1
|
Xie Z, Chen F, Li P, Xiao Y, Ruan Z. Tracking the gastrointestinal fate and prebiotic effect of ellagic acid-rich fruit byproducts: A comprehensive evaluation. Food Chem 2025; 484:144290. [PMID: 40250217 DOI: 10.1016/j.foodchem.2025.144290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/21/2025] [Accepted: 04/08/2025] [Indexed: 04/20/2025]
Abstract
This study demonstrated the potential of ellagic acid-rich fruit byproducts, particularly pomegranate peel, as functional ingredients. During digestion, pomegranate peel exhibited superior antioxidant activity owing to its high phenolic content. Twenty-four phenolic compounds were released, with pomegranate peel maintaining higher bioactivity than chestnut and walnut peels. In vitro colonic fermentation with urolithin A-producing microbiota revealed that pomegranate peel stimulated butyrate synthesis (11.94 mM) and urolithin A production (6.31 μM), highlighting the prebiotic role of ellagic acid. Gut microbiota modulation by pomegranate peel increased Bacteroides and Bifidobacterium (a potential key for ellagic acid conversion) while suppressing Alistipes. Functional analyses confirmed its effect on carbohydrate, lipid, and amino acid metabolism. In contrast, chestnut and walnut peels exhibited lower bioactivity and microbial selectivity. These findings positioned pomegranate peel as a superior ingredient for gut health and microbial metabolism optimization, offering targeted nutritional benefits over conventional byproducts.
Collapse
Affiliation(s)
- Zhike Xie
- State Key Laboratory of Food Science and Resources, Institute of Nutrition, Nanchang University, Nanchang 330047, China
| | - Feng Chen
- State Key Laboratory of Food Science and Resources, Institute of Nutrition, Nanchang University, Nanchang 330047, China
| | - Pei Li
- State Key Laboratory of Food Science and Resources, Institute of Nutrition, Nanchang University, Nanchang 330047, China
| | - Yawen Xiao
- State Key Laboratory of Food Science and Resources, Institute of Nutrition, Nanchang University, Nanchang 330047, China
| | - Zheng Ruan
- State Key Laboratory of Food Science and Resources, Institute of Nutrition, Nanchang University, Nanchang 330047, China.
| |
Collapse
|
2
|
Daniel N, Farinella R, Belluomini F, Fajkic A, Rizzato C, Souček P, Campa D, Hughes DJ. The relationship of the microbiome, associated metabolites and the gut barrier with pancreatic cancer. Semin Cancer Biol 2025; 112:43-57. [PMID: 40154652 DOI: 10.1016/j.semcancer.2025.03.002] [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: 11/30/2024] [Revised: 02/26/2025] [Accepted: 03/19/2025] [Indexed: 04/01/2025]
Abstract
Pancreatic cancers have high mortality and rising incidence rates which may be related to unhealthy western-type dietary and lifestyle patterns as well as increasing body weights and obesity rates. Recent data also suggest a role for the gut microbiome in the development of pancreatic cancer. Here, we review the experimental and observational evidence for the roles of the oral, gut and intratumoural microbiomes, impaired gut barrier function and exposure to inflammatory compounds as well as metabolic dysfunction as contributors to pancreatic disease with a focus on pancreatic ductal adenocarcinoma (PDAC) initiation and progression. We also highlight some emerging gut microbiome editing techniques currently being investigated in the context of pancreatic disease. Notably, while the gut microbiome is significantly altered in PDAC and its precursor diseases, its utility as a diagnostic and prognostic tool is hindered by a lack of reproducibility and the potential for reverse causality in case-control cohorts. Future research should emphasise longitudinal and mechanistic studies as well as integrating lifestyle exposure and multi-omics data to unravel complex host-microbiome interactions. This will allow for deeper aetiologic and mechanistic insights that can inform treatments and guide public health recommendations.
Collapse
Affiliation(s)
- Neil Daniel
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomedical and Biomolecular Sciences, University College Dublin, Dublin, Ireland
| | | | | | - Almir Fajkic
- Department of Pathophysiology Faculty of Medicine, University of Sarajevo, Sarajevo, Bosnia and Herzegovina
| | | | - Pavel Souček
- Laboratory of Pharmacogenomics, Biomedical Center, Faculty of Medicine in Pilsen, Charles University, Pilsen, Czech Republic; Toxicogenomics Unit, National Institute of Public Health, Prague, Czech Republic
| | - Daniele Campa
- Department of Biology, University of Pisa, Pisa, Italy
| | - David J Hughes
- Molecular Epidemiology of Cancer Group, UCD Conway Institute, School of Biomedical and Biomolecular Sciences, University College Dublin, Dublin, Ireland.
| |
Collapse
|
3
|
Zhang L, Tuoliken H, Li J, Gao H. Diet, gut microbiota, and health: a review. Food Sci Biotechnol 2025; 34:2087-2099. [PMID: 40351733 PMCID: PMC12064509 DOI: 10.1007/s10068-024-01759-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 10/19/2024] [Accepted: 11/13/2024] [Indexed: 05/14/2025] Open
Abstract
The relationship between diet and human physical and mental health is highly interconnected and has been significantly correlated with the occurrence of various diseases, including neurological disorders, cancer, and chronic inflammatory diseases. Moreover, diet has been demonstrated to play a pivotal role in governing gut microbiota composition, making it one of the most influential factors. The diet is crucial in connecting humans and their gut microorganisms. The nutrients ingested supply energy to the body and serve as substrates for the metabolic processes of the gut microorganisms. Consequently, the gut flora and their metabolites reciprocally impact the host's metabolism, thereby influencing the physiological state of the human body. Extensive investigations on human and mouse models have revealed that diet potentially underlies various effects on human health and disease. Graphical abstract
Collapse
Affiliation(s)
- Longxiang Zhang
- The Second Department of Gastroenterology, the First Affiliated Hospital of Xinjiang, Medical University, Urumqi, 830000 Xinjiang China
| | - Haishaer Tuoliken
- The Second Department of Gastroenterology, the First Affiliated Hospital of Xinjiang, Medical University, Urumqi, 830000 Xinjiang China
| | - Jian Li
- The Second Department of Gastroenterology, the First Affiliated Hospital of Xinjiang, Medical University, Urumqi, 830000 Xinjiang China
| | - Hongliang Gao
- The Second Department of Gastroenterology, the First Affiliated Hospital of Xinjiang, Medical University, Urumqi, 830000 Xinjiang China
| |
Collapse
|
4
|
Neurath MF, Artis D, Becker C. The intestinal barrier: a pivotal role in health, inflammation, and cancer. Lancet Gastroenterol Hepatol 2025; 10:573-592. [PMID: 40086468 DOI: 10.1016/s2468-1253(24)00390-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 11/08/2024] [Accepted: 11/15/2024] [Indexed: 03/16/2025]
Abstract
The intestinal barrier serves as a boundary between the mucosal immune system in the lamina propria and the external environment of the intestinal lumen, which contains a diverse array of microorganisms and ingested environmental factors, including pathogens, food antigens, toxins, and other foreign substances. This barrier has a central role in regulating the controlled interaction between luminal factors and the intestinal immune system. Disruptions of intestinal epithelial cells, which serve as a physical barrier, or the antimicrobial peptides and mucins they produce, which act as a chemical barrier, can lead to a leaky gut. In this state, the intestinal wall is unable to efficiently separate the intestinal flora and luminal contents from the intestinal immune system. The subsequent activation of the immune system has an important role in the pathogenesis of inflammatory bowel disease, as well as in metabolic dysfunction-associated steatohepatitis, primary sclerosing cholangitis, and colorectal cancer. Dysregulated intestinal barrier integrity has also been described in patients with chronic inflammatory diseases outside the gastrointestinal tract, including rheumatoid arthritis and neurodegenerative disorders. Mechanistic studies of barrier dysfunction have revealed that the subsequent local activation and systemic circulation of activated immune cells and the cytokines they secrete, as well as extracellular vesicles, promote proinflammatory processes within and outside the gastrointestinal tract. In this Review, we summarise these findings and highlight several new therapeutic concepts currently being developed that attempt to control inflammatory processes via direct or indirect modulation of intestinal barrier function.
