1
|
Kwon HY, Jung Y, Jeon H, Han HS. Investigation into recent advanced strategies of reactive oxygen species-mediated therapy based on Prussian blue: Conceptualization and prospect. Bioact Mater 2025; 48:71-99. [PMID: 40034810 PMCID: PMC11874232 DOI: 10.1016/j.bioactmat.2025.01.023] [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: 01/16/2025] [Accepted: 01/20/2025] [Indexed: 03/05/2025] Open
Abstract
Prussian blue (PB) has garnered considerable scholarly interest in the field of biomedical research owing to its notably high biocompatibility, formidable multi-enzyme mimetic capabilities, and established clinical safety profile. These properties in combination with its reactive oxygen species (ROS) scavenging activity have facilitated significant progress in disease diagnosis and therapy for various ROS-mediated pathologies, where overproduced ROS exacerbates disease symptoms. Additionally, the underlying ROS-associated mechanisms are disease-specific. Hence, we systematically examined the role of ROS and its basic underlying mechanisms in representative disease categories and comprehensively reviewed the effect of PB-based materials in effectively alleviating pathological states. Furthermore, we present a thorough synthesis of disease-specific design methodologies and prospective directions for PB as a potent ROS-scavenging biotherapeutic material with emphasis on its applications in neurological, cardiovascular, inflammatory, and other pathological states. Through this review, we aim to accelerate the progress of research on disease treatment using PB-based integrated therapeutic system.
Collapse
Affiliation(s)
- Hee-Young Kwon
- Biomaterials Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Yuna Jung
- Biomaterials Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
| | - Hojeong Jeon
- Biomaterials Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- KU-KIST Graduate School of Converging Science and Technology, Korea University, Seoul, 02841, Republic of Korea
| | - Hyung-Seop Han
- Biomaterials Research Center, Korea Institute of Science and Technology, Seoul, 02792, Republic of Korea
- Research and Development Center, Elecell Corporation, Seoul, 02455, Republic of Korea
| |
Collapse
|
2
|
Su M, Jin R, Zhu J, Pei J, Wang Y, Chai X, Jiang M. Composition and antioxidant activity of flavonoids from two different species of Amomi Fructus extracted using natural deep eutectic solvents. Food Chem 2025; 472:142984. [PMID: 39848052 DOI: 10.1016/j.foodchem.2025.142984] [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: 11/11/2024] [Revised: 01/09/2025] [Accepted: 01/18/2025] [Indexed: 01/25/2025]
Abstract
Amomi Fructus, a mature fruit from a ginger family plant, has various species, resulting in inconsistent sourcing and quality. Most studies distinguish species by volatile compounds, yet research shows it also contains flavonoids with notable pharmacological effects. Solely focusing on volatile compounds could lead to considerable resource waste. This study aims to establish flavonoid markers in Amomi Fructus to distinguish its species, assess quality, and promote efficient resource use. Utilizing natural deep eutectic solvents (NADES) and response surface methodology (RSM), an optimal extraction system (choline chloride-ethylene glycol) yielded 41.38 mg RE/g total flavonoids. LC-MS analysis of 18 Amomi Fructus batches identified 26 flavonoids, quantified 19, and highlighted three key markers-epicatechin, procyanidin B2, and procyanidin B4-that effectively differentiate Amomum villosum Lour. (AMV) from Amomum villosum Lour. var. xanthioides T.L. Wu et Senjen (AMVX). Finally, flow cytometry confirmed these markers' antioxidant activity, effectively reducing H₂O₂-induced oxidative damage in GES-1 cells.
Collapse
Affiliation(s)
- Mei Su
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Ruyi Jin
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jian Zhu
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Jierong Pei
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China
| | - Yuefei Wang
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Xin Chai
- Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China
| | - Miaomiao Jiang
- National Key Laboratory of Chinese Medicine Modernization, Tianjin University of Traditional Chinese Medicine, Tianjin 301617, China; Haihe Laboratory of Modern Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, 301617, China.
| |
Collapse
|
3
|
Shen J, Jia G, Wu Q, Yang H, Jiang Y, Wu X, Chai Y, Zhang C, Xu J. Black phosphorus nanosheets fortified with catalase to enhance Schwann cell responses for neural repair. J Control Release 2025; 380:579-598. [PMID: 39938721 DOI: 10.1016/j.jconrel.2025.02.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2024] [Revised: 02/07/2025] [Accepted: 02/08/2025] [Indexed: 02/14/2025]
Abstract
Peripheral nerve injuries (PNI) present a significant clinical challenge due to the complex cellular and molecular activities that hinder functional recovery. Schwann cells (SCs), the principal glial cells in the peripheral nervous system, play a vital role in neural repair by transitioning into a repairing phenotype capable of supporting axonal regrowth. However, these regenerative properties fade over time, leading to poor clinical outcomes. To address this issue, we engineered a black phosphorus nanosheet (BPNS) functionalized with catalase (BPNS@CAT) to modulate SC activity and enhance nerve regeneration. In vitro experiments demonstrated that BPNS@CAT reduced ROS levels, regulated the angiogenic and immunomodulatory functions of SCs. Mechanistically, we identified that BPNS@CAT activated the JAK/STAT pathway, which is crucial for SC-mediated repair processes. To validate its therapeutic potential, a BPNS@CAT-GelMA/PCL hydrogel scaffold was fabricated and applied in a rat sciatic nerve-crush model. The scaffold enhanced axonal regeneration, restored nerve function, and improved sensory, motor, and emotional behaviors. Our study broadens the range of BPNS applications in SC-based nerve repair and pave the way for future applications of BPNS in translational medicine.
Collapse
Affiliation(s)
- Junjie Shen
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China
| | - Guoping Jia
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Qinghe Wu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Huizhen Yang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Yifei Jiang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Xubo Wu
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, PR China
| | - Yimin Chai
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China
| | - Chunfu Zhang
- School of Biomedical Engineering, Shanghai Jiao Tong University, Shanghai 200030, PR China.
| | - Jia Xu
- Department of Orthopedic Surgery, Shanghai Sixth People's Hospital Affiliated to Shanghai Jiao Tong University School of Medicine, Shanghai 200233, PR China.
| |
Collapse
|
4
|
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. [PMID: 40173890 DOI: 10.1021/jacs.5c00100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [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
|
5
|
Qiu M, Man C, Zhao Q, Yang X, Zhang Y, Zhang W, Zhang X, Irudayaraj J, Jiang Y. Nanozymes meet hydrogels: Fabrication, progressive applications, and perspectives. Adv Colloid Interface Sci 2025; 338:103404. [PMID: 39884113 DOI: 10.1016/j.cis.2025.103404] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Revised: 11/19/2024] [Accepted: 01/16/2025] [Indexed: 02/01/2025]
Abstract
Nanozyme, a class of emerging enzyme mimics, is the nanomaterials with enzyme-mimicking activity, which has obtained significant and widespread applications in various fields. However, they still face many challenges in practical applications (e.g., instability and low biocompatibility in the physiological environments), which affect their widespread applications to a certain extent. Hydrogels with superior performances (e.g., the controllable degradability, good biocompatibility, hydrophilic properties, and adjustable physical properties) may provide a promising strategy to make up the existing deficiencies of nanozymes in practical applications. Thus, the sapiential combination of nanozymes with hydrogels endows nanozyme hydrogels with both characteristics of nanozymes and properties of hydrogels, making nanozyme hydrogels become novel multifunctional materials. In this review, we comprehensively summarizes the preparation, properties, and progressive applications of nanozyme hydrogels. First of all, the main design and preparation strategies of nanozyme hydrogels are considerately summarized. Then, the properties of different nanozyme hydrogels are introduced. In addition, sophisticated applications of nanozyme hydrogels in the fields of biosensing, biomedicine applications, and environmental are comprehensively summarized. Most importantly, future obstacles and chances in this emerging field are profoundly proposed. This review will provide a new horizon for the development and future applications of novel nanozyme hydrogels.
Collapse
Affiliation(s)
- Manyan Qiu
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Chaoxin Man
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China.
| | - Qianyu Zhao
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Xinyan Yang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Yu Zhang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Wei Zhang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China
| | - Xianlong Zhang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China.
| | - Joseph Irudayaraj
- Department of Bioengineering, Grainger College of Engineering, Carle-Illinois College of Medicine, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA.
| | - Yujun Jiang
- Key Laboratory of Dairy Science, Ministry of Education, College of Food Science, Northeast Agricultural University, Harbin 150030, China; Food Laboratory of Zhongyuan, Luohe 462300, Henan, China; Key Laboratory of Infant Formula Food, State Administration for Market Regulation, Harbin 150030, China.
| |
Collapse
|
6
|
Mosiej W, Długosz E, Kruk M, Zielińska D. Immunomodulatory Properties of Live and Thermally-Inactivated Food-Origin Lactic Acid Bacteria-In Vitro Studies. Mol Nutr Food Res 2025:e70047. [PMID: 40166824 DOI: 10.1002/mnfr.70047] [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: 10/21/2024] [Revised: 03/03/2025] [Accepted: 03/10/2025] [Indexed: 04/02/2025]
Abstract
The study investigates the strain-specific immunomodulatory properties of live and thermally-inactivated (TI) lactic acid bacteria (LAB) derived from traditional Polish fermented foods, focusing on their potential as probiotics and postbiotics. LAB strains, known for their role in food fermentation, were assessed for their ability to influence cytokine production in THP-1 macrophages, maintain intestinal epithelial barrier integrity in Caco-2 monolayers, exhibit antioxidant activity, and produce specific organic acids and sugars. The research demonstrated that live LAB strains significantly upregulated the anti-inflammatory cytokine IL-10, particularly under inflammatory conditions, while TI strains exhibited notable antioxidant and anti-inflammatory properties. TI strains showed a greater ability to protect epithelial barrier function and reduce pro-inflammatory cytokine secretion than live strains, suggesting a promising role for postbiotics. The findings underscore the potential of LAB from fermented foods, demonstrating that postbiotic derivatives can differently influence inflammation compared to live bacteria, highlighting their potential as immune-enhancing agents, capable of modulating immune responses and offering therapeutic benefits against inflammation-related disorders. However, the limitations of in vitro models highlight the need for further in vivo and clinical studies to validate these effects and fully uncover the health benefits of these LAB strains for humans.
Collapse
Affiliation(s)
- Wioletta Mosiej
- Department of Food Gastronomy and Food Hygiene, Institute of Human Nutrition Sciences, Warsaw University of Life Science - SGGW, Warsaw, Poland
| | - Ewa Długosz
- Department of Preclinical Sciences, Institute of Veterinary Medicine, Warsaw University of Life Science - SGGW, Warsaw, Poland
| | - Marcin Kruk
- Department of Food Gastronomy and Food Hygiene, Institute of Human Nutrition Sciences, Warsaw University of Life Science - SGGW, Warsaw, Poland
| | - Dorota Zielińska
- Department of Food Gastronomy and Food Hygiene, Institute of Human Nutrition Sciences, Warsaw University of Life Science - SGGW, Warsaw, Poland
| |
Collapse
|
7
|
El-Sayed SF, Mahmoud SM, Samy W, Wahid RM, Talaat A, Seada SG. Vitamin D3 mitigates aspirin-induced gastric injury by modulating gastrokines, E-cadherin, and inhibiting NLRP3 and NF-κB/MMP-9 signaling pathway. Tissue Cell 2025; 93:102724. [PMID: 39823708 DOI: 10.1016/j.tice.2025.102724] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 12/23/2024] [Accepted: 01/03/2025] [Indexed: 01/20/2025]
Abstract
BACKGROUND The prevalence of gastric ulcers has grown significantly in the modern era affecting 10 % of global population. Aspirin downregulates gastrokines 1(GKN1) expression in gastric mucosa and GKN1 down-regulation results in gastric cancer. Vitamin D3 (Vit.D3) has anti-inflammatory and antioxidant effects. AIM Study the gastroprotective impact of Vit.D3 following aspirin-induced gastric injury in relation to gastrokines and investigate the possible underlying mechanisms. MATERIALS AND METHODS 24 rats were divided into 4 groups: control, Vit.D3 supplemented normal, aspirin-induced gastric injury, and Vit.D3 supplemented gastric injury groups. Some oxidative stress markers with gene expression of GKN1&2, mucin 5AC (Muc5ac) and NLR family pyrin domain containing 3 (NLRP3) in the gastric tissue were done. Histopathological and immunohistochemical study of E-Cadherin, nuclear factor kappa beta (NFκB), and metalloproteinase-9 (MMP-9) in the stomach mucosa were identified. RESULTS Vit.D3 supplementation significantly upregulated E-Cadherin, GSH, GKN1 and Muc5ac in the gastric tissue. Also, it improved the morphology, histology of gastric tissue, by alleviating oxidative stress and NFκB, MMP-9 and down regulation of inflammasome (NLRP3). CONCLUSION Vitamin D3 has a potential protective effect against aspirin -induced gastric injury via upregulating gastrokine1 and E-cadherin and down regulation of NFKB/MMP-9 signaling pathway and NLRP3 inflammasome.
Collapse
Affiliation(s)
- Sherein F El-Sayed
- Department of Medical Physiology, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Samar Mortada Mahmoud
- Department of Human Anatomy and Embryology, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Walaa Samy
- Department of Medical Biochemistry, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Reham M Wahid
- Department of Medical Physiology, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Aliaa Talaat
- Department of Medical Biochemistry, Faculty of Medicine, Zagazig University, Zagazig, Egypt.
| | - Sara G Seada
- Department of Medical Physiology, Faculty of Medicine, Zagazig University, Zagazig, Egypt
| |
Collapse
|
8
|
Wei J, Tao G, Liu J, Framroze B, Sylvester KG. Protective effect of soluble protein hydrolysate against H 2O 2‑induced intestinal injury: An interventional study. Mol Med Rep 2025; 31:85. [PMID: 39886967 PMCID: PMC11811813 DOI: 10.3892/mmr.2025.13450] [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: 09/20/2024] [Accepted: 12/09/2024] [Indexed: 02/01/2025] Open
Abstract
The present study aimed to investigate whether soluble protein hydrolysate (SPH) protects against intestinal oxidative stress injury. An in vitro lactate dehydrogenase assay was used to assess the cytotoxicity and protective effect of SPH. For in vivo assessment, friend virus B NIH Jackson mouse pups aged 21 days were administered with 5% w/v soluble protein hydrolysate (SPH) through drinking water for 14 days and then luminally injected with 0.3% or 0.6% H2O2. Thereafter, the fecal samples of mice were collected, and the mice were sacrificed. Intestinal epithelial injury was assessed, and the expressions of 84 oxidative stress‑related genes in intestinal tissues was determined. SPH prophylactically protected against H2O2‑induced oxidative stress injury in human intestinal epithelial cells. An animal model of oxidative stress‑induced intestinal injury was established using 0.3 and 0.6% H2O2. SPH treatment reduced oxidative stress (0.3% H2O2)‑induced gut injury in mice. As no accelerated body growth was observed in SPH‑treated mice, it was hypothesized that the underlying protective mechanism of SPH is not related to nutrient oversupply. Treatment with SPH upregulated five oxidative protective genes that were not consistent between the sexes. Some antioxidative genes, including ferritin heavy polypeptide‑1 (Fth1), heme oxygenase‑1 (Hmox1), NAD(P)H dehydrogenase quinone 1 (Nqo1) and superoxide dismutase 1 (Sod1), were commonly upregulated in both male and female mice. Overall, an antioxidative protective effect was observed following SPH treatment, which may be attributed to the upregulation of genes that protect against oxidative damage. The findings of the present study highlight the promising potential of SPH as a functional food for alleviating intestinal oxidative stress injury.
Collapse
Affiliation(s)
- Jingjing Wei
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94304, USA
- Department of Pediatrics, Shanxi Medical University, Taiyuan, Shanxi 030001, P.R. China
| | - Guozhong Tao
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94304, USA
| | - Junlin Liu
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94304, USA
- Department of General Surgery, The People's Hospital of Liuyang City, Liuyang, Hunan 410300, P.R. China
| | - Bomi Framroze
- Department of R&D, Hofseth BioCare ASA, Aalesund 6003, Norway
| | - Karl G. Sylvester
- Department of Surgery, Stanford University School of Medicine, Stanford, CA 94304, USA
| |
Collapse
|
9
|
Gao C, Yang Z, Song R, Sheng H, Zhu L. Nanotechnology-based drug delivery system for targeted therapy of ulcerative colitis from traditional Chinese medicine: A review. Int J Pharm 2025; 673:125375. [PMID: 39965734 DOI: 10.1016/j.ijpharm.2025.125375] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 01/27/2025] [Accepted: 02/15/2025] [Indexed: 02/20/2025]
Abstract
Ulcerative colitis (UC) is a chronic autoimmune disease and seriously affects the normal life of patients. Conventional therapeutic drugs are difficult to meet clinical needs. Traditional Chinese medicine (TCM) ingredients could effectively alleviate the symptoms of UC by anti-inflammatory, anti-oxidative, regulating the gut microbiota, and repairing the colonic epithelial barrier, but their low solubility and bioavailability severely limit their clinical application. Nano-drug delivery systems (NDDS) combined with TCM ingredients is a promising option for treating UC, and they could significantly enhance the stability, solubility, and bioavailability of TCM ingredients. The review describes the anti-UC mechanisms of TCM ingredients, systematically summarizes various kinds of NDDS for TCM ingredients according to different routes of administration, and highlights the advantages of NDDS for TCM ingredients in the treatmentof UC. In addition, we discuss the limitations of existing NDDS for TCM ingredients and the development direction in the future. This review will provide a basis for the future development of anti-UC NDDS for TCM ingredients.
