1
|
Jin Z, Zhang Y, Hu H, Li Q, Zhang L, Zhao K, Liu W, Li L, Gao C. Closed-loop theranostic microgels for immune microenvironment modulation and microbiota remodeling in ulcerative colitis. Biomaterials 2025; 314:122834. [PMID: 39288617 DOI: 10.1016/j.biomaterials.2024.122834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/26/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024]
Abstract
Inflammatory bowel disease (IBD) is characterized by the upregulation of reactive oxygen species (ROS) and dysfunction of gut immune system, and microbiota. The conventional treatments mainly focus on symptom control with medication by overuse of drugs. There is an urgent need to develop a closed-loop strategy that combines in situ monitoring and precise treatment. Herein, we innovatively designed the 'cluster munition structure' theranostic microgels to realize the monitoring and therapy for ulcerative colitis (a subtype of IBD). The superoxide anion specific probe (tetraphenylethylene-coelenterazine, TPC) and ROS-responsive nanogels consisting of postbiotics urolithin A (UA) were loaded into alginate and ion-crosslinked to obtain the theranostic microgels. The theranostic microgels could be delivered to the inflammatory site, where the environment-triggered breakup of the microgels and release of the nanogels were achieved in sequence. The TPC-UA group had optimal results in reducing inflammation, repairing colonic epithelial tissue, and remodeling microbiota, leading to inflammation amelioration and recovery of tight junction between the colonic epithelium, and maintenance of gut microbiota. During the recovery process, the local chemiluminescence intensity, which is proportional to the degree of inflammation, was gradually inhibited. The cluster munition of theranostic microgels displayed promising outcomes in monitoring inflammation and precise therapy, and demonstrated the potential for inflammatory disease management.
Collapse
Affiliation(s)
- Zeyuan Jin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China.
| | - Yaqi Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Haijun Hu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Qian Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Liwen Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Kefei Zhao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Wenxing Liu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China; Center for Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing, 312099, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China; Center for Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing, 312099, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
2
|
Ghosh S, Zheng M, He J, Wu Y, Zhang Y, Wang W, Shen J, Yeung KWK, Neelakantan P, Xu C, Qiao W. Electrically-driven drug delivery into deep cutaneous tissue by conductive microneedles for fungal infection eradication and protective immunity. Biomaterials 2025; 314:122908. [PMID: 39454504 DOI: 10.1016/j.biomaterials.2024.122908] [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: 07/13/2024] [Revised: 10/14/2024] [Accepted: 10/20/2024] [Indexed: 10/28/2024]
Abstract
Fungal infections affect over 13 million people worldwide and are responsible for 1.5 million deaths annually. Some deep cutaneous fungal infections may extend the dermal barriers to cause systemic infection, resulting in substantial morbidity and mortality. However, the management of deep cutaneous fungal infection is challenging and yet overlooked by traditional treatments, which only offer limited drug availability within deep tissue. In this study, we have developed an electrically stimulated microneedle patch to deliver miconazole into the subcutaneous layer. We tested its antifungal efficacy using in vitro and ex vivo models that mimic fungal infection. Moreover, we confirmed its anti-fungal and wound-healing effects in a murine subcutaneous fungal infection model. Furthermore, our findings also showed that the combination of miconazole and applied current synergistically stimulated the nociceptive sensory nerves, thereby activating protective cutaneous immunity mediated by dermal dendritic and γδ-T cells. Collectively, this study provides a new strategy for minimally invasive delivery of therapeutic agents and the modulation of the neuro-immune axis in deep tissue.
Collapse
Affiliation(s)
- Sumanta Ghosh
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China; Division of Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Mengjia Zheng
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, China
| | - Jiahui He
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, China
| | - Yefeng Wu
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China
| | - Yaming Zhang
- Department of Pharmacy and Pharmacology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Weiping Wang
- Department of Pharmacy and Pharmacology, Li Ka Shing Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China
| | - Jie Shen
- Shenzhen Key Laboratory of Spine Surgery, Department of Spine Surgery, Peking University Shenzhen Hospital, Shenzhen, China
| | - Kelvin W K Yeung
- Department of Orthopaedics and Traumatology, Faculty of Medicine, The University of Hong Kong, Hong Kong SAR, China; Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China
| | - Prasanna Neelakantan
- Division of Restorative Dental Sciences, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China; Mike Petryk School of Dentistry, Faculty of Medicine and Dentistry, University of Alberta, Edmonton, Alberta, Canada.
| | - Chenjie Xu
- Department of Biomedical Engineering, City University of Hong Kong, 83 Tat Chee Avenue, Kowloon, Hong Kong SAR, China.
| | - Wei Qiao
- Applied Oral Sciences and Community Dental Care, Faculty of Dentistry, The University of Hong Kong, Hong Kong SAR, China; Shenzhen Key Laboratory for Innovative Technology in Orthopaedic Trauma, The University of Hong Kong-Shenzhen Hospital, Shenzhen, China.
| |
Collapse
|
3
|
Bezold MG, Dollinger BR, DeJulius CR, Keech MC, Hanna AR, Kittel AR, Yu F, Gupta MK, D'Arcy R, Brunger JM, Duvall CL. Shear-thinning hydrogel for allograft cell transplantation and externally controlled transgene expression. Biomaterials 2025; 314:122812. [PMID: 39288619 DOI: 10.1016/j.biomaterials.2024.122812] [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: 06/20/2024] [Revised: 08/22/2024] [Accepted: 09/02/2024] [Indexed: 09/19/2024]
Abstract
This work establishes the design of a fully synthetic, shear-thinning hydrogel platform that is injectable and can isolate engineered, allogeneic cell therapies from the host. We utilized RAFT to generate a library of linear random copolymers of N,N-dimethylacrylamide (DMA) and 2-vinyl-4,4-dimethyl azlactone (VDMA) with variable mol% VDMA and degree of polymerization. Poly(DMA-co-VDMA) copolymers were subsequently modified with either adamantane (Ad) or β-cyclodextrin (Cd) through amine-reactive VDMA to prepare hydrogel precursor macromers containing complementary guest-host pairing pendant groups that, when mixed, form shear-thinning hydrogels. Rheometric evaluation of the hydrogel library enabled identification of lead macromer structures comprising 15 mol% pendants (Ad or Cd) and a degree of polymerization of 1000; mixing of these Ad and Cd functionalized precursors yielded hydrogels possessing storage modulus above 1000 Pa, tan(δ) values below 1 and high yield strain, which are target characteristics of robust but injectable shear-thinning gels. This modular system proved amenable to nanoparticle integration with surface-modified nanoparticles displaying Ad. The addition of the Ad-functionalized nanoparticles simultaneously improved mechanical properties of the hydrogels and enabled extended hydrogel retention of a model small molecule in vivo. In studies benchmarking against alginate, a material traditionally used for cell encapsulation, the lead hydrogel showed significantly less fibrous encapsulation in a subcutaneous implant site. Finally, this platform was utilized to encapsulate and extend in vivo longevity of inducible transgene-engineered mesenchymal stem cells in an allogeneic transplant model. The hydrogels remained intact and blocked infiltration by host cells, consequently extending the longevity of grafted cell function relative to a benchmark, shear-thinning hyaluronic acid-based gel. In sum, the new synthetic, shear-thinning hydrogel system presented here shows potential for further development as an injectable platform for delivery and in situ drug modulation of allograft and engineered cell therapies.
Collapse
Affiliation(s)
- Mariah G Bezold
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Bryan R Dollinger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Carlisle R DeJulius
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Megan C Keech
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Andrew R Hanna
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Anna R Kittel
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Fang Yu
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Mukesh K Gupta
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Richard D'Arcy
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Jonathan M Brunger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, TN, 37235, USA.
| |
Collapse
|
4
|
Hughes MDG, West D, Wurr R, Cussons S, Cook KR, Mahmoudi N, Head D, Brockwell DJ, Dougan L. Competition between cross-linking and force-induced local conformational changes determines the structure and mechanics of labile protein networks. J Colloid Interface Sci 2025; 678:1259-1269. [PMID: 39357245 DOI: 10.1016/j.jcis.2024.09.183] [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: 07/17/2024] [Revised: 09/03/2024] [Accepted: 09/21/2024] [Indexed: 10/04/2024]
Abstract
Folded protein hydrogels are emerging as promising new materials for medicine and healthcare applications. Folded globular proteins can be modelled as colloids which exhibit site specific cross-linking for controlled network formation. However, folded proteins have inherent mechanical stability and unfolded in response to an applied force. It is not yet understood how colloidal network theory maps onto folded protein hydrogels and whether it models the impact of protein unfolding on network properties. To address this, we study a hybrid system which contains folded proteins (patchy colloids) and unfolded proteins (biopolymers). We use a model protein, bovine serum albumin (BSA), to explore network architecture and mechanics in folded protein hydrogels. We alter both the photo-chemical cross-linking reaction rate and the mechanical properties of the protein building block, via illumination intensity and redox removal of robust intra-protein covalent bonds, respectively. This dual approach, in conjunction with rheological and structural techniques, allows us to show that while reaction rate can 'fine-tune' the mechanical and structural properties of protein hydrogels, it is the force-lability of the protein which has the greatest impact on network architecture and rigidity. To understand these results, we consider a colloidal model which successfully describes the behaviour of the folded protein hydrogels but cannot account for the behaviour observed in force-labile hydrogels containing unfolded protein. Alternative models are needed which combine the properties of colloids (folded proteins) and biopolymers (unfolded proteins) in cross-linked networks. This work provides important insights into the accessible design space of folded protein hydrogels without the need for complex and costly protein engineering, aiding the development of protein-based biomaterials.
Collapse
Affiliation(s)
- Matt D G Hughes
- School of Physics and Astronomy, Faculty of Engineering and Physical Sciences, University of Leeds, UK
| | - Daniel West
- School of Physics and Astronomy, Faculty of Engineering and Physical Sciences, University of Leeds, UK
| | - Rebecca Wurr
- School of Physics and Astronomy, Faculty of Engineering and Physical Sciences, University of Leeds, UK; Department of Physics, King's College London, London, WC2R 2LS, UK
| | - Sophie Cussons
- Astbury Centre for Structural Molecular Biology, University of Leeds, UK; School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, UK
| | - Kalila R Cook
- School of Physics and Astronomy, Faculty of Engineering and Physical Sciences, University of Leeds, UK
| | - Najet Mahmoudi
- ISIS Neutron and Muon Spallation Source, STFC Rutherford Appleton Laboratory, Oxfordshire, UK
| | - David Head
- School of Computer Science, Faculty of Engineering and Physical Science, University of Leeds, UK
| | - David J Brockwell
- Astbury Centre for Structural Molecular Biology, University of Leeds, UK; School of Molecular and Cellular Biology, Faculty of Biological Sciences, University of Leeds, UK
| | - Lorna Dougan
- School of Physics and Astronomy, Faculty of Engineering and Physical Sciences, University of Leeds, UK; Astbury Centre for Structural Molecular Biology, University of Leeds, UK.
| |
Collapse
|
5
|
Zöller K, To D, Bernkop-Schnürch A. Biomedical applications of functional hydrogels: Innovative developments, relevant clinical trials and advanced products. Biomaterials 2025; 312:122718. [PMID: 39084097 DOI: 10.1016/j.biomaterials.2024.122718] [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: 02/08/2024] [Revised: 07/08/2024] [Accepted: 07/26/2024] [Indexed: 08/02/2024]
Abstract
Functional hydrogels are used for numerous biomedical applications such as tissue engineering, wound dressings, lubricants, contact lenses and advanced drug delivery systems. Most of them are based on synthetic or natural polymers forming a three-dimensional network that contains aqueous media. Among synthetic polymers, poly(meth)acrylates, polyethyleneglycols, poly(vinylalcohols), poly(vinylpyrrolidones), PLGA and poly(urethanes) are of high relevance, whereas natural polymers are mainly polysaccharides such as hyaluronic acid, alginate or chitosan and proteins such as albumin, collagen or elastin. In contrast to most synthetic polymers, natural polymers are biodegradable. Both synthetic and natural polymers are often chemically modified in order to improve or induce favorable properties and functions like high mechanical strength, stiffness, elasticity, high porosity, adhesive properties, in situ gelling properties, high water binding capacity or drug release controlling properties. Within this review we provide an overview about the broad spectrum of biomedical applications of functional hydrogels, summarize innovative approaches, discuss the concept of relevant functional hydrogels that are in clinical trials and highlight advanced products as examples for successful developments.
Collapse
Affiliation(s)
- Katrin Zöller
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, Innsbruck, 6020, Austria
| | - Dennis To
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, Innsbruck, 6020, Austria
| | - Andreas Bernkop-Schnürch
- Department of Pharmaceutical Technology, Institute of Pharmacy, University of Innsbruck, Innrain 80/82, Innsbruck, 6020, Austria.
| |
Collapse
|
6
|
Sparks Z, Chauhan A. Polymerization of hydroxyethyl methacrylate (HEMA) under rotation to form core-annular hydrogels. J Colloid Interface Sci 2025; 677:294-306. [PMID: 39094490 DOI: 10.1016/j.jcis.2024.07.105] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 07/11/2024] [Accepted: 07/12/2024] [Indexed: 08/04/2024]
Abstract
HYPOTHESIS We propose to polymerize high water content hydroxyethyl methacrylate (HEMA) formulations in a rotating cylinder to explore the effect of the rotation on microstructure and critical parameters such as diffusivity of model proteins in porous poly-HEMA gels. EXPERIMENTS Cylindrical molds were partially filled with water-HEMA-initiator-crosslinker mixtures and exposed to UV light while undergoing rotation to polymerize into a cylindrical tube. The process was repeated multiple times to manufacture a core annular rod with multiple concentric rings, in which at least one ring was porous. The porous gels were imaged by scanning electron microscopy to explore the microstructure. The transport of model proteins bovine serum albumin and human γ-globulin was measured and modeled, in radial and axial directions, to obtain the effective diffusivity and partition coefficient. Also, the true diffusivity of proteins was calculated by accounting for the effects of porosity and tortuosity. FINDINGS The porous gels exhibited diffusion-controlled release of both model proteins. The hydrogels prepared with 55% water in the monomer mixture were porous with non-isotropic structure likely due to axially oriented pores with minimal radial connectivity. The gels with higher water content were isotropic with interconnected pores in both directions. The pore volume increased with water content, but the partition coefficient was relatively constant and less than one likely due to presence of isolated unconnected pores.
Collapse
Affiliation(s)
- Zachary Sparks
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, CO 80401, United States
| | - Anuj Chauhan
- Department of Chemical and Biological Engineering, Colorado School of Mines, Golden, CO 80401, United States.
| |
Collapse
|
7
|
Yang H, Whitby CP, Travas-Sejdic J. Dual-network hydrogel capsules for controlled molecular transport via pH and temperature responsiveness. J Colloid Interface Sci 2025; 677:942-951. [PMID: 39178673 DOI: 10.1016/j.jcis.2024.08.119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2024] [Revised: 08/12/2024] [Accepted: 08/15/2024] [Indexed: 08/26/2024]
Abstract
We have developed innovative core-shell hydrogel capsules with a dual-network shell structure designed for precise control of molecular transport in response to external stimuli such as pH and temperature. The capsules were fabricated using a combination of microfluidic electrospray techniques and water-in-water (w/w) core-shell droplets templating. The primary network of the shell, calcium alginate (Ca-Alg), with a pKa around 3.4, exhibits sensitivity to pH. The secondary network of the shell, poly(ethylene glycol) methyl ether methacrylate (PEGMA), undergoes a volume phase transition near 60 °C. These properties enable precise molecular transport control in/out of the capsules by modulating the surface charges through varying pH and modifying pore size through temperature changes. Moreover, the dual-network shell structure not only significantly enhances the mechanical strength of the capsules but also improves their stability under external stimulus, ensuring structural integrity during the transport of molecules. This research lays the groundwork for further investigations into the multimodal stimuli-responsive hydrogel systems to control molecular transport, important in applications such as sensors and reactors for chemical cascade reactions.