Collapse
Affiliation(s)
- Markus F Neurath
- Medical Clinic 1, Department of Gastroenterology, Ludwig Demling Endoscopy Center of Excellence, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany.
| | - David Artis
- Jill Roberts Institute for Research in Inflammatory Bowel Disease, Weill Cornell Medicine, Cornell University, New York, NY, USA; Friedman Center for Nutrition and Inflammation, Weill Cornell Medicine, Cornell University, New York, NY, USA; Joan and Sanford I Weill Department of Medicine, Weill Cornell Medicine, Cornell University, New York, NY, USA; Department of Microbiology and Immunology, Weill Cornell Medicine, Cornell University, New York, NY, USA; Allen Discovery Center for Neuroimmune Interactions, Weill Cornell Medicine, Cornell University, New York, NY, USA
| | - Christoph Becker
- Medical Clinic 1, Department of Gastroenterology, Ludwig Demling Endoscopy Center of Excellence, University Hospital Erlangen, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany; Deutsches Zentrum Immuntherapie (DZI), Friedrich-Alexander University Erlangen-Nürnberg, Erlangen, Germany
| |
Collapse
|
5
|
Xiong L, Huang YX, Mao L, Xu Y, Deng YQ. Targeting gut microbiota and its associated metabolites as a potential strategy for promoting would healing in diabetes. World J Diabetes 2025; 16:98788. [DOI: 10.4239/wjd.v16.i5.98788] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Revised: 01/03/2025] [Accepted: 03/05/2025] [Indexed: 04/25/2025] Open
Abstract
Impaired healing of diabetic wounds is one of the most important complications of diabetes, often leading to lower limb amputations and incurring significant economic and psychosocial costs. Unfortunately, there are currently no effective prevention or treatment strategies available. Recent research has reported that an imbalance in the gut microbiota, known as dysbiosis, was linked to the onset of type 2 diabetes, as well as the development and progression of diabetic complications. Indeed, the gut microbiota has emerged as a promising therapeutic approach for treating type 2 diabetes and related diseases. However, there is few of literatures specifically discussing the relationship between gut microbiota and diabetic wounds. This review aims to explore the potential role of the gut microbiota, especially probiotics, and its associated byproducts such as short chain fatty acids, bile acids, hydrogen sulfide, and tryptophan metabolites on wound healing to provide fresh insights and novel perspectives for the treatment of chronic wounds in diabetes.
Collapse
Affiliation(s)
- Ling Xiong
- Department of Dermatology & STD, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Ya-Xin Huang
- Department of Dermatology & STD, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Lan Mao
- Department of Dermatology & STD, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Yong Xu
- Department of Endocrinology and Metabolism, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, Sichuan Province, China
| | - Yong-Qiong Deng
- Department of Dermatology & STD, Chengdu Integrated TCM & Western Medicine Hospital, Chengdu 610000, Sichuan Province, China
- Institute of Cardiovascular Research, Southwest Medical University, Luzhou 646000, Sichuan Province, China
| |
Collapse
|
6
|
Xu H, Lv D, Guan Y. Appeal of Urolithins from Synthesis to Biological Activities. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:11477-11494. [PMID: 40300072 DOI: 10.1021/acs.jafc.5c00634] [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: 05/01/2025]
Abstract
Urolithins (Uros), a series of natural polyphenols derived from ellagic acid through gut bacteria metabolism, have gathered significant attention due to their diverse bioactivities such as maintaining mitochondrial health and anti-inflammatory and antioxidative effects. However, the ability to metabolize Uros varies among individuals. This Review provides a comprehensive insight into the synthesis, encapsulation and bioactivities of Uros, focusing on their biotransformation in vivo. We highlight the critical role of gut microbiota in the biotransformation of urolithins, including primary bacterial species such as Gordonibacter urolithinfaciens, Enterocloster bolteae and Enterococcus faecium. Furthermore, the therapeutic potential of Uros in alleviating neurodegenerative diseases, cancer, and Duchenne muscular dystrophy is discussed. Finally, several encapsulation strategies for enhancing the solubility and bioavailability of Uros are summarized. Future research direction includes identifying key genes involved in Uros biotransformation, elucidating the bioactive mechanisms of Uros, and improving their bioavailability. In conclusion, we synthesized biosynthetic pathways and bioactive properties of Uros for better utilization in health management.
Collapse
Affiliation(s)
- Huanyu Xu
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510641, China
| | - Danyu Lv
- School of Food Science and Engineering, South China University of Technology, Guangzhou 510641, China
| | - Yongguang Guan
- Department of Food Science, Foshan University, Foshan 528000, China
| |
Collapse
|
7
|
Song L, Xu L, Zhang P, Li S, Qu Y, Zhao Y, Shi Z, Ma R, Li Y, Chen Y, Wang Y, Jiang Z, Wei G, Shen X. A Dual-Targeting T6SS DNase Drives Bacterial Antagonism and Eukaryotic Apoptosis via the cGAS-STING-TNF Axis. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025:e2504086. [PMID: 40365777 DOI: 10.1002/advs.202504086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/04/2025] [Revised: 04/27/2025] [Indexed: 05/15/2025]
Abstract
The Type VI secretion system (T6SS) is a key virulence mechanism utilized by many Gram-negative bacteria to mediate the microbial competition and host pathogenesis. Despite the identification of diverse T6SS effectors targeting eukaryotic or prokaryotic cells, the trans-kingdom T6SS effectors that simultaneously target both eukaryotic and prokaryotic cells remain rarely reported. In this study, it is demonstrated that Yersinia pseudotuberculosis (Yptb) T6SS secretes a DNase effector, TkeA, which induces apoptosis in host cells. The translocation of TkeA into host cells causes nuclear DNA damage. This, in turn, activates the DNA-sensing cyclic GMP-AMP synthase (cGAS)/stimulator of interferon genes (STING) pathway. The activation of the cGAS-STING pathway by TkeA subsequently triggers apoptosis in host cells via extrinsic pathways, with tumor necrosis factor (TNF) signaling playing a critical role. Additionally, TkeA enhances bacterial competition by targeting rival bacteria, thereby promoting host colonization. These findings reveal that the transkingdom T6SS effector TkeA executes a "one weapon, two battlefields" strategy, acting as a trans-kingdom effector that enhances interbacterial competition while inducing apoptosis in host cells through the activation of the cGAS-STING-TNF axis. This highlights a previously unrecognized dimension of bacterial virulence strategies and expands the understanding of host-pathogen interactions involving T6SS effectors.
Collapse
Affiliation(s)
- Li Song
- Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Natural Resources and Environment, Northwest A&F University, Yangling, Shaanxi, 12100, P. R. China
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Lei Xu
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Pengfei Zhang
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Shuying Li
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Yichen Qu
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Yixin Zhao
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Zhenkun Shi
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Ruiqi Ma
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Yongdong Li
- Ningbo Municipal Center for Disease Control and Prevention, Ningbo, Zhejiang, 315010, P. R. China
| | - Yi Chen
- Ningbo Municipal Center for Disease Control and Prevention, Ningbo, Zhejiang, 315010, P. R. China
| | - Yao Wang
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Zhengfan Jiang
- Key Laboratory of Cell Proliferation and Differentiation of the Ministry of Education, School of Life Sciences, Peking University, Beijing, 100871, P. R. China
- Peking-Tsinghua Center for Life Sciences, Peking University, Beijing, 100871, P. R. China
| | - Gehong Wei
- Shaanxi Key Laboratory of Agricultural and Environmental Microbiology, College of Natural Resources and Environment, Northwest A&F University, Yangling, Shaanxi, 12100, P. R. China
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| | - Xihui Shen
- College of Life Sciences, Northwest A&F University, Yangling, Shaanxi, 712100, P. R. China
| |
Collapse
|
8
|
Maity D, Rahi V, Dorai ST, Chandrashekharappa S, Kaundal RK. Urolithin-A Derivative UAS03 Improves Cognitive Deficits and Memory by Activating Nrf2 Pathways to Alleviate Oxidative Stress and Neuroinflammation. ACS Chem Neurosci 2025; 16:1815-1826. [PMID: 40227891 DOI: 10.1021/acschemneuro.4c00886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/16/2025] Open
Abstract
Neuroinflammation is a key factor in age-related cognitive decline and memory impairment. UAS03, a potent synthetic analogue of Urolithin-A, has demonstrated anti-inflammatory and antioxidant properties. This investigation examined the neuroprotective effect of UAS03 on lipopolysaccharide (LPS) induced neuroinflammation, and its associated cognitive impairments, memory deficits, and depression-like behaviors. Intracerebroventricular administration of LPS (12 μg/kg) was performed to induce neuroinflammation in mice, followed by a 7 day treatment with UAS03 at 10 and 30 mg/kg doses. Mice were evaluated for depressive and anxiety-like behavior, spatial memory, and learning functions using a series of neurobehavioral test paradigms. Histopathological and molecular analyses were conducted using hematoxylin-eosin and cresyl violet staining, immunohistochemistry, ELISA, and Western blotting techniques. We have found that, UAS03 significantly enhanced cognitive and memory functions impaired by LPS while concurrently reducing depressive symptoms. Furthermore, the compound attenuated neuronal damage and decreased the expression of IBA-1 and GFAP in hippocampal region. Through the activation of the nuclear factor erythroid 2-related factor 2 (Nrf2) signaling pathway, UAS03 effectively mitigated markers of oxidative stress and reduced levels of pro-inflammatory factors, including IL-1β, TNF-α, and COX-2. Cumulatively, this study provides compelling evidence that UAS03 exerts neuroprotective effects by regulating essential pathways involved in anti-inflammatory and neuroprotective mechanisms, suggesting its potential as a preventative measure against age-related cognitive decline and memory impairments associated with neuroinflammation.