Collapse
Affiliation(s)
- Chengcheng Gao
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Zerun Yang
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Ruirui Song
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China
| | - Huagang Sheng
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| | - Liqiao Zhu
- College of Pharmacy, Shandong University of Traditional Chinese Medicine, Jinan 250355, China.
| |
Collapse
|
10
|
Shafi FAA, Imeer ATA, Nassrullah HAA, Naeemah AM. The impact of extracellular glucose concentrations on antioxidant capacity, viability, and microRNA expression in TM4 Sertoli cells. Reprod Biol 2025; 25:101015. [PMID: 40163939 DOI: 10.1016/j.repbio.2025.101015] [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: 11/30/2024] [Revised: 03/09/2025] [Accepted: 03/24/2025] [Indexed: 04/02/2025]
Abstract
This study investigates the impact of extracellular glucose concentrations on antioxidant capacity, viability, and microRNA (miR) expression in TM4 Sertoli cell lines. TM4 cells were cultured in high-glucose (115 µm) and low-glucose (<505 µm) conditions to simulate hyperglycemia and glucose starvation, respectively. The study measured total antioxidant capacity (TAC), malondialdehyde (MDA), total oxidant status (TOS), glutathione (GSH), glutathione disulfide (GSSG), NADP/NADPH, glutathione peroxidase (GPX), and glutathione reductase (GR) levels. MiR-17, miR-34, miR-106a, and miR-200a expression levels were assessed. Cell viability and apoptosis were evaluated using MTT assay and acridine-orange staining. Results indicated that high glucose reduced miR-17 expression while low glucose increased it. Both glucose conditions elevated miR-34, miR-106a, and miR-200a expressions. TAC levels decreased, while TOS and MDA levels increased significantly under both conditions. High glucose had no significant effect on GPX and GR levels, whereas low glucose decreased them. Both conditions led to reduced GSH levels, increased GSSG levels, and altered NADP/NADPH ratio. Increased apoptosis and decreased cell viability were observed under both glucose conditions. These findings suggest that extracellular glucose levels significantly dysregulate miRNA expression, antioxidant capacities, and redox buffer systems in TM4 cells. High glucose conditions suppress miR-17 expression, increase miR-34 and miR-106a levels, and induce reductive buffer imbalance. Conversely, low glucose conditions trigger compensatory mechanisms via increased miR-17 expression to enhance antioxidant status while reducing GPX and GR levels. These results provide insights into the molecular responses of Sertoli cells under varying glucose environments, highlighting potential therapeutic pathways for conditions like diabetes and metabolic dysfunctions.
Collapse
Affiliation(s)
- Farha A Ali Shafi
- Department of Biology College of Science Mustansiriyah University, Baghdad, Iraq.
| | | | | | - Ali Mutashar Naeemah
- Department of Histology and Embryology, Faculty of Veterinary Medicine, Urmia University, Urmia, Iran
| |
Collapse
|
11
|
Ülger Y, Delik A. Gene expression profile in ulcerative colitis patients: FOXO4, ALDOB, SLC26A3, SOD2 genes as potential biomarkers. Genes Genomics 2025:10.1007/s13258-025-01625-y. [PMID: 40153227 DOI: 10.1007/s13258-025-01625-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 02/14/2025] [Indexed: 03/30/2025]
Abstract
BACKGROUND Ulcerative colitis (UC) is a complex, chronic inflammatory disease that primarily impacts the colon mucosa. One of the key pathological contributors to the development and progression of inflammatory bowel disease (IBD) is oxidative stress, which results in reactive oxygen species (ROS)-induced mucosal damage. This stress leads to dysfunction of the intestinal barrier. OBJECTIVES The purpose of this study is to examine the expression levels of genes involved in various inflammatory pathways, including autophagy, unfolded protein response (UPR), ubiquitination, metabolic pathways, and immune responses in the colon mucosa of patients with UC. MATERIAL AND METHODS Patients diagnosed with UC at Çukurova University, Balcalı Hospital, Gastroenterology Department between December 2023 and January 2024 were included in this prospective study. A total of 40 participants were included in the study: 27 ulcerative colitis patients and 13 controls. To isolate high-quality RNA, colon biopsy material obtained during colonoscopy was immediately placed in stabilization solution and stored at - 80 degrees Celsius. The relative quantification of target gene mRNA was determined using a Light Cycler. Subsequently, differences in gene expression between patients and the control group were evaluated using the Mann-Whitney U and Kruskal-Wallis tests. RESULTS In our study, FOXO4 gene expression increased in UC patients during both active and remission phases compared to the control group. The high expression of this gene is associated with its role in inflammation and cell death processes. Additionally, the high expression of ALDOB and SLC26A genes is linked to increased inflammation and energy demand. Lastly, the elevated expression of the SOD2 gene can be considered a response to oxidative stress-related inflammatory processes in the disease. CONCLUSION These findings propose that these genes could serve as potential biomarkers for genomic identification and understanding the pathogenesis of UC.
Collapse
Affiliation(s)
- Yakup Ülger
- Division of Gastroenterology, Faculty of Medicine, Cukurova University, 01330, Adana, Turkey.
- Balcalı Hospital, Sarıcam, Adana, Turkey.
| | - Anıl Delik
- Division of Gastroenterology, Faculty of Medicine, Cukurova University, 01330, Adana, Turkey
- Balcalı Hospital, Sarıcam, Adana, Turkey
- Division of Biology, Faculty of Science and Literature, Cukurova University, 01330, Adana, Turkey
| |
Collapse
|
12
|
Bakhtiyari-Ramezani M, Nasiri M, Baniasadi M. Helium and argon cold plasma effects on the 4T1 cancer cells and a triple negative mouse model of breast cancer. Sci Rep 2025; 15:10569. [PMID: 40148523 PMCID: PMC11950318 DOI: 10.1038/s41598-025-95065-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Accepted: 03/18/2025] [Indexed: 03/29/2025] Open
Abstract
Nowadays, cold atmospheric plasma (CAP) technology has developed as an innovative tool for cancer therapy. Although many studies have reported the antitumor effects of plasma in vivo and in vitro, there are many challenges, including standardization of plasma devices and treatment time for different tumors. For the first time, we aimed to evaluate and compare optimal exposure time and direction-dependent cellular effects of helium and argon plasma on the 4T1 cancer cells and a triple-negative mouse model of breast cancer. This study used two types of helium and argon plasma jet devices with different input parameters. In vitro evaluations on 4T1 cell line using the MTT assays and flow cytometry analysis demonstrate CAP-induced apoptosis in all treated groups, especially in the direct approach. These changes were concurrent with increased intracellular reactive oxygen species levels and decreased total antioxidant capacity in these cells. In vivo studies concurrent with in vitro results revealed that CAP therapy reduces tumor size, decreases Nottingham histological score, prevents weight loss, and increases the survival rate in all treated groups. These results suggest that plasma therapy may overcome the adverse effects of approved cancer therapeutic strategies and seems to be a significant issue for cancer patients in the clinical stage, alone or in combination with current therapeutic programs.
Collapse
Affiliation(s)
- Mahdiyeh Bakhtiyari-Ramezani
- Plasma Physics and Nuclear Fusion Research School, Nuclear Science and Technology Research Institute (NSTRI), Tehran, Iran.
| | - Meysam Nasiri
- Department of Cellular and Molecular Biology, School of Biology, Damghan University, Damghan, Iran
| | - Mansoureh Baniasadi
- Department of Biotechnology, Knowledge-Intensive Plasma Technology Development Company, Tehran, Iran
| |
Collapse
|
13
|
Azevedo T, Silva-Reis R, Medeiros-Fonseca B, Gonçalves M, Mendes G, Roboredo M, Rocha MJ, Peixoto F, Pinto MDL, Matos M, Sousa JR, Oliveira PA, Coimbra AM. Do (xeno)estrogens pose a risk to earthworms? Soy isoflavones and estradiol impact gonad structure and induce oxidative stress in Eisenia fetida. CHEMOSPHERE 2025; 377:144315. [PMID: 40147346 DOI: 10.1016/j.chemosphere.2025.144315] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/06/2024] [Revised: 02/04/2025] [Accepted: 03/12/2025] [Indexed: 03/29/2025]
Abstract
Understanding the impact of endocrine disruptor compounds (EDCs) across a wide range of species is crucial, given their ubiquitous presence. Although invertebrate species lack sex steroid hormone pathways, they exhibit sensitivity to EDCs, which could affect population dynamics. This study assessed reproductive endpoints and oxidative stress parameters in Eisenia fetida following exposure to estradiol and soy isoflavones, resembling the concentrations found in livestock manure. The experiment used artificial soil, as recommended by OECD guidelines (7:2:1 sand, kaolin and peat). Adult specimens were randomly divided into seven groups (n = 11/replicate): one control, three estradiol (156.1, 283.4 and 633.8 μg/kg of dry soil) and three soy isoflavones (113.0, 215.3 and 405.0 mg/kg of dry soil) concentrations. After eight weeks, samples were collected for cytological, histological and biochemical analysis. Offspring development was assessed after 12 additional weeks. Higher estradiol and isoflavone concentrations led to lower germ cell number and increased abnormalities, especially in the seminal vesicles and ovaries. Catalase and peroxidase activities were significantly increased in all treated groups. The exposure did not significantly affect the number of E. fetida offspring. These findings highlight E. fetida's sensitivity to EDCs at a biochemical and tissue level, suggesting its use as a bioindicator for assessing EDC contamination in soils.
Collapse
Affiliation(s)
- Tiago Azevedo
- UTAD - University of Trás-os-Montes and Alto Douro, 5000-801, Vila Real, Portugal; CITAB - Centre for the Research and Technology of Agro-Environmental and Biological Sciences, 5000-801, Vila Real, Portugal.
| | - Rita Silva-Reis
- UTAD - University of Trás-os-Montes and Alto Douro, 5000-801, Vila Real, Portugal; CITAB - Centre for the Research and Technology of Agro-Environmental and Biological Sciences, 5000-801, Vila Real, Portugal; LAQV-REQUIMTE, Department of Chemistry, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Beatriz Medeiros-Fonseca
- UTAD - University of Trás-os-Montes and Alto Douro, 5000-801, Vila Real, Portugal; CITAB - Centre for the Research and Technology of Agro-Environmental and Biological Sciences, 5000-801, Vila Real, Portugal; Molecular Oncology and Viral Pathology Group, Research Center of IPO Porto (CI-IPOP) & RISE@CI-IPOP (Health Research Network), Portuguese Oncology Institute of Porto (IPO Porto), Porto Comprehensive Cancer Center (Porto.CCC), 4200-072, Porto, Portugal
| | - Mariana Gonçalves
- UTAD - University of Trás-os-Montes and Alto Douro, 5000-801, Vila Real, Portugal; CITAB - Centre for the Research and Technology of Agro-Environmental and Biological Sciences, 5000-801, Vila Real, Portugal; Research and Development Unit, Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge, INSA I.P, 4000-055, Porto, Portugal; CECA - Center for the Study of Animal Science, University of Porto, 4051-401, Porto, Portugal; Al4AnimalS- Associate Laboratory for Animal and Veterinary Sciences, Faculdade de Medicina Veterinária, Lisboa, 1300-477, Portugal
| | - Gabriel Mendes
- UTAD - University of Trás-os-Montes and Alto Douro, 5000-801, Vila Real, Portugal
| | - Marta Roboredo
- UTAD - University of Trás-os-Montes and Alto Douro, 5000-801, Vila Real, Portugal; CITAB - Centre for the Research and Technology of Agro-Environmental and Biological Sciences, 5000-801, Vila Real, Portugal
| | - Maria J Rocha
- Laboratory of Histology and Embryology, Department of Microscopy, ICBAS - School of Medicine and Biomedical Sciences - University of Porto, 4050-313, Porto, Portugal; Animal Morphology and Toxicology Team, CIMAR/CIIMAR - Interdisciplinary Centre of Marine and Environmental Research, University of Porto, 4450-208, Matosinhos, Portugal
| | - Francisco Peixoto
- UTAD - University of Trás-os-Montes and Alto Douro, 5000-801, Vila Real, Portugal; CQ-VRV - Chemistry Center-Vila Real, 5001-801, Vila Real, Portugal
| | - Maria de Lurdes Pinto
- UTAD - University of Trás-os-Montes and Alto Douro, 5000-801, Vila Real, Portugal; CECAV - Animal and Veterinary Research Center, Al4AnimalS - Associate Laboratory for Animal and Veterinary Sciences, 5000-801, Vila Real, Portugal
| | - Manuela Matos
- UTAD - University of Trás-os-Montes and Alto Douro, 5000-801, Vila Real, Portugal; CITAB - Centre for the Research and Technology of Agro-Environmental and Biological Sciences, 5000-801, Vila Real, Portugal; Inov4Agro - Institute for Innovation, Capacity Building and Sustainability of Agri-food Production, 5000-801, Vila Real, Portugal
| | - João R Sousa
- UTAD - University of Trás-os-Montes and Alto Douro, 5000-801, Vila Real, Portugal; CITAB - Centre for the Research and Technology of Agro-Environmental and Biological Sciences, 5000-801, Vila Real, Portugal; Inov4Agro - Institute for Innovation, Capacity Building and Sustainability of Agri-food Production, 5000-801, Vila Real, Portugal
| | - Paula A Oliveira
- UTAD - University of Trás-os-Montes and Alto Douro, 5000-801, Vila Real, Portugal; CITAB - Centre for the Research and Technology of Agro-Environmental and Biological Sciences, 5000-801, Vila Real, Portugal; Inov4Agro - Institute for Innovation, Capacity Building and Sustainability of Agri-food Production, 5000-801, Vila Real, Portugal
| | - Ana M Coimbra
- UTAD - University of Trás-os-Montes and Alto Douro, 5000-801, Vila Real, Portugal; CITAB - Centre for the Research and Technology of Agro-Environmental and Biological Sciences, 5000-801, Vila Real, Portugal; Inov4Agro - Institute for Innovation, Capacity Building and Sustainability of Agri-food Production, 5000-801, Vila Real, Portugal.
| |
Collapse
|
14
|
Eladawy RM, Ahmed LA, Salem MB, El-Sayed RM, Salem HA, Mohamed AF. Probiotics reverse gut dysbiosis and memory impairment associated with esomeprazole use in chronically stressed rats: A significant neuroprotective role for cholecystokinin. Int Immunopharmacol 2025; 150:114227. [PMID: 39952008 DOI: 10.1016/j.intimp.2025.114227] [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: 11/27/2024] [Revised: 01/30/2025] [Accepted: 02/02/2025] [Indexed: 02/17/2025]
Abstract
Recent studies propose an association between the prolonged usage of gastric acid suppressants (GAS) and the incidence of dementia in stressed patients. This association was confirmed in our investigation recently published with a significant role for gut dysbiosis using different GAS, especially PPI esomeprazole (Esom). Hence, this work explored the influence of different probiotics on gut dysbiosis associated with Esom use in unpredictable chronic mild stress (UCMS)-induced cognitive impairment. Rats were given Esom (3.7 mg/kg/day orally) with exposure to UCMS for 7 weeks and treated with Lactobacillus delbrukii and Lactobacillus fermentum (LB) (1 × 1010 CFU/day orally) or Bacillus clausii (BC) (1 × 109 CFU/day orally) in the last 3 weeks of the experiment. LB and BC attenuated the cognitive impairment associated with Esom use in the presence of UCMS, where BC showed more remarkable results. These results were correlated with improvement of dysbiosis and gut membrane integrity by reducing colonic inflammation via hampering the NLRP3 inflammasome pathway. The improvement of gut dysbiosis was further interrelated with decrease in systemic inflammation and improvement of cholecystokinin (CCK) level. The neuroprotective effects in LB and BC groups were achieved via enhancement of brain-derived neurotrophic factor (BDNF) by 2.7 and 3.4-folds, respectively, through CCK activation with decline of hippocampal amyloid β accumulation by 67.29 % and 97.9 %, respectively, compared to UCMS with Esom group. Our study supports the neuroprotective effect of probiotics on cognitive impairment attributed to long-term use of GAS in the presence of stress, with a significant role for gut microbiota modulation.
Collapse
Affiliation(s)
- Reem M Eladawy
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Sinai University - Arish Branch 45511 Arish, Egypt.
| | - Lamiaa A Ahmed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt.
| | - Maha B Salem
- Pharmacology Department, Theodor Bilharz Research Institute, Giza, Egypt
| | - Rehab M El-Sayed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Sinai University - Arish Branch 45511 Arish, Egypt
| | - Hesham A Salem
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Ahmed F Mohamed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Cairo University, Cairo, Egypt; Faculty of Pharmacy, King Salman International University (KSIU), South Sinai 46612, Egypt
| |
Collapse
|
15
|
Xian S, Meng F, Chen X, Zhu L, Wang H. Reduction of colitis in mice by chemically programmed supramolecular nanoassemblies of vitamin-lipid conjugates. J Nanobiotechnology 2025; 23:247. [PMID: 40128782 PMCID: PMC11934663 DOI: 10.1186/s12951-025-03322-0] [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: 11/11/2024] [Accepted: 03/11/2025] [Indexed: 03/26/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a relapsing disorder characterized by uncontrolled chronic inflammation of the gastrointestinal tract, posing a significant therapeutic challenge owing to the limited efficacy and undesirable side effects of current therapeutic options. A key pathological hallmark of IBD is the excessive production of reactive oxygen species (ROS). Hence, therapeutic strategies aimed at reducing ROS levels are promising for relieving these inflammatory conditions. Vitamin C-a natural nutrient for the human body-is well known for its potent antioxidant effects. However, the clinical development of vitamin C as a therapeutic drug has been hindered by its poor stability, rapid metabolism, and inadequate tissue accumulation. Herein, we report that the bioavailability of vitamin C can be enhanced by chemically reprogramming it with a small panel of long-chain fatty acids that aid in the aqueous self-assembly of the resulting drug conjugates to create self-deliverable nanoassemblies, enhancing their inflammation disease-oriented delivery and cellular uptake. In mice with dextran sulfate sodium-induced colitis, the optimal vitamin C-lipid nanoassemblies preferentially accumulated in inflamed colonic tissues following systemic administration and substantially ameliorated disease severity. We extended this strategy to incorporate the clinically approved glucocorticoid budesonide into the vitamin C nanosystem, facilitating a synergistic combination. In the chronic colitis model, the combination treatment reduced inflammation without compromising global immunity. Mechanistically, the treatment modulated the intestinal inflammatory microenvironment and altered the immune cell landscape, partly through regulation of the gut microbiome. Given its anticipated negligible side effects, this novel nanoassembly platform leveraging small-molecule lipidation may become a promising therapeutic for treating various inflammatory diseases.
