1
|
Qi W, Zhao T, Liu M, Shi X, Yang Y, Huang Y, Li N, Ai K, Huang Q. Engineered tantalum sulfide nanosheets for effective acute liver injury treatment by regulating oxidative stress and inflammation. J Colloid Interface Sci 2025; 693:137596. [PMID: 40250115 DOI: 10.1016/j.jcis.2025.137596] [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: 02/04/2025] [Revised: 04/11/2025] [Accepted: 04/12/2025] [Indexed: 04/20/2025]
Abstract
INTRODUCTION Tantalum sulfide (TaS2), a two-dimensional layered material, shows significant promise for treating acute liver injury (ALI) due to its exceptional biocompatibility and potent reactive oxygen species (ROS) scavenging capacity. However, the clinical translation of TaS2-based therapy remains limited by challenges in optimizing its stability, bioavailability, and particle size to match the liver's complex architecture. OBJECTIVES This study investigated the mechanisms by which serum albumin (SA)-modified TaS2 nanosheets (S-TaS2) modulate oxidative stress, apoptosis, and inflammation to achieve therapeutic efficacy in ALI. METHODS S-TaS2 was synthesized via a top-down exfoliation strategy and comprehensively characterized using transmission electron microscopy (TEM), X-ray photoelectron spectroscopy (XPS), X-ray diffraction (XRD), Fourier-transform infrared spectroscopy (FTIR), ultraviolet-visible (UV-Vis) spectroscopy, and Zeta potential analysis. In vivo therapeutic performance was evaluated through liver function tests, Hematoxylin-Eosin staining (HE), Dihydroethidium (DHE) staining, 8-Hydroxy-2'-deoxyguanosine (8-OHdG) staining, and ROS level assessments. Biodistribution, mitochondrial protection, and anti-inflammatory effects of S-TaS2 were assessed via in vivo fluorescence imaging, immunohistochemistry, western blotting, JC-1 and Mitochondrial Superoxide (MitoSOX) staining, Annexin V-fluorescein isothiocyanate (FITC)/Propidium Iodide (PI) apoptosis assays, enzyme-linked immunosorbent assays (ELISA), and other complementary techniques. RESULTS The exfoliation process successfully reduced TaS2 to monolayer nanosheets, yielding a nanoscale formulation with improved bioactivity. SA modification significantly enhanced aqueous stability and enabled targeted liver delivery. This targeting effect is attributed to two factors: the inherent liver affinity of SA and the optimal particle size of S-TaS2 (∼185 nm), which facilitates passage through hepatic sinusoids (50-200 nm) and, in pathological conditions such as ALI, through damaged vascular endothelium. In an acetaminophen (APAP)-induced ALI model, S-TaS2 preferentially accumulated in the injured liver, where it scavenged excessive ROS, mitigated mitochondrial dysfunction, and significantly preserved hepatocyte integrity. Notably, S-TaS2 also attenuated liver inflammation, reduced pro-inflammatory cytokine levels, and promoted tissue repair. Furthermore, it demonstrated adequate biosafety both in vitro and in vivo. CONCLUSIONS This study presents the first successful synthesis of S-TaS2, a liver-targeting nanotherapeutic engineered through SA modification and size optimization. S-TaS2 preferentially accumulates in damaged hepatic tissue and effectively combats ALI by suppressing oxidative stress and inflammation, while preventing their pathological amplification. These findings offer new therapeutic insights and a promising platform for future liver-targeted interventions.
Collapse
Affiliation(s)
- Weimin Qi
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Tianjiao Zhao
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Min Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Xiaojing Shi
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Yongqi Yang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Yunying Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Niansheng Li
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China.
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China; Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Qiong Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
2
|
Yang J, Ye J, Li R, Li R, Liu X, Han J, Yang Y, Ran N, Yuan M, Zhang Z, Chong W, Ji X. Nanozyme-functionalized microalgal biohybrid microrobots in inflammatory bowel disease treatment. Biomaterials 2025; 319:123231. [PMID: 40037206 DOI: 10.1016/j.biomaterials.2025.123231] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/24/2025] [Accepted: 02/28/2025] [Indexed: 03/06/2025]
Abstract
Oral drugs are the most direct and effective strategy for the treatment of gastrointestinal diseases. However, the harsh environment of gastric juice, lack of targeted lesion sites, and rapid metabolism present difficulties in the development of oral drugs. This research introduces a nanozyme-functionalized microalgal biohybrid microrobot (Hp@CS-PNAs@PAA) with a novel mechanism for treating inflammatory bowel disease (IBD) by leveraging the therapeutic advantages of microalgae and nanozymes. The microrobot uniquely combines the natural antioxidant capacity of Hematococcus pluvialis (Hp) microalgae and the catalytically active enzyme-mimicking properties of platinum-based nanoparticle assemblies (PNAs), enabling enhanced scavenging of reactive oxygen species (ROS) and targeted anti-inflammatory effects. Through its layered design, the Hp@CS-PNAs@PAA microrobot can navigate the gastrointestinal tract, resist degradation, and target inflamed colon tissues via electrostatic interactions, achieving extended retention and prolonged therapeutic action at inflammation sites. This study demonstrated that the synergistic anti-inflammatory effects of the microrobot derive from its ability to reduce ROS, inhibit proinflammatory cytokines, and promote the expression of tight junction proteins critical for preserving the integrity of the intestinal barrier. Both in vitro and in vivo tests in a DSS-induced colitis mouse model revealed that this system effectively restores damaged tissues by reducing oxidative stress and inflammation, indicating significant potential for clinical application in the management of colitis and similar inflammatory diseases.
Collapse
Affiliation(s)
- Jinmei Yang
- State Key Laboratory of Advanced Medical Materials and Devices, Medical College, Tianjin University, Tianjin, 300072, China
| | - Jiamin Ye
- State Key Laboratory of Advanced Medical Materials and Devices, Medical College, Tianjin University, Tianjin, 300072, China
| | - Runtan Li
- State Key Laboratory of Advanced Medical Materials and Devices, Medical College, Tianjin University, Tianjin, 300072, China
| | - Ruiyan Li
- State Key Laboratory of Advanced Medical Materials and Devices, Medical College, Tianjin University, Tianjin, 300072, China
| | - Xinting Liu
- State Key Laboratory of Advanced Medical Materials and Devices, Medical College, Tianjin University, Tianjin, 300072, China
| | - Jingwen Han
- State Key Laboratory of Advanced Medical Materials and Devices, Medical College, Tianjin University, Tianjin, 300072, China
| | - Yiwen Yang
- State Key Laboratory of Advanced Medical Materials and Devices, Medical College, Tianjin University, Tianjin, 300072, China
| | - Nana Ran
- State Key Laboratory of Advanced Medical Materials and Devices, Medical College, Tianjin University, Tianjin, 300072, China
| | - Mengyu Yuan
- State Key Laboratory of Advanced Medical Materials and Devices, Medical College, Tianjin University, Tianjin, 300072, China
| | - Zhuhong Zhang
- School of Pharmacy, Key Laboratory of Molecular Pharmacology and Drug Evaluation (Yantai University), Ministry of Education, Collaborative Innovation Center of Advanced Drug Delivery System and Biotech Drugs in Universities of Shandong, Yantai University, Yantai, 264005, China.
| | - Wei Chong
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, 250021, Jinan, China; Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, 250021, Jinan, China.
| | - Xiaoyuan Ji
- State Key Laboratory of Advanced Medical Materials and Devices, Medical College, Tianjin University, Tianjin, 300072, China; Medical College, Linyi University, Linyi, 276000, China.
| |
Collapse
|
3
|
Zhu Y, Fang Z, Bai J, Wang L, Chen J, Zhang Z, Wang Q, Sheng W, Pan X, Gao Z, Xu D, Wu P, Sun B. Orally Administered Functional Polyphenol-Nanozyme-Armored Probiotics for Enhanced Amelioration of Intestinal Inflammation and Microbiota Dysbiosis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2411939. [PMID: 40067175 PMCID: PMC12061243 DOI: 10.1002/advs.202411939] [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] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/23/2025] [Indexed: 05/04/2025]
Abstract
Maintaining microbiota balance and enhancing the antioxidant performance of nanozyme-based probiotic systems are crucial for effective inflammatory bowel disease (IBD) therapy. Despite significant advancements, developing a green and safe coating technology that functionalizes probiotics with nanozymes while preserving the activity of both components remains a challenge. To address this, chitosan-modified epigallocatechin gallate (EGCG-CS, EC)is synthesized, leveraging the intrinsic adhesive and coordination properties of polyphenols to capture gold nanozymes (AuNPs), forming ECA complexes that enhance nanozyme activity. When coated onto Escherichia coli Nissle 1917 (EcN), the resulting ECA@EcN system effectively scavenged reactive oxygen species (ROS), improving probiotic viability and promoting colon accumulation. Mechanistically, ECA protected EcN by suppressing the activation of the Flagellar Assembly and Branched-Chain Amino Acid Synthesis pathways, ultimately alleviating inflammation and modulating intestinal microbial communities to relieve IBD symptoms. Given the biocompatibility of its components and the environmentally friendly assembly approach, this polyphenol-nanozyme-armored probiotic system represents a promising platform for IBD treatment.
Collapse
Affiliation(s)
- Yong Zhu
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Ziqu Fang
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Jie Bai
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Longhui Wang
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Jiaqing Chen
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Zehua Zhang
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Qiang Wang
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Weiwei Sheng
- Department of General SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
| | - Xueyin Pan
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Zhenyuan Gao
- Department of General SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefei230022China
| | - Dengqiu Xu
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Pengkai Wu
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| | - Beicheng Sun
- Department of Hepatobiliary SurgeryThe First Affiliated Hospital of Anhui Medical UniversityHefeiAnhui230022China
- MOE Innovation Center for Basic Research in Tumor ImmunotherapyHefeiAnhui230022China
- Anhui Province Key Laboratory of Tumor Immune Microenvironment and ImmunotherapyHefeiAnhui230022China
| |
Collapse
|
4
|
Long M, Li J, Yang M, Chen W, Qiu L, Cheng X, Bi L. Rosmarinic acid-chondroitin sulfate nanoconjugate for targeted treatment of ulcerative colitis. Int J Biol Macromol 2025; 306:141008. [PMID: 39971022 DOI: 10.1016/j.ijbiomac.2025.141008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 01/18/2025] [Accepted: 02/12/2025] [Indexed: 02/21/2025]
Abstract
Rosmarinic acid (RA) is an attractive candidate for ulcerative colitis (UC) application due to its bioactive properties, including antioxidant and anti-inflammatory functions, however, the poor water solubility and on-targeting hamper its therapeutic outcome. Therefore, this work reported the synthesis and preparation of novel water-soluble rosmarinic acid-chondroitin sulfate A (RA-CSA) nanoconjugate, which was used for the treatment of UC in dextran sulfate sodium (DSS)-induced acute colitis mouse model. RA was functionalized with CSA as confirmed by FTIR and 1H NMR, and self-assembled to form nanoassemblies with a diameter of 247.3 ± 2.99 nm. RA-CSA nanoassemblies exhibited radical scavenging and antioxidant capacity. RA-CSA remarkably inhibited lipopolysaccharide-induced nitric oxide and TNF-α production in RAW 264.7 cells without cytotoxicity, whose inhibition rate was <5 % at 200 μg mL-1. Oral administration of RA-CSA nanoassemblies significantly attenuated colonic inflammation compared to the parent RA, as evidenced by significantly reduced the shortening of colon length (4.20 ± 0.15 cm), body weight loss, and colonic inflammatory damage in DSS-induced colitis mice. In addition, RA-CSA nanoassemblies suppressed the expression and production of typical pro-inflammatory cytokines of ulcerative colitis. These results suggest that RA-CSA nanoassemblies deserve further consideration as a potential therapeutic drug for the treatment of UC.
Collapse
Affiliation(s)
- Miaomiao Long
- Institute of Chemical Industry of Forest Products, Chinese Academy of Forestry, Nanjing 210042, China; Department of Pharmacy, Wuxi Higher Health Vocational Technology School, Wuxi 214028, China
| | - Jie Li
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Meiyang Yang
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China.
| | - Weijun Chen
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Lipeng Qiu
- School of Life Sciences and Health Engineering, Jiangnan University, Wuxi 214122, China
| | - Xian Cheng
- Institute of Chemical Industry of Forest Products, Chinese Academy of Forestry, Nanjing 210042, China.
| | - Liangwu Bi
- Institute of Chemical Industry of Forest Products, Chinese Academy of Forestry, Nanjing 210042, China.
| |
Collapse
|
5
|
Wang B, Tao M, Zhu W, Li J, Hai Z. In Situ Self-Assembled Probe for Antioxidant and Anti-Inflammatory Therapy of Inflammatory Bowel Disease. ACS APPLIED BIO MATERIALS 2025. [PMID: 40299753 DOI: 10.1021/acsabm.5c00394] [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: 05/01/2025]
Abstract
Inflammatory bowel disease (IBD) is a chronic and relapsing disease of the gastrointestinal tract. At present, antioxidant therapy is a promising strategy for IBD treatment. Since low-molecular-weight antioxidants (e.g., 2,2,6,6-tetramethylpiperidin-N-oxyl (TEMPO)) have a short in vivo half-life and inadequate cellular uptake, researchers have focused on loading antioxidants into nanostructures for improving their antioxidant and anti-inflammatory activities. As we know, in situ self-assembly with the formation of nanostructures under intracellular specific stimuli is a convenient delivery strategy to enhance the accumulation and retention of molecules at target sites in vivo. Herein, we developed an in situ self-assembled TEMPO probe TPP-FFYp-O to improve the antioxidant and anti-inflammatory effects of TEMPO in IBD. Compared to the control probe TPP-O without a self-assembly moiety, TPP-FFYp-O could successfully self-assemble into nanoparticles (NPs) under alkaline phosphatase (ALP)-guided dephosphorylation, with significantly enhanced antioxidant capacity in vitro. Cell experiments confirmed that intracellular formation of NPs by TPP-FFYp-O could improve the antioxidant and anti-inflammatory abilities of TEMPO and alleviate cellular damage. Moreover, TPP-FFYp-O exhibited good biocompatibility in vivo and significantly relieved pathological injury and inflammatory factors in the colon tissues of an IBD model compared to TPP-O. We envision that the in situ self-assembly platform can be used to load various active molecules for more applications in the future.