Collapse
Affiliation(s)
- Dipan Maity
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, Uttar Pradesh 226002, India
| | - Vikrant Rahi
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, Uttar Pradesh 226002, India
| | - Sandya Tambi Dorai
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, Uttar Pradesh 226002, India
| | - Sandeep Chandrashekharappa
- Department of Medicinal Chemistry, National Institute of Pharmaceutical Education and Research Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, Uttar Pradesh 226002, India
| | - Ravinder K Kaundal
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, Uttar Pradesh 226002, India
- Department of Regulatory Toxicology, National Institute of Pharmaceutical Education and Research, Raebareli (NIPER-R), Transit Campus, Bijnor-Sisendi Road, Sarojini Nagar, Near CRPF Base Camp, Lucknow, Uttar Pradesh 226002, India
| |
Collapse
|
9
|
Chen YM, Chuang SY, Tsai CY. The Impact of Daily Walnut Consumption on Gastrointestinal Symptoms: A Mixed-Method Study in Healthy Adults. JOURNAL OF THE AMERICAN NUTRITION ASSOCIATION 2025; 44:332-337. [PMID: 39778130 DOI: 10.1080/27697061.2024.2431287] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 10/31/2024] [Accepted: 11/13/2024] [Indexed: 01/11/2025]
Abstract
BACKGROUND Common gastrointestinal (GI) symptoms such as abdominal pain, indigestion, and constipation affect a significant portion of the global population and can substantially impair quality of life. Despite these widespread issues, research specifically investigating the effects of walnuts on gut function and GI symptoms remain limited. OBJECTIVE This study investigates the effects of walnuts on gastrointestinal symptoms in healthy adults. DESIGN An experimental baseline-end study with an equivalent group design was employed. SETTING The experimental group consumed 42 grams of walnuts daily, and their gastrointestinal symptoms were compared with those of a control group that did not consume walnuts over a 3-week period. PARTICIPANTS Sixty university students were recruited as volunteer subjects, consisting of 30 males and 30 females. INTERVENTION(S) Participants were randomly assigned to either an experimental group or a control group. MAIN OUTCOME MEASURE(S) The independent variable was walnut consumption, and the dependent variable was gastrointestinal health, assessed using the Gastrointestinal Symptom Rating Scale (GSRS) and a qualitative questionnaire to collect participants' perceived changes in GI symptoms. ANALYSIS A t-test with a p-value of less than 0.05 and verbatim analysis were utilized. RESULTS This mixed-methods study provides evidence for the beneficial effects of walnuts in promoting normal digestive function. CONCLUSIONS AND IMPLICATIONS The study provides alternative evidence for the beneficial effects of walnuts in promoting normal digestive function.
Collapse
Affiliation(s)
- Yi-Mei Chen
- School of Foreign Languages, Jiaying University, Meizhou City, Guangdong, China
| | - Shu-Yu Chuang
- Department of Education, University of Taipei, Taipei, Taiwan
| | - Chih-Yung Tsai
- Department of Education, University of Taipei, Taipei, Taiwan
| |
Collapse
|
10
|
Fan Y, Li Y, Gu X, Chen N, Chen Y, Fang C, Wang Z, Yin Y, Deng H, Dai L. Intestinal metabolites in colitis-associated carcinogenesis: Building a bridge between host and microbiome. Chin Med J (Engl) 2025:00029330-990000000-01527. [PMID: 40287783 DOI: 10.1097/cm9.0000000000003430] [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: 11/18/2024] [Indexed: 04/29/2025] Open
Abstract
ABSTRACT Microbial-derived metabolites are important mediators of host-microbial interactions. In recent years, the role of intestinal microbial metabolites in colorectal cancer has attracted considerable attention. These metabolites, which can be derived from bacterial metabolism of dietary substrates, modification of host molecules such as bile acids, or directly from bacteria, strongly influence the progression of colitis-associated cancer (CAC) by regulating inflammation and immune response. Here, we review how microbiome metabolites short-chain fatty acids (SCFAs), secondary bile acids, polyamines, microbial tryptophan metabolites, and polyphenols are involved in the tumorigenesis and development of CAC through inflammation and immunity. Given the heated debate on the metabolites of microbiota in maintaining gut homeostasis, serving as tumor molecular markers, and affecting the efficacy of immune checkpoint inhibitors in recent years, strategies for the prevention and treatment of CAC by targeting intestinal microbial metabolites are also discussed in this review.
Collapse
Affiliation(s)
- Yating Fan
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China
| | - Yang Li
- Department of Gastrointestinal Surgery, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Xiangshuai Gu
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China
| | - Na Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China
- School of Pharmacy, Chengdu Medical College, Chengdu, Sichuan 610500, China
| | - Ye Chen
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China
| | - Chao Fang
- Colorectal Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Ziqiang Wang
- Colorectal Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Yuan Yin
- Gastric Cancer Center, West China Hospital, Sichuan University, Chengdu, Sichuan 610041, China
| | - Hongxin Deng
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China
| | - Lei Dai
- State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, Sichuan University and Collaborative Innovation Center for Biotherapy, Chengdu, Sichuan 610041, China
| |
Collapse
|
11
|
Wang X, Liu Y, Chang H, Tun HM, Xia X, Peng Y, Qin N. Goat Milk-Derived Extracellular Vesicles Alleviate Colitis Potentially Through Improved Gut Microbiota in Mice. Foods 2025; 14:1514. [PMID: 40361597 PMCID: PMC12071645 DOI: 10.3390/foods14091514] [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: 02/28/2025] [Revised: 04/18/2025] [Accepted: 04/21/2025] [Indexed: 05/15/2025] Open
Abstract
Ulcerative colitis (UC) is characterized clinically by intestinal inflammation and gut microbiota dysbiosis. The consumption of biologics, although effective in inflammation control, may lead to adverse effects and is inconvenient for at-home administration. Goat milk-derived extracellular vesicles (GMEVs) have been proposed as a supplement to prevent intestinal inflammation. However, their therapeutic potential for colitis remains elusive. This study aimed to explore the preventive effect of GMEVs on colitis and its underlying mechanisms through the microbiota-immune axis using a dextran sodium sulfate (DSS)-induced colitis mouse model. We found that a pre-treatment of 20 mg/kg/d GMEVs effectively prevented body weight loss, colon shortening, the depletion of colonic goblet cells, and the disappearance of crypts, while enhancing the intestinal mucosal barrier. Consistent with these phenotypes, GMEV pre-treatment increased levels of IL-22 and IL-10 and decreased levels of IL-1β, TNF-α, IL-6, and iNOS. However, GMEVs themselves had no effect on normal mice. Paralleling the alleviation of intestinal inflammation, GMEV pre-treatment also restored the reduction in unclassified Muribaculaceae, Dubosiella, and Lactobacillus and suppressed the expansion of Alistipes and Proteobacteria following DSS treatment. Additionally, GMEV intake significantly downregulated the expression of proteins in the NF-κB signaling pathway induced by DSS. In summary, GMEVs could prevent colitis by regulating intestinal inflammation, the intestinal mucosal barrier, gut microbiota, organ damage, and the immune microenvironment. This study demonstrated that GMEVs have potential application prospects for UC prevention.
Collapse
Affiliation(s)
- Xinru Wang
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Yi Liu
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Hong Chang
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Hein-Min Tun
- Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
- Microbiota I-Center (MagIC), Hong Kong SAR 999077, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Xiaodong Xia
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Ye Peng
- Jockey Club School of Public Health and Primary Care, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
- Microbiota I-Center (MagIC), Hong Kong SAR 999077, China
- Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Ningbo Qin
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| |
Collapse
|
12
|
Minato I, Mena P, Ricciardiello L, Scaioli E, Belluzzi A, Rotondo E, Derlindati E, Montanini B, Michelini C, Tosi N, Agullò Garcià V, Picone G, Mengucci C, Dobani S, Salamanca P, Rosi A, Dall'Asta M, Bresciani L, Curti C, Spisni E, Dei Cas A, Bordoni A, Tomás-Barberán FA, Ferguson LR, Del Rio D, Danesi F. Evidence for a Modulatory Effect of a 12-Week Pomegranate Juice Intervention on the Transcriptional Response in Inflammatory Bowel Disease Patients Reducing Fecal Calprotectin Levels: Findings From a Proof-of-Principle Study. Mol Nutr Food Res 2025:e70067. [PMID: 40255128 DOI: 10.1002/mnfr.70067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 03/27/2025] [Accepted: 04/02/2025] [Indexed: 04/22/2025]
Abstract
This study aimed at investigating the effects of pomegranate juice (POMJ) consumption on inflammatory biomarkers and gene expression in patients with inflammatory bowel disease (IBD) in clinical remission. In this randomized, placebo-controlled trial, 16 subjects with IBD in remission consumed POMJ or placebo for 12 weeks. POMJ consumption significantly reduced fecal calprotectin (FC) and plasma endotoxin levels. Transcriptomic analysis of peripheral blood mononuclear cells revealed upregulation of genes involved in mucosal immunity, including aryl hydrocarbon receptor (AHR), neutrophil cytosolic factor 4 (NCF4), and nuclear factor, interleukin 3 regulated (NFIL3). Urolithin metabotypes were predominantly of the B type, associated with intestinal dysbiosis. No significant changes were observed in serum inflammatory markers or colonic mucosal cytokine expression. POMJ consumption reduced markers of intestinal inflammation and modulated gene expression related to mucosal immunity and barrier function in patients with IBD. These findings suggest the potential of POMJ as a beneficial dietary intervention for maintaining remission in IBD, highlighting the promise of targeted nutritional strategies in managing chronic inflammatory conditions. Further research is needed to elucidate the long-term clinical implications of these molecular changes. TRIAL REGISTRATION: ClinicalTrials.gov identifier: NCT03000101.
