedics, West China Hospital, Sichuan University, Chengdu 610041, P. R. China
| | - Xiaolin Cui
- Cardiac and Osteochondral Tissue Engineering (COTE) Group, School of medicine, the Chinese University of Hong Kong, Shenzhen 518172, China
- Department of Orthopedic Surgery & Musculoskeletal Medicine, Centre for Bioengineering & Nanomedicine, University of Otago, Christchurch 8011, New Zealand
| | - Pengcheng Liu
- Chengdu Xiangyakanglin Biotechnology Co., Ltd, Chengdu 610213, China
| | - Jie Liang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
- Sichuan Testing Center for Biomaterials and Medical Devices, Sichuan University, 29 Wangjiang Road, Chengdu 610000, China
| | - Yujiang Fan
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
| | - Qiguang Wang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
| | - Xingdong Zhang
- National Engineering Research Center for Biomaterials, College of Biomedical Engineering, Sichuan University, 29 Wangjiang Road, Chengdu 610064, P. R. China
| |
Collapse
|
5
|
Chen S, Hu C, Lu W, Zhang J. A lubcan cross-linked polyethylene glycol dimethyl ether hydrogel for hyaluronic acid replacement as soft tissue engineering fillers. Int J Biol Macromol 2025; 298:140061. [PMID: 39832602 DOI: 10.1016/j.ijbiomac.2025.140061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/07/2025] [Accepted: 01/17/2025] [Indexed: 01/22/2025]
Abstract
The structure of soft tissues is often destroyed by injury and aging. Injectable fillers eliminate the need for surgery and enhance repair. Hyaluronic acid-based hydrogels are commonly employed for their effectiveness and biocompatibility. However, hyaluronidase breaks them down quickly. Lubcan, a naturally sourced microbial extracellular polysaccharide, has demonstrated significant water absorption and retention capabilities, as well as lubricating properties comparable to those of hyaluronic acid. In this study, a novel injectable and implantable hydrogel was created from lubcan by adding polyethylene glycol diglycidyl ether as a cross-linking agent. Lubcan hydrogels exhibit exceptional thermal stability, favorable swelling behavior, in vitro degradation, compressive strength, injectability, and rheological properties, all while preserving the integrity of their three-dimensional porous structure. In vitro tests indicated that the lubcan hydrogel was non-cytotoxic, did not adhere to blood cells, and exhibited good hemocompatibility. Compared to the subcutaneous injection of commercially available hyaluronic acid hydrogels, lubcan hydrogels demonstrated superior integrity, persistence, and a softer texture in Balb/c mice after 16 weeks. At the same time, lubcan hydrogel is non-toxic to organs, does not affect blood biochemical test values, and is non-immunogenic in mice. These findings suggest that lubcan hydrogel may be a promising new superficial soft tissue filler.
Collapse
Affiliation(s)
- Silu Chen
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing 210094, China; Key Laboratory of Metabolic Engineering and Biosynthesis Technology, Ministry of Industry and Information Technology, 210094, China
| | - Chengtao Hu
- Nanjing Southern Element Biotechnology Co., Ltd, Nanjing 211899, China
| | - Weiling Lu
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing 210094, China; Key Laboratory of Metabolic Engineering and Biosynthesis Technology, Ministry of Industry and Information Technology, 210094, China
| | - Jianfa Zhang
- Center for Molecular Metabolism, Nanjing University of Science & Technology, Nanjing 210094, China; Key Laboratory of Metabolic Engineering and Biosynthesis Technology, Ministry of Industry and Information Technology, 210094, China.
| |
Collapse
|
6
|
Wang L, Wang Y, Peng S, Li S, Wu S. Study on Highly Sensitive Capacitive Pressure Sensor Based on Silk Fibroin-Lignin Nanoparticles Hydrogel. Biomacromolecules 2025. [PMID: 39789849 DOI: 10.1021/acs.biomac.4c01334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2025]
Abstract
Silk fibroin (SF) hydrogel has been proven to have excellent applications in the field of pressure sensors, but its sensing performance still needs improvement. A flexible hydrogel prepared from natural macromolecular materials was developed, and lignin nanoparticles (LNPs) were introduced during the preparation of the SF hydrogel. When LNPs account for 3% of SF, the sensing unit of the SF-LNPs3% hydrogel exhibits high stress sensitivity (1.32 kPa-1), fast response speed (<0.1 s), and superior cycle stability (≥8000 cycles). The sensor can detect human motion information, such as finger bending, elbow bending, and pulse signals. When worn at the vocal cord position, it can detect the peak value of the characteristic signal during the wearer speaks. This work demonstrates that the SF-LNPs3% hydrogel has high sensitivity and shows great potential in the field of pressure sensors.
Collapse
Affiliation(s)
- Lei Wang
- Hubei Provincial Key Laboratory of Green Materials for Light Industry, Hubei University of Technology, Wuhan 430068, China
| | - Yue Wang
- Hubei Provincial Key Laboratory of Green Materials for Light Industry, Hubei University of Technology, Wuhan 430068, China
| | - Simin Peng
- Hubei Provincial Key Laboratory of Green Materials for Light Industry, Hubei University of Technology, Wuhan 430068, China
| | - Shuangyan Li
- Hubei Provincial Key Laboratory of Green Materials for Light Industry, Hubei University of Technology, Wuhan 430068, China
| | - Silong Wu
- Hubei Provincial Key Laboratory of Green Materials for Light Industry, Hubei University of Technology, Wuhan 430068, China
| |
Collapse
|
7
|
Lei P, Yu H, Jiang T, Ma J, Du J, Fang Y, Wang H, Chen R, Yang Q, Cheng Y, Wu W, Sun D. Development of a sodium hyaluronate-enriched therapeutic formulation with stevia glycoside and mogroside V for the comprehensive management of diabetes and its complications. Int J Biol Macromol 2025; 293:139487. [PMID: 39756763 DOI: 10.1016/j.ijbiomac.2025.139487] [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: 10/13/2024] [Revised: 12/27/2024] [Accepted: 01/02/2025] [Indexed: 01/07/2025]
Abstract
Diabetes prevalence continues to increase as a result of people's increasing sugar intake. Diabetes mellitus and its complications (dry skin, constipation, depression, and dental caries), as well as the prohibition of sweets ingestion, seriously affect patients' physical and mental health. Therefore, it is crucial to develop a long-term food for special medical purposes (FSMP) that aids in managing diabetes and its complications. To ensure effective biomedical function and taste, we developed a FSMP beverage formulation containing stevia glycoside, mogroside V, and sodium hyaluronate (SMH-B), each at a concentration of 0.1 mg/mL. Meanwhile, this study verified that SMH-B is an environmentally friendly and biocompatible formulation. Furthermore, both in vivo and in vitro studies have demonstrated that SMH-B significantly lowers blood glucose and lipid levels, enhances skin moisture and elasticity, prevents dental caries, alleviates constipation, reduces oxidative stress, and mitigates depressive symptoms. Notably, the SMH-B compound formula exhibits a more effective adjuvant therapeutic effect compared to single-ingredient formulation composed of stevia glycosides, mogroside V, and sodium hyaluronate. Moreover, SMH-B provides the sweetness desired by diabetic patients without affecting blood glucose levels, while also offering an auxiliary therapeutic role, making it a potential FSMP for diabetes management.
Collapse
Affiliation(s)
- Pengyu Lei
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Haiyang Yu
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Tao Jiang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Jiahui Ma
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Jiao Du
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Yimeng Fang
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China
| | - Hanbing Wang
- Department of Biotechnology, The University of Hong Kong, 999077, Hong Kong
| | - Rongbing Chen
- Department of Biomedical Engineering, City University of Hong Kong, 999077, Hong Kong
| | - Qinsi Yang
- Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou 325000, China
| | - Yongwei Cheng
- National Engineering Research Center of Cell Growth Factor Drugs and Protein Biologics, Wenzhou Medical University, Wenzhou 325000, China; MedTech (Wenzhou) Health Innovation Achievement Transformation Institute, Wenzhou Institute of Industry & Science, Wenzhou 325000, China.
| | - Wei Wu
- Key Laboratory for Biorheological Science and Technology of Ministry of Education, State and Local Joint Engineering Laboratory for Vascular Implants, Bioengineering College of Chongqing University, Chongqing 400044, China.
| | - Da Sun
- Institute of Life Sciences & Biomedical Collaborative Innovation Center of Zhejiang Province, Wenzhou University, Wenzhou 325035, China.
| |
Collapse
|
8
|
Chen H, Zhang R, Zhang G, Liang X, Xu C, Li Y, Xu FJ. Naturally Inspired Tree-Ring Structured Dressing Provides Sustained Wound Tightening and Accelerates Closure. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2410845. [PMID: 39533478 DOI: 10.1002/adma.202410845] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/18/2024] [Indexed: 11/16/2024]
Abstract
Mechanically regulated wound dressings require a rational combination of contraction and adhesion functions as well as balancing exudate-induced swelling issues. However, many of the reported dressings face the dilemma of impaired function and impeded wound self-contraction due to fluid-absorbing swelling. In this study, inspired by the tree ring, a core-ring structured hydrogel dressing capable of mechanical modulation is designed, and prepare it using a simple two-step photopolymerization process. The core covers the center of the wound, contracts spontaneously at body temperature to generate a contractile force of 3.4 kPa, and resists swelling. Meanwhile, the ring adheres to the normal epidermis around the wound and transfers the contraction stress to the wound edge. The integration of a functionally independent core and ring ultimately achieves effective wound traction and avoids dressing swelling. In murine and porcine skin wound-healing models, this hydrogel with a closely connected core and ring promotes healing by accelerating epidermal closure (50% closure in mouse skin on day 2, 85% closure in pig skin on day 8), collagen deposition, vascular maturation, and extracellular matrix remodeling. These results can guide further research on mechanical force modulation in wound healing, with the potential for clinical translation.
Collapse
Affiliation(s)
- Honggui Chen
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Rui Zhang
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Guo Zhang
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Xiaoyang Liang
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Chen Xu
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Yang Li
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| | - Fu-Jian Xu
- State Key Laboratory of Chemical Resource Engineering, Key Lab of Biomedical Materials of Natural Macromolecules (Beijing University of Chemical Technology, Ministry of Education), Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing, 100029, China
| |
Collapse
|
9
|
Dini C, Borges MHR, Malheiros SS, Piazza RD, van den Beucken JJJP, de Avila ED, Souza JGS, Barão VAR. Progress in Designing Therapeutic Antimicrobial Hydrogels Targeting Implant-associated Infections: Paving the Way for a Sustainable Platform Applied to Biomedical Devices. Adv Healthc Mater 2025; 14:e2402926. [PMID: 39440583 DOI: 10.1002/adhm.202402926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 09/19/2024] [Indexed: 10/25/2024]
Abstract
Implantable biomedical devices have found widespread use in restoring lost functions or structures within the human body, but they face a significant challenge from microbial-related infections, which often lead to implant failure. In this context, antimicrobial hydrogels emerge as a promising strategy for treating implant-associated infections owing to their tunable physicochemical properties. However, the literature lacks a comprehensive analysis of antimicrobial hydrogels, encompassing their development, mechanisms, and effect on implant-associated infections, mainly in light of existing in vitro, in vivo, and clinical evidence. Thus, this review addresses the strategies employed by existing studies to tailor hydrogel properties to meet the specific needs of each application. Furthermore, this comprehensive review critically appraises the development of antimicrobial hydrogels, with a particular focus on solving infections related to metallic orthopedic or dental implants. Then, preclinical and clinical studies centering on providing quantitative microbiological results associated with the application of antimicrobial hydrogels are systematically summarized. Overall, antimicrobial hydrogels benefit from the tunable properties of polymers and hold promise as an effective strategy for the local treatment of implant-associated infections. However, future clinical investigations, grounded on robust evidence from in vitro and preclinical studies, are required to explore and validate new antimicrobial hydrogels for clinical use.
Collapse
Affiliation(s)
- Caroline Dini
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, Universidade Estadual de Campinas (UNICAMP), Piracicaba, São Paulo, 13414-903, Brazil
| | - Maria Helena Rossy Borges
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, Universidade Estadual de Campinas (UNICAMP), Piracicaba, São Paulo, 13414-903, Brazil
| | - Samuel Santana Malheiros
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, Universidade Estadual de Campinas (UNICAMP), Piracicaba, São Paulo, 13414-903, Brazil
| | - Rodolfo Debone Piazza
- Physical Chemistry Department, Institute of Chemistry, São Paulo State University (UNESP), Araraquara, São Paulo, 14800-900, Brazil
| | | | - Erica Dorigatti de Avila
- Department of Dental Materials and Prosthodontics, School of Dentistry at Araçatuba, São Paulo State University (UNESP), Araçatuba, São Paulo, 16015-050, Brazil
| | - João Gabriel S Souza
- Dental Research Division, Guarulhos University (UNG), Guarulhos, São Paulo, 07023-070, Brazil
| | - Valentim A R Barão
- Department of Prosthodontics and Periodontology, Piracicaba Dental School, Universidade Estadual de Campinas (UNICAMP), Piracicaba, São Paulo, 13414-903, Brazil
| |
Collapse
|
10
|
Li A, Nicolas J, Mura S. Unlocking the Potential of Hybrid Nanocomposite Hydrogels: Design, Mechanical Properties and Biomedical Performances. ADVANCED FUNCTIONAL MATERIALS 2025; 35. [DOI: 10.1002/adfm.202409670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Indexed: 01/06/2025]
Abstract
AbstractHybrid nanocomposite hydrogels consist of the homogeneous incorporation of nano‐objects in a hydrogel matrix. The latter, whether made of natural or synthetic materials, possesses a microporous, soft structure that makes it an ideal host for a variety of polymer and lipid‐based nano‐objects as well as metal‐ and silica‐based ones. By carefully choosing the composition and the proportions of the different constituents, hybrid hydrogels can display a wide array of properties, from simple enhancement of mechanical characteristics to specific bioactivity. This review aims to provide an overview of the state of the art in hybrid hydrogels highlighting key aspects that make them a promising choice for a variety of biomedical applications. Strategies for the preparation of hybrid hydrogels are discussed by covering the selection of individual components. The review will also explore the physico‐chemical and rheological characterization of these materials, which is essential for understanding their structure and function, ultimately satisfying specifications for the intended use. Successful examples of biomedical applications will also be presented, and the main challenges to be met will be discussed, with the aim of stimulating the research community to exploit the full potential of these materials.
