1
|
Wang X, Liu X, Li C, Li J, Qiu M, Wang Y, Han W. Effects of molecular weights on the bioactivity of hyaluronic acid: A review. Carbohydr Res 2025; 552:109472. [PMID: 40186950 DOI: 10.1016/j.carres.2025.109472] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 03/19/2025] [Accepted: 03/27/2025] [Indexed: 04/07/2025]
Abstract
Hyaluronic acid (HA), the only non-sulfated glycosaminoglycan (GAG), is essential for maintaining the extracellular matrix's structural and functional integrity. Its bioactivity is determined by interactions between HA fragments of different molecular weights and specific receptors, which influence downstream signaling pathways. This review systematic summarizes the correlation between HA molecular weight dynamic changes and bioactivities focusing on imbalance of HA degradation and metabolism due to various pathological processes. Outline the core transduction mechanisms of HA receptors and signaling pathways, and innovatively hypothesize that discrepancies in cellular distribution with HA-molecular weights dependent lead to the activation of different signaling pathways from the perspective of molecular weight affecting cellular distribution. Finally, it addresses challenges in studying HA's biofunctions and provides new perspectives for future research.
Collapse
Affiliation(s)
- Xiaoyun Wang
- College of Life Science and Health, University of Health and Rehabilitation Sciences, Qingdao, 266113, China; Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266024, China; Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, and College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao, 266100, China
| | - Xiaojun Liu
- College of Life Science and Health, University of Health and Rehabilitation Sciences, Qingdao, 266113, China; Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266024, China
| | - Chao Li
- College of Life Science and Health, University of Health and Rehabilitation Sciences, Qingdao, 266113, China; Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266024, China
| | - Jiangtao Li
- College of Life Science and Health, University of Health and Rehabilitation Sciences, Qingdao, 266113, China
| | - Meng Qiu
- Key Laboratory of Marine Chemistry Theory and Technology, Ministry of Education, and College of Chemistry and Chemical Engineering, Ocean University of China, Qingdao, 266100, China.
| | - Yongliang Wang
- College of Life Science and Health, University of Health and Rehabilitation Sciences, Qingdao, 266113, China; Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266024, China.
| | - Wenwei Han
- College of Life Science and Health, University of Health and Rehabilitation Sciences, Qingdao, 266113, China; Qingdao Municipal Hospital, University of Health and Rehabilitation Sciences, Qingdao, 266024, China.
| |
Collapse
|
2
|
Rouco H, Permuy M, Muñoz F, Vázquez JA, Caeiro JR, Landin M, Diaz-Rodriguez P. Micelle-to-Gel: Thermosensitive intra-articular hydrogels for osteoarthritis management. J Control Release 2025; 381:113639. [PMID: 40107515 DOI: 10.1016/j.jconrel.2025.113639] [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: 09/24/2024] [Revised: 02/17/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025]
Abstract
Osteoarthritis (OA) is a chronic and degenerative joint disease with a rising incidence worldwide. Current therapeutic approaches primarily focus on symptom relief through systemic administration, which raises safety concerns related to side effects and long-term use. In this context, the local administration of natural compounds with anti-inflammatory and anti-arthritic properties, such as β-Lapachone constitutes an interesting alternative. In this work, we prepared and characterized injectable thermosensitive hybrid hydrogels loaded with β-Lapachone. A comprehensive characterization of the hydrogel systems was performed, including micellar diameter, mechanical properties at different temperatures, the ability to control drug release and microstructure. The anti-inflammatory activity of the free drug, as well as that of the blank or loaded hydrogels was then evaluated ex vivo, using OA cartilage explants. Additionally, in vivo studies were carried out in a rabbit model of OA to assess their clinical potential. The results suggest that the hydrogel systems possess a composite microstructure integrating micelles, together with a temperature-responsive stiffness and the ability to modulate drug release. In addition, β-Lapachone-loaded hydrogels display an interesting immunomodulatory potential ex vivo, as they were able to efficiently reduce the secretion of several proinflammatory mediators, such as IL-6, MMP9, MMP13 and CXCL8. Furthermore, the drug-loaded hydrogels were found to improve in vivo cartilage and bone histomorphometric markers, such as subchondral bone thickness, as well as early signs of cartilage damage, such as the fibrillation index. Therefore, the developed β-Lapachone-loaded thermosensitive hydrogels constitute a promising alternative for OA management.
Collapse
Affiliation(s)
- Helena Rouco
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Grupo I+D Farma (GI-1645), Facultad de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain; Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), IDIS Research Institute, 15706 Santiago de Compostela, Spain; Instituto de Materiais da Universidade de Santiago de Compostela (iMATUS), Spain
| | - Maria Permuy
- Anatomy, Animal Production and Veterinary Clinical Sciences Department, Veterinary Faculty, Universidade de Santiago de Compostela, Campus Universitario s/n, 27002 Lugo, Spain; iBoneLab SL, Avenida da Coruña 500, 27003 Lugo, Spain
| | - Fernando Muñoz
- Anatomy, Animal Production and Veterinary Clinical Sciences Department, Veterinary Faculty, Universidade de Santiago de Compostela, Campus Universitario s/n, 27002 Lugo, Spain; iBoneLab SL, Avenida da Coruña 500, 27003 Lugo, Spain
| | - José Antonio Vázquez
- Group of Recycling and Valorisation of Waste Materials (REVAL), Marine Research Institute (IIM-CSIC), Vigo, Spain
| | - José R Caeiro
- Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), IDIS Research Institute, 15706 Santiago de Compostela, Spain; Department of Orthopaedic Surgery and Traumatology, Complexo Hospitalario Universitario de Santiago de Compostela, Santiago de Compostela, Spain
| | - Mariana Landin
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Grupo I+D Farma (GI-1645), Facultad de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain; Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), IDIS Research Institute, 15706 Santiago de Compostela, Spain; Instituto de Materiais da Universidade de Santiago de Compostela (iMATUS), Spain.
| | - Patricia Diaz-Rodriguez
- Departamento de Farmacología, Farmacia y Tecnología Farmacéutica, Grupo I+D Farma (GI-1645), Facultad de Farmacia, Universidade de Santiago de Compostela, Santiago de Compostela 15782, Spain; Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), IDIS Research Institute, 15706 Santiago de Compostela, Spain; Instituto de Materiais da Universidade de Santiago de Compostela (iMATUS), Spain.
| |
Collapse
|
3
|
Steel R. Identification of hyaluronic acid in seized samples by Hypercarb chromatography-high resolution mass spectrometry. Drug Test Anal 2025; 17:585-588. [PMID: 38978182 DOI: 10.1002/dta.3761] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 06/17/2024] [Accepted: 06/20/2024] [Indexed: 07/10/2024]
Abstract
Hyaluronic acid (HA) has been commonly used to treat osteoarthritis and joint injuries in horses and dogs since the 1970s. HA is a polysaccharide made up of alternating N-acetyl-d-glucosamine and d-glucuronic acid residues, with polymeric molecules achieving molecular weights as high as 20 MDa. High molecular weight HA forms a viscous hydrogel when dissolved in water, making HA solutions distinct from most other pharmaceutical preparations. Clear viscous solutions are often encountered during stable and kennel inspections, but in the absence of an analytical method, it is not possible to identify if these substances contain HA or other unknown compounds. This paper presents a simple method for the identification of HA in seized materials based on chemical hydrolysis followed by Hypercarb chromatography and MS/MS analysis.
Collapse
Affiliation(s)
- Rohan Steel
- Biological Research Unit, Racing Analytical Services Ltd., Flemington, Victoria, Australia
| |
Collapse
|
4
|
Rodella G, Ma Z, Ucakar B, Joudiou N, Préat V, Gallez B, Malfanti A. Repurposing Chemotherapeutics in a Hyaluronic Acid-conjugate Combination Treatment Approach for the Local Immunomodulation of the Glioblastoma Microenvironment. Int J Pharm 2025; 676:125612. [PMID: 40252866 DOI: 10.1016/j.ijpharm.2025.125612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 04/15/2025] [Accepted: 04/15/2025] [Indexed: 04/21/2025]
Abstract
The immunosuppressive tumor immune microenvironment (TIME) renders glioblastoma (GBM) refractory to current chemo-immunotherapeutics. We sought to explore a novel approach for local GBM-associated TIME immunomodulation based on a synergistic combination of the repurposed chemotherapeutic drugs doxorubicin (DOX), which acts to induce immunogenic cell death (ICD) and gemcitabine (GEM), which depletes immunosuppressive myeloid-derived suppressor cells (MDSCs). We conjugated DOX and GEM to hyaluronic acid (HA) to improve efficacy, given this polymer's ability to target CD44 which are overexpressed on cancer cells. The HA-DOX and HA-GEM polymer-drug conjugates provided synergistic cytotoxic effects and maintained ICD-related properties in GBM cells compared to a combination of free drugs. HA-DOX and HA-GEM also reverted the immunosuppressive GBM-associated TIME in orthotopic GL261 tumor-bearing mice by selectively depleting MDSCs and reprogramming M2-like macrophages towards a pro-inflammatory M1-like state, resulting in controlled tumor growth. Local HA-DOX and HA-GEM delivery also increased median survival and controlled tumor growth in an immune refractory SB28-GBM orthotopic mouse GBM model. These findings highlight the potential of repurposing clinically applicable chemotherapeutics in the context of polymer-drug combination treatments for novel immunomodulation strategies in unresectable GBM, which may open new avenues for developing innovative therapies.
Collapse
Affiliation(s)
- Giulia Rodella
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200 Brussels, Belgium; UCLouvain, Louvain Drug Research Institute, Biomedical Magnetic Resonance, Avenue Mounier 73 B1.73.08, 1200 Brussels, Belgium
| | - Zhanjun Ma
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200 Brussels, Belgium
| | - Bernard Ucakar
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200 Brussels, Belgium
| | - Nicolas Joudiou
- UCLouvain, Louvain Drug Research Institute, Biomedical Magnetic Resonance, Avenue Mounier 73 B1.73.08, 1200 Brussels, Belgium
| | - Véronique Préat
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200 Brussels, Belgium
| | - Bernard Gallez
- UCLouvain, Louvain Drug Research Institute, Biomedical Magnetic Resonance, Avenue Mounier 73 B1.73.08, 1200 Brussels, Belgium.
| | - Alessio Malfanti
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73 B1.73.12, 1200 Brussels, Belgium; Departement of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo, 5, 35131 Padova, Italy.
| |
Collapse
|
5
|
Maxim ME, Toma RM, Aricov L, Leonties AR, Precupas A, Tatia R, Oprita EI. Unlocking the Rich Potential of a Soft Gel-Cream Enriched with Royal Jelly for Topical Use. Gels 2025; 11:294. [PMID: 40277730 PMCID: PMC12027417 DOI: 10.3390/gels11040294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2025] [Revised: 04/11/2025] [Accepted: 04/14/2025] [Indexed: 04/26/2025] Open
Abstract
For decades, royal jelly achieved notoriety and became an ultra-rich ingredient with numerous pharmacological properties especially for its use in production of topical ointments and creams. A novel formulation enriched with 2% royal jelly has been developed and characterized. Rheological results highlight a gel-like behavior of the product in the packaging, as it does not flow from the costumer's hand after application and behaves like a liquid, spreading evenly onto clean skin. A clear comparison in size distribution of pure and cream samples was noticed by dynamic light scattering analysis and completed further by Fourier transform infrared spectroscopy (FTIR-ATR) which showed off shift changes in the gel sample as compared to pure compounds. MTT assays were conducted in quintuplicate on murine fibroblasts cell line (NCTC L-929) for testing the biocompatibility of the product in the range of 50-1000 μg/mL over 24, 48 and 72 h. The designed formulation is typically intended to deliver active compounds to the skin surface and potentially into deeper layers. A molecular docking study was performed for binding mode prediction of P-gp protein residues with two ligands, quercetin and myricetin, in order to investigate their role in the internal modulation of drug transport across cell membranes within the skin.
Collapse
Affiliation(s)
- Monica-Elisabeta Maxim
- Romanian Academy, Ilie Murgulescu—Institute of Physical Chemistry, 202 Splaiul Independentei, 060021 Bucharest, Romania; (M.-E.M.); (L.A.); (A.-R.L.); (A.P.)
| | - Raluca-Marieta Toma
- Romanian Academy, Ilie Murgulescu—Institute of Physical Chemistry, 202 Splaiul Independentei, 060021 Bucharest, Romania; (M.-E.M.); (L.A.); (A.-R.L.); (A.P.)
| | - Ludmila Aricov
- Romanian Academy, Ilie Murgulescu—Institute of Physical Chemistry, 202 Splaiul Independentei, 060021 Bucharest, Romania; (M.-E.M.); (L.A.); (A.-R.L.); (A.P.)
| | - Anca-Ruxandra Leonties
- Romanian Academy, Ilie Murgulescu—Institute of Physical Chemistry, 202 Splaiul Independentei, 060021 Bucharest, Romania; (M.-E.M.); (L.A.); (A.-R.L.); (A.P.)
| | - Aurica Precupas
- Romanian Academy, Ilie Murgulescu—Institute of Physical Chemistry, 202 Splaiul Independentei, 060021 Bucharest, Romania; (M.-E.M.); (L.A.); (A.-R.L.); (A.P.)
| | - Rodica Tatia
- National Institute of Research and Development for Biological Sciences, 296 Splaiul Independentei, 060031 Bucharest, Romania; (R.T.); (E.I.O.)
| | - Elena Iulia Oprita
- National Institute of Research and Development for Biological Sciences, 296 Splaiul Independentei, 060031 Bucharest, Romania; (R.T.); (E.I.O.)
| |
Collapse
|
6
|
Ai W, Yang C, Wang Q, Han W, Wang Y, Hou J, Zhu Z, Zhao J, Zhang Y, Zhang Y, Li X. Stable Silicon Anodes Enabled by Innovative Biobased Binder with Cross-Linking Network in Lithium-Ion Batteries. LANGMUIR : THE ACS JOURNAL OF SURFACES AND COLLOIDS 2025; 41:9469-9479. [PMID: 40177947 DOI: 10.1021/acs.langmuir.5c00314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
Graphite anodes, with their capacity nearing the theoretical maximum of 372 mA h g-1, are increasingly being complemented by silicon-based materials, which offer a 10-fold higher capacity. Nevertheless, extreme volume expansion (>300%) of Si during cycling poses significant challenges to its practical deployment. Previous studies on the synthesis of binders are often intricate and not conducive to large-scale implementation. In this study, an innovative binder, denoted as HM, is developed by combining the macromolecular polysaccharide sodium hyaluronate with the small organic molecule malic acid without the need for any external triggers. A cross-linked network structure is formed in situ after heat treatment with silicon under vacuum conditions. The contact interface establishes a robust network structure through multiple macromolecular hydrogen bonds and chemical interactions. Consequently, the HM binder exhibits exceptional mechanical properties and efficiently lessens the volumetric change of silicon particles, thereby benefiting the generation of a stable solid electrolyte interphase. Electrochemical characterization demonstrates that the exceptional cycling stability of Si@HM electrodes can maintain a high capacity of 1949 mA h g-1 at 0.1 C and 1426 mA h g-1 at 0.5 C after 100 cycles. Furthermore, silicon anodes employing the HM binder demonstrate superior rate performance and reduced internal resistance compared to those of conventional binders, representing a significant advancement in performance. This research provides crucial perspectives for binder design and experimental evidence for the commercial utilization of silicon anodes within lithium-ion batteries.