Collapse
Affiliation(s)
- Shiyun Xian
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong Province, 250118, P. R. China
- The First Affiliated Hospital, NHC Key Laboratory of Combined Multi-Organ Transplantation, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310003, P. R. China
| | - Fanchao Meng
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong Province, 250118, P. R. China
- The First Affiliated Hospital, NHC Key Laboratory of Combined Multi-Organ Transplantation, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310003, P. R. China
| | - Xiaona Chen
- Department of Medical Oncology, Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310003, P. R. China
| | - Liqing Zhu
- Department of Clinical Laboratory, Peking University Cancer Hospital and Institute, Beijing, P. R. China.
| | - Hangxiang Wang
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong Province, 250118, P. R. China.
- The First Affiliated Hospital, NHC Key Laboratory of Combined Multi-Organ Transplantation, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, Zhejiang University School of Medicine, Hangzhou, Zhejiang Province, 310003, P. R. China.
| |
Collapse
|
16
|
Byrne SR, DeMott MS, Yuan Y, Ghanegolmohammadi F, Kaiser S, Fox JG, Alm EJ, Dedon PC. Temporal dynamics and metagenomics of phosphorothioate epigenomes in the human gut microbiome. MICROBIOME 2025; 13:81. [PMID: 40128848 PMCID: PMC11931770 DOI: 10.1186/s40168-025-02071-4] [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: 09/19/2024] [Accepted: 02/24/2025] [Indexed: 03/26/2025]
Abstract
BACKGROUND Epigenetic regulation of gene expression and host defense is well established in microbial communities, with dozens of DNA modifications comprising the epigenomes of prokaryotes and bacteriophage. Phosphorothioation (PT) of DNA, in which a chemically reactive sulfur atom replaces a non-bridging oxygen in the sugar-phosphate backbone, is catalyzed by dnd and ssp gene families widespread in bacteria and archaea. However, little is known about the role of PTs or other microbial epigenetic modifications in the human microbiome. Here we optimized and applied fecal DNA extraction, mass spectrometric, and metagenomics technologies to characterize the landscape and temporal dynamics of gut microbes possessing PT modifications. RESULTS Exploiting the nuclease-resistance of PTs, mass spectrometric analysis of limit digests of PT-containing DNA reveals PT dinucleotides as part of genomic consensus sequences, with 16 possible dinucleotide combinations. Analysis of mouse fecal DNA revealed a highly uniform spectrum of 11 PT dinucleotides in all littermates, with PTs estimated to occur in 5-10% of gut microbes. Though at similar levels, PT dinucleotides in fecal DNA from 11 healthy humans possessed signature combinations and levels of individual PTs. Comparison with a widely distributed microbial epigenetic mark, m6dA, suggested temporal dynamics consistent with expectations for gut microbial communities based on Taylor's Power Law. Application of PT-seq for site-specific metagenomic analysis of PT-containing bacteria in one fecal donor revealed the larger consensus sequences for the PT dinucleotides in Bacteroidota, Bacillota (formerly Firmicutes), Actinomycetota (formerly Actinobacteria), and Pseudomonadota (formerly Proteobacteria), which differed from unbiased metagenomics and suggested that the abundance of PT-containing bacteria did not simply mirror the spectrum of gut bacteria. PT-seq further revealed low abundance PT sites not detected as dinucleotides by mass spectrometry, attesting to the complementarity of the technologies. Video Abstract CONCLUSIONS: The results of our studies provide a benchmark for understanding the behavior of an abundant and chemically reactive epigenetic mark in the human gut microbiome, with implications for inflammatory conditions of the gut.
Collapse
Affiliation(s)
- Shane R Byrne
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Michael S DeMott
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yifeng Yuan
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | | | - Stefanie Kaiser
- Pharmaceutical Chemistry, Goethe University, Frankfurt, Germany
| | - James G Fox
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA
- Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Eric J Alm
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA, USA
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Singapore-MIT Alliance for Research and Technology, Antimicrobial Resistance IRG, Singapore, Singapore
| | - Peter C Dedon
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Center for Environmental Health Sciences, Massachusetts Institute of Technology, Cambridge, MA, USA.
- Singapore-MIT Alliance for Research and Technology, Antimicrobial Resistance IRG, Singapore, Singapore.
| |
Collapse
|
17
|
Bano N, Khan S, Ahamad S, Dar NJ, Alanazi HH, Nazir A, Bhat SA. Microglial NOX2 as a therapeutic target in traumatic brain injury: Mechanisms, consequences, and potential for neuroprotection. Ageing Res Rev 2025; 108:102735. [PMID: 40122395 DOI: 10.1016/j.arr.2025.102735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2025] [Revised: 03/08/2025] [Accepted: 03/16/2025] [Indexed: 03/25/2025]
Abstract
Traumatic brain injury (TBI) is a leading cause of long-term disability worldwide, with secondary injury mechanisms, including neuroinflammation and oxidative stress, driving much of its chronic pathology. While NADPH oxidase 2 (NOX2)-mediated reactive oxygen species (ROS) production is a recognized factor in TBI, the specific role of microglial NOX2 in perpetuating oxidative and inflammatory damage remains underexplored. Addressing this gap is critical, as current therapeutic approaches primarily target acute symptoms and fail to interrupt the persistent neuroinflammation that contributes to progressive neurodegeneration. Besides NOX, other ROS-generating enzymes, such as CYP1B1, COX2, and XO, also play crucial roles in triggering oxidative stress and neuroinflammatory conditions in TBI. However, this review highlights the pathophysiological role of microglial NOX2 in TBI, focusing on its activation following injury and its impact on ROS generation, neuroinflammatory signaling, and neuronal loss. These insights reveal NOX2 as a critical driver of secondary injury, linked to worsened outcomes, particularly in aged individuals where NOX2 activation is more pronounced. In addition, this review evaluates emerging therapeutic approaches targeting NOX2, such as GSK2795039 and other selective NOX2 inhibitors, which show potential in reducing ROS levels, limiting neuroinflammation, and preserving neurological functions. By highlighting the specific role of NOX2 in microglial ROS production and secondary neurodegeneration, this study advocates for NOX2 inhibition as a promising strategy to improve TBI outcomes by addressing the unmet need for therapies targeting long-term inflammation and neuroprotection. Our review highlights the potential of NOX2-targeted interventions to disrupt the cycle of oxidative stress and inflammation, ultimately offering a pathway to mitigate the chronic impact of TBI.
Collapse
Affiliation(s)
- Nargis Bano
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Sameera Khan
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India
| | - Shakir Ahamad
- Department of Chemistry, Aligarh Muslim University, Aligarh 202002, India
| | - Nawab John Dar
- CNB, SALK Institute of Biological Sciences, La Jolla, CA 92037, USA
| | - Hamad H Alanazi
- Department of Clinical Laboratory Science, College of Applied Medical Sciences, Al Jouf University 77455, Saudi Arabia
| | - Aamir Nazir
- Division of Neuroscience and Ageing Biology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh, India; Academy of Scientific and Innovative Research, New Delhi, India.
| | - Shahnawaz Ali Bhat
- Department of Zoology, Aligarh Muslim University, Aligarh 202002, India.
| |
Collapse
|
18
|
Yakout SM, AlSubhi MA, Hussain SD, Alnaami AM, Alamro AA, Al-Daghri NM. Oxidative stress biomarkers of neonates in Saudi population: An observational study. Medicine (Baltimore) 2025; 104:e41926. [PMID: 40128061 PMCID: PMC11936561 DOI: 10.1097/md.0000000000041926] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 03/05/2025] [Indexed: 03/26/2025] Open
Abstract
Oxidative stress results from an imbalance between free radical production and antioxidant defense, significantly impacting neonatal health, particularly in preterm infants with immature antioxidant systems. This study aims to assess oxidative stress in Saudi neonates by measuring key antioxidants, both enzymatic (superoxide dismutase, catalase, glutathione peroxidase) and nonenzymatic (glutathione, bilirubin, uric acid), and comparing them across sex (male vs female) and term status (full term vs preterm). A total of 110 Saudi neonates (55 normal neonates and 55 preterm neonates; 52 females and 58 males) were included in this study. The gestational age of preterm neonates ranged from 28 to 36 weeks, with a mean of 32 weeks. Serum samples were retrieved from the chair for biomarkers of chronic diseases BioBank. Ethical approval was obtained from the College of Medicine, King Saud University. GSH levels were higher in preterm neonates compared to normal neonates (16.4 vs 11.0 µmol/L, P < .001), and uric acid levels were higher in normal neonates compared to preterm neonates (246.2 vs 206.2 µmol/L, P < .015). SOD1 levels were higher in preterm neonates compared to normal neonates (291.5 vs 225.4 ng/mL, P < .040). In terms of both term and sex of neonates, GSH levels were higher in preterm female neonates compared to normal female neonates (16.8 vs 13.8 µmol/L, P < .054), and in preterm male neonates compared to normal male neonates (16.4 vs 9.2 µmol/L, P < .001). SOD1 levels were higher in preterm male neonates compared to normal male neonates (300.1 vs 198.8 ng/mL, P < .038), and uric acid levels were higher in normal male neonates compared to preterm male neonates (243.9 vs 200.1 µmol/L, P < .011). GPx-1 levels were higher in preterm neonates compared to normal neonates (14.6 vs 7.9 ng/mL, P < .006). There are no differences in antioxidant parameters between female and male neonates. However, some antioxidants differ between preterm and normal neonates. The comparison according to both sex and term status also showed differences in some antioxidant parameters.
Collapse
Affiliation(s)
- Sobhy M. Yakout
- Chair for Biomarkers of Chronic Diseases, Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Malak A. AlSubhi
- Chair for Biomarkers of Chronic Diseases, Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Syed D. Hussain
- Chair for Biomarkers of Chronic Diseases, Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Abdullah M. Alnaami
- Chair for Biomarkers of Chronic Diseases, Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Abir A. Alamro
- Chair for Biomarkers of Chronic Diseases, Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| | - Nasser M. Al-Daghri
- Chair for Biomarkers of Chronic Diseases, Department of Biochemistry, College of Science, King Saud University, Riyadh, Saudi Arabia
| |
Collapse
|
19
|
Chen D, Yu R, Cai Y, Zhang H, Jiang Y, Wu Y, Peng XE. Additive interaction between hepatitis B virus infection and tobacco smoking on the risk of gastric cancer in a Chinese population. Infect Agent Cancer 2025; 20:19. [PMID: 40114245 PMCID: PMC11927330 DOI: 10.1186/s13027-025-00648-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
OBJECTIVE Although hepatitis B virus (HBV) infection was regarded as a risk factor for liver cancer, the association of HBV infection with gastric cancer (GC) is unclear. In this study, we aim to assess the association of HBV infection with the risk of GC and explore the interaction between HBV infection and other risk factors. METHODS A case-control study was conducted and 409 GC cases and 1275 healthy controls were enrolled in Fujian province, China. Serum hepatitis B surface antigen (HBsAg) was measured and epidemiological data were collected. The association between HBV infection and GC risk was analyzed using logistic regression and meta-analysis method was employed to make estimates more conservative. Meanwhile, multiplicative and additive models were used to explore the interaction between HBV infection and other risk factors. RESULTS The prevalence of serum HBsAg positivity was 13.20% among GC cases and 6.20% among controls. Compared to HBsAg-negative subjects, the adjusted odds ratios (OR) for HBsAg positive were 3.30 [95% confidence interval (CI): (1.84-5.91)]. Compared to HBsAg-negative never smokers, the adjusted OR was 2.00 (95%CI: 1.19-3.34) for HBsAg-negative ever smokers,4.27 (95%CI: 1.97-9.26) for HBsAg-positive never smokers, and 4.73 (95%CI: 1.85-12.08) for HBsAg-positive ever smokers. These evidences indicated super-additive [API (95%CI): 0.78 (0.67-0.90), S (95%CI): 5.45 (3.26-9.08)] between HBV infection and tobacco smoking. No interaction between HBV infection and alcohol drinking was found on the risk of GC. CONCLUSIONS HBV infection increased the risk of GC, and tobacco smoking and HBV infection may positively interact in the development of GC.
Collapse
Affiliation(s)
- Danjing Chen
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, People's Republic of China
| | - Rong Yu
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, People's Republic of China
- School and Hospital of Stomatology, Fujian Medical University, Fuzhou, 350002, People's Republic of China
| | - Yongfeng Cai
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, People's Republic of China
| | - He Zhang
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, People's Republic of China
| | - Yijun Jiang
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, People's Republic of China
| | - Yunli Wu
- Key Laboratory of Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Ministry of Education, Fujian Medical University, Fuzhou, 350108, People's Republic of China
| | - Xian-E Peng
- Department of Epidemiology and Health Statistics, Fujian Provincial Key Laboratory of Environment Factors and Cancer, School of Public Health, Fujian Medical University, Fuzhou, 350122, People's Republic of China.
- Key Laboratory of Gastrointestinal Cancer, School of Basic Medical Sciences, Fujian Medical University, Ministry of Education, Fujian Medical University, Fuzhou, 350108, People's Republic of China.
- Department of Epidemiology and Health Statistics, Minhou Country, Xuefu North Road 1st, Shangjie Town, Minhou Country, Fuzhou, 350108, Fujian, China.
| |
Collapse
|
20
|
Xu L, Wang Y, Hu Y, Dai X, Sun C, Cheng J. ROS-responsive oridonin and dihydroartemisinin hetero-polymeric prodrug NPs for potentiating ferroptosis in gastric cancer by disrupting redox balance. Colloids Surf B Biointerfaces 2025; 252:114637. [PMID: 40132335 DOI: 10.1016/j.colsurfb.2025.114637] [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: 11/30/2024] [Revised: 03/10/2025] [Accepted: 03/12/2025] [Indexed: 03/27/2025]
Abstract
Gastric cancer presents a significant global health concern, with conventional therapies often limited in effectiveness. The abnormal redox balance in gastric cancer cells may represent a breakthrough in the treatment of gastric cancer. In this study, we report for the first time the development of reactive oxygen species (ROS)-sensitive hetero-polymeric prodrug nanoparticles (NPs) designed for the co-delivery of the Chinese herbal extract oridonin (ORI) and dihydroartemisinin (DHA) in combination therapy for gastric cancer. This strategy aims to disrupt the intracellular redox balance and ultimately induce ferroptosis in gastric cancer cells. The ROS-responsive ORI and DHA polymeric prodrug were synthesised by conjugating ORI or DHA to poly(ethylene glycol)-block-poly(L-lysine) (PEG-b-PLL) via a ROS-sensitive linker thioketal (TK). The resulting polymeric prodrugs self-assemble in water to form NPs OD-M. After internalization by gastric cancer cells, OD-M released ORI and DHA in response to high ROS conditions within cancer cells. The released ORI reacts with GSH to induce GSH depletion while DHA amplifies intracellular ROS levels, ultimately inducing ferroptosis in gastric cancer cells. Experimental results demonstrate that OD-M acts as both a GSH scavenger and ROS generator, effectively disrupting intracellular redox balance, inducing ferroptosis, and exhibiting effective anticancer efficacy in vitro and in vivo, offering a departure from traditional methods.
Collapse
Affiliation(s)
- Luzhou Xu
- Department of Gastroenterology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China.
| | - Yan Wang
- Department of Gastroenterology, Affiliated Hospital of Nanjing University of Chinese Medicine, Jiangsu Province Hospital of Chinese Medicine, Nanjing 210029, China
| | - Yanqin Hu
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Xinyi Dai
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Cheng Sun
- The First School of Clinical Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, China
| | - Jun Cheng
- Jiangsu Hongdian Chinese Medicine Industry Research Institute, Nanjing 210042, China
| |
Collapse
|
21
|
Liu J, Dai Y, Yang W, Chen ZY. Role of Mushroom Polysaccharides in Modulation of GI Homeostasis and Protection of GI Barrier. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:6416-6441. [PMID: 40063730 PMCID: PMC11926878 DOI: 10.1021/acs.jafc.5c00745] [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: 03/20/2025]
Abstract
Edible and medicinal mushroom polysaccharides (EMMPs) have been widely studied for their various biological activities. It has been shown that EMMPs could modulate microbiota in the large intestine and improve intestinal health. However, the role of EMMPs in protecting the gastric barrier, regulating gastric microbiota, and improving gastric health cannot be ignored. Hence, this review will elucidate the effect of EMMPs on gastric and intestinal barriers, with emphasis on the interaction of EMMPs with microbiota in maintaining overall gastrointestinal health. Additionally, this review highlights the gastroprotective effects and underlying mechanisms of EMMPs against gastric mucosa injury, gastritis, gastric ulcer, and gastric cancer. Furthermore, the effects of EMMPs on intestinal diseases, including inflammatory bowel disease, colorectal cancer, and intestinal infection, are also summarized. This review will also discuss the future perspective and challenges in the use of EMMPs as a dietary supplement or a nutraceutical in preventing and treating gastrointestinal diseases.