Collapse
Affiliation(s)
- Beibei Wang
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China
| | - Menglin Tao
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China
| | - Wujuan Zhu
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China
| | - Jin Li
- Department of Gastroenterology, The Eighth Affiliated Hospital, Sun Yat-Sen University, Shenzhen 518033, China
| | - Zijuan Hai
- Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui 230601, China
| |
Collapse
|
6
|
Feng Y, Liu Y, Liu L, Yang Q, An M, Yang H. Magnetite Micro/Nanorobots for Efficient Targeted Alleviation of Inflammatory Bowel Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2503307. [PMID: 40277443 DOI: 10.1002/advs.202503307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 03/30/2025] [Indexed: 04/26/2025]
Abstract
Millions of people worldwide have inflammatory bowel disease (IBD). Self-driven micro/nanorobots (MNRs) are efficient in the treatment of IBD. However, their lack of controllability regarding direction of motion in the organism and their inability to achieve continuous navigation limits their further application. In this study, polydopamine is wrapped around the magnetite surface, loaded with an anti-inflammatory drug resveratrol, and wrapped with pH-responsive sodium alginate to obtain magnetic MNRs. MNRs can be driven by magnetic fields to achieve directional movement and targeted transportation. In addition, MNRs can effectively remove reactive oxygen species from the inflammation site, repair intestinal damage, inhibit the cellular pathway of pro-inflammatory factors, such as MAPK and NF-κB pathways, and restore intestinal flora, thereby relieving IBDs. MNRs are safe and effective for in vivo treatment of IBD and have proven to be a promising therapeutic platform. This MNRs therapeutic strategy provides new insights into comprehensive IBD therapy.
Collapse
Affiliation(s)
- Ying Feng
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, China University of Geosciences, Wuhan, 430074, China
- Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
- Laboratory of Advanced Mineral Materials, China University of Geosciences, Wuhan, 430074, China
| | - Yang Liu
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, China University of Geosciences, Wuhan, 430074, China
- Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
- Laboratory of Advanced Mineral Materials, China University of Geosciences, Wuhan, 430074, China
| | - Linlin Liu
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, China University of Geosciences, Wuhan, 430074, China
- Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
- Laboratory of Advanced Mineral Materials, China University of Geosciences, Wuhan, 430074, China
| | - Qian Yang
- Centre for Immune-oncology, Nuffield Department of Medicine, University of Oxford, Old Road Campus, Oxford, OX3 7BN, UK
| | - Miao An
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, China University of Geosciences, Wuhan, 430074, China
- Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
- Laboratory of Advanced Mineral Materials, China University of Geosciences, Wuhan, 430074, China
| | - Huaming Yang
- Engineering Research Center of Nano-Geomaterials of Ministry of Education, China University of Geosciences, Wuhan, 430074, China
- Faculty of Materials Science and Chemistry, China University of Geosciences, Wuhan, 430074, China
- Laboratory of Advanced Mineral Materials, China University of Geosciences, Wuhan, 430074, China
| |
Collapse
|
7
|
Chen Z, Zhu YX, You Y, Ge M, Chen Y, Lin H, Shi J. Enzyme-Mimic Activities of RuCo Bimetallic Nanosheets for Inflammatory Bowel Disease Treatment. J Am Chem Soc 2025; 147:13424-13436. [PMID: 40173890 DOI: 10.1021/jacs.5c00100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/04/2025]
Abstract
The presence of abnormal levels of reactive oxygen species (ROS) is a recognized pathological feature of inflammatory bowel disease (IBD). Therefore, the development of orally administered antioxidants with high antioxidative capacity and gastric acid tolerance for the treatment of IBD is of great significance. Here, we present the design and synthesis of a bimetallic ruthenium-cobalt (RuCo) nanosheet for the treatment of IBD. The Ru-Co atoms within the nanosheet structure exhibit significant electron transfer properties owing to their electronegativity feature. Density functional theory calculations indicate that the RuCo nanosheets have higher d-band centers than the corresponding Ru and Co metal monoliths, which increases the catalytic activity. Such RuCo nanosheets exhibit superoxide dismutase and catalase-like cascade enzyme activities and show robust stability in gastric fluid over a 4 h period when exposed to simulated gastric fluid, ensuring desirable retention of antioxidative activity. Cellular and animal studies show that RuCo nanosheets are capable of effectively reducing oxidative stress, preventing inflammatory responses triggered by an abnormal increase in ROS at intestinal sites, and thus protecting cells from inflammatory damages. This research presents a gastric-acid-stabilized antioxidative nanocatalytic platform for the efficient treatment of inflammatory diseases of the digestive system.
Collapse
Affiliation(s)
- Zhixin Chen
- Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai Tenth People's Hospital, Shanghai 200331, P. R. China
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure; Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences, Shanghai Institute of Ceramics Chinese Academy of Sciences, Shanghai 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Ya-Xuan Zhu
- Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai Tenth People's Hospital, Shanghai 200331, P. R. China
| | - Yanling You
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure; Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences, Shanghai Institute of Ceramics Chinese Academy of Sciences, Shanghai 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Min Ge
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure; Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences, Shanghai Institute of Ceramics Chinese Academy of Sciences, Shanghai 200050, P. R. China
| | - Yihan Chen
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure; Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences, Shanghai Institute of Ceramics Chinese Academy of Sciences, Shanghai 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Han Lin
- Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai Tenth People's Hospital, Shanghai 200331, P. R. China
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure; Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences, Shanghai Institute of Ceramics Chinese Academy of Sciences, Shanghai 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| | - Jianlin Shi
- Shanghai Frontiers Science Center of Nanocatalytic Medicine, School of Medicine, Tongji University, Shanghai Tenth People's Hospital, Shanghai 200331, P. R. China
- State Key Laboratory of High Performance Ceramics and Superfine Microstructure; Research Unit of Nanocatalytic Medicine in Specific Therapy for Serious Disease, Chinese Academy of Medical Sciences, Shanghai Institute of Ceramics Chinese Academy of Sciences, Shanghai 200050, P. R. China
- Center of Materials Science and Optoelectronics Engineering, University of Chinese Academy of Sciences, Beijing 100049, P. R. China
| |
Collapse
|
8
|
Park J, Wu Y, Le QV, Kim JS, Xu E, Lee J, Oh YK. Self-disassembling nanoparticles as oral nanotherapeutics targeting intestinal microenvironment. Nat Commun 2025; 16:3365. [PMID: 40204740 PMCID: PMC11982569 DOI: 10.1038/s41467-025-58513-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 03/25/2025] [Indexed: 04/11/2025] Open
Abstract
Inspired by the survival strategies of pyomelanin-producing microbes, we synthesize pyomelanin nanoparticles (PMNPs) from homogentisic acid- γ-lactone via auto-oxidation and investigate their biomedical potential. PMNPs possess distinct physicochemical properties, including reactive oxygen species scavenging and microenvironment-responsive self-disassembly. Under intestinal conditions, PMNPs self-disassemble and penetrate the nanoscale pores of the mucin layer. In an inflammatory bowel disease model, orally administered PMNPs withstand gastric acidity and, in their solubilized form, interact with macrophages and epithelial cells. They significantly reduce reactive oxygen species levels, exert anti-inflammatory effects, and restore gut microbiota composition. Compared to conventional nanoparticles and 5-aminosalicylic acid, PMNPs exhibit greater therapeutic efficacy. Clinical symptoms and intestinal inflammation are alleviated, and the gut microbiota is restored to near-normal levels. These findings underscore the therapeutic potential of PMNPs for inflammatory bowel disease treatment and suggest broader biomedical applications.
Collapse
Affiliation(s)
- Jinwon Park
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Yina Wu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Quoc-Viet Le
- Faculty of Pharmacy, Ton Duc Thang University, Ho Chi Minh City, Vietnam
| | - Jung Suk Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Enzhen Xu
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea
| | - Jaiwoo Lee
- College of Pharmacy, Korea University, Sejong, Republic of Korea.
| | - Yu-Kyoung Oh
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
9
|
Zhao C, Wen S, Xu R, Wang K, Zhong Y, Huang D, Zhao B, Chen W. Oral delivery of ultra-small zwitterionic nanoparticles to overcome mucus and epithelial barriers for macrophage modulation and colitis therapy. Acta Biomater 2025; 196:399-409. [PMID: 39983856 DOI: 10.1016/j.actbio.2025.02.039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 02/23/2025]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory disease of the colon that poses significant therapeutic challenges due to the intestinal mucus and epithelial barriers. In this study, ultra-small zwitterionic nanoparticles (HC-CB NPs) is developed based on glutathione (GSH)-responsive hyperbranched polycarbonate to enhance the oral delivery of drugs and overcome these physiological barriers. HC-CB NPs demonstrate high colloidal stability across a wide range of pH environments and physiological fluids, preventing premature drug release within the gastrointestinal tract. The ultra-small sized HC-CB NPs demonstrate minimal mucin adsorption and effectively penetrate through the mucus layer, and the zwitterion surface further facilitate epithelial barrier crossing via the proton-assisted amino acid transporter 1 (PAT1) pathway. HC-CB NPs mediate enhanced macrophage uptake via monocarboxylate transporters (MCTs) pathway and ultimately improved therapy efficacy on colitis. The in vivo results reveal that FK506-loaded HC-CB NPs (HC-CB NPs@FK506) significantly reduce inflammatory markers (TNF-α, IL-6) and myeloperoxidase (MPO) levels, while promoting epithelial integrity by increasing E-cadherin expression. This study offers a promising approach to overcoming intestinal barriers in oral UC treatment, offering biocompatibility and potential for clinical translation. STATEMENT OF SIGNIFICANCE: Ulcerative colitis (UC) is a chronic inflammatory disease of the colon that poses significant therapeutic challenges due to the intestinal mucus and epithelial barriers. This study explores an oral UC therapy using ultra-small zwitterionic nanoparticles (HC-CB NPs) constructed from GSH-responsive hyperbranched polycarbonate. Compared to existing strategies, HC-CB NPs demonstrate minimal mucin adsorption and effectively penetrate through the mucus layer, and the zwitterion surface further facilitate epithelial barrier crossing via the proton-assisted amino acid transporter 1 (PAT1) pathway. Additionally, HC-CB NPs mediate enhanced macrophage uptake via monocarboxylate transporters (MCTs) pathway, resulting in improved therapeutic efficacy. These findings underscore the potential of HC-CB NPs as a transformative platform for overcoming intestinal barriers in UC treatment.
Collapse
Affiliation(s)
- Changshun Zhao
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Suchen Wen
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Rui Xu
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Ke Wang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Yinan Zhong
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| | - Dechun Huang
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China; Engineering Research Center for Smart Pharmaceutical Manufacturing Technologies, Ministry of Education, School of Engineering, China Pharmaceutical University, Nanjing 210009, China
| | - Bingbing Zhao
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| | - Wei Chen
- Department of Pharmaceutical Engineering, School of Engineering, China Pharmaceutical University, Nanjing 210009, China; Engineering Research Center for Smart Pharmaceutical Manufacturing Technologies, Ministry of Education, School of Engineering, China Pharmaceutical University, Nanjing 210009, China.
| |
Collapse
|
10
|
Sun J, Wu J, Zhao W, Zhang L, Han Y, Dong J, Zhang R, Shi Y. Multienzyme active melanin nanodots for antioxidant-immunomodulatory therapy of hyperoxia lung injury. Mater Today Bio 2025; 31:101609. [PMID: 40104637 PMCID: PMC11919337 DOI: 10.1016/j.mtbio.2025.101609] [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: 11/16/2024] [Revised: 02/18/2025] [Accepted: 02/23/2025] [Indexed: 03/20/2025] Open
Abstract
Supraphysiological oxygen is the most conventional method of treating patients with acute respiratory failure, but prolonged exposure to hyperoxia generates large amounts of reactive oxygen species (ROS) in the lungs, leading to hyperoxia lung injury (HLI). Nevertheless, there is no safe and effective prevention strategy. Herein, multienzyme active melanin nanodots were developed as an antioxidant-immunomodulatory defense nanoplatform for the treatment of HLI. The prepared nanodots are about 4 nm in size and are mainly composed of carbon, nitrogen and oxygen elements with high stability and multi-enzymatic activity for scavenging various reactive oxygen and reactive nitrogen radicals. Cellular experiments showed that melanin nanodots increased cell viability and ameliorated hyperoxia-induced morphological changes, mitochondrial damage and apoptosis. Meanwhile, by activating the Nrf2/Keap1/HO-1 signaling pathway, the treatment of melanin nanodots significantly inhibited the overproduction of ROS, reduced malondialdehyde, and increased the endogenous antioxidant enzyme activity in BEAS-2B cells. Interestingly, the antioxidant properties of melanin nanodots indirectly promoted the phenotypic shift of macrophages, and reduced hyperoxia-induced inflammatory responses in the damaged environment. In vivo NIR-II fluorescence imaging confirms the high retention of nanodots in the lungs and low accumulation in other major organs after inhalation administration, as well as the high biosafety of the melanin nanodots as they are metabolized out of the body over time via the liver and intestines. In addition, the melanin nanodots exhibited satisfactory antioxidant protection and inhibition of inflammatory cell infiltration in the lungs of HLI mouse models. Therefore, the melanin nanodots provide a potential and effective strategy for the treatment of HLI, showing great promise for application.