Collapse
Affiliation(s)
- Wengxiang Ai
- National Local Joint Engineering Research Center for Lithium-ion Batteries and Materials Preparation Technology, Key Laboratory of Advanced Batteries Materials of Yunnan Province, Faculty of Metallurgical and Energy Engineering, Kunming University of Science and Technology, Kunming 650093, PR China
| | - Chunman Yang
- National Local Joint Engineering Research Center for Lithium-ion Batteries and Materials Preparation Technology, Key Laboratory of Advanced Batteries Materials of Yunnan Province, Faculty of Metallurgical and Energy Engineering, Kunming University of Science and Technology, Kunming 650093, PR China
| | - Qian Wang
- National Local Joint Engineering Research Center for Lithium-ion Batteries and Materials Preparation Technology, Key Laboratory of Advanced Batteries Materials of Yunnan Province, Faculty of Metallurgical and Energy Engineering, Kunming University of Science and Technology, Kunming 650093, PR China
| | - Wenchang Han
- National Local Joint Engineering Research Center for Lithium-ion Batteries and Materials Preparation Technology, Key Laboratory of Advanced Batteries Materials of Yunnan Province, Faculty of Metallurgical and Energy Engineering, Kunming University of Science and Technology, Kunming 650093, PR China
| | - Yongqi Wang
- National Local Joint Engineering Research Center for Lithium-ion Batteries and Materials Preparation Technology, Key Laboratory of Advanced Batteries Materials of Yunnan Province, Faculty of Metallurgical and Energy Engineering, Kunming University of Science and Technology, Kunming 650093, PR China
| | - Jiyue Hou
- National Local Joint Engineering Research Center for Lithium-ion Batteries and Materials Preparation Technology, Key Laboratory of Advanced Batteries Materials of Yunnan Province, Faculty of Metallurgical and Energy Engineering, Kunming University of Science and Technology, Kunming 650093, PR China
| | - Ziyi Zhu
- National Local Joint Engineering Research Center for Lithium-ion Batteries and Materials Preparation Technology, Key Laboratory of Advanced Batteries Materials of Yunnan Province, Faculty of Metallurgical and Energy Engineering, Kunming University of Science and Technology, Kunming 650093, PR China
| | - Jinbao Zhao
- State Key Laboratory of Physical Chemistry of Solid Surfaces, State-Province Joint Engineering Laboratory of Power Source Technology for New Energy Vehicle, Collaborative Innovation Center of Chemistry for Energy Materials, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
| | - Yiyong Zhang
- National Local Joint Engineering Research Center for Lithium-ion Batteries and Materials Preparation Technology, Key Laboratory of Advanced Batteries Materials of Yunnan Province, Faculty of Metallurgical and Energy Engineering, Kunming University of Science and Technology, Kunming 650093, PR China
| | - Yingjie Zhang
- National Local Joint Engineering Research Center for Lithium-ion Batteries and Materials Preparation Technology, Key Laboratory of Advanced Batteries Materials of Yunnan Province, Faculty of Metallurgical and Energy Engineering, Kunming University of Science and Technology, Kunming 650093, PR China
| | - Xue Li
- National Local Joint Engineering Research Center for Lithium-ion Batteries and Materials Preparation Technology, Key Laboratory of Advanced Batteries Materials of Yunnan Province, Faculty of Metallurgical and Energy Engineering, Kunming University of Science and Technology, Kunming 650093, PR China
| |
Collapse
|
7
|
Hu K, Xiao M, Chen S, Huang Y, Hou Z, Li X, Yang L. Innovative applications of natural polysaccharide polymers in intravesical therapy of bladder diseases. Carbohydr Polym 2025; 354:123307. [PMID: 39978897 DOI: 10.1016/j.carbpol.2025.123307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2024] [Revised: 01/17/2025] [Accepted: 01/20/2025] [Indexed: 02/22/2025]
Abstract
Natural polysaccharide polymers, characterized by their remarkable biocompatibility, biodegradability, and structural versatility, hold great promise for intravesical therapy in treating of bladder diseases. Conditions such as bladder cancer and interstitial cystitis compromise drug efficacy by affecting the permeability of the bladder wall. Traditional therapeutic approaches are often hindered by physiological challenges, including rapid drug clearance and the intrinsic permeability barrier of the bladder. Polysaccharides like hyaluronic acid (HA) and chitosan (CS) have emerged as promising materials for intravesical drug delivery systems (IDDS), owing to their ability to repair tight junctions in the bladder wall, mitigate inflammation, and enhance permeability. This review provides a comprehensive overview of the mechanisms through which polysaccharide-based natural polymers regulate bladder wall permeability and highlights their advancements in delivery platforms, including nanoparticles, hydrogels, floating systems, and composite materials. By improving drug retention, enhancing bioavailability, and promoting patient adherence, these materials offer a solid foundation for the development of innovative therapeutic strategies for bladder diseases.
Collapse
Affiliation(s)
- Ke Hu
- Research Center for Biomedical Materials, Shenyang Key Laboratory of Biomedical Polymers, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 11004, China; Department of Urology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Miaomiao Xiao
- Research Center for Biomedical Materials, Shenyang Key Laboratory of Biomedical Polymers, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 11004, China
| | - Siwen Chen
- Research Center for Biomedical Materials, Shenyang Key Laboratory of Biomedical Polymers, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 11004, China
| | - Yuanbing Huang
- Department of Urology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China
| | - Zhipeng Hou
- Research Center for Biomedical Materials, Shenyang Key Laboratory of Biomedical Polymers, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 11004, China.
| | - Xiancheng Li
- Department of Urology, The Second Affiliated Hospital of Dalian Medical University, Dalian, China.
| | - Liqun Yang
- Research Center for Biomedical Materials, Shenyang Key Laboratory of Biomedical Polymers, Engineering Research Center of Ministry of Education for Minimally Invasive Gastrointestinal Endoscopic Techniques, Shengjing Hospital of China Medical University, Shenyang 11004, China.
| |
Collapse
|
8
|
Joshi N, Yan J, Dang M, Slaughter K, Wang Y, Wu D, Ung T, Bhingaradiya N, Pandya V, Chen MX, Kaur S, Bhagchandani S, Alfassam HA, Joseph J, Gao J, Dewani M, Chu RWC, Yip RCS, Weldon E, Shah P, Pisal ND, Shukla C, Sherman NE, Luo JN, Conway T, Eickhoff JP, Botelho L, Alhasan AH, Karp JM, Ermann J. A mechanically resilient soft hydrogel improves drug delivery for treating post-traumatic osteoarthritis in physically active joints. Proc Natl Acad Sci U S A 2025; 122:e2409729122. [PMID: 40163719 PMCID: PMC12002200 DOI: 10.1073/pnas.2409729122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Accepted: 02/12/2025] [Indexed: 04/02/2025] Open
Abstract
Intra-articular delivery of disease-modifying osteoarthritis drugs (DMOADs) is likely to be most effective in the early stages of post-traumatic osteoarthritis (PTOA), when symptoms are minimal, and patients remain physically active. To ensure effective therapy, DMOAD delivery systems therefore must withstand repeated mechanical loading without altering the kinetics of drug release. While soft materials are typically preferred for DMOAD delivery, mechanical loading can compromise their structural integrity and disrupt controlled drug release. In this study, we present a mechanically resilient soft hydrogel that rapidly self-heals under conditions simulating human running while maintaining sustained release of the cathepsin-K inhibitor L-006235, used as a proof-of-concept DMOAD. This hydrogel demonstrated superior performance compared to a previously reported hydrogel designed for intra-articular drug delivery, which, in our study, neither recovered its structure nor maintained drug release under mechanical loading. When injected into mouse knee joints, the hydrogel provided consistent release kinetics of the encapsulated drug in both treadmill-running and nonrunning mice. In a mouse model of severe PTOA exacerbated by treadmill-running, the L-006235 hydrogel significantly reduced cartilage degeneration, whereas the free drug did not. Overall, our data underscore the hydrogel's potential for treating PTOA in physically active patients.
Collapse
Affiliation(s)
- Nitin Joshi
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
- Harvard Medical School, Boston, MA02115
| | - Jing Yan
- Harvard Medical School, Boston, MA02115
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Mickael Dang
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Kai Slaughter
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Yufeng Wang
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Dana Wu
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Trevor Ung
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Nutan Bhingaradiya
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
- Harvard Medical School, Boston, MA02115
| | - Virja Pandya
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Mu Xian Chen
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Shahdeep Kaur
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
- Harvard Medical School, Boston, MA02115
| | - Sachin Bhagchandani
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Haya A. Alfassam
- National Center for Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology, Riyadh11442, Saudi Arabia
- King Abdulaziz City for Science and Technology Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology, Riyadh11442, Saudi Arabia
| | - John Joseph
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
- Harvard Medical School, Boston, MA02115
| | - Jingjing Gao
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
- Harvard Medical School, Boston, MA02115
| | - Mahima Dewani
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Rachel Wai Chun Chu
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Ryan Chak Sang Yip
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Eli Weldon
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Purna Shah
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Nishkal Dhiraj Pisal
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Chetan Shukla
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - Nicholas E. Sherman
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | - James N. Luo
- Harvard Medical School, Boston, MA02115
- Department of Surgery, Brigham and Women’s Hospital, Boston, MA02115
| | - Thomas Conway
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
| | | | | | - Ali H. Alhasan
- National Center for Biotechnology, Life Science and Environment Research Institute, King Abdulaziz City for Science and Technology, Riyadh11442, Saudi Arabia
- King Abdulaziz City for Science and Technology Center of Excellence for Biomedicine, Joint Centers of Excellence Program, King Abdulaziz City for Science and Technology, Riyadh11442, Saudi Arabia
| | - Jeffrey M. Karp
- Center for Nanomedicine, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Boston, MA02115
- Harvard Medical School, Boston, MA02115
- Harvard–Massachusetts Institute of Technology Division of Health Sciences and Technology, Massachusetts Institute of Technology, Cambridge, MA02139
- Broad Institute, Cambridge, MA02142
- Harvard Stem Cell Institute, Cambridge, MA02138
| | - Joerg Ermann
- Harvard Medical School, Boston, MA02115
- Division of Rheumatology, Inflammation and Immunity, Department of Medicine, Brigham and Women’s Hospital, Boston, MA02115
| |
Collapse
|
9
|
Kumaran K, Vasudevan AK, Jayakumar R, Biswas R. Injectable vancomycin loaded hyaluronic acid-chitosan hydrogel for the treatment of Staphylococcus aureus septic arthritis. Carbohydr Res 2025; 550:109384. [PMID: 39826274 DOI: 10.1016/j.carres.2025.109384] [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: 09/11/2024] [Revised: 12/09/2024] [Accepted: 01/13/2025] [Indexed: 01/22/2025]
Abstract
Staphylococcus aureus (S. aureus) is a Gram positive opportunistic pathogen and a major cause for bacterial septic arthritis. Vancomycin is the preferred antibiotic for the treatment of methicillin resistance S. aureus septic arthritis. Patients undergoing vancomycin treatment need to be hospitalized and their serum vancomycin level should be monitored, as increase in vancomycin concentration in serum may lead to hepatotoxicity. To overcome vancomycin mediated cytotoxicity, we have prepared a local injectable delivery system by incorporating vancomycin into hyaluronic acid (HA)-chitosan (van-HA-chitosan) hydrogel. The prepared van-HA-chitosan hydrogel was characterized using Fourier Transform Infrared Spectroscopy (FTIR) and rheometer. The van-HA-chitosan hydrogel is injectable, has shear thinning behaviour; and is hemo- and cyto-compatible. In vitro drug release assay showed that 95 % of vancomycin was released from the hydrogel in 8 days. Under in vitro conditions the load of S. aureus decreased from 6.4 Log10 CFU/ml to 3.5 Log10 CFU/ml when treated with van-HA-chitosan hydrogel for 6 h. Significant decrease in bacterial counts was observed when S. aureus infected synovial fluid and bone samples were treated with van-HA-chitosan hydrogel. Our results suggest that the prepared van-HA-chitosan could be used for the treatment of septic arthritis caused by S. aureus.
Collapse
Affiliation(s)
- K Kumaran
- School of Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, 682041, India
| | - Anil Kumar Vasudevan
- Department of Microbiology, Amrita Institute of Medical Sciences and Research Centre (AIMS), Amrita Vishwa Vidyapeetham, Kochi, 682041, India
| | - R Jayakumar
- School of Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, 682041, India.
| | - Raja Biswas
- School of Nanosciences and Molecular Medicine, Amrita Vishwa Vidyapeetham, Kochi, 682041, India.
| |
Collapse
|
10
|
Rathore SS, Leno Jenita JJ, Dotherabandi M. A systematic review on hyaluronic acid coated nanoparticles: recent strategy in breast cancer management. JOURNAL OF BIOMATERIALS SCIENCE. POLYMER EDITION 2025; 36:605-646. [PMID: 39429014 DOI: 10.1080/09205063.2024.2416293] [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: 08/09/2024] [Accepted: 10/09/2024] [Indexed: 10/22/2024]
Abstract
Hyaluronic acid, a non-sulphated glycosaminoglycan has attracted its usage in the management of breast cancer. Drug-loaded nanoparticles with hyaluronic acid surface modifications show potential as a promising method for targeting and delivering drugs to the tumor site. The aim of this study was to conduct a systematic review of articles and assess the impact of hyaluronic acid coated nanoparticles on breast cancer. The various database were used for this comprehensive review. The inclusion and exclusion criteria were selected according to the PRISMA guidelines. Studies associated with characterization, in vitro, and in vivo studies were collected and subjected for further analysis. According to the inclusion criteria, 41 literature were selected for analysis. From all the studies, it was observed that the nanoparticles coated with hyaluronic acid produced better particle size, shape, zeta potential, increased in vitro cytotoxicity, cellular uptake, cell apoptosis, and anti-tumor effect in vivo. Research has shown that hyaluronic acid exhibits a higher affinity for CD44 receptors, resulting in enhanced targeted nanoparticle activity on cancer cells while sparing normal cells.
Collapse
Affiliation(s)
- Seema S Rathore
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Dayananda Sagar University, Bangalore, India
| | - J Josephine Leno Jenita
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Dayananda Sagar University, Bangalore, India
| | - Manjula Dotherabandi
- Department of Pharmaceutics, College of Pharmaceutical Sciences, Dayananda Sagar University, Bangalore, India
| |
Collapse
|
11
|
Sherman SL, Gudeman AS, Kelly JD, Dimeff RJ, Farr J. Mechanisms of Action of Intra-articular Hyaluronic Acid Injections for Knee Osteoarthritis: A Targeted Review of the Literature. Am J Sports Med 2025:3635465241302820. [PMID: 40108507 DOI: 10.1177/03635465241302820] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
BACKGROUND The knee is the most commonly afflicted joint in osteoarthritis (OA). Injection of intra-articular of hyaluronic acid (IAHA) is a frequently used therapy for the management of knee OA with varying product characteristics. PURPOSE To describe and characterize the mechanism of action (MoA) of IAHA products concerning nociception, chondroprotection, and anti-inflammatory properties via a targeted literature review. STUDY DESIGN Systematic review; Level of evidence, 2. METHODS We followed the standard methodologies for conducting and reporting targeted reviews as recommended by the Cochrane Handbook for Systematic Reviews of Interventions, adapted for conducting a targeted literature review. Relevant studies were identified by searching the Embase database using predefined search strategies via the Ovid platform. The results of the review were reported according to the PRISMA guidelines (Preferred Reporting Items for Systematic Reviews and Meta-analyses). RESULTS A total of 182 studies were included in this targeted literature review. Of these, 107 reported chondroprotective action, 59 anti-inflammatory activity, 18 analgesic properties, 30 proteoglycan or glycosaminoglycan synthesis, 8 subchondral bone effects, 2 mechanical effects, and 1 other effects of IAHA. These MoAs were studied through diverse types of studies: in vitro biochemistry, animal physiological studies, or human physiological and clinical studies. The chondroprotective effect was the most studied MoA and showed an increase in anabolic biomarkers, such as collagen types II, IX, and XI, and a reduction in catabolic biomarkers, such as matrix metalloproteinases, which play a primary role in the downstream signaling pathways in OA and cartilage degradation in the synovial fluid. IAHA was widely reported by studies to reduce soluble inflammatory mediators, such as interleukins 1β and 6 and tumor necrosis factor α, thereby decreasing the production of degradative enzymes (eg, matrix metalloproteinases, aggrecanases). IAHA was also reported to enhance the synthesis of intrinsic proteoglycan (eg, aggrecan) and glycosaminoglycans, thus delaying the progression of OA. IAHA also reported improvement in the mechanical function of the knee by increasing the viscosity of the synovial fluid, reducing the coefficient of friction, and improving its lubrication. Overall, a significant decrease in knee pain was observed after IAHA treatments. CONCLUSION Preclinical and clinical studies established evidence for varied MoAs by which IAHA preparations may produce a desired effect in patients with knee OA.
