1
|
Xuanyuan X, Liu W, Jiang M, Zhang X, Wen B, Zheng R, Yao N, Zhang T, Feng Y, Qiao C, Zhang H, Luo D, Feng S, Li M, Gao J, Lu Z. Harnessing prazosin for tumors: Liposome hybrid nanovesicles activate tumor immunotherapy via autophagy inhibition. Biomaterials 2025; 319:123184. [PMID: 39985978 DOI: 10.1016/j.biomaterials.2025.123184] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/01/2025] [Accepted: 02/09/2025] [Indexed: 02/24/2025]
Abstract
Prazosin (Prz), an antagonist of alpha-1 adrenergic receptors, is conventionally employed in the treatment of hypertension. Our study pioneers the exploration of Prz in oncology, examining its impact on cellular autophagy and its potential to trigger antitumor immune responses. We have developed a novel Prz-loaded liposome hybrid nanovesicle (Prz@LINV) system, integrating tumor-derived nanovesicles (TNV) with liposomes (LIP) to facilitate targeted Prz delivery to tumor sites. This formulation enhances Prz bioavailability and markedly inhibits tumor cell autophagy, leading to immunogenic cell death (ICD) and the activation of antitumor immune responses. Furthermore, Prz@LINV modulates dendritic cells (DCs), augmenting their antigen cross-presentation capacity and thereby potentiating antitumor immunity. These effects were validated in a colorectal cancer mouse model, demonstrating the good biocompatibility of Prz@LINV and its significant inhibition in tumor growth, along with the enhancement of antitumor immune responses. Our findings elucidate a novel mechanism by which Prz inhibits autophagy and enhances the antitumor immune response, providing a foundation for the development of innovative immunotherapeutic strategies. The efficacy of Prz@LINV suggests that Prz may emerge as a pivotal component in future immunotherapeutic regimens, offering patients more potent therapeutic options.
Collapse
Affiliation(s)
- Xinyang Xuanyuan
- Department of Dermatology, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Wenshang Liu
- Department of Dermatology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China
| | - Min Jiang
- The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Xin Zhang
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - BeiBei Wen
- School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Rui Zheng
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Ning Yao
- Department of General Surgery, Joint Support Force 903rd Hospital, Hangzhou, 310013, China
| | - Tinglin Zhang
- Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Yu Feng
- School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Chaofeng Qiao
- School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Huiqi Zhang
- School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Dong Luo
- School of Pharmacy, Henan University, Kaifeng, 475004, China
| | - Sa Feng
- School of Pharmacy, Henan University, Kaifeng, 475004, China.
| | - Meng Li
- Department of Dermatology, Shanghai Children's Medical Center, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200127, China.
| | - Jie Gao
- Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China; Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, China.
| | - Zhengmao Lu
- Department of Gastrointestinal Surgery, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China.
| |
Collapse
|
2
|
Xie Z, Yang C, Zhang J, Zhang Y, Wang O, Wang Y, Liu F. From victims to protectors: Microalgae's unexpected capacity for diuron elimination. JOURNAL OF HAZARDOUS MATERIALS 2025; 489:137693. [PMID: 40015043 DOI: 10.1016/j.jhazmat.2025.137693] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Revised: 02/04/2025] [Accepted: 02/19/2025] [Indexed: 03/01/2025]
Abstract
Diuron is a widespread PSII-inhibiting herbicide that persists in aquatic ecosystems and threatens environmental health. However, cost-effective removal solutions remain elusive. This study revealed that microalgae have the ability to remove diuron efficiently under mixotrophic conditions, e.g., Parachlorella kessleri could completely remove 60 μg·L-1 of diuron within 60 hours. The primary process of this removal is bioabsorption, which relies more on new biomass generation than on total biomass. Additionally, diuron significantly enhances the mixotrophic growth of P. kessleri by maintaining a stable pH environment, stabilizing between 7.0 and 7.5 without the need for buffers or adjustments. This pH stabilization is attributed to diuron's reduction of the alkalizing effect of microalgal photosynthetic carbon assimilation. Other microalgae species, including Chlorella vulgaris, Chlorella ellipsoidea and Scenedesmus quadricauda, also show high diuron removal efficiencies and growth promotion under mixotrophic conditions. These findings collectively suggest that the use of mixotrophic microalgae could represent a new potential method for diuron removal.
Collapse
Affiliation(s)
- Zhangzhang Xie
- National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangdong Key Laboratory of Integrated Agro-environmental Pollution Control and Management, Institute of Eco-environmental and Soil Sciences, Guangdong Academy of Sciences, Guangzhou 510650, PR China
| | - Cuiyun Yang
- Laboratory for Marine Ecology and Environmental Science, Qingdao National Laboratory for Marine Science and Technology, Qingdao 266071, PR China
| | - Ju Zhang
- National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangdong Key Laboratory of Integrated Agro-environmental Pollution Control and Management, Institute of Eco-environmental and Soil Sciences, Guangdong Academy of Sciences, Guangzhou 510650, PR China; School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, PR China
| | - Yaping Zhang
- School of Environmental Science and Engineering, Guangdong University of Technology, Guangzhou 510006, PR China
| | - Oumei Wang
- National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangdong Key Laboratory of Integrated Agro-environmental Pollution Control and Management, Institute of Eco-environmental and Soil Sciences, Guangdong Academy of Sciences, Guangzhou 510650, PR China
| | - Ying Wang
- National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangdong Key Laboratory of Integrated Agro-environmental Pollution Control and Management, Institute of Eco-environmental and Soil Sciences, Guangdong Academy of Sciences, Guangzhou 510650, PR China
| | - Fanghua Liu
- National-Regional Joint Engineering Research Center for Soil Pollution Control and Remediation in South China, Guangdong Key Laboratory of Integrated Agro-environmental Pollution Control and Management, Institute of Eco-environmental and Soil Sciences, Guangdong Academy of Sciences, Guangzhou 510650, PR China; Laboratory for Marine Biology and Biotechnology, Qingdao Marine Science and Technology Center, Qingdao 266237, PR China.
| |
Collapse
|
3
|
Cai J, Zhou H, Liu M, Zhang D, Lv J, Xue H, Zhou H, Zhang W. Host immunity and intracellular bacteria evasion mechanisms: Enhancing host-directed therapies with drug delivery systems. Int J Antimicrob Agents 2025; 65:107492. [PMID: 40107461 DOI: 10.1016/j.ijantimicag.2025.107492] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 03/04/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025]
Abstract
Host-directed therapies (HDTs) have been investigated as a potential solution to combat intracellular and drug-resistant bacteria. HDTs stem from extensive research on the intricate interactions between the host and intracellular bacteria, leading to a treatment approach that relies on immunoregulation. To improve the bioavailability and safety of HDTs, researchers have utilized diverse drug delivery systems (DDS) to encapsulate and transport therapeutic agents to target cells. In this review, we first introduce the three mechanisms of bactericidal action and intracellular bacterial evasion: autophagy, reactive oxygen species (ROS), and inflammatory cytokines, with a particular focus on autophagy. Special attention is given to the detailed mechanism of xenophagy in clearing intracellular bacteria, a crucial selective autophagy process that specifically targets and degrades intracellular pathogens. Following this, we present the application of DDS to modulate these regulatory methods for intracellular bacteria elimination. By integrating insights from immunology and nanomedicine, this review highlights the emerging role of DDS in advancing HDTs for intracellular bacterial infections and paving the way for innovative therapeutic interventions.