Collapse
Affiliation(s)
- Hui Yang
- Centre for Innovative Materials for Health, School of Chemical Sciences, The University of Auckland, 23 Symonds Street, Auckland, New Zealand; MacDiarmid Institute for Advanced Materials and Nanotechnology, Victoria University of Wellington, PO Box 600, Wellington, New Zealand
| | - Catherine P Whitby
- MacDiarmid Institute for Advanced Materials and Nanotechnology, Victoria University of Wellington, PO Box 600, Wellington, New Zealand; School of Food Technology and Natural Sciences, Massey University, Palmerston North, New Zealand
| | - Jadranka Travas-Sejdic
- Centre for Innovative Materials for Health, School of Chemical Sciences, The University of Auckland, 23 Symonds Street, Auckland, New Zealand; MacDiarmid Institute for Advanced Materials and Nanotechnology, Victoria University of Wellington, PO Box 600, Wellington, New Zealand.
| |
Collapse
|
8
|
Wang M, Bergès R, Malfanti A, Préat V, Bastiancich C. Local delivery of doxorubicin prodrug via lipid nanocapsule-based hydrogel for the treatment of glioblastoma. Drug Deliv Transl Res 2024; 14:3322-3338. [PMID: 37889402 PMCID: PMC11499358 DOI: 10.1007/s13346-023-01456-y] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/17/2023] [Indexed: 10/28/2023]
Abstract
Glioblastoma (GBM) recurrences appear in most cases around the resection cavity borders and arise from residual GBM cells that cannot be removed by surgery. Here, we propose a novel treatment that combines the advantages of nanomedicine and local drug delivery to target these infiltrating GBM cells. We developed an injectable lipid nanocapsule (LNC)-based formulation loaded with lauroyl-doxorubicin prodrug (DOXC12). Firstly, we demonstrated the efficacy of intratumoral administration of DOXC12 in GL261 GBM-bearing mice, which extended mouse survival. Then, we formulated an injectable hydrogel by mixing the appropriate amount of prodrug with the lipophilic components of LNC. We optimized the hydrogel by incorporating cytidine-C16 (CytC16) to achieve a mechanical stiffness adapted for an application in the brain post-surgery (DOXC12-LNCCL). DOXC12-LNCCL exhibited high DOXC12 encapsulation efficiency (95%) and a size of approximately 60 nm with sustained drug release for over 1 month in vitro. DOXC12-LNCCL exhibited enhanced cytotoxicity compared to free DOXC12 (IC50 of 349 and 86 nM, respectively) on GL261 GBM cells and prevented the growth of GL261 spheroids cultured on organotypic brain slices. In vivo, post-surgical treatment with DOXC12-LNCCL significantly improved the survival of GL261-bearing mice. The combination of this local treatment with the systemic administration of anti-inflammatory drug ibuprofen further delayed the onset of recurrences. In conclusion, our study presents a promising therapeutic approach for the treatment of GBM. By targeting residual GBM cells and reducing the inflammation post-surgery, we present a new strategy to delay the onset of recurrences in the gap period between surgery and standard of care therapy.
Collapse
Affiliation(s)
- Mingchao Wang
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73, 1200, Brussels, Belgium
| | - Raphaël Bergès
- Aix-Marseille University, CNRS, INP, Inst Neurophysiopathol, 27 Boulevard Jean Moulin, Marseille, 13005, France
| | - Alessio Malfanti
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73, 1200, Brussels, Belgium
| | - Véronique Préat
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73, 1200, Brussels, Belgium.
| | - Chiara Bastiancich
- UCLouvain, Louvain Drug Research Institute, Advanced Drug Delivery and Biomaterials, Avenue Mounier 73, 1200, Brussels, Belgium.
- Aix-Marseille University, CNRS, INP, Inst Neurophysiopathol, 27 Boulevard Jean Moulin, Marseille, 13005, France.
- Department of Drug Science and Technology, University of Turin, Via Pietro Giuria 9, Turin, 10125, Italy.
| |
Collapse
|
9
|
Pohjola J, Jokinen M, Soukka T, Stolt M. Polymer microsphere inks for semi-solid extrusion 3D printing at ambient conditions. J Mech Behav Biomed Mater 2024; 160:106783. [PMID: 39486301 DOI: 10.1016/j.jmbbm.2024.106783] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Revised: 10/02/2024] [Accepted: 10/15/2024] [Indexed: 11/04/2024]
Abstract
Extrusion-based 3D printing methods have great potential for manufacturing of personalized polymer-based drug-releasing systems. However, traditional melt-based extrusion techniques are often unsuitable for processing thermally labile molecules. Consequently, methods that utilize the extrusion of semi-solid inks under mild conditions are frequently employed. The rheological properties of the semi-solid inks have a substantial impact on the 3D printability, making it necessary to evaluate and tailor these properties. Here, we report a novel semi-solid extrusion 3D printing method based on utilization of a Carbopol gel matrix containing various concentrations of polymeric microspheres. We also demonstrate the use of a solvent vapor-based post-processing method for enhancing the mechanical strength of the printed objects. As our approach enables room-temperature processing of polymers typically used in the pharmaceutical industry, it may also facilitate the broader application of 3D printing and microsphere technologies in preparation of personalized medicine.
Collapse
Affiliation(s)
- Juuso Pohjola
- Biotechnology, Department of Life Technologies, Faculty of Technology, University of Turku, FI-20014 Turku, Finland; Pharmaceutical Sciences, Bayer Oy, FI-20210 Turku, Finland.
| | | | - Tero Soukka
- Biotechnology, Department of Life Technologies, Faculty of Technology, University of Turku, FI-20014 Turku, Finland
| | - Mikael Stolt
- Pharmaceutical Sciences, Bayer Oy, FI-20210 Turku, Finland
| |
Collapse
|
10
|
Guo Z, Xiu L, Li Y, Tan J, Wei C, Sui J, Zhang S, Zhu R, Li JL. Injectable nanocomposite hydrogel with cascade drug release for treatment of uveal melanoma. J Control Release 2024; 376:1086-1099. [PMID: 39500408 DOI: 10.1016/j.jconrel.2024.11.001] [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: 06/06/2024] [Revised: 10/28/2024] [Accepted: 11/01/2024] [Indexed: 11/11/2024]
Abstract
Uveal melanoma (UM) is the most common malignant intraocular tumor with the trait of distant metastases. Currently, the standard clinical therapy results in suboptimal outcomes due to ineffective inhibition of tumor metastasis. Thus, developing novel therapeutic modalities for UM remains a critical priority. Herein, we have developed an injectable nanocomposite hydrogel (HA-DOX/LAP gel) through integrating hyaluronic acid-based drug-loaded nanoparticles into an alginate-dopamine gel, delivering the chemotherapeutic drugs, lapatinib and doxorubicin for combinational treatment of UM. HA-DOX/LAP gel is fabricated in situ by a simple injection of the mixed precursor solution into tumor sites and maintains in vivo for more than 21 days. The entrapped drug-loaded nanoparticles can gradually release from HA-DOX/LAP gel, enhancing tumor targeting and penetration, and synchronously releasing lapatinib and doxorubicin into the acidic intracellular environment to synergistically destroy UM cells. In vivo anti-tumor studies conducted in MuM-2B tumor models demonstrated that HA-DOX/LAP gel significantly impedes tumor growth, diminishes postoperative recurrence, and prolongs overall survivals of UM tumor-bearing mice through only single injection. Remarkably, the escaped drug-loaded nanoparticles effectively reduce the risk of tumor metastases. Our findings provide new insights for the development of multifunctional nanocomposite-incorporating combination therapy against UM by targeting tumor recurrence and metastases.
Collapse
Affiliation(s)
- Zhihao Guo
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China.
| | - Linyun Xiu
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Yumei Li
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Jiangcheng Tan
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Cailing Wei
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Junhui Sui
- Beijing Tide Pharmaceutical Co., Ltd., Beijing 100176, China
| | - Shijin Zhang
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Ruohua Zhu
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China
| | - Ji-Liang Li
- National Engineering Research Center of Ophthalmology and Optometry, School of Biomedical Engineering, Eye Hospital, Wenzhou Medical University, Wenzhou 325027, China; University of Chinese Academy of Sciences Wenzhou Institute, Wenzhou 325001, China.
| |
Collapse
|
11
|
Qiu W, Gehre C, Nepomuceno JP, Bao Y, Li Z, Müller R, Qin XH. Coumarin-Based Photodegradable Hydrogels Enable Two-Photon Subtractive Biofabrication at 300 mm s -1. Angew Chem Int Ed Engl 2024; 63:e202404599. [PMID: 39023389 DOI: 10.1002/anie.202404599] [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: 03/06/2024] [Indexed: 07/20/2024]
Abstract
Spatiotemporally controlled two-photon photodegradation of hydrogels has gained increasing attention for high-precision subtractive tissue engineering. However, conventional photolabile hydrogels often have poor efficiency upon two-photon excitation in the near-infrared (NIR) region and thus require high laser dosage that may compromise cell activity. As a result, high-speed two-photon hydrogel erosion in the presence of cells remains challenging. Here we introduce the design and synthesis of efficient coumarin-based photodegradable hydrogels to overcome these limitations. A set of photolabile coumarin-functionalized polyethylene glycol linkers are synthesized through a Passerini multicomponent reaction. After mixing these linkers with thiolated hyaluronic acid, semi-synthetic photodegradable hydrogels are formed in situ via Michael addition crosslinking. The efficiency of photodegradation in these hydrogels is significantly higher than that in nitrobenzyl counterparts upon two-photon irradiation at 780 nm. A complex microfluidic network mimicking the bone microarchitecture is successfully fabricated in preformed coumarin hydrogels at high speeds of up to 300 mm s-1 and low laser dosage down to 10 mW. Further, we demonstrate fast two-photon printing of hollow microchannels inside a hydrogel to spatiotemporally direct cell migration in 3D. Collectively, these hydrogels may open new avenues for fast laser-guided tissue fabrication at high spatial resolution.
Collapse
Affiliation(s)
- Wanwan Qiu
- Institute for Biomechanics, ETH Zurich, Gloriastrasse 39, 8092, Zurich, Switzerland
| | - Christian Gehre
- Institute for Biomechanics, ETH Zurich, Gloriastrasse 39, 8092, Zurich, Switzerland
| | | | - Yinyin Bao
- Institute for Chemical and Bioengineering, ETH Zurich, Vladimir-Prelog-Weg 1-5/10, 8093, Zurich, Switzerland
| | - Zhiquan Li
- School of Materials and Energy, Guangdong University of Technology, Guangzhou, Guangdong, 510006, China
| | - Ralph Müller
- Institute for Biomechanics, ETH Zurich, Gloriastrasse 39, 8092, Zurich, Switzerland
| | - Xiao-Hua Qin
- Institute for Biomechanics, ETH Zurich, Gloriastrasse 39, 8092, Zurich, Switzerland
| |
Collapse
|
12
|
Hosseinlou R, Dargahi M, Keshtkar Vanashi A. Alkaline range pH sensor based on chitosan hydrogel: A novel approach to pH sensing. Int J Biol Macromol 2024; 279:135199. [PMID: 39218193 DOI: 10.1016/j.ijbiomac.2024.135199] [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: 06/22/2024] [Revised: 08/14/2024] [Accepted: 08/28/2024] [Indexed: 09/04/2024]
Abstract
Monitoring of pH under extreme alkaline range is still a challenge due to the lack of accuracy and validity. This research developed a novel pH sensor (hydrogel/BTB) based on the transition of bromothymol blue from the hydrogel matrix into the pH-examining sample solution. The hydrogel/BTB sensor was synthesized through the solvent casting of chitosan, citric acid as the crosslinker, and bromothymol blue as a pH-sensitive dye. The structure of hydrogel/BTB was characterized using Fourier-transform infrared spectroscopy (FT-IR), Energy-dispersive X-ray spectroscopy (EDS), Field emission scanning electron microscopy (FE-SEM), Brunauer-Emmett-Teller (BET) analysis, and thermogravimetric analysis (TGA). The effect of various parameters on pH determination was investigated. The developed pH sensor demonstrated a linear detection range validated from pH 10 to 14 using the gravimetric method, and from pH 11 to 14 using the colorimetric method. The sensor successfully detected pH in alkaline tap water, carbonate buffer, and ethanol amine buffer. The transition of bromothymol blue is described by the Peppas-Korsmeyer kinetic model. The activation and Gibbs free energy were obtained as 357.1 J/mol and 260 J/mol, respectively. This work furnished a new mechanism for pH detection, and it has excellent potential for developing novel sensors in this field.
Collapse
Affiliation(s)
- Rasool Hosseinlou
- Department of Chemistry, Faculty of Science, Imam Khomeini International University, Qazvin 34148-96818, Islamic Republic of Iran
| | - Maryam Dargahi
- Department of Chemistry, Faculty of Science, Imam Khomeini International University, Qazvin 34148-96818, Islamic Republic of Iran.
| | - Abolfazl Keshtkar Vanashi
- Department of Chemistry, Faculty of Science, Imam Khomeini International University, Qazvin 34148-96818, Islamic Republic of Iran
| |
Collapse
|
13
|
Zhao N, Yuan W. Injectable and self-healable hydrogel based on pullulan polysaccharide loading platelet-rich plasma and metal-phenol network nanoparticles for infectious wound healing. Int J Biol Macromol 2024; 279:135361. [PMID: 39244111 DOI: 10.1016/j.ijbiomac.2024.135361] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2024] [Revised: 09/03/2024] [Accepted: 09/04/2024] [Indexed: 09/09/2024]
Abstract
Due to elevated glucose levels, oxidative stress, weakened immune function, and delayed angiogenesis, diabetic wounds are difficult to heal. However, current dressings often do not successfully achieve the desired therapeutic outcome for diabetic wounds. Platelet rich plasma (PRP) is widely used in the treatment of diabetic wounds. Even so, the sudden release of growth factors or proteins in PRP frequently hampers the therapeutic efficacy. Therefore, it is of considerable clinical value to achieve long-term release of active molecules in PRP and to create more effective diabetic wound dressings. Here, PRP was combined with pullulan polysaccharide derivatives (OPD) and polylysine derivatives (EPL-BA) to form a multifunctional hydrogel backbone. Tea polyphenols (TP), gallic acid (GA), and metal ions (Cu2+) were utilized to prepare metal-phenol network nanoparticles (TGMPN) which were encapsulated in the hydrogel system. The EPL-BA/OPD/PRP/TGMPN (EOPM) composite hydrogel showed injectable and self-healing properties. The hydrogel system could effectively remove reactive oxygen species (ROS) and showed excellent antibacterial properties against different bacteria. The results showed that EOPM hydrogel was effective in reducing the inflammatory response and promoting the regeneration of blood vessels and neoplastic tissues, thus greatly accelerating the repair of infected wounds.
Collapse
Affiliation(s)
- Nuoya Zhao
- School of Materials Science and Engineering, Key Laboratory of Advanced Civil Materials of Ministry of Education, Tongji University, Shanghai 201804, People's Republic of China
| | - Weizhong Yuan
- School of Materials Science and Engineering, Key Laboratory of Advanced Civil Materials of Ministry of Education, Tongji University, Shanghai 201804, People's Republic of China.
| |
Collapse
|
14
|
Ding X, Li W, Shang L, Zhao Y, Sun W. Controllable Contact-Destructive Hydrogel Actuators. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2409965. [PMID: 39252674 DOI: 10.1002/adma.202409965] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 08/19/2024] [Indexed: 09/11/2024]
Abstract
Constructing hydrogels with spatially heterogeneous structures are crucial for unlocking novel applications. To this end, selectively removing a specific portion of hydrogels by facile and intricate destructive strategies is worth exploring. Herein, a "contact-destructive" hydrogel actuator is presented, composed of a dynamic hydrogel network doped with hydrophilic polyethylene glycol (PEG). The destructive behavior of the hydrogel actuator is attributed to the surface tension-induced spreading effect and the enhanced water absorption due to the additive PEG. Parameters that act on these mechanisms are used to control the destruction of the hydrogel. During the destructive process, the hydrogel actuator exhibits locomotion routes predetermined by the graphic pattern with the aid of 3D printing. Additionally, such self-destructive behavior can be terminated by UV light irradiation when PEG is replaced with poly(ethylene glycol) diacrylate (PEGDA). Significantly, diverse applications including controllable 3D structures collapse, self-erasing, and on-demand cell release, are realized with such self-destructive hydrogel. These results demonstrate that the present hydrogel has great values in soft robotics, anti-counterfeiting, controlled drug delivery, and other related fields.