Collapse
Affiliation(s)
| | - Andrew S Gudeman
- School of Medicine, Indiana University, Indianapolis, Indiana, USA
| | - John D Kelly
- University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | | | | |
Collapse
|
12
|
Woo I, Park JJ, Park CH. Dual intra-articular injections of corticosteroid and hyaluronic acid versus single corticosteroid injection for ankle osteoarthritis: a randomized comparative trial. BMC Musculoskelet Disord 2025; 26:239. [PMID: 40069683 PMCID: PMC11895146 DOI: 10.1186/s12891-025-08488-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2024] [Accepted: 03/03/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND Intra-articular corticosteroid injection is commonly used for pain relief in ankle osteoarthritis (OA). The effects of corticosteroids (CS) are short-lived, whereas hyaluronic acid (HA) have longer-lasting effects. The objective was to compare the efficacy of dual injections of CS and HA to CS alone. We hypothesized that intra-articular injections of dual agents would be more effective than CS alone. METHODS A single-blind, randomized, controlled trial was designed to investigate this hypothesis. 135 patients with ankle OA were enrolled into an intra-articular CS injection group (CS group, n = 61) or dual HA plus CS injection group (CS + HA group, n = 74). The CS group received 1 mL of corticosteroid and 1 mL of 0.5% bupivacaine and 1 mL of normal saline once, and the CS + HA group received 3 mL of a total of 5 mL mixtures containing 2 mL of HA, or 1 mL of corticosteroid, 0.5% bupivacaine, and normal saline in the first week, followed by 2 mL of HA in the second and third weeks. Clinical evaluations were performed before injection, 6 and 12 weeks after the first injections. The Ankle Osteoarthritis Scale (AOS) was used as the primary outcome measure, and the Visual Analogue Scale (VAS), Short Form Health Survey (SF-36), and complications were used as secondary outcomes. RESULTS The mean AOS change from baseline was significantly greater in the CS + HA group than in the CS group at 6 (p ≤ 0.01) and 12 weeks (p ≤ 0.01). The mean VAS change from baseline was significantly greater in the CS group than in the CS + HA group at 6 weeks (p = 0.023), but not at 12 weeks (p = 0.731). The mean SF-36 change from baseline was not significant between the CS and CS + HA groups at 6 (p = 0.416) and 12 weeks (p = 0.215). CONCLUSIONS The combination of corticosteroid and HA injection is more effective than corticosteroid alone in relieving pain in ankle OA. TRIAL REGISTRATION Clinical Research Information Service in South Korea, KCT0008690 // Registration Date (First Posted): July 21th, 2023 ( http://cris.nih.go.kr ).
Collapse
Affiliation(s)
- Inha Woo
- The Armed Forces Daejeon Hospital, Daejeon, Republic of Korea
| | - Jeong-Jin Park
- Korea Armed Forces Athletic Corps, Mungyeong, Gyeongsangbuk-do Province, Republic of Korea
| | - Chul Hyun Park
- Department of Orthopaedic Surgery, College of Medicine, Yeungnam University, 170 Hyeonchung-ro, Nam-gu, Daegu, 42415, Republic of Korea.
| |
Collapse
|
13
|
Wang X, Xue Y, Hao K, Peng B, Chen H, Liu H, Wang J, Cao J, Dong W, Zhang S, Yang Q, Li J, Lei W, Feng Y. Sustained therapeutic effects of self-assembled hyaluronic acid nanoparticles loaded with α-Ketoglutarate in various osteoarthritis stages. Biomaterials 2025; 314:122845. [PMID: 39326362 DOI: 10.1016/j.biomaterials.2024.122845] [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: 06/03/2024] [Revised: 09/14/2024] [Accepted: 09/16/2024] [Indexed: 09/28/2024]
Abstract
Osteoarthritis (OA) is a prevalent degenerative disease characterized by irreversible destruction of articular cartilage, for which no current drugs are known to modify its progression. While intra-articular (IA) injections of hyaluronic acid (HA) offer temporary relief, their effectiveness and long-term benefits are debated. Alpha-ketoglutarate (αKG) has potential chondroprotective properties, but its use is limited by a short half-life and poor cartilage-targeting efficiency. Here, we developed self-assembled HA-αKG nanoparticles (NPs) to combine the benefits of both HA and αKG, showing stability, bioavailability, and sustained pH-responsive release in the knee joint. In both early and advanced OA stages in mice, HA, αKG, and HA-αKG NPs could relieve pain, enhance mobility, and reduce cartilage damage, with HA-αKG NPs demonstrating the best efficacy. Mechanistically, αKG not only promotes cartilage matrix synthesis but also inhibits degradation by activating the PERK-ATF4 signaling pathway to reduce endoplasmic reticulum stress (ERS) in chondrocytes. This study highlights the therapeutic potential of HA-αKG NPs for treating various OA stages, with efficient and sustained effects, suggesting rapid clinical adoption and high acceptability among clinicians and patients.
Collapse
Affiliation(s)
- Xinli Wang
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Yufei Xue
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, Xi'an, 710072, China
| | - Kaili Hao
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Bo Peng
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, Xi'an, 710072, China
| | - Hongli Chen
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Hui Liu
- Frontiers Science Center for Flexible Electronics, Xi'an Institute of Flexible Electronics (IFE), Xi'an Institute of Biomedical Materials & Engineering (IBME), Northwestern Polytechnical University, Xi'an, 710072, China
| | - Jing Wang
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Jiahao Cao
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Wengang Dong
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China; Department of Emergency Surgery, Shaanxi Provincial People's Hospital, Xi'an, 710068, China
| | - Siqi Zhang
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Qian Yang
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China
| | - Jia Li
- Key Laboratory of Aerospace Medicine of the Ministry of Education, School of Aerospace Medicine, Fourth Military Medical University, Xi'an, 710000, China; Key Lab of Hazard Assessment and Control in Special Operational Environment, Ministry of Education, Fourth Military Medical University, Xi'an, 710000, China; Department of Health Statistics, School of Public Health, Fourth Military Medical University, Xi'an, 710000, China.
| | - Wei Lei
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China.
| | - Yafei Feng
- Department of Orthopedics, Xijing Hospital, The Fourth Military Medical University, Xi'an, 710032, China.
| |
Collapse
|
14
|
Saadh MJ, Ahmed HH, Kareem RA, Bishoyi AK, Roopashree R, Shit D, Arya R, Joshi KK, Sameer HN, Yaseen A, Athab ZH, Adil M, Narmani A, Farhood B. Recent advances of hyaluronic acid-based materials in drug delivery systems and regenerative medicine: A review. Arch Pharm (Weinheim) 2025; 358:e2400903. [PMID: 40091562 DOI: 10.1002/ardp.202400903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/31/2025] [Accepted: 02/18/2025] [Indexed: 03/19/2025]
Abstract
Nowadays, diseases have a high rate of incidence and mortality worldwide. On the other side, the drawbacks of conventional modalities in the suppression of diseases have encountered serious problematic issues for the health of human beings. For instance, although various approaches have been applied for the treatment of cancer, it has an ever-increasing rate of incidence and mortality throughout the globe. Thus, there is a fundamental requirement for the development of breakthrough technologies in the inhibition of diseases. Hyaluronic acid (HA) is one of the most practical biopolymers in the suppression of diseases. HA has lots of potential physicochemical (like rheological, structural, molecular weight, and ionization, etc.) and biomedical properties (bioavailability, biocompatibility, CD44 targeting and signaling pathways, components of biological organs, mucoadhesion, immunomodulation, etc.), which made it a potential candidate for the development of breakthrough tools in pharmaceutical and biomedical sciences. The ease of surface modification (carboxylation, amidation, hydroxylation, and esterification), high bioavailability and synthesis routes, and various administration routes are considered as other merits of HA-based vehicles. These mucopolysaccharide HA-based materials have been considerably developed for use in drug delivery systems (DDSs), cancer therapy, wound healing, antiaging, and tissue engineering. This review summarizes the advantages of HA-based DDS and scaffolds in the treatment of diseases.
Collapse
Affiliation(s)
| | | | | | - Ashok Kumar Bishoyi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot, Gujarat, India
| | - R Roopashree
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Debasish Shit
- Centre for Research Impact & Outcome, Chitkara University Institute of Engineering and Technology, Chitkara University, Rajpura, Punjab, India
| | - Renu Arya
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, India
| | - Kamal Kant Joshi
- Department of Allied Science, Graphic Era Hill University, Dehradun, Uttarakhand, India
- Graphic Era Deemed to be University, Dehradun, Uttarakhand, India
| | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar, Iraq
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Mohaned Adil
- Pharmacy college, Al-Farahidi University, Baghdad, Iraq
| | - Asghar Narmani
- Department of Medical Nanotechnology, School of Advanced Technologies in Medicine, Tehran University of Medical Sciences, Tehran, Iran
| | - Bagher Farhood
- Department of Medical Physics and Radiology, Faculty of Paramedical Sciences, Kashan University of Medical Sciences, Kashan, Iran
| |
Collapse
|
15
|
Glinkowski WM, Tomaszewski W. Intra-Articular Hyaluronic Acid for Knee Osteoarthritis: A Systematic Umbrella Review. J Clin Med 2025; 14:1272. [PMID: 40004802 PMCID: PMC11856182 DOI: 10.3390/jcm14041272] [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: 12/10/2024] [Revised: 02/06/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
Objective: to evaluate the efficacy, safety, and cost-effectiveness of intra-articular hyaluronic acid (IAHA) in treating osteoarthritis (OA), considering innovations in formulations, comparative outcomes, and variability in guidelines. This review aims to synthesize evidence supporting the role of IAHA in multimodal treatment strategies. Materials and Methods: A general, narrative, umbrella review of systematic reviews and meta-analyses was conducted. Clinical practice recommendations and guidelines for IAHA use were also reviewed and evaluated. A comprehensive search was conducted across the main medical data sources. Inclusion criteria focused on studies evaluating the efficacy, safety, and impact of IAHA. Key outcomes included pain reduction (e.g., WOMAC, VAS), functional improvement, safety, and cost-effectiveness. Results: IAHA showed moderate efficacy in pain relief and functional improvement, especially in early-to-moderate OA. The results indicate that hybrid formulations and combination therapies show better clinical outcomes, with expanded efficacy and potential chondroprotection. However, heterogeneity between studies was noted, reflecting variability in patient populations and intervention protocols. International guidelines varied significantly, with some opposing routine use (e.g., AAOS, NICE) and others endorsing IAHA more or less conditionally (e.g., ESCEO, OARSI). Conclusions: IAHA remains a treatment modality in the arsenal of selected populations of people with OA, especially for early and moderate disease. High-quality, standardized studies are still needed to refine IAHA's role and establish personalized guidelines for individual patients. A concerted effort to harmonize global recommendations and economic strategies, such as tiered pricing, can increase equitable access and optimize IAHA's integration of multimodal treatment for OA.
Collapse
Affiliation(s)
- Wojciech Michał Glinkowski
- Center of Excellence “TeleOrto” for Telediagnostics and Treatment of Disorders and Injuries of the Locomotor System, Department of Medical Informatics and Telemedicine, Medical University of Warsaw, 02-091 Warsaw, Poland
- Stichting Med Partners, 1098 XH Amsterdam, The Netherlands
| | - Wiesław Tomaszewski
- Ars Medica Foundation for Medical Education, Health Promotion, Art and Culture, 03-301 Warsaw, Poland
- College of Physiotherapy, 50-038 Wrocław, Poland
| |
Collapse
|
16
|
Li CS, Xu Y, Li J, Qin SH, Huang SW, Chen XM, Luo Y, Gao CT, Xiao JH. Ultramodern natural and synthetic polymer hydrogel scaffolds for articular cartilage repair and regeneration. Biomed Eng Online 2025; 24:13. [PMID: 39920742 PMCID: PMC11804105 DOI: 10.1186/s12938-025-01342-3] [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: 10/07/2024] [Accepted: 01/20/2025] [Indexed: 02/09/2025] Open
Abstract
Articular cartilage injury is a serious bone disease that can result in disabilities. With the rapid increase in the aging population, this disorder has become an increasingly important public health issue. Recently, stem cell-based cartilage tissue engineering has emerged as a promising therapeutic option for treating articular cartilage damage. Cellular scaffolds, which are among three key elements of tissue engineering, play significant roles in the repair of damaged articular cartilage by regulating cellular responses and promoting cartilage tissue regeneration. Biological macromolecules are commonly used as scaffold materials owing to their unique properties. For example, natural and synthetic polymer hydrogel scaffolds can effectively mimic the microenvironment of the natural extracellular matrix; exhibit high cytocompatibility, biocompatibility, and biodegradability; and have attracted increasing attention in bone and cartilage tissue engineering and regeneration medicine. Several types of hydrogel scaffolds have been fabricated to treat articular cartilage abnormalities. This article outlines the recent progress in the field of hydrogel scaffolds manufactured from various biomaterials for repairing damaged articular cartilage, discusses their advantages and disadvantages, and proposes directions for their future development.
Collapse
Affiliation(s)
- Chun-Sheng Li
- Institute of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, 563003, China
| | - Yan Xu
- Institute of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, 563003, China
- Guizhou Provincial Key Laboratory of Medicinal Biotechnology and Research Center for Translational Medicine in Colleges and Universities, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, 563003, China
| | - Juan Li
- National Engineering Research Center for Compounding and Modification of Polymer Materials, Guizhou Material Industrial Technology Research Institute, Guiyang, 550014, China
| | - Shu-Hao Qin
- Institute of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, 563003, China.
- National Engineering Research Center for Compounding and Modification of Polymer Materials, Guizhou Material Industrial Technology Research Institute, Guiyang, 550014, China.
| | - Shao-Wen Huang
- National Engineering Research Center for Compounding and Modification of Polymer Materials, Guizhou Material Industrial Technology Research Institute, Guiyang, 550014, China
| | - Xue-Mei Chen
- Institute of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, 563003, China
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, 563003, China
| | - Yi Luo
- Institute of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, 563003, China
- Guizhou Provincial Key Laboratory of Medicinal Biotechnology and Research Center for Translational Medicine in Colleges and Universities, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, 563003, China
| | - Cheng-Tao Gao
- National Engineering Research Center for Compounding and Modification of Polymer Materials, Guizhou Material Industrial Technology Research Institute, Guiyang, 550014, China
| | - Jian-Hui Xiao
- Institute of Medicinal Biotechnology, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, 563003, China.
- Guizhou Provincial Key Laboratory of Medicinal Biotechnology and Research Center for Translational Medicine in Colleges and Universities, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, 563003, China.
- Department of Pediatrics, Affiliated Hospital of Zunyi Medical University, 149 Dalian Road, Huichuan District, Zunyi, 563003, China.
| |
Collapse
|
17
|
Ariyoshi W, Takeuchi J, Mitsugi S, Koga A, Nagai-Yoshioka Y, Yamasaki R. Mechanisms Underlying the Stimulation of DUSP10/MKP5 Expression in Chondrocytes by High Molecular Weight Hyaluronic Acid. Biomedicines 2025; 13:376. [PMID: 40002789 PMCID: PMC11852791 DOI: 10.3390/biomedicines13020376] [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: 12/29/2024] [Revised: 01/25/2025] [Accepted: 02/03/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Previously, we reported that high molecular weight hyaluronic acid (HMW-HA) exerts chondroprotective effects by enhancing dual specificity protein phosphatase 10/mitogen-activated protein kinase (MAPK) phosphatase 5 (DUSP10/MKP5) expression and suppressing inflammatory cytokine-induced matrix metalloproteinase-13 (MMP13) expression in a human immortalized chondrocyte line (C28/I2 cells) via inhibition of MAPKs. The aim of this study was to elucidate the molecular mechanisms underlying the enhancement of DUSP10/MKP5 expression by HMW-HA in C28/I2 cells. Methods: C28/I2 cells were treated with HMW-HA, and the activation of intracellular signaling molecules was determined using Western blot analysis. The expression levels of mRNAs and microRNAs (miRNAs) were evaluated through real-time quantitative reverse transcription PCR analysis. Results: HMW-HA treatment induced Akt phosphorylation via interaction with CD44, and pretreatment with specific inhibitors of phosphatidylinositol-3 kinase/protein kinase B (PI3K/Akt) signaling attenuated the HMW-HA-induced expression of DUSP10/MKP5. HMW-HA suppressed the expression of miR-92a, miR-181a, and miR-181d. Loss-of-function and gain-of-function analyses of these miRNAs indicate that miR-92a, miR-181a, and miR-181d negatively regulate DUSP10/MKP5 expression. Moreover, HMW-HA-induced Akt phosphorylation was partially suppressed by miR-181a and miR-181d mimics. Finally, we found that HMW-HA activates RhoA-associated protein kinase (ROK) signaling, which contributes to Akt phosphorylation. Conclusions: These findings suggest that the induction of DUSP10/MKP5 expression by HMW-HA binding to CD44, leading to MMP13 suppression, involves multiple regulatory mechanisms, including PI3K/Akt and RhoA-activated ROK signaling, in addition to miRNA-mediated regulation. Elucidating these detailed molecular mechanisms may reveal novel biological activities that contribute to the therapeutic efficacy of HMW-HA against osteoarthritis.