Collapse
Affiliation(s)
- Jiayang Cai
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Han Zhou
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Mingwei Liu
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Dingjian Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Jingxuan Lv
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Haokun Xue
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Houcheng Zhou
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China
| | - Wenli Zhang
- Department of Pharmaceutics, China Pharmaceutical University, Jiangsu, China.
| |
Collapse
|
4
|
Brignone SG, Tampucci S, Maletto BA, Allemandi DA, Chetoni P, Palma SD. Self-assembled structures of ascorbic acid derivatives: encapsulation properties and controlled drug delivery applications. Expert Opin Drug Deliv 2025:1-18. [PMID: 40397471 DOI: 10.1080/17425247.2025.2510301] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2025] [Revised: 05/09/2025] [Accepted: 05/20/2025] [Indexed: 05/22/2025]
Abstract
INTRODUCTION The chemical instability and low liposolubility of ascorbic acid have driven the development of hydrophobic derivatives that not only enhance its stability but also enable the formation of self-assembled systems for controlled drug delivery. These systems have garnered increasing interest due to their potential to improve drug administration by enhancing solubility, protecting against degradation, and enabling controlled or targeted release. AREAS COVERED This review critically explores the development and application of self-assembled systems derived from ascorbic acid compounds, including micelles, microemulsions, aspasomes, and other nanostructures. A comprehensive literature search was conducted across PubMed, Scopus, and ScienceDirect databases, covering publications from January 2000 to February 2025. The discussion focuses on the physicochemical characteristics, biological behavior, and relevance of these systems in the delivery of both hydrophilic and hydrophobic drugs. EXPERT OPINION Self-assembled drug delivery systems based on hydrophobic ascorbic acid derivatives represent a promising strategy in the field of pharmaceutical development. These platforms combine biocompatibility with functional versatility, offering enhanced membrane permeability, reduced systemic toxicity, and the potential for sustained or targeted drug release. Their suitability for oral, ophthalmic, and parenteral administration further highlights their applicability across a range of therapeutic areas, including immunological, anti-inflammatory, anti-infective, and oncological treatments.
Collapse
Affiliation(s)
| | - Silvia Tampucci
- Department of Pharmacy, University of Pisa, Pisa, Italy
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), University of Pisa, Pisa, Italy
| | - Belkys A Maletto
- Departamento de Bioquímica Clínica, Facultad de Ciencias QuímicasUniversidad Nacional de Córdoba, Córdoba, Argentina
- Centro de Investigaciones en Bioquímica Clínica e Inmunología (CIBICI), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Daniel A Allemandi
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| | - Patrizia Chetoni
- Department of Pharmacy, University of Pisa, Pisa, Italy
- Inter-University Center for the Promotion of the 3Rs Principles in Teaching & Research (Centro 3R), University of Pisa, Pisa, Italy
| | - Santiago D Palma
- Departamento de Ciencias Farmacéuticas, Facultad de Ciencias Químicas, Universidad Nacional de Córdoba, Córdoba, Argentina
- Unidad de Investigación y Desarrollo en Tecnología Farmacéutica (UNITEFA), Consejo Nacional de Investigaciones Científicas y Técnicas (CONICET), Córdoba, Argentina
| |
Collapse
|
5
|
Han C, Zhang Z, Adiham A, Huang F, Yan Y, Li D, Gong P. Santali Albi Lignum: From traditional efficacies to pharmacological properties and modern therapeutic applications. JOURNAL OF ETHNOPHARMACOLOGY 2025; 350:120031. [PMID: 40419205 DOI: 10.1016/j.jep.2025.120031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 04/10/2025] [Accepted: 05/22/2025] [Indexed: 05/28/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Santali Albi Lignum (SA), the dry heartwood of the trunk of Santalum album L., was originally discovered in India. It has a long history of use in Ayurveda and traditional Chinese medicine (TCM), mainly to treat skin, cardiovascular and lung diseases. In recent years, SA has received worldwide attention because of its diverse pharmacological effects, including anti-tumour, neuroprotective and gastrointestinal regulatory effects. PURPOSE This paper aims to systematically review progress in the research on SA, focusing on its ethnopharmacology, phytochemical ingredients, pharmacological effects, quality control and clinical applications; directions for further research and development of this herbal medicine are also discussed. METHODS Information on SA was obtained mainly from published materials, classic ancient books on TCM and electronic databases (PubMed, Google Scholar, Science Direct, Web of Science China National Knowledge Infrastructure and traditional Chinese medicine classics). RESULTS A total of 216 molecules have been identified in SA, including terpenoids, fatty acids, organic acids, phenols, aldehydes, alkanes, esters, ketones, steroids, alcohols, phenylpropanoids and other compounds, of which sesquiterpenes have emerged as the primary bioactive ingredients. A wide spectrum of biological activities of extracts or compounds of SA, including neuroprotective, antitumour, anti-inflammatory, antioxidant and gastrointestinal effects, have been verified in in vitro and in vivo pharmacological studies. Quality is monitored by the quantification and identification of α⁃santalol, β⁃santalol and volatile oils. TCM formulations that contain SA are commonly used to treat coronary heart disease, heart failure, ischaemic stroke, skin diseases and others. CONCLUSIONS This systematic review demonstrates that modern bioactivities and clinical research reports provide scientific evidence for the efficacy of SA, especially the ability to circulate qi and alleviate pain. Current studies have focused mainly on the chemical composition and pharmacological effects of the volatile oil fraction of SA. Moreover, the integrated pharmacological mechanisms of the active compounds and extracts of SA still need to be comprehensively elucidated. Furthermore, research on its toxicology and pharmacokinetics should be expanded to ensure the reasonable and safe clinical use of SA.