Collapse
Affiliation(s)
- Ilaria Minato
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
- Interdepartmental Center for Innovation in Health Products, Biopharmanet-TEC, University of Parma, Parma, Italy
| | - Pedro Mena
- Department of Food and Drug, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Luigi Ricciardiello
- IRCCS - St. Orsola-Malpighi Hospital, Bologna, Italy
- Department of Medical and Surgical Sciences, University of Bologna, Bologna, Italy
| | | | | | - Enrica Rotondo
- Department of Agricultural and Food Sciences, University of Bologna, Cesena, Italy
| | - Eleonora Derlindati
- Department of Agricultural and Food Sciences, University of Bologna, Cesena, Italy
| | - Barbara Montanini
- Department of Chemistry, Life Sciences and Environmental Sustainability, University of Parma, Parma, Italy
- Interdepartmental Center for Innovation in Health Products, Biopharmanet-TEC, University of Parma, Parma, Italy
| | | | - Nicole Tosi
- Department of Food and Drug, University of Parma, Parma, Italy
| | | | - Gianfranco Picone
- Department of Agricultural and Food Sciences, University of Bologna, Cesena, Italy
| | - Carlo Mengucci
- Department of Agricultural and Food Sciences, University of Bologna, Cesena, Italy
| | - Sara Dobani
- Department of Food and Drug, University of Parma, Parma, Italy
| | | | - Alice Rosi
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Margherita Dall'Asta
- Department of Food and Drug, University of Parma, Parma, Italy
- Department of Animal Science, Food and Nutrition, Università Cattolica del Sacro Cuore, Piacenza, Italy
| | | | - Claudio Curti
- Department of Food and Drug, University of Parma, Parma, Italy
| | - Enzo Spisni
- Department of Biological, Geological, and Environmental Sciences, University of Bologna, Bologna, Italy
| | - Alessandra Dei Cas
- Department of Medicine and Surgery - Division of Endocrinology and Metabolic Diseases, University of Parma, Parma, Italy
| | - Alessandra Bordoni
- Department of Agricultural and Food Sciences, University of Bologna, Cesena, Italy
| | | | - Lynnette R Ferguson
- Department of Nutrition, Faculty of Medical and Health Sciences, University of Auckland, Auckland, New Zealand
| | - Daniele Del Rio
- Department of Food and Drug, University of Parma, Parma, Italy
- Microbiome Research Hub, University of Parma, Parma, Italy
| | - Francesca Danesi
- Department of Agricultural and Food Sciences, University of Bologna, Cesena, Italy
| |
Collapse
|
13
|
Ionescu VA, Diaconu CC, Gheorghe G, Mihai MM, Diaconu CC, Bostan M, Bleotu C. Gut Microbiota and Colorectal Cancer: A Balance Between Risk and Protection. Int J Mol Sci 2025; 26:3733. [PMID: 40332367 PMCID: PMC12028331 DOI: 10.3390/ijms26083733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Revised: 04/11/2025] [Accepted: 04/14/2025] [Indexed: 05/08/2025] Open
Abstract
The gut microbiome, a complex community of microorganisms residing in the intestinal tract, plays a dual role in colorectal cancer (CRC) development, acting both as a contributing risk factor and as a protective element. This review explores the mechanisms by which gut microbiota contribute to CRC, emphasizing inflammation, oxidative stress, immune evasion, and the production of genotoxins and microbial metabolites. Fusobacterium nucleatum, Escherichia coli (pks+), and Bacteroides fragilis promote tumorigenesis by inducing chronic inflammation, generating reactive oxygen species, and producing virulence factors that damage host DNA. These microorganisms can also evade the antitumor immune response by suppressing cytotoxic T cell activity and increasing regulatory T cell populations. Additionally, microbial-derived metabolites such as secondary bile acids and trimethylamine-N-oxide (TMAO) have been linked to carcinogenic processes. Conversely, protective microbiota, including Lactobacillus, Bifidobacterium, and Faecalibacterium prausnitzii, contribute to intestinal homeostasis by producing short-chain fatty acids (SCFAs) like butyrate, which exhibit anti-inflammatory and anti-carcinogenic properties. These beneficial microbes enhance gut barrier integrity, modulate immune responses, and inhibit tumor cell proliferation. Understanding the dynamic interplay between pathogenic and protective microbiota is essential for developing microbiome-based interventions, such as probiotics, prebiotics, and fecal microbiota transplantation, to prevent or treat CRC. Future research should focus on identifying microbial biomarkers for early CRC detection and exploring personalized microbiome-targeted therapies. A deeper understanding of host-microbiota interactions may lead to innovative strategies for CRC management and improved patient outcomes.
Collapse
Affiliation(s)
- Vlad Alexandru Ionescu
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila Bucharest, 050474 Bucharest, Romania; (V.A.I.); (G.G.); (M.-M.M.)
- Internal Medicine Department, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania
| | - Camelia Cristina Diaconu
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila Bucharest, 050474 Bucharest, Romania; (V.A.I.); (G.G.); (M.-M.M.)
- Internal Medicine Department, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania
- Academy of Romanian Scientists, 050085 Bucharest, Romania;
| | - Gina Gheorghe
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila Bucharest, 050474 Bucharest, Romania; (V.A.I.); (G.G.); (M.-M.M.)
- Internal Medicine Department, Clinical Emergency Hospital of Bucharest, 105402 Bucharest, Romania
| | - Mara-Madalina Mihai
- Faculty of Medicine, University of Medicine and Pharmacy Carol Davila Bucharest, 050474 Bucharest, Romania; (V.A.I.); (G.G.); (M.-M.M.)
- Department of Oncologic Dermathology, “Elias” University Emergency Hospital, 010024 Bucharest, Romania
| | - Carmen Cristina Diaconu
- Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (C.C.D.); (M.B.)
| | - Marinela Bostan
- Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (C.C.D.); (M.B.)
- Department of Immunology, “Victor Babes” National Institute of Pathology, 050096 Bucharest, Romania
| | - Coralia Bleotu
- Academy of Romanian Scientists, 050085 Bucharest, Romania;
- Stefan S. Nicolau Institute of Virology, Romanian Academy, 030304 Bucharest, Romania; (C.C.D.); (M.B.)
- Research Institute of the University of Bucharest (ICUB), University of Bucharest, 060023 Bucharest, Romania
| |
Collapse
|
14
|
Zhao Z, Lu H, Wang J, Wu T, Xu S, Ge Y, You Q, Jiang Z, Lu M. Discovery of β-amino acid substituted naphthalene sulfonamide derivatives as potent Kelch-like ECH-associated protein 1-nuclear factor erythroid 2-related factor 2 (Keap1-Nrf2) protein-protein interaction inhibitors for ulcerative colitis management. Eur J Med Chem 2025; 288:117384. [PMID: 39965408 DOI: 10.1016/j.ejmech.2025.117384] [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: 12/20/2024] [Revised: 01/24/2025] [Accepted: 02/02/2025] [Indexed: 02/20/2025]
Abstract
The transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) is a key regulator of cellular defense system against oxidative insults. Directly inhibiting the Kelch-like ECH-associated protein 1 (Keap1)-Nrf2 protein-protein interaction (PPI) has emerged as a promising approach to activate Nrf2 for the treatment of diseases associated with oxidative stress. Herein, we identified β-amino acids as privileged structural fragments for designing novel naphthalene sulfonamide-based Keap1-Nrf2 PPI inhibitors. Comprehensive structure-activity relationship (SAR) exploration identified compound 19 as the optimal inhibitor with an IC50 of 0.55 μM for disrupting the Keap1-Nrf2 interaction and a Kd of 0.50 μM for binding to Keap1. Further studies demonstrated that 19 effectively activated the Nrf2-regulated cytoprotective system and provided protective effects against dextran sulfate sodium (DSS)-induced ulcerative colitis (UC) in both in vitro and in vivo models. These findings highlight the potential of β-amino acid substituted naphthalene sulfonamide Keap1-Nrf2 inhibitor 19 as a prospective therapeutic agent for UC via Keap1 targeting.