Collapse
Affiliation(s)
- Anqi Li
- Université Paris‐Saclay CNRS Institut Galien Paris‐Saclay Orsay 91400 France
| | - Julien Nicolas
- Université Paris‐Saclay CNRS Institut Galien Paris‐Saclay Orsay 91400 France
| | - Simona Mura
- Université Paris‐Saclay CNRS Institut Galien Paris‐Saclay Orsay 91400 France
| |
Collapse
|
11
|
Pal P, Sambhakar S, Paliwal S. Revolutionizing Ophthalmic Care: A Review of Ocular Hydrogels from Pathologies to Therapeutic Applications. Curr Eye Res 2025; 50:1-17. [PMID: 39261982 DOI: 10.1080/02713683.2024.2396385] [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: 10/08/2023] [Revised: 08/12/2024] [Accepted: 08/20/2024] [Indexed: 09/13/2024]
Abstract
PURPOSE This comprehensive review is designed to elucidate the transformative role and multifaceted applications of ocular hydrogels in contemporary ophthalmic therapeutic strategies, with a particular emphasis on their capability to revolutionize drug delivery mechanisms and optimize patient outcomes. METHODS A systematic and structured methodology is employed, initiating with a succinct exploration of prevalent ocular pathologies and delineating the corresponding therapeutic agents. This serves as a precursor for an extensive examination of the diverse methodologies and fabrication techniques integral to the design, development, and application of hydrogels specifically tailored for ophthalmic pharmaceutical delivery. The review further scrutinizes the pivotal manufacturing processes that significantly influence hydrogel efficacy and delves into an analysis of the current spectrum of hydrogel-centric ocular formulations. RESULTS The review yields illuminating insights into the escalating prominence of ocular hydrogels within the medical community, substantiated by a plethora of ongoing clinical investigations. It reveals the dynamic and perpetually evolving nature of hydrogel research and underscores the extensive applicability and intricate progression of transposing biologics-loaded hydrogels from theoretical frameworks to practical clinical applications. CONCLUSIONS This review accentuates the immense potential and promising future of ocular hydrogels in the realm of ophthalmic care. It not only serves as a comprehensive guide but also as a catalyst for recognizing the transformative potential of hydrogels in augmenting drug delivery mechanisms and enhancing patient outcomes. Furthermore, it draws attention to the inherent challenges and considerations that necessitate careful navigation by researchers and clinicians in this progressive field.
Collapse
Affiliation(s)
- Pankaj Pal
- Department of Pharmacy, Banasthali Vidyapith, Vanasthali, India
- IIMT College of Pharmacy, IIMT Group of Colleges, Greater Noida, India
| | | | | |
Collapse
|
12
|
Maity S, Bhuyan T, Jewell C, Kawakita S, Sharma S, Nguyen HT, Najafabadi AH, Ermis M, Falcone N, Chen J, Mandal K, Khorsandi D, Yilgor C, Choroomi A, Torres E, Mecwan M, John JV, Akbari M, Wang Z, Moniz-Garcia D, Quiñones-Hinojosa A, Jucaud V, Dokmeci MR, Khademhosseini A. Recent Developments in Glioblastoma-On-A-Chip for Advanced Drug Screening Applications. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2405511. [PMID: 39535474 PMCID: PMC11719323 DOI: 10.1002/smll.202405511] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/03/2024] [Revised: 09/08/2024] [Indexed: 11/16/2024]
Abstract
Glioblastoma (GBM) is an aggressive form of cancer, comprising ≈80% of malignant brain tumors. However, there are no effective treatments for GBM due to its heterogeneity and the presence of the blood-brain barrier (BBB), which restricts the delivery of therapeutics to the brain. Despite in vitro models contributing to the understanding of GBM, conventional 2D models oversimplify the complex tumor microenvironment. Organ-on-a-chip (OoC) models have emerged as promising platforms that recapitulate human tissue physiology, enabling disease modeling, drug screening, and personalized medicine. There is a sudden increase in GBM-on-a-chip models that can significantly advance the knowledge of GBM etiology and revolutionize drug development by reducing animal testing and enhancing translation to the clinic. In this review, an overview of GBM-on-a-chip models and their applications is reported for drug screening and discussed current challenges and potential future directions for GBM-on-a-chip models.
Collapse
Affiliation(s)
- Surjendu Maity
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
- Department of Orthopedic Surgery, Duke University School of
Medicine, Duke University, Durham, NC 27705
| | - Tamanna Bhuyan
- Department of Applied Biology, School of Biological
Sciences, University of Science & Technology Meghalaya, Meghalaya, 793101,
India
| | - Christopher Jewell
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Satoru Kawakita
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Saurabh Sharma
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Huu Tuan Nguyen
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | | | - Menekse Ermis
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
- Center of Excellence in Biomaterials and Tissue
Engineering, Middle East Technical University, Ankara, Turkey
| | - Natashya Falcone
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Junjie Chen
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Kalpana Mandal
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Danial Khorsandi
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Can Yilgor
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Auveen Choroomi
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Emily Torres
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Marvin Mecwan
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Johnson V. John
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | - Mohsen Akbari
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
- Laboratoryfor Innovations in Micro Engineering (LiME),
Department of Mechanical Engineering, University of Victoria, Victoria, BC V8P 5C2,
Canada
- Biotechnology Center, Silesian University of Technology,
Akademicka 2A, 44-100 Gliwice, Poland
| | - Zhaohui Wang
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | | | | | - Vadim Jucaud
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| | | | - Ali Khademhosseini
- Terasaki Institute for Biomedical Innovation, Los Angeles,
CA, 90064 USA
| |
Collapse
|
13
|
Hong LQ, Ho TNT, Cu ST, Ngan LT, Tran NQ, Dang TT. Effective Strategies in Designing Chitosan-hyaluronic Acid Nanocarriers: From Synthesis to Drug Delivery Towards Chemotherapy. Curr Drug Deliv 2025; 22:41-62. [PMID: 38310441 DOI: 10.2174/0115672018275983231207101222] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 10/05/2023] [Accepted: 10/26/2023] [Indexed: 02/05/2024]
Abstract
The biomedical field faces an ongoing challenge in developing more effective anti-cancer medication due to the significant burden that cancer poses on human health. Extensive research has been conducted on the utilization of natural polysaccharides in nanomedicine owing to their properties of biocompatibility, biodegradability, non-immunogenicity, and non-toxicity. These characteristics make them a potent drug delivery system for cancer therapy. The chitosan hyaluronic acid nanoparticle (CSHANp) system, consisting of chitosan and hyaluronic acid nanoparticles, has exhibited considerable potential as a nanocarrier for various cancer drugs, rendering it one of the most auspicious systems presently accessible. The CSHANps demonstrate remarkable drug loading capacity, precise control over drug release, and exceptional selectivity towards cancer cells. These properties enhance the therapeutic effectiveness against cancerous cells. This article aims to provide a comprehensive analysis of CSHANp, focusing on its characteristics, production techniques, applications, and future prospects.
Collapse
Affiliation(s)
- Long-Quy Hong
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, 1B TL29, District 12, Ho Chi Minh City, Vietnam
| | - Thao N T Ho
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, 1B TL29, District 12, Ho Chi Minh City, Vietnam
| | - Son T Cu
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, 1B TL29, District 12, Ho Chi Minh City, Vietnam
| | - Lien Tuyet Ngan
- Institute of Applied Technology and Sustainable Development, Nguyen Tat Thanh University, Ho Chi Minh City 700000, Vietnam
| | - Ngoc Quyen Tran
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, 1B TL29, District 12, Ho Chi Minh City, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Hanoi 100000, Vietnam
| | - Tien T Dang
- Institute of Applied Materials Science, Vietnam Academy of Science and Technology, 1B TL29, District 12, Ho Chi Minh City, Vietnam
- Graduate University of Science and Technology, Vietnam Academy of Science and Technology, Hanoi 100000, Vietnam
| |
Collapse
|
14
|
Binsi P, Parvathy U, Jeyakumari A, George Thomas N, Zynudheen A. Marine biopolymers in cosmetics. MARINE BIOPOLYMERS 2025:677-752. [DOI: 10.1016/b978-0-443-15606-9.00023-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
15
|
Gordián‐Vélez WJ, Browne KD, Galarraga JH, Chouhan D, Duda JE, España RA, Chen HI, Burdick JA, Cullen DK. Dopaminergic Axon Tracts Within a Hyaluronic Acid Hydrogel Encasement to Restore the Nigrostriatal Pathway. Adv Healthc Mater 2025; 14:e2402997. [PMID: 39494983 PMCID: PMC11730070 DOI: 10.1002/adhm.202402997] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Indexed: 11/05/2024]
Abstract
Parkinson's disease is characterized by motor deficits emerging from insufficient dopamine in the striatum after degeneration of dopaminergic neurons and their long-projecting axons comprising the nigrostriatal pathway. To address this, a tissue-engineered nigrostriatal pathway (TE-NSP) featuring a tubular hydrogel with a collagen/laminin core that encases aggregated dopaminergic neurons and their axonal tracts is developed. This engineered microtissue can be implanted to replace neurons and axons with fidelity to the lost pathway and thus may provide dopamine according to feedback from host circuitry. While TE-NSPs have traditionally been fabricated with agarose, here a hyaluronic acid (HA) hydrogel is utilized to have a more bioactive encasement while expanding control over physical and biochemical properties. Using rat ventral midbrain neurons, it is found that TE-NSPs exhibited improved neurite growth with HA relative to agarose, with no differences in electrically-evoked dopamine release. When transplanted, HA hydrogels reduced average host neuron loss and inflammation around the implant compared to agarose, and TE-NSP neurons and axonal tracts survived for at least 2 weeks to structurally emulate the lost pathway. This study represents an innovative use of HA hydrogels for neuroregenerative medicine and enables future studies expanding the control and functionality of TE-NSPs.
Collapse
Affiliation(s)
- Wisberty J. Gordián‐Vélez
- Department of BioengineeringSchool of Engineering and Applied ScienceUniversity of PennsylvaniaPhiladelphiaPA19104USA
- Center for Brain Injury & RepairDepartment of NeurosurgeryPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
- Center for NeurotraumaNeurodegeneration & RestorationCorporal Michael Crescenz Veterans Affairs Medical CenterPhiladelphiaPA19104USA
| | - Kevin D. Browne
- Center for Brain Injury & RepairDepartment of NeurosurgeryPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
- Center for NeurotraumaNeurodegeneration & RestorationCorporal Michael Crescenz Veterans Affairs Medical CenterPhiladelphiaPA19104USA
| | - Jonathan H. Galarraga
- Department of BioengineeringSchool of Engineering and Applied ScienceUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Dimple Chouhan
- Center for Brain Injury & RepairDepartment of NeurosurgeryPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
- Center for NeurotraumaNeurodegeneration & RestorationCorporal Michael Crescenz Veterans Affairs Medical CenterPhiladelphiaPA19104USA
| | - John E. Duda
- Center for NeurotraumaNeurodegeneration & RestorationCorporal Michael Crescenz Veterans Affairs Medical CenterPhiladelphiaPA19104USA
- Department of NeurologyPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
| | - Rodrigo A. España
- Department of Neurobiology & AnatomyCollege of MedicineDrexel UniversityPhiladelphiaPA19104USA
| | - H. Isaac Chen
- Center for Brain Injury & RepairDepartment of NeurosurgeryPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
- Center for NeurotraumaNeurodegeneration & RestorationCorporal Michael Crescenz Veterans Affairs Medical CenterPhiladelphiaPA19104USA
| | - Jason A. Burdick
- Department of BioengineeringSchool of Engineering and Applied ScienceUniversity of PennsylvaniaPhiladelphiaPA19104USA
- BioFrontiers Institute and Department of Chemical and Biological EngineeringUniversity of Colorado BoulderBoulderCO80303USA
| | - D. Kacy Cullen
- Department of BioengineeringSchool of Engineering and Applied ScienceUniversity of PennsylvaniaPhiladelphiaPA19104USA
- Center for Brain Injury & RepairDepartment of NeurosurgeryPerelman School of MedicineUniversity of PennsylvaniaPhiladelphiaPA19104USA
- Center for NeurotraumaNeurodegeneration & RestorationCorporal Michael Crescenz Veterans Affairs Medical CenterPhiladelphiaPA19104USA
| |
Collapse
|
16
|
Steudter T, Lam T, Pirmahboub H, Stoppel C, Kloke L, Pearson S, Del Campo A. A Comparative Study between Thiol-Ene and Acrylate Photocrosslinkable Hyaluronic Acid Hydrogel Inks for Digital Light Processing. Macromol Biosci 2024:e2400535. [PMID: 39741116 DOI: 10.1002/mabi.202400535] [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: 11/01/2024] [Revised: 12/13/2024] [Indexed: 01/02/2025]
Abstract
Photocrosslinkable formulations based on the radical thiol-ene reaction are considered better alternatives than methacrylated counterparts for light-based fabrication processes. This study quantifies differences between thiol-ene and methacrylated crosslinked hydrogels in terms of precursors stability, the control of the crosslinking process, and the resolution of printed features particularized for hyaluronic acid (HA) inks at concentrations relevant for bioprinting. First, the synthesis of HA functionalized with norbornene, allyl ether, or methacrylate groups with the same molecular weight and comparable degrees of functionalization is presented. The thiol-ene hydrogel precursors show storage stability over 15 months, 3.8 times higher than the methacrylated derivative. Photorheology experiments demonstrate up to 4.7-times faster photocrosslinking. Network formation in photoinitiated thiol-ene HA crosslinking allows higher temporal control than in methacrylated HA, which shows long post-illumination hardening. Using digital light processing, 4% w/v HA hydrogels crosslinked with a dithiol allowed printing of 13.5 × 4 × 1 mm3 layers with holes of 100 µm resolution within 2 s. This is the smallest feature size demonstrated in DLP printing with HA-based thiol-ene hydrogels. The results are important to estimate the extent to which the synthetic effort of introducing -ene functions can pay off in the printing step.