Collapse
Affiliation(s)
- Jinghua Sun
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Juan Wu
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Wenjing Zhao
- School of Pharmacy, Shanxi Medical University, Taiyuan, 030001, China
| | - Liyan Zhang
- School of Pharmacy, Shanxi Medical University, Taiyuan, 030001, China
| | - Yahong Han
- The Radiology Department of Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
| | - Jie Dong
- The Radiology Department of Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
| | - Ruiping Zhang
- The Radiology Department of Shanxi Provincial People's Hospital Affiliated to Shanxi Medical University, Taiyuan, 030001, China
| | - Yiwei Shi
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
11
|
Wang D, Wang S, Liu J, Shi X, Xiong T, Li R, Wei W, Ji L, Huang Q, Gong X, Ai K. Nanomedicine Penetrating Blood-Pancreas Barrier for Effective Treatment of Acute Pancreatitis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413925. [PMID: 39950925 PMCID: PMC11967758 DOI: 10.1002/advs.202413925] [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: 10/29/2024] [Revised: 02/01/2025] [Indexed: 04/05/2025]
Abstract
Acute pancreatitis (AP) is a primary contributor to hospitalization and in-hospital mortality worldwide. Targeted elimination of mitochondrial reactive oxygen species (mtROS) within pancreatic acinar cells (PACs) represents an ideal strategy for treating AP. However, existing drugs fail to overcome the physiological barriers of the pancreas to effectively reach PACs mitochondria due to the trade-off between conventional positively charged mitochondrial-targeting groups and their inability to penetrate the blood-pancreas barrier (BPB). Here, a tungsten-based heteropolyacid nano-antioxidant (mTWNDs) is introduced, co-modified with tannic acid (TA) and melanin, enabling site-specific clearance of mtROS in PACs, offering a highly effective treatment for AP. TA exhibits a strong affinity for proline-rich type III collagen and the mitochondrial outer membrane protein TOM20. This unique property allows mTWNDs to traverse the damaged BPB-exposing type III collagen to reach PACs and subsequently penetrate mitochondria for targeted mtROS elimination. In cerulein-induced AP mice, mTWNDs reversed AP at 1/50th the dose of N-acetylcysteine, suppressing PACs apoptosis and inflammation by blocking the stimulator of the interferon genes pathway activation in macrophage. This study establishes a mitochondrial-targeting antioxidant nanomedicine strategy for AP treatment.
Collapse
Affiliation(s)
- Dan Wang
- Department of General SurgeryXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Shuya Wang
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
| | - Jinjin Liu
- Department of General SurgeryXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Xiaojing Shi
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
| | - Tingli Xiong
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
| | - Ruishi Li
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
| | - Wei Wei
- Department of General SurgeryXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Liandong Ji
- Department of General SurgeryXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Qiong Huang
- Department of PharmacyXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Xuejun Gong
- Department of General SurgeryXiangya HospitalCentral South UniversityChangsha410008China
- National Clinical Research Center for Geriatric DisordersXiangya HospitalCentral South UniversityChangsha410008China
| | - Kelong Ai
- Xiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Hunan Provincial Key Laboratory of Cardiovascular ResearchXiangya School of Pharmaceutical SciencesCentral South UniversityChangsha410013China
- Key Laboratory of Aging‐related Bone and Joint Diseases Prevention and TreatmentMinistry of EducationXiangya HospitalCentral South UniversityChangsha410008China
| |
Collapse
|
12
|
Zhao X, Wang L, Fu YJ, Yu F, Li K, Wang YQ, Guo Y, Zhou S, Yang W. Inflammatory Microenvironment-Responsive Microsphere Vehicles Modulating Gut Microbiota and Intestinal Inflammation for Intestinal Stem Cell Niche Remodeling in Inflammatory Bowel Disease. ACS NANO 2025; 19:12063-12079. [PMID: 40125581 DOI: 10.1021/acsnano.4c17999] [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: 03/25/2025]
Abstract
Intestinal stem cells (ISCs) engage in proliferation to maintain a stable stem cell population and differentiate into functional epithelial subpopulations. This intricate process is upheld by various signals derived from the host and gut microbiota, establishing an ISC niche. However, during inflammatory bowel disease (IBD), this signaling niche undergoes dramatic changes, leading to impaired ISC and hindered restoration of the damaged intestinal epithelial barrier. This study introduces intestinal inflammatory microenvironment-responsive microsphere vehicles designed to remodel the ISC niche, offering an approach to treat IBD. Using an advanced emulsion technique, these microsphere vehicles specifically target colonic inflammation sites, delivering a responsive release of MXene and l-arginine. This delivery system is formulated to modulate intestinal flora and immune responses effectively. l-arginine is converted into nitric oxide to regulate the gut microbiome, while MXene serves as a nanoimmunomodulator to stabilize immune homeostasis. Our findings demonstrate that the anti-inflammatory properties of the microspheres are key to promoting epithelial repair and remodeling of the ISC niche. This study highlights the role of antioxidant microspheres as anti-inflammatory agents that indirectly support ISC function and gut regeneration.
Collapse
Affiliation(s)
- Xing Zhao
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu 610041, China
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Liya Wang
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu 610041, China
| | - Ya-Jun Fu
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| | - Fei Yu
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu 610032, China
| | - Kai Li
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041 , China
| | - Yu-Qiang Wang
- Department of Cardiovascular Surgery and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Yingqiang Guo
- Department of Cardiovascular Surgery and National Clinical Research Center for Geriatrics, West China Hospital, Sichuan University, Chengdu 610065, China
| | - Shaobing Zhou
- Institute of Biomedical Engineering, College of Medicine, Southwest Jiaotong University, Chengdu 610031, China
| | - Wei Yang
- College of Polymer Science and Engineering, Sichuan University, Chengdu 610065, China
| |
Collapse
|
13
|
Chen Y, Li J, Zhang X, Li S, Cheng Y, Fu X, Li J, Zhu L. Mesenteric adipose-derived exosomal TINAGL1 enhances intestinal fibrosis in Crohn's Disease via SMAD4. J Adv Res 2025; 70:139-158. [PMID: 38750695 PMCID: PMC11976418 DOI: 10.1016/j.jare.2024.05.016] [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: 02/04/2024] [Revised: 04/23/2024] [Accepted: 05/11/2024] [Indexed: 03/23/2025] Open
Abstract
INTRODUCTION Crohn's Disease (CD) is a chronic inflammatory condition characterized by intestinal fibrosis, severely impacting patient quality of life. The molecular mechanisms driving this fibrosis remain inadequately understood. Recent evidence implicates mesenteric adipose tissue (MAT) in CD pathogenesis, particularly through its exosome secretion, which may influence fibrogenic pathways. Understanding the role of MAT-derived exosomes is crucial for unraveling these molecular processes. OBJECTIVES This study aims to elucidate the role of MAT-derived exosomes in CD-related intestinal fibrosis. We focus on investigating their molecular composition and the potential impact on fibrosis progression, with an emphasis on identifying novel therapeutic targets. METHODS We induced chronic intestinal inflammation in mice using dinitrobenzene sulfonic acid (DNBS), simulating CD-like fibrosis. Exosomes were isolated from DNBS-treated mice (MG) and normal controls (NG) for characterization using electron microscopy and proteomic analysis. Additionally, human colonic fibroblasts were exposed to exosomes from CD patients and healthy individuals, with subsequent assessment of fibrogenesis through proteomic and RNA sequencing analyses. RESULTS Proteomic analyses revealed a significant activation of the TGF-β signaling pathway in MG-treated mice compared to controls, correlating with enhanced intestinal fibrosis. In vitro experiments demonstrated that colonic fibroblasts exposed to CD patient-derived exosomes exhibited increased fibrogenic activity. Protein docking and co-immunoprecipitation studies suggested a critical interaction between TINAGL1 and SMAD4, enhancing fibrosis. Importantly, in vivo experiments corroborated that recombinant TINAGL1 protein exacerbated DNBS-induced intestinal fibrosis. CONCLUSION Our findings highlight the pivotal role of MAT-derived exosomes, particularly those carrying TINAGL1, in the progression of intestinal fibrosis in CD. The involvement of the TGF-β signaling pathway, especially the SMAD4 protein, offers new insights into the molecular mechanisms of CD-related fibrosis and presents potential targets for therapeutic intervention.
Collapse
Affiliation(s)
- Yidong Chen
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Junrong Li
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaopeng Zhang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Shuang Li
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Yiyu Cheng
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Xiaoyu Fu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Jiamin Li
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China
| | - Liangru Zhu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China.
| |
Collapse
|
14
|
Gao J, He Y, Shi F, Hou F, Wu X, Yi Y, Zhang Y, Gong Q. Activation of Sirt6 by icariside Ⅱ alleviates depressive behaviors in mice with poststroke depression by modulating microbiota-gut-brain axis. J Adv Res 2025:S2090-1232(25)00142-0. [PMID: 40037430 DOI: 10.1016/j.jare.2025.03.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 02/24/2025] [Accepted: 03/01/2025] [Indexed: 03/06/2025] Open
Abstract
BACKGROUND Sirt6-mediated gut microbiota plays a vital role in poststroke depression (PSD). Icariside Ⅱ (ICS Ⅱ) is a naturally-occurring neuroprotectant with Sirt6 induction potency. However, it is unknown whether ICS Ⅱ protects against PSD through modulation of gut microbiota. OBJECTIVE This study aimed to reveal the effect and potential mechanisms of ICS Ⅱ on PSD, and the role of the microbiota-gut-brain axis was investigated. METHODS Using middle cerebral artery occlusion (MCAO) and chronic unpredictable mild stress (CUMS) to establish post-stroke depression (PSD) mice, we assessed anti-depressant effects of ICS Ⅱ via behavioral tests, immunohistochemistry, and western blot. Transcriptome profiling, molecular docking, and surface plasmon resonance were used to identify key targets. 16S rDNA genomic-derived taxonomic profiling and fecal microbiota transplantation (FMT) were conducted to figure out the mechanistic role of the gut microbiota and short-chain fatty acids (SCFAs). RESULTS ICS Ⅱ ameliorated depressive-like behaviors in PSD mice as evidenced by sucrose preference test, forced swimming test and tail suspension test. ICS Ⅱ restored mitochondrial function, reduced oxidative damage and pro-inflammatory cytokines both in brain and intestine through regulation of Sirt6/NF-κB pathway. ICS Ⅱ significantly increased the abundance of gut microbiota (such asAkkermansia and Ligilactobacillus), enhanced SCFAs concentrations, repaired intestinal barrier integrity and upreglated the tight junction protein expression. FMT from ICS II-treated mice replicated these benefits, confirming gut microbiota's role. Mechanistically, ICS Ⅱ directly bound to Sirt6 and enhanced its activity. However, ICS Ⅱ-mediated neuroprotection was neutralized in PSD mice or hydrogen peroxide-induced enteric glial cells when Sirt6 was absent. CONCLUSION Our findings expand the pharmacological properties of ICS II by demonstrating its ability to ameliorate PSD through modulation of the microbiota-gut-brain axis. ICS Ⅱ, as a novel Sirt6 activator, could be translated into an alternative microbiota-targeted avenue for coping with PSD.