Collapse
Affiliation(s)
- Jianhui Liu
- Collaborative Innovation Center for Modern Grain Circulation and Safety, Jiangsu Province Engineering Research Center of Edible Fungus Preservation and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong 999077, China
| | - Yi Dai
- Collaborative Innovation Center for Modern Grain Circulation and Safety, Jiangsu Province Engineering Research Center of Edible Fungus Preservation and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China
| | - Wenjian Yang
- Collaborative Innovation Center for Modern Grain Circulation and Safety, Jiangsu Province Engineering Research Center of Edible Fungus Preservation and Intensive Processing, College of Food Science and Engineering, Nanjing University of Finance and Economics, Nanjing 210023, China
| | - Zhen-Yu Chen
- School of Life Sciences, The Chinese University of Hong Kong, Shatin, NT, Hong Kong 999077, China
| |
Collapse
|
22
|
Lin CH, Jiang WP, Itokazu N, Huang GJ. Chlorogenic acid attenuates 5-fluorouracil-induced intestinal mucositis in mice through SIRT1 signaling-mediated oxidative stress and inflammatory pathways. Biomed Pharmacother 2025; 186:117982. [PMID: 40106967 DOI: 10.1016/j.biopha.2025.117982] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2025] [Revised: 02/17/2025] [Accepted: 03/10/2025] [Indexed: 03/22/2025] Open
Abstract
Mucositis, a common side effect of the chemotherapeutic drug 5-Fluorouracil (5-FU), causes severe and aggravating effects on mucosal cells in the oral cavity and intestine. This study in mice aimed to assess the antioxidant, anti-inflammatory, and mucosal protective properties of chlorogenic acid in mitigating 5-FU-induced intestinal mucositis. To investigate these potential protective effects, we developed a mouse model by administering an initial intraperitoneal (i.p.) injection of 5-FU, followed by daily i.p. injections of chlorogenic acid (10 and 20 mg/kg) for 10 consecutive days. Chlorogenic acid mitigated intestinal histopathological damage, reduced proinflammatory mediators and malondialdehyde (MDA) levels, and increased the glutathione (GSH) level by 5-FU. Chlorogenic acid treatment led to a significant reduction in the expression of inflammation-related proteins decreased oxidative stress-related proteins and, attenuated the expression of apoptosis and autophagy-related proteins in small intestinal tissues. Additional investigations are necessary to verify our findings and enhance our comprehension of how SIRT1 inhibition (EX-527) counteracts the anti-inflammatory effects of chlorogenic acid in intestinal tissues. In conclusion, our mice study has shown that chlorogenic acid exerts its protective effects on 5-FU-induced intestinal tissue damage, by reducing oxidative stress and inflammation through the modulation of multiple signaling pathways, including the TLR4/NF-κB/MAPK, AMPK/ SIRT1, and PI3K/AKT axis. These findings highlight the potential of chlorogenic acid as a therapeutic agent for mucositis, given its anti-inflammatory and antioxidant properties.
Collapse
Affiliation(s)
- Che-Hsuan Lin
- Department of Otolaryngology, School of Medicine, College of Medicine, Taipei Medical University, Taipei 11031, Taiwan; Department of Otolaryngology, Taipei Medical University Hospital, Taipei Medical University, Taipei 11031, Taiwan
| | - Wen-Ping Jiang
- School of Pharmacy, College of Pharmacy, China Medical University, Taichung 404, Taiwan
| | - Nanae Itokazu
- Department of Pharmaceutical Sciences, Nihon Pharmaceutical University, Saitama 3620806, Japan
| | - Guan-Jhong Huang
- Department of Food Nutrition and Healthy Biotechnology, Asia University, Asia University, Taichung 413, Taiwan; Department of Chinese Pharmaceutical Sciences and Chinese Medicine Resources, College of Chinese Medicine, China Medical University, Taichung 404, Taiwan.
| |
Collapse
|
23
|
Sun J, Cao X, Li Y, Yu K, Cong Y, Pan Q, Yin Y, Wang J. Oxidative stress in the liver of chicken during fowl adenovirus serotype 4 infection. Poult Sci 2025; 104:105054. [PMID: 40120244 DOI: 10.1016/j.psj.2025.105054] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2025] [Revised: 03/11/2025] [Accepted: 03/15/2025] [Indexed: 03/25/2025] Open
Abstract
Hepatitis is a significant pathological manifestation of fowl adenovirus serotype-4 (FAdV-4) infection, which is a crucial factor contributing to the mortality of chickens. The pathophysiology of liver disease is rooted in oxidative stress. The present study aims to investigate the presence of oxidative stress during the liver lesion process in FAdV-4 infection. Specifically, one-day-old specific pathogen-free (SPF) chickens were allocated into three groups, the control group, the infection group, and the quercetin group. The quercetin group received daily oral administration of quercetin. At the age of 12 days, the chickens belonging to both the infection and quercetin groups were subjected to intramuscular injection of FAdV-4 (0.3 mL103TCID50/mL). Samples were collected from each group at 2, 4, and 6 days post-infection (dpi), and sera were collected to measure the levels of ALT and AST. A portion of liver tissue was fixed to examine the histological changes, cell apoptosis, and mitochondrial morphology, while another portion was homogenized and mitochondria were isolated. The levels of MDA, SOD, H2O2, and GSH-Px in the homogenate supernatants of livers and isolated mitochondria were measured, and the viral load in the liver was studied. And Cyt C levels in the mitochondria and cytosolic supernatant were recorded. The results showed that AST and ALT in the serum of chicken in the infection group were significantly higher than those in the control and quercetin group at 6 dpi. Obvious swelling, steatosis, necrosis, and inflammatory cell infiltration were observed in the liver of the infection group. Administered with quercetin can significantly decrease the viral load in the liver at 4 and 6 dpi. H2O2 in the liver, and MDA, H2O2, GSH and SOD levels in mitochondria in the hepatocyte of the infection group were significantly higher than those in the control and quercetin groups. Cyt C in the mitochondria of the hepatocyte of infection and quercetin groups were significantly lower than those in the control group at 2 dpi. Cyt C in the cytoplasm of the liver in chicken in the quercetin group was significantly higher than those in the control and infection groups. It was found that the outer mitochondrial membrane in hepatocytes was fractured in the infection group. The proportion of apoptotic cells in the liver in the infection groups was significantly higher than those in the control and quercetin group at 4 dpi, and that in the control group was significantly lower than in the infection and quercetin group. The results suggested that during liver injury induced by FAdV-4 infection, oxidative damage occurred obviously in the liver and mitochondria, and hepatocyte apoptosis was observed. Quercetin, as an antioxidant, can inhibit virus replication to some extent, and alleviate oxidative damage, liver damage, and the mortality caused by FAdV-4 infection.
Collapse
Affiliation(s)
- Jiayu Sun
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Xu Cao
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Yufeng Li
- Institute of Poultry Science, Shandong Academy of Agricultural Sciences, Jinan, Shandong 250023, PR China
| | - Kexiang Yu
- Institute of Poultry Science, Shandong Academy of Agricultural Sciences, Jinan, Shandong 250023, PR China
| | - Yanfang Cong
- Qingdao VL and Biotech Inc, Qingdao 266000, PR China
| | - Qing Pan
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Yanbo Yin
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, PR China
| | - Jianlin Wang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao 266109, PR China.
| |
Collapse
|
24
|
Dibakoane SR, Mhlongo G, Moonsamy G, Wokadala OC, Mnisi CM, Mlambo V. Phenomenological and mechanistic insights into potential dietary nucleotide - probiotic synergies in layer chickens: A review. Poult Sci 2025; 104:105049. [PMID: 40106904 PMCID: PMC11964621 DOI: 10.1016/j.psj.2025.105049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2025] [Revised: 03/08/2025] [Accepted: 03/15/2025] [Indexed: 03/22/2025] Open
Abstract
Despite their growing popularity as alternatives to antibiotic growth promoters (AGPs), the individual effects of nucleotides and probiotics on poultry gut functionality remain poorly understood. In addition, inconsistent outcomes are quite common in studies where these two additives have been used separately to modify gut function and related parameters in birds. These inconsistencies, which have limited the potential of probiotics and nucleotides as AGP replacements, stem from various factors and need to be addressed. Combining probiotics and nucleotides could potentially enhance their effectiveness and lead to more consistent outcomes in layer chickens. Since their mechanisms of action complement each other, some level of synergy is expected when used together. Both additives have been shown to support gut health, boost immune function, and improve performance in chickens when used individually. However, no studies have investigated the possible synergistic effects of nucleotides and probiotics in poultry. This review makes the case for combined use of probiotics and nucleotides in layer chickens by providing phenomenological and mechanistic insights into hypothetical synergistic effects. This paper highlights the need for AGP alternatives and reviews studies on the effects and mechanisms of probiotics and nucleotides in layer chickens when used individually. We then propose potential mechanisms for their synergistic effects on gut health, performance, and egg quality based on logical deductions from observed biological responses. These proposed mechanisms are hypothetical and require experimental validation. Finally, the review explores how this synergy could lead to more consistent outcomes and enhance the feasibility of AGP-free egg production.
Collapse
Affiliation(s)
- Siphosethu R Dibakoane
- School of Agricultural Sciences, Faculty of Agriculture and Natural Sciences, University of Mpumalanga, Nelspruit 1200, South Africa
| | - Godfrey Mhlongo
- School of Agricultural Sciences, Faculty of Agriculture and Natural Sciences, University of Mpumalanga, Nelspruit 1200, South Africa
| | - Ghaneshree Moonsamy
- Council for Scientific and Industrial Research (CSIR); Future production: Chemicals, Meiring Naude Drive, Pretoria 0081, South Africa
| | - Obiro Cuthbert Wokadala
- School of Agricultural Sciences, Faculty of Agriculture and Natural Sciences, University of Mpumalanga, Nelspruit 1200, South Africa
| | - Caven Mguvane Mnisi
- Department of Animal Science, Faculty of Natural and Agricultural Sciences, North-West University, Mafikeng 2735, South Africa; Food Security and Safety Focus Area, Faculty of Natural and Agricultural Sciences, North-West University, Mafikeng 2735, South Africa
| | - Victor Mlambo
- School of Agricultural Sciences, Faculty of Agriculture and Natural Sciences, University of Mpumalanga, Nelspruit 1200, South Africa.
| |
Collapse
|
25
|
Bollu VS, Chen YC, Zhang F, Gowda K, Amin S, Sharma AK, Schell TD, Zhu J, Robertson GP. Managing telomerase and telomere dysfunction in acral melanoma. Pharmacol Res 2025; 215:107700. [PMID: 40097124 DOI: 10.1016/j.phrs.2025.107700] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 03/04/2025] [Accepted: 03/12/2025] [Indexed: 03/19/2025]
Abstract
Acral Lentiginous Melanoma is a rare and aggressive subtype of melanoma that commonly affects the palms, soles, and nail beds. It is more prevalent in individuals with darker skin tones, including Asian, African, and Hispanic populations. Unlike cutaneous melanomas, acral melanoma is not associated with UV exposure and has a distinct genetic and molecular profile, underscoring the need for tailored research and treatment strategies. Standard treatments, such as surgery, chemotherapy, immunotherapy, and targeted therapies, have shown limited success for this melanoma subtype, highlighting the urgency of developing more effective interventions. Telomerase is an enzyme that extends telomeres and is a key target in acral melanoma which exhibits' high telomerase activity, driven by mutations in the telomerase reverse transcriptase TERT promoter, which contributes to uncontrolled tumor cell proliferation, cancer cell immortality, and resistance to conventional therapies. Therefore, targeting telomerase presents a promising therapeutic avenue for acral melanoma patients who do not respond well to current treatments. Several approaches for targeting telomerase deregulation have been developed, and their potential for the management of acral melanoma is discussed in this review. Specifically, the promise of telomerase-targeted therapies for acral melanoma is emphasized and explores how these strategies could improve outcomes for patients with this challenging skin cancer. By focusing on the role of telomerase in tumorigenesis and treatment resistance, telomerase-targeted strategies hold potential as a foundational component of therapies for acral melanoma, complementing existing approaches.
Collapse
Affiliation(s)
- Vishnu Sravan Bollu
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Yu-Chi Chen
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Fan Zhang
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Pharmaceutical Sciences, Washington State University College of Pharmacy and Pharmaceutical Sciences, Spokane, WA 99202, United States
| | - Krishne Gowda
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Shantu Amin
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Arun K Sharma
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Todd D Schell
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Pathology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States
| | - Jiyue Zhu
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Pharmaceutical Sciences, Washington State University College of Pharmacy and Pharmaceutical Sciences, Spokane, WA 99202, United States
| | - Gavin P Robertson
- Department of Pharmacology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Dermatology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Pathology, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Department of Surgery, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Foreman Foundation for Melanoma Research, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Melanoma and Skin Cancer Center, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States; Melanoma Therapeutics Program, Pennsylvania State University College of Medicine, Hershey, PA 17033, United States.
| |
Collapse
|
26
|
Novi S, Caponigro V, Miranda MR, Aquino G, Carri MD, Salviati E, Franceschelli S, Sardo C, Basilicata MG, Vestuto V, Tecce MF, Marini F, Pepe G, Campiglia P, Manfra M. Metabolomics insights into the protective molecular mechanism of Vaccinium myrtillus against oxidative stress in intestinal cells. Sci Rep 2025; 15:8643. [PMID: 40082563 PMCID: PMC11906781 DOI: 10.1038/s41598-025-93722-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2025] [Accepted: 03/10/2025] [Indexed: 03/16/2025] Open
Abstract
Blueberry (Vaccinium myrtillus L.) is a rich source of secondary metabolites known for their potent antioxidant, anti-inflammatory, and cytoprotective properties. These compounds are essential in neutralizing reactive oxygen species (ROS), which are implicated in oxidative stress-related diseases. In this study, we induced oxidative stress in IEC-6 small intestine cells using hydrogen peroxide (H2O2), creating a cellular model to investigate the biochemical response. The obtained results showed that a blueberry extract (BLUBE) significantly exhibited strong antioxidant capacity, as evidenced by DPPH, FRAP and ABTS in vitro tests. Additionally, BLUBE effectively inhibited the release of reactive species in cells and enhanced cytoprotective response, as indicated by improved wound healing and clonogenic potential reduction of stress fibers rearrangement and apoptosis. Metabolomic analysis, specifically High-Resolution Mass Spectrometry (HR-MS), was employed to elucidate the metabolic alterations associated with the protective activity of BLUBE against oxidative stress in IEC-6 cells. Chemometric approaches were applied to preprocess the data, explore variability, and identify systematic biases, ensuring the removal of batch effects and other experimental artifacts. A Partial Least Squares Discriminant Analysis classification model confirmed clear group stratifications with high accuracy (98.75 ± 2.31%), sensitivity, and specificity, aiding in the identification of significant metabolites for pathway enrichment analysis. Key metabolic pathways, including sphingolipid metabolism, taurine and hypotaurine metabolism, glycerophospholipid metabolism, and cysteine and methionine metabolism, were significantly modulated, supporting the biochemical basis of BLUBE's protective effects. In fact, BLUBE was able to partially reverse the downregulation of these pathways, effectively reducing oxidative stress and promoting cell survival. This study highlights the power of HR-MS-based metabolomics in uncovering the mechanisms of nutraceuticals and emphasizes the potential of BLUBE as a protective agent for oxidative stress-related diseases. It also underscores the growing significance of metabolomics in the food and pharmaceutical industries.
Collapse
Affiliation(s)
- Sara Novi
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084, Fisciano, Salerno, Italy
| | - Vicky Caponigro
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084, Fisciano, Salerno, Italy
| | - Maria Rosaria Miranda
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084, Fisciano, Salerno, Italy
- Drug Discovery and Development, University of Salerno, 84084, Fisciano, Salerno, Italy
- National Biodiversity Future Center (NBFC), 90133, Palermo, Italy
| | - Giovanna Aquino
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084, Fisciano, Salerno, Italy
- Drug Discovery and Development, University of Salerno, 84084, Fisciano, Salerno, Italy
| | - Matteo Delli Carri
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084, Fisciano, Salerno, Italy
- Drug Discovery and Development, University of Salerno, 84084, Fisciano, Salerno, Italy
| | - Emanuela Salviati
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084, Fisciano, Salerno, Italy
| | - Silvia Franceschelli
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084, Fisciano, Salerno, Italy
| | - Carla Sardo
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084, Fisciano, Salerno, Italy
| | - Manuela Giovanna Basilicata
- Department of Advanced Medical and Surgical Sciences, University of Campania "Luigi Vanvitelli", Naples, Italy.
| | - Vincenzo Vestuto
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084, Fisciano, Salerno, Italy.
| | - Mario Felice Tecce
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084, Fisciano, Salerno, Italy
| | - Federico Marini
- Department of Chemistry, University of Rome "La Sapienza", Piazzale Aldo Moro 5, 00185, Rome, Italy
| | - Giacomo Pepe
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084, Fisciano, Salerno, Italy
- National Biodiversity Future Center (NBFC), 90133, Palermo, Italy
| | - Pietro Campiglia
- Department of Pharmacy, University of Salerno, Via G. Paolo II, 84084, Fisciano, Salerno, Italy
| | - Michele Manfra
- Department Health Science, University of Basilicata, Viale dell'Ateneo Lucano, 85100, Potenza, Italy
| |
Collapse
|
27
|
Prayoga DK, Pitaloka DAE, Aulifa DL, Budiman A, Levita J, Jiranusornkul S, Nguyen BP. Phytochemical Analysis, Computational Study, and in vitro Assay of Etlingera elatior Inflorescence Extract Towards Inducible Nitric Oxide Synthase. J Exp Pharmacol 2025; 17:123-141. [PMID: 40078169 PMCID: PMC11899951 DOI: 10.2147/jep.s505658] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2024] [Accepted: 12/11/2024] [Indexed: 03/14/2025] Open
Abstract
Background Overproduction of nitric oxide (NO), catalyzed by inducible nitric oxide synthase (iNOS), in the gastric mucosa, contributes to the inflammatory process caused by oxidative stress. Current medications for gastric ulcers, such as proton pump inhibitors and histamine-2 receptor antagonists, have been reported to generate adverse reactions. Purpose To obtain the phytochemical profile of Etlingera elatior inflorescence extract, computational studies, and in vitro assay of the extract towards iNOS. Methods Fresh E. elatior inflorescence petals collected from West Java, Indonesia, were extracted using ethanol, and their nutritional composition, anthocyanin content, and levels of vitamin C, C3G, and quercetin in the extract were determined. Drug-likeness and ADMET properties were predicted, and the binding affinity and stability of the phytoconstituents towards iNOS were studied using molecular docking and molecular dynamic simulation, and in vitro assay of the extract towards human iNOS. Results The extract contains protein 21.81%, fat 0.99%, carbohydrate 38.27%, water 24.56%, and ash 14.37%. The total anthocyanin and vitamin C levels were 47.535 mg/100 g and 985.250 mg/100 g, respectively. The levels of C3G and quercetin were 0.0007% w/w, 0.004% w/w, and 0.0005% w/w, respectively. Drug-likeness and ADMET properties of the constituents showed that most followed Lipinski Rules of Five (Ro5), with few violations. All phytoconstituents occupied the catalytic site by binding to Glu377, and Trp372, similar to S-ethylisothiourea (SEITU) and quinazoline, the iNOS inhibitors. Among these, the flavylium cation of cyanidin, demethoxycurcumin, C3G, cyanidin, and quercetin showed the best binding affinities. Root mean square deviation (RMSD), root mean square fluctuation (RMSF), solvent-accessible surface area (SASA), and radius of gyration (Rg) graphs confirmed the stability of the complexes. E. elatior inflorescence extract inhibited human iNOS with an IC50 value of 24.718 µg/mL. Conclusion Etlingera elatior inflorescence may inhibit iNOS activity due to its anthocyanin and flavonoid content. The flavylium cation of cyanidin, demethoxycurcumin, C3G, cyanidin, and quercetin play leading roles in the interaction with iNOS.