Collapse
Affiliation(s)
- Xiaoya Ding
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
- Department of Rheumatology and Immunology, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health) Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Wenzhao Li
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health) Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
| | - Luoran Shang
- Department of Rheumatology and Immunology, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Shanghai Xuhui Central Hospital, Zhongshan-Xuhui Hospital, and the Shanghai Key Laboratory of Medical Epigenetics, the International Co-laboratory of Medical Epigenetics and Metabolism (Ministry of Science and Technology), Institutes of Biomedical Sciences Fudan University, Shanghai, 200032, China
| | - Yuanjin Zhao
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
- Department of Rheumatology and Immunology, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
- Oujiang Laboratory (Zhejiang Lab for Regenerative Medicine, Vision and Brain Health) Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, 325001, China
- Shenzhen Research Institute, Southeast University, Shenzhen, 518071, China
| | - Weijian Sun
- Department of Gastrointestinal Surgery, The First Affiliated Hospital, Wenzhou Medical University, Wenzhou, 325035, China
- Department of Rheumatology and Immunology, School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, China
| |
Collapse
|
15
|
Chen Y, Zhu Z, Shi K, Jiang Z, Guan C, Zhang L, Yang T, Xie F. Shellac-based materials: Structures, properties, and applications. Int J Biol Macromol 2024; 279:135102. [PMID: 39197605 DOI: 10.1016/j.ijbiomac.2024.135102] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/20/2024] [Accepted: 08/25/2024] [Indexed: 09/01/2024]
Abstract
Shellac stands out among natural polymers as the sole animal-derived resin, boasting a complex polyester composition comprising polyhydroxy long-chain fatty acids and sesquiterpene acids. Its unique attributes include biocompatibility, non-toxicity, distinctive amphiphilicity, superb film-forming and adhesive properties, excellent dielectric properties, rapid drying, and solubility in alkaline solutions while resisting acidic ones. These exceptional qualities have propelled shellac beyond its traditional role as a varnish and decorative material, positioning it as a viable option for diverse applications such as food packaging, pharmaceutical formulations, electronic devices, fiber dyeing, and wood restoration. Furthermore, shellac serves as a crucial carbon source for graphene materials. This review comprehensively explores shellac's contributions to prolonging food shelf life, enhancing the carbon sourcing of graphene materials, facilitating the delivery of active substances, boosting the performance of organic field-effect transistors, enabling environmentally friendly textile dyeing, and providing protective coatings for wood. Additionally, it delves into the current limitations and future directions of shellac's applications. By disseminating this knowledge, we aim to deepen researchers' comprehension of shellac and inspire further exploration, thereby fostering sustainable advancements across various industries.
Collapse
Affiliation(s)
- Ying Chen
- School of Food Science and Engineering, Yangzhou University, Huayang Xilu 196, Yangzhou, Jiangsu 225127, China
| | - Zhu Zhu
- School of Food Science and Engineering, Yangzhou University, Huayang Xilu 196, Yangzhou, Jiangsu 225127, China
| | - Kunbo Shi
- School of Food Science and Engineering, Yangzhou University, Huayang Xilu 196, Yangzhou, Jiangsu 225127, China
| | - Zhiyao Jiang
- School of Food Science and Engineering, Yangzhou University, Huayang Xilu 196, Yangzhou, Jiangsu 225127, China
| | - Chengran Guan
- School of Food Science and Engineering, Yangzhou University, Huayang Xilu 196, Yangzhou, Jiangsu 225127, China; Jiangsu Dairy Biotechnology Engineering Research Center, Yangzhou, Jiangsu 225127, China
| | - Liang Zhang
- School of Food Science and Engineering, Yangzhou University, Huayang Xilu 196, Yangzhou, Jiangsu 225127, China.
| | - Tao Yang
- School of Pharmacy, Hainan Medical University, Haikou 571199, China.
| | - Fengwei Xie
- Department of Chemical Engineering, University of Bath, Bath BA2 7AY, United Kingdom.
| |
Collapse
|
16
|
Qin L, Zhu Y, Zhang H, Ren H, Zhai H. Lignin-modified cellulose nanofibers hydrogel under adjustable binary solvent systems with excellent adhesion, self-healing and anti-freeze properties. Int J Biol Macromol 2024; 279:135559. [PMID: 39349328 DOI: 10.1016/j.ijbiomac.2024.135559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2024] [Revised: 09/05/2024] [Accepted: 09/09/2024] [Indexed: 10/02/2024]
Abstract
Hydrogels with remarkable flexibility have gained popularity as materials for current research. However, the unfavorable properties of short-term adhesion, susceptibility to damage, and freezing in low-temperature presented by conventional hydrogels have become bottlenecks for further applications. In this work, an anti-freezing hydrogel with excellent mechanical, adhesion, and self-healing properties were developed by constructing a persistent semiquinone/quinone-catechol redox equilibrium environment. The introduction of lignin-modified cellulose nanofibers (LCNFs) significantly improved the overall mechanical properties of the material, driven by strong hydrogen bond interactions. This enhancement was evident in the tensile properties (97.74 ± 1.72 kPa, 783 %) and compression properties (> 90 %). Within the internal network of the gel, the synergistic action of lignin and ammonium persulfate resulted in the production of catechol, which imparted the gel with excellent adhesion properties (28.26 ± 2.13 KPa) and broad adhesion applicability. In addition, the incorporation of ethylene glycol (EG) positively contributed to the strengthening of the gel while endowed with tunable anti-freezing properties. Given the exceptional advantages of the prepared hydrogels, they were used to assemble flexible strain sensors with outstanding sensitivity for monitoring human motions.
Collapse
Affiliation(s)
- Linli Qin
- Jiangsu Provincial Key Lab of Pulp and Paper Science and Technology, Nanjing Forestry University, Nanjing, Jiangsu Province 210037, China
| | - Yanchen Zhu
- Jiangsu Provincial Key Lab of Pulp and Paper Science and Technology, Nanjing Forestry University, Nanjing, Jiangsu Province 210037, China
| | - Haonan Zhang
- Jiangsu Provincial Key Lab of Pulp and Paper Science and Technology, Nanjing Forestry University, Nanjing, Jiangsu Province 210037, China
| | - Hao Ren
- Jiangsu Provincial Key Lab of Pulp and Paper Science and Technology, Nanjing Forestry University, Nanjing, Jiangsu Province 210037, China.
| | - Huamin Zhai
- Jiangsu Provincial Key Lab of Pulp and Paper Science and Technology, Nanjing Forestry University, Nanjing, Jiangsu Province 210037, China
| |
Collapse
|
17
|
Habibah T, Matonohová J, Kulhánek J, Fitzgerald U, Ingr M, Pravda M, Pandit A, Velebný V. In situ formed aldehyde-modified hyaluronic acid hydrogel with polyelectrolyte complexes of aldehyde-modified chondroitin sulfate and gelatin: An approach for minocycline delivery. Carbohydr Polym 2024; 343:122455. [PMID: 39174092 DOI: 10.1016/j.carbpol.2024.122455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 06/28/2024] [Accepted: 07/01/2024] [Indexed: 08/24/2024]
Abstract
Polysaccharides like hyaluronan (HA) and chondroitin sulfate (CS) are native of the brain's extracellular matrix crucial for myelination and brain maturation. Despite extensive research on HA and CS as drug delivery systems (DDS), their high water solubility limits their application as drug carriers. This study introduces an injectable DDS using aldehyde-modified hyaluronic acid (HAOX) hydrogel containing polyelectrolyte complexes (PEC) formed with calcium, gelatin, and either CS or aldehyde-modified CS (CSOX) to deliver minocycline for Multiple Sclerosis therapy. PECs with CSOX enable covalent crosslinking to HAOX, creating immobilized PECs (HAOX_PECOX), while those with CS remain unbound (HAOX_PECS). The in situ forming DDS can be administered via a 20 G needle, with rapid gelation preventing premature leakage. The system integrates into an implanted device for minocycline release through either Fickian or anomalous diffusion, depending on PEC immobilization. HAOX_PECOX reduced burst release by 88 %, with a duration of 127 h for 50 % release. The DDS exhibited an elastic modulus of 3800 Pa and a low swelling ratio (0-1 %), enabling precise control of minocycline release kinetics. Released minocycline reduced IL-6 secretion in the Whole Blood Monocytes Activation Test, suggesting that DDS formation may not alter the biological activity of the loaded drug.
Collapse
Affiliation(s)
- Tutut Habibah
- Contipro a.s. Dolní Dobrouč 401, Dolní Dobrouč, 56102, Czechia; Faculty of Technology, Tomas Bata University in Zlin, Vavrečkova, 5669, Czechia
| | - Jana Matonohová
- Contipro a.s. Dolní Dobrouč 401, Dolní Dobrouč, 56102, Czechia
| | | | - Una Fitzgerald
- CURAM, SFI Centre for Research on Biomedical Devices, Biomedical Engineering, University of Galway, Upper Newcastle, H91 W2TY, Ireland
| | - Marek Ingr
- Faculty of Technology, Tomas Bata University in Zlin, Vavrečkova, 5669, Czechia
| | - Martin Pravda
- Contipro a.s. Dolní Dobrouč 401, Dolní Dobrouč, 56102, Czechia.
| | - Abhay Pandit
- CURAM, SFI Centre for Research on Biomedical Devices, Biomedical Engineering, University of Galway, Upper Newcastle, H91 W2TY, Ireland
| | | |
Collapse
|
18
|
Nguyen HL, Holderbaum Do Amaral R, Lerouge S, De Roo AK, Zehtabi F, Vikkula M, Soulez G. Injectable chitosan hydrogel effectively controls lesion growth in a venous malformation murine model. Diagn Interv Imaging 2024; 105:430-438. [PMID: 39095271 DOI: 10.1016/j.diii.2024.07.004] [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: 02/20/2024] [Revised: 07/03/2024] [Accepted: 07/08/2024] [Indexed: 08/04/2024]
Abstract
PURPOSE The purpose of this study was to evaluate the safety and efficacy of intralesional injection of chitosan hydrogel (CH) combined with sodium tetradecyl sulfate (STS) to sclerose and embolize venous malformations (VMs) by comparison with 3% STS foam and placebo in a mouse model. MATERIALS AND METHODS Subcutaneous VMs were created by injecting HUVEC_TIE2-L914F cells, mixed with matrigel, into the back of athymic mice (Day [D] 0). After VM-like lesions were established at D10, 70 lesions were randomly assigned to one of six treatment groups (untreated, saline, 3% STS-foam, CH, 1% STS-CH, 3% STS-CH). For 3% STS-foam, the standard Tessari technique was performed. VMs were regularly evaluated every 2-3 days to measure lesion size until the time of collection at D30 (primary endpoint). At D30, VM lesions including the matrigel plugs were culled and evaluated by histological analysis to assess vessel size, chitosan distribution and endothelial expression. One-way analysis of variance (ANOVA) test was performed to compare quantitative variables with normal distribution, otherwise Kruskal-Wallis test followed by pairwise comparisons by a Wilcoxon rank sum test was performed. RESULTS All VMs were successfully punctured and injected. Six VMs injected with 3% STS-CH showed early skin ulceration with an extrusion of the matrigel plug and were excluded from final analysis. In the remaining 64 VMs, skin ulceration occurred on 26 plugs, resulting in the loss of three 3% STS-foam and one 1% STS-CH plugs. Both chitosan formulations effectively controlled growth of VMs by the end of follow-up compared to untreated or 3% STS-foam groups (P < 0.05). Vessel sizes were smaller with both CH formulations compared to untreated and saline groups (P < 0.05). Additionally, there were smaller vascular channels within the 1% STS-CH group compared to the 3% STS-foam group (P < 0.05). CONCLUSION Chitosan's ability to control the growth of VMs suggests a promising therapeutic effect that outperforms the gold standard (STS-foam) on several variables.
Collapse
Affiliation(s)
- Ha-Long Nguyen
- Human Molecular Genetics, de Duve Institute, University of Louvain, B-1200 Brussels, Belgium
| | - Ricardo Holderbaum Do Amaral
- Centre Hospitalier de l'Université de Montréal Research Center (CRCHUM), Montreal, QC, H2 × 0A9, Canada; Department of Radiology, Radiation Oncology and Nuclear Medicine, Université de Montreal, Montreal, QC, H3T 1J4, Canada
| | - Sophie Lerouge
- Centre Hospitalier de l'Université de Montréal Research Center (CRCHUM), Montreal, QC, H2 × 0A9, Canada; Department of Radiology, Radiation Oncology and Nuclear Medicine, Université de Montreal, Montreal, QC, H3T 1J4, Canada; Department of Mechanical Engineering, Ecole de Technologie Supérieure (ÉTS), Montreal, QC, H3C 1K3, Canada
| | - An-Katrien De Roo
- Department of Pathology, Saint Luc University Hospital, B-1200 Brussels, Belgium; Institute of Experimental and Clinical Research, University of Louvain, B-1348 Brussels, Belgium; Center for Vascular Anomalies, Member of VASCERN (European Reference Network on Rare Multisystemic Vascular Diseases) VASCA European Reference Centre Cliniques Universitaires Saint-Luc, B-1200 Brussels, Belgium
| | - Fatemeh Zehtabi
- Centre Hospitalier de l'Université de Montréal Research Center (CRCHUM), Montreal, QC, H2 × 0A9, Canada; Department of Mechanical Engineering, Ecole de Technologie Supérieure (ÉTS), Montreal, QC, H3C 1K3, Canada
| | - Miikka Vikkula
- Human Molecular Genetics, de Duve Institute, University of Louvain, B-1200 Brussels, Belgium; WELBIO Department, WEL Research Institute, B-1300 Wavre, Belgium
| | - Gilles Soulez
- Centre Hospitalier de l'Université de Montréal Research Center (CRCHUM), Montreal, QC, H2 × 0A9, Canada; Department of Radiology, Radiation Oncology and Nuclear Medicine, Université de Montreal, Montreal, QC, H3T 1J4, Canada.
| |
Collapse
|
19
|
Torkashvand A, Izadian A, Hajrasouliha A. Advances in ophthalmic therapeutic delivery: A comprehensive overview of present and future directions. Surv Ophthalmol 2024; 69:967-983. [PMID: 38986847 PMCID: PMC11392635 DOI: 10.1016/j.survophthal.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2023] [Revised: 06/26/2024] [Accepted: 07/01/2024] [Indexed: 07/12/2024]
Abstract
Ophthalmic treatment demands precision and consistency in delivering therapeutic agents over extended periods to address many conditions, from common eye disorders to complex diseases. This diversity necessitates a range of delivery strategies, each tailored to specific needs. We delve into various delivery cargos that are pivotal in ophthalmic care. These cargos encompass biodegradable implants that gradually release medication, nonbiodegradable implants for sustained drug delivery, refillable tools allowing flexibility in treatment, hydrogels capable of retaining substances while maintaining ocular comfort, and advanced nanotechnology devices that precisely target eye tissues. Within each cargo category, we explore cutting-edge research-level approaches and FDA-approved methods, providing a thorough overview of the current state of ophthalmic drug delivery. In particular, our focus on nanotechnology reveals the promising potential for gene delivery, cell therapy administration, and the implantation of active devices directly into the retina. These advancements hold the key to more effective, personalized, and minimally- invasive ophthalmic treatments, revolutionizing the field of eye care.
Collapse
Affiliation(s)
- Ali Torkashvand
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Afshin Izadian
- Electrical and Computer Engineering Technology, Purdue University, West Lafayette, IN, United States
| | - Amir Hajrasouliha
- Department of Ophthalmology, Indiana University School of Medicine, Indianapolis, IN, United States.
| |
Collapse
|
20
|
Liu Y, Liu T, Zhu Z, Xie L, Bai D, Liu T, Gu W, Li W, Shu Y, Zhang J. An advanced hydrogel dressing system with progressive delivery and layer-to-layer response for diabetic wound healing. Acta Biomater 2024:S1742-7061(24)00638-X. [PMID: 39486779 DOI: 10.1016/j.actbio.2024.10.046] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Revised: 10/27/2024] [Accepted: 10/29/2024] [Indexed: 11/04/2024]
Abstract
Wound healing in diabetic patients presents a significant challenge due to delayed inflammatory responses, which obstruct subsequent healing stages. In response, we have developed a progressive, layer-by-layer responsive hydrogel, specifically designed to meet the dynamic requirements of diabetic wounds throughout different healing phases. This hydrogel initiates with a glucose-responsive layer formed by boronate ester bonds between 4-arm-poly (ethylene glycol) succinimidyl glutarate (4arm-PEG-SG) and 3-aminophenylboronic acid. This configuration ensures precise control over the physicochemical properties, facilitating accurate drug release during the healing process. Furthermore, we have incorporated an active pharmaceutical ingredient ionic liquid (API) composed of diclofenac and L-carnitine. This combination effectively tackles the solubility and stability issues commonly associated with anti-inflammatory drugs. To further refine drug release, we integrated matrix metalloproteinase-9 (MMP-9)-sensitive gelatin microcapsules, ensuring a controlled release and preventing the abrupt, uneven drug distribution often seen in other systems. Our hydrogel's rheological properties closely resemble human skin, offering a more harmonious approach to diabetic wound healing. Overall, this progressive layer-by-layer responsive wound management system, which is a safe, efficient, and intelligent approach, holds significant potential for the clinical treatment of diabetic wounds. STATEMENT OF SIGNIFICANCE: The two main problems of diabetic wounds are the long-term infiltration of inflammation and the delayed repair process. In this experiment, a glucose-responsive hierarchical drug delivery system was designed to intelligently adjust gel properties to meet the needs of inflammation and repair stage of wound healing, accelerate the transformation of inflammation and repair stage, and accelerate the process of repair stage. In addition, in order to achieve accurate drug release in anti-inflammatory layer hydrogels and avoid sudden drug release due to poor solubility of anti-inflammatory small molecule drugs, we constructed a ionic liquid of active pharmaceutical ingredients (API-ILs) using diclofenac and L-carnitine as raw materials. It was wrapped in MMP-9 enzyme active gelatin microcapsule to construct a double-reaction anti-inflammatory layer gel to achieve accurate drug release. These findings highlight the potential of our system in treating diabetic wounds, providing a significant advance in wound treatment.