Collapse
Affiliation(s)
- Jianmei Gao
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China; Department of Pharmacology, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China; Chinese Pharmacological Society-Guizhou Province Joint Laboratory for Pharmacology, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yifan He
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China; Department of Pharmacology, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China; Chinese Pharmacological Society-Guizhou Province Joint Laboratory for Pharmacology, Zunyi Medical University, Zunyi, Guizhou, China
| | - Fuguo Shi
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China; Department of Pharmacology, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China; Chinese Pharmacological Society-Guizhou Province Joint Laboratory for Pharmacology, Zunyi Medical University, Zunyi, Guizhou, China
| | - Fangqin Hou
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China; Department of Pharmacology, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China; Chinese Pharmacological Society-Guizhou Province Joint Laboratory for Pharmacology, Zunyi Medical University, Zunyi, Guizhou, China
| | - Xiaoyu Wu
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China; Department of Pharmacology, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China; Chinese Pharmacological Society-Guizhou Province Joint Laboratory for Pharmacology, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yang Yi
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China; Department of Pharmacology, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China; Chinese Pharmacological Society-Guizhou Province Joint Laboratory for Pharmacology, Zunyi Medical University, Zunyi, Guizhou, China
| | - Yuandong Zhang
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China; Department of Pharmacology, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China; Chinese Pharmacological Society-Guizhou Province Joint Laboratory for Pharmacology, Zunyi Medical University, Zunyi, Guizhou, China
| | - Qihai Gong
- Key Laboratory of Basic Pharmacology of Ministry of Education and Joint International Research Laboratory of Ethnomedicine of Ministry of Education, Zunyi Medical University, Zunyi, China; Department of Pharmacology, Key Laboratory of Basic Pharmacology of Guizhou Province and School of Pharmacy, Zunyi Medical University, Zunyi, Guizhou, China; Chinese Pharmacological Society-Guizhou Province Joint Laboratory for Pharmacology, Zunyi Medical University, Zunyi, Guizhou, China.
| |
Collapse
|
15
|
Zhao Y, Zhu M, Ling Y, Zhao Y, Lu X, Chu B, He Y, Wang H. A DNA Nanopatch-Bacteriophage System Targeting Streptococcus Gallolyticus for Inflammatory Bowel Disease Treatment and Colorectal Cancer Prevention. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2417334. [PMID: 39924920 DOI: 10.1002/adma.202417334] [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: 11/10/2024] [Revised: 01/28/2025] [Indexed: 02/11/2025]
Abstract
Persistent inflammation in inflammatory bowel disease (IBD) increases Streptococcus gallolyticus (Sg) colonization, increasing the risk of colorectal cancer progression via the Sg-activated cyclooxygenase-2 (COX-2) pathway and β-catenin upregulation. This study presents Sg-specific bacteriophages modified with DNA nanopatches (DNPs@P) designed to treat IBD and prevent Sg-induced malignancy. The DNPs are composed of DNA origami nanosheets and phage capture strands. The DNPs scavenge reactive oxygen species, enhancing the therapeutic efficacy of the phages while targeting and lysing pathogenic bacteria. Coating with an enteric polymer, DNPs@P ensures effective delivery in the gastrointestinal tract. These findings demonstrate significant restoration of colonic length, reduced inflammation, and improved gut microbiota diversity compared with current clinical treatments. Additionally, DNPs@P effectively prevents colonic tumourigenesis in mouse models. This approach presents a promising strategy for treating gastrointestinal diseases by remodeling the gut microenvironment, addressing a critical gap in current therapies.
Collapse
Affiliation(s)
- Yadan Zhao
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| | - Mengna Zhu
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| | - Yufan Ling
- State Key Laboratory of Radiation Medicine and Protection, School of Radiation Medicine and Protection, Suzhou Medical College of Soochow University, Suzhou, Jiangsu, 215123, P. R. China
| | - Yingying Zhao
- Department of Ultrasound, the First Affiliated Hospital of Soochow University, Suzhou, Jiangsu, 215006, China
| | - Xing Lu
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| | - Binbin Chu
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| | - Yao He
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
- Macao Translational Medicine Center, Macau University of Science and Technology, Taipa, Macau SAR, 999078, China
- Macao Institute of Materials Science and Engineering, Macau University of Science and Technology, Taipa, Macau SAR, 999078, China
| | - Houyu Wang
- Suzhou Key Laboratory of Nanotechnology and Biomedicine, Institute of Functional Nano & Soft Materials & Collaborative Innovation Center of Suzhou Nano Science and Technology (NANO-CIC), Soochow University, Suzhou, 215123, China
| |
Collapse
|
16
|
Zhao X, Yu Y, Xu X, Zhang Z, Chen Z, Gao Y, Zhong L, Chen J, Huang J, Qin J, Zhang Q, Tang X, Yang D, Zhu Z. Machine Learning-Assisted High-Throughput Screening of Nanozymes for Ulcerative Colitis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2417536. [PMID: 39801185 DOI: 10.1002/adma.202417536] [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: 11/12/2024] [Revised: 12/13/2024] [Indexed: 03/06/2025]
Abstract
Ulcerative colitis (UC) is a chronic gastrointestinal inflammatory disorder with rising prevalence. Due to the recurrent and difficult-to-treat nature of UC symptoms, current pharmacological treatments fail to meet patients' expectations. This study presents a machine learning-assisted high-throughput screening strategy to expedite the discovery of efficient nanozymes for UC treatment. Therapeutic requirements, including antioxidant property, acid stability, and zeta potential, are quantified and predicted by using a machine learning model. Non-quantifiable attributes, including intestinal barrier repair efficacy and biosafety, are assessed via high-throughput screening. Feature significance analysis, sure independence screening, and sparsifying operator symbolic regression reveal the high-dimensional structure-activity relationships between material features and therapeutic needs. SrDy2O4 with high stability, low toxicity, targeting ability, and reactive oxygen species (ROS) scavenging capability is identified, which reduces ROS production, lowers cytochrome C levels in cytoplasm, and inhibits apoptosis in intestinal epithelial cells by stabilizing the mitochondrial membrane potential. Mice treated with SrDy2O4 show improvements in colon length and body weight compared with dextran sodium sulfate salt-treated model group. Transcriptomic and 16S rRNA sequencing analyses show that SrDy2O4 boosts beneficial gut bacteria, and decreases pathogenic bacteria, thereby effectively restoring gut microbiota balance. Moreover, SrDy2O4 offers the advantage of X-ray imaging without side effects.
Collapse
Affiliation(s)
- Xianguang Zhao
- Department of Digestive Diseases, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Yixin Yu
- College of Materials Science and Engineering, Qingdao University of Science and Technology, 53 Zhengzhou Road, Qingdao, Shandong, 266042, China
| | - Xudong Xu
- Department of Digestive Diseases, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Ziqi Zhang
- College of Materials Science and Engineering, Qingdao University of Science and Technology, 53 Zhengzhou Road, Qingdao, Shandong, 266042, China
| | - Zhen Chen
- College of Materials Science and Engineering, Qingdao University of Science and Technology, 53 Zhengzhou Road, Qingdao, Shandong, 266042, China
| | - Yubo Gao
- College of Materials Science and Engineering, Qingdao University of Science and Technology, 53 Zhengzhou Road, Qingdao, Shandong, 266042, China
| | - Liang Zhong
- Department of Digestive Diseases, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Jiajie Chen
- Department of Digestive Diseases, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Jiaxin Huang
- Department of Digestive Diseases, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Jie Qin
- Department of Radiology, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Qingyun Zhang
- Central Laboratory Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Xuemei Tang
- Central Laboratory Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Dongqin Yang
- Department of Digestive Diseases, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
- Central Laboratory Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
- Department of Laboratory Medicine, Huashan Hospital, Fudan University, 12 Middle Urumqi Road, Shanghai, 200040, China
| | - Zhiling Zhu
- College of Materials Science and Engineering, Qingdao University of Science and Technology, 53 Zhengzhou Road, Qingdao, Shandong, 266042, China
- Key Laboratory of Optic-electric Sensing and Analytical Chemistry for Life Science, MOE, Shandong Key Laboratory of Biochemical Analysis, Qingdao University of Science and Technology, 53 Zhengzhou Road, Qingdao, Shandong, 266042, China
| |
Collapse
|
17
|
Zhang X, Yang H, He Y, Zhang D, Lu G, Ren M, Lyu Y, Yuan Z, He S. Yeast-Inspired Orally-Administered Nanocomposite Scavenges Oxidative Stress and Restores Gut Immune Homeostasis for Inflammatory Bowel Disease Treatment. ACS NANO 2025; 19:7350-7369. [PMID: 39943645 DOI: 10.1021/acsnano.4c18099] [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: 02/26/2025]
Abstract
Excessive oxidative stress, dysregulated immune homeostasis, and disruption of the intestinal epithelial barrier are crucial features of inflammatory bowel disease (IBD). Traditional treatments focusing solely on inflammation resolution remain unsatisfactory. Herein, a yeast-inspired orally administered nanocomposite was developed. First, the MD@MPDA core was fabricated by integrating manganese dioxide (MnO2) nanozymes onto diallyl trisulfide (H2S prodrug)-loaded mesoporous polydopamine nanoparticles (MPDA). Then, yeast cell wall (YCW) was chosen to encapsulate MD@MPDA, namely, YMD@MPDA. The β-glucan embedded in the YCW shell not only protected the nanocomposite from the harsh gastrointestinal environment but also allowed the targeting enrichment in the inflamed colon. Furthermore, M1 macrophages triggered the intracellular GSH-responsive H2S release in the pathological microenvironment. MD@MPDA effectively alleviated inflammatory responses by MnO2-mediated ROS-scavenging and H2S-participated immunomodulation. The synergistic action contributed to macrophage mitochondrial function restoration and M2 polarization by suppressing NOX4 signaling and p38 MAPK pro-inflammatory signaling. In the mice model of dextran sulfate sodium (DSS)-induced IBD, the multipronged manner of scavenging oxidative stress, remodeling innate and adaptive immune homeostasis, and reshaping gut microbiota caused by YMD@MPDA effectively ameliorated inflammation and restored intestinal barrier functions. Overall, the YMD@MPDA nanocomposite provides a promising codelivery strategy of antioxidative nanozymes and gas prodrugs for the comprehensive management of IBD.
Collapse
Affiliation(s)
- Xu Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
| | - Huan Yang
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Ye He
- Thomas Lord Department of Mechanical Engineering and Materials Science, Duke University, Durham, North Carolina 27708, United States
| | - Dan Zhang
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
| | - Guifang Lu
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
| | - Mudan Ren
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
| | - Yi Lyu
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an 710061, P. R. China
| | - Zhang Yuan
- Xi'an Key Laboratory of Stem Cell and Regenerative Medicine, Institute of Medical Research, Northwestern Polytechnical University, 127 West Youyi Road, Beilin District, Xi'an, Shaanxi 710072, P. R. China
| | - Shuixiang He
- Department of Gastroenterology, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
- National Local Joint Engineering Research Center for Precision Surgery and Regenerative Medicine, The First Affiliated Hospital of Xi'an Jiaotong University, No. 277, West Yanta Road, Yanta District, Xi'an, Shaanxi 710061, P. R. China
| |
Collapse
|
18
|
Jiang B, Peng C, Li X, Sun C, Lu W, Fang Y. Banana Starch Nanoparticles Disrupt the Integrity of the Intestinal Barrier by Opening Tight Junctions in Mice. SMALL (WEINHEIM AN DER BERGSTRASSE, GERMANY) 2025; 21:e2408298. [PMID: 39668449 DOI: 10.1002/smll.202408298] [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: 09/12/2024] [Revised: 12/04/2024] [Indexed: 12/14/2024]
Abstract
The banana-derived starch nanoparticles have been extensively used in food and biomedicine industries, due to their unique physicochemical properties and functional benefits. With their pervasive presence in food, people are significantly exposed to these nanoparticles, raising concerns about their potential health risks and impact on intestinal health. However, there is still limited understanding of the direct interaction between native banana starch nanoparticles (BSNs) and the intestinal systems. Here, it is demonstrated that BSNs can cause tight junctions to loosen, increase intestinal permeability, and disrupt the intestinal barrier. This increased permeability is closely linked to the size of BSNs, with smaller BSNs (d = 60 nm) having a stronger effect on permeation. Furthermore, the disruption of the intestinal barrier integrity caused by BSNs is connected to a reduced amount of tight junction proteins. Mechanistically, BSNs disrupt tight junctions by affecting mitochondrial function and activating myosin light chain kinase (MLCK) signaling pathway. These findings indicate that BSNs have the potential to pose health risks by compromising the integrity of the intestinal barrier, reminding the safety of food biopolymer nanoparticles in living organisms needs to be re-assessed.