Collapse
Affiliation(s)
- Cairong Han
- College of Pharmacy and Food, Southwest Minzu University, Chengdu, 610041, China.
| | - Zhongrui Zhang
- College of Pharmacy and Food, Southwest Minzu University, Chengdu, 610041, China.
| | - Akida Adiham
- College of Pharmacy and Food, Southwest Minzu University, Chengdu, 610041, China.
| | - Feifei Huang
- College of Pharmacy and Food, Southwest Minzu University, Chengdu, 610041, China.
| | - Yulu Yan
- College of Pharmacy and Food, Southwest Minzu University, Chengdu, 610041, China.
| | - Dapeng Li
- Key Laboratory of Drug-Targeting and Drug Delivery System of Sichuan Province, Sichuan Engineering Laboratory for Plant-Source Drug, Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu, 610041, China.
| | - Puyang Gong
- College of Pharmacy and Food, Southwest Minzu University, Chengdu, 610041, China.
| |
Collapse
|
6
|
Fang X, Chen X, Dong W, Ye F. A poly(tannic acid) particle-supported β-glucan/chitosan hydrogel for managing oral ulcers in diabetes. Int J Biol Macromol 2025; 306:141609. [PMID: 40024420 DOI: 10.1016/j.ijbiomac.2025.141609] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2024] [Revised: 02/06/2025] [Accepted: 02/27/2025] [Indexed: 03/04/2025]
Abstract
In the management of diabetic oral ulcers, current treatments frequently fall short due to complications from bacterial contamination, oxidative damage, and impaired angiogenesis. These shortcomings highlight the pressing need for more efficacious interventions that not only circumvent these issues but also expedite healing processes. To bridge this gap, we have introduced a novel hydrogel patch, POQ2 [a poly(tannic acid) particle-incorporated oxidized β-glucan/quaternized chitosan hydrogel]. POQ2 demonstrated significant improvements in healing outcomes by enhancing bacterial clearance (up to 99.7 % against Escherichia coli and 99.2 % against Staphylococcus aureus), reducing oxidative stress through efficient ROS scavenging (70.2 % reduction in DPPH radicals), and promoting angiogenesis. In vivo experiments on diabetic rat models showed accelerated wound closure and reduced IL-6 inflammatory markers, with nearly complete ulcer healing within seven days. The introduction of POQ2 into clinical protocols represents a substantial advancement in the management of diabetic oral ulcers, promising not only improved patient outcomes but also a shift in the standard of care in this challenging clinical area. This innovation not only fills a significant gap in diabetic wound care but also sets a new benchmark for the development of future treatments, potentially influencing broader therapeutic strategies for managing complex diabetic wounds.
Collapse
Affiliation(s)
- Xiaobi Fang
- Department of Otolaryngology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Xiaojing Chen
- Department of Otolaryngology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China
| | - Wei Dong
- School of Chemical Engineering, Nanjing University of Science & Technology, Nanjing, Jiangsu 210094, China.
| | - Fan Ye
- Department of Otolaryngology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang 325000, China.
| |
Collapse
|
7
|
Cheng X, Hemmati S, Pirhayati M, Zangeneh MM, Veisi H. Decoration of copper nanoparticles (Cu 2O NPs) over chitosan-guar gum: Its application in the Sonogashira cross-coupling reactions and treatment of human lung adenocarcinoma. Int J Biol Macromol 2025; 305:141122. [PMID: 39965696 DOI: 10.1016/j.ijbiomac.2025.141122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 02/02/2025] [Accepted: 02/14/2025] [Indexed: 02/20/2025]
Abstract
This study outlines the sustainable synthesis of hybrid biopolymer hydrogels supported with octahedral Cu2O nanoparticles (NPs), alongside their biological assessments and characterizations. A composite hydrogel made of chitosan and guar gum (CS-GG) was employed as a template for the environmentally friendly synthesis of nanoparticles. Leveraging their electron-rich functional groups, the biopolymers acted as stabilizing agents for the Cu2O NPs and as green reductants, facilitating the reduction of copper ions. The material's physicochemical properties were thoroughly examined using advanced techniques, such as X-ray diffraction (XRD), Field-Emission Scanning Electron Microscopes (FE-SEM), Eneregy Dispersive X-ray Electron Spectroscopy (EDX), Fourier Transformed Infrared Spectroscopy (FT-IR), Transmission Electron Microscopy (TEM) and ICP-OES. The resulting CS-GG/Cu2O NPs nanocomposite was investigated as a reusable heterogeneous nanocatalyst, demonstrating its efficiency in the phosphine-free, palladium-free, and ligand-free synthesis of various stilbene derivatives with high yields through the Sonogashira coupling reaction. The catalyst showed no significant reduction in activity after being reused seven times consecutively. The cytotoxic effects of the CS-GG/Cu2O NPs nanocomposite on NCI-H661 lung cancer cells and normal cells (HUVEC) were assessed over 48 h using MTT assay. The cancer cell's viability decreased after exposure to the CS-GG/Cu2O NPs, with an IC50 value of 82 μg/mL. The CS-GG/Cu2O NPs nanocomposite controls the phosphatidylinositol 3-kinase (PI3K)/protein kinase B (AKT)/mammalian target of rapamycin (mTOR) system, which in turn impacts apoptosis and cell proliferation in NCI-H661 cells, according to a detailed examination of the mTOR pathway. The pathway could act a role in the cell cycle inhibition and apoptosis induced by the CS-GG/Cu2O NPs nanocomposite. The CS-GG/Cu2O NPs nanocomposite could be a useful natural anti-cancer agent for the treatment of lung cancer.