Collapse
Affiliation(s)
| | - Dian Ayu Eka Pitaloka
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
| | - Diah Lia Aulifa
- Department of Pharmaceutical Analysis and Medicinal Chemistry, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
| | - Arif Budiman
- Department of Pharmaceutics and Pharmaceutical Technology, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
| | - Jutti Levita
- Department of Pharmacology and Clinical Pharmacy, Faculty of Pharmacy, Universitas Padjadjaran, Sumedang, Indonesia
| | - Supat Jiranusornkul
- Department of Pharmaceutical Science, Faculty of Pharmacy, Chiang Mai University, Chiang Mai, Thailand
| | - Binh Phu Nguyen
- School of Mathematics and Statistics, Victoria University of Wellington, Wellington, New Zealand
| |
Collapse
|
28
|
Liao J, Chen H, Liao Y, Luo C, Wang Z, Zhang F, Fu C. Neuroprotective effects of hirudin against cerebral ischemia-reperfusion injury via inhibition of CCL2-mediated ferroptosis and inflammatory pathways. Brain Res Bull 2025; 224:111293. [PMID: 40064243 DOI: 10.1016/j.brainresbull.2025.111293] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/24/2025] [Accepted: 03/05/2025] [Indexed: 03/27/2025]
Abstract
Cerebral ischemia-reperfusion injury (CIRI) is a leading cause of neurological impairment in stroke, primarily correlated to oxidative stress, inflammation, and ferroptosis. This study investigates the neuroprotective effects of hirudin on CIRI, focusing on its role in modulating neuronal survival, oxidative stress, and ferroptosis markers through inhibition of CCL2. A middle cerebral artery occlusion (MCAO) model in mice and an oxygen-glucose deprivation/reoxygenation (OGD/R) model in HT22 cells were used to simulate ischemic conditions. Hirudin significantly improved neurological function and reduced cerebral edema and infarct size in the MCAO model. In vitro, hirudin enhanced neuronal viability and reduced apoptosis in OGD/R-stimulated cells. Integrative network pharmacology and transcriptomic analysis identified CCL2 as a potential target of hirudin. Hirudin treatment suppressed CCL2 expression, which in turn reduced the TLR4/NF-κB signaling activation, thereby mitigating ferroptosis and inflammatory responses in ischemic neurons. Overexpression of CCL2 partially reversed these protective effects, underscoring its role in ischemic injury. These findings suggest that hirudin alleviates CIRI by modulating CCL2 and preventing ferroptosis, offering insights into its potential as a therapeutic agent for ischemic conditions.
Collapse
Affiliation(s)
- Junbao Liao
- Department of Cerebrovascular Disease, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, PR China
| | - Huahui Chen
- Department of Cerebrovascular Disease, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, PR China
| | - Yiwei Liao
- Department of Neurosurgery, Xiangya Hospital, Central South University, Changsha, Hunan 410008, PR China
| | - Can Luo
- Department of Cerebrovascular Disease, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, PR China
| | - Zhi Wang
- Department of Cerebrovascular Disease, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, PR China
| | - Fan Zhang
- Department of Cerebrovascular Disease, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, PR China
| | - Chuanyi Fu
- Department of Cerebrovascular Disease, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou, Hainan 570311, PR China.
| |
Collapse
|
29
|
Dong J, Jin Z, Li C, Yang J, Jiang Y, Li Z, Chen C, Zhang B, Ye Z, Hu Y, Ma J, Li P, Li Y, Wang D, Ji Z. Machine Learning Models With Prognostic Implications for Predicting Gastrointestinal Bleeding After Coronary Artery Bypass Grafting and Guiding Personalized Medicine: Multicenter Cohort Study. J Med Internet Res 2025; 27:e68509. [PMID: 40053791 PMCID: PMC11926454 DOI: 10.2196/68509] [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: 11/07/2024] [Revised: 02/04/2025] [Accepted: 02/13/2025] [Indexed: 03/09/2025] Open
Abstract
BACKGROUND Gastrointestinal bleeding is a serious adverse event of coronary artery bypass grafting and lacks tailored risk assessment tools for personalized prevention. OBJECTIVE This study aims to develop and validate predictive models to assess the risk of gastrointestinal bleeding after coronary artery bypass grafting (GIBCG) and to guide personalized prevention. METHODS Participants were recruited from 4 medical centers, including a prospective cohort and the Medical Information Mart for Intensive Care IV (MIMIC-IV) database. From an initial cohort of 18,938 patients, 16,440 were included in the final analysis after applying the exclusion criteria. Thirty combinations of machine learning algorithms were compared, and the optimal model was selected based on integrated performance metrics, including the area under the receiver operating characteristic curve (AUROC) and the Brier score. This model was then developed into a web-based risk prediction calculator. The Shapley Additive Explanations method was used to provide both global and local explanations for the predictions. RESULTS The model was developed using data from 3 centers and a prospective cohort (n=13,399) and validated on the Drum Tower cohort (n=2745) and the MIMIC cohort (n=296). The optimal model, based on 15 easily accessible admission features, demonstrated an AUROC of 0.8482 (95% CI 0.8328-0.8618) in the derivation cohort. In external validation, the AUROC was 0.8513 (95% CI 0.8221-0.8782) for the Drum Tower cohort and 0.7811 (95% CI 0.7275-0.8343) for the MIMIC cohort. The analysis indicated that high-risk patients identified by the model had a significantly increased mortality risk (odds ratio 2.98, 95% CI 1.784-4.978; P<.001). For these high-risk populations, preoperative use of proton pump inhibitors was an independent protective factor against the occurrence of GIBCG. By contrast, dual antiplatelet therapy and oral anticoagulants were identified as independent risk factors. However, in low-risk populations, the use of proton pump inhibitors (χ21=0.13, P=.72), dual antiplatelet therapy (χ21=0.38, P=.54), and oral anticoagulants (χ21=0.15, P=.69) were not significantly associated with the occurrence of GIBCG. CONCLUSIONS Our machine learning model accurately identified patients at high risk of GIBCG, who had a poor prognosis. This approach can aid in early risk stratification and personalized prevention. TRIAL REGISTRATION Chinese Clinical Registry Center ChiCTR2400086050; http://www.chictr.org.cn/showproj.html?proj=226129.
Collapse
Affiliation(s)
- Jiale Dong
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Department of Acute Abdomen Surgery, Beijing ChaoYang Hospital, Capital Medical University, Beijing, China
- Department of General Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Zhechuan Jin
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Chengxiang Li
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Jian Yang
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Yi Jiang
- Department of Cardiovascular Surgery, Nanjing Drum Tower Hospital, Chinese Academy of Medical Science & Peking Union Medical College, Nanjing, China
| | - Zeqian Li
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Cheng Chen
- Department of Cardiovascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Beijing, China
| | - Bo Zhang
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Department of General Surgery, Beijing Luhe Hospital, Capital Medical University, Beijing, China
| | - Zhaofei Ye
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yang Hu
- Department of General Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Jianguo Ma
- School of Instrumentation and Optoelectronic Engineering, Beihang University, Beijing, China
| | - Ping Li
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Yulin Li
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Dongjin Wang
- Department of Cardiovascular Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Beijing, China
| | - Zhili Ji
- Beijing Institute of Heart, Lung and Blood Vessel Diseases, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
- Department of Acute Abdomen Surgery, Beijing ChaoYang Hospital, Capital Medical University, Beijing, China
- Department of General Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
- Department of Hepatobiliary and Pancreaticosplenic Surgery, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| |
Collapse
|
30
|
Chen W, Huang Y, Li W, Fan G, Tang Y, Zhao W, Chen K, Chen Z, Zhou K, Li Z, Zhang H. The potential of pomegranate peel supplementation in Yellow-feathered broilers: effects on growth performance, serum biochemistry, antioxidant capacity, intestinal health, intestinal microbiota, and duodenal mucosal metabolites. Poult Sci 2025; 104:104983. [PMID: 40058007 PMCID: PMC11930591 DOI: 10.1016/j.psj.2025.104983] [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: 01/11/2025] [Revised: 02/24/2025] [Accepted: 03/03/2025] [Indexed: 03/28/2025] Open
Abstract
This study aimed to investigate the effects of dietary supplementation with pomegranate peel powder (PP) on the growth performance, serum biochemistry, antioxidant capacity, intestinal microbiota, and duodenal mucosal metabolites of yellow-feathered broilers. A total of 360 yellow-feathered broilers were randomly divided into three groups, with their diets supplemented with different levels of PP (0, 1, and 4 g/kg) for 42 days. Dietary supplementation with PP significantly increased the average body weight and average daily gain of yellow-feathered broilers during the periods of 1-21 and 22-42 days, while reducing the feed conversion ratio (p < 0.05). It also decreased the serum levels of aspartate aminotransferase, alanine aminotransferase, creatinine, and uric acid, increased the activities of glutathione peroxidase and superoxide dismutase, and reduced malondialdehyde content in the serum, liver, and intestinal mucosa (p < 0.05). Furthermore, PP supplementation promoted the mRNA expression of farnesoid X receptor, peroxisome proliferator-activated receptor alpha, fatty acid-binding protein 4, epidermal growth factor/epidermal growth factor receptor, and B-cell lymphoma 2, while decreasing the mRNA expression of caspase-1 and interleukin-1 beta (p < 0.05). Regarding mucosal metabolites, PP supplementation increased the contents of polyunsaturated fatty acids (cis-11-eicosenoic acid, cis-13,16-docosadienoic acid, and cis-11,14-eicosadienoic acid), prostaglandin E2/G2, and secondary bile acids (apocholic, hyodeoxycholic, 7-ketodeoxycholic, and omega-muricholic acids) in the mucosa (p < 0.05). In terms of cecal microbiota, PP supplementation increased the β-diversity index (p < 0.05), elevated the relative abundances of Bacteroidota, Alistipes, Bacilli, and Actinobacteriota, and reduced the relative abundances of Clostridia and Gammaproteobacteria (p < 0.05). In conclusion, dietary supplementation of PP can improve intestinal health and growth performance of yellow-feathered broilers by regulating the composition of the gut microbiota.
Collapse
Affiliation(s)
- Wang Chen
- School of Animal Science and Technology, Foshan University, No. 33 Guangyun Road, Shishan Town, Nanhai District, Foshan, Guangdong 528000, China.
| | - Yurong Huang
- School of Animal Science and Technology, Foshan University, No. 33 Guangyun Road, Shishan Town, Nanhai District, Foshan, Guangdong 528000, China.
| | - Wenlong Li
- School of Animal Science and Technology, Foshan University, No. 33 Guangyun Road, Shishan Town, Nanhai District, Foshan, Guangdong 528000, China.
| | - Gao Fan
- Wen's Food Group, No. 9, North Dongdi Road, Xincheng Town, Yunfu, Guangdong 527400, China.
| | - Yanfang Tang
- School of Animal Science and Technology, Foshan University, No. 33 Guangyun Road, Shishan Town, Nanhai District, Foshan, Guangdong 528000, China.
| | - Weiru Zhao
- School of Animal Science and Technology, Foshan University, No. 33 Guangyun Road, Shishan Town, Nanhai District, Foshan, Guangdong 528000, China.
| | - Kexin Chen
- School of Animal Science and Technology, Foshan University, No. 33 Guangyun Road, Shishan Town, Nanhai District, Foshan, Guangdong 528000, China.
| | - Zifan Chen
- School of Animal Science and Technology, Foshan University, No. 33 Guangyun Road, Shishan Town, Nanhai District, Foshan, Guangdong 528000, China.
| | - Keyue Zhou
- Wen's Food Group, No. 9, North Dongdi Road, Xincheng Town, Yunfu, Guangdong 527400, China.
| | - Zhaoyao Li
- Wen's Food Group, No. 9, North Dongdi Road, Xincheng Town, Yunfu, Guangdong 527400, China; College of Veterinary Medicine, South China Agricultural University, No. 483, Wushan Road, Tianhe District, Guangzhou, Guangdong, 510642, China.
| | - Huihua Zhang
- School of Animal Science and Technology, Foshan University, No. 33 Guangyun Road, Shishan Town, Nanhai District, Foshan, Guangdong 528000, China.
| |
Collapse
|
31
|
Cui G, Jiao X, Wang Z, Zhang Z. Association between tobacco smoke exposure and constipation among American adults: a National Health and Nutrition Examination Survey. Front Public Health 2025; 13:1502341. [PMID: 40109432 PMCID: PMC11920834 DOI: 10.3389/fpubh.2025.1502341] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Accepted: 02/13/2025] [Indexed: 03/22/2025] Open
Abstract
Background Studies have shown that there is a relationship between smoke exposure and constipation. However, there are limited data on the associations between constipation and smoking or serum cotinine levels, a metabolite of nicotine commonly used as a marker of tobacco exposure. Methods This study thoroughly examined the cross-sectional data from the National Health and Nutrition Examination Survey (NHANES) from 2005 to 2010. We applied multivariate logistic regression models to assess the association between tobacco smoking status, serum cotinine levels, and constipation. Results In this cross-sectional study, 11,651 participants were included. The average age of the participants was 48.8 ± 17.9 years. After controlling for covariates, there was no significant relationship between serum cotinine levels, smoking status, and constipation risk. According to the categorical analysis, the incidence of constipation was 36% higher in participants with serum cotinine levels between 0.05 and 2.99 ng/mL (model 1: OR = 1.45 [1.13-1.85]; model 2: OR = 1.44 [1.12-1.83]; and model 3: OR = 1.36 [1.06-1.74]; p < 0.05) after adjusting for covariates by using the lowest levels of serum cotinine (<0.05 ng/mL) as a reference. In the non-smokers, serum cotinine levels were linearly and positively associated with the risk of constipation (p > 0.05), and the relationship between smoke exposure and constipation remained relatively stable in across all subgroups. Conclusion The study suggests that serum cotinine levels have a promoting effect on stool frequency-related constipation in non-smokers. Therefore, avoiding passive smoking as much as possible may reduce the effect of smoke exposure on constipation and serve as a preventive measure for treatment.
Collapse
Affiliation(s)
- Guoce Cui
- Department of Traditional Chinese Medicine Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Xia Jiao
- Department of Traditional Chinese Medicine Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Zhenbiao Wang
- Department of Traditional Chinese Medicine Surgery, Beijing Shijitan Hospital, Capital Medical University, Beijing, China
| | - Zhuhui Zhang
- Department of Anorectology, Guang'anmen Hospital, China Academy of Traditional Chinese Medicine, Beijing, China
| |
Collapse
|
32
|
Yuan J, Su J, Zhong S, Yuan X, Zhu J, Lu S, Zhang D, Li G, Xue H, Yan M, Yue L, Zhang T. Dictamnine alleviates DSS-induced colitis mice by inhibiting ferroptosis of enterocytes via activating Nrf2-Gpx4 signaling pathway. Eur J Pharmacol 2025; 997:177464. [PMID: 40049578 DOI: 10.1016/j.ejphar.2025.177464] [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: 11/11/2024] [Revised: 02/22/2025] [Accepted: 03/04/2025] [Indexed: 03/12/2025]
Abstract
BACKGROUND The treatment of ulcerative colitis (UC) remains a huge challenge worldwide. Dictamnine is a natural product derived from Dictamnus dasycarpus Turcz. root bark and possesses multi-pharmacological properties, including anti-inflammation effects. However, its protective effect on UC and its underlying mechanisms are unknown. PURPOSE Here we explored the protective effect and underlying mechanism of dictamnine against dextran sulfate sodium (DSS)-induced colitis in mice. METHODS The experimental colitis was established by adding 3% DSS on drinking water of mice and the effects of dictamnine (10, 20, 40 mg/kg, p.o, once a day by 10 days) in colon tissues was analyzed. NCM460 cell was induced by RSL3 to detect the effect of dictamnine on ferroptosis and the underlying mechanism. Pathological damage was determined by H&E. Indicators related to intestinal permeability were detected by FITC and immunofluorescence. Cytokines levels (TNF-α、IL-1β and IL-6), antioxidant enzymes activities (MDA and GSH), the level of Fe2+ Cytokines levels and Gpx4 activity were detected by ELISA. Finally, the activation of nuclear factor erythroid 2-like 2 (Nrf2) was detected to explore the mechanism. RESULTS The results indicated that dictamnine significantly attenuated DSS-induced colon pathological damage, intestinal barrier, cytokines levels, and increased the antioxidant enzymes activities. Moreover, dictamnine attenuated ferroptosis in DSS-induced colon injury and upregulated Gpx4 expression in DSS-induced mice. Mechanistic experiments revealed that dictamnine activated Nrf2 in mice. CONCLUSION Taken together, this study evaluates that dictamnine alleviates DSS-induced colitis mice by inhibiting ferroptosis of enterocytes and its protective effects are associated with activating the Nrf2-Gpx4 signaling pathway.