Collapse
Affiliation(s)
- Therese Steudter
- INM - Leibniz Institute for New Materials, 66123, Saarbrücken, Germany
- Chemistry Department, Saarland University, 66123, Saarbrücken, Germany
| | | | | | | | | | - Samuel Pearson
- INM - Leibniz Institute for New Materials, 66123, Saarbrücken, Germany
| | - Aránzazu Del Campo
- INM - Leibniz Institute for New Materials, 66123, Saarbrücken, Germany
- Chemistry Department, Saarland University, 66123, Saarbrücken, Germany
| |
Collapse
|
17
|
Xian J, Xiao F, Zou J, Luo W, Han S, Liu Z, Chen Y, Zhu Q, Li M, Yu C, Saiding Q, Tao W, Kong N, Xie T. Elemene Hydrogel Modulates the Tumor Immune Microenvironment for Enhanced Treatment of Postoperative Cancer Recurrence and Metastases. J Am Chem Soc 2024; 146:35252-35263. [PMID: 39625467 DOI: 10.1021/jacs.4c12531] [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: 12/26/2024]
Abstract
As a representative active ingredient of traditional Chinese medicine (TCM) and a clinically approved anticancer drug, elemene (ELE) exhibits exciting potential in the antitumor field; however, appropriate drug formulations still need to be explored for specific diseases such as postoperative cancer recurrence and metastasis. Herein, we report an ELE hydrogel with controlled drug release kinetics that can allow ELE to maintain effective concentrations at local lesion sites for extended periods to enhance the bioavailability of ELE. Concretely, dopamine-conjugated hyaluronic acid is synthesized and utilized to prepare ELE nanodrug-embedded hydrogels. In a model of postoperative breast cancer recurrence and metastasis, the ELE hydrogel demonstrates a 96% inhibition rate of recurrence; in contrast, the free ELE nanodrug shows only a 65.5% inhibition rate of recurrence. Importantly, the ELE hydrogel markedly stimulates a potent antitumor immune response in the microenvironment of cancer lesions, increasing antitumor immune cells such as CD8+ T cells, CD4+ T cells, and M1-type macrophages, as well as elevating antitumor cytokines including TNF-α, IFN-γ, and IL-6. Overall, this study not only advances the field of TCM but also highlights the transformative impact of controlled-release hydrogels in improving antitumor therapy.
Collapse
Affiliation(s)
- Jing Xian
- School of Pharmacy; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang 311121, China
| | - Fan Xiao
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang 311121, China
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Jianhua Zou
- School of Pharmacy; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Wei Luo
- School of Pharmacy; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Shiqi Han
- School of Pharmacy; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Ziwei Liu
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang 311121, China
| | - Yiquan Chen
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang 311121, China
| | - Qianru Zhu
- School of Pharmacy; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Meng Li
- School of Pharmacy; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Chuao Yu
- School of Pharmacy; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
| | - Qimanguli Saiding
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Na Kong
- Liangzhu Laboratory, Zhejiang University, Hangzhou, Zhejiang 311121, China
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, United States
| | - Tian Xie
- School of Pharmacy; Key Laboratory of Elemene Class Anti-Cancer Chinese Medicines; Engineering Laboratory of Development and Application of Traditional Chinese Medicines; Collaborative Innovation Center of Traditional Chinese Medicines of Zhejiang Province, Hangzhou Normal University, Hangzhou, Zhejiang 311121, China
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, Sichuan 611137, China
| |
Collapse
|
18
|
Rudzinska-Radecka M, Turos-Korgul L, Mukherjee D, Podszywalow-Bartnicka P, Piwocka K, Guzowski J. High-throughput formulation of reproducible 3D cancer microenvironments for drug testing in myeloid leukemia. Biofabrication 2024; 17:015035. [PMID: 39622161 DOI: 10.1088/1758-5090/ad998d] [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: 06/28/2024] [Accepted: 12/02/2024] [Indexed: 12/17/2024]
Abstract
Leukemic microenvironment has been recognized as a factor that strongly supports the mechanisms of resistance. Therefore, targeting the microenvironment is currently one of the major directions in drug development and preclinical studies in leukemia. Despite the variety of available leukemia 3D culture models, the reproducible generation of miniaturized leukemic microenvironments, suitable for high-throughput drug testing, has remained a challenge. Here, we use droplet microfluidics to generate tens of thousands of highly monodisperse leukemic-bone marrow microenvironments within minutes. We employ gelatin methacryloyl (GelMA) as a model extracellular matrix (ECM) and tune the concentration of the biopolymer, check the impact of other components of the ECM (hyaluronic acid), cell concentration and the ratio of leukemic cells to bone marrow cells within the microbeads to establish the optimal conditions for microtissue formation. We administer model kinase inhibitor, imatinib, at various concentrations to the encapsulated leukemic microtissues, and, via comparing mono- and co-culture conditions (cancer alone vs cancer-stroma), we find that the stroma-leukemia crosstalk systematically protects the encapsulated cells against the drug-induced cytotoxicity. With that we demonstrate that our system mimics the physiological stroma-dependent protection. We discuss applicability of our model to (i) studying the role of direct- or close-contact interactions between the leukemia and bone marrow cells embedded in microscale 3D ECM on the stroma-mediated protection, and (ii) high-throughput screening of anti-cancer therapeutics in personalized leukemia therapies.
Collapse
Affiliation(s)
- M Rudzinska-Radecka
- Institute of Physical Chemistry, Polish Academy of Sciences, ul. Kasprzaka 44/52, 01-224 Warsaw, Poland
- Equal contribution.v
| | - L Turos-Korgul
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Str., Warsaw 02-093, Poland
- Equal contribution.v
| | - D Mukherjee
- Institute of Physical Chemistry, Polish Academy of Sciences, ul. Kasprzaka 44/52, 01-224 Warsaw, Poland
| | - P Podszywalow-Bartnicka
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Str., Warsaw 02-093, Poland
| | - K Piwocka
- Laboratory of Cytometry, Nencki Institute of Experimental Biology, Polish Academy of Sciences, 3 Pasteur Str., Warsaw 02-093, Poland
| | - J Guzowski
- Institute of Physical Chemistry, Polish Academy of Sciences, ul. Kasprzaka 44/52, 01-224 Warsaw, Poland
| |
Collapse
|
19
|
Kwon M, Lee Y, Kim KS. Biomimetic gradient hydrogel with fibroblast spheroids for full-thickness skin regeneration. BIOMATERIALS ADVANCES 2024; 169:214152. [PMID: 39708659 DOI: 10.1016/j.bioadv.2024.214152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Revised: 12/12/2024] [Accepted: 12/14/2024] [Indexed: 12/23/2024]
Abstract
Hydrogel-based scaffolds have been widely investigated for their use in tissue engineering to accelerate tissue regeneration. However, replicating the physiological microenvironments of tissues with appropriate biological cues remains challenging. Recent advances in gradient hydrogels have transformed tissue-engineering research by providing precise structures that mimic the extracellular matrix of natural tissues. Unlike conventional homogeneously structured hydrogels, gradient hydrogels provide a better bio-mimicking microenvironment for combined cell therapies in chronic wound treatment by regulating various cell behaviors, such as proliferation, migration, and differentiation. Here, we present the integration of L929 mouse fibroblast spheroids into gradient hydrogels to mimic the dermal stiffness microenvironment and we investigated their impact on full-thickness skin regeneration. A stiffness gradient was achieved by modulating the concentration of methacrylated hyaluronic acid (HA-MA) with varying degrees of methacrylation, using a dual-syringe pump system. The encapsulation of L929 spheroids with gradient hydrogel facilitated skin cell organization in a hierarchically ordered configuration, leading to full-thickness wound healing that was 1.53 times faster than the untreated group in a rat model. This study provides a method for investigating the potential role of gradient hydrogels in various tissue engineering and regeneration applications.
Collapse
Affiliation(s)
- Mina Kwon
- School of Chemical Engineering, Pusan National University, Busan 46241, Republic of Korea.
| | - Yuhan Lee
- Department of Anaesthesiology, Perioperative and Pain Medicine, Center for Accelerated Medical Innovation, Center for Nanomedicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, United States of America.
| | - Ki Su Kim
- School of Chemical Engineering, Pusan National University, Busan 46241, Republic of Korea; Department of Organic Materials Science and Engineering, Pusan National University, Busan 46241, Republic of Korea; Institute of Advanced Organic Materials, Pusan National University, Busan 46241, Republic of Korea.
| |
Collapse
|
20
|
Kim M, Jung MY, Lee DY, Ahn SM, Lee GM, Park CY. How to Fabricate Hyaluronic Acid for Ocular Drug Delivery. Pharmaceutics 2024; 16:1604. [PMID: 39771582 PMCID: PMC11680071 DOI: 10.3390/pharmaceutics16121604] [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: 11/26/2024] [Revised: 12/13/2024] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
This review aims to examine existing research on the development of ocular drug delivery devices utilizing hyaluronic acid (HA). Renowned for its exceptional biocompatibility, viscoelastic properties, and ability to enhance drug bioavailability, HA is a naturally occurring biopolymer. The review discussed specific mechanisms by which HA enhances drug delivery, including prolonging drug residence time on ocular surfaces, facilitating controlled drug release, and improving drug penetration through ocular tissues. By focusing on these unique functionalities, this review highlights the potential of HA-based systems to revolutionize ocular treatment. Various fabrication techniques for HA-based ocular drug delivery systems, including hydrogels, nanoparticles, and microneedles, are discussed, highlighting their respective advantages and limitations. Additionally, this review explores the clinical applications of HA-based devices in treating a range of ocular diseases, such as dry eye syndrome, glaucoma, retinal disorders, and ocular infections. By comparing the efficacy and safety profiles of these devices with traditional ocular drug delivery methods, this review aims to provide a comprehensive understanding of the potential benefits and challenges associated with HA-based systems. Moreover, this review discusses current limitations and future directions in the field, such as the need for standardized fabrication protocols, long-term biocompatibility studies, and large-scale clinical trials. The insights and advancements presented in this review aim to guide future research and development efforts, ultimately enhancing the effectiveness of ocular drug delivery and improving patient outcomes.
Collapse
Affiliation(s)
- Martha Kim
- Department of Ophthalmology, Dongguk University Ilsan Hospital, Goyang 10326, Republic of Korea; (M.K.); (M.-Y.J.); (D.-Y.L.); (S.M.A.); (G.M.L.)
| | - Mi-Young Jung
- Department of Ophthalmology, Dongguk University Ilsan Hospital, Goyang 10326, Republic of Korea; (M.K.); (M.-Y.J.); (D.-Y.L.); (S.M.A.); (G.M.L.)
| | - Do-Yeon Lee
- Department of Ophthalmology, Dongguk University Ilsan Hospital, Goyang 10326, Republic of Korea; (M.K.); (M.-Y.J.); (D.-Y.L.); (S.M.A.); (G.M.L.)
| | - So Min Ahn
- Department of Ophthalmology, Dongguk University Ilsan Hospital, Goyang 10326, Republic of Korea; (M.K.); (M.-Y.J.); (D.-Y.L.); (S.M.A.); (G.M.L.)
| | - Gyeong Min Lee
- Department of Ophthalmology, Dongguk University Ilsan Hospital, Goyang 10326, Republic of Korea; (M.K.); (M.-Y.J.); (D.-Y.L.); (S.M.A.); (G.M.L.)
| | - Choul Yong Park
- Department of Ophthalmology, Samsung Medical Center, Sungkyunkwan University School of Medicine, 81, Irwon-ro, Gangnam-gu, Seoul 06351, Republic of Korea
| |
Collapse
|
21
|
Prakash G, Clasky AJ, Gadani K, Nazeri M, Gu FX. Ion-Mediated Cross-Linking of Hyaluronic Acid into Hydrogels without Chemical Modification. Biomacromolecules 2024; 25:7723-7735. [PMID: 39485907 DOI: 10.1021/acs.biomac.4c00985] [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: 11/03/2024]
Abstract
Hyaluronic acid (HA) is a biomedically relevant polymer widely explored as a component of hydrogels. The prevailing approaches for cross-linking HA into hydrogels require chemically modifying the polymer, which can increase processing steps and complicate biocompatibility. Herein, we demonstrate an alternative approach to cross-link HA that eliminates the need for chemical modifications by leveraging the interactions between metal cations and the negatively charged, ionizable functional groups on HA. We demonstrate that HA can be cross-linked with the bivalent metal cations Mn(II), Fe(II), Co(II), Ni(II), Cu(II), Zn(II), Pd(II), and notably Mg(II). Using Mg(II) as a model, we show that ion-HA hydrogel rheological properties can be tuned by altering the HA molecular weight and concentrations of ions, NaOH, and HA. Mg(II)-HA hydrogels showed the potential for self-healing and stimulus response. Our findings lay the groundwork for developing a new class of HA-based hydrogels for use in biomedical applications and beyond.
Collapse
Affiliation(s)
- Gayatri Prakash
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
| | - Aaron J Clasky
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
| | - Kunal Gadani
- Department of Chemistry, University of Waterloo, 200 University Avenue West, Waterloo, Ontario N2L 3G1, Canada
| | - Mohammad Nazeri
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
| | - Frank X Gu
- Department of Chemical Engineering & Applied Chemistry, University of Toronto, 200 College Street, Toronto, Ontario M5S 3E5, Canada
- Institute of Biomedical Engineering, University of Toronto, 164 College Street, Toronto, Ontario M5S 3G9, Canada
| |
Collapse
|
22
|
Jiang F, Fu M, Gan Y, Bu C, Guo Z, Lv X, Ding X. A multifunctional hydrogel dressing loaded with antibiotics for healing of infected wound. Int J Pharm 2024; 666:124770. [PMID: 39349226 DOI: 10.1016/j.ijpharm.2024.124770] [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: 09/15/2024] [Accepted: 09/25/2024] [Indexed: 10/02/2024]
Abstract
Wound bacterial infections can significantly delay the healing process and even lead to fetal sepsis. There is a need for multifunctional dressings that possess antibacterial property, tissue adhesive property, self-healing capability, and biocompatibility to effectively treat bacteria-infected wound. In this study, we report a dual dynamically crosslinked hydrogel, OHA-PBA/PVA/Gen, which incorporates the antibiotic gentamicin (Gen) as a dynamic crosslinker. The hydrogel is formed through the formation of Schiff base bonds between phenylboronic acid-grafted oxidized hyaluronic acid (OHA-PBA) and Gen, as well as boronic acid ester bonds between OHA-PBA and polyvinyl alcohol (PVA). This unique composition imparts tissue adhesiveness, injectability and self-healing property to the hydrogel. The hydrogel also exhibits pH-responsive antibiotic release behavior due to the acid-responsive dissociation of Schiff base bonds. As a result, it demonstrates strong antibacterial activity against both Gram-positive bacteria S. aureus and Gram-negative bacteria E. coli through contact killing and diffusion killing mechanisms. Importantly, the OHA-PBA/PVA/Gen hydrogel avoids incorporation of toxic small molecular crosslinking agents, and all the components of the hydrogel are biocompatible, ensuring its biosafety. In a S. aureus-infected wound mouse model, this hydrogel effectively eradicated bacteria and promoted angiogenesis, leading to significantly accelerated wound healing. These results highlight the potential of the dual dynamically crosslinking hydrogel OHA-PBA/PVA/Gen as a multifunctional wound dressing for the treatment of bacteria-infected wound.