Collapse
Affiliation(s)
- Bing Jiang
- Department of food science and engineering, School of agriculture and biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Chenglu Peng
- Department of food science and engineering, School of agriculture and biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Xiaoyang Li
- Department of food science and engineering, School of agriculture and biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Cuixia Sun
- Department of food science and engineering, School of agriculture and biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Wei Lu
- Department of food science and engineering, School of agriculture and biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Yapeng Fang
- Department of food science and engineering, School of agriculture and biology, Shanghai Jiao Tong University, Shanghai, 200240, China
| |
Collapse
|
19
|
Yang Y, Chen Q, Liu Z, Huang T, Hong Y, Li N, Ai K, Huang Q. Novel reduced heteropolyacid nanoparticles for effective treatment of drug-induced liver injury by manipulating reactive oxygen and nitrogen species and inflammatory signals. J Colloid Interface Sci 2025; 678:174-187. [PMID: 39243718 DOI: 10.1016/j.jcis.2024.08.239] [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: 06/14/2024] [Revised: 08/07/2024] [Accepted: 08/29/2024] [Indexed: 09/09/2024]
Abstract
With the rapid advancements in biomedicine, the use of clinical drugs has surged sharply. However, potential hepatotoxicity limits drug exploitation and widespread usage, posing serious threats to patient health. Hepatotoxic drugs disrupt liver enzyme levels and cause refractory pathological damage, creating a challenge in the application of diverse first-line drugs. The activation and deterioration of reactive oxygen and nitrogen species (RONS) and inflammatory signals are key pathological mechanisms of drug-induced liver injury (DILI). Herein, a novel reduced heteropolyacid nanoparticle (RNP) has been developed, possessing high RONS-scavenging ability, strong anti-inflammatory activity, and excellent biosafety. These features enable it to swiftly restore the redox and immune balance of the liver. Intravenous administration of RNP effectively scavenged RONS storm, reversing liver oxidative stress and restoring normal mitochondrial membrane potential and function. Furthermore, by inhibiting c-Jun-N-terminal kinase phosphorylation, RNP facilitated the restoration of nuclear factor erythroid 2-related factor 2-mediated endogenous antioxidant signaling, ultimately rescuing the liver function and tissue morphology in acetaminophen-induced DILI mice. Crucially, the high biocompatible RNP exhibited superior efficacy in the DILI mouse model compared to the clinical antioxidant N-acetylcysteine. This targeted therapeutic approach, tailored to address the onset and progression of DILI, offers valuable new insights into controlling the condition and restoring liver structure and function.
Collapse
Affiliation(s)
- Yongqi Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Qiaohui Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Zerun Liu
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ting Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Ying Hong
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Niansheng Li
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China.
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China; Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Qiong Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha 410008, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China.
| |
Collapse
|
20
|
Zhang X, Zuo L, Song X, Zhang W, Yang Z, Wang Z, Guo Y, Ge S, Wang L, Wang Y, Geng Z, Li J, Hu J. The mesenteric adipokine SFRP5 alleviated intestinal epithelial apoptosis improving barrier dysfunction in Crohn's disease. iScience 2024; 27:111517. [PMID: 39759008 PMCID: PMC11699250 DOI: 10.1016/j.isci.2024.111517] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2024] [Revised: 09/21/2024] [Accepted: 11/28/2024] [Indexed: 01/07/2025] Open
Abstract
The hypertrophic mesenteric adipose tissue (htMAT) of Crohn disease (CD) participates in inflammation through the expression of adipokines, but the exact mechanism of this action in the intestine is unknown. Here, we analyzed the expression of secreted frizzled-related protein 5 (SFRP5), an adipokine with cytoprotective effects, in htMAT and its role in CD. The results of this study revealed that the level of SFPR5 increased in the diseased MAT (htMAT) of CD patients and aggregated among intestinal epithelial cells in the diseased intestine and that it could ameliorate intestinal barrier dysfunction in tumor necrosis factor alpha (TNF-α)-stimulated colonic organoids and 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced mice at least in part through the inhibition of Wnt5a-mediated apoptosis in epithelial cells. This study elucidates possible mechanisms by which mesenteric adipokines influence the progression of enteritis and provides a new theoretical basis for the treatment of CD via the mesenteric pathway.
Collapse
Affiliation(s)
- Xiaofeng Zhang
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Lugen Zuo
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Xue Song
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Wenjing Zhang
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Zi Yang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Zhiyuan Wang
- Clinical Medical College, Bengbu Medical University, Bengbu, China
| | - Yibing Guo
- Clinical Medical College, Bengbu Medical University, Bengbu, China
| | - Sitang Ge
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Lian Wang
- Department of Gastrointestinal Surgery, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Yueyue Wang
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Zhijun Geng
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Jing Li
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Jianguo Hu
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-related Diseases, Bengbu, China
- Inflammatory Bowel Disease Research Center, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| |
Collapse
|
21
|
Huai M, Pei M, Chen J, Duan X, Zhu Y, Yang F, Ge W. Oral creatine-modified selenium-based hyaluronic acid nanogel mediated mitochondrial energy recovery to drive the treatment of inflammatory bowel disease. J Nanobiotechnology 2024; 22:740. [PMID: 39609811 PMCID: PMC11603945 DOI: 10.1186/s12951-024-03007-0] [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: 06/18/2024] [Accepted: 11/11/2024] [Indexed: 11/30/2024] Open
Abstract
The damnification of mitochondrion is often considered to be an important culprit of inflammatory bowel disease (IBD), however, there are fewer reports of mechanisms of mitochondria-mediated IBD treatment. Therefore, we first proposed to reboot mitochondrial energy metabolism to treat IBD by capturing the double-sided factor of ROS and creatine (Cr)-assisted energy adjustment. Herein, an oral Cr-modified selenium-based hyaluronic acid (HA) nanogel (HASe-Cr nanogel) was fabricated for treatment of IBD, through ROS elimination and energy metabolism upgradation. More concretely, due to IBD lesion-specific positive charge and the high expression of CD44, HASe-Cr nanogel exhibited dual targeted inflammatory bio-functions, and ROS-driven degradation properties in high-yield ROS levels in inflammation areas. As expected, multifunctional HASe-Cr nanogel could effectively ameliorate IBD-related symptoms, such as mitochondrial biological function restoration, inhibition of M1-like macrophage polarization, gut mucosal reconstruction, microbial ecological repair, etc., thus excellently treating IBD. Overall, the proposed strategy underlined that the great potentiality of HASe-Cr nanogel by restarting mitochondrial metabolic energy in colitis lesions, providing new a pavement of mitochondrion-mediated colitis treatment in clinical applications.
Collapse
Affiliation(s)
- Manxiu Huai
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, P. R. China
| | - Mingliang Pei
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Jie Chen
- Shanghai Chest Hospital, Shanghai Jiao Tong University School of Medicine, No.241 West Huaihai Road, Shanghai, 200030, P. R. China
| | - Xiaoyan Duan
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, P. R. China
| | - Yun Zhu
- Department of Oral and Maxillofacial Surgery, Zhongshan Hospital, Fudan University, Shanghai, China
| | - Fan Yang
- Department of Orthopaedics, Shanghai Key Laboratory for Prevention and Treatment of Bone and Joint Diseases, Shanghai Institute of Traumatology and Orthopaedics, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, 200025, China.
| | - Wensong Ge
- Department of Gastroenterology, Xinhua Hospital, Shanghai Jiaotong University School of Medicine, No. 1665 Kongjiang Road, Shanghai, 200092, P. R. China.
| |
Collapse
|
22
|
Feig VR, Zhang S, Patel A, Santos B, Kang Z, Wasan S, Beloqui A, Traverso G. Designing for medication adherence in inflammatory bowel disease: multi-disciplinary approaches for self-administrable biotherapeutics. EClinicalMedicine 2024; 77:102850. [PMID: 39763512 PMCID: PMC11701474 DOI: 10.1016/j.eclinm.2024.102850] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 09/05/2024] [Accepted: 09/10/2024] [Indexed: 03/15/2025] Open
Abstract
Biotherapeutics are among the therapeutics that have revolutionized standard inflammatory bowel disease (IBD) treatment, which was previously limited to mesalamine, 5-aminosalicylic acid, corticosteroids, and classical immunosuppressants. Self-administrable biotherapeutics for IBD would enable home-based treatment and reduce the burden on medical infrastructure. Self-administration is made possible through subcutaneous injectable, oral, and rectal dosage forms. Nevertheless, the full benefits of self-administration cannot be realized without first addressing the issue of medication adherence, which remains woefully inadequate for IBD biotherapies. Some of the major barriers to medication adherence in IBD are the route of administration, frequency of administration, and undesired side effects. In this review, we identify the main physiological and engineering constraints that underlie these three barriers to adherence. We then highlight key technological and behavioral innovations-spanning multiple scientific disciplines-that can be leveraged to design novel therapies and interventions that improve adherence to self-administered IBD biotherapies.
Collapse
Affiliation(s)
- Vivian Rachel Feig
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Mechanical Engineering, Stanford University, Stanford, CA, USA
| | - Sufeng Zhang
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Biomedical Engineering, Stony Brook University, Stony Brook, NY, USA
| | - Ashka Patel
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Department of Bioengineering, Northeastern University, Boston, MA, USA
| | - Bruna Santos
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Ziliang Kang
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Boston, MA, USA
| | - Sharmeel Wasan
- Department of Gastroenterology, Boston Medical Center, Boston, MA, USA
| | - Ana Beloqui
- Advanced Drug Delivery and Biomaterials, Louvain Drug Research Institute, Université catholique de Louvain, Brussels, Belgium
- WEL Research Institute, Wavre, Belgium
| | - Giovanni Traverso
- Division of Gastroenterology, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
- Koch Institute for Integrative Cancer Research, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Mechanical Engineering, Massachusetts Institute of Technology, Boston, MA, USA
| |
Collapse
|
23
|
Chi X, Chen T, Luo F, Zhao R, Li Y, Hu S, Li Y, Jiang W, Chen L, Wu D, Du Y, Hu J. Targeted no-releasing L-arginine-induced hesperetin self-assembled nanoparticles for ulcerative colitis intervention. Acta Biomater 2024:S1742-7061(24)00628-7. [PMID: 39461688 DOI: 10.1016/j.actbio.2024.10.037] [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: 07/19/2024] [Revised: 10/07/2024] [Accepted: 10/23/2024] [Indexed: 10/29/2024]
Abstract
Overproduction of reactive oxygen species (ROS) plays a crucial role in initiating and advancing ulcerative colitis (UC), and the persistent cycle between ROS and inflammation accelerates disease development. Therefore, developing strategies that can effectively scavenge ROS and provide targeted intervention are crucial for the management of UC. In this study, we synthesized natural carrier-free nanoparticles (HST-Arg NPs) using the Mannich reaction and π-π stacking for the intervention of UC. HST-Arg NPs are an oral formulation that exhibit good antioxidant capabilities and gastrointestinal stability. Benefiting from the negatively charged characteristics, HST-Arg NPs can specifically accumulate in positively charged inflamed regions of the colon. Furthermore, in the oxidative microenvironment of colonic inflammation, HST-Arg NPs respond to ROS by releasing nitric oxide (NO). In mice model of UC induced by dextran sulfate sodium (DSS), HST-Arg NPs significantly mitigated colonic injury by modulating oxidative stress, lowering pro-inflammatory cytokines, and repairing intestinal barrier integrity. In summary, this convenient and targeted oral nanoparticle can effectively scavenge ROS at the site of inflammation and achieve gas intervention, offering robust theoretical support for the development of subsequent oral formulations in related inflammatory interventions. STATEMENT OF SIGNIFICANCE: Nanotechnology has been extensively explored in the biomedical field, but the application of natural carrier-free nanotechnology in this area remains relatively rare. In this study, we developed a natural nanoparticle system based on hesperetin (HST), L-arginine (L-Arg), and vanillin (VA) to scavenge ROS and alleviate inflammation. In the context of ulcerative colitis (UC), the synthesized nanoparticles exhibited excellent intervention effects, effectively protecting the colon from damage. Consequently, these nanoparticles provide a promising and precise nutritional intervention strategy by addressing both oxidative stress and inflammatory pathways simultaneously, demonstrating significant potential for application.
Collapse
Affiliation(s)
- Xuesong Chi
- 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 116034, 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 116034, 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 116034, China
| | - Runan Zhao
- 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 116034, China; College of Biosystems Engineering and Food Science, Fuli Institute of Food Science, Zhejiang University, Hangzhou 310058, China
| | - Yangjing Li
- 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 116034, China
| | - Shumeng 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 116034, China; College of Food Science and Engineering, Jilin Agricultural University, Changchun 130118, China
| | - Yanfei Li
- 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 116034, China
| | - Wen Jiang
- 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 116034, 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 116034, 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 116034, China
| | - Yinan Du
- 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 116034, 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 116034, China.
| |
Collapse
|
24
|
Fan S, Zhao Y, Yao Y, Shen X, Chai X, Li J, Pi J, Huang X, Jin H, Zhou Z. Oral colon-targeted pH-responsive polymeric nanoparticles loading naringin for enhanced ulcerative colitis therapy. J Transl Med 2024; 22:878. [PMID: 39350164 PMCID: PMC11440766 DOI: 10.1186/s12967-024-05662-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/09/2024] [Indexed: 10/04/2024] Open
Abstract
An oral colon-targeted drug delivery system holds great potential in preventing systemic toxicity and preserving the therapeutic benefits of ulcerative colitis (UC) treatment. In this study, we developed a negatively charged PLGA-PEG nanoparticle system for encapsulating naringin (Nar). Additionally, chitosan and mannose were coated on the surface of these nanoparticles to enhance their mucosal adsorption and macrophage targeting abilities. The resulting nanoparticles, termed MC@Nar-NPs, exhibited excellent resistance against decomposition in the strong acidic gastrointestinal environment and specifically accumulated at inflammatory sites. Upon payload release, MC@Nar-NPs demonstrated remarkable efficacy in alleviating colon inflammation as evidenced by reduced levels of pro-inflammatory cytokines in both blood and colon tissues, as well as the scavenging of reactive oxygen species (ROS) in the colon. This oral nanoparticle delivery system represents a novel approach to treating UC by utilizing Chinese herbal ingredient-based oral delivery and provides a theoretical foundation for local and precise intervention in specific UC treatment.