Collapse
Affiliation(s)
- Ziquan Zhao
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Hongjin Lu
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Junjie Wang
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Tingting Wu
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Shicheng Xu
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yuxin Ge
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Qidong You
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China; Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University Medical College, Suzhou, 215123, China.
| | - Zhengyu Jiang
- Jiang Su Key Laboratory of Drug Design and Optimization and Department of Medicinal Chemistry, School of Pharmacy, China Pharmaceutical University, Nanjing, 210009, China.
| | - Mengchen Lu
- Department of Medicinal Chemistry, College of Pharmaceutical Sciences, Soochow University Medical College, Suzhou, 215123, China.
| |
Collapse
|
15
|
Zhou F, Feng X, Xu Z, Yan F, Song G, Tang L. Design, synthesis and biological activity of 8-hydroxy modified urolithin A derivatives as phosphodiesterase type II (PDE2) inhibitors. Bioorg Med Chem 2025; 121:118127. [PMID: 40015121 DOI: 10.1016/j.bmc.2025.118127] [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: 11/03/2024] [Revised: 02/17/2025] [Accepted: 02/18/2025] [Indexed: 03/01/2025]
Abstract
Urolithin A (UA) is a naturally occurring polyphenolic compound.Due to its remarkable efficacy in safeguarding the central nervous system, UA has emerged as a promising candidate for drug development targeting neurodegenerative diseases such as Alzheimer's. However, the source of UA is limited and the activity of UA to inhibit PDE2 needs to be further improved. Therefore, this study will be optimized on the basis of UA to seek PDE2 inhibitors with better activity. In this study, we designed a series of UA derivatives based on 4HTX as the target protein and UA as the lead compound, utilizing the binding crystal structures of 4HTX and BAY60-7550 as references. After thorough screening, we successfully identified the 8-hydroxyl group as the precise site of modification. Utilizing 2-bromo-5-hydroxybenzoic acid as our primary raw material, we synthesized a series of the 8-hydroxyl modified UA. Subsequently, we evaluated the inhibitory activity of these synthesized UA derivatives using a phosphodiesterase assay kit. Ultimately, we screened a total of 34 derivatives; among them, compounds 1f, 1q, 2d, and 2j exhibited significant inhibitory activity against PDE2 with half-maximal inhibitory concentrations of 3.05 μM, 0.67 μM, 0.57 μM, and 4.96 μM, respectively.
Collapse
Affiliation(s)
- Feng Zhou
- School of Pharmacy & School of Biological and Food Engineering, Changzhou University, Changzhou 213164, China.
| | - Xiaoqing Feng
- School of Pharmacy & School of Biological and Food Engineering, Changzhou University, Changzhou 213164, China.
| | - Zhongqiu Xu
- School of Pharmacy & School of Biological and Food Engineering, Changzhou University, Changzhou 213164, China.
| | - Fen Yan
- School of Pharmacy & School of Biological and Food Engineering, Changzhou University, Changzhou 213164, China.
| | - Guoqiang Song
- School of Pharmacy & School of Biological and Food Engineering, Changzhou University, Changzhou 213164, China.
| | - Long Tang
- School of Pharmacy & School of Biological and Food Engineering, Changzhou University, Changzhou 213164, China.
| |
Collapse
|
16
|
Luo Y, Lan C, Ren W, Wu A, Yu B, He J, Chen D. Bacteroides thetaiotaomicron: A symbiotic ally against diarrhea along with modulation of gut microbial ecological networks via tryptophan metabolism and AHR-Nrf2 signaling. J Adv Res 2025:S2090-1232(25)00260-7. [PMID: 40233891 DOI: 10.1016/j.jare.2025.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2024] [Revised: 02/01/2025] [Accepted: 04/11/2025] [Indexed: 04/17/2025] Open
Abstract
INTRODUCTION Bacteroides is a crucial mucosal symbiotic bacterium in mammals, with Bacteroides thetaiotaomicron (B. thetaiotaomicron) being particularly noteworthy as a glyco-specialist due to its significant nutritional impact. However, the potential effects of B. thetaiotaomicron on host health remain underexplored. OBJECTIVES This study aimed to investigate the patterns of microbial community changes and the molecular mechanisms mediated by microbial metabolites in alleviating piglet diarrhea through B. thetaiotaomicron intervention. METHODS Cold stress was induced in piglets to trigger stress-induced diarrhea. The control group and B group were administered a blank medium and 1 × 108 CFU of B. thetaiotaomicron, respectively, on days 1, 3, and 5. The diarrhea rate and growth performance of the piglets were recorded during the experimental period. Based on 16S rRNA gene amplicon sequencing, microbial ecological networks analysis, and metabolomics analysis, the composition and changes of the colonic microbiota and metabolites were analyzed. The antibacterial capacity and anti-inflammatory molecular mechanisms of B. thetaiotaomicron metabolites were analyzed through in vitro antibacterial assays and inflammatory cell models. RESULTS B. thetaiotaomicron administration alleviated diarrhea and improved the growth performance of piglets. It modulated the composition and interactions of the intestinal microbiota, with microbial metabolites primarily enriched in the tryptophan metabolism pathway-especially indole and its derivatives, which were closely associated with host phenotypes. In vitro co-culture experiments showed that B. thetaiotaomicron metabolites inhibited the growth of pathogenic bacteria. Further experiments demonstrated that these metabolites, including indole, enhanced epithelial barrier function and attenuated TNF-α-induced inflammation and apoptosis in Caco-2 cells, highlighting the involvement of the AHR-Nrf2 signaling pathway in mediating these protective effects. CONCLUSION In conclusion, this study offers a theoretical framework for understanding the role of the symbiotic bacterium B. thetaiotaomicron in the gut microbiota ecosystem during diarrhea and its interactions with the host's intestinal tract.
Collapse
Affiliation(s)
- Yuheng Luo
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education of China, Key Laboratory for Animal Disease-Resistance Nutrition and Feed of Ministry of Agriculture of China, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Engineering Research Center of Animal Disease-Resistance Nutrition Biotechnology of Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Cong Lan
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education of China, Key Laboratory for Animal Disease-Resistance Nutrition and Feed of Ministry of Agriculture of China, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Engineering Research Center of Animal Disease-Resistance Nutrition Biotechnology of Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Wen Ren
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education of China, Key Laboratory for Animal Disease-Resistance Nutrition and Feed of Ministry of Agriculture of China, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Engineering Research Center of Animal Disease-Resistance Nutrition Biotechnology of Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Aimin Wu
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education of China, Key Laboratory for Animal Disease-Resistance Nutrition and Feed of Ministry of Agriculture of China, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Engineering Research Center of Animal Disease-Resistance Nutrition Biotechnology of Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Bing Yu
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education of China, Key Laboratory for Animal Disease-Resistance Nutrition and Feed of Ministry of Agriculture of China, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Engineering Research Center of Animal Disease-Resistance Nutrition Biotechnology of Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Jun He
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education of China, Key Laboratory for Animal Disease-Resistance Nutrition and Feed of Ministry of Agriculture of China, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Engineering Research Center of Animal Disease-Resistance Nutrition Biotechnology of Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China
| | - Daiwen Chen
- Key Laboratory for Animal Disease-Resistance Nutrition of Ministry of Education of China, Key Laboratory for Animal Disease-Resistance Nutrition and Feed of Ministry of Agriculture of China, Key Laboratory of Animal Disease-Resistant Nutrition of Sichuan Province, Engineering Research Center of Animal Disease-Resistance Nutrition Biotechnology of Ministry of Education of China, Animal Nutrition Institute, Sichuan Agricultural University, Chengdu, China.
| |
Collapse
|
17
|
Mishra S, Jain S, Agadzi B, Yadav H. A Cascade of Microbiota-Leaky Gut-Inflammation- Is it a Key Player in Metabolic Disorders? Curr Obes Rep 2025; 14:32. [PMID: 40208464 DOI: 10.1007/s13679-025-00624-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/21/2025] [Indexed: 04/11/2025]
Abstract
PURPOSE OF REVIEW This review addresses critical gaps in knowledge and provides a literature overview of the molecular pathways connecting gut microbiota dysbiosis to increased intestinal permeability (commonly referred to as "leaky gut") and its contribution to metabolic disorders. Restoring a healthy gut microbiota holds significant potential for enhancing intestinal barrier function and metabolic health. These interventions offer promising therapeutic avenues for addressing leaky gut and its associated pathologies in metabolic syndrome. RECENT FINDINGS In metabolic disorders such as obesity and type 2 diabetes (T2D), beneficial microbes such as those producing short-chain fatty acids (SCFAs) and other key metabolites like taurine, spermidine, glutamine, and indole derivatives are reduced. Concurrently, microbes that degrade toxic metabolites such as ethanolamine also decline, while proinflammatory, lipopolysaccharide (LPS)-enriched microbes increase. These microbial shifts place a higher burden on intestinal epithelial cells, which are in closest proximity to the gut lumen, inducing detrimental changes that compromise the structural and functional integrity of the intestinal barrier. Such changes include exacerbation of tight junction protein (TJP)s dysfunction, particularly through mechanisms such as destabilization of zona occludens (Zo)-1 mRNA or post-translational modifications. Emerging therapeutic strategies including ketogenic and Mediterranean diets, as well as probiotics, prebiotics, synbiotics, and postbiotics have demonstrated efficacy in restoring beneficial microbial populations, enhancing TJP expression and function, supporting gut barrier integrity, reducing leaky gut and inflammation, and ultimately improving metabolic disorders. This review summarizes the mechanisms by which gut microbiota contribute to the development of leaky gut and inflammation associated with metabolic syndrome. It also explores strategies for restoring gut microbiota balance and functionality by promoting beneficial microbes, increasing the production of beneficial metabolites, clearing toxic metabolites, and reducing the proportion of proinflammatory microbes. These approaches can alleviate the burden on intestinal epithelial cells, reduce leaky gut and inflammation, and improve metabolic health.