Collapse
Affiliation(s)
- Fenglin Jiang
- School of Medicine, Sun Yat-sen University, Shenzhen 518107, China; Department of Pharmacy, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang 621000, China
| | - Mengjing Fu
- School of Pharmaceutical Science (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, PR China
| | - Yingying Gan
- School of Pharmaceutical Science (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, PR China
| | - Changxin Bu
- School of Pharmaceutical Science (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, PR China
| | - Zhihao Guo
- School of Pharmaceutical Science (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, PR China
| | - Xue Lv
- School of Pharmaceutical Science (Shenzhen), Sun Yat-sen University, Shenzhen, 518107, PR China
| | - Xin Ding
- School of Medicine, Sun Yat-sen University, Shenzhen 518107, China; State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou 510006, China.
| |
Collapse
|
23
|
Chen S, Xia J, Hou Z, Wu P, Yang Y, Cui L, Xiang Z, Sun S, Yang L. Natural polysaccharides combined with mussel-inspired adhesion for multifunctional hydrogels in wound hemostasis and healing: A review. Int J Biol Macromol 2024; 282:136965. [PMID: 39476886 DOI: 10.1016/j.ijbiomac.2024.136965] [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: 07/12/2024] [Revised: 10/09/2024] [Accepted: 10/25/2024] [Indexed: 11/03/2024]
Abstract
As naturally derived macromolecular polymers, polysaccharides have garnered significant attention in recent years as promising candidates for fabricating multifunctional hydrogels, particularly for wound healing applications, owing to their inherent biocompatibility, biodegradability, and structural diversity. However, the inherently weak skin adhesion of natural polysaccharide hydrogels has motivated the exploration of mussel-inspired catechol-based adhesion strategies to overcome this limitation. Incorporating mussel-inspired modifications into natural polysaccharides can imbue them with unique properties such as enhanced adhesion, antioxidant activity, antibacterial properties, and chelation capabilities, considerably broadening their potential for wound hemostasis and healing applications. This review comprehensively overviews recent advances in mussel-inspired polysaccharide hydrogels, focusing on the combination of natural polysaccharides, including chitosan, alginate, hyaluronic acid, cellulose, and dextran, with mussel-inspired catechol. We delve into their fabrication strategies and highlight their promising biomedical applications, with a particular emphasis on wound hemostasis and diverse wound healing processes. Mussel-inspired modification strategies for polysaccharide hydrogels are expected to remain a focal point within the fields of wound hemostasis and healing, paving the way for more impactful research endeavors.
Collapse
Affiliation(s)
- Siwen Chen
- Research Center for Biomedical Materials, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, PR China; Center for Molecular Science and Engineering, College of Science, Northeastern University, Shenyang 110819, PR China
| | - Jiangli Xia
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, PR China
| | - Zhipeng Hou
- Research Center for Biomedical Materials, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, PR China
| | - Peng Wu
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, PR China
| | - Yuanyuan Yang
- Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, PR China
| | - Longwei Cui
- Department of Plastic Surgery, The First Hospital of China Medical University, Shenyang, Liaoning 110002, PR China
| | - Zheng Xiang
- School of Pharmaceutical Science, Liaoning University, Shenyang 110036, PR China.
| | - Siyu Sun
- Research Center for Biomedical Materials, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, PR China; Department of Gastroenterology, Endoscopic Center, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, PR China.
| | - Liqun Yang
- Research Center for Biomedical Materials, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 110004, PR China.
| |
Collapse
|
24
|
Liu Y, Huang T, Yap NA, Lim K, Ju LA. Harnessing the power of bioprinting for the development of next-generation models of thrombosis. Bioact Mater 2024; 42:328-344. [PMID: 39295733 PMCID: PMC11408160 DOI: 10.1016/j.bioactmat.2024.08.040] [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: 05/19/2024] [Revised: 08/07/2024] [Accepted: 08/29/2024] [Indexed: 09/21/2024] Open
Abstract
Thrombosis, a leading cause of cardiovascular morbidity and mortality, involves the formation of blood clots within blood vessels. Current animal models and in vitro systems have limitations in recapitulating the complex human vasculature and hemodynamic conditions, limiting the research in understanding the mechanisms of thrombosis. Bioprinting has emerged as a promising approach to construct biomimetic vascular models that closely mimic the structural and mechanical properties of native blood vessels. This review discusses the key considerations for designing bioprinted vascular conduits for thrombosis studies, including the incorporation of key structural, biochemical and mechanical features, the selection of appropriate biomaterials and cell sources, and the challenges and future directions in the field. The advancements in bioprinting techniques, such as multi-material bioprinting and microfluidic integration, have enabled the development of physiologically relevant models of thrombosis. The future of bioprinted models of thrombosis lies in the integration of patient-specific data, real-time monitoring technologies, and advanced microfluidic platforms, paving the way for personalized medicine and targeted interventions. As the field of bioprinting continues to evolve, these advanced vascular models are expected to play an increasingly important role in unraveling the complexities of thrombosis and improving patient outcomes. The continued advancements in bioprinting technologies and the collaboration between researchers from various disciplines hold great promise for revolutionizing the field of thrombosis research.
Collapse
Affiliation(s)
- Yanyan Liu
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Tao Huang
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
| | - Nicole Alexis Yap
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
| | - Khoon Lim
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
- School of Medical Sciences, The University of Sydney, Darlington, NSW 2008, Australia
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, 2006, Australia
| | - Lining Arnold Ju
- School of Biomedical Engineering, The University of Sydney, Darlington, NSW, 2008, Australia
- Charles Perkins Centre, The University of Sydney, Camperdown, NSW 2006, Australia
- The University of Sydney Nano Institute (Sydney Nano), The University of Sydney, Camperdown, NSW, 2006, Australia
- Heart Research Institute, Camperdown, Newtown, NSW 2042, Australia
| |
Collapse
|
25
|
Carvalho EM, Ding EA, Saha A, Garcia DC, Weldy A, Zushin PJH, Stahl A, Aghi MK, Kumar S. Viscoelastic High-Molecular-Weight Hyaluronic Acid Hydrogels Support Rapid Glioblastoma Cell Invasion with Leader-Follower Dynamics. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2404885. [PMID: 39508297 PMCID: PMC11637900 DOI: 10.1002/adma.202404885] [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: 04/04/2024] [Revised: 09/25/2024] [Indexed: 11/15/2024]
Abstract
Hyaluronic acid (HA), the primary component of brain extracellular matrix, is increasingly used to model neuropathological processes, including glioblastoma (GBM) tumor invasion. While elastic hydrogels based on crosslinked low-molecular-weight (LMW) HA are widely exploited for this purpose and have proven valuable for discovery and screening, brain tissue is both viscoelastic and rich in high-MW (HMW) HA, and it remains unclear how these differences influence invasion. To address this question, hydrogels comprised of either HMW (1.5 MDa) or LMW (60 kDa) HA are introduced, characterized, and applied in GBM invasion studies. Unlike LMW HA hydrogels, HMW HA hydrogels relax stresses quickly, to a similar extent as brain tissue, and to a greater extent than many conventional HA-based scaffolds. GBM cells implanted within HMW HA hydrogels invade much more rapidly than in their LMW HA counterparts and exhibit distinct leader-follower dynamics. Leader cells adopt dendritic morphologies similar to invasive GBM cells observed in vivo. Transcriptomic, pharmacologic, and imaging studies suggest that leader cells exploit hyaluronidase, an enzyme strongly enriched in human GBMs, to prime a path for followers. This study offers new insight into how HA viscoelastic properties drive invasion and argues for the use of highly stress-relaxing materials to model GBM.
Collapse
Affiliation(s)
- Emily M Carvalho
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, 94720, USA
| | - Erika A Ding
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, 94720, USA
| | - Atul Saha
- Department of Neurosurgery, University of California, San Francisco, CA, 94158, USA
| | - Diana Cruz Garcia
- Department of Bioengineering, University of California, Berkeley, CA, 94720, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, 94158, USA
| | - Anna Weldy
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, 94720, USA
| | - Peter-James H Zushin
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, 94720, USA
| | - Andreas Stahl
- Department of Nutritional Sciences and Toxicology, University of California, Berkeley, 94720, USA
| | - Manish K Aghi
- Department of Neurosurgery, University of California, San Francisco, CA, 94158, USA
| | - Sanjay Kumar
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, 94720, USA
- Department of Bioengineering, University of California, Berkeley, CA, 94720, USA
- Department of Bioengineering and Therapeutic Sciences, University of California, San Francisco, CA, 94158, USA
| |
Collapse
|
26
|
Walker M, Morton JP. Hydrogel models of pancreatic adenocarcinoma to study cell mechanosensing. Biophys Rev 2024; 16:851-870. [PMID: 39830124 PMCID: PMC11735828 DOI: 10.1007/s12551-024-01265-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Accepted: 12/06/2024] [Indexed: 01/22/2025] Open
Abstract
Pancreatic adenocarcinoma (PDAC) is the predominant form of pancreatic cancer and one of the leading causes of cancer-related death worldwide, with an extremely poor prognosis after diagnosis. High mortality from PDAC arises partly due to late diagnosis resulting from a lack of early-stage biomarkers and due to chemotherapeutic drug resistance, which arises from a highly fibrotic stromal response known as desmoplasia. Desmoplasia alters tissue mechanics, which triggers changes in cell mechanosensing and leads to dysregulated transcriptional activity and disease phenotypes. Hydrogels are effective in vitro models to mimic mechanical changes in tissue mechanics during PDAC progression and to study the influence of these changes on mechanosensitive cell responses. Despite the complex biophysical changes that occur within the PDAC microenvironment, carefully designed hydrogels can very closely recapitulate these properties during PDAC progression. Hydrogels are relatively inexpensive, highly reproducible and can be designed in a humanised manner to increase their relevance for human PDAC studies. In vivo models have some limitations, including species-species differences, high variability, expense and legal/ethical considerations, which make hydrogel models a promising alternative. Here, we comprehensively review recent advancements in hydrogel bioengineering for developing our fundamental understanding of mechanobiology in PDAC, which is critical for informing advanced therapeutics.
Collapse
Affiliation(s)
- M Walker
- Centre for the Cellular Microenvironment, Advanced Research Centre, 11 Chapel Lane, James Watt School of Engineering, University of Glasgow, Glasgow, G11 6EW UK
| | - JP Morton
- Cancer Research UK Scotland Institute, Garscube Estate, Switchback Rd, Glasgow, G61 1BD UK
- School of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Rd, Glasgow, G61 1QH UK
| |
Collapse
|
27
|
Choi S, Cho JC, Lee S, Lee SJ. Development of Dispersion Process to Improve Quality of Hyaluronic Acid Filler Crosslinked with 1,4-Butanediol Diglycidyl Ether. Polymers (Basel) 2024; 16:3323. [PMID: 39684068 DOI: 10.3390/polym16233323] [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: 10/25/2024] [Revised: 11/21/2024] [Accepted: 11/22/2024] [Indexed: 12/18/2024] Open
Abstract
This study proposes a new and simple process that improves the quality of a hyaluronic acid (HA) filler crosslinked with 1,4-butanediol diglycidyl ether (BDDE) using solution dispersion at a low temperature. This process involves the solvent being dispersed among the solute naturally after the mixing process. The process used in this study involved two reactions. First, the solution was dispersed among HA molecules (Mw = ~0.7 MDa) creating a well-homogenized mixture. Second, the decomposition and synthesis of HA occurred naturally in an aqueous alkaline solution (>pH 11), the weight average molar mass (Mw) was adjusted (Mw = ~143,000), and the crosslinking surface area was expanded, allowing for a high degree of crosslinking. Therefore, the viscoelasticity and cohesion of the filler increased with the new method compared to the previous process both at the lab scale (previous process:new process, viscosity (cP) = 24M:43M, storage modulus (Pa) = 306:538, loss modulus (Pa) = 33:61, and tack (N) = 0.24:0.43) and at the factory scale (previous process:new process, complex viscosity (cP) = 19M:26M, storage modulus (Pa) = 229:314, loss modulus (Pa) = 71:107, and tack (N) = 0.35:0.43).
Collapse
Affiliation(s)
- Sunglim Choi
- CHA Meditech Co., Ltd., 119 Techno 2-ro (#206, Migeun Techno World, Yongsan-Dong), Yuseong-gu, Daejeon 34116, Republic of Korea
| | - Jin Cheol Cho
- CHA Meditech Co., Ltd., 119 Techno 2-ro (#206, Migeun Techno World, Yongsan-Dong), Yuseong-gu, Daejeon 34116, Republic of Korea
| | - Seunghwa Lee
- CHA Meditech Co., Ltd., 119 Techno 2-ro (#206, Migeun Techno World, Yongsan-Dong), Yuseong-gu, Daejeon 34116, Republic of Korea
| | - Seong Jin Lee
- CHA Meditech Co., Ltd., 119 Techno 2-ro (#206, Migeun Techno World, Yongsan-Dong), Yuseong-gu, Daejeon 34116, Republic of Korea
| |
Collapse
|
28
|
Goto R, Terasawa M, Kojima M, Matsuda K, Nishiura K, Sakai S. Synthesis and application of phenol-grafted rhamnan sulfate for 3D bioprinting. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2024:1-17. [PMID: 39588981 DOI: 10.1080/09205063.2024.2427499] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Accepted: 10/29/2024] [Indexed: 11/27/2024]
Abstract
Rhamnan sulfate (RS) is a sulfated polysaccharide extracted from the cell wall of the green alga Monostroma nitidum. Owing to its negative charge, RS interacts with a variety of proteins, enabling various biological activities, such as antiviral, anticoagulant, and antitumor effects. However, RS does not form a stable hydrogel under physiological conditions, which is required for its beneficial biological activities in tissue engineering. To address this limitation, we developed phenol-grafted rhamnan sulfate (RS-Ph), which allows hydrogelation via horseradish peroxidase (HRP)-mediated cross-linking reactions and can be used for 3D bioprinting. Specifically, we synthesized RS-Ph with three different -Ph content: RS-LPh (16.4 mmol/g), RS-MPh (21.3 mmol/g), and RS-HPh (31.7 mmol/g). Surface plasmon resonance measurements revealed that RS-Ph exhibited a maximum binding capacity of more than 8.3 times higher than that of sodium alginate as a negative control. Additionally, a 10% w/v RS-HPh solution formed a hydrogel within 8.2 ± 0.7 s in the presence of 10 U/mL HRP. Furthermore, high-fidelity 3D bioprinting was achieved using an RS-Ph/cellulose nanofiber composite bioink. Our results demonstrate the potential use of bioactive RS-Ph hydrogels in a wide range of tissue engineering fields, including not only bioprinting but also drug delivery systems and wound dressings.