Collapse
Affiliation(s)
- Shilong Fan
- Guangdong Provincial Key laboratory of research and development of Natural drugs, School of Pharmacology, Guangdong Medical University, Dongguan, 523808, China
- Research Center of Nano Technology and Application Engineering, Dongguan Innovation Institute, Guangdong Medical University, Dongguan, 523808, China
| | - Yue Zhao
- Guangdong Provincial Key laboratory of research and development of Natural drugs, School of Pharmacology, Guangdong Medical University, Dongguan, 523808, China
- Research Center of Nano Technology and Application Engineering, Dongguan Innovation Institute, Guangdong Medical University, Dongguan, 523808, China
| | - Yinlian Yao
- Guangdong Provincial Key laboratory of research and development of Natural drugs, School of Pharmacology, Guangdong Medical University, Dongguan, 523808, China
- Research Center of Nano Technology and Application Engineering, Dongguan Innovation Institute, Guangdong Medical University, Dongguan, 523808, China
| | - Xin Shen
- Guangdong Provincial Key laboratory of research and development of Natural drugs, School of Pharmacology, Guangdong Medical University, Dongguan, 523808, China
- Research Center of Nano Technology and Application Engineering, Dongguan Innovation Institute, Guangdong Medical University, Dongguan, 523808, China
| | - Xingxing Chai
- Laboratory Animal Center, School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China
| | - Jiahui Li
- Guangdong Provincial Key laboratory of research and development of Natural drugs, School of Pharmacology, Guangdong Medical University, Dongguan, 523808, China
- Research Center of Nano Technology and Application Engineering, Dongguan Innovation Institute, Guangdong Medical University, Dongguan, 523808, China
| | - Jiang Pi
- Laboratory Animal Center, School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China
| | - Xueqin Huang
- Laboratory Animal Center, School of Medical Technology, Guangdong Medical University, Dongguan, 523808, China
| | - Hua Jin
- Guangdong Provincial Key laboratory of research and development of Natural drugs, School of Pharmacology, Guangdong Medical University, Dongguan, 523808, China.
- Research Center of Nano Technology and Application Engineering, Dongguan Innovation Institute, Guangdong Medical University, Dongguan, 523808, China.
| | - Zhikun Zhou
- Guangdong Provincial Key laboratory of research and development of Natural drugs, School of Pharmacology, Guangdong Medical University, Dongguan, 523808, China.
- Research Center of Nano Technology and Application Engineering, Dongguan Innovation Institute, Guangdong Medical University, Dongguan, 523808, China.
| |
Collapse
|
25
|
Feng G, Zhang H, Liu H, Zhang X, Jiang H, Liao S, Luo X, Yao H, Xiang B, Liu S, Zhang J, Zhang J, Fang J. Natural Flavonoid-Derived Enzyme Mimics DHKNase Balance the Two-Edged Reactive Oxygen Species Function for Wound Healing and Inflammatory Bowel Disease Therapy. RESEARCH (WASHINGTON, D.C.) 2024; 7:0464. [PMID: 39253100 PMCID: PMC11381673 DOI: 10.34133/research.0464] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/07/2024] [Indexed: 09/11/2024]
Abstract
Rational regulation of reactive oxygen species (ROS) plays a vital importance in maintaining homeostasis of living biological systems. For ROS-related pathologies, chemotherapy technology derived from metal nanomaterials currently occupies a pivotal position. However, they suffer from inherent issues such as complicated synthesis, batch-to-batch variability, high cost, and potential biological toxicity caused by metal elements. Here, we reported for the first time that dual-action 3,5-dihydroxy-1-ketonaphthalene-structured small-molecule enzyme imitator (DHKNase) exhibited 2-edged ROS regulation, catering to the execution of physiology-beneficial ROS destiny among diverse pathologies in living systems. Based on this, DHKNase is validated to enable remarkable therapeutic effects in 2 classic disease models, including the pathogen-infected wound-healing model and the dextran sulfate sodium (DSS)-caused inflammatory bowel disease (IBD). This work provides a guiding landmark for developing novel natural small-molecule enzyme imitator and significantly expands their application potential in the biomedical field.
Collapse
Affiliation(s)
- Guangfu Feng
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| | - Huaizu Zhang
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| | - Huipeng Liu
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| | - Xiaoyan Zhang
- College of Life Science, Shihezi University, Shihezi, Xinjiang 832003, P.R. China
| | - Hongmei Jiang
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| | - Sijie Liao
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| | - Xingyu Luo
- School of Chemistry and Chemical Engineering, Guangxi University, Nanning 530004, P.R. China
| | - Hao Yao
- Changsha IMADEK Intelligent Technology Co. Ltd., Changsha, Hunan 410081, P.R. China
| | - Bo Xiang
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| | - Shiyu Liu
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| | - Jiali Zhang
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| | - Jiaheng Zhang
- College of Chemistry, Food Laboratory of Zhongyuan, Flavour Science Research Center of Zhengzhou University, Zhengzhou University, Zhengzhou, Henan 450001, P.R. China
| | - Jun Fang
- School of Bioscience and Biotechnology, Hunan Agricultural University, Changsha, Hunan 410128, P.R. China
| |
Collapse
|
26
|
Deng B, Liu Y, Chen Y, He P, Ma J, Tan Z, Zhang J, Dong W. Exploring the butyrate metabolism-related shared genes in metabolic associated steatohepatitis and ulcerative colitis. Sci Rep 2024; 14:15949. [PMID: 38987612 PMCID: PMC11237055 DOI: 10.1038/s41598-024-66574-0] [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: 03/20/2024] [Accepted: 07/02/2024] [Indexed: 07/12/2024] Open
Abstract
Metabolic-associated steatohepatitis (MASH) and ulcerative colitis (UC) exhibit a complex interconnection with immune dysfunction, dysbiosis of the gut microbiota, and activation of inflammatory pathways. This study aims to identify and validate critical butyrate metabolism-related shared genes between both UC and MASH. Clinical information and gene expression profiles were sourced from the Gene Expression Omnibus (GEO) database. Shared butyrate metabolism-related differentially expressed genes (sBM-DEGs) between UC and MASH were identified via various bioinformatics methods. Functional enrichment analysis was performed, and UC patients were categorized into subtypes using the consensus clustering algorithm based on sBM-DEGs. Key genes within sBM-DEGs were screened through Random Forest, Support Vector Machines-Recursive Feature Elimination, and Light Gradient Boosting. The diagnostic efficacy of these genes was evaluated using receiver operating characteristic (ROC) analysis on independent datasets. Additionally, the expression levels of characteristic genes were validated across multiple independent datasets and human specimens. Forty-nine shared DEGs between UC and MASH were identified, with enrichment analysis highlighting significant involvement in immune, inflammatory, and metabolic pathways. The intersection of butyrate metabolism-related genes with these DEGs produced 10 sBM-DEGs. These genes facilitated the identification of molecular subtypes of UC patients using an unsupervised clustering approach. ANXA5, CD44, and SLC16A1 were pinpointed as hub genes through machine learning algorithms and feature importance rankings. ROC analysis confirmed their diagnostic efficacy in UC and MASH across various datasets. Additionally, the expression levels of these three hub genes showed significant correlations with immune cells. These findings were validated across independent datasets and human specimens, corroborating the bioinformatics analysis results. Integrated bioinformatics identified three significant biomarkers, ANXA5, CD44, and SLC16A1, as DEGs linked to butyrate metabolism. These findings offer new insights into the role of butyrate metabolism in the pathogenesis of UC and MASH, suggesting its potential as a valuable diagnostic biomarker.
Collapse
Affiliation(s)
- Beiying Deng
- Department of Gastroenterology, Renmin Hospital of Wuhan University, No 99 Zhangzhidong Road, Wuhan, 430060, Hubei Province, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Yinghui Liu
- Department of Geriatric, Renmin Hospital of Wuhan University, Wuhan, China
| | - Ying Chen
- Department of Gastroenterology, Renmin Hospital of Wuhan University, No 99 Zhangzhidong Road, Wuhan, 430060, Hubei Province, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Pengzhan He
- Department of Gastroenterology, Renmin Hospital of Wuhan University, No 99 Zhangzhidong Road, Wuhan, 430060, Hubei Province, China
- Central Laboratory, Renmin Hospital of Wuhan University, Wuhan, China
| | - Jingjing Ma
- Department of Geriatric, Renmin Hospital of Wuhan University, Wuhan, China
| | - Zongbiao Tan
- Department of Gastroenterology, Renmin Hospital of Wuhan University, No 99 Zhangzhidong Road, Wuhan, 430060, Hubei Province, China
| | - Jixiang Zhang
- Department of Gastroenterology, Renmin Hospital of Wuhan University, No 99 Zhangzhidong Road, Wuhan, 430060, Hubei Province, China
| | - Weiguo Dong
- Department of Gastroenterology, Renmin Hospital of Wuhan University, No 99 Zhangzhidong Road, Wuhan, 430060, Hubei Province, China.
| |
Collapse
|
27
|
Ma C, Xu C, Zhang T, Mu Q, Lv J, Xing Q, Yang Z, Xu Z, Guan Y, Chen C, Ni K, Dai X, Ding W, Hu J, Bao Z, Wang S, Liu P. Tracking the hologenome dynamics in aquatic invertebrates by the holo-2bRAD approach. Commun Biol 2024; 7:827. [PMID: 38972908 PMCID: PMC11228047 DOI: 10.1038/s42003-024-06509-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Accepted: 06/26/2024] [Indexed: 07/09/2024] Open
Abstract
The "hologenome" concept is an increasingly popular way of thinking about microbiome-host for marine organisms. However, it is challenging to track hologenome dynamics because of the large amount of material, with tracking itself usually resulting in damage or death of the research object. Here we show the simple and efficient holo-2bRAD approach for the tracking of hologenome dynamics in marine invertebrates (i.e., scallop and shrimp) from one holo-2bRAD library. The stable performance of our approach was shown with high genotyping accuracy of 99.91% and a high correlation of r > 0.99 for the species-level profiling of microorganisms. To explore the host-microbe association underlying mass mortality events of bivalve larvae, core microbial species changed with the stages were found, and two potentially associated host SNPs were identified. Overall, our research provides a powerful tool with various advantages (e.g., cost-effective, simple, and applicable for challenging samples) in genetic, ecological, and evolutionary studies.
Collapse
Affiliation(s)
- Cen Ma
- Fang Zongxi Center for Marine Evo-Devo & MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Laoshan Laboratory, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory, Qingdao, China
| | - Chang Xu
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya, China
| | - Tianqi Zhang
- Fang Zongxi Center for Marine Evo-Devo & MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Qianqian Mu
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya, China
| | - Jia Lv
- Fang Zongxi Center for Marine Evo-Devo & MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Laoshan Laboratory, Qingdao, China
| | - Qiang Xing
- Laboratory for Marine Biology and Biotechnology, Laoshan Laboratory, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory, Qingdao, China
| | - Zhihui Yang
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya, China
| | - Zhenyuan Xu
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya, China
| | - Yalin Guan
- Fang Zongxi Center for Marine Evo-Devo & MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Chengqin Chen
- Fang Zongxi Center for Marine Evo-Devo & MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Kuo Ni
- Fang Zongxi Center for Marine Evo-Devo & MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Xiaoting Dai
- Department of Molecular & Integrative Physiology, University of Michigan, Ann Arbor, MI, USA
- Institute of Gerontology, Geriatrics Center, University of Michigan, Ann Arbor, MI, USA
| | - Wei Ding
- Fang Zongxi Center for Marine Evo-Devo & MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China
| | - Jingjie Hu
- Fang Zongxi Center for Marine Evo-Devo & MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya, China
| | - Zhenmin Bao
- Fang Zongxi Center for Marine Evo-Devo & MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China
- Laboratory for Marine Fisheries Science and Food Production Processes, Laoshan Laboratory, Qingdao, China
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya, China
| | - Shi Wang
- Fang Zongxi Center for Marine Evo-Devo & MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China
- Laboratory for Marine Biology and Biotechnology, Laoshan Laboratory, Qingdao, China
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya, China
| | - Pingping Liu
- Fang Zongxi Center for Marine Evo-Devo & MOE Key Laboratory of Marine Genetics and Breeding, College of Marine Life Sciences, Ocean University of China, Qingdao, China.
- Laboratory for Marine Biology and Biotechnology, Laoshan Laboratory, Qingdao, China.