Collapse
Affiliation(s)
- Jin Yuan
- Puer Hospital of Traditional Chinese Medicine, Puer, Yunnan, China
| | - Junwei Su
- Department of Anesthesiology, Sir Run Run Shaw Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Shaowen Zhong
- School of Pharmaceutical Sciences, Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, 510006, China; Adverse Drug Reaction Monitoring Center, Zhongshan Food and Drug Inspection Institute, Zhongshan, Guangdong, China
| | - Xin Yuan
- Department of Pharmacy, The Second Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou, Guangdong, China
| | - Jianping Zhu
- Puer Hospital of Traditional Chinese Medicine, Puer, Yunnan, China
| | - Shuangxi Lu
- Puer Hospital of Traditional Chinese Medicine, Puer, Yunnan, China
| | - Di Zhang
- Puer Hospital of Traditional Chinese Medicine, Puer, Yunnan, China
| | - Guiling Li
- Puer Hospital of Traditional Chinese Medicine, Puer, Yunnan, China
| | - Hanyu Xue
- Puer Hospital of Traditional Chinese Medicine, Puer, Yunnan, China.
| | - Min Yan
- Department of Pathogen Biology and Immunology, Faculty of Basic Medical Science, Kunming Medical University, Kunming, Yunnan, China.
| | - Lei Yue
- Institute of Medical Biology, Chinese Academy of Medical Sciences and Peking Union Medical College, Kunming, Yunnan, China.
| | - Tianwu Zhang
- Puer Hospital of Traditional Chinese Medicine, Puer, Yunnan, China.
| |
Collapse
|
33
|
Pan D, Sun Y, Zhang J, Zeng J, Yu S, Zhao D, Dong Z, Liu M, Liu S, Wang W, Wang S. An active protein from Dendrobium officinale residue: Protects the gastric mucosa and stabilized in the gastrointestinal tract. Int J Biol Macromol 2025; 294:139387. [PMID: 39753178 DOI: 10.1016/j.ijbiomac.2024.139387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2024] [Revised: 12/17/2024] [Accepted: 12/29/2024] [Indexed: 01/11/2025]
Abstract
A large number of by-products generated in the food industry is discarded as waste, especially the residue left after extracting plant resources, which is typically repurposed as fertilizer. In this study, we extracted and purified a new protein, DOP1, from the residue of Dendrobium officinale Kimura & Migo (D. officinale), and explored the protective effect of DOP1 on alcohol-induced gastric mucosal injury. Its amino acid composition and the stability of secondary structure were measured by amino acid analysis and various spectroscopic methods. The microscopic morphology of DOP1 was observed by electron microscopy and its particle distribution was determined to be in the range of 3.7-5.5 nm. In addition, DOP1 was found to exhibit excellent protective activity against alcohol-induced gastric mucosal injury, as well as anti-inflammatory and antioxidant activities by in vitro and in vivo activity assays, which may be related to the upregulation of TFF2 through the activation of PPARγ. Intriguingly, DOP1 does not degrade and remains active after being digested by the gastrointestinal tract. From an economic and sustainable perspective, the discovery of DOP1 will provide new opportunities for the enhanced utilization and sustainable development of D. officinale resources.
Collapse
Affiliation(s)
- Daian Pan
- Changchun University of Chinese Medicine, Key Laboratory of Ginseng Efficacy Substance Base and Biological Mechanism Research, Ministry of Education, Changchun 130117, China; Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China; Research Center of Traditional Chinese Medicine, The Affiliated Hospital to Changchun University of Chinese Medicine, Changchun 130021, China
| | - Yanling Sun
- Changchun University of Chinese Medicine, Key Laboratory of Ginseng Efficacy Substance Base and Biological Mechanism Research, Ministry of Education, Changchun 130117, China; Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Jiayi Zhang
- Changchun University of Chinese Medicine, Key Laboratory of Ginseng Efficacy Substance Base and Biological Mechanism Research, Ministry of Education, Changchun 130117, China; Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Jing Zeng
- Changchun University of Chinese Medicine, Key Laboratory of Ginseng Efficacy Substance Base and Biological Mechanism Research, Ministry of Education, Changchun 130117, China; Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Shiting Yu
- Changchun University of Chinese Medicine, Key Laboratory of Ginseng Efficacy Substance Base and Biological Mechanism Research, Ministry of Education, Changchun 130117, China; Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Daqing Zhao
- Changchun University of Chinese Medicine, Key Laboratory of Ginseng Efficacy Substance Base and Biological Mechanism Research, Ministry of Education, Changchun 130117, China; Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Zhengqi Dong
- State Key Laboratory for Quality Ensurance and Sustainable Use of Dao-di Herbs, Chinese Academy of Medical Sciences & Peking Union Medical College, Institute of Medicinal Plant Development, Beijing 100193, China
| | - Meichen Liu
- Changchun University of Chinese Medicine, Key Laboratory of Ginseng Efficacy Substance Base and Biological Mechanism Research, Ministry of Education, Changchun 130117, China; Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China
| | - Shichao Liu
- Changchun University of Chinese Medicine, Key Laboratory of Ginseng Efficacy Substance Base and Biological Mechanism Research, Ministry of Education, Changchun 130117, China; College of Pharmacy, Changchun University of Chinese Medicine, Changchun 130117, China.
| | - Weinan Wang
- Guangdong Provincial Key Laboratory of Natural Drugs Research and Development, The Affiliated Dongguan Songshan Lake Central Hospital, Guangdong Medical University, Dongguan 523808, China; Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China.
| | - Siming Wang
- Changchun University of Chinese Medicine, Key Laboratory of Ginseng Efficacy Substance Base and Biological Mechanism Research, Ministry of Education, Changchun 130117, China; Northeast Asia Research Institute of Traditional Chinese Medicine, Changchun University of Chinese Medicine, Changchun 130117, China.
| |
Collapse
|
34
|
Mitchell TM, Burdick Sanchez NC, Carroll JA, Broadway PR, Legako JF, Bowen BM, Petry AL. Prenatal lipopolysaccharide stimulation modulates gastrointestinal immunity and oxidative status in weaned pigs. Am J Physiol Gastrointest Liver Physiol 2025; 328:G197-G205. [PMID: 39853237 DOI: 10.1152/ajpgi.00268.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/21/2024] [Accepted: 01/09/2025] [Indexed: 01/26/2025]
Abstract
Gastrointestinal immunity and antioxidant defenses may be bolstered in young animals through prenatal immune stimulation (PIS), but this is largely uninvestigated in swine. This study tested the hypothesis that PIS could regulate offspring's gastrointestinal immune response and oxidative stress profile. To this end, a PIS model was utilized in sows, delivering low-dose lipopolysaccharide (LPS) during the final third of gestation to target the developing immune system. On day 78 ± 1.8 of gestation, 14 Camborough sows (parity = 2.6 ± 1.4) received either saline (Control, CON) or LPS from Escherichia coli O111:B4 (2.5 µg/kg of body wt). A subset of 34 weaned barrows (n = 17 CON, PIS), weaned at 21 ± 1.3 days, were anesthetized for subcutaneous temperature loggers and jugular catheter placement. Following recovery, all pigs received an intravenous injection of LPS (10 µg/kg·body wt) from E. coli O111:B4. Our findings demonstrate that PIS enhances the gut immune response by upregulating key inflammatory cytokines, indicative of a proinflammatory profile. Consistently across the jejunum and ileum, stem cell factor was modulated with heightened expression in PIS than CON (P ≤ 0.05). In the ileum alone, PIS exhibited heightened expression of proinflammatory cytokines and chemokines, including TNFα, IL-6, IL-1β, and CCL3L1, compared with CON (P ≤ 0.05). Exposure to PIS resulted in reduced systemic total antioxidant capacity at hours 2 and 4 postchallenge (P = 0.004). Piglets exposed to PIS had decreased jejunal tissue malondialdehyde concentrations (P = 0.049). Together, these data indicate that exposure to PIS alters the inflammatory profile of the gastrointestinal immune response and oxidative status in weaned pigs.NEW & NOTEWORTHY These studies represent novel investigations into the influence of prenatal immune stimulation (PIS) in swine on the gastrointestinal immune response and oxidative status of offspring following subsequent immune challenge. Notable alterations were observed in gut protein biomarkers, particularly the upregulation of proinflammatory cytokines TNFα, IL-6, and IL-1β in PIS-exposed pigs, but has variable effects on oxidative status. Altered intestinal immune development may contribute to an increased risk for inflammatory disease associated with prenatal immune stimulation.
Collapse
Affiliation(s)
- Ty M Mitchell
- Department of Large Animal Clinical Sciences, College of Veterinary Medicine, Michigan State University, East Lansing, Michigan, United States
| | - Nicole C Burdick Sanchez
- Livestock Issues Research Unit, Agriculture Research Service, United States Department of Agriculture, Lubbock, Texas, United States
| | - Jeff A Carroll
- Livestock Issues Research Unit, Agriculture Research Service, United States Department of Agriculture, Lubbock, Texas, United States
| | - Paul R Broadway
- Livestock Issues Research Unit, Agriculture Research Service, United States Department of Agriculture, Lubbock, Texas, United States
| | - Jerrad F Legako
- Department of Animal and Food Sciences, Texas Tech University, Lubbock, Texas, United States
| | - Brooke M Bowen
- Department of Animal and Food Sciences, Texas Tech University, Lubbock, Texas, United States
| | - Amy L Petry
- Division of Animal Sciences, University of Missouri, Columbia, Missouri, United States
| |
Collapse
|
35
|
Yu Y, Zheng Z, Gao X, Gu Y, Zhang M, Hu B, Gao Q, Li Z, Chen Y, Li Q, Shen F, Zhu M, Hang D, Zhan Q, Wang L, Shen C, Lu X, Gu D, Ma H, Shen H, Jin G, Yan C. Plasma Metabolomic Signatures of H. pylori Infection, Alcohol Drinking, Smoking, and Risk of Gastric Cancer. Mol Carcinog 2025; 64:463-474. [PMID: 39630052 DOI: 10.1002/mc.23851] [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: 07/26/2024] [Revised: 10/25/2024] [Accepted: 11/05/2024] [Indexed: 02/13/2025]
Abstract
Circulating metabolic profiles have shown promising potential in identifying high-risk populations for various diseases, while metabolic perturbation plays an important role in gastric cancer. In this study, we conducted a cross-sectional study with 1800 participants to identify plasma metabolite signatures associated with environmental risk factors of gastric cancer. Subsequently, we evaluated the association between these signatures and gastric cancer risk in a nested case-control study involving 326 gastric cancer cases and 326 matched cancer-free controls. We conducted mediation analyses to elucidate the potential impact of metabolites on the association between environmental factors and gastric cancer. In the cross-sectional study, we identified 46 metabolites associated with Helicobacter pylori (H. pylori) infection, 365 with alcohol drinking, and 154 with smoking status. In the nested case-control study, 60 plasma metabolites, comprising 30 lipids, 15 amino acids, 6 xenobiotics, 3 nucleotides, 2 cofactors and vitamins, 2 carbohydrate, 1 energy, and 1 peptide, were associated with gastric cancer risk. A one-standard deviation increment in the H. pylori infection-related metabolomic signature was associated with an increased risk of gastric cancer (OR = 1.66, 95% CI: 1.32-2.09, p = 1.62 × 10-5). Furthermore, the effect of H. pylori infection on gastric cancer was partially mediated by the metabolomic signature (23.28%, 95% CI: 0.09-0.56) or adenine (13.69%, 95% CI: 0.05-0.31). In conclusion, we have identified metabolites associated with environmental factors and demonstrated the association between the H. pylori infection signature and gastric cancer risk. The findings provide novel insights into characterizing high-risk population for gastric cancer.
Collapse
Affiliation(s)
- Yuhui Yu
- Department of Epidemiology, State Key Laboratory Cultivation Base of Biomarkers for Cancer Precision Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Zhonghua Zheng
- Department of Epidemiology, State Key Laboratory Cultivation Base of Biomarkers for Cancer Precision Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xinxiang Gao
- Department of Epidemiology, State Key Laboratory Cultivation Base of Biomarkers for Cancer Precision Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yuanliang Gu
- Department of Epidemiology, State Key Laboratory Cultivation Base of Biomarkers for Cancer Precision Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Min Zhang
- Department of Epidemiology, State Key Laboratory Cultivation Base of Biomarkers for Cancer Precision Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Beiping Hu
- Department of Epidemiology, State Key Laboratory Cultivation Base of Biomarkers for Cancer Precision Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qian Gao
- Department of Epidemiology, State Key Laboratory Cultivation Base of Biomarkers for Cancer Precision Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Zhe Li
- Department of Epidemiology, State Key Laboratory Cultivation Base of Biomarkers for Cancer Precision Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Yan Chen
- Department of Epidemiology, State Key Laboratory Cultivation Base of Biomarkers for Cancer Precision Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qian Li
- Department of Epidemiology, State Key Laboratory Cultivation Base of Biomarkers for Cancer Precision Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Fang Shen
- Department of Epidemiology, State Key Laboratory Cultivation Base of Biomarkers for Cancer Precision Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Meng Zhu
- Department of Epidemiology, State Key Laboratory Cultivation Base of Biomarkers for Cancer Precision Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Chronic Non-Communicable Disease Control, Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, China
| | - Dong Hang
- Department of Epidemiology, State Key Laboratory Cultivation Base of Biomarkers for Cancer Precision Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Qiang Zhan
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| | - Lu Wang
- Department of Chronic Non-Communicable Disease Control, Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, China
| | - Chong Shen
- Department of Epidemiology, State Key Laboratory Cultivation Base of Biomarkers for Cancer Precision Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Xiangfeng Lu
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Cardiovascular Epidemiology, Chinese Academy of Medical Sciences, Beijing, China
- Research Units of Cohort Study on Cardiovascular Diseases and Cancers, Chinese Academy of Medical Sciences, Beijing, China
| | - Dongfeng Gu
- Department of Epidemiology, Fuwai Hospital, National Center for Cardiovascular Diseases, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
- Key Laboratory of Cardiovascular Epidemiology, Chinese Academy of Medical Sciences, Beijing, China
- Research Units of Cohort Study on Cardiovascular Diseases and Cancers, Chinese Academy of Medical Sciences, Beijing, China
| | - Hongxia Ma
- Department of Epidemiology, State Key Laboratory Cultivation Base of Biomarkers for Cancer Precision Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
| | - Hongbing Shen
- Department of Epidemiology, State Key Laboratory Cultivation Base of Biomarkers for Cancer Precision Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
- Research Units of Cohort Study on Cardiovascular Diseases and Cancers, Chinese Academy of Medical Sciences, Beijing, China
| | - Guangfu Jin
- Department of Epidemiology, State Key Laboratory Cultivation Base of Biomarkers for Cancer Precision Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Chronic Non-Communicable Disease Control, Affiliated Wuxi Center for Disease Control and Prevention of Nanjing Medical University, Wuxi Center for Disease Control and Prevention, Wuxi, China
| | - Caiwang Yan
- Department of Epidemiology, State Key Laboratory Cultivation Base of Biomarkers for Cancer Precision Prevention and Treatment, Collaborative Innovation Center for Cancer Personalized Medicine, School of Public Health, Nanjing Medical University, Nanjing, China
- Department of Gastroenterology, The Affiliated Wuxi People's Hospital of Nanjing Medical University, Wuxi Medical Center, Nanjing Medical University, Wuxi, China
| |
Collapse
|
36
|
Xia Y, Li X, Huang F, Wu Y, Liu J, Liu J. Design and advances in antioxidant hydrogels for ROS-induced oxidative disease. Acta Biomater 2025; 194:80-97. [PMID: 39900274 DOI: 10.1016/j.actbio.2025.01.057] [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: 09/30/2024] [Revised: 01/14/2025] [Accepted: 01/29/2025] [Indexed: 02/05/2025]
Abstract
Reactive oxygen species (ROS) play a crucial role in human physiological processes, but oxidative stress caused by excessive ROS may lead to a variety of acute and chronic diseases. Despite the development of various strategies and biomaterials, an efficiently and broadly applied method for treatment of ROS-induced oxidative disease remains a bottleneck. Aiming to improve the local oxidative stress environment, numerous bioactive hydrogels with antioxidant properties have emerged and are proven to quickly and continuously eliminate excessive ROS. To deeply understand the design principles and applications of antioxidant hydrogels is highly beneficial for designing antioxidant hydrogels for treatment of oxidative disease. This review provides a detailed summary of recent advances in design and applications of antioxidant hydrogels for various ROS-induced oxidative diseases. In this review, the kinds of antioxidant components in antioxidant hydrogels are outlined in detail. Additionally, the crosslinking methods and the biomedical applications of antioxidant hydrogels are widely summarized and discussed, especially focusing on their usage in different types of diseases and the attention given to the treatment of diseases such as skin wounds, myocardial infarction, and osteoarthritis. Finally, the future development direction of antioxidant hydrogel is further proposed. STATEMENT OF SIGNIFICANCE: Oxidative stress is a pivotal biochemical process that plays a critical role in cellular homeostasis. Excessive cellular oxidative stress triggers an inflammatory response, which is implicated in a spectrum of associated diseases. Given the critical need for managing oxidative stress, antioxidant therapies have become a vital focus in medical research. Hydrogels have garnered substantial interest among biomaterial scientists due to their hydrophilic nature and biocompatibility. The review delves into the realm of antioxidant hydrogels, encompassing the classification of antioxidant components, the synthesis and fabrication of hydrogels, and a comprehensive overview of the biological applications and challenges of these antioxidant hydrogels. Aiming to provide new perspectives for researchers in developing cutting-edge therapeutic approaches that leverage antioxidant hydrogels.