Collapse
Affiliation(s)
- Ying Liu
- State Key Laboratory of Advanced Welding and Joining, Research Centre of Printed Flexible Electronics, School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, PR China; Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, PR China; Changzhou Zhitai Shengchuang Technology Co., Ltd., Changzhou 213000, PR China
| | - Tianqi Liu
- State Key Laboratory of Advanced Welding and Joining, Research Centre of Printed Flexible Electronics, School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, PR China; Changzhou Zhitai Shengchuang Technology Co., Ltd., Changzhou 213000, PR China.
| | - Zhenye Zhu
- State Key Laboratory of Advanced Welding and Joining, Research Centre of Printed Flexible Electronics, School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, PR China
| | - Lin Xie
- State Key Laboratory of Advanced Welding and Joining, Research Centre of Printed Flexible Electronics, School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, PR China
| | - De Bai
- State Key Laboratory of Advanced Welding and Joining, Research Centre of Printed Flexible Electronics, School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, PR China
| | - Tonglin Liu
- Division of Biosciences, University College London, London WC1E 6BT, United Kingdom
| | - Wenting Gu
- Changzhou Zhitai Shengchuang Technology Co., Ltd., Changzhou 213000, PR China
| | - Wei Li
- Changzhou Zhitai Shengchuang Technology Co., Ltd., Changzhou 213000, PR China
| | - Yang Shu
- Department of Chemistry, College of Sciences, Northeastern University, Shenyang 110819, PR China.
| | - Jiaheng Zhang
- State Key Laboratory of Advanced Welding and Joining, Research Centre of Printed Flexible Electronics, School of Materials Science and Engineering, Harbin Institute of Technology (Shenzhen), Shenzhen 518055, PR China; Changzhou Zhitai Shengchuang Technology Co., Ltd., Changzhou 213000, PR China; Shenzhen Shinehigh Innovation Technology Co., Ltd., Shenzhen 518055, PR China.
| |
Collapse
|
21
|
Mace MAM, Reginatto CL, Silva VRJ, Pinheiro ACS, Silva LSD, Moura DJ, Fuentefria AM, Soares RMD. 3D Printable Alginate-Chitosan Hydrogel Loaded With Ketoconazole Exhibits Anticryptococcal Activity. Biopolymers 2024:e23638. [PMID: 39470150 DOI: 10.1002/bip.23638] [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: 04/16/2024] [Revised: 10/01/2024] [Accepted: 10/13/2024] [Indexed: 10/30/2024]
Abstract
Natural polymers have recently been investigated for various applications, such as 3D printing and healthcare, including treating infections. Among microbial infections, fungal diseases remain overlooked, with limited therapeutic options and high recurrence. Cutaneous cryptococcosis is an opportunistic fungal infection triggered by mechanical inoculation or hematogenous dissemination of the yeast that causes cryptococcal pneumonia and meningitis. Every year, Cryptococcus neoformans endanger the lives of immunosuppressed hosts, resulting in 180,000 deaths per year. Nonetheless, healthy individuals can also be affected by this fungal infection. Cryptococcosis has a restricted and expensive therapeutic regimen with no topical approach to skin manifestations. This study sought to create a 3D printable biodegradable polymeric hydrogel carrying ketoconazole, a low-cost antifungal drug with reported anticryptococcal activity. The developed hydrogel exhibited good 3D printability and rheological properties, including a pseudoplastic behavior. The FTIR spectra of cross-linked hydrogels revealed interactions between alginate and Ca+2, referred to as the egg-box model, indicated by the decrease in peaks at 1600 and 1410 cm-1. Furthermore, the hydrogel loaded with ketoconazole showed remarkable antifungal activity against C. neoformans strains indicated by inhibition zones, which cross-linking did not seem to affect its antifungal performance. The developed material remained structurally stable for up to 12 days (288 h) in swelling studies, and preliminary cytotoxicity performed with V79 cells indicates potential for in vivo studies and topical application.
Collapse
Affiliation(s)
- Manoela Almeida Martins Mace
- Programa de Pós-Graduação Em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Laboratório de Pesquisa Em Micologia Aplicada (LPMA), Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Laboratório de Biomateriais Poliméricos (Poli-BIO), Instituto de Química, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Camila Leites Reginatto
- Laboratório de Biomateriais Poliméricos (Poli-BIO), Instituto de Química, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Programa de Pós-Graduação Em Ciência dos Materiais, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Victória Rapack Jacinto Silva
- Laboratório de Genética Toxicológica, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
- Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Ana Carolina Silva Pinheiro
- Laboratório de Genética Toxicológica, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
- Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Laiane Souza da Silva
- Laboratório de Biomateriais Poliméricos (Poli-BIO), Instituto de Química, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Programa de Pós-Graduação Em Química, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Dinara Jaqueline Moura
- Laboratório de Genética Toxicológica, Universidade Federal de Ciências da Saúde de Porto Alegre (UFCSPA), Porto Alegre, Brazil
- Universidade Federal de Ciências da Saúde de Porto Alegre, Porto Alegre, Brazil
| | - Alexandre Meneghello Fuentefria
- Programa de Pós-Graduação Em Ciências Farmacêuticas, Universidade Federal do Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
- Laboratório de Pesquisa Em Micologia Aplicada (LPMA), Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Rosane Michele Duarte Soares
- Laboratório de Biomateriais Poliméricos (Poli-BIO), Instituto de Química, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Programa de Pós-Graduação Em Ciência dos Materiais, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
- Programa de Pós-Graduação Em Química, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
22
|
Dai C, Shi Z, Xu Y, Su L, Li X, Deng P, Wen H, Wang J, Ye Q, Han RPS, Liu Q. Wearable Multifunctional Hydrogel for Oral Microenvironment Visualized Sensing Coupled with Sonodynamic Bacterial Elimination and Tooth Whitening. Adv Healthc Mater 2024:e2401269. [PMID: 39468859 DOI: 10.1002/adhm.202401269] [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: 04/06/2024] [Revised: 10/14/2024] [Indexed: 10/30/2024]
Abstract
Bacterial-driven dental caries and tooth discoloration are growing concerns as the most common oral health problems. Current diagnostic methods and treatment strategies hardly allow simultaneous early detection and non-invasive treatment of these oral diseases. Herein, a wearable multifunctional double network hydrogel combined with polyaniline and barium titanate (PANI@BTO) nanoparticles is developed for oral microenvironment visualized sensing and sonodynamic therapy. Due to the colorimetric properties of polyaniline, the hydrogel displays a highly sensitive and selective response for visualized sensing of oral acidic microenvironment. Meanwhile, the barium titanate in the hydrogel efficiently generates reactive oxygen species (ROS) under ultrasound irradiation, realizing non-invasive treatment in the oral cavity. Through bacterial elimination experiments and tooth whitening studies, the hydrogel can achieve the dual effect of effectively inhibiting the growth of cariogenic bacteria and degrading tooth surface pigments. Owing to the visualized sensing of the oral acidic microenvironment and efficient sonodynamic therapy function, the proposed hydrogel system offers a solution for the prevention of caries and tooth whitening, which is promising in developing the biomedical system targeting the simultaneous sensing and therapy for oral diseases.
Collapse
Affiliation(s)
- Chaobo Dai
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, P. R. China
| | - Zhenghan Shi
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, P. R. China
| | - Yi Xu
- Stomatology Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, P. R. China
| | - Lingkai Su
- Stomatology Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, P. R. China
| | - Xin Li
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, P. R. China
| | - Peixue Deng
- Life Sciences Institute, Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, Guangxi, 530021, P.R. China
| | - Hao Wen
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, P. R. China
| | - Jiahao Wang
- Cancer Research Center, College of Computer Science, Jiangxi University of Chinese Medicine, Nanchang, 330004, P. R. China
| | - Qing Ye
- Cancer Research Center, College of Computer Science, Jiangxi University of Chinese Medicine, Nanchang, 330004, P. R. China
| | - Ray P S Han
- Cancer Research Center, College of Computer Science, Jiangxi University of Chinese Medicine, Nanchang, 330004, P. R. China
| | - Qingjun Liu
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, P. R. China
| |
Collapse
|
23
|
Deng P, Shi Z, Fang F, Xu Y, Zhou LA, Liu Y, Jin M, Chen T, Wang Y, Cao Y, Su L, Liang H, Liu Q. Wireless matrix metalloproteinase-9 sensing by smart wound dressing with controlled antibacterial nanoparticles release toward chronic wound management. Biosens Bioelectron 2024; 268:116860. [PMID: 39489012 DOI: 10.1016/j.bios.2024.116860] [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: 08/14/2024] [Revised: 10/12/2024] [Accepted: 10/16/2024] [Indexed: 11/05/2024]
Abstract
Chronic wounds cause serious health and economic burdens on patients and society. Herein, a wireless and flexible smart wound dressing was developed for matrix metalloproteinase-9 (MMP-9) monitoring and antimicrobial treatment toward chronic wound management. The highly sensitive radio frequency MMP-9 sensor was realized based on a bioresponsive hydrogel with the bioactive peptide sequences. Taking advantage of the flexible inductive-capacitive (LC) circuit and bioresponsive hydrogel, the wireless and wearable smart wound dressing offered an efficient strategy for in-situ wound analysis. Besides, the controlled release of silver nanoparticles (AgNPs) from the degradable hydrogel exhibited significant antimicrobial efficacy against typical bacteria in wound infection including Escherichia coli and Staphylococcus aureus. The analysis of MMP-9 in wound exudate from diabetic foot ulcer (DFU) patients demonstrated good accuracy cross-validated with gold-standard fluorescent measurements, providing great potential for personalized wound management.
Collapse
Affiliation(s)
- Peixue Deng
- Life Sciences Institute, Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, Guangxi, 530021, PR China; Taizhou Key Laboratory of Medical Devices and Advanced Materials, Research Institute of Zhejiang University, Taizhou, 318000, PR China
| | - Zhenghan Shi
- Taizhou Key Laboratory of Medical Devices and Advanced Materials, Research Institute of Zhejiang University, Taizhou, 318000, PR China; Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, PR China
| | - Feiyue Fang
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, PR China
| | - Yi Xu
- Stomatology Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, PR China
| | - Li-Ang Zhou
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, PR China
| | - Ye Liu
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, PR China
| | - Meng Jin
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, PR China
| | - Tao Chen
- Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, PR China
| | - Yuzhen Wang
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Yemin Cao
- Shanghai Traditional Chinese Medicine Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, PR China
| | - Lingkai Su
- Stomatology Hospital, School of Medicine, Zhejiang University, Hangzhou, 310016, PR China
| | - Hao Liang
- Life Sciences Institute, Guangxi Key Laboratory of AIDS Prevention and Treatment, Guangxi Medical University, Nanning, Guangxi, 530021, PR China.
| | - Qingjun Liu
- Taizhou Key Laboratory of Medical Devices and Advanced Materials, Research Institute of Zhejiang University, Taizhou, 318000, PR China; Biosensor National Special Laboratory, Key Laboratory for Biomedical Engineering of Education Ministry, Department of Biomedical Engineering, Zhejiang University, Hangzhou, 310027, PR China.
| |
Collapse
|
24
|
Meissner S, Lopez S, Rees S, O'Carroll S, Barker D, Harland B, Raos B, Svirskis D. Safe subdural administration and retention of a neurotrophin-3-delivering hydrogel in a rat model of spinal cord injury. Sci Rep 2024; 14:25424. [PMID: 39455822 PMCID: PMC11511924 DOI: 10.1038/s41598-024-77423-5] [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: 06/27/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024] Open
Abstract
Neurotrophic growth factor (GF) loaded hydrogels have shown promise as a treatment approach for spinal cord injury (SCI). However, SCI presents complex challenges for the direct administration of treatment due to the spinal cord's intricate anatomy and highly sensitive environment. Many current hydrogel administration approaches overlook this complexity, limiting their translational potential. To address this, we propose a novel intrathecal administration method using an in situ gelling, hyaluronic acid-modified heparin-poloxamer hydrogel loaded with neurotrophin-3 (NT-3) for the direct delivery of NT-3 to the spinal cord. We injected a NT-3 loaded hydrogel into the intrathecal space immediately after contusion SCI in Sprague Dawley (SpD) rats. Our results indicate that injecting the NT-3 loaded hydrogel into the intrathecal space was safe and that the gel was retained alongside the cord for at least one week. Additionally, no adverse effects were observed on rat behaviour. While functional improvement trends were noted, statistical significance was not reached, and immunohistochemistry results showed no significant difference between treatment groups. Overall, our findings suggest the feasibility, safety, and potential of the developed intrathecal administration technique for delivering diverse therapeutic molecules for SCI recovery.
Collapse
Affiliation(s)
- Svenja Meissner
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Grafton, 1023, Auckland, New Zealand
| | - Salvador Lopez
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Grafton, 1023, Auckland, New Zealand
| | - Shaun Rees
- School of Chemical Sciences, University of Auckland, Auckland, 1010, New Zealand
| | - Simon O'Carroll
- Department of Anatomy and Medical Imaging, School of Medical Sciences, Faculty of Medical and Health Sciences, University of Auckland, Grafton, 1023, Auckland, New Zealand
| | - David Barker
- School of Chemical Sciences, University of Auckland, Auckland, 1010, New Zealand
- The MacDiarmid Institute for Advanced Materials and Nanotechnology, Wellington, New Zealand
| | - Bruce Harland
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Grafton, 1023, Auckland, New Zealand
| | - Brad Raos
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Grafton, 1023, Auckland, New Zealand
| | - Darren Svirskis
- School of Pharmacy, Faculty of Medical and Health Sciences, University of Auckland, Grafton, 1023, Auckland, New Zealand.
- , Level 3, Building 505, 85 Park Road, Auckland, 1023, New Zealand.
| |
Collapse
|
25
|
Akbari A, Bahram M, Dadashi R, Ehsanimehr S. Sensitive and Selective Determination of Benzidine by Synthesized tragacanth-poly (Acrylic acid-co-acrylamide-GQD) Hydrogel Nanocomposite as a Highly Stable Fluorescent Probe. J Fluoresc 2024:10.1007/s10895-024-03996-z. [PMID: 39441255 DOI: 10.1007/s10895-024-03996-z] [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: 08/03/2024] [Accepted: 10/07/2024] [Indexed: 10/25/2024]
Abstract
Benzidine is known as a toxic and highly carcinogenic substance, so its determination is an essential issue. Until now, no effective and stable fluorescent probe based on hydrogel nanocomposite has been reported for the determination of this substance. In this work, for the first time, the synthesis and use of tragacanth-poly (acrylic acid-co-acrylamide-GQD) hydrogel nanocomposite (H-GQD) as a novel, high-stable, and selective fluorescence hydrogel nanocomposite for the identification of benzidine is reported. To achieve the maximum responsiveness of this hydrogel nanocomposite to determine benzidine, various parameters such as pH, ionic strength, hydrogel nanocomposite concentration, sensitivity, and selectivity were investigated. The results of the investigations showed that the synthesized H-GQD has excellent stability, selectivity, and linearity range of 0.3 - 12 ppm with a limit of detection of 0.098 ppm. The results of the investigation of real water samples showed that the H-GQD has excellent recovery in the range of 93.3 - 106.6%. Finally, we believe that this H-GQD as a new and highly stable fluorescent probe can be a starting point for its application in various fields and industries to identify benzidine in water samples.