Collapse
Affiliation(s)
- Xiongtao Cheng
- Graduate School, Hunan University of Chinese Medicine, Changsha, Hunan 410208, China
| | - Saba Hemmati
- Department of Chemistry, Payame Noor University, Tehran, Iran
| | - Mozhgan Pirhayati
- Department of Applied Chemistry, Faculty of Science, Malayer University, Malayer, Iran.
| | - Mohamad Mehdi Zangeneh
- Biotechnology and Medicinal Plants Research Center, Ilam University of Medical Sciences, Ilam, Iran
| | - Hojat Veisi
- Department of Chemistry, Payame Noor University, Tehran, Iran.
| |
Collapse
|
8
|
Meng F, Yang L, Ji M, Zhu S, Tao H, Wang G. Nanomaterials: A Prospective Strategy for Biofilm-Forming Helicobacter pylori Treatment. Int J Nanomedicine 2025; 20:5209-5229. [PMID: 40292401 PMCID: PMC12034278 DOI: 10.2147/ijn.s512066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2024] [Accepted: 04/07/2025] [Indexed: 04/30/2025] Open
Abstract
Helicobacter pylori (H. pylori) is prevalent in over 50% of the global population and is recognized as the primary etiological agent for the development of gastric cancer. With the increasing incidence of antibiotic resistance, clinical treatment of H. pylori is a significant challenge. The formation of H. pylori biofilm is an important reason for antibiotic resistance and chronic infection, and it is also one of the key obstacles to eradicating H. pylori. H. pylori biofilm acts as a physical barrier, preventing the penetration of antibiotics and increasing the expression of efflux pump genes and drug-resistant gene mutations. Therefore, the treatment of H. pylori biofilm is extremely challenging. Nanomaterials, such as inorganic nanoparticles, lipid-based nanoparticles, and polymeric nanoparticles, which have properties including disrupting bacterial cell membranes, controlling drug release, and overcoming antibiotic resistance, have attracted significant interest. Furthermore, nanomaterials have the ability to treat H. pylori biofilm owing to their unique size, structure, and physical properties, including the inhibition of biofilm formation, enhancement of biofilm permeability, and disruption of mature biofilm. Moreover, nanomaterials have targeting functions and can carry antimicrobial drugs that play a synergistic role, thus providing a prospective strategy for treating H. pylori biofilm. In this review, we summarize the formation and antibiotic-resistance mechanisms of H. pylori biofilm and outline the latest progress in nanomaterials against H. pylori biofilm with the aim of laying the foundation for the development and clinical application of nanomaterials for anti-H. pylori biofilm.
Collapse
Affiliation(s)
- Fansen Meng
- Department of Gastroenterology, Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, People’s Republic of China
- Medical School of Chinese PLA, Beijing, 100853, People’s Republic of China
| | - Lyukun Yang
- Medical School of Chinese PLA, Beijing, 100853, People’s Republic of China
| | - Mingzhong Ji
- Medical School of Chinese PLA, Beijing, 100853, People’s Republic of China
| | - Siying Zhu
- Medical School of Chinese PLA, Beijing, 100853, People’s Republic of China
| | - Hongjin Tao
- Medical School of Chinese PLA, Beijing, 100853, People’s Republic of China
| | - Gangshi Wang
- Department of Gastroenterology, Second Medical Center and National Clinical Research Center for Geriatric Diseases, Chinese PLA General Hospital, Beijing, 100853, People’s Republic of China
| |
Collapse
|
9
|
Zhang H, Wang Y, Qiao W, Hu X, Qiang H, Xia K, Du L, Yang L, Bao Y, Gao J, Zhang T, Yu Z. An injectable multifunctional nanocomposite hydrogel promotes vascularized bone regeneration by regulating macrophages. J Nanobiotechnology 2025; 23:283. [PMID: 40197239 PMCID: PMC11978117 DOI: 10.1186/s12951-025-03358-2] [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: 02/04/2025] [Accepted: 03/26/2025] [Indexed: 04/10/2025] Open
Abstract
The local inflammatory microenvironment, insufficient vascularization, and inadequate bone repair materials are the three key factors that constrain the repair of bone defects. Here, we synthesized a composite nanoparticle, TPQ (TCP-PDA-QK), with a core‒shell structure. The core consists of nanotricalcium phosphate (TCP), and the shell is derived from polydopamine (PDA). The surface of the shell is modified with a vascular endothelial growth factor (VEGF) mimic peptide (QK peptide). TPQ was then embedded in porous methacrylate gelatin (GelMA) to form a TPQGel hydrogel. In the inflammatory environment, the TPQGel hydrogel can gradually release drugs through pH responsiveness, promoting M2 macrophage polarization, vascularization and bone regeneration in turn. In addition, reprogrammed M2 macrophages stimulate the generation of anti-inflammatory and pro-healing growth factors, which provide additional support for angiogenesis and bone regeneration. The TPQGel hydrogel can not only accurately fill irregular bone defects but also has excellent biocompatibility, making it highly suitable for the minimally invasive treatment of bone defects. Transcriptomic tests revealed that the TPQGel hydrogel achieved macrophage reprogramming by regulating the PI3K-AKT signalling pathway. Overall, the TPQGel hydrogel can be harnessed for safe and efficient therapeutics that accelerate the repair of bone defects.
Collapse
Affiliation(s)
- Huaiyuan Zhang
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Yu Wang
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Wenyu Qiao
- Department of General Surgery, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Xueneng Hu
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Huifen Qiang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China
| | - Kuo Xia
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Longhai Du
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, China
| | - Luling Yang
- Digestive Endoscopy Center, School of Medicine, Shanghai Tongren Hospital, Shanghai Jiao Tong University, Shanghai, 200336, China
| | - Yi Bao
- Biological Safety Protection 3-Level Laboratory, Guangxi Medical University, Nanning, Guangxi Zhuang, 530021, China
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, China.
| | - Tinglin Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai, 200433, China.
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, China.
| | - Zuochong Yu
- Department of Orthopedics, Jinshan Hospital, Fudan University, Shanghai, 201508, China.
| |
Collapse
|
10
|
Zhou J, Li Y, Jiang X, Xin Z, Liu W, Zhang X, Zhai Y, Zhang Z, Shi T, Xue M, Zhang M, Wu Y, Chu Y, Wang S, Jin X, Zhu W, Gao J. PD-L1 siRNA incorporation into a cationic liposomal tumor mRNA vaccine enhances cytotoxic T cell activation and prevents immune evasion. Mater Today Bio 2025; 31:101603. [PMID: 40124340 PMCID: PMC11926701 DOI: 10.1016/j.mtbio.2025.101603] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 01/23/2025] [Accepted: 02/20/2025] [Indexed: 03/25/2025] Open
Abstract
Engaging antigen-presenting cells and T lymphocytes is essential for invigorating the immune system's response to cancer. Nonetheless, challenges such as the low immunogenicity of tumor antigens, the genetic heterogeneity of tumor cells, and the elevated expression of immune checkpoint molecules frequently result in resistance to immunotherapy or enable immune evasion by tumors. To overcome this resistance, we developed a therapeutic tumor vaccine employing cationic liposomes to encapsulate MC38 total RNA alongside PD-L1 siRNA (siPD-L1). The encapsulated total RNA, enriched with tumor mRNA, effectively transduces dendritic cells (DCs), thereby enhancing antigen presentation. The incorporation of siPD-L1 specifically targets and diminishes PD-L1 expression on both DCs and tumor cells, synergistically amplifying the cytotoxic capabilities of CD8+ T cells. Furthermore, cationic liposomes play dual roles as carriers crucial for preserving the integrity of nucleic acids for antigen translation and as inhibitors of autophagy-a process essential for both promoting antigen cross-presentation and revitalizing MHC-I expression on tumor cells, thereby increasing their immunogenicity. This cationic liposomal vaccine represents a promising strategy in cancer immunotherapy, launching a multidimensional offensive against tumor cells that enhances cytotoxic T lymphocyte (CTL) activation and prevents tumor immune evasion.