Collapse
Affiliation(s)
- Yi Xia
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Xinyi Li
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Fan Huang
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China
| | - Yuanhao Wu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China.
| | - Jinjian Liu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China.
| | - Jianfeng Liu
- State Key Laboratory of Advanced Medical Materials and Devices, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Key Laboratory of Radiopharmacokinetics for Innovative Drugs, Tianjin Institutes of Health Science, Institute of Radiation Medicine, Chinese Academy of Medical Sciences & Peking Union Medical College, Tianjin 300192, PR China.
| |
Collapse
|
37
|
Liu D, Wei M, Fang Y, Yuan T, Sun Y, Xie H, Yan W, Yuan B, Zhuang B, Jin Y. Oral colon-retentive inulin gels protect against radiation-induced hematopoietic and gastrointestinal injury by improving gut homeostasis. Int J Biol Macromol 2025; 292:139199. [PMID: 39730057 DOI: 10.1016/j.ijbiomac.2024.139199] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2024] [Revised: 12/19/2024] [Accepted: 12/23/2024] [Indexed: 12/29/2024]
Abstract
Ionizing radiation-induced injury often occurs in nuclear accidents or large-dose radiotherapy, leading to acute radiation syndromes characterized by hematopoietic and gastrointestinal injuries even to death. However, current radioprotective drugs are only used in hospitals with unavoidable side effects. Here, we heated the aqueous solution of inulin, a polysaccharide dietary fiber, forming colon-retentive gel as a radiation protector in radiotherapy. Mouse models were established after 60Co γ-ray irradiation of the total body or abdomen. Inulin gels were orally administered to the mice every day from 3 days pre-radiation to 3 days post-radiation. The hematopoietic system was well protected with good blood cell recovery and cell proliferation in the femur and spleen. Oral inulin gels increased the relative abundances of key commensal microorganisms including f_Lachnospiraceae, Akkermansia, Blautia, and short-chain fatty acid metabolites. The secretion of the anti-inflammation cytokines IL-22 and IL-10 in the intestinal cells also increased. Similarly, the expression of the tight junction proteins claudin-1 and occludin in the gut mucosa was affected. In an orthotopic murine colorectal cancer model, oral inulin gels followed by 10-Gy abdomen radiation improved the radiotherapy efficiency with low attenuated radiation injury. Taken the data together, these results suggest that oral inulin gels are a bioactive material against ionizing radiation-induced injury.
Collapse
Affiliation(s)
- Dongdong Liu
- Beijing Institute of Radiation Medicine, Beijing 100850, China; China Rehabilitation Science Institute, China Rehabilitation Research Center, Beijing Key Laboratory of Neural Injury and Rehabilitation, Beijing 100068, China
| | - Meng Wei
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yubao Fang
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Tianyu Yuan
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Yingbao Sun
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Hua Xie
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Wenrui Yan
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Bochuan Yuan
- Beijing Institute of Radiation Medicine, Beijing 100850, China
| | - Bo Zhuang
- Institute of NBC Defense, Beijing 102205, China.
| | - Yiguang Jin
- Beijing Institute of Radiation Medicine, Beijing 100850, China.
| |
Collapse
|
38
|
Bu X, Wang L. Iron metabolism and the tumor microenvironment: A new perspective on cancer intervention and therapy (Review). Int J Mol Med 2025; 55:39. [PMID: 39749705 PMCID: PMC11722052 DOI: 10.3892/ijmm.2024.5480] [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: 09/09/2024] [Accepted: 10/29/2024] [Indexed: 01/04/2025] Open
Abstract
Iron metabolism plays a crucial role in the tumor microenvironment, influencing various aspects of cancer cell biology and tumor progression. This review discusses the regulatory mechanisms of iron metabolism within the tumor microenvironment and highlights how tumor cells and associated stromal cells manage iron uptake, accumulation and regulation. The sources of iron within tumors and the biological importance of ferroptosis in cancer were explored, focusing on its mechanisms, biological effects and, in particular, its tumor‑suppressive properties. Furthermore, the protective strategies employed by cancer cells to evade ferroptosis were examined. This review also delves into the intricate relationship between iron metabolism and immune modulation within the tumor microenvironment, detailing the impact on tumor‑associated immune cells and immune evasion. The interplay between ferroptosis and immunotherapy is discussed and potential strategies to enhance cancer immunotherapy by modulating iron metabolism are presented. Finally, the current ferroptosis‑based cancer therapeutic approaches were summarized and future directions for therapies that target iron metabolism were proposed.
Collapse
Affiliation(s)
- Xiaorui Bu
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| | - Lufang Wang
- Department of Obstetrics and Gynecology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei 430022, P.R. China
| |
Collapse
|
39
|
Mivehchi H, Eskandari-Yaghbastlo A, Pour Bahrami P, Elhami A, Faghihinia F, Nejati ST, Kazemi KS, Nabi Afjadi M. Exploring the role of oral bacteria in oral cancer: a narrative review. Discov Oncol 2025; 16:242. [PMID: 40009328 DOI: 10.1007/s12672-025-01998-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 02/19/2025] [Indexed: 02/27/2025] Open
Abstract
A growing body of research indicates that a wide range of cancer types may correlate with human microbiome components. On the other hand, little is known about the potential contribution of the oral microbiota to oral cancer. However, some oral microbiome components can stimulate different tumorigenic processes associated with the development of cancer. In this line, two prevalent oral infections, Porphyromonas gingivalis, and Fusobacterium nucleatum can increase tumor growth. The microbiome can impact the course of the illness through direct interactions with the human body and major modifications to the toxicity and responsiveness to different kinds of cancer therapy. Recent research has demonstrated a relationship between specific phylogenetic groupings and the results of immunotherapy treatment for particular tumor types. Conversely, there has been a recent upsurge in interest in the possibility of using microbes to treat cancer. At the moment, some species, such as Salmonella typhimurium and Clostridium spp., are being explored as possible cancer treatment vectors. Thus, understanding these microbial interactions highlights the importance of maintaining a healthy oral microbiome in preventing oral cancers. From this perspective, this review will discuss the role of the microbiome on oral cancers and their possible application in oral cancer treatment/improvement.
Collapse
Affiliation(s)
- Hassan Mivehchi
- Faculty of Dentistry, University of Debrecen, Debrecen, Hungary
| | | | | | - Anis Elhami
- Faculty of Dentistry, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Farbod Faghihinia
- School of Dentistry, Yasuj University of Medical Sciences, Yasuj, Iran
| | | | - Kimia Sadat Kazemi
- Faculty of Dentistry, Shahid Beheshti University of Medical Sciences, Tehran, Iran.
| | - Mohsen Nabi Afjadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| |
Collapse
|
40
|
Lim JS, Li X, Lee DY, Yao L, Yoo G, Kim Y, Eum SM, Cho YC, Yoon S, Park SJ. Antioxidant and Anti-Inflammatory Activities of Methanol Extract of Senna septemtrionalis (Viv.) H.S. Irwin & Barneby Through Nrf2/HO-1-Mediated Inhibition of NF-κB Signaling in LPS-Stimulated Mouse Microglial Cells. Int J Mol Sci 2025; 26:1932. [PMID: 40076558 PMCID: PMC11900505 DOI: 10.3390/ijms26051932] [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: 12/24/2024] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 03/14/2025] Open
Abstract
Botanical extracts are recognized in traditional medicine for their therapeutic potential and safety standards. Botanical extracts are viable and sustainable alternatives to synthetic drugs, being essential in drug discovery for various diseases. Senna septemtrionalis (Viv.) H.S. Irwin & Barneby is a medical plant traditionally used to treat inflammation. However, its antioxidant and anti-inflammatory properties and the molecular pathways activated in microglial cells require further investigation. Therefore, this study examines the antioxidant and anti-inflammatory properties of Senna septemtrionalis (Viv.) H.S. Irwin & Barneby methanol extracts (SMEs) in lipopolysaccharide (LPS)-stimulated mouse microglial cells. SMEs significantly inhibit LPS-induced nitric oxide (NO) and proinflammatory cytokine production, which are mediated through the dephosphorylation of mitogen-activated protein kinases and inhibition of nuclear factor kappa B (NF-κB) translocation into the nucleus. Additionally, SME treatment upregulated the expression of nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase (HO)-1, reducing oxidative stress, indicated by a decrease in reactive oxygen species and restoration of the total glutathione content in LPS-stimulated BV2 cells. The inhibitory effects of SMEs on inflammatory mediator production and NF-κB nuclear translocation were significantly reversed by Sn-protoporphyrin, a specific HO-1 inhibitor. These findings demonstrate that SME protects microglial cells by activating the Nrf2/HO-1 pathway and inhibiting NF-κB translocation.
Collapse
Affiliation(s)
- Jae Sung Lim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, 77 Yongbong-ro, Gwangju 61186, Republic of Korea; (J.S.L.); (X.L.); (L.Y.); (Y.K.); (Y.-C.C.)
| | - Xiangying Li
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, 77 Yongbong-ro, Gwangju 61186, Republic of Korea; (J.S.L.); (X.L.); (L.Y.); (Y.K.); (Y.-C.C.)
| | - Da Young Lee
- R&D Center, CUOME BIO Co., Ltd., Sandan-gil, Hwasun-eup, Hwasun-gun 58141, Jeollanam-do, Republic of Korea;
| | - Lulu Yao
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, 77 Yongbong-ro, Gwangju 61186, Republic of Korea; (J.S.L.); (X.L.); (L.Y.); (Y.K.); (Y.-C.C.)
| | - Guijae Yoo
- Korea Food Research Institute, 245, Nongsaengmyeong-ro, Iseo-myeon, Wanju-gun 55365, Jeollabuk-do, Republic of Korea;
| | - Yunyeong Kim
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, 77 Yongbong-ro, Gwangju 61186, Republic of Korea; (J.S.L.); (X.L.); (L.Y.); (Y.K.); (Y.-C.C.)
| | - Sang Mi Eum
- International Biological Material Research Center, Korea Research Institute of Bioscience & Biotechnology, 125 Gwahak-ro, Daejeon 34141, Republic of Korea;
| | - Young-Chang Cho
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, 77 Yongbong-ro, Gwangju 61186, Republic of Korea; (J.S.L.); (X.L.); (L.Y.); (Y.K.); (Y.-C.C.)
| | - Somy Yoon
- College of Pharmacy and Research Institute of Pharmaceutical Sciences, Chonnam National University, 77 Yongbong-ro, Gwangju 61186, Republic of Korea; (J.S.L.); (X.L.); (L.Y.); (Y.K.); (Y.-C.C.)
| | - Su-Jin Park
- Functional Biomaterial Research Center, Korea Research Institute of Bioscience and Biotechnology, 181 Ipsin-gil, Jeongeup-si 56212, Republic of Korea
| |
Collapse
|
41
|
Catalano T, Selvaggi F, Cotellese R, Aceto GM. The Role of Reactive Oxygen Species in Colorectal Cancer Initiation and Progression: Perspectives on Theranostic Approaches. Cancers (Basel) 2025; 17:752. [PMID: 40075600 PMCID: PMC11899472 DOI: 10.3390/cancers17050752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Altered levels of reactive oxygen species (ROS) are recognized as one of the key factors in mediating tumor cell survival in the tissue microenvironment, where they play a role in the initiation, progression and recurrence/relapse of colorectal cancer (CRC). Tumor cells can adapt to oxidative stress (OS) using genetic or metabolic reprogramming in the long or short term. In addition, tumor cells defend themselves through positive regulation of antioxidant molecules, enhancing ROS-driven proliferation. Balanced oxidative eustress levels can influence chemotherapy resistance, allowing tumor cells to survive treatment. Secondary effects of chemotherapy include increased ROS production and redox stress, which can kill cancer cells and eliminate drug resistance. Anticancer treatments based on manipulating ROS levels could represent the gold standard in CRC therapy. Therefore, exploring the modulation of the response to OS in deregulated signaling pathways may lead to the development of new personalized CRC treatments to overcome therapy resistance. In this review, we explore the role of ROS in the initiation and progression of CRC and their diagnostic implications as biomarkers of disease. Furthermore, we focused on the involvement of ROS in different CRC therapeutic options, such as surgery, radiotherapy, theranostic imaging, chemotherapy and immunotherapy and other precision medicine approaches.
Collapse
Affiliation(s)
- Teresa Catalano
- Department of Clinical and Experimental Medicine, University of Messina, Via Consolare Valeria, 98125 Messina, Italy
| | - Federico Selvaggi
- Villa Serena Foundation for Research, 65013 Città Sant’Angelo, Italy; (F.S.); (R.C.)
| | - Roberto Cotellese
- Villa Serena Foundation for Research, 65013 Città Sant’Angelo, Italy; (F.S.); (R.C.)
| | - Gitana Maria Aceto
- Villa Serena Foundation for Research, 65013 Città Sant’Angelo, Italy; (F.S.); (R.C.)
- Department of Sciences, University “G. d’Annunzio” Chieti-Pescara, Via dei Vestini 31, 66100 Chieti, Italy
| |
Collapse
|
42
|
Liu X, Yang K, Jia Y, Yeertai Y, Wu C, Wang X, Jia Q, Gu Z, Cong J, Ling J. Chaihushugan powder regulates the gut microbiota to alleviate mitochondrial oxidative stress in the gastric tissues of rats with functional dyspepsia. Front Immunol 2025; 16:1549554. [PMID: 40040709 PMCID: PMC11876139 DOI: 10.3389/fimmu.2025.1549554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Accepted: 01/29/2025] [Indexed: 03/06/2025] Open
Abstract
Introduction Functional dyspepsia (FD) is a prevalent functional gastrointestinal disorder associated with oxidative stress (OS) and dysbiosis. Chaihushugan powder (CHSGP) demonstrates efficacy in treating FD; however, the underlying therapeutic mechanism is not yet elucidated. This study aims to investigate the effects of CHSGP on OS and gut microbiota (GM) in FD rats, with a particular emphasis on the role of GM as a potential target for the antioxidant properties of CHSGP. Methods The FD rat model was established with a modified tail-clamp stimulation and the administration of the CHSGP decoction at a dosage of 9.6 g/kg via gavage for a duration of 4 weeks. The GM was depleted by the administration of a cocktail of metronidazole (200 mg/kg), ampicillin (200 mg/kg), neomycin sulfate (200 mg/kg), and vancomycin (100 mg/kg). Fecal microbiota transplantation (FMT) was performed with CHSGP-treated fecal supernatant at a dosage of 10 mL/kg. The gastrointestinal motility was measured using the rates of gastric emptying and small intestine propulsion. Hematoxylin and eosin staining was employed to elucidate the pathological changes, while the transmission electron microscope was used to examine the microstructures of the interstitial cells of Cajal (ICC). Chemiluminescence, colorimetric assay, immunofluorescence co-staining, and western blot assay were employed to identify the OS-related markers (ROS, SOD, NOX4, PRDX1, and TRX2). Sequencing of fecal microbiota was performed utilizing 16S rDNA. Results The CHSGP decoction promoted gastrointestinal motility, protected the microstructure of ICC, and reduced OS in FD rats. The GM composition was also regulated by CHSGP. However, these effects disappeared after microbiota depletion. Fortunately, the FMT therapy reinstated them. Conclusion Chaihushugan powder decoction might regulate the GM to alleviate mitochondrial OS in the gastric tissues of FD rats.
Collapse
Affiliation(s)
- Xuejiao Liu
- Department of Gastroenterology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Keming Yang
- Department of Gastroenterology, Shenzhen Traditional Chinese Medicine Hospital, Shenzhen, Guangdong, China
| | - Yuebo Jia
- Department of Gastroenterology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yeliya Yeertai
- Department of Gastroenterology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Chenheng Wu
- Department of Gastroenterology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Xiangxiang Wang
- School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Qingling Jia
- Department of Gastroenterology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Zhijian Gu
- Department of Gastroenterology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jun Cong
- Department of Gastroenterology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Jianghong Ling
- Department of Gastroenterology, Shuguang Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
43
|
Kong W, Ding X, Wang Z, Lu L, Fan S. NVP-AUY922 relieves radiation-induced intestinal injury via regulating EPHX1. Life Sci 2025; 363:123382. [PMID: 39798648 DOI: 10.1016/j.lfs.2025.123382] [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: 11/01/2024] [Revised: 01/06/2025] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
As a common side effect of radiotherapy, radiation-induced intestinal injury (RIII) greatly affects the prognosis of patients and the efficacy of radiotherapy. Current therapeutic strategies for RIII are still very limited. Thus, the identification of effective radioprotective agents is of great importance. NVP-AUY922 is an HSP90 inhibitor with favorable anti-inflammatory and antioxidant activities. It has been proven to mitigate radiation-induced lung injury. However, its effects on the alleviation of RIII remain unclear. In this study, our data indicated that NVP-AUY922 remarkably increased the survival rate after radiation exposure. NVP-AUY922 treatment could enhance the viability of intestinal stem cells (ISCs) and promote the recovery of the small intestine. In addition, it also inhibited intestinal inflammation and reshaped the gut microbiota structure. We found that the radioprotective effect of NVP-AUY922 is partially dependent on EPHX1. In addition, NVP-AUY922 could attenuate dextran sulfate sodium (DSS)-induced colitis and promote intestinal barrier recovery. Thus, our results suggest that NVP-AUY922 contributes to the amelioration of intestinal injury after radiation exposure, which offers a new approach for the prevention of RIII.