Collapse
Affiliation(s)
- Ryota Goto
- Department of Materials Engineering Science, Graduate School of Engineering Science, Osaka University, Toyonaka, Japan
| | | | - Masaru Kojima
- Department of Materials Engineering Science, Graduate School of Engineering Science, Osaka University, Toyonaka, Japan
| | - Koichi Matsuda
- Konan Chemical Manufacturing Co., Ltd., Yokkaichi, Japan
| | - Kaoru Nishiura
- Konan Chemical Manufacturing Co., Ltd., Yokkaichi, Japan
| | - Shinji Sakai
- Department of Materials Engineering Science, Graduate School of Engineering Science, Osaka University, Toyonaka, Japan
| |
Collapse
|
29
|
Lai Y, Xiao X, Huang Z, Duan H, Yang L, Yang Y, Li C, Feng L. Photocrosslinkable Biomaterials for 3D Bioprinting: Mechanisms, Recent Advances, and Future Prospects. Int J Mol Sci 2024; 25:12567. [PMID: 39684279 DOI: 10.3390/ijms252312567] [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: 10/28/2024] [Revised: 11/15/2024] [Accepted: 11/16/2024] [Indexed: 12/18/2024] Open
Abstract
Constructing scaffolds with the desired structures and functions is one of the main goals of tissue engineering. Three-dimensional (3D) bioprinting is a promising technology that enables the personalized fabrication of devices with regulated biological and mechanical characteristics similar to natural tissues/organs. To date, 3D bioprinting has been widely explored for biomedical applications like tissue engineering, drug delivery, drug screening, and in vitro disease model construction. Among different bioinks, photocrosslinkable bioinks have emerged as a powerful choice for the advanced fabrication of 3D devices, with fast crosslinking speed, high resolution, and great print fidelity. The photocrosslinkable biomaterials used for light-based 3D printing play a pivotal role in the fabrication of functional constructs. Herein, this review outlines the general 3D bioprinting approaches related to photocrosslinkable biomaterials, including extrusion-based printing, inkjet printing, stereolithography printing, and laser-assisted printing. Further, the mechanisms, advantages, and limitations of photopolymerization and photoinitiators are discussed. Next, recent advances in natural and synthetic photocrosslinkable biomaterials used for 3D bioprinting are highlighted. Finally, the challenges and future perspectives of photocrosslinkable bioinks and bioprinting approaches are envisaged.
Collapse
Affiliation(s)
- Yushang Lai
- Division of Vascular Surgery, Department of General Surgery and Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiong Xiao
- Division of Vascular Surgery, Department of General Surgery and Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ziwei Huang
- Division of Vascular Surgery, Department of General Surgery and Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hongying Duan
- Division of Vascular Surgery, Department of General Surgery and Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Liping Yang
- Division of Vascular Surgery, Department of General Surgery and Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Yuchu Yang
- Division of Vascular Surgery, Department of General Surgery and Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Chenxi Li
- Division of Vascular Surgery, Department of General Surgery and Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Li Feng
- Division of Vascular Surgery, Department of General Surgery and Regenerative Medicine Research Center, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
30
|
Kim MH, Singh YP, Celik N, Yeo M, Rizk E, Hayes DJ, Ozbolat IT. High-throughput bioprinting of spheroids for scalable tissue fabrication. Nat Commun 2024; 15:10083. [PMID: 39572584 PMCID: PMC11582690 DOI: 10.1038/s41467-024-54504-7] [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: 07/08/2024] [Accepted: 11/12/2024] [Indexed: 11/24/2024] Open
Abstract
Tissue biofabrication mimicking organ-specific architecture and function requires physiologically-relevant cell densities. Bioprinting using spheroids can achieve this, but is limited due to the lack of practical, scalable techniques. This study presents HITS-Bio (High-throughput Integrated Tissue Fabrication System for Bioprinting), a multiarray bioprinting technique for rapidly positioning multiple spheroids simultaneously using a digitally-controlled nozzle array (DCNA). HITS-Bio achieves an unprecedented speed, ten times faster compared to existing techniques while maintaining high cell viability ( > 90%). The utility of HITS-Bio was exemplified in multiple applications, including intraoperative bioprinting with microRNA transfected human adipose-derived stem cell spheroids for calvarial bone regeneration ( ~ 30 mm3) in a rat model achieving a near-complete defect closure (bone coverage area of ~ 91% in 3 weeks and ~96% in 6 weeks). Additionally, the successful fabrication of scalable cartilage constructs (1 cm3) containing ~600 chondrogenic spheroids highlights its high-throughput efficiency (under 40 min per construct) and potential for repairing volumetric defects.
Collapse
Affiliation(s)
- Myoung Hwan Kim
- Department of Biomedical Engineering, Penn State University, University Park, PA, USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, USA
| | - Yogendra Pratap Singh
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, USA
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, USA
| | - Nazmiye Celik
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, USA
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, USA
| | - Miji Yeo
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, USA
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, USA
| | - Elias Rizk
- Department of Neurosurgery, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA
| | - Daniel J Hayes
- Department of Biomedical Engineering, Penn State University, University Park, PA, USA
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, USA
- Materials Research Institute, Penn State University, University Park, PA, USA
| | - Ibrahim T Ozbolat
- Department of Biomedical Engineering, Penn State University, University Park, PA, USA.
- The Huck Institutes of the Life Sciences, Penn State University, University Park, PA, USA.
- Engineering Science and Mechanics Department, Penn State University, University Park, PA, USA.
- Department of Neurosurgery, Penn State Milton S. Hershey Medical Center, Hershey, PA, USA.
- Materials Research Institute, Penn State University, University Park, PA, USA.
- Department of Medical Oncology, Cukurova University, Adana, Turkey.
| |
Collapse
|
31
|
Saletti M, Pepi S, Paolino M, Venditti J, Giuliani G, Bonechi C, Leone G, Magnani A, Rossi C, Cappelli A. Crosslinking by Click Chemistry of Hyaluronan Graft Copolymers Involving Resorcinol-Based Cinnamate Derivatives Leading to Gel-like Materials. Gels 2024; 10:751. [PMID: 39590107 PMCID: PMC11594237 DOI: 10.3390/gels10110751] [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: 10/07/2024] [Revised: 11/06/2024] [Accepted: 11/14/2024] [Indexed: 11/28/2024] Open
Abstract
The well-known "click chemistry" reaction copper(I)-catalyzed azide-alkyne 1,3-dipolar cycloaddition (CuAAC) was used to transform under very mild conditions hyaluronan-based graft copolymers HA(270)-FA-Pg into the crosslinked derivatives HA(270)-FA-TEGERA-CL and HA(270)-FA-HEGERA-CL. In particular, medium molecular weight (i.e., 270 kDa) hyaluronic acid (HA) grafted at various extents (i.e., 10, 20, and 40%) with fluorogenic ferulic acid (FA) residue bonding propargyl groups were used in the CuAAC reaction with novel azido-terminated crosslinking agents Tri(Ethylene Glycol) Ethyl Resorcinol Acrylate (TEGERA) and Hexa(Ethylene Glycol) Ethyl Resorcinol Acrylate (HEGERA). The resulting HA(270)-FA-TEGERA-CL and HA(270)-FA-HEGERA-CL materials were characterized from the point of view of their structure by performing NMR studies. Moreover, the swelling behavior and rheological features were assessed employing TGA and DSC analysis to evaluate the potential gel-like properties of the resulting crosslinked materials. Despite the 3D crosslinked structure, HA(270)-FA-TEGERA-CL and HA(270)-FA-HEGERA-CL frameworks showed adequate swelling performance, the required shear thinning behavior, and coefficient of friction values close to those of the main commercial HA solutions used as viscosupplements (i.e., 0.20 at 10 mm/s). Furthermore, the presence of a crosslinked structure guaranteed a longer residence time. Indeed, HA(270)-FA-TEGERA-CL-40 and HA(270)-FA-HEGERA-CL-40 after 48 h showed a four times greater enzymatic resistance than the commercial viscosupplements. Based on the promising obtained results, the crosslinked materials are proposed for their potential applicability as novel viscosupplements.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | - Andrea Cappelli
- Dipartimento di Biotecnologie, Chimica e Farmacia, Università degli Studi di Siena, Via Aldo Moro 2, 53100 Siena, Italy; (M.S.); (S.P.); (M.P.); (J.V.); (G.G.); (C.B.); (G.L.); (A.M.); (C.R.)
| |
Collapse
|
32
|
Han EH, Cho SH, Lee SN, Cho MY, Lee H, Lee SY, Ngoc Thi Tran C, Park HS, Min JY, Kim HM, Park MS, Kim TD, Lim YT, Hong KS. 3D Scaffold-Based Culture System Enhances Preclinical Evaluation of Natural Killer Cell Therapy in A549 Lung Cancer Cells. ACS APPLIED BIO MATERIALS 2024; 7:7194-7206. [PMID: 39392900 DOI: 10.1021/acsabm.4c00800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2024]
Abstract
Cell-based immunotherapies have emerged as promising cancer treatment modalities, demonstrating remarkable clinical efficacy. As interest in applying immune cell-based therapies to solid tumors has gained momentum, experimental models that enable long-term monitoring and mimic clinical administration are increasingly necessary. This study explores the potential of scaffold-based cell culture technologies, specifically three-dimensional (3D) extracellular matrix (ECM)-like frameworks, as promising solutions. These frameworks facilitate unhindered immune cell growth and enable continuous cancer cell culture. The three-dimensional (3D) cell culture model was developed using tailored scaffolds for natural killer (NK) cell culture. Within this framework, A549 lung cancer cells were cocultured with NK cells, allowing real-time monitoring for up to 28 days. The expression of critical markers associated with anticancer drug resistance and epithelial-mesenchymal transition (EMT) was evaluated in cancer cells within this 3D culture context. Compared to conventional 2D monolayer cultures, this 3D scaffold-based culture revealed that solid tumor cells, specifically A549 cells, exhibited heightened resistance to anticancer drugs. Additionally, the 3D culture environment upregulated the expression of EMT markers namely vimentin, N-cadherin, and fibronectin, while NK and zEGFR-CAR-NK cells displayed anticancer effects. In the two-dimensional (2D) coculture, only zEGFR-CAR-NK cells exhibited such effects in the 3D coculture system, highlighting an intriguing inconsistency with the 2D culture model, further confirmed by in vivo experiments. This in vitro 3D cell culture model reliably predicts outcomes in NK immunotherapy experiments. Thus, it represents a valuable tool for investigating drug resistance mechanisms and assessing the efficacy of immune cell-based therapies. By bridging the gap between in vitro and in vivo investigations, this model effectively translates potential treatments into animal models and facilitates rigorous preclinical evaluations.
Collapse
Affiliation(s)
- Eun Hee Han
- Biopharmaceutical Research Center, Ochang Institute of Biological and Environmental Science, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea
- Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Sun-Hee Cho
- Biopharmaceutical Research Center, Ochang Institute of Biological and Environmental Science, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea
| | - Sang Nam Lee
- SKKU Advanced Institute of Nanotechnology (SAINT) and School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Mi Young Cho
- Biopharmaceutical Research Center, Ochang Institute of Biological and Environmental Science, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea
- SKKU Advanced Institute of Nanotechnology (SAINT) and School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hyunseung Lee
- Biopharmaceutical Research Center, Ochang Institute of Biological and Environmental Science, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea
| | - Soo Yun Lee
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Chau Ngoc Thi Tran
- Biopharmaceutical Research Center, Ochang Institute of Biological and Environmental Science, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea
- Department of Chemistry, Chung-Ang University, Seoul 06974, Republic of Korea
| | - Hye Sun Park
- Biopharmaceutical Research Center, Ochang Institute of Biological and Environmental Science, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea
| | - Jin Young Min
- Biopharmaceutical Research Center, Ochang Institute of Biological and Environmental Science, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea
| | - Hye Min Kim
- Biopharmaceutical Research Center, Ochang Institute of Biological and Environmental Science, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea
- Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Min Sung Park
- Biopharmaceutical Research Center, Ochang Institute of Biological and Environmental Science, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea
- Korea University of Science and Technology (UST), Daejeon 34113, Republic of Korea
| | - Tae-Don Kim
- Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon 34141, Republic of Korea
| | - Yong Taik Lim
- SKKU Advanced Institute of Nanotechnology (SAINT) and School of Chemical Engineering, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Kwan Soo Hong
- Biopharmaceutical Research Center, Ochang Institute of Biological and Environmental Science, Korea Basic Science Institute (KBSI), Cheongju 28119, Republic of Korea
- Department of Chemistry, Chung-Ang University, Seoul 06974, Republic of Korea
| |
Collapse
|
33
|
张 秀, 王 家, 解 慧. [Application and progress of bio-derived materials in bladder regeneration and repair]. ZHONGGUO XIU FU CHONG JIAN WAI KE ZA ZHI = ZHONGGUO XIUFU CHONGJIAN WAIKE ZAZHI = CHINESE JOURNAL OF REPARATIVE AND RECONSTRUCTIVE SURGERY 2024; 38:1299-1306. [PMID: 39542618 PMCID: PMC11563737 DOI: 10.7507/1002-1892.202404099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 08/21/2024] [Accepted: 08/21/2024] [Indexed: 11/17/2024]
Abstract
Objective To summarize the research progress of bio-derived materials used for bladder regeneration and repair. Methods The recent domestic and foreign sutudies on bio-derived materials used for bladder regeneration and repair, including classification, morphology optimization process, tissue regeneration strategies, and relevant clinical trials, were summarized and analyzed. Results Numerous types of bio-derived materials are employed in bladder regeneration and repair, characterized by their low immunogenicity and high inducible activity. Surface modification, gelation, and other morphology optimization process have significantly broadened the application scope of bio-derived materials. These advancements have effectively addressed complications, such as perforation and urolith formation, that may arise during bladder regeneration and repair. The strategy of tissue regeneration utilizing bio-derived materials, targeting the regeneration of bladder epithelium, smooth muscle, blood vessels, and nerves, offers a novel approach to achieving functional regeneration of bladder. Bio-derived materials show great promise for use in bladder regeneration and repair, yet the results from clinical trials with these materials have been less than satisfactory. Conclusion Bio-derived materials are widely used in bladder regeneration and repair due to the good biocompatibility, low immunogenicity, and degradable properties, yet face a series of problems, and there are no commercialized bladder tissue engineering grafts used in clinical treatment.