Collapse
Affiliation(s)
- Jingsheng Zhou
- Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
- Department of Pathology, Chongqing University Cancer Hospital, Chongqing, 400030, China
| | - Yuanyuan Li
- Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Xianghe Jiang
- Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Zhongyuan Xin
- Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Wenshang Liu
- Department of Dermatology, Shanghai Children's Medical Center, Shanghai Jiaotong University School of Medicine, Shanghai, 200127, China
| | - Xinyi Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Yonghua Zhai
- Department of Cardiovascular Medicine, Department of Hypertension, Ruijin Hospital and State Key Laboratory of Medical Genomics, Shanghai Key Laboratory of Hypertension, Shanghai Institute of Hypertension, Shanghai Jiao Tong University School of Medicine, 197 Ruijin 2nd Road, Shanghai, 200025, China
| | - Zhuanzhuan Zhang
- Institute of Translational Medicine, Shanghai University, Shanghai, 200444, China
| | - Te Shi
- Department of Gastroenterology, Chinese People's Liberation Army Naval Medical Center, Shanghai, 200052, China
| | - Minghao Xue
- Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Mengya Zhang
- Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
| | - Yan Wu
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Yanhui Chu
- College of Life Science, Mudanjiang Medical University, Mudanjiang, 157011, China
| | - Shimin Wang
- Department of Gastroenterology, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200000, China
| | - Xin Jin
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Weiping Zhu
- Department of Hepatic Surgery, Fudan University Shanghai Cancer Center, Fudan University, Shanghai, 200032, China
| | - Jie Gao
- Changhai Clinical Research Unit, The First Affiliated Hospital of Naval Medical University, Shanghai, 200433, China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, 200433, China
| |
Collapse
|
11
|
Zhang D, Zhu B, Xu Y, Luo F, Chen T, Chen L, Wang X, Wu D, Hu J. Oleanolic acid-based nanoparticles for the treatment of ulcerative colitis. Nanomedicine (Lond) 2025; 20:677-690. [PMID: 39988882 PMCID: PMC11970778 DOI: 10.1080/17435889.2025.2467019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Accepted: 02/11/2025] [Indexed: 02/25/2025] Open
Abstract
AIM This study aims to develop and assess the therapeutic potential of oleanolic acid nanoparticles (OA NPs) in treating ulcerative colitis (UC). MATERIALS & METHODS OA NPs were synthesized using an emulsion solvent evaporation method, forming spherical nanoparticles with an average diameter of 138.1 nm. The nanoparticles were designed to target the colon through the enhanced permeability and retention (EPR) effect. Network pharmacology and molecular docking identified key inflammatory pathways, and in vitro (RAW264.7 cells) and in vivo (DSS-induced UC mouse model) experiments evaluated their anti-inflammatory effects and therapeutic efficacy. RESULTS OA NPs successfully targeted the colon and demonstrated improved bioavailability. In vitro experiments showed that OA NPs reduced oxidative stress and inflammation by downregulating pro-inflammatory cytokines (TNF-α, IL-6, and IL-1β) and promoting macrophage polarization from M1 to M2. In the DSS-induced UC mouse model, oral administration of OA NPs significantly alleviated colitis symptoms, improved colon length, reduced inflammation, and mitigated tissue damage. CONCLUSION OA NPs mitigate UC pathology through targeted delivery, enhanced stability, and modulation of inflammatory pathways, providing a promising approach for UC treatment. Further studies are needed to evaluate their long-term safety and clinical applicability.
Collapse
Affiliation(s)
- Dan Zhang
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
| | - Beiwei Zhu
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
| | - Yu Xu
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
| | - Fengxian Luo
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
| | - Tao Chen
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
| | - Lihang Chen
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
| | - Xinchuang Wang
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
| | - Di Wu
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
| | - Jiangning Hu
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, School of Food Science and Technology, Dalian Polytechnic University, Dalian, China
| |
Collapse
|
12
|
Lai Y, Shen H, Wang S, Ouyang Y, Zhang X, Hu B, Zhang X, Li G, Xu L, Zhao J. Hydrogel-Transformable Probiotic Powder for Targeted Eradication of Helicobacter pylori with Enhanced Gastric Mucosal Repair and Microbiota Preservation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2500478. [PMID: 40091425 DOI: 10.1002/advs.202500478] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/25/2025] [Indexed: 03/19/2025]
Abstract
Lactobacillus reuteri (L. reuteri) therapies represent a potentially effective approach to eradicating Helicobacter pylori (H. pylori). However, the difficulty in bacterial viability preservation and harsh gastric environment compromises the survival and on-target delivery of L. reuteri. This study presents a novel bacterium-mediated bacterial elimination strategy using an edible L. reuteri@HTP probiotic powder for targeted bacterial elimination. The probiotic powder is obtained by grinding a lyophilized hydrogel composed of L. reuteri, hyaluronic acid (HA), tannic acid (TA), and polyvinyl alcohol (PVA). Upon contact with water, the powder quickly transforms into a hydrogel, enhancing L. reuteri's survival in the harsh gastric environment and ensuring selective release at H. pylori-infected inflammatory sites. L. reuteri targets and reduces H. pylori colonization while secreting reuterin to eliminate the bacteria. Additionally, TA's antioxidant properties help alleviate inflammation, and HA supports gastric mucosal repair. L. reuteri@HTP powder preserves the integrity of the gut microbiota, facilitating the restoration of a healthy microbiome. In particular, the probiotic powder remains stable at room temperature for at least six months, providing a promising alternative to traditional antibiotics for H. pylori treatment. This strategy combines targeted eradication, mucosal healing, and microbiome restoration, offering a new approach to treating gastric infections.