Collapse
Affiliation(s)
- Azra Akbari
- Department of Analytical Chemistry, Faculty of Chemistry, Urmia University, Urmia, Iran
| | - Morteza Bahram
- Department of Analytical Chemistry, Faculty of Chemistry, Urmia University, Urmia, Iran.
| | - Reza Dadashi
- Department of Analytical Chemistry, Faculty of Chemistry, Urmia University, Urmia, Iran
| | - Sedigheh Ehsanimehr
- Department of Organic Chemistry, Faculty of Chemistry, Urmia University, Urmia, Iran
| |
Collapse
|
26
|
Faleye OO, Lee JH, Kim YG, Faleye OS, Lee J. Antibiofilm and antivirulence potentials of iodinated fmoc-phenylalanine against Staphylococcus aureus. Microb Pathog 2024; 197:107080. [PMID: 39454802 DOI: 10.1016/j.micpath.2024.107080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 10/21/2024] [Accepted: 10/22/2024] [Indexed: 10/28/2024]
Abstract
Staphylococcus aureus poses significant risks to public health due to its ability to form biofilm and produce virulence factors, contributing to the increase in antibiotic resistance and treatment complications. This emphasizes the urgent need for novel antimicrobial controls. Based on the premise that halogenation improves antimicrobial efficacy, this study investigated the ability of halogenated phenylalanine to effectively inhibit S. aureus biofilm formation and virulence activities. Among 29 halogenated compounds, Fmoc-4-iodo-phenylalanine (Fmoc-Iodo-Phe) displayed the highest antibiofilm effect against S. aureus, achieving 94.3 % reduction at 50 μg/mL. Microscopic studies confirmed its ability to prevent and disrupt mature biofilms. At 10 μg/mL, Fmoc-Iodo-Phe markedly inhibited virulence factors, such as cell surface hydrophobicity, hemolysin and slime production. It showed low propensity for resistance development and effectively inhibited biofilms formed by methicillin-resistant S. aureus (MRSA) and S. epidermidis, but was inactive against Gram-negative bacteria. Gene expression analysis complemented by molecular docking suggest that Fmoc-Iodo-Phe could target the AgrA quorum sensing cascade due to strong interactions with key residues at its DNA binding sites. Notably, it was non-cytotoxic in Caenorhabditis elegans model and satisfied drug-likeliness criteria based on ADMET prediction. Therefore, our findings position Fmoc-Iodo-Phe as a promising antimicrobial candidate against S. aureus infections, underscoring its potential as an alternative to traditional antibiotics.
Collapse
Affiliation(s)
| | - Jin-Hyung Lee
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan, 38541, Republic of Korea
| | - Yong-Guy Kim
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan, 38541, Republic of Korea
| | - Olajide Sunday Faleye
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan, 38541, Republic of Korea
| | - Jintae Lee
- School of Chemical Engineering, Yeungnam University, 280 Daehak-Ro, Gyeongsan, 38541, Republic of Korea.
| |
Collapse
|
27
|
Tang R, Zhang Z, Liu X, Zhu L, Xu Y, Chai R, Zhan W, Shen S, Liang G. Fibroblast Growth Factor Receptor 1-Specific Dehydrogelation to Release Its Inhibitor for Enhanced Lung Tumor Therapy. ACS NANO 2024; 18:29223-29232. [PMID: 39392940 DOI: 10.1021/acsnano.4c11548] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/13/2024]
Abstract
Fibroblast growth factor receptor 1 (FGFR1) is emerging as a promising molecular target of lung cancer, and various FGFR1 inhibitors have exhibited significant therapeutic effects on lung cancer in preclinical research. Due to their low targeting ability or bioavailability, direct administration of these inhibitors may cause side effects. Herein, a hydrogelator, Nap-Phe-Phe-Phe-Glu-Thr-Glu-Leu-Tyr-OH (Nap-Y), was rationally designed to coassemble with an FGFR1 inhibitor nintedanib (Nin) to form a peptide hydrogel Gel Y/Nin for localized administration and FGFR1-triggered release of Nin. Upon specific phosphorylation by FGFR1 overexpressed on lung cancer cells, Nap-Y in Gel Y/Nin is converted to the hydrophilic product Nap-Phe-Phe-Phe-Glu-Thr-Glu-Leu-Tyr(H2PO3)-OH (Nap-Yp), leading to dehydrogelation of the gel and subsequent Nin release. In vitro experiments demonstrate that the release of Nin in a sustained manner from Gel Y/Nin significantly suppresses the survival, migration, and invasion of A549 cells by inhibiting FGFR1 expression and its phosphorylation function on downstream signaling molecules. Nude mouse studies show that Gel Y/Nin exhibits enhanced therapeutic efficacy on lung tumor than free Nin. We anticipate that Gel Y/Nin will be utilized for lung cancer treatment in clinical settings in the near future.
Collapse
Affiliation(s)
- Runqun Tang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China
| | - Ziyi Zhang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China
| | - Xiaoyang Liu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China
| | - Liangxi Zhu
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China
| | - Yuting Xu
- Breast Surgery, Wenzhou Central Hospital, Wenzhou 325000, China
| | - Renjie Chai
- State Key Laboratory of Digital Medical Engineering, Department of Otolaryngology Head and Neck Surgery, Zhongda Hospital, Advanced Institute for Life and Health, Southeast University, Nanjing 210096, China
| | - Wenjun Zhan
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China
| | - Shurong Shen
- Breast Surgery, Wenzhou Central Hospital, Wenzhou 325000, China
| | - Gaolin Liang
- State Key Laboratory of Digital Medical Engineering, School of Biological Science and Medical Engineering, Southeast University, Nanjing 211189, China
| |
Collapse
|
28
|
Buwalda SJ. 'Click' hydrogels from renewable polysaccharide resources: Bioorthogonal chemistry for the preparation of alginate, cellulose and other plant-based networks with biomedical applications. Int J Biol Macromol 2024:136695. [PMID: 39447791 DOI: 10.1016/j.ijbiomac.2024.136695] [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: 07/05/2024] [Revised: 10/11/2024] [Accepted: 10/17/2024] [Indexed: 10/26/2024]
Abstract
Click chemistry refers to a class of highly selective reactions that occur in one pot, are not disturbed by water or oxygen, proceed quickly to high yield and generate only inoffensive byproducts. Since its first definition by Barry Sharpless in 2001, click chemistry has increasingly been used for the preparation of hydrogels, which are water-swollen polymer networks with numerous biomedical applications. Polysaccharides, which can be obtained from renewable resources including plants, have drawn growing attention for use in hydrogels due to the recent focus on the development of a sustainable society and the reduction of the environmental impact of the chemical industry. Importantly, plant-based polysaccharides are often bioresorbable and exhibit excellent biocompatibility and biomimicry. This comprehensive review describes the synthesis, characterization and biomedical applications of hydrogels which combine the renewable and biocompatible aspects of polysaccharides with the chemically and biomedically favorable characteristics of click crosslinking. The manuscript focuses on click hydrogels prepared from alginate and cellulose, the most widely used polysaccharides for this type of hydrogel, but also click hydrogels based on other plant-derived polymers (e.g. pectin) are discussed. In addition, the challenges are described that should be overcome to facilitate translation from academia to the clinic.
Collapse
Affiliation(s)
- Sytze J Buwalda
- MINES Paris, PSL University, Center for Materials Forming (CEMEF), UMR CNRS 7635, CS 10207, 06904 Sophia Antipolis, France.
| |
Collapse
|
29
|
Wu M, Zhao Y, Zhang C, Pu K. Advancing Proteolysis Targeting Chimera (PROTAC) Nanotechnology in Protein Homeostasis Reprograming for Disease Treatment. ACS NANO 2024; 18:28502-28530. [PMID: 39377250 DOI: 10.1021/acsnano.4c09800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/09/2024]
Abstract
Proteolysis targeting chimeras (PROTACs) represent a transformative class of therapeutic agents that leverage the intrinsic protein degradation machinery to modulate the hemostasis of key disease-associated proteins selectively. Although several PROTACs have been approved for clinical application, suboptimal therapeutic efficacy and potential adverse side effects remain challenging. Benefiting from the enhanced targeted delivery, reduced systemic toxicity, and improved bioavailability, nanomedicines can be tailored with precision to integrate with PROTACs which hold significant potential to facilitate PROTAC nanomedicines (nano-PROTACs) for clinical translation with enhanced efficacy and reduced side effects. In this review, we provide an overview of the recent progress in the convergence of nanotechnology with PROTAC design, leveraging the inherent properties of nanomaterials, such as lipids, polymers, inorganic nanoparticles, nanohydrogels, proteins, and nucleic acids, for precise PROTAC delivery. Additionally, we discuss the various categories of PROTAC targets and provide insights into their clinical translational potential, alongside the challenges that need to be addressed.
Collapse
Affiliation(s)
- Mengyao Wu
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Yilan Zhao
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Chi Zhang
- Hubei Key Laboratory of Bioinorganic Chemistry and Materia Medica, Hubei Engineering Research Center for Biomaterials and Medical Protective Materials, School of Chemistry and Chemical Engineering, Huazhong University of Science and Technology, Wuhan 430074, China
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, 70 Nanyang Drive, 637457, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, 59 Nanyang Drive, 636921, Singapore
| |
Collapse
|
30
|
Noor L, Hafeez A, Rahman MA, Vishwakarma KK, Kapoor A, Ara N, Aqeel R. Demystifying the Potential of Embelin-Loaded Nanoformulations: a Comprehensive Review. AAPS PharmSciTech 2024; 25:249. [PMID: 39433611 DOI: 10.1208/s12249-024-02968-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 10/01/2024] [Indexed: 10/23/2024] Open
Abstract
Phytoconstituent based therapies have the potential to reduce the adverse effects and enhance overall patient compliance for different diseased conditions. Embelin (EMB) is a natural compound extracted from Embelia ribes that has demonstrated high therapeutic potential, particularly as anti-inflammatory and anticancer therapeutic applications. However, its poor water solubility and low oral bioavailability limitations make it challenging to use in biomedical applications. Nanostructure-based novel formulations have shown the potential to improve physicochemical and biological characteristics of active pharmaceutical ingredients obtained from plants. Different nanoformulations that have been utilized to encapsulate/entrap EMB for various therapeutic applications are nanoliposomes, nanostructured lipid carriers, niosomes, polymeric nanoparticles, nanosuspensions, phytosomes, self nanoemulsifying drug delivery system, silver nanoparticles, microparticles, solid lipid nanoparticle, gold nanoparticles and nanomicelles. The common methods reported for the preparation of EMB nanoformulations are thin film hydration, nanoprecipitation, ethanol injection, emulsification followed by sonication. The size of nanoformulations ranged in between 50 and 345 nm. In this review, the mentioned EMB loaded nanocarriers are methodically discussed for size, shape, drug entrapment, zeta potential, in vitro release & permeation and in vivo studies. Potential of EMB with other drugs (dual drug approach) incorporated in nanocarriers are also discussed (physicochemical and preclinical characteristics). Patents related to EMB nanoformulations are also presented which showed the clinical translation of this bioactive for future utilization in different indications.
Collapse
Affiliation(s)
- Layba Noor
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India
| | - Abdul Hafeez
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India.
| | - Md Azizur Rahman
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India
| | | | - Archita Kapoor
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India
| | - Nargis Ara
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India
| | - Rabia Aqeel
- Faculty of Pharmacy, Integral University, Lucknow, 226026, India
| |
Collapse
|
31
|
Lara-Espinoza C, Rascón-Chu A, Micard V, Antoine-Assor C, Carvajal-Millan E, Troncoso-Rojas R, Ohlmaier-Delgadillo F, Brown-Bojorquez F. Covalent Pectin/Arabinoxylan Hydrogels: Rheological and Microstructural Characterization. Polymers (Basel) 2024; 16:2939. [PMID: 39458767 PMCID: PMC11510807 DOI: 10.3390/polym16202939] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 10/03/2024] [Accepted: 10/18/2024] [Indexed: 10/28/2024] Open
Abstract
This research aimed to evaluate the gelation process of ferulated pectin (FP) and ferulated arabinoxylan (AXF) in a new mixed hydrogel and determine its microstructural characteristics. FP from sugar beet (Beta vulgaris) and arabinoxylan from maize (Zea mays) bran were gelled via oxidative coupling using laccase as a crosslinking agent. The dynamic oscillatory rheology of the mixed hydrogel revealed a maximum storage modulus of 768 Pa after 60 min. The scanning electron microscopy images showed that mixed hydrogels possess a microstructure of imperfect honeycomb. The ferulic acid content of the mixed hydrogel was 3.73 mg/g, and ferulic acid dimer 8-5' was the most abundant. The presence of a trimer was also detected. This study reports the distribution and concentration of ferulic acid dimers, and the rheological and microstructural properties of a mixed hydrogel based on FP and AXF, which has promising features as a new covalent biopolymeric material.
Collapse
Affiliation(s)
- Claudia Lara-Espinoza
- Research Center for Food and Development, CIAD, A.C., Carretera Gustavo Enrique Astiazaran Rosas No. 46, Col. La Victoria, Hermosillo 83304, Sonora, Mexico; (C.L.-E.); (E.C.-M.)
| | - Agustín Rascón-Chu
- Research Center for Food and Development, CIAD, A.C., Carretera Gustavo Enrique Astiazaran Rosas No. 46, Col. La Victoria, Hermosillo 83304, Sonora, Mexico; (C.L.-E.); (E.C.-M.)
| | - Valérie Micard
- IATE, INRAE, Institut SupAgro, University Montpellier, 34000 Montpellier, France
| | - Carole Antoine-Assor
- IATE, INRAE, Institut SupAgro, University Montpellier, 34000 Montpellier, France
| | - Elizabeth Carvajal-Millan
- Research Center for Food and Development, CIAD, A.C., Carretera Gustavo Enrique Astiazaran Rosas No. 46, Col. La Victoria, Hermosillo 83304, Sonora, Mexico; (C.L.-E.); (E.C.-M.)
| | - Rosalba Troncoso-Rojas
- Research Center for Food and Development, CIAD, A.C., Carretera Gustavo Enrique Astiazaran Rosas No. 46, Col. La Victoria, Hermosillo 83304, Sonora, Mexico; (C.L.-E.); (E.C.-M.)
| | - Federico Ohlmaier-Delgadillo
- Research Center for Food and Development, CIAD, A.C., Carretera Gustavo Enrique Astiazaran Rosas No. 46, Col. La Victoria, Hermosillo 83304, Sonora, Mexico; (C.L.-E.); (E.C.-M.)
| | - Francisco Brown-Bojorquez
- Departamento de Investigación en Polímeros y Materiales, University of Sonora, Rosales y Blvd. Luis D. Colosio, Hermosillo 83000, Sonora, Mexico;
| |
Collapse
|
32
|
Rodrigues FAP, Oliveira CS, Sá SC, Tavaria FK, Lee SJ, Oliveira AL, Costa JB. Molecules in Motion: Unravelling the Dynamics of Vascularization Control in Tissue Engineering. Macromol Biosci 2024:e2400139. [PMID: 39422632 DOI: 10.1002/mabi.202400139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2024] [Revised: 08/14/2024] [Indexed: 10/19/2024]
Abstract
Significant progress has been made in tissue engineering (TE), aiming at providing personalized solutions and overcoming the current limitations of traditional tissue and organ transplantation. 3D bioprinting has emerged as a transformative technology in the field, able to mimic key properties of the natural architecture of the native tissues. However, most successes in the area are still limited to avascular or thin tissues due to the difficulties in controlling the vascularization of the engineered tissues. To address this issue, several molecules, biomaterials, and cells with pro- and anti-angiogenic potential have been intensively investigated. Furthermore, different bioreactors capable to provide a dynamic environment for in vitro vascularization control have been also explored. The present review summarizes the main molecules and TE strategies used to promote and inhibit vascularization in TE, as well as the techniques used to deliver them. Additionally, it also discusses the current challenges in 3D bioprinting and in tissue maturation to control in vitro/in vivo vascularization. Currently, this field of investigation is of utmost importance and may open doors for the design and development of more precise and controlled vascularization strategies in TE.