Collapse
Affiliation(s)
- 秀珍 张
- 四川大学华西医院干细胞与组织工程研究中心(成都 610041)Stem Cell and Tissue Engineering Research Center, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P. R. China
| | - 家玮 王
- 四川大学华西医院干细胞与组织工程研究中心(成都 610041)Stem Cell and Tissue Engineering Research Center, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P. R. China
| | - 慧琪 解
- 四川大学华西医院干细胞与组织工程研究中心(成都 610041)Stem Cell and Tissue Engineering Research Center, West China Hospital, Sichuan University, Chengdu Sichuan, 610041, P. R. China
| |
Collapse
|
34
|
Abou-Okeil A, Refaei R, Moustafa SE, Ibrahim HM. Magnetic hydrogel scaffold based on hyaluronic acid/chitosan and gelatin natural polymers. Sci Rep 2024; 14:28206. [PMID: 39548201 PMCID: PMC11567962 DOI: 10.1038/s41598-024-78696-6] [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: 08/23/2024] [Accepted: 11/04/2024] [Indexed: 11/17/2024] Open
Abstract
Owing to their native extracellular matrix-like features, magnetic hydrogels have been proven to be promising biomaterials as tissue engineering templates In the present work, magnetic hydrogels scaffold based on chitosan, gelatin, hyaluronic acid, containing Fe3O4 as magnetic nanoparticles (IONPs) were prepared. The prepared hydrogels were loaded with ciprofloxacin hydrochloride as a model drug. The magnetic hydrogel was prepared using different volumes of chitosan, 1%, gelatin, 10%, and hyaluronic acid, 1% in glutaraldehyde as the crosslinking agent and Fe3O4 as magnetic nanoparticles. The hydrogel scaffold and magnetic scaffold hydrogel samples were characterized by scanning electron microscopy (SEM), vibrating sample magnetometry (VSM), and Fourier-transform infrared spectroscopy (FTIR). The porosity, mechanical properties, swelling degree, and antibacterial activity of the hydrogel scaffold were also determined as well as the drug release profiles of the hydrogels. SEM imaging revealed that the magnetic hydrogel scaffold showed a relatively rough morphology with an irregular surface. The data obtained indicated that the hydrogel surface has three-dimensional porous microstructures and the porosity varied depending on the hydrogel formulation. The breaking load of the hydrogel scaffold increased from 1.361 Kgf to 4.98 Kgf by increasing the glutaraldehyde concentration from 0.2 mL to 0.8 mL. Swelling degree values in water were from 250 to 2000% after 24 h. The antibacterial activity of the hydrogel scaffold ranged from 54% to about 97% for Gram-positive bacteria (S. aureus) and from about 26-92% for Gram-negative bacteria (E. coli). The ciprofloxacin hydrochloride loaded hydrogel has a sustained release of ciprofloxacin hydrochloride over 10 h. The presence of IONPs gave a faster release of ciprofloxacin hydrochloride.
Collapse
Affiliation(s)
- Ashraf Abou-Okeil
- National Research Centre, Textile Research and Technology Institute, 33 El Bohouth St., Dokki, Cairo, P.O.12622, Egypt
| | - Rakia Refaei
- National Research Centre, Textile Research and Technology Institute, 33 El Bohouth St., Dokki, Cairo, P.O.12622, Egypt
| | - Shaimaa E Moustafa
- National Research Centre, Textile Research and Technology Institute, 33 El Bohouth St., Dokki, Cairo, P.O.12622, Egypt
| | - Hassan M Ibrahim
- National Research Centre, Textile Research and Technology Institute, 33 El Bohouth St., Dokki, Cairo, P.O.12622, Egypt.
- National Research Centre, Textile Research and Technology Institute, 33 El Bohouth St., Dokki, Cairo, P.O. 12622, , Egypt.
| |
Collapse
|
35
|
Nakagawa T, Ono S, Sakuma M, Ishida Y, Mizuta K, Uetsuki R, Odan K, Shintaku Y, Higuchi M, Matsushita Y, Kojima S, Hirata I, Kato K, Aikawa T. A case of bone resorption in the mentum caused by hyaluronic acid filler in a patient with skeletal Class II jaw deformity. Clin Case Rep 2024; 12:e9150. [PMID: 39525805 PMCID: PMC11543548 DOI: 10.1002/ccr3.9150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2024] [Revised: 06/04/2024] [Accepted: 06/22/2024] [Indexed: 11/16/2024] Open
Abstract
Key Clinical Message Chin augmentation by hyaluronic acid filler injection rarely causes abnormal bone resorption in the mentum. Thus, when taking the history of a patient with jaw deformity, it is imperative to check the history of treatment of the mentum. Abstract Hyaluronic acid (HA) filler injection is a common procedure in nonsurgical cosmetic chin augmentation. Due to its high biocompatibility and simple injection technique, HA filler injection is a popular procedure. However, adverse events such as allergic reactions and foreign body reactions have been reported in some cases. In this study, we report a case of skeletal Class II jaw deformity in which bone resorption was observed in the mentum after HA filler injection. The purpose of this study is to discuss the indications for HA filler injection in skeletal Class II cases that require orthognathic surgery. The patient was a 30-year-old woman. To improve retrusion of the mentum, she has been receiving HA filler injections in the mentum three times every 6 months in the cosmetic surgery clinic since 2015. However, the retrusion of the mentum did not improve, which prompted here to visit the orthodontic clinic. Radiographs and CT revealed bowl-shaped bone resorption surrounding the foreign bodies in the mentum. She was diagnosed with maxillary protrusion, vertical maxillary excess, mandibular retrusion, and bilateral condylar resorption. Bimaxillary orthognathic surgery (BOGS) and removal of residual HA fillers were planned after completion of the preoperative orthodontic treatment. After BOGS, the foreign bodies were completely removed, and the resorption cavities were filled with excess bone segments from the surgical sites. X-ray photoelectron spectroscopy analysis of the foreign bodies suggested the presence of HA. One year after the BOGS, no progression of condylar resorption occurred, and bone healing at the mentum had a good prognosis. Therefore, the patient underwent reduction and advancement genioplasty. She was satisfied with her facial profile and occlusal function. Unexpected bone resorption in the mentum caused by HA filler injection is often discovered incidentally. Although, patients may feel hesitant to confess their history of treatment of the mentum, it is important to ensure that they are carefully interviewed.
Collapse
Affiliation(s)
- Takayuki Nakagawa
- Department of Oral and Maxillofacial Surgery, Program of Dentistry, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Shigehiro Ono
- Department of Oral and Maxillofacial Surgery, Program of Dentistry, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Miyuki Sakuma
- Department of Oral and Maxillofacial Surgery, Program of Dentistry, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Yoko Ishida
- Department of Oral and Maxillofacial Surgery, Program of Dentistry, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Kuniko Mizuta
- Department of Oral and Maxillofacial Surgery, Program of Dentistry, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Ryo Uetsuki
- Department of Oral and Maxillofacial Surgery, Program of Dentistry, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Keijyuro Odan
- Department of Oral and Maxillofacial Surgery, Program of Dentistry, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Yuko Shintaku
- Department of Oral and Maxillofacial SurgerySuita Tokusyukai HospitalOsakaJapan
| | - Masataka Higuchi
- Department of Oral and Maxillofacial SurgerySuita Tokusyukai HospitalOsakaJapan
| | - Yutaka Matsushita
- Department of Oral and Maxillofacial SurgerySuita Tokusyukai HospitalOsakaJapan
| | | | - Isao Hirata
- Department of Biomaterials, Program of Dentistry, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Koichi Kato
- Department of Biomaterials, Program of Dentistry, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| | - Tomonao Aikawa
- Department of Oral and Maxillofacial Surgery, Program of Dentistry, Graduate School of Biomedical and Health SciencesHiroshima UniversityHiroshimaJapan
| |
Collapse
|
36
|
Munar-Bestard M, Vargas-Alfredo N, Ramis JM, Monjo M. Mangostanin hyaluronic acid hydrogel as an effective biocompatible alternative to chlorhexidine. Int J Biol Macromol 2024; 279:135187. [PMID: 39216568 DOI: 10.1016/j.ijbiomac.2024.135187] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2023] [Revised: 08/16/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Periodontal disease (PD) prevention and treatment products typically demonstrate excellent antibacterial activity, but recent studies have raised concerns about their toxicity on oral tissues. Therefore, finding a biocompatible alternative that retains antimicrobial properties is imperative. In this study, a chemically modified hyaluronic acid (HA) hydrogel containing mangostanin (MGTN) was developed. Native HA was chemically modified, incorporating amino and aldehyde groups in different batches of HA, allowing spontaneous crosslinking and gelation when combined at room temperature. MGTN at different concentrations was incorporated before gelation. The structure, swelling characteristics MGTN release, rheological parameters, and in vitro degradation performance of the loaded hydrogel were first evaluated in the study. Then, antimicrobial properties were tested on Porphyromonas gingivalis and its biocompatibility in 3D-engineered human gingiva. HA hydrogel was very stable and showed a sustained release for MGTN for at least 7 days. MGTN-loaded HA hydrogel showed equivalent antimicrobial activity compared to a commercial gel of HA containing 0.2 % chlorhexidine (CHX). In contrast, while MGTN HA hydrogel was biocompatible, CHX gel showed high cytotoxicity, causing cell death and tissue damage. Modified HA hydrogel allows controlled release of MGTN, resulting in a highly biocompatible hydrogel with antibacterial properties. This hydrogel is a suitable alternative therapy to prevent and treat PD.
Collapse
Affiliation(s)
- Marta Munar-Bestard
- Group of Cell Therapy and Tissue Engineering, Research Institute on Health Sciences (IUNICS), University of the Balearic Islands (UIB), Ctra. Valldemossa km 7.5, 07122 Palma, Spain; Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain.
| | - Nelson Vargas-Alfredo
- Group of Cell Therapy and Tissue Engineering, Research Institute on Health Sciences (IUNICS), University of the Balearic Islands (UIB), Ctra. Valldemossa km 7.5, 07122 Palma, Spain; Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain
| | - Joana Maria Ramis
- Group of Cell Therapy and Tissue Engineering, Research Institute on Health Sciences (IUNICS), University of the Balearic Islands (UIB), Ctra. Valldemossa km 7.5, 07122 Palma, Spain; Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain; Department of Fundamental Biology and Health Sciences Fundamental Biology, UIB, Spain.
| | - Marta Monjo
- Group of Cell Therapy and Tissue Engineering, Research Institute on Health Sciences (IUNICS), University of the Balearic Islands (UIB), Ctra. Valldemossa km 7.5, 07122 Palma, Spain; Health Research Institute of the Balearic Islands (IdISBa), 07120 Palma, Spain; Department of Fundamental Biology and Health Sciences Fundamental Biology, UIB, Spain.
| |
Collapse
|
37
|
Abedanzadeh M, Abolmaali SS, Heidari R, Aalaei E, Kaviani M, Dara M, Mohammadi S, Azarpira N, Tamaddon AM. Photo-crosslinked hyaluronic acid hydrogels designed for simultaneous delivery of mesenchymal stem cells and tannic acid: Advancing towards scarless wound healing. Int J Biol Macromol 2024; 281:136394. [PMID: 39406324 DOI: 10.1016/j.ijbiomac.2024.136394] [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: 06/11/2024] [Revised: 09/26/2024] [Accepted: 10/05/2024] [Indexed: 10/25/2024]
Abstract
The quest for scarless wound healing is imperative in healthcare, aiming to diminish the challenges of conventional wound treatment. Hyaluronic acid (HA), a key component of the skin's extracellular matrix, plays a pivotal role in wound healing and skin rejuvenation. Leveraging the advantages of HA hydrogels, this research focuses first on tuning the physicochemical and mechanical properties of photo-crosslinkable methacrylated HA (MAHA) by varying the methacrylation degree, polymer concentration, photo-crosslinker concentration, and UV exposure time. The optimized hydrogel, featuring suitable porosity, swelling ratio, degradability, and mechanical properties, was then used for the combined delivery of tannic acid (TA), known for its hemostatic, antibacterial, and antioxidant properties, and Wharton's jelly-derived mesenchymal stem cells (WJ-MSCs) cultured on the MAHA-TA hydrogel to enhance skin regeneration. The composite MAHA-TA-MSC hydrogel demonstrated favorable pores and biocompatibility, evidenced by cell viability, and promoted cell proliferation. When applied to dorsal wounds in rats, this composite hydrogel accelerated wound healing and reduced scarring. Additionally, molecular and histopathological analyses revealed increased expression of IL-10, the TGF-β3/TGF-β1 ratio, and the Collagen III/Collagen I ratio. These findings suggest that the MAHA-TA-MSC hydrogel is a promising candidate for scarless acute wound healing.