Collapse
Affiliation(s)
- Wenzhe Kong
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin 300192, China
| | - Xudong Ding
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin 300192, China
| | - Zhaoyu Wang
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin 300192, China
| | - Lu Lu
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin 300192, China.
| | - Saijun Fan
- Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Tianjin 300192, China.
| |
Collapse
|
44
|
Perra M, Manca ML. Recent Trends in Nanoantioxidants. Antioxidants (Basel) 2025; 14:207. [PMID: 40002393 PMCID: PMC11852173 DOI: 10.3390/antiox14020207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
The term "oxidative stress" refers to an imbalance between reactive oxygen species (ROS) generation and the antioxidant system, resulting in the increased formation of ROS and the reduced and/or inadequate efficiency of the physiological processes responsible for their elimination and homeostasis maintenance [...].
Collapse
Affiliation(s)
- Matteo Perra
- Department of Life and Environmental Sciences, University of Cagliari, University Campus, S.P. Monserrato-Sestu Km 0.700, 09042 Monserrato, CA, Italy
| | - Maria Letizia Manca
- Department of Life and Environmental Sciences, University of Cagliari, University Campus, S.P. Monserrato-Sestu Km 0.700, 09042 Monserrato, CA, Italy
| |
Collapse
|
45
|
He Z, Deng S, Wu Z, Cui Z, Mei H, Wang J, Wang K, Zhang Y. Angelica sinensis polysaccharide could alleviate the gastrointestinal damage in alcoholic fatty liver disease mice: Regulation of alcohol metabolism and enhancement of short-chain fatty acids utilization. JOURNAL OF ETHNOPHARMACOLOGY 2025; 338:119117. [PMID: 39551279 DOI: 10.1016/j.jep.2024.119117] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/11/2024] [Revised: 10/25/2024] [Accepted: 11/14/2024] [Indexed: 11/19/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Dysfunction of the intestinal barrier was an important trigger for alcoholic liver damage and alcohol had brought about intestinal damage before causing liver damage. The root of Angelica sinensis (Oliv.) Diels, crucial traditional medicinal material, was widely utilized for its blood-invigorating, intestinal-lubricating and gynecological benefits. Angelica sinensis polysaccharide (ASP) was an essential natural active ingredient of Angelica sinensis and exhibited considerable potential for gastrointestinal protection. Nevertheless, the systematic research of ASP on the gastrointestinal tract remained insufficient. AIM OF THIS STUDY To systematically explore the protective effect and underlying mechanisms of ASP against alcohol-induced gastrointestinal injury, including the stomach, ileum and colon. MATERIALS AND METHODS The AFLD mice model was established via the intragastric administration of alcohol twice a day for one week. The protective effect of ASP on the representative segments of the gastrointestinal tract (stomach, ileum and colon) was subsequently studied after confirming its hepatoprotective activity. The impact of ASP on gastrointestinal alcohol metabolism was examined to explain its antioxidant and antiapoptotic activities. Furthermore, the effect of ASP on short-chain fatty acids (SCFA) in the colon and colonic contents was investigated to further enhance the understanding of the underlying mechanisms. RESULTS ASP could reduce oxidative stress and apoptosis in the gastrointestinal tract via regulating CYP2E1-mediated alcohol metabolism. Additionally, ASP could significantly increase the levels of FFAR2, FFAR3 and HCAR2 in colon, thereby promoting the utilization of SCFA. CONCLUSION ASP was proven for the first time to improve gastrointestinal damage caused by alcohol, indicating its enormous potential as a candidate medicine for the treatment of alcohol related gastrointestinal injury.
Collapse
Affiliation(s)
- Zihao He
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, PR China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, 430030, Wuhan, PR China
| | - Siyuan Deng
- Hubei Key Laboratory of Nature Medicinal Chemistry and Resource Evaluation, Tongji Medical College of Pharmacy, Huazhong University of Science and Technology, 430030, Wuhan, PR China
| | - Zhijing Wu
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, PR China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, 430030, Wuhan, PR China
| | - Zheng Cui
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, PR China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, 430030, Wuhan, PR China
| | - Hao Mei
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, PR China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, 430030, Wuhan, PR China
| | - Jinglin Wang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, PR China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, 430030, Wuhan, PR China.
| | - Kaiping Wang
- Hubei Key Laboratory of Nature Medicinal Chemistry and Resource Evaluation, Tongji Medical College of Pharmacy, Huazhong University of Science and Technology, 430030, Wuhan, PR China.
| | - Yu Zhang
- Department of Pharmacy, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 430030, Wuhan, PR China; Hubei Province Clinical Research Center for Precision Medicine for Critical Illness, 430030, Wuhan, PR China.
| |
Collapse
|
46
|
Chen Y, Fan W, Lyu Y, Liao J, Zhou Y. METTL14 modulates the progression and ferroptosis of colitis by regulating the stability of m6A-modified GPX4. Eur J Med Res 2025; 30:88. [PMID: 39920858 PMCID: PMC11806865 DOI: 10.1186/s40001-025-02334-8] [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: 11/12/2024] [Accepted: 01/26/2025] [Indexed: 02/09/2025] Open
Abstract
Ulcerative colitis (UC) is non-specific inflammatory bowel disease. UC development and progression were closely associated with epigenetic modifications. Nevertheless, the specific relationship between N6-methyladenosine (m6A) modification at RNA transcription levels and UC pathogenesis remains unclear. We established UC cell models and mouse models through dextran sulfate sodium (DSS) induction. The expression levels of METTL14 were analyzed via qRT-PCR and western blot. In vitro functional experiments evaluated the effects of METTL14 overexpression on the viability of DSS-induced NCM460 cells and ferroptosis markers. Use of the m6A methylation detection kit, MeRIP-qPCR, and RNA stability experiments confirmed the molecular mechanism controlled by METTL14. In vivo experiments with inflammatory mice models elucidated the interaction between METTL14 and GPX4. Findings from this study indicated a notable reduction in m6A methyltransferase METTL14 expression in DSS-induced NCM460 cells and DSS-induced mice models. METTL14 overexpression effectively suppressed ferroptosis in DSS-induced NCM460 cells. In addition, METTL14 enhanced GPX4 mRNA stability through mediating m6A modification, and the interplay between METTL14 and GPX4 through m6A modification introduced innovative therapeutic approaches for UC management.
Collapse
Affiliation(s)
- Yuhua Chen
- Anorectal Department, Dongguan Hospital of Traditional Chinese Medicine, 1st Floor, No. 3, Dongcheng Section, Songshanhu Avenue, Dongcheng Street, Dongguan, 523000, Guangdong, China
| | - Weicong Fan
- Anorectal Department, Dongguan Hospital of Traditional Chinese Medicine, 1st Floor, No. 3, Dongcheng Section, Songshanhu Avenue, Dongcheng Street, Dongguan, 523000, Guangdong, China
| | - Ying Lyu
- Anorectal Department, Dongguan Hospital of Traditional Chinese Medicine, 1st Floor, No. 3, Dongcheng Section, Songshanhu Avenue, Dongcheng Street, Dongguan, 523000, Guangdong, China
| | - Jingsheng Liao
- Medical Oncology, The Tenth Affiliated Hospital of Southern Medical University (Dongguan People's Hospital), No. 78 Wandao Road, Dongguan, 523059, Guangdong, China.
| | - Ying Zhou
- Anorectal Department, Dongguan Hospital of Traditional Chinese Medicine, 1st Floor, No. 3, Dongcheng Section, Songshanhu Avenue, Dongcheng Street, Dongguan, 523000, Guangdong, China.
| |
Collapse
|
47
|
Ma Y, Hui KL, Ambaw YA, Walther TC, Farese RV, Lengyel M, Gelashvili Z, Lu D, Niethammer P. DHRS7 Integrates NADP +/NADPH Redox Sensing with Inflammatory Lipid Signalling via the Oxoeicosanoid Pathway. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.05.636725. [PMID: 39975387 PMCID: PMC11839141 DOI: 10.1101/2025.02.05.636725] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/21/2025]
Abstract
During the innate immune response at epithelial wound sites, oxidative stress acts microbicidal and-mechanistically less well understood-as an immune and resilience signal. The reversible sulfhydryl (SH) oxidation of kinases, phosphatases, and transcription factors constitute the perhaps best-known redox signalling paradigm, whereas mechanisms that transduce metabolic redox cues, such as redox cofactor balance, remain little explored. Here, using mammalian cells, microsomes, and live zebrafish, we identify DHRS7, a short-chain fatty acid dehydrogenase/reductase (SDR), as conserved, 5-hydroxyeicosanoid dehydrogenase (5-HEDH). Under oxidative stress, DHRS7 consumes NADP+ to convert arachidonic acid (AA)-derived 5(S)-HETE into the inflammatory lipid 5-KETE, which activates leukocyte chemotaxis via the OXER1 receptor. While Dhrs7 acts as a NADPH-dependent 5-KETE sink in unstressed, healthy tissue, it promotes rapid, 5-KETE dependent leukocytic inflammation in wounded zebrafish skin. Thus, DHRS7 epitomizes an underappreciated mode of redox signalling-beyond classic SH oxidation-that leverages NADPH metabolism to generate or quench a paracrine lipid signal. Metabolic redox sensors like DHRS7 might be promising therapeutic targets in diseases characterized by disturbed redox balance.
Collapse
Affiliation(s)
- Yanan Ma
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - King Lam Hui
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Yohannes A. Ambaw
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Tobias C. Walther
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Howard Hughes Medical Institute, New York, NY 10065, USA
| | - Robert V. Farese
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Miklos Lengyel
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Zaza Gelashvili
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
- Gerstner Sloan Kettering Graduate School of Biomedical Sciences, New York, NY 10065, USA
| | - Dajun Lu
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| | - Philipp Niethammer
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, New York, NY 10065, USA
| |
Collapse
|
48
|
Wang AYL, Aviña AE, Liu YY, Chang YC, Kao HK. Transcription Factor Blimp-1: A Central Regulator of Oxidative Stress and Metabolic Reprogramming in Chronic Inflammatory Diseases. Antioxidants (Basel) 2025; 14:183. [PMID: 40002370 PMCID: PMC11851694 DOI: 10.3390/antiox14020183] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/17/2025] [Accepted: 02/03/2025] [Indexed: 02/27/2025] Open
Abstract
B-lymphocyte-induced maturation protein 1 (Blimp-1) is a transcription factor that, among other functions, modulates metabolism and helps to regulate antioxidant pathways, which is important in the context of chronic inflammatory diseases like diabetes, cardiovascular disease, and autoimmune disease. In immune cell function, Blimp-1 has a modulatory role in the orchestration of metabolic reprogramming and as a promoter of anti-inflammatory cytokines, including IL-10, responsible for modulating oxidative stress and immune homeostasis. Moreover, Blimp-1 also modulates key metabolic aspects, such as glycolysis and fatty acid oxidation, which regulate reactive oxygen species levels, as well as tissue protection. This review depicts Blimp-1 as an important regulator of antioxidant defenses and anti-inflammation and suggests that the protein could serve as a therapeutic target in chronic inflammatory and metabolic dysregulation conditions. The modulation of Blimp-1 in diseases such as diabetic coronary heart disease and atherosclerosis could alleviate oxidative stress, augment the protection of tissues, and improve disease outcomes. The therapeutic potential for the development of new treatments for these chronic conditions lies in the synergy between the regulation of Blimp-1 and antioxidant therapies, which are future directions that may be pursued. This review emphasizes Blimp-1's emerging importance as a novel regulator in the pathogenesis of inflammatory diseases, providing new opportunities for therapeutic intervention.
Collapse
Affiliation(s)
- Aline Yen Ling Wang
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (A.E.A.); (Y.-Y.L.)
| | - Ana Elena Aviña
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (A.E.A.); (Y.-Y.L.)
- International PhD Program in Medicine, College of Medicine, Taipei Medical University, Taipei 110, Taiwan
| | - Yen-Yu Liu
- Center for Vascularized Composite Allotransplantation, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan; (A.E.A.); (Y.-Y.L.)
| | - Yun-Ching Chang
- Department of Health Industry Technology Management, Chung Shan Medical University, Taichung 402, Taiwan;
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung 402, Taiwan
| | - Huang-Kai Kao
- Department of Plastic and Reconstructive Surgery, Chang Gung Memorial Hospital, Taoyuan 333, Taiwan;
- College of Medicine, Chang Gung University, Taoyuan 333, Taiwan
| |
Collapse
|
49
|
Liao W, Wang J, Li Y. Natural products based on Correa's cascade for the treatment of gastric cancer trilogy: Current status and future perspective. J Pharm Anal 2025; 15:101075. [PMID: 39957902 PMCID: PMC11830317 DOI: 10.1016/j.jpha.2024.101075] [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: 04/02/2024] [Revised: 07/18/2024] [Accepted: 08/16/2024] [Indexed: 02/18/2025] Open
Abstract
Gastric carcinoma (GC) is a malignancy with multifactorial involvement, multicellular regulation, and multistage evolution. The classic Correa's cascade of intestinal GC specifies a trilogy of malignant transformation of the gastric mucosa, in which normal gastric mucosa gradually progresses from inactive or chronic active gastritis (Phase I) to gastric precancerous lesions (Phase II) and finally to GC (Phase III). Correa's cascade highlights the evolutionary pattern of GC and the importance of early intervention to prevent malignant transformation of the gastric mucosa. Intervening in early gastric mucosal lesions, i.e., Phase I and II, will be the key strategy to prevent and treat GC. Natural products (NPs) have been an important source for drug development due to abundant sources, tremendous safety, and multiple pharmacodynamic mechanisms. This review is the first to investigate and summarize the multi-step effects and regulatory mechanisms of NPs on the Correa's cascade in gastric carcinogenesis. In phase I, NPs modulate Helicobacter pylori urease activity, motility, adhesion, virulence factors, and drug resistance, thereby inhibiting H. pylori-induced gastric mucosal inflammation and oxidative stress, and facilitating ulcer healing. In Phase II, NPs modulate multiple pathways and mediators regulating gastric mucosal cell cycle, apoptosis, autophagy, and angiogenesis to reverse gastric precancerous lesions. In Phase III, NPs suppress cell proliferation, migration, invasion, angiogenesis, and cancer stem cells, induce apoptosis and autophagy, and enhance chemotherapeutic drug sensitivity for the treatment of GC. In contrast to existing work, we hope to uncover NPs with sequential therapeutic effects on multiple phases of GC development, providing new ideas for gastric cancer prevention, treatment, and drug development.
Collapse
Affiliation(s)
- Wenhao Liao
- Department of Nephrology, the Key Laboratory for the Prevention and Treatment of Chronic Kidney Disease of Chongqing, Chongqing Clinical Research Center of Kidney and Urology Diseases, Xinqiao Hospital, Army Medical University (Third Military Medical University), Chongqing, 400037, China
| | - Jing Wang
- Department of Obstetrics and Gynecology, Chongqing Bishan Hospital of Traditional Chinese Medicine, Chongqing, 402760, China
| | - Yuchen Li
- Department of Laboratory Medicine, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| |
Collapse
|
50
|
Nie W, Zhong W, Qian L, Zhong H, Hou Y, Xu H, Qi S, Dai L, Han X, Yang X, Xu R, He Y, Lin D, Gao F. Oral chitosan-cyclodextrin "shell-core" nanoparticles co-loaded Rhein and chlorogenic acid for ulcerative colitis treatment. Int J Biol Macromol 2025; 288:138493. [PMID: 39647762 DOI: 10.1016/j.ijbiomac.2024.138493] [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: 06/02/2024] [Revised: 12/02/2024] [Accepted: 12/05/2024] [Indexed: 12/10/2024]
Abstract
The food-derived ingredients Rhein (RH) and chlorogenic acid (CGA) have DEMONSTRATED a potential synergistic effect in the treatment of ulcerative colitis (UC) through their anti-inflammatory and antioxidant properties. However, the oral co-delivery of RH and CGA faces challenges such as differences in hydrophilicity and hydrophobicity, gastrointestinal instability, and inadequate colonic targeting. To address these issues, shell-core nanoparticles were developed for the co-encapsulation of RH and CGA (CP@CGA-FA/TA@RH NPs). These nanoparticles utilize cyclodextrin-based polymers and folate-amantadine polymers to form a supramolecular core that targets macrophages for anti-inflammatory action with RH, while chitosan cross-link to CGA in the outer shell provides microenvironment-sensitive antioxidant release. The results indicate that CP@CGA-FA/TA@RH NPs could effectively inhibit the classical TLR4/MyD88/NF-κB-mediated anti-inflammatory pathway and activate the Nrf2/HO-1-mediated antioxidant pathway, offering a novel approach to UC treatment. Q-value analysis confirms the substantial co-medication effect between RH and CGA. This study is the first to develop a nano-system combining two food-derived ingredients for the integrated treatment of UC.
Collapse
Affiliation(s)
- Wenbiao Nie
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Wenzhen Zhong
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Lin Qian
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu 610072, China
| | - Huiyun Zhong
- Sichuan Vocational College of Health and Rehabilitation, Zigong 643000, China
| | - Yusen Hou
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China.
| | - Haiting Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Shanshan Qi
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Linxin Dai
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Xiaoqin Han
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Xinyue Yang
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Runchun Xu
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China
| | - Yao He
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China.
| | - Dasheng Lin
- Chengdu Huashen Technology Group Co., Ltd., Chengdu 611137, China.
| | - Fei Gao
- State Key Laboratory of Southwestern Chinese Medicine Resources, Pharmacy School, Chengdu University of Traditional Chinese Medicine, Chengdu 611130, China.
| |
Collapse
|