Collapse
Affiliation(s)
- Francisco A P Rodrigues
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto, 4169-005, Portugal
| | - Cláudia S Oliveira
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto, 4169-005, Portugal
| | - Simone C Sá
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto, 4169-005, Portugal
| | - Freni K Tavaria
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto, 4169-005, Portugal
| | - Sang Jin Lee
- Wake Forest Institute for Regenerative Medicine, Wake Forest University School of Medicine, Winston-Salem, NC, 27157, USA
| | - Ana L Oliveira
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto, 4169-005, Portugal
| | - João B Costa
- CBQF-Centro de Biotecnologia e Química Fina-Laboratório Associado, Universidade Católica Portuguesa, Escola Superior de Biotecnologia, Rua Diogo Botelho 1327, Porto, 4169-005, Portugal
| |
Collapse
|
33
|
Solanki R, Makwana N, Kumar R, Joshi M, Patel A, Bhatia D, Sahoo DK. Nanomedicines as a cutting-edge solution to combat antimicrobial resistance. RSC Adv 2024; 14:33568-33586. [PMID: 39439838 PMCID: PMC11495475 DOI: 10.1039/d4ra06117a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2024] [Accepted: 10/14/2024] [Indexed: 10/25/2024] Open
Abstract
Antimicrobial resistance (AMR) poses a critical threat to global public health, necessitating the development of novel strategies. AMR occurs when bacteria, viruses, fungi, and parasites evolve to resist antimicrobial drugs, making infections difficult to treat and increasing the risk of disease spread, severe illness, and death. Over 70% of infection-causing microorganisms are estimated to be resistant to one or several antimicrobial drugs. AMR mechanisms include efflux pumps, target modifications (e.g., mutations in penicillin-binding proteins (PBPs), ribosomal subunits, or DNA gyrase), drug hydrolysis by enzymes (e.g., β-lactamase), and membrane alterations that reduce the antibiotic's binding affinity and entry. Microbes also resist antimicrobials through peptidoglycan precursor modification, ribosomal subunit methylation, and alterations in metabolic enzymes. Rapid development of new strategies is essential to curb the spread of AMR and microbial infections. Nanomedicines, with their small size and unique physicochemical properties, offer a promising solution by overcoming drug resistance mechanisms such as reduced drug uptake, increased efflux, biofilm formation, and intracellular bacterial persistence. They enhance the therapeutic efficacy of antimicrobial agents, reduce toxicity, and tackle microbial resistance effectively. Various nanomaterials, including polymeric-based, lipid-based, metal nanoparticles, carbohydrate-derived, nucleic acid-based, and hydrogels, provide efficient solutions for AMR. This review addresses the epidemiology of microbial resistance, outlines key resistance mechanisms, and explores how nanomedicines overcome these barriers. In conclusion, nanomaterials represent a versatile and powerful approach to combating the current antimicrobial crisis.
Collapse
Affiliation(s)
- Raghu Solanki
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar Palaj Gujarat 382355 India
| | - Nilesh Makwana
- School of Life Sciences, Jawaharlal Nehru University New Delhi India
| | - Rahul Kumar
- Dr B. R. A. Institute Rotary Cancer Hospital, All India Institute of Medical Sciences New Delhi India
| | - Madhvi Joshi
- Gujarat Biotechnology Research Centre (GBRC) Gandhinagar Gujarat India
| | - Ashish Patel
- Department of Life Sciences, Hemchandracharya North Gujarat University Patan 384265 Gujarat India
| | - Dhiraj Bhatia
- Department of Biological Sciences and Engineering, Indian Institute of Technology Gandhinagar Palaj Gujarat 382355 India
| | - Dipak Kumar Sahoo
- Department of Veterinary Clinical Sciences, College of Veterinary Medicine, Iowa State University Ames IA USA
| |
Collapse
|
34
|
Pan S, Yuan H, Zhai Q, Zhang Y, He H, Yin T, Tang X, Gou J. The journey of nanoparticles in the abdominal cavity: Exploring their in vivo fate and impact factors. J Control Release 2024; 376:266-285. [PMID: 39396710 DOI: 10.1016/j.jconrel.2024.10.011] [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: 07/24/2024] [Revised: 10/03/2024] [Accepted: 10/05/2024] [Indexed: 10/15/2024]
Abstract
Peritoneal carcinomatosis (PC) is caused by metastasis of primary tumor cells from intra-abdominal organs to the peritoneal surface. Intraperitoneal (IP) chemotherapy allows close contact of high concentrations of therapeutic agents with cancer cells in the peritoneal cavity to prolong patient survival. However, conventional IP chemotherapy is prone to rapid elimination from the peritoneal cavity and lacks specificity towards cancer cells. To address these challenges, there is an imperative demand for exploiting novel drug delivery systems to enhance drug retention in the peritoneal cavity and target PC cells. Therefore, in this review, we first recapitulate the physiological structures and barriers associated with IP drug delivery, highlighting the in vivo fate of nanoparticles (NPs) after IP administration. Furthermore, the influence of physicochemical properties (particle size, charge, surface modification, and carrier composition) on the in vivo fate of NPs is discussed. Perspectives on the rational design of NPs for IP therapy and recent clinical progress are also provided.
Collapse
Affiliation(s)
- Shu Pan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Haoyang Yuan
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Qiyao Zhai
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Yu Zhang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Haibing He
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Tian Yin
- School of Functional Food and Wine, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China
| | - Xing Tang
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China.
| | - Jingxin Gou
- Department of Pharmaceutics, School of Pharmacy, Shenyang Pharmaceutical University, 103 Wenhua Road, Shenyang 110016, Liaoning, PR China.
| |
Collapse
|
35
|
Wu KY, Qian SY, Faucher A, Tran SD. Advancements in Hydrogels for Corneal Healing and Tissue Engineering. Gels 2024; 10:662. [PMID: 39451315 PMCID: PMC11507397 DOI: 10.3390/gels10100662] [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: 08/31/2024] [Revised: 10/08/2024] [Accepted: 10/14/2024] [Indexed: 10/26/2024] Open
Abstract
Hydrogels have garnered significant attention for their versatile applications across various fields, including biomedical engineering. This review delves into the fundamentals of hydrogels, exploring their definition, properties, and classification. Hydrogels, as three-dimensional networks of crosslinked polymers, possess tunable properties such as biocompatibility, mechanical strength, and hydrophilicity, making them ideal for medical applications. Uniquely, this article offers original insights into the application of hydrogels specifically for corneal tissue engineering, bridging a gap in current research. The review further examines the anatomical and functional complexities of the cornea, highlighting the challenges associated with corneal pathologies and the current reliance on donor corneas for transplantation. Considering the global shortage of donor corneas, this review discusses the potential of hydrogel-based materials in corneal tissue engineering. Emphasis is placed on the synthesis processes, including physical and chemical crosslinking, and the integration of bioactive molecules. Stimuli-responsive hydrogels, which react to environmental triggers, are identified as promising tools for drug delivery and tissue repair. Additionally, clinical applications of hydrogels in corneal pathologies are explored, showcasing their efficacy in various trials. Finally, the review addresses the challenges of regulatory approval and the need for further research to fully realize the potential of hydrogels in corneal tissue engineering, offering a promising outlook for future developments in this field.
Collapse
Affiliation(s)
- Kevin Y. Wu
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada; (K.Y.W.)
| | - Shu Yu Qian
- Faculty of Medicine, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada
| | - Anne Faucher
- Department of Surgery, Division of Ophthalmology, University of Sherbrooke, Sherbrooke, QC J1G 2E8, Canada; (K.Y.W.)
| | - Simon D. Tran
- Faculty of Dental Medicine and Oral Health Sciences, McGill University, Montreal, QC H3A 1G1, Canada
| |
Collapse
|
36
|
Haider M, Jagal J, Ali Alghamdi M, Haider Y, Hassan HAFM, Najm MB, Jayakuma MN, Ezzat H, Greish K. Erlotinib and curcumin-loaded nanoparticles embedded in thermosensitive chitosan hydrogels for enhanced treatment of head and neck cancer. Int J Pharm 2024; 666:124825. [PMID: 39401579 DOI: 10.1016/j.ijpharm.2024.124825] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2024] [Revised: 10/08/2024] [Accepted: 10/10/2024] [Indexed: 10/20/2024]
Abstract
Head and neck squamous cell carcinoma (HNSCC) remain a major oncological challenge with significant morbidity and mortality rates. Erlotinib (Er) and Curcumin (Cm) are potential therapeutic agents for HNSCC, yet they are hindered by poor solubility and bioavailability. This study explored the optimization of poly(lactic-co-glycolic acid) nanoparticles co-loaded with Er and Cm (Er/Cm-NP), prepared via a D-optimal response surface design-guided nanoprecipitation process. The optimized formulation, optEr/Cm-NP, was then incorporated into chitosan/β-glycerophosphate hydrogels (optEr/Cm-NP-HG) to create an injectable intratumoral (IT) nanocomposite hydrogel (HG) delivery system. Physicochemical properties of the formulations, including gelation time, injectability, mechanical strength and drug release profiles were assessed alongside hemolytic activity. Compared to optEr/Cm-NP alone, the NP-loaded HG formulation exhibited a more pronounced modulation effect, enabling sustained and controlled drug release. The cytotoxicity of the developed formulations was evaluated using the FaDu HNSCC cancer cell line. Both optEr/Cm-NP and optEr/Cm-NP-HG21 displayed enhanced cytotoxicity compared to free drugs. Confocal laser microscopy and flow cytometry confirmed superior cellular uptake of Er and Cm when delivered via NPs or NP-loaded HG. Furthermore, a significant increase in apoptotic cell death upon treatment with optEr/Cm-NP was observed, highlighting its potential for HNSCC therapy. In vivo studies conducted on a xenograft HNSCC mouse model revealed the significant capacity of the intratumorally-injected optEr/Cm-NP-HG21 formulation to retard the tumor growth. Conclusively, the results presented herein report the successful development of a nanocomposite HG system incorporating NPs co-loaded with Er and Cm that could be efficiently utilized in the treatment of HNSCC.
Collapse
Affiliation(s)
- Mohamed Haider
- Department of Pharmaceutics and Pharmaceutical Technology, College of Pharmacy, University of Sharjah, 27272 Sharjah, United Arab Emirates; Research Institute of Medical & Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates.
| | - Jayalakshmi Jagal
- Research Institute of Medical & Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates
| | - Maha Ali Alghamdi
- Department of Biotechnology, College of Science, Taif University, Taif 21974, Kingdom of Saudi Arabia; Department of Molecular Medicine, Princess Al-Jawhara Centre for Molecular Medicine, School of Medicine and Medical Sciences Arabian Gulf University, Manama 329, Bahrain
| | - Youssef Haider
- College of Engineering, Boston University, Boston, MA, USA
| | - Hatem A F M Hassan
- Medway School of Pharmacy, University of Kent, Central Avenue, Chatham Maritime, Canterbury ME4 4TB, UK; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Cairo University, 11562, Cairo, Egypt
| | - Muna B Najm
- Research Institute of Medical & Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates
| | - Manju N Jayakuma
- Research Institute of Medical & Health Sciences, University of Sharjah, 27272 Sharjah, United Arab Emirates
| | - Helal Ezzat
- Research Institute of Sciences and Engineering, University of Sharjah, 27272, Sharjah, United Arab Emirates; Civil Engineering Department, Delta Higher Institute for Engineering and Technology, Mansoura, Egypt
| | - Khaled Greish
- Department of Molecular Medicine, Princess Al-Jawhara Centre for Molecular Medicine, School of Medicine and Medical Sciences Arabian Gulf University, Manama 329, Bahrain.
| |
Collapse
|
37
|
Zhao N, Huang J, Pei J, Fu X, Ling B, Liu K. Modeling Mass Transport Dynamics in Deformable Hydrogels during Evaporation. J Phys Chem B 2024; 128:9798-9804. [PMID: 39324395 DOI: 10.1021/acs.jpcb.4c05885] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/27/2024]
Abstract
Hydrogels possess exceptional mechanical properties and biocompatibility, making them widely used in contemporary bioengineering. Specifically, in the development of wearable and implantable health monitoring devices as well as drug delivery systems, hydrogels are utilized to enable precise control over the transport of solutes. Nonetheless, predicting the distribution of substances within hydrogels still poses a significant challenge due to the complex interplay between the movement of water content, migration of solutes, and deformability of the hydrogel polymer network, which presents challenges to theoretical modeling. Our work introduces a numerical model that addresses the movement of water and solute within a flexible hydrogel, accounting for evaporation and/or moisture absorption at the boundary. The model solves for water saturation, solute concentration, and hydrogel deformation iteratively at each time step while computing the boundary movement velocity based on the transport process. By comparing the modeled results of geometry deformation and water and solute distributions during evaporation with our experiments, we demonstrate the accuracy and applicability of our proposed model. This capability to precisely analyze water and solute concentrations in deformable and nonuniform hydrogel environments paves the way for advancements in biosensing and drug delivery methods that rely on elastic porous materials.
Collapse
Affiliation(s)
- Na Zhao
- MOE Key Laboratory of Hydraulic Machinery Transients, School of Power and Mechanical Engineering, Wuhan University, Wuhan 430072, China
| | - Jun Huang
- MOE Key Laboratory of Hydraulic Machinery Transients, School of Power and Mechanical Engineering, Wuhan University, Wuhan 430072, China
| | - Junxian Pei
- State Key Laboratory of Hydraulics and Mountain River Engineering, College of Water Resource & Hydropower, Sichuan University, Chengdu, Sichuan 610065, China
| | - Xiangqian Fu
- MOE Key Laboratory of Hydraulic Machinery Transients, School of Power and Mechanical Engineering, Wuhan University, Wuhan 430072, China
| | - Bowen Ling
- Institute of Mechanics, Chinese Academy of Sciences, 15 Beisihuanxi Rd., Beijing 100190, China
- School of Engineering Science, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Kang Liu
- MOE Key Laboratory of Hydraulic Machinery Transients, School of Power and Mechanical Engineering, Wuhan University, Wuhan 430072, China
| |
Collapse
|
38
|
Yousfan A, Al Khatib AO, Salman AMH, Abu Elella MH, Barrett G, Michael N, Zariwala MG, Al-Obaidi H. Innovative Microencapsulation of Polymyxin B for Enhanced Antimicrobial Efficacy via Coated Spray Drying. Mol Pharm 2024. [PMID: 39378315 DOI: 10.1021/acs.molpharmaceut.4c00594] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2024]
Abstract
This study aims to develop an innovative microencapsulation method for coated Polymyxin B, utilizing various polysaccharides such as hydroxypropyl β-cyclodextrin, alginate, and chitosan, implemented through a three-fluid nozzle (3FN) spray drying process. High-performance liquid chromatography (HPLC) analysis revealed that formulations with a high ratio of sugar cage, hydroxypropyl β-cyclodextrin (HPβCD), and sodium alginate (coded as ALGHCDHPLPM) resulted in a notable 16-fold increase in Polymyxin B recovery compared to chitosan microparticles. Morphological assessments using fluorescence labeling confirmed successful microparticle formation with core/shell structures. Alginate-based formulations exhibited distinct layers, while chitosan formulations showed uniform fluorescence throughout the microparticles. Focused beam reflectance and histograms from fluorescence microscopic measurements provided insights into physical size analysis, indicating consistent sizes of 6.8 ± 1.2 μm. Fourier-transform infrared (FTIR) spectra unveiled hydrogen bonding between Polymyxin B and other components within the microparticle structures. The drug release study showed sodium alginate's sustained release capability, reaching 26 ± 3% compared to 94 ± 3% from the free solution at the 24 h time point. Furthermore, the antimicrobial properties of the prepared microparticles against two Gram-negative bacteria, Escherichia coli and Pseudomonas aeruginosa, were investigated. The influence of various key excipients on the minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) values was evaluated. Results demonstrated effective bactericidal effects of ALGHCDHPLPM against both E. coli and P. aeruginosa. Additionally, the antibiofilm assay highlighted the potential efficacy of ALGHCDHPLPM against the biofilm viability of E. coli and P. aeruginosa, with concentrations ranging from 3.9 to 500 μg/m. This signifies a significant advancement in antimicrobial drug delivery systems, promising improved precision and efficacy in combating bacterial infections.