Collapse
Affiliation(s)
- Wataru Ariyoshi
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Fukuoka 803-8580, Japan; (A.K.); (Y.N.-Y.); (R.Y.)
| | - Jun Takeuchi
- Medical Affairs, Seikagaku Corporation, Tokyo 100-0005, Japan;
| | - Sho Mitsugi
- Second Department of Oral and Maxillofacial Surgery, Osaka Dental University, Osaka 540-0008, Japan;
| | - Ayaka Koga
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Fukuoka 803-8580, Japan; (A.K.); (Y.N.-Y.); (R.Y.)
- School of Oral Health Sciences, Faculty of Dentistry, Kyushu Dental University, Fukuoka 803-8580, Japan
| | - Yoshie Nagai-Yoshioka
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Fukuoka 803-8580, Japan; (A.K.); (Y.N.-Y.); (R.Y.)
| | - Ryota Yamasaki
- Division of Infections and Molecular Biology, Department of Health Promotion, Kyushu Dental University, Fukuoka 803-8580, Japan; (A.K.); (Y.N.-Y.); (R.Y.)
| |
Collapse
|
18
|
Hou H, Liu X, Liu J, Wang Y. Carbohydrate polymer-based nanoparticles with cell membrane camouflage for cancer therapy: A review. Int J Biol Macromol 2025; 289:138620. [PMID: 39674458 DOI: 10.1016/j.ijbiomac.2024.138620] [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: 10/08/2024] [Revised: 11/21/2024] [Accepted: 12/08/2024] [Indexed: 12/16/2024]
Abstract
Recent developments in biomimetic nanoparticles, specifically carbohydrate polymer-coated cell membrane nanoparticles, have demonstrated considerable promise in treating cancer. These systems improve drug delivery by imitating natural cell actions, enhancing biocompatibility, and decreasing immune clearance. Conventional drug delivery methods frequently face challenges with non-specific dispersal and immune detection, which can hinder their efficiency and safety. These biomimetic nanoparticles improve target specificity, retention times, and therapeutic efficiency by using biological components like chitosan, hyaluronic acid, and alginate. Chitosan-based nanoparticles, which come from polysaccharides found in nature, have self-assembly abilities that make them better drug carriers. Hyaluronic acid helps target tissues more effectively, especially in cancer environments where there are high levels of hyaluronic acid receptors. Alginate-based systems also enhance drug delivery by being biocompatible and degradable, making them ideal choices for advanced therapeutic uses. Moreover, these particles hold potential for overcoming resistance to multiple drugs and boosting the body's immune reaction to tumors through precise delivery and decreased side effects of chemotherapy drugs. This review delves into the possibilities of using carbohydrate polymer-functionalized nanoparticles and their impact on enhancing the efficacy of cancer treatment.
Collapse
Affiliation(s)
- Haijia Hou
- Department of Respiratory and Critical Care Medicine, The First Affiliated Hospital of China Medical University, Shenyang, China
| | - Xuejian Liu
- Department of Pulmonary and Critical Care Medicine, Shengjing Hospital of China Medical University, Shenyang, China
| | - Jun Liu
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, China.
| | - Yudong Wang
- Department of Thoracic Surgery, Shengjing Hospital of China Medical University, Shenyang, China.
| |
Collapse
|
19
|
Biswas KB, Kawai Y, Nakagawa S, Kanai K, Kojima H, Masutani T, Oyama M, Iddamalgoda A, Sakamoto K. Artemisia capillaris with two novel active compounds, Kawarayomogin I and II, inhibits HYBID (KIAA1199) expression as well as hyaluronic acid degradation. Sci Rep 2025; 15:2042. [PMID: 39820064 PMCID: PMC11739587 DOI: 10.1038/s41598-025-86320-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2024] [Accepted: 01/09/2025] [Indexed: 01/19/2025] Open
Abstract
Hyaluronic acid (HA) is an important component of the skin's extracellular matrix, and its degradation leads to wrinkles. Hyaluronan-binding protein involved in hyaluronan depolymerization (HYBID) is the main factor responsible for HA degradation in dermis. This study aimed to identify natural plant materials that can effectively suppress HYBID expression and protect HA from degradation. Screening of various plant extracts was performed for the inhibition of histamine-induced mRNA expression of HYBID in normal human dermal fibroblasts (NHDF). The molecular size distribution of HA was evaluated by incubating fluorescein isothiocyanate (FITC)-labeled large HA (1200-1600 kDa) in NHDF for certain time followed by measuring different sizes of FITC-labeled HA in the cultured medium by HPLC. Among 380 plant extracts, we found that Artemisia capillaris flower extract (ACFE) was the most effective agent in both suppressing HYBID expression as well as protecting large HA from degradation. Subsequent mechanism elucidation studies showed that ACFE epigenetically regulates the expression of HYBID by modulating the expression of a specific miRNA, miR-486-5p, which is known to directly target and inhibit HYBID expression. Our active compound search identified 1-caffeoyl-3-hydroxybutane and 3-caffeoyl-1-hydroxybutane in ACFE as new compounds, which we named Kawarayomogin I and Kawarayomogin II, respectively. This is the first report to show that Artemisia capillaris with two novel active compounds inhibits HYBID expression as well as hyaluronic acid degradation, and therefore, could be used as possible agent for cosmeceutical potential.
Collapse
Affiliation(s)
- Kazal Boron Biswas
- Department of Research and Development, Ichimaru Pharcos Co. Ltd., Motosu, Gifu, Japan.
| | - Yuka Kawai
- Department of Research and Development, Ichimaru Pharcos Co. Ltd., Motosu, Gifu, Japan
| | - Satoshi Nakagawa
- Department of Research and Development, Ichimaru Pharcos Co. Ltd., Motosu, Gifu, Japan
| | - Kyoko Kanai
- Department of Research and Development, Ichimaru Pharcos Co. Ltd., Motosu, Gifu, Japan
| | - Hiroyuki Kojima
- Department of Research and Development, Ichimaru Pharcos Co. Ltd., Motosu, Gifu, Japan
| | - Teruaki Masutani
- Department of Research and Development, Ichimaru Pharcos Co. Ltd., Motosu, Gifu, Japan
| | - Masayoshi Oyama
- Laboratory of Pharmacognosy, Gifu Pharmaceutical University, Gifu, Japan
| | - Arunasiri Iddamalgoda
- Department of Research and Development, Ichimaru Pharcos Co. Ltd., Motosu, Gifu, Japan
| | - Kotaro Sakamoto
- Department of Research and Development, Ichimaru Pharcos Co. Ltd., Motosu, Gifu, Japan.
| |
Collapse
|
20
|
Heller S, Seemann RJ, Burgkart R, Obermeier A, Locher H. Lidocaine Shows Significant Antimicrobial Effects Against Staphylococcus Species: An In-Vitro Study Comparing Different Combinations of Lidocaine and Clinically Used Injectables, like Steroids and Hyaluronan, in the Context of Arthritis Management. Biomedicines 2025; 13:106. [PMID: 39857690 PMCID: PMC11762494 DOI: 10.3390/biomedicines13010106] [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: 12/09/2024] [Revised: 12/31/2024] [Accepted: 01/03/2025] [Indexed: 01/27/2025] Open
Abstract
INTRODUCTION Intra-articular injections, commonly used in osteoarthritis treatment, are debated due to their potential link to septic arthritis, though its incidence remains low. Lidocaine, used as a "carrier" for therapeutic substances like hyaluronan or triamcinolone, has pain-relieving and antimicrobial properties. This study investigates the concentration-dependent antimicrobial effects of lidocaine in combination with hyaluronan and triamcinolone in both standard and synovial fluid cultures. METHODS The antimicrobial efficacy of lidocaine against Staphylococcus aureus was investigated, with variations in bacterial and lidocaine concentrations. Bacterial growth was monitored using a UV/VIS spectrometer at 600 nm. Lidocaine solutions of 1% and 2% were tested, both alone and in combination with hyaluronic acid or Triam40, in tryptic soy broth (TSB), to reflect knee joint applications. The groups included pure lidocaine (L), Triam (T), hyaluronan (H), and combinations (LT, LH, TH, LTH) with 1% or 2% lidocaine. A bacterial inoculum of 300 CFU/mL was used, and samples were incubated for 12 and 24 h. Additional tests were conducted on Staphylococcus epidermidis and methicillin-resistant Staphylococcus aureus (MRSA), as well as on S. aureus in human synovial fluid. RESULTS Lidocaine showed a concentration-dependent antimicrobial effect, with greater inhibition at higher concentrations and lower bacterial densities. All lidocaine-containing combinations significantly reduced the bacterial levels of S. aureus in TSB. Similar results were seen for S. epidermidis and MRSA, with notable inhibition in synovial fluid after 12 h, especially with 2% lidocaine. CONCLUSIONS Lidocaine exhibits dose-dependent antimicrobial effects against key pathogens responsible for septic arthritis. Its combination with Triam40 and hyaluronan may reduce the risk of septic arthritis, supporting its clinical relevance.
Collapse
Affiliation(s)
- Stephan Heller
- Department of Orthopaedics and Sports Orthopaedics, TUM School of Medicine and Health, TUM Universitätsklinikum Klinikum Rechts der Isar, 81675 Munich, Germany
| | - Ricarda Johanna Seemann
- Centre for Orthopaedics and Specialized Pain Therapy, 88069 Tettnang, Germany
- Charité–Universitätsmedizin Berlin, Corporate Member of Frei Universität Berlin und Humboldt-Universität zu Berlin, Centre for Musculoskeletal Surgery, 10117 Berlin, Germany
| | - Rainer Burgkart
- Department of Orthopaedics and Sports Orthopaedics, TUM School of Medicine and Health, TUM Universitätsklinikum Klinikum Rechts der Isar, 81675 Munich, Germany
| | - Andreas Obermeier
- Department of Orthopaedics and Sports Orthopaedics, TUM School of Medicine and Health, TUM Universitätsklinikum Klinikum Rechts der Isar, 81675 Munich, Germany
| | - Hermann Locher
- Department of Orthopaedics and Sports Orthopaedics, TUM School of Medicine and Health, TUM Universitätsklinikum Klinikum Rechts der Isar, 81675 Munich, Germany
- Centre for Orthopaedics and Specialized Pain Therapy, 88069 Tettnang, Germany
| |
Collapse
|
21
|
Sanati M, Manavi MA, Noruzi M, Behmadi H, Akbari T, Jalali S, Sharifzadeh M, Khoobi M. Carbohydrates and neurotrophic factors: A promising partnership for spinal cord injury rehabilitation. BIOMATERIALS ADVANCES 2025; 166:214054. [PMID: 39332344 DOI: 10.1016/j.bioadv.2024.214054] [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: 07/11/2024] [Revised: 09/19/2024] [Accepted: 09/21/2024] [Indexed: 09/29/2024]
Abstract
Spinal cord injury (SCI) leaves a temporary or enduring motor, sensory, and autonomic function loss, significantly impacting the patient's quality of life. Given their biocompatibility, bioactivity, and tunable attributes, three-dimensional scaffolds frequently employ carbohydrates to facilitate spinal cord regeneration. These scaffolds have also been engineered to be novel local delivery platforms that present distinct advantages in the targeted transportation of drug candidates to the damaged spinal cord, ensuring the right dosage and duration of administration. Neurotrophic factors have emerged as promising therapeutic candidates, preserved neuron survival and encouraged severed axons repair, although their local and continuous delivery is believed to produce considerable spinal cord rehabilitation. This study aims to discuss breakthroughs in scaffold engineering, exploiting carbohydrates as an essential part of their structure, and highlight their impact on spinal cord regeneration and sustained neurotrophic factors delivery to treat SCI.
Collapse
Affiliation(s)
- Mehdi Sanati
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Birjand University of Medical Sciences, Birjand, Iran
| | - Mohammad Amin Manavi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Marzieh Noruzi
- School of Pharmacy, Semnan University of Medical Sciences, Semnan, Iran
| | - Homayoon Behmadi
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Tayebeh Akbari
- Department of Microbiology, Islamic Azad University, North Tehran Branch, Tehran, Iran
| | - Sara Jalali
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
| | - Mohammad Sharifzadeh
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran.
| | - Mehdi Khoobi
- Department of Radiopharmacy, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran; Drug Design and Development Research Center, The Institute of Pharmaceutical Sciences (TIPS), Tehran University of Medical Sciences, 1417614411 Tehran, Iran.
| |
Collapse
|
22
|
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
|
23
|
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]
|
24
|
Wójcik-Pastuszka D, Iwaszkiewicz R, Musiał W. The Effects of Synthetic Polymers on the Release Patterns of Bupivacaine Hydrochloride from Sodium Hyaluronate Hydrogels. Biomedicines 2024; 13:39. [PMID: 39857623 PMCID: PMC11760862 DOI: 10.3390/biomedicines13010039] [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: 11/05/2024] [Revised: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 01/27/2025] Open
Abstract
BACKGROUND Using hydrogels for the controlled release of drugs is beneficial for patients, who then receive the proper dose of the medicinal substance. In addition, the formulation can provide more consistent drug absorption while reducing the frequency of dosing. OBJECTIVES The aim of this investigation is to propose a novel HA (sodium hyaluronate)-based hydrogel for intra-articular injection doped with synthetic polymers and incorporated with bupivacaine hydrochloride (Bu) as a local anesthetic. The other aim of this study is to reveal the effects of the formulation's ingredients on its viscosity and the relationship between the hydrogel's viscosity and drug release. METHODS First, HA-based hydrogels doped with synthetic polymers and incorporated with Bu were prepared. A study of the hydrogels' viscosities was performed using a rotational viscometer. Release tests were carried out by employing a paddle-over-disk apparatus following the USP/Ph.Eur guidelines. The drug concentrations in the acceptor fluid were analyzed spectrophotometrically. RESULTS It was found that the viscosity of the hydrogels doped with synthetic polymers was higher than the viscosity of the hydrogels made with only HA. The viscosity of the hydrogels doped with AX (ammonium acryloyldimethyltaurate/VP copolymer) was the highest, measuring 6750 ± 160 cP and 12623 ± 379 cP with and without Bu, respectively. The results of the kinetic experiment indicate that the Higuchi and Korsmeyer-Peppas models best described the drug release. Bu was released the most slowly from the formulation doped with AX. The release rate constants obtained from the Higuchi and Korsmeyer-Peppas models were kH = 4.4 ± 0.2 mg × min-1/2 and kK-P = 3.4 ± 0.2 × 10-2 min-N, respectively. The half-release time, calculated using the Higuchi model, was the longest for the formulation doped with AX, at 199.5 ± 17.6 min. CONCLUSIONS This indicates that the incorporation of AX into the hydrogel may prolong the drug dissolution. The hydrogel doped with AX was the optimal formulation for the controlled release of Bu.
Collapse
Affiliation(s)
| | | | - Witold Musiał
- Department of Physical Chemistry and Biophysics, Faculty of Pharmacy, Wroclaw Medical University, ul. Borowska 211A, 55-556 Wrocław, Poland; (D.W.-P.); (R.I.)
| |
Collapse
|
25
|
Kanayama T, Nakase J, Yoshioka K, Takata Y, Ishida Y, Yanatori Y, Takemoto N, Demura S. Histological evaluation of medial collateral ligament bursa injection, a new conservative treatment, in a rabbit model of medial meniscus horizontal tear. J Orthop Sci 2024:S0949-2658(24)00267-7. [PMID: 39694739 DOI: 10.1016/j.jos.2024.11.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Revised: 11/09/2024] [Accepted: 11/22/2024] [Indexed: 12/20/2024]
Abstract
BACKGROUND The ultrasound-guided medial collateral ligament bursa injection technique is safe, reproducible, and effective in treating symptomatic degenerative medial meniscal tears. However, the mechanisms of action and optimal drug combinations remain unclear. This study aimed to evaluate and compare the histological changes caused by injections of corticosteroids and hyaluronic acid into the medial collateral ligament bursa in a rabbit model of medial meniscus horizontal tears. Furthermore, we compared neovascularization and neurogenesis between different treatments. METHODS The medial meniscus horizontal tear rabbit model was created. Medial collateral ligament bursa injection with triamcinolone acetonide, purified sodium hyaluronate, saline, or needle alone was performed after model creation. The area of the medial collateral ligament bursa was measured. Immunostaining validation (cluster of differentiation31, smooth muscle alpha-actin, calcitonin gene-related peptide, and 4',6-diamidino-2-phenylindole) of the medial collateral ligament bursa injections was performed 2 and 4 weeks after injection. RESULTS The group injected with triamcinolone acetonide had a smaller area of the medial collateral ligament bursa than did the other groups. The groups injected with triamcinolone acetonide and purified sodium hyaluronate had lower neovascularization levels than did the other groups. The calcitonin gene-related peptide count was lower in the group injected with triamcinolone acetonide than in the other groups. Corticosteroid and hyaluronic acid injections into the medial collateral ligament bursa suppressed neovascularization and calcitonin gene-related peptide expression, while steroid injections caused adipose tissue and synovial tissue atrophy. CONCLUSIONS The present study revealed that ultrasound-guided triamcinolone or hyaluronic injections into the medial collateral ligament bursa inhibited neovascularization within it and concomitantly reduced calcitonin gene-related peptide release from neurogenesis. Hence, medial collateral ligament bursa injection should be considered a new treatment option for symptomatic horizontal meniscal tears.