Collapse
Affiliation(s)
- Yongkang Lai
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital; National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, P. R. China
| | - Hanchun Shen
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, P. R. China
| | - Shige Wang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, P. R. China
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong, SAR, 999077, P. R. China
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong, SAR, 999077, P. R. China
| | - Yongliang Ouyang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, P. R. China
| | - Xinyuan Zhang
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, P. R. China
| | - Bin Hu
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, P. R. China
| | - Xiaoyi Zhang
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong, SAR, 999077, P. R. China
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong, SAR, 999077, P. R. China
| | - Guisheng Li
- School of Materials and Chemistry, University of Shanghai for Science and Technology, Shanghai, 200093, P. R. China
| | - Lizhi Xu
- Department of Mechanical Engineering, The University of Hong Kong, Hong Kong, SAR, 999077, P. R. China
- Advanced Biomedical Instrumentation Centre, Hong Kong Science Park, Shatin, New Territories, Hong Kong, SAR, 999077, P. R. China
- Materials Innovation Institute for Life Sciences and Energy (MILES), The University of Hong Kong Shenzhen Institute of Research and Innovation (HKU-SIRI), Shenzhen, 518057, P. R. China
| | - Jiulong Zhao
- Department of Gastroenterology, Shanghai Institute of Pancreatic Diseases, Changhai Hospital; National Key Laboratory of Immunity and Inflammation, Naval Medical University, Shanghai, 200433, P. R. China
| |
Collapse
|
13
|
Zheng Y, Zhang T, Shao J, Du Y, Li Z, Zhang L, Gao J. Antibiotic-free responsive biomaterials for specific and targeted Helicobacter pylori eradication. J Control Release 2025; 379:708-729. [PMID: 39863021 DOI: 10.1016/j.jconrel.2025.01.054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2024] [Revised: 12/17/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
Gastric cancer is highly correlated with Helicobacter pylori (H. pylori) infection. Approximately 50 % of the population worldwide is infected with H. pylori. However, current treatment regimens face severe challenges including drug resistance and gut microbiota disruption. An integrative treatment with slight negative influences on intestinal flora, conforming with concepts of integrative prevention of gastric cancer, is urgently needed. Non-antibiotic responsive biomaterials can respond to different stimuli, including pH, enzymes, light, ultrasound and magnetism, under which biomaterials are specifically activated to perform antibacterial capabilities, while neutral intestinal microenvironments differ from gastric microenvironments or inflammatory sites and have no or minimal irradiation via precisely controlled exogenous stimuli, which may not only overcome antibiotic resistance but also avoid gut microbiota disorders. First, the latest progress in responsive biomaterials against H. pylori without gut microbiome disturbance and their anti-H. pylori performances are profoundly summarized. Second, the mechanisms against planktonic bacteria, biofilms and intracellular bacteria are discussed respectively. Finally, the strategies of specific and targeted H. pylori elimination by responsive biomaterials are introduced. Additionally, the challenges and the focus of future research on translation into clinical application are fully proposed. Antibiotic-free responsive biomaterials for specific and targeted H. pylori eradication represent an innovative approach.
Collapse
Affiliation(s)
- Yating Zheng
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China; Yangzhou Branch of Jiangsu Provincial Corps of Chinese People's Armed Police Force, Yangzhou 225007, Jiangsu, China
| | - Tinglin Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China; Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, China
| | - Juan Shao
- Yangzhou Branch of Jiangsu Provincial Corps of Chinese People's Armed Police Force, Yangzhou 225007, Jiangsu, China
| | - Yiqi Du
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China; Department of Gastroenterology, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China; Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, China
| | - Zhaoshen Li
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China; Department of Gastroenterology, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China; Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, China
| | - Li Zhang
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China; Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, China.
| | - Jie Gao
- Changhai Clinical Research Unit, Shanghai Changhai Hospital, Naval Medical University, Shanghai 200433, China; Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical Devices, Shanghai, China.
| |
Collapse
|
14
|
Liu T, Gu Y, Zhao Y, Li Y. Nanomaterials in gastric cancer: pioneering precision medicine for diagnosis, therapy, and prevention. Med Oncol 2025; 42:93. [PMID: 40050498 DOI: 10.1007/s12032-025-02650-3] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Accepted: 02/24/2025] [Indexed: 03/29/2025]
Abstract
Gastric cancer (GC) continues to be a major health issue globally due to its high rates of both occurrence and mortality. Despite advancements in treatment, the outlook for those affected remains poor, highlighting the critical need for new diagnostic and treatment methods. Nanotechnology, especially nanoparticles, is emerging as a crucial innovation in cancer care by improving imaging, targeting drug delivery, and enhancing early detection. These nanoparticles are also enhancing the effectiveness of treatments like phototherapy, chemotherapy, and immunotherapy. Notably, they show potential in addressing infections like Helicobacter pylori (H. pylori), which is known to increase the risk of developing GC. This review underscores the pivotal role of nanotechnology in enhancing the integrated management of GC, offering a basis for future advancements in the field.
Collapse
Affiliation(s)
- Tiantian Liu
- Lanzhou University Second Clinical Medical School, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Yanmei Gu
- Lanzhou University Second Clinical Medical School, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Yang Zhao
- Lanzhou University Second Clinical Medical School, Lanzhou University Second Hospital, Lanzhou, Gansu, China
| | - Yumin Li
- Lanzhou University Second Clinical Medical School, Lanzhou University Second Hospital, Lanzhou, Gansu, China.
| |
Collapse
|
15
|
Li Y, Xiang Y, Chen Y, Wang Y, Dong W, Liu Y, Qi X, Shen J. A Natural Eumelanin-Assisted Pullulan/Chitosan Hydrogel for the Management of Diabetic Oral Ulcers. Macromol Biosci 2025; 25:e2400526. [PMID: 39601462 DOI: 10.1002/mabi.202400526] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/13/2024] [Indexed: 11/29/2024]
Abstract
Existing methods for treating diabetic oral ulcers often fall short in clinical environments due to potential bacterial contamination, oxidative harm, and hindered angiogenesis throughout the healing process. Here, a hydrogel patch (HYG2) have been developed for local in situ application. HYG2 comprises oxidized pullulan, quaternized chitosan, and eumelanin nanoparticles derived from cuttlefish ink. These components work together to efficiently heal wounds associated with diabetic oral ulcers. Application begins with a simple local injection that quickly forms a protective barrier over the mucosa, effectively stopping bleeding and counteracting inflammatory agents. HYG2 is distinguished by its strong antibacterial properties and capacity to eliminate reactive oxygen species, promoting bacteria clearance and managing oxidative stress, which accelerates the healing phase from inflammation to tissue regeneration. Additionally, HYG2's 3D structure, incorporating elements from natural sources, offers exemplary support for structural and nutritional cell needs. This enhancement fosters cell adhesion, migration, and proliferation, along with further angiogenesis during mucosal remodeling. Ultimately, HYG2 is fully absorbed by the body after serving its therapeutic functions. Evidence from in vitro and in vivo studies shows that HYG2 hydrogel markedly accelerates mucosal wound repair, making it a promising treatment for diabetic oral ulcers.