Collapse
Affiliation(s)
- Amal Yousfan
- School of Pharmacy, University of Reading, Reading RG6 6AD, U.K
| | | | - Afrah M H Salman
- School of Biological Sciences, University of Reading, Reading RG6 6AD, U.K
- College of Pharmacy, Pharmacology and Toxicology Department, Mustansiriyha University, Baghdad 14132, Iraq
| | | | - Glyn Barrett
- School of Biological Sciences, University of Reading, Reading RG6 6AD, U.K
| | - Nicholas Michael
- Chemical Analysis Facility, University of Reading, Reading RG6 6AD, U.K
| | - Mohammed Gulrez Zariwala
- Centre for Nutraceuticals, School of Life Sciences, University of Westminster, 115 New, Cavendish Street, London W1W 6UW, U.K
| | | |
Collapse
|
39
|
Yang T, Li D, Luo Z, Wang J, Xiao F, Xu Y, Lin X. Space-Confined Amplification for In Situ Imaging of Single Nucleic Acid and Single Pathogen on Biological Samples. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2407055. [PMID: 39373849 DOI: 10.1002/advs.202407055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 09/20/2024] [Indexed: 10/08/2024]
Abstract
Direct in situ imaging of nucleic acids on biological samples is advantageous for rapid analysis without DNA extraction. However, traditional nucleic acid amplification in aqueous solutions tends to lose spatial information because of the high mobility of molecules. Similar to a cellular matrix, hydrogels with biomimetic 3D nanoconfined spaces can limit the free diffusion of nucleic acids, thereby allowing for ultrafast in situ enzymatic reactions. In this study, hydrogel-based in situ space-confined interfacial amplification (iSCIA) is developed for direct imaging of single nucleic acid and single pathogen on biological samples without formaldehyde fixation. With a polyethylene glycol hydrogel coating, nucleic acids on the sample are nanoconfined with restricted movement, while in situ amplification can be successfully performed. As a result, the nucleic acids are lighted-up on the large-scale surface in 20 min, with a detection limit as low as 1 copy/10 cm2. Multiplex imaging with a deep learning model is also established to automatically analyze multiple targets. Furthermore, the iSCIA imaging of pathogens on plant leaves and food is successfully used to monitor plant health and food safety. The proposed technique, a rapid and flexible system for in situ imaging, has great potential for food, environmental, and clinical applications.
Collapse
Affiliation(s)
- Tao Yang
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Dong Li
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
- The Rural Development Academy, Zhejiang University, Hangzhou, 310058, China
| | - Zisheng Luo
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Jingjing Wang
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Fangbin Xiao
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Yanqun Xu
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
| | - Xingyu Lin
- College of Biosystems Engineering and Food Science, Zhejiang University, Hangzhou, 310058, China
- State Key Laboratory of Fluid Power and Mechatronic Systems, Zhejiang University, Hangzhou, 310058, China
| |
Collapse
|
40
|
Najm A, Moldoveanu ET, Niculescu AG, Grumezescu AM, Beuran M, Gaspar BS. Advancements in Drug Delivery Systems for the Treatment of Sarcopenia: An Updated Overview. Int J Mol Sci 2024; 25:10766. [PMID: 39409095 PMCID: PMC11476378 DOI: 10.3390/ijms251910766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 10/04/2024] [Accepted: 10/04/2024] [Indexed: 10/20/2024] Open
Abstract
Since sarcopenia is a progressive condition that leads to decreased muscle mass and function, especially in elderly people, it is a public health problem that requires attention from researchers. This review aims to highlight drug delivery systems that have a high and efficient therapeutic potential for sarcopenia. Current as well as future research needs to consider the barriers encountered in the realization of delivery systems, such as the route of administration, the interaction of the systems with the aggressive environment of the human body, the efficient delivery and loading of the systems with therapeutic agents, and the targeted delivery of therapeutic agents into the muscle tissue without creating undesirable adverse effects. Thus, this paper sets the framework of existing drug delivery possibilities for the treatment of sarcopenia, serving as an inception point for future interdisciplinary studies.
Collapse
Affiliation(s)
- Alfred Najm
- Department of Surgery, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (A.N.); (M.B.); (B.S.G.)
- Emergency Hospital Floreasca Bucharest, 014461 Bucharest, Romania
| | - Elena-Theodora Moldoveanu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania; (E.-T.M.); (A.-G.N.)
| | - Adelina-Gabriela Niculescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania; (E.-T.M.); (A.-G.N.)
- Romania Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050657 Bucharest, Romania
| | - Alexandru Mihai Grumezescu
- Department of Science and Engineering of Oxide Materials and Nanomaterials, National University of Science and Technology Politehnica Bucharest, 011061 Bucharest, Romania; (E.-T.M.); (A.-G.N.)
- Romania Research Institute of the University of Bucharest—ICUB, University of Bucharest, 050657 Bucharest, Romania
| | - Mircea Beuran
- Department of Surgery, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (A.N.); (M.B.); (B.S.G.)
- Emergency Hospital Floreasca Bucharest, 014461 Bucharest, Romania
| | - Bogdan Severus Gaspar
- Department of Surgery, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (A.N.); (M.B.); (B.S.G.)
- Emergency Hospital Floreasca Bucharest, 014461 Bucharest, Romania
| |
Collapse
|
41
|
Rafati N, Zarepour A, Bigham A, Khosravi A, Naderi-Manesh H, Iravani S, Zarrabi A. Nanosystems for targeted drug Delivery: Innovations and challenges in overcoming the Blood-Brain barrier for neurodegenerative disease and cancer therapy. Int J Pharm 2024; 666:124800. [PMID: 39374818 DOI: 10.1016/j.ijpharm.2024.124800] [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: 07/18/2024] [Revised: 09/30/2024] [Accepted: 10/02/2024] [Indexed: 10/09/2024]
Abstract
The evolution of sophisticated nanosystems has revolutionized biomedicine, notably in treating neurodegenerative diseases and cancer. These systems show potential in delivering medication precisely to affected tissues, improving treatment effectiveness while minimizing side effects. Nevertheless, a major hurdle in targeted drug delivery is breaching the blood-brain barrier (BBB), a selective shield separating the bloodstream from the brain and spinal cord. The tight junctions between endothelial cells in brain capillaries create a formidable physical barrier, alongside efflux transporters that expel harmful molecules. This presents a notable challenge for brain drug delivery. Nanosystems present distinct advantages in overcoming BBB challenges, offering enhanced drug efficacy, reduced side effects, improved stability, and controlled release. Despite their promise, challenges persist, such as the BBB's regional variability hindering uniform drug distribution. Efflux transporters can also limit therapeutic agent efficacy, while nanosystem toxicity necessitates rigorous safety evaluations. Understanding the long-term impact of nanomaterials on the brain remains crucial. Additionally, addressing nanosystem scalability, cost-effectiveness, and safety profiles is vital for widespread clinical implementation. This review delves into the advancements and obstacles of advanced nanosystems in targeted drug delivery for neurodegenerative diseases and cancer therapy, with a focus on overcoming the BBB.
Collapse
Affiliation(s)
- Nesa Rafati
- Department of Nanobiotechnology, Faculty of Biological Science, Tarbiat Modares University, 14115-154, Tehran, Iran
| | - Atefeh Zarepour
- Department of Research Analytics, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai 600 077, India
| | - Ashkan Bigham
- Institute of Polymers, Composites, and Biomaterials, National Research Council (IPCB-CNR), Naples 80125, Italy; Department of Chemical, Materials and Production Engineering, University of Naples Federico II, Piazzale V. Tecchio 80, 80125 Naples, Italy
| | - Arezoo Khosravi
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, Istanbul Okan University, Istanbul 34959, Turkiye
| | - Hossein Naderi-Manesh
- Department of Nanobiotechnology, Faculty of Biological Science, Tarbiat Modares University, 14115-154, Tehran, Iran; Departments of Biophysics, Faculty of Biological Science, Tarbiat Modares University, 14115-154, Tehran, Iran.
| | - Siavash Iravani
- Independent Researcher, W Nazar ST, Boostan Ave, Isfahan, Iran.
| | - Ali Zarrabi
- Department of Biomedical Engineering, Faculty of Engineering and Natural Sciences, Istinye University, Istanbul 34396, Turkiye; Graduate School of Biotechnology and Bioengineering, Yuan Ze University, Taoyuan 320315, Taiwan.
| |
Collapse
|
42
|
Zhang S, Zhong R, Younis MR, He H, Xu H, Li G, Yang R, Lui S, Wang Y, Wu M. Hydrogel Applications in the Diagnosis and Treatment of Glioblastoma. ACS APPLIED MATERIALS & INTERFACES 2024. [PMID: 39366948 DOI: 10.1021/acsami.4c11855] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/06/2024]
Abstract
Glioblastoma multiforme (GBM), a common malignant neurological tumor, has boundaries indistinguishable from those of normal tissue, making complete surgical removal ineffective. The blood-brain barrier (BBB) further impedes the efficacy of radiotherapy and chemotherapy, leading to suboptimal treatment outcomes and a heightened probability of recurrence. Hydrogels offer multiple advantages for GBM diagnosis and treatment, including overcoming the BBB for improved drug delivery, controlled drug release for long-term efficacy, and enhanced relaxation properties of magnetic resonance imaging (MRI) contrast agents. Hydrogels, with their excellent biocompatibility and customizability, can mimic the in vivo microenvironment, support tumor cell culture, enable drug screening, and facilitate the study of tumor invasion and metastasis. This paper reviews the classification of hydrogels and recent research for the diagnosis and treatment of GBM, including their applications as cell culture platforms and drugs including imaging contrast agents carriers. The mechanisms of drug release from hydrogels and methods to monitor the activity of hydrogel-loaded drugs are also discussed. This review is intended to facilitate a more comprehensive understanding of the current state of GBM research. It offers insights into the design of integrated hydrogel-based GBM diagnosis and treatment with the objective of achieving the desired therapeutic effect and improving the prognosis of GBM.
Collapse
Affiliation(s)
- Shuaimei Zhang
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Renming Zhong
- Radiotherapy Physics & Technology Center, Cancer Center, West China Hospital, Chengdu, Sichuan 610041, P. R. China
| | - Muhammad Rizwan Younis
- Department of Chemical and Biomolecular Engineering, Samueli School of Engineering, University of California at Los Angeles, Los Angeles, California 90095, United States
| | - Hualong He
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Hong Xu
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Gaocan Li
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Ruiyan Yang
- Department of Biology, Macalester College, Saint Paul, Minnesota 55105, United States
| | - Su Lui
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Yunbing Wang
- National Engineering Research Center for Biomaterials, Sichuan University, Chengdu, Sichuan 610041, P. R. China
| | - Min Wu
- Department of Radiology and Huaxi MR Research Center (HMRRC), Functional and Molecular Imaging Key Laboratory of Sichuan Province, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, P. R. China
- Research Unit of Psychoradiology, Chinese Academy of Medical Sciences, Chengdu, Sichuan 610041, P. R. China
| |
Collapse
|
43
|
Ristić I, Nikolić L, Cakić S, Nikolić V, Tanasić J, Zvezdanović J, Krstić M. Eco-Friendly Microwave Synthesis of Sodium Alginate-Chitosan Hydrogels for Effective Curcumin Delivery and Controlled Release. Gels 2024; 10:637. [PMID: 39451290 PMCID: PMC11507994 DOI: 10.3390/gels10100637] [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: 08/08/2024] [Revised: 09/03/2024] [Accepted: 09/12/2024] [Indexed: 10/26/2024] Open
Abstract
In this study, we developed sodium alginate-chitosan hydrogels using a microwave-assisted synthesis method, aligning with green chemistry principles for enhanced sustainability. This eco-friendly approach minimizes chemical use and waste while boosting efficiency. A curcumin:2-hydroxypropyl-β-cyclodextrin complex was incorporated into the hydrogels, significantly increasing the solubility and bioavailability of curcumin. Fourier Transform Infrared Spectroscopy (FTIR) analysis confirmed the structure and successful incorporation of curcumin, in both its pure and complexed forms, into the polymer matrix. Differential scanning calorimetry revealed distinct thermal transitions influenced by the hydrogel composition and physical cross-linking. Hydrogels with higher alginate content had higher swelling ratios (338%), while those with more chitosan showed the lowest swelling ratios (254%). Scanning Electron Microscopy (SEM) micrographs showed a porous structure as well as successful incorporation of curcumin or its complex. Curcumin release studies indicated varying releasing rates between its pure and complexed forms. The chitosan-dominant hydrogel exhibited the slowest release rate of pure curcumin, while the alginate-dominant hydrogel exhibited the fastest. Conversely, for curcumin from the inclusion complex, a higher chitosan proportion led to the fastest release rate, while a higher alginate proportion resulted in the slowest. This study demonstrates that the form of curcumin incorporation and gel matrix composition critically influence the release profile. Our findings offer valuable insights for designing effective curcumin delivery systems, representing a significant advancement in biodegradable and sustainable drug delivery technologies.
Collapse
Affiliation(s)
- Ivan Ristić
- Faculty of Technology Novi Sad, University of Novi Sad, Bulevar cara Lazara 1, 21000 Novi Sad, Serbia;
| | - Ljubiša Nikolić
- Faculty of Technology, University of Niš, Bulevar Oslobodjenja 124, 16000 Leskovac, Serbia; (L.N.); (S.C.); (V.N.); (J.Z.)
| | - Suzana Cakić
- Faculty of Technology, University of Niš, Bulevar Oslobodjenja 124, 16000 Leskovac, Serbia; (L.N.); (S.C.); (V.N.); (J.Z.)
| | - Vesna Nikolić
- Faculty of Technology, University of Niš, Bulevar Oslobodjenja 124, 16000 Leskovac, Serbia; (L.N.); (S.C.); (V.N.); (J.Z.)
| | - Jelena Tanasić
- Faculty of Technology Novi Sad, University of Novi Sad, Bulevar cara Lazara 1, 21000 Novi Sad, Serbia;
| | - Jelena Zvezdanović
- Faculty of Technology, University of Niš, Bulevar Oslobodjenja 124, 16000 Leskovac, Serbia; (L.N.); (S.C.); (V.N.); (J.Z.)
| | - Marija Krstić
- Faculty of Technology Novi Sad, University of Novi Sad, Bulevar cara Lazara 1, 21000 Novi Sad, Serbia;
| |
Collapse
|
44
|
Nawrotek K, Chyb M, Gatkowska J, Rudnicka K, Michlewska S, Jóźwiak P. Effect of sodium L-lactate on bioactive properties of chitosan-hydroxyapatite/polycaprolactone conduits for peripheral nerve tissue engineering. Int J Biol Macromol 2024; 281:136254. [PMID: 39366606 DOI: 10.1016/j.ijbiomac.2024.136254] [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: 02/22/2024] [Revised: 09/26/2024] [Accepted: 10/01/2024] [Indexed: 10/06/2024]
Abstract
Biomaterials and synthetic polymers have been widely used to replicate the regenerative microenvironment of the peripheral nervous system. Chitosan-based conduits have shown promise in the regeneration of nerve injuries. However, to mimic the regenerative microenvironment, the scaffold structure should possess bioactive properties. This can be achieved by the incorporation of biomolecules (e.g., proteins, peptides) or trophic factors that should preferably be aligned and/or released with controlled kinetics to activate the process of positive axon chemotaxis. In this study, sodium L-lactate has been used to enhance the bioactive properties of chitosan-hydroxyapatite/polycaprolactone electrodeposits. Next, two methods have been developed to incorporate NGF-loaded microspheres - Method 1 involves entrapment and co-deposition of NGF-loaded microspheres, while Method 2 is based on absorption of NGF-loaded microspheres. The study shows that modification of chitosan-hydroxyapatite/polycaprolactone conduits by sodium L-lactate significantly improves their bioactive, biological, and physicochemical properties. The obtained implants are cytocompatible, enhancing the neurite regeneration process by stimulating its elongation. The absorption of NGF-loaded microspheres into the conduit structure may be considered more favorable for the stimulation of axonal elongation compared to entrapment, as it allows for trophic factor dose-dependent controlled release. The developed conduits possess properties essential for the successful treatment of peripheral nerve discontinuities.