Collapse
Affiliation(s)
- Tomoyuki Kanayama
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Junsuke Nakase
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan.
| | - Kazuaki Yoshioka
- Department of Physiology, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Ishikawa, Japan
| | - Yasushi Takata
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yoshihiro Ishida
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Yusuke Yanatori
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Naoki Takemoto
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| | - Satoru Demura
- Department of Orthopedic Surgery, Graduate School of Medical Sciences, Kanazawa University, Kanazawa, Japan
| |
Collapse
|
26
|
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
|
27
|
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
|
28
|
Banigo AT, Konings IBM, Nauta L, Zoetebier B, Karperien M. Synthesis and Engineering of Hyaluronic Acid-Gelatin Hydrogels with Improved Cellular Attachment and Growth. Polymers (Basel) 2024; 16:3410. [PMID: 39684154 DOI: 10.3390/polym16233410] [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: 10/17/2024] [Revised: 11/22/2024] [Accepted: 11/27/2024] [Indexed: 12/18/2024] Open
Abstract
Injectable hydrogels are promising materials for cartilage regeneration in tissue engineering due to their tunable crosslinking rates, mechanical properties, and biodegradation profiles. This study investigates the chondrogenic potential of hyaluronic acid (HA) hydrogels crosslinked via tyramine (TA) moieties, with and without gelatin modified with TA (Gel-TA). Incorporating Gel-TA improved cell viability, spreading, and cartilage matrix deposition, particularly in medium and high molecular weight (MMW and HMW) HA-TA/Gel-TA hydrogels. Although the hydrogels' molecular weight did not significantly alter stiffness, MMW and HMW HA-TA/Gel-TA formulations exhibited enhanced functional properties such as slower degradation and superior cartilage matrix deposition. These attributes, coupled with Gel-TA's effects, underscore the importance of both molecular weight and biofunctional components in hydrogel design for cartilage regeneration. While low molecular weight (LMW) HA-TA hydrogels offered excellent injectability and supported high cell viability, they degraded rapidly and exhibited reduced cartilage matrix formation. Gel-TA enhanced cell adhesion and spreading by providing integrin-binding sites and promoted collagen type II deposition, crucial for cartilage regeneration. Moreover, the increased stiffness of MMW and HMW HA-TA/Gel-TA hydrogels facilitated extracellular matrix production. These findings show the potential of Gel-TA-modified HA-TA hydrogels for cartilage tissue engineering, with the opportunity for further optimization through the incorporation of bioactive components.
Collapse
Affiliation(s)
- Alma Tamunonengiofori Banigo
- Department of Developmental BioEngineering, Faculty of Science and Technology and TechMed Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands
| | - Irene B M Konings
- Department of Developmental BioEngineering, Faculty of Science and Technology and TechMed Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands
| | - Laura Nauta
- Department of Developmental BioEngineering, Faculty of Science and Technology and TechMed Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands
| | - Bram Zoetebier
- Department of Developmental BioEngineering, Faculty of Science and Technology and TechMed Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands
| | - Marcel Karperien
- Department of Developmental BioEngineering, Faculty of Science and Technology and TechMed Centre, University of Twente, Drienerlolaan 5, 7522 NB Enschede, The Netherlands
| |
Collapse
|
29
|
Carvalho DN, Gonçalves C, Sousa RO, Reis RL, Oliveira JM, Silva TH. Extraction and Purification of Biopolymers from Marine Origin Sources Envisaging Their Use for Biotechnological Applications. MARINE BIOTECHNOLOGY (NEW YORK, N.Y.) 2024; 26:1079-1119. [PMID: 39254780 PMCID: PMC11541305 DOI: 10.1007/s10126-024-10361-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2023] [Accepted: 08/19/2024] [Indexed: 09/11/2024]
Abstract
Biopolymers are a versatile and diverse class of materials that has won high interest due to their potential application in several sectors of the economy, such as cosmetics, medical materials/devices, and food additives. In the last years, the search for these compounds has explored a wider range of marine organisms that have proven to be a great alternative to mammal sources for these applications and benefit from their biological properties, such as low antigenicity, biocompatibility, and biodegradability, among others. Furthermore, to ensure the sustainable exploitation of natural marine resources and address the challenges of 3R's policies, there is a current necessity to valorize the residues and by-products obtained from food processing to benefit both economic and environmental interests. Many extraction methodologies have received significant attention for the obtention of diverse polysaccharides, proteins, and glycosaminoglycans to accomplish the increasing demands for these products. The present review gives emphasis to the ones that can be obtained from marine biological resources, as agar/agarose, alginate and sulfated polysaccharides from seaweeds, chitin/chitosan from crustaceans from crustaceans, collagen, and some glycosaminoglycans such as chondroitin sulfate and hyaluronic acids from fish. It is offered, in a summarized and easy-to-interpret arrangement, the most well-established extraction and purification methodologies used for obtaining the referred marine biopolymers, their chemical structure, as well as the characterization tools that are required to validate the extracted material and respective features. As supplementary material, a practical guide with the step-by-step isolation protocol, together with the various materials, reagents, and equipment, needed for each extraction is also delivered is also delivered. Finally, some remarks are made on the needs still observed, despite all the past efforts, to improve the current extraction and purification procedures to achieve more efficient and green methodologies with higher yields, less time-consuming, and decreased batch-to-batch variability.
Collapse
Affiliation(s)
- Duarte Nuno Carvalho
- 3B´S Research Group, I3B´s - Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark 4805-017, Barco, Guimarães, Portugal
- ICVS/3B´s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Cristiana Gonçalves
- 3B´S Research Group, I3B´s - Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark 4805-017, Barco, Guimarães, Portugal
- ICVS/3B´s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rita O Sousa
- 3B´S Research Group, I3B´s - Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark 4805-017, Barco, Guimarães, Portugal
- ICVS/3B´s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Rui L Reis
- 3B´S Research Group, I3B´s - Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark 4805-017, Barco, Guimarães, Portugal
- ICVS/3B´s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - J Miguel Oliveira
- 3B´S Research Group, I3B´s - Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark 4805-017, Barco, Guimarães, Portugal
- ICVS/3B´s - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Tiago H Silva
- 3B´S Research Group, I3B´s - Research Institute on Biomaterials, Biodegradables and Biomimetics of University of Minho, Headquarters of the European Institute of Excellence On Tissue Engineering and Regenerative Medicine, AvePark 4805-017, Barco, Guimarães, Portugal.
- ICVS/3B´s - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
30
|
Mansur H, Maranho DA, de Castro Junior IM, Gomes FF. May the Symptomatic Subtalar Joint Be Conservatively Treated With Intra-Articular Hyaluronic Acid Injections After a Calcaneus Fracture? Foot Ankle Spec 2024; 17:537-544. [PMID: 35125018 DOI: 10.1177/19386400211068256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND Subtalar pain following intra-articular calcaneus fractures may be associated with disability, pain, and a negative impact on the quality of life. Salvage procedures as subtalar fusion are associated with further consequences as stiffness, altered ankle biomechanics, and adjacent articular overloading with degenerative changes. The objective of the present study is to evaluate the short-term effects of viscosupplementation with intra-articular hyaluronic acid (HA) on function and pain, in patients with painful subtalar joint after calcaneus fracture. METHODS We searched for patients who underwent osteosynthesis of intra-articular calcaneus fracture between January 2011 and July 2015 and were diagnosed during the follow-up with pain and subtalar osteoarthritis. Between January and December of 2018, 13 patients (50 ± 10 years) accepted to participate in this study and received intra-articular HA injections. Three consecutive doses of 20 mg of HA were administered within a week interval, through anterolateral injections into the subtalar joint. We prospectively evaluated the function using the ankle/hindfoot American Orthopaedic Foot & Ankle Society score (AOFAS) and level of pain using the visual analog scale (VAS) before the intervention and 4, 12, and 24 weeks after the first injection. RESULTS Hindfoot function improved with an increase of AOFAS from 55 ± 19 before the intervention to 88 ± 20 at the 24th week (P = .001). Similarly, we observed relief of pain during the 24 weeks following intra-articular hyaluronic acid injection, with a decrease in VAS from 8.3 ± 1.3 before treatment to 2.2 ± 3.0 at the 24th week (P = .001). CONCLUSION For patients experiencing pain and dysfunction with subtalar osteoarthritis after intra-articular calcaneus fracture, viscosupplementation with intra-articular HA may be associated with improvement in function and pain in the short term. Furthermore, patients with higher grades of osteoarthritis may have limited benefit in pain relief and function improvement. LEVEL OF EVIDENCE IV, Case series.
Collapse
Affiliation(s)
- Henrique Mansur
- Department of Orthopedic Surgery, Hospital DF Star and Santa Helena, Brasília, Brazil
- Department of Foot and Ankle Surgery, National Institute of Traumatology and Orthopedics, Rio de Janeiro, Brazil
| | | | | | - Fernanda Ferreira Gomes
- Department of Foot and Ankle Surgery, National Institute of Traumatology and Orthopedics, Rio de Janeiro, Brazil
| |
Collapse
|
31
|
Zhang BK, Gines L. Analysis of Cancer-Resisting Evolutionary Adaptations in Wild Animals and Applications for Human Oncology. J Mol Evol 2024; 92:685-694. [PMID: 39256250 DOI: 10.1007/s00239-024-10204-w] [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: 06/07/2024] [Accepted: 08/28/2024] [Indexed: 09/12/2024]
Abstract
This literature review is to present a new direction in developing better treatment or preventive measures. The larger the body of an organism, the more numerous the cells, which theoretically lead to a higher risk of cancer. However, observational studies suggest the lack of correlation between body size and cancer risk, which is known as Peto's paradox. The corollary of Peto's paradox is that large organisms must be cancer-resistant. Further investigation of the anti-cancer mechanisms in each species could be potentially rewarding, and how the anti-cancer mechanisms found in wild animals can help influence and develop more effective cancer treatment in humans is the main focus of this literature review. Due to a lack of research and understanding of the exact molecular mechanisms of the researched species, only a few (Elephants and rodents) that have been extensively researched have made substantive contributions to human oncology. A new research direction is to investigate the positively selective genes that are related to cancer resistance and see if homologous genes are presented in humans. Despite the great obstacle of applying anti-cancer mechanisms to the human body from phylogenetically distant species, this research direction of gaining insights through investigating cancer-resisting evolutionary adaptations in wild animals has great potential in human oncology research.
Collapse
Affiliation(s)
- Bokai K Zhang
- Brigham Young University, Brigham Young University, Provo, UT, 84602, USA.
| | - Leoned Gines
- Shoreline Community College, 16101 Greenwood Avenue North, Shoreline, WA, 98133-5696, USA
| |
Collapse
|
32
|
Vishwanath K, McClure SR, Bonassar LJ. Heterogeneous distribution of viscosupplements in vivo is correlated to ex vivo frictional properties of equine cartilage. J Biomed Mater Res A 2024; 112:2149-2159. [PMID: 38923105 DOI: 10.1002/jbm.a.37766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2024] [Revised: 06/03/2024] [Accepted: 06/08/2024] [Indexed: 06/28/2024]
Abstract
Intra-articular injections of hyaluronic acid (HA) are the cornerstone of osteoarthritis (OA) treatments. However, the mechanism of action and efficacy of HA viscosupplementation are debated. As such, there has been recent interest in developing synthetic viscosupplements. Recently, a synthetic 4 wt% polyacrylamide (pAAm) hydrogel was shown to effectively lubricate and bind to the surface of cartilage in vitro. However, its ability to localize to cartilage and alter the tribological properties of the tissue in a live articulating large animal joint is not known. The goal of this study was to quantify the distribution and extent of localization of pAAm in the equine metacarpophalangeal or metatarsophalangeal joint (fetlock joint), and determine whether preferential localization of pAAm influences the tribological properties of the tissue. An established planar fluorescence imaging technique was used to visualize and quantify the distribution of fluorescently labeled pAAm within the joint. While the pAAm hydrogel was present on all surfaces, it was not uniformly distributed, with more material present near the site of the injection. The lubricating ability of the cartilage in the joint was then assessed using a custom tribometer across two orders of magnitude of sliding speed in healthy synovial fluid. Cartilage regions with a greater coverage of pAAm, that is, higher fluorescent intensities, exhibited friction coefficients nearly 2-fold lower than regions with lesser pAAm (Rrm = -0.59, p < 0.001). Collectively, the findings from this study indicate that intra-articular viscosupplement injections are not evenly distributed inside a joint, and the tribological outcomes of these materials is strongly determined by the ability of the material to localize to the articulating surfaces in the joint.
Collapse
Affiliation(s)
- Karan Vishwanath
- Department of Materials Science and Engineering, Cornell University, Ithaca, New York, USA
| | | | - Lawrence J Bonassar
- Meinig School of Biomedical Engineering, Cornell University, Ithaca, New York, USA
- Sibley School of Mechanical and Aerospace Engineering, Cornell University, Ithaca, New York, USA
| |
Collapse
|
33
|
Wu D, Yang S, Gong Z, Zhu X, Hong J, Wang H, Xu W, Lai J, Wang X, Lu J, Fang X, Jiang G, Zhu J. Enhanced therapeutic potential of a self-healing hyaluronic acid hydrogel for early intervention in osteoarthritis. Mater Today Bio 2024; 29:101353. [PMID: 39687801 PMCID: PMC11647215 DOI: 10.1016/j.mtbio.2024.101353] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 11/09/2024] [Accepted: 11/18/2024] [Indexed: 12/18/2024] Open
Abstract
Osteoarthritis (OA) is characterized by symptoms such as abnormal lubrication function of synovial fluid and heightened friction on the cartilage surface in its early stages, prior to evident cartilage damage. Current early intervention strategies employing lubricated hydrogels to shield cartilage from friction often overlook the significance of hydrogel-cartilage adhesion and enhancement of the cartilage extracellular matrix (ECM). Herein, we constructed a hydrogel based on dihydrazide-modified hyaluronic acid (HA) (AHA) and catechol-conjugated aldehyde-modified HA (CHA), which not only adheres to the cartilage surface as an effective lubricant but also improves the extracellular environment of chondrocytes in OA. Material characterization experiments on AHA/CHA hydrogels with varying concentrations validated their exceptional self-healing capabilities, superior injectability and viscoelasticity, sustained adhesion strength to cartilage, and a low friction coefficient. Chondrocytes exhibited robust adhesion and proliferation on the AHA/CHA hydrogel surface, with the upregulation of cartilage matrix protein expression. Intra-articular injection of AHA/CHA hydrogels was performed following destabilization of the medial meniscus (DMM) surgery in mice to assess its protective effect on cartilage. The AHA/CHA hydrogel effectively attenuated the degree of cartilage wear, facilitated chondrocytes' anabolic metabolism, and restored the ECM of cartilage. Therefore, the AHA/CHA hydrogel emerges as a promising therapeutic approach in clinical practices of OA treatment.