Collapse
Affiliation(s)
- Mozhgan Abedanzadeh
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Samira Sadat Abolmaali
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Reza Heidari
- Pharmaceutical Sciences Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Ehsan Aalaei
- Department of Mechanical Engineering, University of Washington, Seattle, WA, USA.
| | - Maryam Kaviani
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Mahintaj Dara
- Stem Cells Technology Research Center, Shiraz University of Medical Sciences, Shiraz, Iran.
| | - Samaneh Mohammadi
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Negar Azarpira
- Transplant Research Center, Shiraz University of Medical Sciences, Shiraz, Iran
| | - Ali Mohammad Tamaddon
- Department of Pharmaceutical Nanotechnology, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran; Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz, Iran; Pharmaceutics Departments, School of Pharmacy, Shiraz University of Medical Sciences, Shiraz, Iran
| |
Collapse
|
38
|
Nakamura K, Di Caprio N, Burdick JA. Engineered Shape-Morphing Transitions in Hydrogels Through Suspension Bath Printing of Temperature-Responsive Granular Hydrogel Inks. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2410661. [PMID: 39358935 PMCID: PMC11588557 DOI: 10.1002/adma.202410661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 09/12/2024] [Indexed: 10/04/2024]
Abstract
4D printing of hydrogels is an emerging technology used to fabricate shape-morphing soft materials that are responsive to external stimuli for use in soft robotics and biomedical applications. Soft materials are technically challenging to process with current 4D printing methods, which limits the design and actuation potential of printed structures. Here, a simple multi-material 4D printing technique is developed that combines dynamic temperature-responsive granular hydrogel inks based on hyaluronic acid, whose actuation is modulated via poly(N-isopropylacrylamide) crosslinker design, with granular suspension bath printing that provides structural support during and after the printing process. Granular hydrogels are easily extruded upon jamming due to their shear-thinning properties and their porous structure enables rapid actuation kinetics (i.e., seconds). Granular suspension baths support responsive ink deposition into complex patterns due to shear-yielding to fabricate multi-material objects that can be post-crosslinked to obtain anisotropic shape transformations. Dynamic actuation is explored by varying printing patterns and bath shapes, achieving complex shape transformations such as 'S'-shaped and hemisphere structures. Furthermore, stepwise actuation is programmed into multi-material structures by using microgels with varied transition temperatures. Overall, this approach offers a simple method to fabricate programmable soft actuators with rapid kinetics and precise control over shape morphing.
Collapse
Affiliation(s)
- Keisuke Nakamura
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303 USA
| | - Nikolas Di Caprio
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104 USA
| | - Jason A. Burdick
- BioFrontiers Institute, University of Colorado Boulder, Boulder, CO, 80303 USA
- Department of Bioengineering, University of Pennsylvania, Philadelphia, PA, 19104 USA
- Department of Chemical and Biological Engineering, University of Colorado Boulder, Boulder, CO, 80303 USA
| |
Collapse
|
39
|
Lan X, Du T, Zhuo J, Wang T, Shu R, Li Y, Zhang W, Ji Y, Wang Y, Yue X, Wang J. Advances of biomacromolecule-based antibacterial hydrogels and their performance evaluation for wound healing: A review. Int J Biol Macromol 2024; 279:135577. [PMID: 39270907 DOI: 10.1016/j.ijbiomac.2024.135577] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2024] [Revised: 09/09/2024] [Accepted: 09/10/2024] [Indexed: 09/15/2024]
Abstract
Biomacromolecule hydrogels possess excellent mechanical properties and biocompatibility, but their inability to combat bacteria restricts their application in the biomedical field. With the increasing requirements and demands for hydrogel dressings, wound dressings with antibacterial properties of biomacromolecule hydrogels reinforced by adding antibacterial agents have attracted much attention, and related reviews are emerging. In this paper, the advances of biomacromolecule antibacterial hydrogels (including chitosan, sodium alginate, Hyaluronic acid, cellulose and gelatin) were first overviewed, and the antibacterial agents incorporated into hydrogels were classified (including metals and their derivatives, carbon-based materials, and native compounds). A series of performance evaluations of antibacterial hydrogels in the process of promoting wound healing were then reviewed, including basic properties (mechanical, rheological, injectable and self-healing, etc.), in vitro experiments (hemostasis, antibacterial, anti-inflammatory, anti-oxidation, biocompatibility) and in vivo experiments (in vivo model, histomorphology analysis, cytokines). Finally, the future development of biomacromolecule-based antibacterial hydrogels for wound healing is prospected. This work can provide a useful reference for researchers to prepare practical new wound hydrogel dressings.
Collapse
Affiliation(s)
- Xi Lan
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Rode, Yangling 712100, Shaanxi, China
| | - Ting Du
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Rode, Yangling 712100, Shaanxi, China
| | - Junchen Zhuo
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Rode, Yangling 712100, Shaanxi, China
| | - Tianyu Wang
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Rode, Yangling 712100, Shaanxi, China
| | - Rui Shu
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Rode, Yangling 712100, Shaanxi, China
| | - Yuechun Li
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Rode, Yangling 712100, Shaanxi, China
| | - Wentao Zhang
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Rode, Yangling 712100, Shaanxi, China
| | - Yanwei Ji
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Rode, Yangling 712100, Shaanxi, China
| | - Yanru Wang
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Rode, Yangling 712100, Shaanxi, China
| | - Xiaoyue Yue
- College of Food and Bioengineering, Zhengzhou University of Light Industry, Zhengzhou, Henan Province 450001, China; Key Laboratory of Cold Chain Food Processing and Safety Control (Zhengzhou University of Light Industry), Ministry of Education, Zhengzhou 450001, China
| | - Jianlong Wang
- College of Food Science and Engineering, Northwest A&F University, 22 Xinong Rode, Yangling 712100, Shaanxi, China.
| |
Collapse
|
40
|
Moghaddam MM, Jooybar E, Imani R, Ehrbar M. Development of injectable microgel-based scaffolds via enzymatic cross-linking of hyaluronic acid-tyramine/gelatin-tyramine for potential bone tissue engineering. Int J Biol Macromol 2024; 279:135176. [PMID: 39214205 DOI: 10.1016/j.ijbiomac.2024.135176] [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: 05/08/2024] [Revised: 08/22/2024] [Accepted: 08/27/2024] [Indexed: 09/04/2024]
Abstract
Currently, the healing of large bone defects relies on invasive surgeries and the transplantation of autologous bone. As a less invasive treatment option, the provision of microenvironments that promote the regeneration of defective bones holds great promise. Here, we developed hyaluronic acid (HA)/gelatin (Ge) microgel-based scaffolds to guide bone regeneration. To enable the formation of microgels by enzymatic cross-linking in the presence of horseradish peroxidase (HRP) and hydrogen peroxide (H2O2), we modified the polymers with tyramine (TA). Spectrophotometry and proton nuclear magnetic resonance (1H NMR) spectroscopy analysis confirmed successful tyramine substitution on polymer backbones. To enable the formation of microgels by a water-in-oil emulsion approach, the HRP and H2O2 concentrations were tuned to achieve the gelation in a few seconds. By varying the stirring speed from 600 to 1000 rpm, spherical microgels were produced with an average size of 116 ± 8.7 and 68 ± 4.7 μm, respectively. The results showed that microgels were injectable through needles and showed good biocompatibility with the cultured human osteosarcoma cell line (MG-63). HA/Ge-TA microgels served as a promising substrate for MG-63 cells since they improved the alkaline phosphatase activity and level of calcium deposition. In summary, the developed HA/Ge-TA microgels are promising injectable microgel-based scaffolds in bone tissue engineering.
Collapse
Affiliation(s)
- Melika Mansouri Moghaddam
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran
| | - Elaheh Jooybar
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran.
| | - Rana Imani
- Department of Biomedical Engineering, Amirkabir University of Technology (Tehran Polytechnic), Tehran, Iran.
| | - Martin Ehrbar
- Department of Obstetrics, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| |
Collapse
|
41
|
Wang F, Xu Y, Zhou Q, Xie L. Biomolecule-based hydrogels as delivery systems for limbal stem cell transplantation: A review. Int J Biol Macromol 2024; 280:135778. [PMID: 39304050 DOI: 10.1016/j.ijbiomac.2024.135778] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 08/25/2024] [Accepted: 09/17/2024] [Indexed: 09/22/2024]
Abstract
Limbal stem cell deficiency (LSCD) is a complex disease of the cornea resulting from dysfunction and/or loss of limbal stem cells (LSCs) and their niche. Most patients with LSCD cannot be treated by conventional corneal transplants because the donor tissue lacks the LSCs necessary for corneal epithelial regeneration. Successful treatment of LSCD depends on effective stem cell transplantation to the ocular surface for replenishment of the LSC reservoir. Thus, stem cell therapies employing carrier substrates for LSCs have been widely explored. Hydrogel biomaterials have many favorable characteristics, including hydrophilicity, flexibility, cytocompatibility, and optical properties suitable for the transplantation of LSCs. Therefore, due to these properties, along with the necessary signals for stem cell proliferation and differentiation, hydrogels are ideal carrier substrates for LSCD treatment. This review summarizes the use of different medical-type hydrogels in LSC transplantation from 2001 to 2024. First, a brief background of LSCD is provided. Then, studies that employed various hydrogel scaffolds as LSC carriers are highlighted to provide a multimodal strategic reference for LSCD treatment. Finally, an analysis of prospective future developments and challenges in the field of hydrogels as LSC carriers for treating LSCD is presented.
Collapse
Affiliation(s)
- Fuyan Wang
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, School of Ophthalmology, Shandong First Medical University, Qingdao 266071, China
| | - Yuehe Xu
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, School of Ophthalmology, Shandong First Medical University, Qingdao 266071, China
| | - Qingjun Zhou
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, School of Ophthalmology, Shandong First Medical University, Qingdao 266071, China.
| | - Lixin Xie
- Eye Institute of Shandong First Medical University, Qingdao Eye Hospital of Shandong First Medical University, State Key Laboratory Cultivation Base, Shandong Provincial Key Laboratory of Ophthalmology, School of Ophthalmology, Shandong First Medical University, Qingdao 266071, China.
| |
Collapse
|
42
|
Ye Z, Sun L, Xiang Q, Hao Y, Liu H, He Q, Yang X, Liao W. Advancements of Biomacromolecular Hydrogel Applications in Food Nutrition and Health. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:23689-23708. [PMID: 39410660 DOI: 10.1021/acs.jafc.4c05903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/01/2024]
Abstract
Hydrogels exhibit remarkable degradability, biocompatibility and functionality, which position them as highly promising materials for applications within the food and pharmaceutical industries. Although many relevant studies on hydrogels have been reported in the chemical industry, materials, and other fields, there have been few reviews on their potential applications in food nutrition and human health. This study aims to address this gap by reviewing the functional properties of hydrogels and assessing their value in terms of food nutrition and human health. The use of hydrogels in preserving bioactive ingredients, food packaging and food distribution is delved into specifically in this review. Hydrogels can serve as cutting-edge materials for food packaging and delivery, ensuring the preservation of nutritional activity within food products, facilitating targeted delivery of bioactive compounds and regulating the digestion and absorption processes in the human body, thereby promoting human health. Moreover, hydrogels find applications in in vitro cell and tissue culture, human tissue repair, as well as chronic disease prevention and treatment. These broad applications have attracted great attention in the fields of human food nutrition and health. Ultimately, this paper serves as a valuable reference for further utilization and exploration of hydrogels in these respective fields.
Collapse
Affiliation(s)
- Zichong Ye
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Linye Sun
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Qianru Xiang
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Yuting Hao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Hongji Liu
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| | - Qi He
- Food Safety and Health Research Center, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, P. R. China
| | - Xingfen Yang
- Food Safety and Health Research Center, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou 510515, P. R. China
| | - Wenzhen Liao
- Department of Nutrition and Food Hygiene, Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, Guangdong 510515, P. R. China
| |
Collapse
|
43
|
Zhang M, Ye Q, Zhu Z, Shi S, Xu C, Xie R, Li Y. Hyaluronic Acid-Based Dynamic Hydrogels for Cartilage Repair and Regeneration. Gels 2024; 10:703. [PMID: 39590059 PMCID: PMC11594165 DOI: 10.3390/gels10110703] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 10/27/2024] [Accepted: 10/29/2024] [Indexed: 11/28/2024] Open
Abstract
Hyaluronic acid (HA), an important natural polysaccharide and meanwhile, an essential component of extracellular matrix (ECM), has been widely used in tissue repair and regeneration due to its high biocompatibility, biodegradation, and bioactivity, and the versatile chemical groups for modification. Specially, HA-based dynamic hydrogels, compared with the conventional hydrogels, offer an adaptable network and biomimetic microenvironment to optimize tissue repair and the regeneration process with a striking resemblance to ECM. Herein, this review comprehensively summarizes the recent advances of HA-based dynamic hydrogels and focuses on their applications in articular cartilage repair. First, the fabrication methods and advantages of HA dynamic hydrogels are presented. Then, the applications of HA dynamic hydrogels in cartilage repair are illustrated from the perspective of cell-free and cell-encapsulated and/or bioactive molecules (drugs, factors, and ions). Finally, the current challenges and prospective directions are outlined.
Collapse
Affiliation(s)
- Mingshuo Zhang
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China; (M.Z.); (Q.Y.); (Z.Z.); (S.S.)
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China;
| | - Qianwen Ye
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China; (M.Z.); (Q.Y.); (Z.Z.); (S.S.)
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China;
| | - Zebo Zhu
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China; (M.Z.); (Q.Y.); (Z.Z.); (S.S.)
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China;
| | - Shuanglian Shi
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China; (M.Z.); (Q.Y.); (Z.Z.); (S.S.)
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China;
| | - Chunming Xu
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China;
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| | - Renjian Xie
- School of Medical Information Engineering, Gannan Medical University, Ganzhou 341000, China; (M.Z.); (Q.Y.); (Z.Z.); (S.S.)
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China;
- Key Laboratory of Prevention and Treatment of Cardiovascular and Cerebrovascular Diseases (Ministry of Education), Gannan Medical University, Ganzhou 341000, China
| | - Yumei Li
- Jiangxi Provincial Key Laboratory of Tissue Engineering (2024SSY06291), Gannan Medical University, Ganzhou 341000, China;
- School of Basic Medicine, Gannan Medical University, Ganzhou 341000, China
| |
Collapse
|
44
|
Liu Y, Chen W, Gao Y, Wei K. Anti-inflammatory dressing based on hyaluronic acid and hydroxyethyl starch for wound healing. Int J Biol Macromol 2024; 282:137078. [PMID: 39481723 DOI: 10.1016/j.ijbiomac.2024.137078] [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: 08/27/2024] [Revised: 10/09/2024] [Accepted: 10/28/2024] [Indexed: 11/02/2024]
Abstract
Eliminating persistent inflammation and choosing dressings that provide the best healing environment is key to promoting wound healing. Dynamic and reversible hydrogels have attracted much attention because of their capacity to adapt to irregular wound surfaces. Herein, oxidized hydroxyethyl starch (OHES) and hyaluronic acid (HA-ADH) were crosslinked via the dynamic acylhydrazone bond to form an anti-inflammatory function hydrogel (HA-ADH/OHES@XT) that could release xanthatin (XT) slowly. The HA-ADH/OHES hydrogels showed an appropriate gelation time, notable water-retaining capacity, self-healing, suitable biodegradability, and good biocompatibility for wound healing applications. In vivo experiments demonstrated that HA-ADH/OHES@XT hydrogels promoted tissue regeneration and wound healing at a rate of approximately 89.1 % on day 20 by reducing inflammation, increasing collagen deposition, and promoting re-epithelialization, indicating their great potential as a wound dressing.