Collapse
Affiliation(s)
- Sidharth Mishra
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Center for Excellence of Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Byrd Alzheimer's Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Shalini Jain
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Center for Excellence of Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Byrd Alzheimer's Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Bryan Agadzi
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Hariom Yadav
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
- Center for Excellence of Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
- Byrd Alzheimer's Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
- Director of USF Center for Microbiome Research, Microbiomes Institute, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd, MDC78, Tampa, FL, 33612, USA.
| |
Collapse
|
18
|
Cao Y, Xiao S, Fang Y, Yang J, Hu Z, Zhang H, Liu X, Liu D, Zhou Z, Wang P. Fluxapyroxad induces chronic colonic inflammation via inhibiting intestinal aryl hydrocarbon receptors in mice. THE SCIENCE OF THE TOTAL ENVIRONMENT 2025; 973:179134. [PMID: 40112552 DOI: 10.1016/j.scitotenv.2025.179134] [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: 02/13/2025] [Revised: 03/11/2025] [Accepted: 03/11/2025] [Indexed: 03/22/2025]
Abstract
Fluxapyroxad, the most extensively utilized succinate dehydrogenase inhibitor (SDHI) fungicide, lacks comprehensive research on potential risks associated with chronic toxicity. To investigate its effects on chronic colonic inflammation and elucidate the underlying mechanisms, a mouse model was employed to assess oral exposure to fluxapyroxad at no observed adverse effect level (NOEL) for 13 weeks, in vitro and in silico models were utilized as well. The results revealed reduced body weight gain, colon length reduction, crypt damage, goblet cell loss in the colon, impaired intestinal barrier integrity, and an elevation of proinflammatory cytokines, including IL-6, IL-1β, and TNF-α following fluxapyroxad exposure in mice. These findings suggested that fluxapyroxad induced chronic colonic inflammation. Furthermore, fluxapyroxad decreased interleukin 22 levels and antibacterial peptide secretion by inhibiting Aryl hydrocarbon receptors (AhR) activation, which was confirmed in vitro experiments. Molecular docking analysis indicated that fluxapyroxad spontaneously formed halogen bonds and bound hydrophobic interactions with AhR, which might act as an AhR inhibitor. These results indicated that AhR inhibition may represent one of the primary mechanisms for chronic colonic inflammation induced by fluxapyroxad exposure. This study shed light on the association between low acute pesticide exposure to fluxapyroxad and chronic colonic inflammation development while contributing to pesticide safety assessment.
Collapse
Affiliation(s)
- Yue Cao
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, PR China.
| | - Shouchun Xiao
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, PR China.
| | - Yaofeng Fang
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, PR China
| | - Jiaxing Yang
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, PR China.
| | - Zeyu Hu
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, PR China
| | - Hongjun Zhang
- Institute for the Control of Agrochemicals, Ministry of Agriculture and Rural Affairs, China, No. 22 Maizidian Street, Chaoyang, Beijing 100125, PR China.
| | - Xueke Liu
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, PR China.
| | - Donghui Liu
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, PR China.
| | - Zhiqiang Zhou
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, PR China.
| | - Peng Wang
- Department of Applied Chemistry, College of Science, China Agricultural University, No. 2 West Yuanmingyuan Road, Beijing 100193, PR China.
| |
Collapse
|
19
|
Song Z, Li P, Wu M, Guo S, Wu T, Hou Y, Yi D. Multi-Effects of Natural Plant Bioactive Components on Intestinal Health in Pigs: Promising Feed-Antibiotic Alternatives? J Nutr 2025; 155:1068-1076. [PMID: 39954741 DOI: 10.1016/j.tjnut.2025.02.009] [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] [Revised: 02/03/2025] [Accepted: 02/11/2025] [Indexed: 02/17/2025] Open
Abstract
The poor intestinal health induced by management, stress, or infection remains a substantial challenge restricting the rapid development of the pig industry. Some natural plant bioactive components (NPBCs) have garnered considerable interest owing to their multifarious benefits, including enhancing intestinal morphology, digestion and absorption, barrier function, immune function, and regulating the gut microbiota. However, there are critical factors, such as the lack of standardized production technologies, lower stability and bioavailability, and unclear mechanisms of NPBCs, severely limiting their feeding efficacy and their application in animal production. Here, we conducted a comprehensive review of the recent advances regarding the impacts of NPBCs on pig gut health. Additionally, we highlighted the key areas that warrant further in-depth investigation. Taken together, NPBCs could be green, safe, and effective feed additives by constructively overcoming their limitations, and they are expected to have broader applications in animal husbandry.
Collapse
Affiliation(s)
- Zhuan Song
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, Hubei, China
| | - Peng Li
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, Hubei, China
| | - Mengjun Wu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, Hubei, China
| | - Shuangshuang Guo
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, Hubei, China
| | - Tao Wu
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, Hubei, China
| | - Yongqing Hou
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, Hubei, China
| | - Dan Yi
- Hubei Key Laboratory of Animal Nutrition and Feed Science, Wuhan Polytechnic University, Wuhan, Hubei, China.
| |
Collapse
|
20
|
Cuadrado A, Cazalla E, Bach A, Bathish B, Naidu SD, DeNicola GM, Dinkova-Kostova AT, Fernández-Ginés R, Grochot-Przeczek A, Hayes JD, Kensler TW, León R, Liby KT, López MG, Manda G, Shivakumar AK, Hakomäki H, Moerland JA, Motohashi H, Rojo AI, Sykiotis GP, Taguchi K, Valverde ÁM, Yamamoto M, Levonen AL. Health position paper and redox perspectives - Bench to bedside transition for pharmacological regulation of NRF2 in noncommunicable diseases. Redox Biol 2025; 81:103569. [PMID: 40059038 PMCID: PMC11970334 DOI: 10.1016/j.redox.2025.103569] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/13/2025] [Accepted: 02/24/2025] [Indexed: 03/22/2025] Open
Abstract
Nuclear factor erythroid 2-related factor 2 (NRF2) is a redox-activated transcription factor regulating cellular defense against oxidative stress, thereby playing a pivotal role in maintaining cellular homeostasis. Its dysregulation is implicated in the progression of a wide array of human diseases, making NRF2 a compelling target for therapeutic interventions. However, challenges persist in drug discovery and safe targeting of NRF2, as unresolved questions remain especially regarding its context-specific role in diseases and off-target effects. This comprehensive review discusses the dualistic role of NRF2 in disease pathophysiology, covering its protective and/or destructive roles in autoimmune, respiratory, cardiovascular, and metabolic diseases, as well as diseases of the digestive system and cancer. Additionally, we also review the development of drugs that either activate or inhibit NRF2, discuss main barriers in translating NRF2-based therapies from bench to bedside, and consider the ways to monitor NRF2 activation in vivo.
Collapse
Affiliation(s)
- Antonio Cuadrado
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| | - Eduardo Cazalla
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Anders Bach
- Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, Universitetsparken 2, 2100, Copenhagen, Denmark
| | - Boushra Bathish
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Sharadha Dayalan Naidu
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Gina M DeNicola
- Department of Metabolism and Physiology, H. Lee. Moffitt Cancer Center, Tampa, FL, 33612, USA
| | - Albena T Dinkova-Kostova
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Raquel Fernández-Ginés
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Anna Grochot-Przeczek
- Department of Medical Biotechnology, Faculty of Biochemistry, Biophysics and Biotechnology, Jagiellonian University, Krakow, Poland
| | - John D Hayes
- Jacqui Wood Cancer Centre, Division of Cancer Research, Ninewells Hospital and Medical School, University of Dundee, Dundee, DD1 9SY, Scotland, UK
| | - Thomas W Kensler
- Translational Research Program, Fred Hutchinson Cancer Center, Seattle, WA, 98109, USA
| | - Rafael León
- Instituto de Química Médica, Consejo Superior de Investigaciones Científicas (IQM-CSIC), 28007, Madrid, Spain
| | - Karen T Liby
- Indiana University School of Medicine, Department of Medicine, W. Walnut Street, Indianapolis, IN, 46202, USA
| | - Manuela G López
- Department of Pharmacology, School of Medicine, Universidad Autónoma Madrid, Madrid, Spain; Instituto de Investigación Sanitario (IIS-IP), Hospital Universitario de La Princesa, Madrid, Spain; Instituto Teófilo Hernando, Madrid, Spain
| | - Gina Manda
- Radiobiology Laboratory, Victor Babes National Institute of Pathology, Bucharest, Romania
| | | | - Henriikka Hakomäki
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland
| | - Jessica A Moerland
- Indiana University School of Medicine, Department of Medicine, W. Walnut Street, Indianapolis, IN, 46202, USA
| | - Hozumi Motohashi
- Department of Medical Biochemistry, Graduate School of Medicine Tohoku University, Sendai, Japan; Service of Endocrinology, Diabetology and Metabolism, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Ana I Rojo
- Department of Biochemistry, Medical College, Autonomous University of Madrid (UAM), Madrid, Spain; Instituto de Investigaciones Biomédicas Sols-Morreale (CSIC-UAM), Madrid, Spain; Instituto de Investigación Sanitaria La Paz (IdiPaz), Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | | | - Keiko Taguchi
- Laboratory of Food Chemistry, Department of Applied Biological Chemistry, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Japan; Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Ángela M Valverde
- Instituto de Investigaciones Biomédicas "Sols-Morreale" UAM-CSIC, Instituto de Investigación Sanitaria La Paz (IdiPaz), Centro de Investigación Biomédica en Red de Diabetes y Enfermedades Metabólicas Asociadas (CIBERDEM), ISCIII, Madrid, Spain
| | - Masayuki Yamamoto
- Department of Biochemistry and Molecular Biology, Tohoku Medical Megabank Organization, Tohoku University, Sendai, Japan
| | - Anna-Liisa Levonen
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, Kuopio, Finland.