Collapse
Affiliation(s)
- Dongze Wu
- Department of Spinal Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, 315000, Zhejiang, China
| | - Shuhui Yang
- School of Materials Science and Engineering, Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, Zhejiang, China
| | - Zhe Gong
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine & Zhejiang Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases, Hangzhou, 310016, Zhejiang, China
| | - Xinxin Zhu
- Stomatology Hospital, School of Stomatology, Zhejiang University School of Medicine, Zhejiang Provincial Clinical Research Center for Oral Diseases, Key Laboratory of Oral Biomedical Research of Zhejiang Province, Cancer Center of Zhejiang University, Engineering Research Center of Oral Biomaterials and Devices of Zhejiang Province, Hangzhou, 310000, Zhejiang, China
| | - Juncong Hong
- Department of Anesthesiology, The First People's Hospital of Linping District, Hangzhou, 311100, Zhejiang, China
| | - Haitao Wang
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine & Zhejiang Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases, Hangzhou, 310016, Zhejiang, China
| | - Wenbin Xu
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine & Zhejiang Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases, Hangzhou, 310016, Zhejiang, China
| | - Juncheng Lai
- School of Materials Science and Engineering, Zhejiang-Mauritius Joint Research Center for Biomaterials and Tissue Engineering, Zhejiang Sci-Tech University, Hangzhou, 310018, Zhejiang, China
| | - Xiumei Wang
- State Key Laboratory of New Ceramics and Fine Processing, Key Laboratory of Advanced Materials, School of Materials Science and Engineering, Tsinghua University, Beijing, 100084, China
| | - Jiye Lu
- Department of Spinal Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, 315000, Zhejiang, China
| | - Xiangqian Fang
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine & Zhejiang Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases, Hangzhou, 310016, Zhejiang, China
| | - Guoqiang Jiang
- Department of Spinal Surgery, The First Affiliated Hospital of Ningbo University, Ningbo, 315000, Zhejiang, China
| | - Jinjin Zhu
- Department of Orthopedic Surgery, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine & Zhejiang Key Laboratory of Mechanism Research and Precision Repair of Orthopaedic Trauma and Aging Diseases, Hangzhou, 310016, Zhejiang, China
| |
Collapse
|
34
|
Iaconisi GN, Ahmed A, Lauria G, Gallo N, Fiermonte G, Cowman MK, Capobianco L, Dolce V. Targeting mitochondria in Cancer therapy: Machine learning analysis of hyaluronic acid-based drug delivery systems. Int J Biol Macromol 2024; 283:137840. [PMID: 39566768 DOI: 10.1016/j.ijbiomac.2024.137840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 10/30/2024] [Accepted: 11/17/2024] [Indexed: 11/22/2024]
Abstract
BACKGROUND Mitochondrial alterations play a crucial role in the development and progression of cancer. Dysfunctional mitochondria contribute to the acquisition of key hallmarks of cancer, including sustained proliferative signaling, evasion of growth suppressors, and resistance to cell death. Consequently, targeting mitochondrial dysfunction has emerged as a promising therapeutic strategy. Hyaluronic acid (HA), a naturally occurring glycosaminoglycan, has garnered significant attention due to its multifaceted roles in cancer biology. METHODS We employed a Systematic Literature Review (SLR) approach to examine a collection of 90 scientific publications using a text mining technique leveraging the Latent Dirichlet Allocation (LDA) algorithm. RESULTS The result of this activity, performed through the MySLR digital platform, allowed us to identify a set of two distinct topics representing the research domain. Specifically, Topic 1 comprised 41 papers, while Topic 2 comprised 49 papers. CONCLUSIONS The computational analysis highlighted that the integration of HA into drug delivery systems represents a promising approach to enhance the effectiveness and safety of cancer therapies. The discussed clinical trials provided compelling evidence of the potential of HA-based treatments in targeting cancer cells while minimizing adverse effects on healthy tissues.
Collapse
Affiliation(s)
- Giorgia Natalia Iaconisi
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy.
| | - Amer Ahmed
- Department of Bioscience, Biotechnology and Environment, University of Bari, 70125 Bari, Italy.
| | - Graziantonio Lauria
- Department of Pharmacy, Health, and Nutritional Sciences, University of Calabria, 87036, Arcavacata di Rende, Cosenza, Italy.
| | - Nunzia Gallo
- Department of Engineering for Innovation, University of Salento, Via Monteroni, 73100 Lecce, Italy.
| | - Giuseppe Fiermonte
- Department of Bioscience, Biotechnology and Environment, University of Bari, 70125 Bari, Italy.
| | - Mary K Cowman
- Department of Biomedical Engineering, Tandon School of Engineering, New York University, NY, New York, USA; Department of Orthopedic Surgery, Grossman School of Medicine, New York University, NY, New York, USA.
| | - Loredana Capobianco
- Department of Biological and Environmental Sciences and Technologies, University of Salento, 73100 Lecce, Italy.
| | - Vincenza Dolce
- Department of Pharmacy, Health, and Nutritional Sciences, University of Calabria, 87036, Arcavacata di Rende, Cosenza, Italy.
| |
Collapse
|
35
|
Bianchera A, Borghetti P, Ravanetti F, Bertocchi L, De Angelis E, Bettini R. Effect of Low-Molecular-Weight Hyaluronate-Based Nanoparticles on the In Vitro Expression of Cartilage Markers. Int J Mol Sci 2024; 25:12486. [PMID: 39684203 DOI: 10.3390/ijms252312486] [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: 10/15/2024] [Revised: 11/14/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
Hyaluronic acid (HA) is a key component of synovial fluid as it plays a crucial role in joint physiology. Its biological activity is influenced by molecular weight, local concentration, and persistence in joints. High-molecular-weight HA has a consolidated history of clinical use, whereas little is known about the metabolic effect of low-molecular-weight hyaluronate on cartilage differentiation. This study explores the potential of HA-based nanoparticles (NPs) on chondrocytes differentiation in vitro. Starting from 25 kDa and 250 kDa sodium hyaluronate solutions, two types of NPs were prepared by antisolvent precipitation in ethanol. The resulting NPs were dried in the presence of dipalmitoyl phosphatidylcholine, a natural synovial fluid component, then applied on an in vitro model of horse articular chondrocytes: no toxicity was observed and NPs prepared from 250 kDa HA promoted chondrocyte differentiation to a larger extent with respect to corresponding HA solutions, as evidenced by increased gene expression of chondrogenic markers (Col2a1 and Sox9) and reduced expression of dedifferentiation markers (Col1a1 and Runx2). These findings suggest that HA-based NPs are more effective at promoting the cellular internalization of the molecule and the differentiation of chondrocytes in vitro and could be a promising platform for drug delivery and cartilage repair.
Collapse
Affiliation(s)
- Annalisa Bianchera
- Food and Drug Department, University of Parma, Parco Area Delle Scienze 27/a, 43124 Parma, Italy
| | - Paolo Borghetti
- Department of Veterinary Science, University of Parma, 43124 Parma, Italy
| | | | - Laura Bertocchi
- Food and Drug Department, University of Parma, Parco Area Delle Scienze 27/a, 43124 Parma, Italy
| | - Elena De Angelis
- Department of Veterinary Science, University of Parma, 43124 Parma, Italy
| | - Ruggero Bettini
- Food and Drug Department, University of Parma, Parco Area Delle Scienze 27/a, 43124 Parma, Italy
| |
Collapse
|
36
|
Sharma Y, Bala K. Multifarious Aspect of Cytokines as an Immuno-Therapeutic for Various Diseases. J Interferon Cytokine Res 2024; 44:477-485. [PMID: 39394036 DOI: 10.1089/jir.2024.0090] [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] Open
Abstract
Cytokines are known to be a group of growing small proteins that are majorly responsible for the transmission of signals and communication between hematopoietic cells, the cells of the human immune system, and other types of cells. Cytokines play a dominant role in different types of disorders and in perpetuating the inflammation-related disorders. The production of cytokines is a natural process inside the body of a human being against any foreign invasion or due to some pathogenic state to maintain the homeostasis. Cytokines respond in two ways; in some cases, the production and development of cytokines as a therapeutic discovery or intervention will enhance the treatment process and support the reaction given by the body against any pathogenic activity, and in some cases, overproduction of these cytokines responds in the opposite way and behaves as antagonists toward a typical therapeutic drug and its treatment. Overall, 41 articles were reviewed, and it was found that cytokines have proved to be a therapeutic approach among various diseases and can be utilized as a good candidate or a better choice for cancer therapeutics in future development.
Collapse
Affiliation(s)
- Yash Sharma
- Department of Biotechnology, IILM University, Greater Noida, India
| | - Kumud Bala
- Department of Biotechnology, IILM University, Greater Noida, India
- Therapeutics and Molecular Diagnostic Lab, Centre for Medical Biotechnology, Amity Institute of Biotechnology, Amity University, Noida, India
| |
Collapse
|
37
|
Skierska I, Górski B, Fus Ł. Tunnel technique and subepithelial connective tissue graft, with or without cross-linked hyaluronic acid, in the treatment of multiple gingival recessions: 12-month outcomes of a randomized clinical trial. J Periodontol 2024; 95:1060-1072. [PMID: 38808976 DOI: 10.1002/jper.24-0093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2024] [Revised: 04/26/2024] [Accepted: 05/02/2024] [Indexed: 05/30/2024]
Abstract
BACKGROUND This study evaluated the influence of the adjunctive application of a cross-linked hyaluronic acid (HA) in the treatment of multiple gingival recessions, using a modified coronally advanced tunnel (MCAT) technique and subepithelial connective tissue graft (SCTG) (MCAT+SCTG±HA). METHODS A randomized, split-mouth, double-masked comparison of the effects of MCAT+HA+SCTG (test) versus MCAT+SCTG (control) in the treatment of multiple, contralateral gingival recessions with clinical, esthetic, and histological evaluations was carried out. All samples were stained with hematoxylin and eosin, Masson's trichrome, Verhoeff-Van Gieson, and Alcian blue stain for semiquantitative evaluation. The primary outcome variable was 12-month mean root coverage (MRC). RESULTS Twenty-four patients with 266 gingival recessions received both control and test treatments (133 recessions per group). 12-month MRC of the MCAT+HA+SCTG group was not significantly different from the MCAT+SCTG group with 84.32%± 34.46% and 85.71%± 36.43%, respectively (p = 0.991). Both treatment modes produced favorable esthetic outcomes (root coverage esthetic score [RES] 9.51± 1.01 tests vs. 9.26± 1.10 controls, p = 0.7292). However, the application of HA improved soft tissue texture (p = 0.0091). The remaining end point measures did not differ significantly between groups. Histological evaluation showed a significantly greater number of elastic fibers and a moderate increase in collagen fiber density in biopsy samples taken from the test sides when compared to the control sides (p = 0.0419 and p = 0.300, respectively). CONCLUSIONS MCAT+SCTG is an effective procedure in the treatment of multiple recession Type 1 (RT1) and RT2 recessions. There were no statistically significant differences in evaluated clinical treatment outcomes in the MCAT+HA+SCTG group compared to the MCAT+SCTG group within a period of 12 months. The application of HA increased collagen and elastic fiber density.
Collapse
Affiliation(s)
- Izabela Skierska
- Department of Periodontology and Oral Mucosa Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Bartłomiej Górski
- Department of Periodontology and Oral Mucosa Diseases, Medical University of Warsaw, Warsaw, Poland
| | - Łukasz Fus
- Department of Pathology, Medical University of Warsaw, Warsaw, Poland
| |
Collapse
|
38
|
Świerczyńska M, Kudzin MH, Chruściel JJ. Poly(lactide)-Based Materials Modified with Biomolecules: A Review. MATERIALS (BASEL, SWITZERLAND) 2024; 17:5184. [PMID: 39517460 PMCID: PMC11546716 DOI: 10.3390/ma17215184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024]
Abstract
Poly(lactic acid) (PLA) is characterized by unique features, e.g., it is environmentally friendly, biocompatible, has good thermomechanical properties, and is readily available and biodegradable. Due to the increasing pollution of the environment, PLA is a promising alternative that can potentially replace petroleum-derived polymers. Different biodegradable polymers have numerous biomedical applications and are used as packaging materials. Because the pure form of PLA is delicate, brittle, and is characterized by a slow degradation rate and a low thermal resistance and crystallization rate, these disadvantages limit the range of applications of this polymer. However, the properties of PLA can be improved by chemical or physical modification, e.g., with biomolecules. The subject of this review is the modification of PLA properties with three classes of biomolecules: polysaccharides, proteins, and nucleic acids. A quite extensive description of the most promising strategies leading to improvement of the bioactivity of PLA, through modification with these biomolecules, is presented in this review. Thus, this article deals mainly with a presentation of the major developments and research results concerning PLA-based materials modified with different biomolecules (described in the world literature during the last decades), with a focus on such methods as blending, copolymerization, or composites fabrication. The biomedical and unique biological applications of PLA-based materials, especially modified with polysaccharides and proteins, are reviewed, taking into account the growing interest and great practical potential of these new biodegradable biomaterials.
Collapse
Affiliation(s)
- Małgorzata Świerczyńska
- Łukasiewicz Research Network—Lodz Institute of Technology (ŁIT), 19/27 Marii Skłodowskiej-Curie Str., 90-570 Łódź, Poland; (M.Ś.); (M.H.K.)
- Circular Economy Center (BCG), Environmental Protection Engineering Research Group, Łukasiewicz Research Network—Lodz Institute of Technology (ŁIT), Brzezińska 5/15, 92-103 Łódź, Poland
- Institute of Polymer and Dye Technology, Faculty of Chemistry, Lodz University of Technology, Stefanowskiego 16, 90-537 Łódź, Poland
| | - Marcin H. Kudzin
- Łukasiewicz Research Network—Lodz Institute of Technology (ŁIT), 19/27 Marii Skłodowskiej-Curie Str., 90-570 Łódź, Poland; (M.Ś.); (M.H.K.)
- Circular Economy Center (BCG), Environmental Protection Engineering Research Group, Łukasiewicz Research Network—Lodz Institute of Technology (ŁIT), Brzezińska 5/15, 92-103 Łódź, Poland
| | - Jerzy J. Chruściel
- Łukasiewicz Research Network—Lodz Institute of Technology (ŁIT), 19/27 Marii Skłodowskiej-Curie Str., 90-570 Łódź, Poland; (M.Ś.); (M.H.K.)
- Circular Economy Center (BCG), Environmental Protection Engineering Research Group, Łukasiewicz Research Network—Lodz Institute of Technology (ŁIT), Brzezińska 5/15, 92-103 Łódź, Poland
| |
Collapse
|
39
|
Wang Z, Huang W, Jin S, Gao F, Sun T, He Y, Jiang X, Wang H. Hyaluronic acid/chitin thermosensitive hydrogel loaded with TGF-β1 promotes meniscus repair in rabbit meniscus full-thickness tear model. J Orthop Surg Res 2024; 19:683. [PMID: 39438973 PMCID: PMC11520169 DOI: 10.1186/s13018-024-05144-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 10/04/2024] [Indexed: 10/25/2024] Open
Abstract
Repair of the damaged meniscus is a scientific challenge owing to the poor self-healing potential of the white area of the meniscus. Tissue engineering provides a new method for the repair of meniscus injuries. In this study, we explored the superiority of 2% hyaluronic acid chitin hydrogel in temperature sensitivity, in vitro degradation, biocompatibility, cell adhesion, and other biological characteristics, and investigated the advantages of hyaluronic acid (HA) and Transforming Growth Factor β1 (TGF-β1) in promoting cell proliferation and a matrix formation phenotype. The hydrogel loaded with HA and TGF-β1 promoted cell proliferation. The HA + TGF-β1 mixed group showed the highest glycosaminoglycan (GAG) content and promoted cell migration. Hydroxypropyl chitin (HPCH), HA, and TGF-β1 were combined to form a composite hydrogel with a concentration of 2% after physical cross-linking, and this was injected into a rabbit model of a meniscus full-thickness tear. After 12 weeks of implantation, the TGF-β1 + HA/HPCH composite hydrogel was significantly better than HPCH, HA/HPCH, TGF-β1 + HPCH, and the control group in promoting meniscus repair. In addition, the new meniscus tissue of the TGF-β1 + HA/HPCH composite hydrogel had a tissue structure and biochemical content similar to that of the normal meniscus tissue.