Collapse
Affiliation(s)
- Yuanqi Liu
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, PR China
| | - Wenyu Chen
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, PR China
| | - Yuanyuan Gao
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, PR China
| | - Kun Wei
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou 510006, PR China.
| |
Collapse
|
45
|
Arslan H, Davuluri A, Nguyen HH, So BR, Lee J, Jeon J, Yum K. 3D Bioprinting Using Universal Fugitive Network Bioinks. ACS APPLIED BIO MATERIALS 2024; 7:7040-7050. [PMID: 39291381 DOI: 10.1021/acsabm.4c01220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/19/2024]
Abstract
Three-dimensional (3D) bioprinting has emerged with potential for creating functional 3D tissues with customized geometries. However, the limited availability of bioinks capable of printing 3D structures with both high-shape fidelity and desired biological environments for encapsulated cells remains a key challenge. Here, we present a 3D bioprinting approach that uses universal fugitive network bioinks prepared by loading cells and hydrogel precursors (bioink base materials) into a 3D printable fugitive carrier. This approach constructs 3D structures of cell-encapsulated hydrogels by printing 3D structures using fugitive network bioinks, followed by cross-linking printed structures and removing the carrier from them. The use of the fugitive carrier decouples the 3D printability of bioinks from the material properties of bioink base materials, making this approach readily applicable to a range of hydrogel systems. The decoupling also enables the design of bioinks for the biological functionality of the final printed constructs without compromising the 3D printability. We demonstrate the generalizable 3D printability by printing self-supporting 3D structures of various hydrogels, including conventionally non-3D printable hydrogels and those with a low polymer content. We conduct preprinting screening of bioink base materials through 3D cell culture to select bioinks with high cell compatibility. The selected bioinks produce 3D constructs of cell-encapsulated hydrogels with both high-shape fidelity and high cell viability and proliferation. The universal fugitive network bioink platform could significantly expand 3D printable bioinks with customizable biological functionalities and the adoption of 3D bioprinting in diverse research and applied settings.
Collapse
Affiliation(s)
- Hakan Arslan
- Department of Materials Science and Engineering, University of Texas at Arlington, Arlington, Texas 76019, United States
- Department of Mechanical and Aerospace Engineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Aneela Davuluri
- Department of Materials Science and Engineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Hiep H Nguyen
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Byung Ran So
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Juhyun Lee
- Department of Bioengineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Junha Jeon
- Department of Chemistry and Biochemistry, University of Texas at Arlington, Arlington, Texas 76019, United States
| | - Kyungsuk Yum
- Department of Materials Science and Engineering, University of Texas at Arlington, Arlington, Texas 76019, United States
| |
Collapse
|
46
|
Villani C, Murugan P, George A. Exosome-Laden Hydrogels as Promising Carriers for Oral and Bone Tissue Engineering: Insight into Cell-Free Drug Delivery. Int J Mol Sci 2024; 25:11092. [PMID: 39456873 PMCID: PMC11508290 DOI: 10.3390/ijms252011092] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Revised: 10/08/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Mineralization is a key biological process that is required for the development and repair of tissues such as teeth, bone and cartilage. Exosomes (Exo) are a subset of extracellular vesicles (~50-150 nm) that are secreted by cells and contain genetic material, proteins, lipids, nucleic acids, and other biological substances that have been extensively researched for bone and oral tissue regeneration. However, Exo-free biomaterials or exosome treatments exhibit poor bioavailability and lack controlled release mechanisms at the target site during tissue regeneration. By encapsulating the Exos into biomaterials like hydrogels, these disadvantages can be mitigated. Several tissue engineering approaches, such as those for wound healing processes in diabetes mellitus, treatment of osteoarthritis (OA) and cartilage degeneration, repair of intervertebral disc degeneration, and cardiovascular diseases, etc., have been exploited to deliver exosomes containing a variety of therapeutic and diagnostic cargos to target tissues. Despite the significant efficacy of Exo-laden hydrogels, their use in mineralized tissues, such as oral and bone tissue, is very sparse. This review aims to explore and summarize the literature related to the therapeutic potential of hydrogel-encapsulated exosomes for bone and oral tissue engineering and provides insight and practical procedures for the development of future clinical techniques.
Collapse
Affiliation(s)
| | | | - Anne George
- Brodie Tooth Development Genetics & Regenerative Medicine Research Laboratory, Department of Oral Biology, University of Illinois at Chicago, Chicago, IL 60612, USA; (C.V.); (P.M.)
| |
Collapse
|
47
|
Shirk BD, Heichel DL, Eccles LE, Rodgers LI, Lateef AH, Burke KA, Stoppel WL. Modifying Naturally Occurring, Nonmammalian-Sourced Biopolymers for Biomedical Applications. ACS Biomater Sci Eng 2024; 10:5915-5938. [PMID: 39259773 DOI: 10.1021/acsbiomaterials.4c00689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/13/2024]
Abstract
Natural biopolymers have a rich history, with many uses across the fields of healthcare and medicine, including formulations for wound dressings, surgical implants, tissue culture substrates, and drug delivery vehicles. Yet, synthetic-based materials have been more successful in translation due to precise control and regulation achievable during manufacturing. However, there is a renewed interest in natural biopolymers, which offer a diverse landscape of architecture, sustainable sourcing, functional groups, and properties that synthetic counterparts cannot fully replicate as processing and sourcing of these materials has improved. Proteins and polysaccharides derived from various sources (crustaceans, plants, insects, etc.) are highlighted in this review. We discuss the common types of polysaccharide and protein biopolymers used in healthcare and medicine, highlighting methods and strategies to alter structures and intra- and interchain interactions to engineer specific functions, products, or materials. We focus on biopolymers obtained from natural, nonmammalian sources, including silk fibroins, alginates, chitosans, chitins, mucins, keratins, and resilins, while discussing strategies to improve upon their innate properties and sourcing standardization to expand their clinical uses and relevance. Emphasis will be placed on methods that preserve the structural integrity and native biological functions of the biopolymers and their makers. We will conclude by discussing the untapped potential of new technologies to manipulate native biopolymers while controlling their secondary and tertiary structures, offering a perspective on advancing biopolymer utility in novel applications within biomedical engineering, advanced manufacturing, and tissue engineering.
Collapse
Affiliation(s)
- Bryce D Shirk
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Danielle L Heichel
- Department of Chemical Engineering, University of Connecticut, Storrs, Connecticut 06269-3222, United States
- Polymer Program, Institute of Materials Science, University of Connecticut, Storrs, Connecticut 06269-3136, United States
| | - Lauren E Eccles
- Department of Chemical Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Liam I Rodgers
- Department of Chemical Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Ali H Lateef
- Department of Chemical Engineering, University of Florida, Gainesville, Florida 32611, United States
| | - Kelly A Burke
- Department of Chemical Engineering, University of Connecticut, Storrs, Connecticut 06269-3222, United States
- Polymer Program, Institute of Materials Science, University of Connecticut, Storrs, Connecticut 06269-3136, United States
| | - Whitney L Stoppel
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, Gainesville, Florida 32611, United States
- Department of Chemical Engineering, University of Florida, Gainesville, Florida 32611, United States
| |
Collapse
|
48
|
Merghes P, Ilia G, Maranescu B, Varan N, Simulescu V. The Sol-Gel Process, a Green Method Used to Obtain Hybrid Materials Containing Phosphorus and Zirconium. Gels 2024; 10:656. [PMID: 39451309 PMCID: PMC11507187 DOI: 10.3390/gels10100656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/08/2024] [Accepted: 10/12/2024] [Indexed: 10/26/2024] Open
Abstract
The sol-gel process is a green method used in the last few decades to synthesize new organic-inorganic phosphorus-containing hybrid materials. The sol-gel synthesis is a green method because it takes place in mild conditions, mostly by using water or alcohol as solvents, at room temperature. Therefore, the sol-gel method is, among others, a promising route for obtaining metal-phosphonate networks. In addition to phosphorus, the obtained hybrid materials could also contain titanium, zirconium, boron, and other elements, which influence their properties. The sol-gel process has two steps: first, the sol formation, and second, the transition to the gel phase. In other words, the sol-gel process converts the precursors into a colloidal solution (sol), followed by obtaining a network (gel). By using the sol-gel method, different organic moieties could be introduced into an inorganic matrix, resulting in organic-inorganic hybrid structures (sometimes they are also referred as organic-inorganic copolymers).
Collapse
Affiliation(s)
- Petru Merghes
- “King Mihai I” University of Life Sciences from Timisoara, Calea Aradului 119, 300645 Timisoara, Romania; (P.M.); (N.V.)
| | - Gheorghe Ilia
- Faculty of Chemistry, Biology, Geography, West University Timisoara, 16 Pestalozzi Street, 300115 Timisoara, Romania;
| | - Bianca Maranescu
- Faculty of Chemistry, Biology, Geography, West University Timisoara, 16 Pestalozzi Street, 300115 Timisoara, Romania;
| | - Narcis Varan
- “King Mihai I” University of Life Sciences from Timisoara, Calea Aradului 119, 300645 Timisoara, Romania; (P.M.); (N.V.)
| | - Vasile Simulescu
- Faculty of Chemistry, Biology, Geography, West University Timisoara, 16 Pestalozzi Street, 300115 Timisoara, Romania;
| |
Collapse
|
49
|
Morgan FC, Beeren IAO, Bauer J, Moroni L, Baker MB. Structure-Reactivity Relationships in a Small Library of Imine-Type Dynamic Covalent Materials: Determination of Rate and Equilibrium Constants Enables Model Prediction and Validation of a Unique Mechanical Softening in Dynamic Hydrogels. J Am Chem Soc 2024; 146:27499-27516. [PMID: 39350717 PMCID: PMC11467966 DOI: 10.1021/jacs.4c08099] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2024] [Revised: 09/12/2024] [Accepted: 09/17/2024] [Indexed: 10/10/2024]
Abstract
The development of next generation soft and recyclable materials prominently features dynamic (reversible) chemistries such as host-guest, supramolecular, and dynamic covalent. Dynamic systems enable injectability, reprocessability, and time-dependent mechanical properties. These properties arise from the inherent relationship between the rate and equilibrium constants (RECs) of molecular junctions (cross-links) and the resulting macroscopic behavior of dynamic networks. However, few examples explicitly measure RECs while exploring this connection between molecular and material properties, particularly for polymeric hydrogel systems. Here we use dynamic covalent imine formation to study how single-point compositional changes in NH2-terminated nucleophiles affect binding constants and resulting hydrogel mechanical properties. We explored both model small molecule studies and model polymeric macromers, and found >3-decade change in RECs. Leveraging established relationships in the literature, we then developed a simple model to describe the cross-linking equilibrium and predict changes in hydrogel mechanical properties. Interestingly, we observed that a narrow ≈2-decade range of Keq's determine the bound fraction of imines. Our model allowed us to uncover a regime where adding cross-linker before saturation can decrease the cross-link density of a hydrogel. We then demonstrated the veracity of this predicted behavior experimentally. Notably this emergent behavior is not accounted for in covalent hydrogel theory. This study expands upon structure-reactivity relationships for imine formation, highlighting how quantitative determination of RECs facilitates predicting macroscopic behavior. Furthermore, while the present study focuses on dynamic covalent imine formation, the underlying principles of this work are applicable to the general bottom-up design of soft and recyclable dynamic materials.
Collapse
Affiliation(s)
- Francis
L. C. Morgan
- Department
of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department
of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Ivo A. O. Beeren
- Department
of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department
of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Jurica Bauer
- Department
of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Lorenzo Moroni
- Department
of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| | - Matthew B. Baker
- Department
of Instructive Biomaterials Engineering, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
- Department
of Complex Tissue Regeneration, MERLN Institute for Technology-Inspired
Regenerative Medicine, Maastricht University, 6229 ER Maastricht, The Netherlands
| |
Collapse
|
50
|
He X, He S, Xiang G, Deng L, Zhang H, Wang Y, Li J, Lu H. Precise Lubrication and Protection of Cartilage Damage by Targeting Hydrogel Microsphere. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2405943. [PMID: 39155588 DOI: 10.1002/adma.202405943] [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: 04/25/2024] [Revised: 07/31/2024] [Indexed: 08/20/2024]
Abstract
Osteoarthritis (OA) is a degenerative bone and joint disease characterized by decreased cartilage lubrication, leading to continuous wear and ultimately irreversible damage. This situation is particularly challenging for early-stage OA, as current bio-lubricants lack precise targeting for small inflammatory lesions. In this work, an antibody-mediated targeting hydrogel microspheres (HMS) is developed to precisely lubricate the local injury site of cartilage and prevent the progression of early OA. Anti-Collagen type I (Anti-Col1) is an antibody that targets cartilage injury sites in early OA stages. It is anchored on a HMS matrix made of Gelatin methacrylate (GelMA) and poly (sulfobetaine methacrylate) (PSBMA) to create targeted HMS (T-G/S HMS). The T-G/S HMS's high hydrophilicity, along with the dynamic interaction between its surficial Anti-Col1 and the Col1 on cartilage injury site, ensures its precise and effective lubrication of early OA lesions. Consequently, injecting T-G/S HMS into rats with early OA significantly slows disease progression and reduces symptoms. In conclusion, the developed injectable targeted lubricating HMS and the precisely targeted lubrication strategy represent a promising, convenient technique for treating OA, particularly for slowing the early-stage OA progression.
Collapse
Affiliation(s)
- Xiangming He
- Laboratory for Advanced Lubricating Materials, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201210, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Sihan He
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Gang Xiang
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Linhua Deng
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Hongqi Zhang
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Yunjia Wang
- Department of Spine Surgery and Orthopaedics, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, Hunan, 410008, China
| | - Jiusheng Li
- Laboratory for Advanced Lubricating Materials, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201210, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hengyi Lu
- Laboratory for Advanced Lubricating Materials, Shanghai Advanced Research Institute, Chinese Academy of Sciences, Shanghai, 201210, China
- University of Chinese Academy of Sciences, Beijing, 100049, China
| |
Collapse
|