Collapse
Affiliation(s)
- Katarzyna Nawrotek
- Lodz University of Technology, Faculty of Process and Environmental Engineering, Department of Environmental Engineering, Wolczanska 213, 93-005 Lodz, Poland; Lodz University of Technology, International Centre for Research on Innovative Bio-based Materials, 2/22 Stefanowskiego, 90-537, Poland; Warsaw University of Technology, Centre for Advanced Materials and Technology (CEZAMAT), 19 Poleczki, 02-822 Warsaw, Poland.
| | - Maciej Chyb
- University of Lodz, Faculty of Biology and Environmental Protection, Department of Molecular Microbiology, 12/16 Banacha, 90-237 Lodz, Poland; Bio-Med-Chem Doctoral School of the University of Lodz and Lodz Institutes of the Polish Academy of Sciences Banacha 12/16, 90-237 Lodz, Poland.
| | - Justyna Gatkowska
- University of Lodz, Faculty of Biology and Environmental Protection, Department of Molecular Microbiology, 12/16 Banacha, 90-237 Lodz, Poland.
| | - Karolina Rudnicka
- University of Lodz, Faculty of Biology and Environmental Protection, Department of Immunology and Infectious Biology, 90-237 Lodz, Poland.
| | - Sylwia Michlewska
- University of Lodz, Faculty of Biology and Environmental Protection, Laboratory of Microscopic Imaging and Specialized Biological Techniques, Banacha 12/16, 90-237 Lodz, Poland.
| | - Piotr Jóźwiak
- University of Lodz, Faculty of Biology and Environmental Protection, Department of Invertebrates Zoology and Hydrobiology, Banacha 12/16, 90-324 Lodz, Poland.
| |
Collapse
|
45
|
Naaz A, Turnquist HR, Gorantla VS, Little SR. Drug delivery strategies for local immunomodulation in transplantation: Bridging the translational gap. Adv Drug Deliv Rev 2024; 213:115429. [PMID: 39142608 DOI: 10.1016/j.addr.2024.115429] [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: 02/15/2024] [Revised: 08/07/2024] [Accepted: 08/11/2024] [Indexed: 08/16/2024]
Abstract
Drug delivery strategies for local immunomodulation hold tremendous promise compared to current clinical gold-standard systemic immunosuppression as they could improve the benefit to risk ratio of life-saving or life-enhancing transplants. Such strategies have facilitated prolonged graft survival in animal models at lower drug doses while minimizing off-target effects. Despite the promising outcomes in preclinical animal studies, progression of these strategies to clinical trials has faced challenges. A comprehensive understanding of the translational barriers is a critical first step towards clinical validation of effective immunomodulatory drug delivery protocols proven for safety and tolerability in pre-clinical animal models. This review overviews the current state-of-the-art in local immunomodulatory strategies for transplantation and outlines the key challenges hindering their clinical translation.
Collapse
Affiliation(s)
- Afsana Naaz
- Department of Chemical Engineering, University of Pittsburgh, Pittsburgh, PA 15261, United States; Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, 15213, United States.
| | - Heth R Turnquist
- Thomas E. Starzl Transplantation Institute, University of Pittsburgh, Pittsburgh, PA, 15213, United States; Department of Surgery, University of Pittsburgh, Pittsburgh, PA, 15213, United States; Department of Immunology, University of Pittsburgh, Pittsburgh, PA, 15213, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, United States.
| | - Vijay S Gorantla
- McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, United States; Departments of Surgery, Ophthalmology and Bioengineering, Wake Forest School of Medicine, Wake Forest Institute of Regenerative Medicine, Winston Salem, NC, 27101, United States.
| | - Steven R Little
- Department of Chemical Engineering, University of Pittsburgh, Pittsburgh, PA 15261, United States; Department of Surgery, University of Pittsburgh, Pittsburgh, PA, 15213, United States; Department of Immunology, University of Pittsburgh, Pittsburgh, PA, 15213, United States; McGowan Institute for Regenerative Medicine, University of Pittsburgh, Pittsburgh, PA, 15219, United States; Department of Bioengineering, University of Pittsburgh, Pittsburgh, PA 15261, United States; Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA 15213, United States; Department of Pharmaceutical Sciences, University of Pittsburgh, Pittsburgh, PA 15261, United States.
| |
Collapse
|
46
|
Cui Z, Wang L, Liu W, Xu D, Zhang T, Ma B, Zhang K, Yuan L, Bing Z, Liu J, Liu B, Wu W, Tian L. Imageable Brachytherapy with Chelator-Free Radiolabeling Hydrogel. Adv Healthc Mater 2024; 13:e2401438. [PMID: 38744050 DOI: 10.1002/adhm.202401438] [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: 04/19/2024] [Revised: 05/09/2024] [Indexed: 05/16/2024]
Abstract
Brachytherapy stands as an essential clinical approach for combating locally advanced tumors. Here, an injectable brachytherapy hydrogel is developed for the treatment of both local and metastatic tumor. Fe-tannins nanoparticles are efficiently and stably radiolabeled with clinical used therapeutic radionuclides (such as 131I, 90Y, 177Lu, and 225Ac) without a chelator, and then chemically cross-linked with 4-armPEG-SH to form brachytherapy hydrogel. Upon intratumoral administration, magnetic resonance imaging (MRI) signal from ferric ions embedded within the hydrogel directly correlates with the retention dosage of radionuclides, which can real-time monitor radionuclides emitting short-range rays in vivo without penetration limitation during brachytherapy. The hydrogel's design ensures the long-term tumor retention of therapeutic radionuclides, leading to the effective eradication of local tumor. Furthermore, the radiolabeled hydrogel is integrated with an adjuvant to synergize with immune checkpoint blocking therapy, thereby activating potent anti-tumor immune responses and inhibiting metastatic tumor growth. Therefore, this work presents an imageable brachytherapy hydrogel for real-time monitoring therapeutic process, and expands the indications of brachytherapy from treatment of localized tumors to metastatic tumors.
Collapse
Affiliation(s)
- Zhencun Cui
- MOE Frontiers Science Center for Rare Isotopes, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China
- School of Nuclear Science and Technology, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China
- Department of Nuclear Medicine, Second Hospital of Lanzhou University, 82 Cuiying Gate, Lanzhou, 730000, China
| | - Liqin Wang
- Department of Nuclear Medicine, Second Hospital of Lanzhou University, 82 Cuiying Gate, Lanzhou, 730000, China
| | - Wei Liu
- MOE Frontiers Science Center for Rare Isotopes, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China
- School of Nuclear Science and Technology, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China
| | - Dan Xu
- School of Nuclear Science and Technology, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou, 730000, China
| | - Taofeng Zhang
- School of Life Science and Engineering, Lanzhou University of Technology, 287 Langongping Road, Lanzhou, 730000, China
| | - Baoliang Ma
- Department of Nuclear Medicine, Second Hospital of Lanzhou University, 82 Cuiying Gate, Lanzhou, 730000, China
| | - Kai Zhang
- Department of Nuclear Medicine, Second Hospital of Lanzhou University, 82 Cuiying Gate, Lanzhou, 730000, China
| | - Lingyan Yuan
- Key Laboratory of Heavy Ion Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou, 730000, China
| | - Zhitong Bing
- Key Laboratory of Heavy Ion Radiation Medicine, Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou, 730000, China
| | - Jiangyan Liu
- Department of Nuclear Medicine, Second Hospital of Lanzhou University, 82 Cuiying Gate, Lanzhou, 730000, China
| | - Bin Liu
- School of Nuclear Science and Technology, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China
- School of Stomatology, Lanzhou University, 199 Donggang West Road, Lanzhou, 730000, China
| | - Wangsuo Wu
- MOE Frontiers Science Center for Rare Isotopes, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China
- School of Nuclear Science and Technology, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China
| | - Longlong Tian
- MOE Frontiers Science Center for Rare Isotopes, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China
- School of Nuclear Science and Technology, Lanzhou University, 222 Tianshui South Road, Lanzhou, 730000, China
| |
Collapse
|
47
|
Luo Q, Li Z, Liu B, Ding J. Hydrogel formulations for orthotopic treatment of myocardial infarction. Expert Opin Drug Deliv 2024; 21:1463-1478. [PMID: 39323051 DOI: 10.1080/17425247.2024.2409906] [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: 07/25/2024] [Revised: 08/25/2024] [Accepted: 09/24/2024] [Indexed: 09/27/2024]
Abstract
INTRODUCTION Myocardial infarction (MI) causes extensive structural and functional damage to the cardiac tissue due to the significant loss of cardiomyocytes. Early reperfusion is the standard treatment strategy for acute MI, but it is associated with adverse effects. Additionally, current therapies to alleviate pathological changes post-MI are not effective. Subsequent pathological remodeling of the damaged myocardium often results in heart failure. Oral drugs aimed at reducing myocardial damage and remodeling require repeated administration of high doses to maintain therapeutic levels. This compromises efficacy and patient adherence and may cause adverse effects, such as hypotension and liver and/or kidney dysfunction. Hydrogels have emerged as an effective delivery platform for orthotopic treatment of MI due to their high water content and excellent tissue compatibility. AREA COVERED Hydrogels create an optimal microenvironment for delivering drugs, proteins, and cells, preserving their efficacy and increasing their bioavailability. Current research focuses on discovering functional hydrogels for mitigating myocardial damage and regulating repair processes in MI treatment. EXPERT OPINION Hydrogels offer a promising approach in enhancing cardiac repair and improving patient outcomes post-MI. Advancements in hydrogel technology are poised to transform MI therapy, paving the way for personalized treatment strategies and enhanced recovery.
Collapse
Affiliation(s)
- Qiang Luo
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, P. R. China
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, P. R. China
| | - Zhibo Li
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, P. R. China
| | - Bin Liu
- Department of Cardiology, The Second Hospital of Jilin University, Changchun, P. R. China
| | - Jianxun Ding
- Key Laboratory of Polymer Ecomaterials, Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, P. R. China
- School of Applied Chemistry and Engineering, University of Science and Technology of China, Hefei, P. R. China
| |
Collapse
|
48
|
Ren J, Wu Z, Wang B, Zheng L, Han S, Hu J. Multifunctional chitosan-based composite hydrogels engineered for sensing applications. Int J Biol Macromol 2024; 278:134956. [PMID: 39179061 DOI: 10.1016/j.ijbiomac.2024.134956] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 08/05/2024] [Accepted: 08/20/2024] [Indexed: 08/26/2024]
Abstract
Chitosan-based hydrogels, as natural high-molecular-weight flexible materials, are widely utilized due to their outstanding properties. In this research, we developed a one-pot method for synthesizing a novel PVA/CS@PPy-PDAx% conductive hydrogel and explored the internal bonding patterns through molecular dynamics simulations. By adding PPy-PDA nanoparticles into a hydrogel matrix, an interpenetrating conductive network established successfully. The uniform distribution of PPy-PDA nanoparticles endowed the hydrogel with good electrical conductivity (0.171 S/m), significantly enhanced mechanical properties, and strain sensing (S = 5.04), as well as near-infrared photothermal responsiveness (temperature increase of 41.9 °C within 30 s). Additionally, due to the hydrogel's significant photothermal conversion efficiency under near-infrared radiation, it exhibits rapid elimination of Escherichia coli with an antibacterial efficiency exceeding 90 %. The unique hydrogen-bonded crosslinked structure provides the hydrogel with excellent re-healing properties, allowing for restoration through a freeze-thaw process after damage. The conductivity remains nearly unchanged after re-healing, maintaining the material's integrity and functionality. The flexible sensor based on this hydrogel has a response time of 100 ms and can sensitively detect large-scale deformations (e.g., joint bending at various angles), different gravitational forces, and recognize human handwriting. These characteristics make this hydrogel a promising candidate for advancing intelligent wearable technologies and human-machine interaction systems.
Collapse
Affiliation(s)
- Jie Ren
- Center for Molecular Science and Engineering, College of Science, Northeastern University, Shenyang 110819, PR China
| | - Zengyang Wu
- Center for Molecular Science and Engineering, College of Science, Northeastern University, Shenyang 110819, PR China
| | - Bai Wang
- Shenyang Fire Science and Technology Research Institute of MEM, Shenyang 110034, PR China; National Engineering Laboratory for Fire and Emergency Rescue, Shenyang 110034, PR China
| | - Liuping Zheng
- College of Chemistry and Materials Science, Fujian Normal University, Fuzhou, Fujian 350007, PR China.
| | - Siyu Han
- Center for Molecular Science and Engineering, College of Science, Northeastern University, Shenyang 110819, PR China.
| | - Jianshe Hu
- Center for Molecular Science and Engineering, College of Science, Northeastern University, Shenyang 110819, PR China.
| |
Collapse
|
49
|
Rothe R, Xu Y, Wodtke J, Brandt F, Meister S, Laube M, Lollini PL, Zhang Y, Pietzsch J, Hauser S. Programmable Release of Chemotherapeutics from Ferrocene-Based Injectable Hydrogels Slows Melanoma Growth. Adv Healthc Mater 2024; 13:e2400265. [PMID: 39007274 DOI: 10.1002/adhm.202400265] [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: 01/23/2024] [Revised: 07/02/2024] [Indexed: 07/16/2024]
Abstract
Hydrogel-based injectable drug delivery systems provide temporally and spatially controlled drug release with reduced adverse effects on healthy tissues. Therefore, they represent a promising therapeutic option for unresectable solid tumor entities. In this study, a peptide-starPEG/hyaluronic acid-based physical hydrogel is modified with ferrocene to provide a programmable drug release orchestrated by matrix-drug interaction and local reactive oxygen species (ROS). The injectable ROS-responsive hydrogel (hiROSponse) exhibits adequate biocompatibility and biodegradability, which are important for clinical applications. HiROSponse is loaded with the two cytostatic drugs (hiROSponsedox/ptx) doxorubicin (dox) and paclitaxel (ptx). Dox is a hydrophilic compound and its release is mainly controlled by Fickian diffusion, while the hydrophobic interactions between ptx and ferrocene can control its release and thus be regulated by the oxidation of ferrocene to the more hydrophilic state of ferrocenium. In a syngeneic malignant melanoma-bearing mouse model, hiROSponsedox/ptx slows tumor growth without causing adverse side effects and doubles the relative survival probability. Programmable release is further demonstrated in a tumor model with a low physiological ROS level, where dox release, low dose local irradiation, and the resulting ROS-triggered ptx release lead to tumor growth inhibition and increased survival.
Collapse
Affiliation(s)
- Rebecca Rothe
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Bautzner Landstrasse 400, 01328, Dresden, Germany
- Technische Universität Dresden, Faculty of Chemistry and Food Chemistry, School of Science, Bergstrasse 66, 01069, Dresden, Germany
| | - Yong Xu
- B CUBE Center for Molecular Bioengineering, Technische Universität Dresden, Tatzberg 41, 01307, Dresden, Germany
| | - Johanna Wodtke
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Bautzner Landstrasse 400, 01328, Dresden, Germany
| | - Florian Brandt
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Bautzner Landstrasse 400, 01328, Dresden, Germany
- Technische Universität Dresden, Faculty of Chemistry and Food Chemistry, School of Science, Bergstrasse 66, 01069, Dresden, Germany
| | - Sebastian Meister
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Bautzner Landstrasse 400, 01328, Dresden, Germany
| | - Markus Laube
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Bautzner Landstrasse 400, 01328, Dresden, Germany
| | - Pier-Luigi Lollini
- Alma Mater Studiorum, University of Bologna, Department of Medical and Surgical Sciences, Viale Filopanti 22, Bologna, 40126, Italy
| | - Yixin Zhang
- B CUBE Center for Molecular Bioengineering, Technische Universität Dresden, Tatzberg 41, 01307, Dresden, Germany
| | - Jens Pietzsch
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Bautzner Landstrasse 400, 01328, Dresden, Germany
- Technische Universität Dresden, Faculty of Chemistry and Food Chemistry, School of Science, Bergstrasse 66, 01069, Dresden, Germany
| | - Sandra Hauser
- Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Department of Radiopharmaceutical and Chemical Biology, Bautzner Landstrasse 400, 01328, Dresden, Germany
| |
Collapse
|
50
|
Widener AE, Roberts A, Phelps EA. Granular Hydrogels for Harnessing the Immune Response. Adv Healthc Mater 2024; 13:e2303005. [PMID: 38145369 PMCID: PMC11196388 DOI: 10.1002/adhm.202303005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2023] [Revised: 11/13/2023] [Indexed: 12/26/2023]
Abstract
This review aims to understand the current progress in immune-instructive granular hydrogels and identify the key features used as immunomodulatory strategies. Published work is systematically reviewed and relevant information about granular hydrogels used throughout these studies is collected. The base polymer, microgel generation technique, polymer crosslinking chemistry, particle size and shape, annealing strategy, granular hydrogel stiffness, pore size and void space, degradability, biomolecule presentation, and drug release are cataloged for each work. Several granular hydrogel parameters used for immune modulation: porosity, architecture, bioactivity, drug release, cell delivery, and modularity, are identified. The authors found in this review that porosity is the most significant factor influencing the innate immune response to granular hydrogels, while incorporated bioactivity is more significant in influencing adaptive immune responses. Here, the authors' findings and summarized results from each section are presented and suggestions are made for future studies to better understand the benefits of using immune-instructive granular hydrogels.
Collapse
Affiliation(s)
- Adrienne E Widener
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Dr., Gainesville, 32611, USA
| | - Abilene Roberts
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Dr., Gainesville, 32611, USA
| | - Edward A Phelps
- J. Crayton Pruitt Family Department of Biomedical Engineering, University of Florida, 1275 Center Dr., Gainesville, 32611, USA
| |
Collapse
|