- Key Laboratory of Tropical Aquatic Germplasm of Hainan Province, Sanya Oceanographic Institution, Ocean University of China, Sanya, China.
| |
Collapse
|
28
|
Luo D, Liu X, Dai S, Yi J, Tang N, Cai Y, Bao X, Hu M, Liu Z. Highly Crystalline Copper Aluminum-Layered Double Hydroxides with Intrinsic Fenton-Like Catalytic Activity for Robust Oral Health Management. Inorg Chem 2024; 63:10691-10704. [PMID: 38805682 DOI: 10.1021/acs.inorgchem.4c01189] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
As the main challenge of dental healthcare, oral infectious diseases are highly associated with the colonization of pathogenic microbes. However, current antibacterial treatments in the field of stomatology still lack a facile, safe, and universal approach. Herein, we report the controllable synthesis of copper aluminum-layered double hydroxides (CuAl-LDHs) with high Fenton-like catalytic activity, which can be utilized in the treatment of oral infectious diseases with negligible side effects. Our strategy can efficiently avoid the unwanted doping of other divalent metal ions in the synthesis of Cu-contained LDHs and result in the formation of binary CuAl-LDHs with high crystallinity and purity. Evidenced by experimental and theoretical results, CuAl-LDHs exhibit excellent catalytic ability toward the ·OH generation in the presence of H2O2 and hold strong affinity toward bacteria, endowing them with great catalytic sterilization against both Gram-positive and Gram-negative bacteria. As expected, these CuAl-LDHs provide outstanding treatments for mucosal infection and periodontitis by promoting wound healing and remodeling of the periodontal microenvironment. Moreover, toxicity investigation demonstrates the overall safety. Accordingly, the current study not only provides a convenient and economic strategy for treating oral infectious diseases but also extends the development of novel LDH-based Fenton or Fenton-like antibacterial reagents for further biomedical applications.
Collapse
Affiliation(s)
- Danfeng Luo
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Xiaocan Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Shuang Dai
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Jingzheng Yi
- Western Dental, Fresno, California 93726, United States
| | - Nan Tang
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| | - Yanting Cai
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Xingfu Bao
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Min Hu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
| | - Zhen Liu
- Jilin Provincial Key Laboratory of Tooth Development and Bone Remodeling, School and Hospital of Stomatology, Jilin University, Changchun 130021, China
- Beijing Advanced Innovation Center for Soft Matter Science and Engineering, College of Life Science and Technology, Beijing University of Chemical Technology, Beijing 100029, China
| |
Collapse
|
29
|
Chen T, Meng W, Li Y, Li X, Yu X, Qi J, Ding D, Li W. Probiotics Armed with In Situ Mineralized Nanocatalysts and Targeted Biocoatings for Multipronged Treatment of Inflammatory Bowel Disease. NANO LETTERS 2024. [PMID: 38787330 DOI: 10.1021/acs.nanolett.4c01143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/25/2024]
Abstract
While oral probiotics show promise in treating inflammatory bowel disease, the primary challenge lies in sustaining their activity and retention within the inflamed gastrointestinal environment. In this work, we develop an engineered probiotic platform that is armed with biocatalytic and inflamed colon-targeting nanocoatings for multipronged management of IBD. Notably, we achieve the in situ growth of artificial nanocatalysts on probiotics through a bioinspired mineralization strategy. The resulting ferrihydrite nanostructures anchored on bacteria exhibit robust catalase-like activity across a broad pH range, effectively scavenging ROS to alleviate inflammation. The further envelopment with fucoidan-based shields confers probiotics with additional inflamed colon-targeting functions. Upon oral administration, the engineered probiotics display markedly improved viability and colonization within the inflamed intestine, and they further elicit boosted prophylactic and therapeutic efficacy against colitis through the synergistic interplay of nanocatalysis-based immunomodulation and probiotics-mediated microbiota reshaping. The robust and multifunctional probiotic platforms offer great potential for the comprehensive management of gastrointestinal disorders.
Collapse
Affiliation(s)
- Ting Chen
- Tianjin Key Laboratory of Biomedical Materials and Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Wen Meng
- Tianjin Key Laboratory of Biomedical Materials and Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Yi Li
- Tianjin Key Laboratory of Biomedical Materials and Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Xueping Li
- Tianjin Key Laboratory of Biomedical Materials and Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Xuya Yu
- Tianjin Key Laboratory of Biomedical Materials and Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Ji Qi
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Frontiers Science Center for Cell Responses, and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Dan Ding
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials, Ministry of Education, Frontiers Science Center for Cell Responses, and College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Wen Li
- Tianjin Key Laboratory of Biomedical Materials and Key Laboratory of Biomaterials and Nanotechnology for Cancer Immunotherapy, Institute of Biomedical Engineering, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| |
Collapse
|
30
|
Fu YJ, Zhao X, Wang LY, Li K, Jiang N, Zhang ST, Wang RK, Zhao YF, Yang W. A Gas Therapy Strategy for Intestinal Flora Regulation and Colitis Treatment by Nanogel-Based Multistage NO Delivery Microcapsules. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2024; 36:e2309972. [PMID: 38324725 DOI: 10.1002/adma.202309972] [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: 09/25/2023] [Revised: 01/28/2024] [Indexed: 02/09/2024]
Abstract
Current approaches to treating inflammatory bowel disease focus on the suppression of overactive immune responses, the removal of reactive intestinal oxygen species, and regulation of the intestinal flora. However, owing to the complex structure of the gastrointestinal tract and the influence of mucus, current small-molecule and biologic-based drugs for treating colitis cannot effectively act at the site of colon inflammation, and as a result, they tend to exhibit low efficacies and toxic side effects. In this study, nanogel-based multistage NO delivery microcapsules are developed to achieve NO release at the inflammation site by targeting the inflammatory tissues using the nanogel. Surprisingly, oral administration of the microcapsules suppresses the growth of pathogenic bacteria and increases the abundance of probiotic bacteria. Metabolomics further show that an increased abundance of intestinal probiotics promotes the production of metabolites, including short-chain fatty acids and indole derivatives, which modulate the intestinal immunity and restore the intestinal barrier via the interleukin-17 and PI3K-Akt signaling pathways. This work reveals that the developed gas therapy strategy based on multistage NO delivery microcapsules modulates the intestinal microbial balance, thereby reducing inflammation and promoting intestinal barrier repair, ultimately providing a new therapeutic approach for the clinical management of colitis.
Collapse
Affiliation(s)
- Ya-Jun Fu
- College of Polymer Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Xing Zhao
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Li-Ya Wang
- Department of Nephrology, Kidney Research Institute, West China Hospital of Sichuan University, Chengdu, 610041, China
| | - Kai Li
- Division of Thoracic Tumor Multimodality Treatment, Cancer Center, West China Hospital, Sichuan University, Chengdu, 610041, China
| | - Niu Jiang
- College of Polymer Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Shu-Ting Zhang
- College of Polymer Science and Engineering, Sichuan University, Chengdu, 610065, China
| | - Rao-Kaijuan Wang
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610032, China
| | - Yi-Fan Zhao
- Department of Orthodontics, West China Hospital of Stomatology, Sichuan University, Chengdu, 610032, China
| | - Wei Yang
- College of Polymer Science and Engineering, Sichuan University, Chengdu, 610065, China
| |
Collapse
|
31
|
Jin T, Lu H, Zhou Q, Chen D, Zeng Y, Shi J, Zhang Y, Wang X, Shen X, Cai X. H 2S-Releasing Versatile Montmorillonite Nanoformulation Trilogically Renovates the Gut Microenvironment for Inflammatory Bowel Disease Modulation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2308092. [PMID: 38308198 PMCID: PMC11005690 DOI: 10.1002/advs.202308092] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2023] [Revised: 01/20/2024] [Indexed: 02/04/2024]
Abstract
Abnormal activation of the intestinal mucosal immune system, resulting from damage to the intestinal mucosal barrier and extensive invasion by pathogens, contributes to the pathogenesis of inflammatory bowel disease (IBD). Current first-line treatments for IBD have limited efficacy and significant side effects. An innovative H2S-releasing montmorillonite nanoformulation (DPs@MMT) capable of remodeling intestinal mucosal immune homeostasis, repairing the mucosal barrier, and modulating gut microbiota is developed by electrostatically adsorbing diallyl trisulfide-loaded peptide dendrimer nanogels (DATS@PDNs, abbreviated as DPs) onto the montmorillonite (MMT) surface. Upon rectal administration, DPs@MMT specifically binds to and covers the damaged mucosa, promoting the accumulation and subsequent internalization of DPs by activated immune cells in the IBD site. DPs release H2S intracellularly in response to glutathione, initiating multiple therapeutic effects. In vitro and in vivo studies have shown that DPs@MMT effectively alleviates colitis by eliminating reactive oxygen species (ROS), inhibiting inflammation, repairing the mucosal barrier, and eradicating pathogens. RNA sequencing revealed that DPs@MMT exerts significant immunoregulatory and mucosal barrier repair effects, by activating pathways such as Nrf2/HO-1, PI3K-AKT, and RAS/MAPK/AP-1, and inhibiting the p38/ERK MAPK, p65 NF-κB, and JAK-STAT3 pathways, as well as glycolysis. 16S rRNA sequencing demonstrated that DPs@MMT remodels the gut microbiota by eliminating pathogens and increasing probiotics. This study develops a promising nanoformulation for IBD management.
Collapse
Affiliation(s)
- Ting Jin
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| | - Hongyang Lu
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| | - Qiang Zhou
- Department of OtolaryngologyRuian People's HospitalThe Third Affiliated Hospital of Wenzhou Medical UniversityWenzhou325016China
| | - Dongfan Chen
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| | - Youyun Zeng
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| | - Jiayi Shi
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| | - Yanmei Zhang
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| | - Xianwen Wang
- School of Biomedical EngineeringResearch and Engineering Center of Biomedical MaterialsAnhui Medical UniversityHefei230032China
| | - Xinkun Shen
- Department of OtolaryngologyRuian People's HospitalThe Third Affiliated Hospital of Wenzhou Medical UniversityWenzhou325016China
| | - Xiaojun Cai
- School and Hospital of StomatologyWenzhou Medical UniversityWenzhou325027China
| |
Collapse
|
32
|
Xu T, Ning X, Wu J, Wang Q, Wang Z, Chen Z, Tang X, Bai P, Pu K, Li L, Zhang R. Metabolic Nanoregulator Remodels Gut Microenvironment for Treatment of Inflammatory Bowel Disease. ACS NANO 2024; 18:7123-7135. [PMID: 38390866 DOI: 10.1021/acsnano.3c11496] [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: 02/24/2024]
Abstract
Inflammatory bowel disease (IBD) is strongly related to the occurrence of accumulation of toxic reactive oxygen species (ROS), inflammation of the mucosa, and an imbalance of intestinal microbes. However, current treatments largely focus on a single factor, yielding unsatisfactory clinical outcomes. Herein, we report a biocompatible and IBD-targeted metabolic nanoregulator (TMNR) that synergistically regulates cellular and bacterial metabolism. The TMNR comprises a melanin-gallium complex (MNR) encapsulated within a thermosensitive and colitis-targeting hydrogel, all composed of natural and FDA-approved components. The TMNR confers superior broad-spectrum antioxidant properties, effectively scavenging reactive oxygen species (ROS) and blocking inflammatory signaling pathways. The presence of Ga3+ in TMNR selectively disrupts iron metabolism in pathogenic microorganisms due to its structural resemblance to the iron atom. Additionally, incorporating a thermosensitive injectable hydrogel enables targeted delivery of TMNR to inflammatory regions, prolonging their retention time and providing a physical barrier function for optimizing IBD treatment efficacy. Collectively, TMNR effectively modulates the redox balance of inflamed colonic epithelial tissue and disrupts iron metabolism in pathogenic microorganisms, thereby eliminating inflammation and restoring intestinal homeostasis against IBD. Hence, this work presents a comprehensive approach for precise spatiotemporal regulation of the intestinal microenvironmental metabolism for IBD treatment.
Collapse
Affiliation(s)
- Ting Xu
- The Radiology Department of Shanxi Provincial People's Hospital, The Fifth Hospital of Shanxi Medical University, Taiyuan, 030012, China
| | - Xiaogang Ning
- School of Chinese Medicine and Food Engineering, Shanxi University of Chinese Medicine, Jinzhong, 030619, China
| | - Jiayan Wu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637457, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 637457, Singapore
| | - Qian Wang
- The Radiology Department of Shanxi Provincial People's Hospital, The Fifth Hospital of Shanxi Medical University, Taiyuan, 030012, China
| | - Zhifei Wang
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Zhiqing Chen
- The Radiology Department of Shanxi Provincial People's Hospital, The Fifth Hospital of Shanxi Medical University, Taiyuan, 030012, China
| | - Xiaoxian Tang
- The Radiology Department of Shanxi Provincial People's Hospital, The Fifth Hospital of Shanxi Medical University, Taiyuan, 030012, China
| | - Peirong Bai
- Third Hospital of Shanxi Medical University, Shanxi Bethune Hospital, Shanxi Academy of Medical Sciences, Tongji Shanxi Hospital, Taiyuan, 030032, China
| | - Kanyi Pu
- School of Chemistry, Chemical Engineering and Biotechnology, Nanyang Technological University, Singapore, 637457, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, 637457, Singapore
| | - Liping Li
- The Fifth Hospital of Shanxi Medical University, Taiyuan, 030012, China
- School of Basic Medical Sciences, Shanxi Medical University, Taiyuan, 030001, China
| | - Ruiping Zhang
- The Radiology Department of Shanxi Provincial People's Hospital, The Fifth Hospital of Shanxi Medical University, Taiyuan, 030012, China
| |
Collapse
|
33
|
Xiang Y, Chen Q, Nan Y, Liu M, Xiao Z, Yang Y, Zhang J, Ying X, Long X, Wang S, Sun J, Huang Q, Ai K. Nitric Oxide‐Based Nanomedicines for Conquering TME Fortress: Say “NO” to Insufficient Tumor Treatment. ADVANCED FUNCTIONAL MATERIALS 2024; 34. [DOI: 10.1002/adfm.202312092] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2023] [Indexed: 01/02/2025]
Abstract
AbstractAlmost all cancer treatments are significantly limited by the strong tumor microenvironment (TME) fortress formed by abnormal vasculature, dense extracellular matrix (ECM), multidrug resistance (MDR) system, and immune “cold” environment. In the huge efforts of dismantling the TME fortress, nitric oxide (NO)‐based nanomedicines are increasingly occupying a central position and have already been identified as super “strong polygonal warriors” to dismantle TME fortress for efficient cancer treatment, benefiting from NO's unique physicochemical properties and extremely fascinating biological effects. However, there is a paucity of systematic review to elaborate on the progress and fundamental mechanism of NO‐based nanomedicines in oncology from this aspect. Herein, the key characteristics of TME fortress and the potential of NO in reprogramming TME are delineated and highlighted. The evolution of NO donors and the advantages of NO‐based nanomedicines are discussed subsequently. Moreover, the latest progress of NO‐based nanomedicines for solid tumors is comprehensively reviewed, including normalizing tumor vasculature, overcoming ECM barrier, reversing MDR, and reactivating the immunosuppression TME. Lastly, the prospects, limitations, and future directions on NO‐based nanomedicines for TME manipulation are discussed to provide new insights into the construction of more applicable anticancer nanomedicines.