| |
Collapse
|
21
|
Luo X, Cheng P, Fang Y, Wang F, Mao T, Shan Y, Lu Y, Wei Z. Yinzhihuang formula modulates the microbe‒gut‒liver axis and bile acid excretion to attenuate cholestatic liver injury. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 139:156495. [PMID: 39978276 DOI: 10.1016/j.phymed.2025.156495] [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: 10/09/2024] [Revised: 02/09/2025] [Accepted: 02/09/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND Cholestatic liver injury is a hepatobiliary disorder primarily characterized by cholestasis, which significantly contributes to liver damage. The Yinzhihuang (YZH) oral preparation is an effective clinical treatment for cholestatic liver injury; however, the specific mechanism of action has not been clarified. PURPOSE This study investigated YZH's pharmacological mechanisms associated with the microbe‒gut‒liver axis in cholestatic mice, offering new perspectives for the treatment of cholestasis. METHODS YZH's protective effects were evaluated by evaluating serum liver injury indices and liver staining in an alpha-nephthyl isothiocyanate (ANIT)-induced intrahepatic cholestasis mouse model. Colon hematoxylin‒eosin (H&E) and alcian blue staining and FITC‒dextran leakage assays were performed to assess intestinal barrier integrity. Fluorescence in situ hybridization was employed to analyze bacterial translocation. Additionally, 16S rRNA sequencing, fecal microbiota transplantation, and bile acid metabolomics analysis were conducted to examine the relationships among the microbiome, bile acid metabolism, and YZH formula. RESULTS We found that YZH administration alleviated symptoms of ANIT-induced hepatic pathological injury and fibrosis. In addition, YZH reduced the transfer of gut bacteria to liver tissue by maintaining an intact intestinal barrier. Notably, YZH influenced the intestinal microbiota composition, upregulated the abundance of bile acid metabolism-associated probiotic bacteria, including Clostridiales, Lachnospiraceae and Bifidobacterium pseudolongum; and downregulated the abundance of Escherichia-Shigella and Serratia, thereby promoting bile acid excretion. CONCLUSION YZH protects against cholestatic liver damage by promoting bile excretion and maintaining intestinal mucosal barrier integrity. Furthermore, YZH alleviates cholestasis in a gut microbiota-dependent manner, and upregulation of probiotics may be crucial for YZH's influence on bile acid metabolism.
Collapse
Affiliation(s)
- Xin Luo
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Peng Cheng
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Yuan Fang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Feihui Wang
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Ting Mao
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China
| | - Yunlong Shan
- Key Laboratory of Drug Metabolism and Pharmacokinetics, State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing 210009, PR China.
| | - Yin Lu
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China; Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China; Jiangsu Collaborative Innovation Center of Traditional Chinese Medicine (TCM) Prevention and Treatment of Tumor, Nanjing University of Chinese Medicine, Nanjing 210023, PR China.
| | - Zhonghong Wei
- Jiangsu Key Laboratory for Pharmacology and Safety Evaluation of Chinese Materia Medica, School of Pharmacy, Nanjing University of Chinese Medicine, Nanjing 210023, PR China; Jiangsu Joint International Research Laboratory of Chinese Medicine and Regenerative Medicine, Nanjing University of Chinese Medicine, Nanjing 210023, PR China.
| |
Collapse
|
22
|
Leng P, Wang Y, Xie M. Ellagic Acid and Gut Microbiota: Interactions, and Implications for Health. Food Sci Nutr 2025; 13:e70133. [PMID: 40196228 PMCID: PMC11972986 DOI: 10.1002/fsn3.70133] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2024] [Revised: 03/17/2025] [Accepted: 03/21/2025] [Indexed: 04/09/2025] Open
Abstract
Ellagic acid (EA), a widely distributed natural polyphenolic acid existing in many kinds of plant-based foods, undergoes complex physical and chemical transformations during digestion and biotransformation. Particularly, EA is metabolized by gut microbiota and transformed into urolithins in the colon. These metabolites exhibit enhanced bioavailability and bioactivity. This review explores the intricate interactions between EA and gut microbiota, emphasizing their implications for human health. We discuss the role of gut microbiota in EA metabolism, resulting in distinct metabolic phenotypes associated with varying urolithin production profiles. EA and its gut-derived metabolites, urolithins, have been reported to have the potential to modulate the microbial community composition and function of gut microbiota, promoting beneficial bacteria while reducing harmful ones. Furthermore, EA and urolithins exhibit a spectrum of beneficial biological activities, including antioxidant, anti-inflammatory, and anticancer properties, along with enhancements to intestinal barrier function and modulatory effects on metabolic and cardiovascular systems, through molecular mechanisms such as activating Nrf2 and inhibiting NF-κB pathways. The review highlights and compares the potential of EA and its gut microbial metabolites in the prevention and treatment of various diseases. However, further studies are required to elucidate the underlying mechanisms of the interactions between EA and gut microbiota and their health benefits. Continued investigation into EA and its metabolites is essential for advancing our understanding of their role in promoting human health and developing novel therapeutic applications.
Collapse
Affiliation(s)
- Pinze Leng
- School of MedicineJiangsu UniversityZhenjiangChina
| | - Ye Wang
- Collaborative Innovation Center for Modern Grain Circulation and Safety, College of Food Science and EngineeringNanjing University of Finance and EconomicsNanjingChina
| | - Minhao Xie
- Collaborative Innovation Center for Modern Grain Circulation and Safety, College of Food Science and EngineeringNanjing University of Finance and EconomicsNanjingChina
- Jiangsu Province Engineering Research Center of Edible Fungus Preservation and Intensive ProcessingNanjingChina
| |
Collapse
|
23
|
Ran Z, Mu BR, Wang DM, Xin-Huang, Ma QH, Lu MH. Parkinson's Disease and the Microbiota-Gut-Brain Axis: Metabolites, Mechanisms, and Innovative Therapeutic Strategies Targeting the Gut Microbiota. Mol Neurobiol 2025; 62:5273-5296. [PMID: 39531191 DOI: 10.1007/s12035-024-04584-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Accepted: 10/22/2024] [Indexed: 11/16/2024]
Abstract
The human gut microbiota is diverse and abundant and plays important roles in regulating health by participating in metabolism and controlling physiological activities. The gut microbiota and its metabolites have been shown to affect the functioning of the gut and central nervous system through the microbiota-gut-brain axis. It is well established that microbiota play significant roles in the pathogenesis and progression of Parkinson's disease (PD). Disorders of the intestinal microbiota and altered metabolite levels are closely associated with PD. Here, the changes in intestinal microbiota and effects of metabolites in patients with PD are reviewed. Potential mechanisms underlying intestinal microbiota disorders in the pathogenesis of PD are briefly discussed. Additionally, we outline the current strategies for the treatment of PD that target the gut microbiota, emphasizing the development of promising novel strategies.