Collapse
Affiliation(s)
- Ze Wang
- Department of Orthopedics, Wuhan Children's Hospital (Wuhan Maternal and Child Healthcare Hospital), Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430015, People's Republic of China
| | - Wei Huang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Shengyang Jin
- Department of Orthopedics, The First Affiliated Hospital of Soochow University, Suzhou, 215006, People's Republic of China
| | - Fei Gao
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Tingfang Sun
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Yu He
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China
| | - Xulin Jiang
- Key Laboratory of Biomedical Polymers of Ministry of Education and Department of Chemistry, Wuhan University, Wuhan, 430072, People's Republic of China.
| | - Hong Wang
- Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430022, People's Republic of China.
| |
Collapse
|
40
|
Di Cicco G, Marzano E, Mastrostefano A, Pitocco D, Castilho RS, Zambelli R, Mascio A, Greco T, Cinelli V, Comisi C, Maccauro G, Perisano C. The Pathogenetic Role of RANK/RANKL/OPG Signaling in Osteoarthritis and Related Targeted Therapies. Biomedicines 2024; 12:2292. [PMID: 39457605 PMCID: PMC11505501 DOI: 10.3390/biomedicines12102292] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2024] [Revised: 09/15/2024] [Accepted: 10/07/2024] [Indexed: 10/28/2024] Open
Abstract
Background: Osteoarthritis (OA) is the most common degenerative joint disease and affects millions of people worldwide, particularly the elderly population. The pathophysiology of OA is complex and involves multiple factors. Methods: Several studies have emphasized the crucial role of inflammation in this process. The receptor activator of NF-κB ligand (RANKL), the receptor activator of NF-κB (RANK), and osteoprotegerin (OPG) trigger a signaling cascade that leads to the excessive production of RANKL in the serum. Conclusions: The aim of this narrative review is (i) to assess the role of the RANK/RANKL/OPG signaling pathway in the context of OA progression, focusing especially on the physiopathology and on all the mechanisms leading to the activation of the inflammatory cascade, and (ii) to evaluate all the potential therapeutic strategies currently available that restore balance to bone formation and resorption, reducing structural abnormalities and relieving pain in patients with OA.
Collapse
Affiliation(s)
- Gabriele Di Cicco
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy; (G.D.C.)
| | - Emanuela Marzano
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy; (G.D.C.)
| | - Andrea Mastrostefano
- Department of Physiology and Pharmacology, Sapienza University of Rome, 00185 Rome, Italy; (G.D.C.)
| | - Dario Pitocco
- Diabetes Care Unit, Endocrinology, University Hospital “A. Gemelli”, Catholic University of the Sacred Heart, 00136 Rome, Italy
| | - Rodrigo Simões Castilho
- Department of Orthopaedics and Traumatology, Mater Dei Hospital, Belo Horizonte 30170-041, Brazil
| | - Roberto Zambelli
- Department of Orthopaedics and Traumatology, Mater Dei Hospital, Belo Horizonte 30170-041, Brazil
| | - Antonio Mascio
- Department of Orthopedics and Geriatric Sciences, Catholic University of the Sacred Heart, 00136 Rome, Italy
- Department of Orthopedics and Rheumatological Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00136 Rome, Italy
| | - Tommaso Greco
- Department of Orthopedics and Rheumatological Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00136 Rome, Italy
- Department of Life Sciences, Health, and Healthcare Professions, Link Campus University, 00165 Rome, Italy
| | - Virginia Cinelli
- Department of Orthopedics and Geriatric Sciences, Catholic University of the Sacred Heart, 00136 Rome, Italy
- Department of Orthopedics and Rheumatological Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00136 Rome, Italy
| | - Chiara Comisi
- Department of Orthopedics and Geriatric Sciences, Catholic University of the Sacred Heart, 00136 Rome, Italy
- Department of Orthopedics and Rheumatological Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00136 Rome, Italy
| | - Giulio Maccauro
- Department of Orthopedics and Geriatric Sciences, Catholic University of the Sacred Heart, 00136 Rome, Italy
- Department of Orthopedics and Rheumatological Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00136 Rome, Italy
| | - Carlo Perisano
- Department of Orthopedics and Geriatric Sciences, Catholic University of the Sacred Heart, 00136 Rome, Italy
- Department of Orthopedics and Rheumatological Sciences, Fondazione Policlinico Universitario A. Gemelli IRCCS, 00136 Rome, Italy
| |
Collapse
|
41
|
Guo J, Su K, Wang L, Feng B, You X, Deng M, Toh WS, Wu J, Cheng B, Xia J. Poly( p-coumaric acid) nanoparticles alleviate temporomandibular joint osteoarthritis by inhibiting chondrocyte ferroptosis. Bioact Mater 2024; 40:212-226. [PMID: 38973989 PMCID: PMC11224931 DOI: 10.1016/j.bioactmat.2024.06.007] [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: 03/28/2024] [Revised: 06/03/2024] [Accepted: 06/03/2024] [Indexed: 07/09/2024] Open
Abstract
Oxidative stress and inflammation are key drivers of osteoarthritis (OA) pathogenesis and disease progression. Herein we report the synthesis of poly(p-coumaric) nanoparticles (PCA NPs) from p-courmaic acid (p-CA), a naturally occurring phytophenolic acid, to be a multifunctional and drug-free therapeutic for temporomandibular joint osteoarthritis (TMJOA). Compared to hyaluronic acid (HA) that is clinically given as viscosupplementation, PCA NPs exhibited long-term efficacy, superior anti-oxidant and anti-inflammatory properties in alleviating TMJOA and repairing the TMJ cartilage and subchondral bone in a rat model of TMJOA. Notably, TMJ repair mediated by PCA NPs could be attributed to their anti-oxidant and anti-inflammatory properties in enhancing cell proliferation and matrix synthesis, while reducing inflammation, oxidative stress, matrix degradation, and chondrocyte ferroptosis. Overall, our study demonstrates a multifunctional nanoparticle, synthesized from natural p-coumaric acid, that is stable and possess potent antioxidant, anti-inflammatory properties and ferroptosis inhibition, beneficial for treatment of TMJOA.
Collapse
Affiliation(s)
- Jiaxin Guo
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, PR China
| | - Kai Su
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, PR China
| | - Liying Wang
- Department of Hematology, The Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, 518107, PR China
| | - Bingyu Feng
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, PR China
| | - Xinru You
- Center for Nanomedicine and Department of Anesthesiology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Miao Deng
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, PR China
| | - Wei Seong Toh
- Department of Orthopaedic Surgery, Yong Loo Lin School of Medicine, National University of Singapore, 119228, Singapore
- Faculty of Dentistry, National University of Singapore, 119085, Singapore
| | - Jun Wu
- Bioscience and Biomedical Engineering Thrust, The Hong Kong University of Science and Technology (Guangzhou), Nansha, Guangzhou, 511455, PR China
- Division of Life Science, The Hong Kong University of Science and Technology, Hong Kong, China
| | - Bin Cheng
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, PR China
| | - Juan Xia
- Hospital of Stomatology, Sun Yat-sen University, Guangzhou, 510055, PR China
- Guangdong Provincial Key Laboratory of Stomatology, Guangzhou, 510055, PR China
| |
Collapse
|
42
|
Senobari F, Abolmaali SS, Farahavr G, Tamaddon AM. Targeting inflammation with hyaluronic acid-based micro- and nanotechnology: A disease-oriented review. Int J Biol Macromol 2024; 280:135923. [PMID: 39322155 DOI: 10.1016/j.ijbiomac.2024.135923] [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: 12/03/2023] [Revised: 08/29/2024] [Accepted: 09/20/2024] [Indexed: 09/27/2024]
Abstract
Inflammation is a pivotal immune response in numerous diseases and presents therapeutic challenges. Traditional anti-inflammatory drugs and emerging cytokine inhibitors encounter obstacles such as limited bioavailability, poor tissue distribution, and adverse effects. Hyaluronic acid (HA), a versatile biopolymer, is widely employed to deliver therapeutic agents, including anti-inflammatory drugs, genes, and cell therapies owing to its unique properties, such as hydrophilicity, biodegradability, and safety. HA interacts with cell receptors to initiate processes such as angiogenesis, cell proliferation, and immune regulation. HA-based drug delivery systems offer dual strategies for effective inflammation management, capitalizing on passive and active mechanisms. This synergy permits the mitigation of inflammation by lowering the doses of anti-inflammatory drugs and their off-target adverse effects. A diverse array of micro- and nanotechnology techniques enable the fabrication of tailored HA-engineered systems, including hydrogels, microgels, nanogels, microneedles, nanofibers, and 3D-printed scaffolds, for diverse formulations and administration routes. This review explores recent insights into HA pharmacology in inflammatory conditions, material design, and fabrication methods, as well as its applications across a spectrum of inflammatory diseases, such as atherosclerosis, psoriasis, dermatitis, wound healing, rheumatoid arthritis, osteoarthritis, inflammatory bowel disease, and colitis, highlighting its potential for clinical translation.
Collapse
Affiliation(s)
- Fatemeh Senobari
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz 71345, Iran
| | - Samira Sadat Abolmaali
- Associate Professor, Pharmaceutical Nanotechnology Department and Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz 71345, Iran
| | - Ghazal Farahavr
- Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz 71345, Iran
| | - Ali Mohammad Tamaddon
- Professor, Pharmaceutics and Pharmaceutical Nanotechnology Department and Center for Nanotechnology in Drug Delivery, Shiraz University of Medical Sciences, Shiraz 71345, Iran.
| |
Collapse
|
43
|
Matalqah S, Lafi Z, Asha SY. Hyaluronic Acid in Nanopharmaceuticals: An Overview. Curr Issues Mol Biol 2024; 46:10444-10461. [PMID: 39329973 PMCID: PMC11431703 DOI: 10.3390/cimb46090621] [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: 08/22/2024] [Revised: 09/14/2024] [Accepted: 09/15/2024] [Indexed: 09/28/2024] Open
Abstract
Hyaluronic acid (HA) is a naturally occurring, long, unbranched polysaccharide that plays a critical role in maintaining skin structure and hydration. Its unique properties make it a valuable component in the field of nanopharmaceuticals. The combination of HA into nanopharmaceuticals enhances its ability to interact with various therapeutic agents, improving the delivery and efficacy of drugs. HA-based nanoparticles, including solid lipid nanoparticles, and polymeric nanogels, offer controlled release, enhanced stability, and targeted delivery of therapeutic agents. These innovations significantly improve therapeutic outcomes and reduce side effects, making HA an essential tool in modern medicine. In general, HA-modified liposomes enhance drug encapsulation and targeting, while HA-modified solid lipid nanoparticles (SLNs) provide a solid lipid core for drug encapsulation, offering controlled release and stability. This article provides an overview of the potential applications and recent advancements of HA in nanopharmaceuticals, emphasizing its significant impact on the evolving field of targeted drug delivery and advanced therapeutic strategies. By delving into the unique properties of HA and its compatibility with various therapeutic agents, this review underscores the promising potential of HA in revolutionizing nanopharmaceuticals.
Collapse
Affiliation(s)
- Sina Matalqah
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan
| | - Zainab Lafi
- Pharmacological and Diagnostic Research Center, Faculty of Pharmacy, Al-Ahliyya Amman University, Amman 19328, Jordan
| | | |
Collapse
|
44
|
Fragassi A, Greco A, Palomba R. Lubricant Strategies in Osteoarthritis Treatment: Transitioning from Natural Lubricants to Drug Delivery Particles with Lubricant Properties. J Xenobiot 2024; 14:1268-1292. [PMID: 39311151 PMCID: PMC11417909 DOI: 10.3390/jox14030072] [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: 07/18/2024] [Revised: 09/06/2024] [Accepted: 09/13/2024] [Indexed: 09/26/2024] Open
Abstract
Osteoarthritis (OA) is a debilitating joint disease characterized by cartilage degradation, leading to pain and functional impairment. A key contributor to OA progression is the decline in cartilage lubrication. In physiological conditions, synovial fluid (SF) macromolecules like hyaluronic acid (HA), phospholipids, and lubricin play a crucial role in the boundary lubrication of articular cartilage. In early OA, cartilage damage triggers inflammation, altering SF composition and compromising the lubrication layer. This increases friction between mating interfaces, worsening cartilage degradation and local inflammation. Therefore, early-stage restoration of lubrication (by injecting in the joint different classes of compounds and formulations) could alleviate, and potentially reverse, OA progression. In the light of this, a broad variety of lubricants have been investigated for their ability to reduce friction in OA joints and promote cartilage repair in clinical and preclinical studies. This review examines recent advancements in lubricant-based therapy for OA, focusing on natural, bioinspired, and alternative products. Starting from the currently applied therapy, mainly based on natural lubricants as HA, we will present their modified versions, either in hydrogel form or with specific biomimetic moieties with the aim of reducing their clearance from the joint and of enhancing their lubricating properties. Finally, the most advanced and recent formulation, represented by alternative strategies, will be proposed. Particular emphasis will be placed on those ones involving new types of hydrogels, microparticles, nanoparticles, and liposomes, which are currently under investigation in preclinical studies. The potential application of particles and liposomes could foster the transition from natural lubricants to Drug Delivery Systems (DDSs) with lubricant features; transition which could provide more complete OA treatments, by simultaneously providing lubrication replacement and sustained release of different payloads and active agents directly at the joint level. Within each category, we will examine relevant preclinical studies, highlighting challenges and future prospects.
Collapse
Affiliation(s)
- Agnese Fragassi
- Department of Pharmaceutical and Pharmacological Sciences, University of Padova, Via F. Marzolo 5, 35131 Padova, Italy
| | - Antonietta Greco
- Department of Medicine and Surgery, NanoMedicine Center (NANOMIB), University of Milano-Bicocca, Via Follereau 3, 20854 Vedano al Lambro, Italy
| | - Roberto Palomba
- Laboratory of Nanotechnology for Precision Medicine, Italian Institute of Technology, Via Morego 30, 16163 Genova, Italy
| |
Collapse
|
45
|
Wang J, Brugnoli B, Foglietta F, Andreana I, Longo G, Dinarelli S, Girasole M, Serpe L, Arpicco S, Francolini I, Di Meo C, Matricardi P. Tuning stiffness of hyaluronan-cholesterol nanogels by mussel-inspired dopamine-Fe 3+ coordination: Preparation and properties evaluation. Int J Biol Macromol 2024; 280:135553. [PMID: 39276885 DOI: 10.1016/j.ijbiomac.2024.135553] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 09/08/2024] [Accepted: 09/09/2024] [Indexed: 09/17/2024]
Abstract
In the evolving field of nanomedicine, tailoring the mechanical properties of nanogels to fine-tune their biological performance is a compelling avenue of research. This work investigates an innovative method for modulating the stiffness of hyaluronan-cholesterol (HACH) nanogels, an area that remains challenging. By grafting dopamine (DOPA) onto the HA backbone, characterized through UV, 1H NMR, and FT-IR analyses, we synthesized a novel polymer that spontaneously forms nanogels in aqueous environments. These HACH-DOPA nanogels are characterized by their small size (~170 nm), negative charge (around -32 mV), high stability, efficient drug encapsulation, and potent antioxidant activities (measured by ABTS test). Leveraging mussel-inspired metal coordination chemistry, the DOPA moieties enable stiffness modulation of the nanogels through catechol-Fe3+ interactions. This modification leads to increased crosslinking and, consequently, nanogels with a significantly increased stiffness, as measured by atomic force microscopy (AFM), with the formation of the HACH-DOPA@Fe3+ complex being pH-dependent and reversible. The cytocompatibility was evaluated via WST-1 cell proliferation assays on HUVEC and HDF cell lines, showing no evident cytotoxicity. Furthermore, the modified nanogels demonstrated enhanced cellular uptake, suggesting their substantial potential for intracellular drug delivery applications, a hypothesis supported by confocal microscopy assays. This work not only provides valuable insight into modulating nanogel stiffness but also advances new nanosystems for promising biomedical applications.