Collapse
Affiliation(s)
- Ying Li
- School of Chemical Engineering, Nanjing University of Science & Technology, Nanjing, Jiangsu, 210094, China
| | - Yajing Xiang
- School & Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Yuxi Chen
- School & Hospital of Stomatology, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Yingying Wang
- Department of Otolaryngology, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, Zhejiang, 325000, China
| | - Wei Dong
- School of Chemical Engineering, Nanjing University of Science & Technology, Nanjing, Jiangsu, 210094, China
| | - Yan Liu
- School of Chemical Engineering, Nanjing University of Science & Technology, Nanjing, Jiangsu, 210094, China
| | - Xiaoliang Qi
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
| | - Jianliang Shen
- National Engineering Research Center of Ophthalmology and Optometry, Eye Hospital, Wenzhou Medical University, Wenzhou, Zhejiang, 325027, China
- Zhejiang Engineering Research Center for Tissue Repair Materials, Wenzhou Institute, University of Chinese Academy of Sciences, Wenzhou, Zhejiang, 325001, China
| |
Collapse
|
16
|
Dharejo SA, Pirzada T, Shah MR, Nadeem A, Thebo KH. In-vitro study of hybrid silver nanoparticles with humic acid extracted from cow dung against pathogens. Heliyon 2025; 11:e41636. [PMID: 39866433 PMCID: PMC11760302 DOI: 10.1016/j.heliyon.2025.e41636] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 12/31/2024] [Accepted: 01/02/2025] [Indexed: 01/28/2025] Open
Abstract
Recently, researchers have used silver nanoparticles (AgNPs) coupled with humic acid (HA) as antimicrobial agents. Herein, AgNPs were prepared and coupled with humic acid for their antimicrobial activities. The as-prepared AgNPs coupled with humic acid (HA) were characterized by an atomic force microscope (AFM), X-ray powder diffraction (XRD), zeta potential, zeta sizer, Fourier-transform infrared (FT-IR) spectroscopy, and UV-VIS spectrophotometer. Moreover, human plasma, varied salt concentrations, and pH levels were used for stability confirmation using a UV-VIS spectrophotometer. The antibacterial activities and minimal bactericidal concentration (MBC) of coupled AgNPs were determined by disk diffusion and broth dilution methods, respectively, against identified Staphylococcus aureus, Streptococcus pyogene, Pseudomonas aeruginosa, and Escherichia coli, which are extracted from cow dung. AgNPs' peak in the UV-Vis spectral range showed maximal absorption at 415 nm. The coupled AgNPs displayed their distinctive peak under all circumstances, demonstrating their stability. The FT-IR displayed functional groups such as hydroxyl, carboxylic acids, carbonyl, ester, and ether groups. The particles were face-centered cubic (FCC) in structure, according to the XRD. Moreover, particles had a spherical shape, high negative zeta potential, and were polydisperse in nature, with an average size of 25.43 nm. The minimal bactericidal concentration (MBC) of AgNPs was found to be 2.5 mg/mL, and the MBC of AgNPs/HA was found to be 5 mg/mL. The result indicated that the as-synthesizedAgNPs/HA are more effective in inhibiting all the studied microorganisms, which can be attributed to the therapeutic use of nanoparticles coated with humic acids.
Collapse
Affiliation(s)
- Shabir Ahmed Dharejo
- Institute of Chemistry, Shah Abdul Latif University, Khairpur Mir's, 66020, Sindh, Pakistan
| | - Tajnees Pirzada
- Institute of Chemistry, Shah Abdul Latif University, Khairpur Mir's, 66020, Sindh, Pakistan
| | - Muhammad Raza Shah
- International Center for Chemical and Biological Sciences, H.E.J. Research Institute of Chemistry, Karachi University Karachi, 74200, Pakistan
| | - Ahmed Nadeem
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, Riyadh, 11451, Saudi Arabia
| | - Khalid Hussain Thebo
- Institute of Metal Research (IMR), Chinese Academy of Science, Wenhua Road, Shenyang, China
| |
Collapse
|
17
|
Taherzadeh E, Arianfar A, Mahdian E, Mohseni S. Impact of nanoemulsion of Ajwain-cardamom essential oils on Mortadella sausage quality during chilling (4°C) storage. Heliyon 2025; 11:e41643. [PMID: 39866456 PMCID: PMC11759640 DOI: 10.1016/j.heliyon.2025.e41643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 12/26/2024] [Accepted: 01/02/2025] [Indexed: 01/28/2025] Open
Abstract
Essential oils application as natural preservatives is challenging owning to low solubility and stability to harsh conditions, while incorporation of essential oils into nanoemulsion systems can effectively improve these issues. Therefore, the nanoemulsion of ajwain (C. copticum) and cardamom essential oils were fabricated through self-emulsification technique and evaluated their size, ζ-potential, antioxidative and antibacterial activities. The effect of double nanomulsion on the textural and sensorial properties of Mortadella sausage was also examined under chilling temperature (4 °C). Our goal was to improve the chilling storage of Mortadella sausage by using ajwain and cardamom nanoemulsion as natural preservative. By increasing the ajwain essential oil in the nanoemulsion, the protein and moisture of sausage increased, while the fat content decreased (17 %). Furthermore, nanoemulsion of ajwain and cardamom essential oils showed particle size less than 100 nm and PDI<0.5 revealing the stability of nanoemulsions. Moreover, double nanoemulsions exhibited higher antibacterial activity against S. aureus and IC50 DPPH value (107 ppm). The nanoemulsion had a greater effect on the textural properties of Mortadella, reduction in hardness (∼5300 g), and chewiness (∼2500 g mm). Ajwain/cardamom nanoemulsion also increased the sensory properties, particularly taste and acceptance of the Mortadella. Consequently, Ajwain/cardamom nanoemulsion not only improve the storage of mortadella sausage at chilling temperature due to their antioxidant and antimicrobial properties, but also has a positive effect on the red color and textural properties created a special herbal aroma, taste and odor in the Mortadella samples, which ultimately contributed to the customer-friendly product. The appropriate dose of these nanoemulsion can develop meat products at lowest amount of nitrite in Mortadella sausage formulations, although, further research should be conducted on the mechanism of action AEO/CEO nanoemulsion concerning appearance and nitrite reduction in the meat products.