Collapse
Affiliation(s)
- Yuting Xiang
- Department of Pharmacy Xiangya Hospital Central South University Changsha Hunan 410008 P. R. China
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
| | - Qiaohui Chen
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research Xiangya School of Pharmaceutical Sciences Central South University Changsha 410078 P. R. China
| | - Yayun Nan
- Geriatric Medical Center People's Hospital of Ningxia Hui Autonomous Region Yinchuan Ningxia 750002 P. R. China
| | - Min Liu
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research Xiangya School of Pharmaceutical Sciences Central South University Changsha 410078 P. R. China
| | - Zuoxiu Xiao
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research Xiangya School of Pharmaceutical Sciences Central South University Changsha 410078 P. R. China
| | - Yuqi Yang
- Department of Pharmacy Xiangya Hospital Central South University Changsha Hunan 410008 P. R. China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Central South University Changsha Hunan 410008 P. R. China
| | - Jinping Zhang
- Department of Pharmacy Xiangya Hospital Central South University Changsha Hunan 410008 P. R. China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Central South University Changsha Hunan 410008 P. R. China
| | - Xiaohong Ying
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research Xiangya School of Pharmaceutical Sciences Central South University Changsha 410078 P. R. China
| | - Xingyu Long
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research Xiangya School of Pharmaceutical Sciences Central South University Changsha 410078 P. R. China
| | - Shuya Wang
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research Xiangya School of Pharmaceutical Sciences Central South University Changsha 410078 P. R. China
| | - Jian Sun
- College of Pharmacy Xinjiang Medical University Urumqi 830017 P. R. China
| | - Qiong Huang
- Department of Pharmacy Xiangya Hospital Central South University Changsha Hunan 410008 P. R. China
- National Clinical Research Center for Geriatric Disorders Xiangya Hospital Central South University Changsha Hunan 410008 P. R. China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences Central South University Changsha Hunan 410078 P. R. China
- Hunan Provincial Key Laboratory of Cardiovascular Research Xiangya School of Pharmaceutical Sciences Central South University Changsha 410078 P. R. China
- Key Laboratory of Aging‐related Bone and Joint Diseases Prevention and Treatment Ministry of Education Xiangya Hospital Central South University Changsha 410078 P. R. China
| |
Collapse
|
34
|
Jiang K, Cao X, Wu H, Xu Y, Liu L, Qian H, Miao Z, Wang H, Ma Y. 2D Nanozymes Modulate Gut Microbiota and T-Cell Differentiation for Inflammatory Bowel Disease Management. Adv Healthc Mater 2024; 13:e2302576. [PMID: 37897434 DOI: 10.1002/adhm.202302576] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2023] [Revised: 10/26/2023] [Indexed: 10/30/2023]
Abstract
Intestinal commensal microbiota dysbiosis and immune dysfunction are significant exacerbating factors in inflammatory bowel disease (IBD). To address these problems, Pluronic F-127-coated tungsten diselenide (WSe2 @F127) nanozymes are developed by simple liquid-phase exfoliation. The abundant valence transitions of elemental selenium (Se2- /Se4+ ) and tungsten (W4+ /W6+ ) enable the obtained WSe2 @F127 nanozymes to eliminate reactive oxygen/nitrogen species. In addition, the released tungsten ions are capable of inhibiting the proliferation of Escherichia coli. In a model of dextran sodium sulfate-induced colitis, WSe2 @F127 nanozymes modulate the gut microbiota by increasing the abundance of bacteria S24-7 and significantly reducing the abundance of Enterobacteriaceae. Moreover, WSe2 @F127 nanozymes inhibit T-cell differentiation and improve intestinal immune barrier function in a model of Crohn's disease. The WSe2 @F127 nanozymes effectively alleviate IBD by reducing oxidative stress damage, modulating intestinal microbial populations, and remodeling the immune barrier.
Collapse
Affiliation(s)
- Kai Jiang
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Xiangjing Cao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Haitao Wu
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yifeng Xu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Lulu Liu
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Haisheng Qian
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230022, China
| | - Zhaohua Miao
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, 230009, China
| | - Hua Wang
- Department of Oncology, The First Affiliated Hospital of Anhui Medical University, Hefei, 230022, China
| | - Yan Ma
- School of Biomedical Engineering, Anhui Medical University, Hefei, 230022, China
| |
Collapse
|
35
|
Wang M, Huang Q, Liu M, Zhao T, Song X, Chen Q, Yang Y, Nan Y, Liu Z, Zhang Y, Wu W, Ai K. Precisely Inhibiting Excessive Intestinal Epithelial Cell Apoptosis to Efficiently Treat Inflammatory Bowel Disease with Oral Pifithrin-α Embedded Nanomedicine (OPEN). ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2023; 35:e2309370. [PMID: 37747308 DOI: 10.1002/adma.202309370] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 09/22/2023] [Indexed: 09/26/2023]
Abstract
The increased incidence of inflammatory bowel disease (IBD) has seriously affected the life quality of patients. IBD develops due to excessive intestinal epithelial cell (IEC) apoptosis, disrupting the gut barrier, colonizing harmful bacteria, and initiating persistent inflammation. The current therapeutic approaches that reduce inflammation are limited. Although IBD can be treated significantly by directly preventing IEC apoptosis, achieving this therapeutic approach remains challenging. Accordingly, the authors are the first to develop an oral pifithrin-α (PFTα, a highly specific p53 inhibitor) embedded nanomedicine (OPEN) to effectively treat IBD by inhibiting excessive IEC apoptosis. As a major hub for various stressors, p53 is a central determinant of cell fate, and its inhibition can effectively reduce excessive IEC apoptosis. The tailored OPEN can precisely inhibit the off-target and inactivation resulting from PFTα entry into the bloodstream. Subsequently, it persistently targets IBD lesions with high specificity to inhibit the pathological events caused by excessive IEC apoptosis. Eventually, OPEN exerts a significant curative effect compared with the clinical first-line drugs 5-aminosalicylic acid (5-ASA) and dexamethasone (DEX). Consequently, the OPEN therapeutic strategy provides new insights into comprehensive IBD therapy.
Collapse
Affiliation(s)
- Mingyuan Wang
- Department of Geriatric Surgery, Xiangya Hospital, Central South University, Changsha, 410078, China
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410078, China
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410078, China
| | - Qiong Huang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410078, China
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410078, China
| | - Min Liu
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
| | - Tianjiao Zhao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
| | - Xiangping Song
- Department of Geriatric Surgery, Xiangya Hospital, Central South University, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410078, China
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410078, China
| | - Qiaohui Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
| | - Yongqi Yang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
| | - Yayun Nan
- Geriatric Medical Center, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, Ningxia, 750002, China
| | - Zerun Liu
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410078, China
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410078, China
| | - Yuntao Zhang
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410078, China
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410078, China
| | - Wei Wu
- Department of Geriatric Surgery, Xiangya Hospital, Central South University, Changsha, 410078, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410078, China
- Department of General Surgery, Xiangya Hospital, Central South University, Changsha, 410078, China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410008, China
- Key Laboratory of Aging-related Bone and Joint Diseases Prevention and Treatment, Ministry of Education, Xiangya Hospital, Central South University, Changsha, 410008, China
| |
Collapse
|
36
|
Yang Y, Nan Y, Chen Q, Xiao Z, Zhang Y, Zhang H, Huang Q, Ai K. Antioxidative 0-dimensional nanodrugs overcome obstacles in AKI antioxidant therapy. J Mater Chem B 2023; 11:8081-8095. [PMID: 37540219 DOI: 10.1039/d3tb00970j] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/05/2023]
Abstract
Acute kidney injury (AKI) is a commonly encountered syndrome associated with various aetiologies and pathophysiological processes leading to enormous health risks and economic losses. In the absence of specific drugs to treat AKI, hemodialysis remains the primary clinical treatment for AKI patients. The revelation of the pathology opens new horizons for antioxidant therapy in the treatment of AKI. However, small molecule antioxidant drugs and common nanozymes have failed to challenge AKI due to their unsatisfactory drug properties and renal physiological barriers. 0-Dimensional (0D) antioxidant nanodrugs stand out at this time thanks to their small size and high performance. Recently, a number of research studies have been carried out around 0D nanodrugs for alleviating AKI, and their multi-antioxidant enzyme mimetic activities, smooth glomerular filtration barrier permeability and excellent biocompatibility have been investigated. Here, we comprehensively summarize recent advances in 0D nanodrugs for AKI antioxidant therapy. We classify these representative studies into three categories according to the characteristics of 0D nanomaterials, namely ultra-small metal nanodots, inorganic non-metallic quantum dots and polymer nanodots. We focus on the antioxidant mechanisms and their distribution in vivo in each inspiring work, and the purpose and ingenuity of each design are rigorously captured and described. Finally, we provide our reflections and prospects for 0D antioxidant nanodrugs in AKI treatment. This mini review provides unique insights and valuable clues in the design of 0D nanodrugs and other kidney absorbable drugs.
Collapse
Affiliation(s)
- Yuqi Yang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Yayun Nan
- Geriatric Medical Center, People's Hospital of Ningxia Hui Autonomous Region, Yinchuan, 750002, China
| | - Qiaohui Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Zuoxiu Xiao
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Yuntao Zhang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Huanan Zhang
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| | - Qiong Huang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, 410008, China.
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, 410008, China
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China.
- Hunan Provincial Key Laboratory of Cardiovascular Research, Xiangya School of Pharmaceutical Sciences, Central South University, Changsha, 410078, China
| |
Collapse
|
37
|
Elzayat EM, Sherif AY, Nasr FA, Attwa MW, Alshora DH, Ahmad SF, Alqahtani AS. Enhanced Codelivery of Gefitinib and Azacitidine for Treatment of Metastatic-Resistant Lung Cancer Using Biodegradable Lipid Nanoparticles. MATERIALS (BASEL, SWITZERLAND) 2023; 16:5364. [PMID: 37570067 PMCID: PMC10419431 DOI: 10.3390/ma16155364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/17/2023] [Revised: 07/20/2023] [Accepted: 07/27/2023] [Indexed: 08/13/2023]
Abstract
Lung cancer is a formidable challenge in clinical practice owing to its metastatic nature and resistance to conventional treatments. The codelivery of anticancer agents offers a potential solution to overcome resistance and minimize systemic toxicity. The encapsulation of these agents within nanostructured lipid carriers (NLCs) provides a promising strategy to enhance lymphatic delivery and reduce the risk of relapse. This study aimed to develop an NLC formulation loaded with Gefitinib and Azacitidine (GEF-AZT-NLC) for the treatment of metastatic-resistant lung cancer. The physicochemical properties of the formulations were characterized, and in vitro drug release was evaluated using the dialysis bag method. The cytotoxic activity of the GEF-AZT-NLC formulations was assessed on a lung cancer cell line, and hemocompatibility was evaluated using suspended red blood cells. The prepared formulations exhibited nanoscale size (235-272 nm) and negative zeta potential values (-15 to -31 mV). In vitro study revealed that the GEF-AZT-NLC formulation retained more than 20% and 60% of GEF and AZT, respectively, at the end of the experiment. Hemocompatibility study demonstrated the safety of the formulation for therapeutic use, while cytotoxicity studies suggested that the encapsulation of both anticancer agents within NLCs could be advantageous in treating resistant cancer cells. In conclusion, the GEF-AZT-NLC formulation developed in this study holds promise as a potential therapeutic tool for treating metastatic-resistant lung cancer.
Collapse
Affiliation(s)
- Ehab M Elzayat
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Abdelrahman Y Sherif
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Fahd A Nasr
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Mohamed W Attwa
- Department of Pharmaceutical Chemistry, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Doaa H Alshora
- Department of Pharmaceutics, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Sheikh F Ahmad
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| | - Ali S Alqahtani
- Department of Pharmacognosy, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh 11451, Saudi Arabia
| |
Collapse
|