Collapse
Affiliation(s)
- Ju Wang
- Departments of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, Rome, 00185, Italy
| | - Benedetta Brugnoli
- Department of Chemistry, Sapienza University of Rome, Piazzale Aldo Moro 5, Rome, 00185, Italy
| | - Federica Foglietta
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria, 9, 10125, Turin, Italy
| | - Ilaria Andreana
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria, 9, 10125, Turin, Italy
| | - Giovanni Longo
- Institute for the Structure of the Matter (ISM), Italian National Research Council (CNR), Via del fosso del Cavaliere 100, 00133, Rome, Italy
| | - Simone Dinarelli
- Institute for the Structure of the Matter (ISM), Italian National Research Council (CNR), Via del fosso del Cavaliere 100, 00133, Rome, Italy
| | - Marco Girasole
- Institute for the Structure of the Matter (ISM), Italian National Research Council (CNR), Via del fosso del Cavaliere 100, 00133, Rome, Italy
| | - Loredana Serpe
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria, 9, 10125, Turin, Italy
| | - Silvia Arpicco
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria, 9, 10125, Turin, Italy
| | - Iolanda Francolini
- Department of Chemistry, Sapienza University of Rome, Piazzale Aldo Moro 5, Rome, 00185, Italy
| | - Chiara Di Meo
- Departments of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, Rome, 00185, Italy
| | - Pietro Matricardi
- Departments of Drug Chemistry and Technologies, Sapienza University of Rome, Piazzale Aldo Moro 5, Rome, 00185, Italy.
| |
Collapse
|
46
|
Yu XJ, Zhao YT, Abudouaini H, Zou P, Li TQ, Bai XF, Wang SX, Guan JB, Li MW, Wang XD, Wang YG, Hao DJ. A novel spherical GelMA-HAMA hydrogel encapsulating APET×2 polypeptide and CFIm25-targeting sgRNA for immune microenvironment modulation and nucleus pulposus regeneration in intervertebral discs. J Nanobiotechnology 2024; 22:556. [PMID: 39267105 PMCID: PMC11391743 DOI: 10.1186/s12951-024-02783-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2024] [Accepted: 08/16/2024] [Indexed: 09/14/2024] Open
Abstract
METHODS Single-cell transcriptomics and high-throughput transcriptomics were used to screen factors significantly correlated with intervertebral disc degeneration (IDD). Expression changes of CFIm25 were determined via RT-qPCR and Western blot. NP cells were isolated from mouse intervertebral discs and induced to degrade with TNF-α and IL-1β. CFIm25 was knocked out using CRISPR-Cas9, and CFIm25 knockout and overexpressing nucleus pulposus (NP) cell lines were generated through lentiviral transfection. Proteoglycan expression, protein expression, inflammatory factor expression, cell viability, proliferation, migration, gene expression, and protein expression were analyzed using various assays (alcian blue staining, immunofluorescence, ELISA, CCK-8, EDU labeling, transwell migration, scratch assay, RT-qPCR, Western blot). The GelMA-HAMA hydrogel loaded with APET×2 polypeptide and sgRNA was designed, and its effects on NP regeneration were assessed through in vitro and mouse model experiments. The progression of IDD in mice was evaluated using X-ray, H&E staining, and Safranin O-Fast Green staining. Immunohistochemistry was performed to determine protein expression in NP tissue. Proteomic analysis combined with in vitro and in vivo experiments was conducted to elucidate the mechanisms of hydrogel action. RESULTS CFIm25 was upregulated in IDD NP tissue and significantly correlated with disease progression. Inhibition of CFIm25 improved NP cell degeneration, enhanced cell proliferation, and migration. The hydrogel effectively knocked down CFIm25 expression, improved NP cell degeneration, promoted cell proliferation and migration, and mitigated IDD progression in a mouse model. The hydrogel inhibited inflammatory factor expression (IL-6, iNOS, IL-1β, TNF-α) by targeting the p38/NF-κB signaling pathway, increased collagen COLII and proteoglycan Aggrecan expression, and suppressed NP degeneration-related factors (COX-2, MMP-3). CONCLUSION The study highlighted the crucial role of CFIm25 in IDD and introduced a promising therapeutic strategy using a porous spherical GelMA-HAMA hydrogel loaded with APET×2 polypeptide and sgRNA. This innovative approach offers new possibilities for treating degenerated intervertebral discs.
Collapse
Grants
- 82302763, 82202764, 82202765 National Natural Science Foundation of China
- 82302763, 82202764, 82202765 National Natural Science Foundation of China
- 82302763, 82202764, 82202765 National Natural Science Foundation of China
- 82302763, 82202764, 82202765 National Natural Science Foundation of China
- 82302763, 82202764, 82202765 National Natural Science Foundation of China
- 82302763, 82202764, 82202765 National Natural Science Foundation of China
- 82302763, 82202764, 82202765 National Natural Science Foundation of China
- 82302763, 82202764, 82202765 National Natural Science Foundation of China
- 82302763, 82202764, 82202765 National Natural Science Foundation of China
- 82302763, 82202764, 82202765 National Natural Science Foundation of China
- 82302763, 82202764, 82202765 National Natural Science Foundation of China
- 82302763, 82202764, 82202765 National Natural Science Foundation of China
Collapse
Affiliation(s)
- Xiao-Jun Yu
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an City, Shaanxi Province, 710054, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, Shaanxi, China
| | - Yuan-Ting Zhao
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an City, Shaanxi Province, 710054, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, Shaanxi, China
| | - Haimiti Abudouaini
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an City, Shaanxi Province, 710054, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, Shaanxi, China
| | - Peng Zou
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an City, Shaanxi Province, 710054, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, Shaanxi, China
| | - Tian-Qi Li
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an City, Shaanxi Province, 710054, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, Shaanxi, China
| | - Xiao-Fan Bai
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an City, Shaanxi Province, 710054, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, Shaanxi, China
| | - Shan-Xi Wang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an City, Shaanxi Province, 710054, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, Shaanxi, China
| | - Jian-Bin Guan
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an City, Shaanxi Province, 710054, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, Shaanxi, China
| | - Meng-Wei Li
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, 430030, China
| | - Xiao-Dong Wang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an City, Shaanxi Province, 710054, China
- Shaanxi Key Laboratory of Spine Bionic Treatment, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, Shaanxi, China
| | - Ying-Guang Wang
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an City, Shaanxi Province, 710054, China.
- Shaanxi Key Laboratory of Spine Bionic Treatment, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, Shaanxi, China.
| | - Ding-Jun Hao
- Department of Spine Surgery, Honghui Hospital, Xi'an Jiaotong University, Xi'an City, Shaanxi Province, 710054, China.
- Shaanxi Key Laboratory of Spine Bionic Treatment, No.555 Friendship East Road, South Gate, Beilin District, Xi'an, Shaanxi, China.
| |
Collapse
|
47
|
Ha YJ, Tak KH, Jung JM, Lee JL, Kim CW, Ah YC, Kim SS, Moon IJ, Yoon YS. The Effect of Polynucleotide-Hyaluronic Acid Hydrogel in the Recovery After Mechanical Skin Barrier Disruption. Skin Res Technol 2024; 30:e70068. [PMID: 39300806 DOI: 10.1111/srt.70068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 08/06/2024] [Accepted: 09/05/2024] [Indexed: 09/22/2024]
Abstract
BACKGROUND The epidermal barrier acts as a defense against external agents as well as helps to maintain body homeostasis. Polynucleotides (PN), exogenous DNA fragments, promote wound repair through their stimulatory and anti-inflammatory effects. Recent findings indicate a synergistic effect of PN and hyaluronic acid (HA) combinations in regulating inflammation and promoting cell proliferation. This study aims to elucidate the effects of PN and HA on repairing the epidermal barrier following its disruption by tape stripping (TS) in a mouse model. MATERIALS AND METHODS After disrupting the epidermal barrier using TS, a formulation containing PN (14 mg/mL) and HA (6 mg/mL) was applied. Trans-epidermal water loss (TEWL) was measured at 0, 3, 6, 24, 48, and 72 h. Mice were euthanized after the final application at 72 h, and tissue samples were analyzed for epidermal/dermal thickness, neutrophil infiltration, and filaggrin expression. RESULTS We observed a significant reduction in TEWL in the PN+HA group compared to that in the control group (20.8 ± 0.5 vs. 43.7 ± 0.5 g/m2h at 72 h, p < 0.05), indicating an improvement in barrier function. Histological evaluation showed decreased epidermal and dermal thickening in the PN+HA group compared to that in the control group (epidermal: 29.4 ± 2.2 vs. 57.9 ± 3.5 μm; dermal: 464.8 ± 25.9 vs. 825.9 ± 44.8 μm, both p < 0.05). Additionally, neutrophil infiltration in the dermis was significantly reduced, and filaggrin protein levels were significantly higher in the PN+HA group compared to those in the control group (4.8 ± 0.4 vs. 21.1 ± 3.3 for neutrophils; 0.84 ± 0.04 vs. 0.42 ± 0.03 for filaggrin, both p < 0.05). CONCLUSION These results suggest that PN+HA may be an effective therapeutic strategy for repairing skin barrier damage.
Collapse
Affiliation(s)
- Ye Jin Ha
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Ka Hee Tak
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jin-Min Jung
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
- Division of Colon and Rectal Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Jong Lyul Lee
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
- Division of Colon and Rectal Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Chan Wook Kim
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
- Division of Colon and Rectal Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | | | | | - Ik Jun Moon
- Department of Dermatology, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| | - Yong Sik Yoon
- Asan Institute for Life Sciences, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
- Division of Colon and Rectal Surgery, Department of Surgery, Asan Medical Center, University of Ulsan College of Medicine, Seoul, South Korea
| |
Collapse
|
48
|
Lu X, Fan M, Ma Y, Feng Y, Pan L. Redox-sensitive hydrogel based on hyaluronic acid with selenocystamine cross-linking for the delivery of Limosilactobacillus reuteri in a DSS-induced colitis mouse model. Int J Biol Macromol 2024; 276:133855. [PMID: 39032895 DOI: 10.1016/j.ijbiomac.2024.133855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2024] [Revised: 06/30/2024] [Accepted: 07/11/2024] [Indexed: 07/23/2024]
Abstract
Disrupted gut microbiota homeostasis is an important cause of inflammatory colitis. Studies have shown that effective supplementation with probiotics can maintain microbial homeostasis and alleviate colitis. Here, to increase the viability of probiotics in the harsh gastrointestinal environments and enable targeted delivery, a redox-sensitive selenium hyaluronic acid (HA-Se) hydrogel encapsulating probiotics was developed. HA was modified with selenocystamine dihydrochloride and crosslinked by an amide reaction to generate a redox-sensitive hydrogel with stable mechanical properties, a low hemolysis rate and satisfactory biocompatibility. The HA-Se hydrogel exhibited suitable sensitivity to 10 mM GSH or 100 μM H2O2. The encapsulation of Limosilactobacillus reuteri (LR) in the HA-Se hydrogel (HA-Se-LR) significantly increased the survival rate of the probiotics in simulated gastric and intestinal fluid. HA-Se-LR administration increased the survival rate of mice with dextran sulfate sodium (DSS)-induced colitis, significantly alleviated oxidative stress and inflammation, and increased the effect of LR on microbiota α diversity. These results indicate that the HA-Se hydrogel constructed in this study can be used as a delivery platform to treat colitis, expanding the targeted applications of the natural polymer HA in disease treatment and the administration of probiotics as drugs to alleviate disease symptoms.
Collapse
Affiliation(s)
- Xi Lu
- College of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an 710000, China.
| | - Mingming Fan
- College of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an 710000, China
| | - Yuzhe Ma
- College of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an 710000, China
| | - Yimeng Feng
- Mathematics Teaching and Research Group, Dajindian Town Junior High School, Zhengzhou 450000, China
| | - Lei Pan
- Tangdu Hospital, Air Force Military Medical University, Xi'an 710000, China
| |
Collapse
|
49
|
Giaretta S, Magni A, Migliore A, Natoli S, Puntillo F, Ronconi G, Santoiemma L, Sconza C, Viapiana O, Zanoli G. A Review of Current Approaches to Pain Management in Knee Osteoarthritis with a Focus on Italian Clinical Landscape. J Clin Med 2024; 13:5176. [PMID: 39274389 PMCID: PMC11396710 DOI: 10.3390/jcm13175176] [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: 06/17/2024] [Revised: 07/24/2024] [Accepted: 08/11/2024] [Indexed: 09/16/2024] Open
Abstract
The global cases of knee osteoarthritis (KOA) are projected to increase by 74.9% by 2050. Currently, over half of patients remain dissatisfied with their pain relief. This review addresses unmet needs for moderate-to-severe KOA pain; it offers evidence and insights for improved management. Italian experts from the fields of rheumatology, physical medicine and rehabilitation, orthopedics, primary care, and pain therapy have identified several key issues. They emphasized the need for standardized care protocols to address inconsistencies in patient management across different specialties. Early diagnosis is crucial, as cartilage responds better to early protective and structural therapies. Faster access to physiatrist evaluation and reimbursement for physical, rehabilitative, and pharmacological treatments, including intra-articular (IA) therapy, could reduce access disparities. Concerns surround the adverse effects of oral pharmacological treatments, highlighting the need for safer alternatives. Patient satisfaction with corticosteroids and hyaluronic acid-based IA therapies reduces over time and there is no consensus on the optimal IA therapy protocol. Surgery should be reserved for severe symptoms and radiographic KOA evidence, as chronic pain post-surgery poses significant societal and economic burdens. The experts advocate for a multidisciplinary approach, promoting interaction and collaboration between specialists and general practitioners, to enhance KOA care and treatment consistency in Italy.
Collapse
Affiliation(s)
- Stefano Giaretta
- UOC Ortopedia e Traumatologia OC San Bortolo di Vicenza (AULSS 8 Berica), 36100 Vicenza, Italy
| | - Alberto Magni
- Local Health Department, Desenzano sul Garda, 25015 Brescia, Italy
| | - Alberto Migliore
- Unit of Rheumatology, San Pietro Fatebenefratelli Hospital, 00189 Rome, Italy
| | - Silvia Natoli
- Department of Clinical-Surgical, Diagnostic, and Pediatric Sciences, University of Pavia, 27100 Pavia, Italy
- Pain Unit, IRCCS Policlinico San Matteo, 27100 Pavia, Italy
| | - Filomena Puntillo
- Anaesthesia, Intensive Care and Pain Unit, Department of Interdisciplinary Medicine, University of Bari "Aldo Moro", 70124 Bari, Italy
| | - Gianpaolo Ronconi
- Department of Rehabilitation, Catholic University of the Sacred Heart, 00168 Rome, Italy
| | | | | | - Ombretta Viapiana
- Rheumatology Unit, Azienda Ospedaliera Universitaria Integrata di Verona, University of Verona, 37126 Verona, Italy
| | - Gustavo Zanoli
- Orthopaedic Ward, Casa di Cura Santa Maria Maddalena, Occhiobello, 45030 Rovigo, Italy
| |
Collapse
|
50
|
Shi R, Zhu Y, Lu W, Zhai R, Zhou M, Shi S, Chen Y. Nanomaterials: innovative approaches for addressing key objectives in periodontitis treatment. RSC Adv 2024; 14:27904-27927. [PMID: 39224639 PMCID: PMC11367407 DOI: 10.1039/d4ra03809f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Accepted: 07/26/2024] [Indexed: 09/04/2024] Open
Abstract
Periodontitis is a chronic inflammatory disease primarily caused by dental plaque, which is a significant global public health concern due to its high prevalence and severe impact on oral, and even systemic diseases. The current therapeutic plan focuses on three objectives: pathogenic bacteria inhibition, inflammation control, and osteogenic differentiation induction. Existing treatments still have plenty of drawbacks, thus, there is a pressing need for novel methods to achieve more effective treatment effects. Nanomaterials, as emerging materials, have been proven to exert their inherent biological properties or serve as stable drug delivery platforms, which may offer innovative solutions in periodontitis treatment. Nanomaterials utilized in periodontitis treatment fall into two categories, organic and inorganic nanomaterials. Organic nanomaterials are known for their biocompatibility and their potential to promote tissue regeneration and cell functions, including natural and synthetic polymers. Inorganic nanomaterials, such as metal, oxides, and mesoporous silica nanoparticles, exhibit unique physicochemical properties that make them suitable as antibacterial agents and drug delivery platforms. The inorganic nanosurface provides terrain induction for cell migration and osteogenic regeneration at defect sites by introducing different surface morphologies. Inorganic nanomaterials also play a role in antibacterial photodynamic therapy (aPDT) for eliminating pathogenic bacteria in the oral cavity. In this review, we will introduce multiple forms and applications of nanomaterials in periodontitis treatment and focus on their roles in addressing the key therapeutic objectives, to emphasize their promising future in achieving more effective and patient-friendly approaches toward periodontal tissue regeneration and overall health.
Collapse
Affiliation(s)
- Ruijianghan Shi
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu 610041 Sichuan China
| | - Yujie Zhu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu 610041 Sichuan China
| | - Weitong Lu
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu 610041 Sichuan China
| | - Ruohan Zhai
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu 610041 Sichuan China
| | - Mi Zhou
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu 610041 Sichuan China
| | - Sirong Shi
- State Key Laboratory of Oral Diseases, National Center for Stomatology, National Clinical Research Center for Oral Diseases, West China Hospital of Stomatology, Sichuan University Chengdu 610041 Sichuan China
| | - Yang Chen
- Department of Pediatric Surgery, Department of Liver Surgery & Liver Transplantation Center, West China Hospital of Sichuan University Chengdu 610041 Sichuan China
| |
Collapse
|