Collapse
Affiliation(s)
- Elmira Taherzadeh
- Department of Food Science and Technology, Quchan Branch, Islamic Azad University, Quchan, Iran
| | - Akram Arianfar
- Department of Food Science and Technology, Quchan Branch, Islamic Azad University, Quchan, Iran
| | - Elham Mahdian
- Department of Food Science and Technology, Quchan Branch, Islamic Azad University, Quchan, Iran
| | - Sharareh Mohseni
- Department of Chemistry, Quchan Branch, Islamic Azad University, Quchan, Iran
| |
Collapse
|
18
|
Yin X, Lai Y, Zhang X, Zhang T, Tian J, Du Y, Li Z, Gao J. Targeted Sonodynamic Therapy Platform for Holistic Integrative Helicobacter pylori Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2408583. [PMID: 39535366 PMCID: PMC11727135 DOI: 10.1002/advs.202408583] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 10/27/2024] [Indexed: 11/16/2024]
Abstract
Helicobacter pylori (H. pylori) is a primary pathogen associated with gastrointestinal diseases, including gastric cancer. The increase in resistance to antibiotics, along with the adverse effects caused by complicated medication protocols, has made the eradication of H. pylori a more formidable challenge, necessitating alternative therapeutics. Herein, a targeted nanoplatform is reported based on sonodynamic therapy, the chitosan-conjugated fucose loaded with indocyanine green (ICG@FCS). It penetrates the gastric mucosa and homes in on H. pylori through dual targeting mechanisms: molecular via fucose and physical via ultrasound. Upon ultrasound activation, it generates singlet oxygen, effectively attacking planktonic bacteria, disrupting biofilms, and facilitating the clearance of intracellular bacteria by promoting autophagy, including multidrug-resistant strains. The ICG@FCS nanoplatform minimally affects the gut microbiota and aids in gastric mucosa repair. a holistic integrative H. pylori therapy strategy is proposed that targets eradication while preserving gastrointestinal health. This strategy emphasizes the importance of maintaining patient health while eradicating the pathogen. This advancement is set to refine the comprehensive antibacterial approach, offering a promising horizon in the ongoing battle against antibiotic resistance and more effective gastric cancer prevention strategies.
Collapse
Affiliation(s)
- Xiaojing Yin
- Department of GastroenterologyShanghai Institute of Pancreatic DiseasesChanghai HospitalShanghai200433China
- Changhai Clinical Research UnitChanghai HospitalNaval Medical UniversityShanghai200433China
| | - Yongkang Lai
- Department of GastroenterologyShanghai Institute of Pancreatic DiseasesChanghai HospitalShanghai200433China
- Changhai Clinical Research UnitChanghai HospitalNaval Medical UniversityShanghai200433China
- Department of GastroenterologyGanzhou People's Hospital Affiliated to Nanchang UniversityGanzhou341000China
| | - Xinyuan Zhang
- School of Health Science and EngineeringUniversity of Shanghai for Science and TechnologyShanghai200093China
| | - Tingling Zhang
- Changhai Clinical Research UnitChanghai HospitalNaval Medical UniversityShanghai200433China
- National Key Laboratory of Immunity and InflammationNaval Medical UniversityShanghai200433China
| | - Jing Tian
- Department of PharmacyShanghai Changhai Hospitalthe First Affiliated Hospital of Navy Medical UniversityShanghai200433China
| | - Yiqi Du
- Department of GastroenterologyShanghai Institute of Pancreatic DiseasesChanghai HospitalShanghai200433China
- Changhai Clinical Research UnitChanghai HospitalNaval Medical UniversityShanghai200433China
- National Key Laboratory of Immunity and InflammationNaval Medical UniversityShanghai200433China
| | - Zhaoshen Li
- Department of GastroenterologyShanghai Institute of Pancreatic DiseasesChanghai HospitalShanghai200433China
- Changhai Clinical Research UnitChanghai HospitalNaval Medical UniversityShanghai200433China
- National Key Laboratory of Immunity and InflammationNaval Medical UniversityShanghai200433China
| | - Jie Gao
- Department of GastroenterologyShanghai Institute of Pancreatic DiseasesChanghai HospitalShanghai200433China
- Changhai Clinical Research UnitChanghai HospitalNaval Medical UniversityShanghai200433China
- National Key Laboratory of Immunity and InflammationNaval Medical UniversityShanghai200433China
- Shanghai Key Laboratory of Nautical Medicine and Translation of Drugs and Medical DevicesShanghai200433China
| |
Collapse
|
19
|
Xu Z, Wu XM, Luo YB, Li H, Zhou YQ, Liu ZQ, Li ZY. Exploring the therapeutic potential of yeast β-glucan: Prebiotic, anti-infective, and anticancer properties - A review. Int J Biol Macromol 2024; 283:137436. [PMID: 39522898 DOI: 10.1016/j.ijbiomac.2024.137436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 10/31/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
Yeast β-glucan (YBG), an indigestible polysaccharide from yeast cell walls, is multifunctional. It plays a pivotal role in regulating gut microbiota (GM) and boosting the immune system, which is central to research on inflammation, cancer, and metabolic diseases. By modulating the GM, YBG exhibits various prebiotic effects, including hypoglycemic, hypolipidemic, and immune-regulating properties. Additionally, acting as a bioreactor modulator, it activates immune responses, demonstrating potential in anti-infection and anticancer applications. This article synthesizes the latest data from in vitro, in vivo, and clinical studies. It comprehensively evaluates the therapeutic potential of YBG, starting from its structure-function relationship. It particularly focuses on the application prospects of yeast β-glucan in probiotic-like effects, anti-infectious properties, and anti-cancer activity, and explores the underlying mechanisms of these actions. The aim of this article is to elucidate the positive impact of YBG on health by modulating the gut microbiota and enhancing immune responses. Simultaneously, it identifies critical areas for future research to provide theoretical support for its development in biomedical applications.
Collapse
Affiliation(s)
- Zhen Xu
- The Second Clinical Medical College, China Three Gorges University, Yichang, Hubei 443002, China
| | - Xiao Meng Wu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
| | - Yan Bin Luo
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
| | - Hui Li
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
| | - Yong Qin Zhou
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443002, China.
| | - Zhao Qi Liu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443002, China.
| | - Zhi Ying Li
- The Second Clinical Medical College, China Three Gorges University, Yichang, Hubei 443002, China.
| |
Collapse
|