Collapse
Affiliation(s)
- Zhao Ran
- Chongqing Key Laboratory of Sichuan-Chongqing Co-Construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Ben-Rong Mu
- Chongqing Key Laboratory of Sichuan-Chongqing Co-Construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Dong-Mei Wang
- College of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China
| | - Xin-Huang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Soochow University, Suzhou, 215021, China
| | - Quan-Hong Ma
- Jiangsu Key Laboratory of Neuropsychiatric Diseases, Institute of Neuroscience, Soochow University, Suzhou, 215021, China.
| | - Mei-Hong Lu
- Chongqing Key Laboratory of Sichuan-Chongqing Co-Construction for Diagnosis and Treatment of Infectious Diseases Integrated Traditional Chinese and Western Medicine, College of Medical Technology, Chengdu University of Traditional Chinese Medicine, Chengdu, 611137, China.
| |
Collapse
|
24
|
Dehghani E, Karimi K, Arekhi S, Ardeshir M, Rezapour R, Shayestehfar M, Memari AH. Effect of nutritional supplements on gut microbiome in individuals with neurodevelopmental disorders: a systematic review and narrative synthesis. BMC Nutr 2025; 11:64. [PMID: 40158118 PMCID: PMC11954342 DOI: 10.1186/s40795-025-01043-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 03/12/2025] [Indexed: 04/01/2025] Open
Abstract
BACKGROUND Neurodevelopmental disorders (NDDs) encompass a range of disruptive conditions with varying prevalence rates and multiple contributing factors. Recent studies have suggested a potential connection between NDDs and the gut-brain axis. Furthermore, there is evidence indicating that nutritional supplements might have an impact on gastrointestinal (GI) and behavioral symptoms. This study aimed to explore the effects of nutritional supplements on the gut microbiota and behavioral symptoms in individuals with NDDs. METHODS A systematic search of databases such as PubMed, Scopus, Web of Science, Embase, and APA PsycINFO was conducted, utilizing relevant keywords until February 2025. In addition, the search for gray literature was carried out on Google Scholar and ProQuest. The risk of bias was assessed using the ROBINS-I tool for non-randomized studies and the RoB-1 tool for randomized controlled trials. Due to the heterogeneity of the studies, a Synthesis without Meta-analysis (SWiM) approach was employed. RESULTS The overall findings from the studies indicated positive effects of supplementation in reducing the Gastrointestinal Severity Index (GIS) score and alleviating GI symptoms. Supplementation with probiotics and vitamins increased good microbiomes (GM) and decrease in bad microbiomes (BM) among individuals with autism spectrum disorder (ASD). Moreover, the Firmicutes to Bacteroidetes ratio (F/R ratio) exhibited significant changes after supplementation. Additionally, improvements were observed in various assessment scores, including ATEC, ABC, CARS, and PGI-2. CONCLUSIONS Nutritional supplementation in individuals with NDDs can have a positive influence by modulating the microbiome, reducing dysbiosis, and enhancing gut barrier integrity. Shifting in the F/R ratio can be considered as the reason for improving gastrointestinal and behavioral symptoms by influencing neurotransmitter activity and neuroinflammation. Targeting the gut-brain axis with interventions that focus on gut microbiota offers a promising adjunct therapy for the management of NDD. Registration of the review protocol. PROSPERO registration no. CRD42023460449.
Collapse
Affiliation(s)
- Elaheh Dehghani
- Department of Clinical Nutrition, School of Nutrition and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Sports Medicine Research Center (SMRC), Neuroscience Institute, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Nutrition and Metabolic Diseases Research Center, Clinical Sciences Research Institute, Ahvaz Jundishapur University of Medical Sciences (AJUMS), Ahvaz, Iran
| | - Keyvan Karimi
- Sports Medicine Research Center (SMRC), Neuroscience Institute, Tehran University of Medical Sciences (TUMS), Tehran, Iran
- Department of Epidemiology and Biostatistics, School of Public Health, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Soheil Arekhi
- Sports Medicine Research Center (SMRC), Neuroscience Institute, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Maryam Ardeshir
- Iranian National Center for Addiction Studies, Tehran University of Medical Sciences, Tehran (TUMS), Tehran, Iran
| | - Reshad Rezapour
- Sports Medicine Research Center (SMRC), Neuroscience Institute, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Monir Shayestehfar
- Sports Medicine Research Center (SMRC), Neuroscience Institute, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Amir Hossein Memari
- Sports Medicine Research Center (SMRC), Neuroscience Institute, Tehran University of Medical Sciences (TUMS), Tehran, Iran.
| |
Collapse
|
25
|
Zhra M, Elahi MA, Tariq A, Abu-Zaid A, Yaqinuddin A. Sirtuins and Gut Microbiota: Dynamics in Health and a Journey from Metabolic Dysfunction to Hepatocellular Carcinoma. Cells 2025; 14:466. [PMID: 40136715 PMCID: PMC11941559 DOI: 10.3390/cells14060466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2025] [Revised: 03/12/2025] [Accepted: 03/13/2025] [Indexed: 03/27/2025] Open
Abstract
Metabolic dysfunction leading to non-alcoholic fatty liver disease (NAFLD) exhibits distinct molecular and immune signatures that are influenced by factors like gut microbiota. The gut microbiome interacts with the liver via a bidirectional relationship with the gut-liver axis. Microbial metabolites, sirtuins, and immune responses are pivotal in different metabolic diseases. This extensive review explores the complex and multifaceted interrelationship between sirtuins and gut microbiota, highlighting their importance in health and disease, particularly metabolic dysfunction and hepatocellular carcinoma (HCC). Sirtuins (SIRTs), classified as a group of NAD+-dependent deacetylases, serve as crucial modulators of a wide spectrum of cellular functions, including metabolic pathways, the inflammatory response, and the process of senescence. Their subcellular localization and diverse functions link them to various health conditions, including NAFLD and cancer. Concurrently, the gut microbiota, comprising diverse microorganisms, significantly influences host metabolism and immune responses. Recent findings indicate that sirtuins modulate gut microbiota composition and function, while the microbiota can affect sirtuin activity. This bidirectional relationship is particularly relevant in metabolic disorders, where dysbiosis contributes to disease progression. The review highlights recent findings on the roles of specific sirtuins in maintaining gut health and their implications in metabolic dysfunction and HCC development. Understanding these interactions offers potential therapeutic avenues for managing diseases linked to metabolic dysregulation and liver pathology.
Collapse
Affiliation(s)
- Mahmoud Zhra
- Department of Anatomy and Genetics, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia;
| | - Muhammad Affan Elahi
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.A.E.); (A.A.-Z.)
| | - Aamira Tariq
- Department of Biosciences, COMSATS University Islamabad, Islamabad Campus, Islamabad 45550, Pakistan
| | - Ahmed Abu-Zaid
- Department of Biochemistry and Molecular Medicine, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia; (M.A.E.); (A.A.-Z.)
| | - Ahmed Yaqinuddin
- Department of Anatomy and Genetics, College of Medicine, Alfaisal University, Riyadh 11533, Saudi Arabia;
| |
Collapse
|
26
|
Wu Y, Xiao W, Xiao B, Wang Y, Li Y, Wu A, Zhang Q, Liu X, Liu S, Yuan Z, Liang Z, Yi J, Wu J. Melatonin Alleviates T-2 Toxin-Induced Intestinal Injury by Enhancing Gut Barrier Function and Modulating Microbiota in Weaned Piglets. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:6903-6916. [PMID: 40048666 DOI: 10.1021/acs.jafc.4c06892] [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
The T-2 toxin, originating from a Fusarium species, is a mycotoxin that can adversely affect animal health. Melatonin (MT) is a natural hormone recognized for its properties that reduce inflammation and act as an antioxidant. However, MT's capacity to alleviate intestinal harm from T-2 toxin remains incompletely explored. Employing postweaning piglets, this research investigates MT's prophylactic impact on T-2 toxin-induced enterotoxicity. The results indicate that MT improved growth performance in piglets exposed to T-2 toxins while also enhancing intestinal barrier function. Such effects probably stem from MT's ability to reduce colonic oxidative stress and inflammation. Further findings suggest that these changes are closely associated with MT-induced remodeling of intestinal microbiota and an increase in short-chain fatty acid (SCFA) levels in the intestine. MT therefore alleviates T-2 toxin intestinal damage; gut microbiota are the key to this process.
Collapse
Affiliation(s)
- You Wu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Wenguang Xiao
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Bo Xiao
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Yongkang Wang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Yuanyuan Li
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Aoao Wu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Qike Zhang
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Xiaofang Liu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Shuiping Liu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Zhihang Yuan
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Zengenni Liang
- Department of Hunan Agricultural Product Processing Institute, Changsha 410128, China
| | - Jine Yi
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| | - Jing Wu
- Hunan Engineering Research Center of Livestock and Poultry Health Care, College of Veterinary Medicine, Hunan Agricultural University, Changsha 410128, China
| |
Collapse
|
27
|
Jin Z, Zhang Y, Hu H, Li Q, Zhang L, Zhao K, Liu W, Li L, Gao C. Closed-loop theranostic microgels for immune microenvironment modulation and microbiota remodeling in ulcerative colitis. Biomaterials 2025; 314:122834. [PMID: 39288617 DOI: 10.1016/j.biomaterials.2024.122834] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 08/26/2024] [Accepted: 09/10/2024] [Indexed: 09/19/2024]
Abstract
Inflammatory bowel disease (IBD) is characterized by the upregulation of reactive oxygen species (ROS) and dysfunction of gut immune system, and microbiota. The conventional treatments mainly focus on symptom control with medication by overuse of drugs. There is an urgent need to develop a closed-loop strategy that combines in situ monitoring and precise treatment. Herein, we innovatively designed the 'cluster munition structure' theranostic microgels to realize the monitoring and therapy for ulcerative colitis (a subtype of IBD). The superoxide anion specific probe (tetraphenylethylene-coelenterazine, TPC) and ROS-responsive nanogels consisting of postbiotics urolithin A (UA) were loaded into alginate and ion-crosslinked to obtain the theranostic microgels. The theranostic microgels could be delivered to the inflammatory site, where the environment-triggered breakup of the microgels and release of the nanogels were achieved in sequence. The TPC-UA group had optimal results in reducing inflammation, repairing colonic epithelial tissue, and remodeling microbiota, leading to inflammation amelioration and recovery of tight junction between the colonic epithelium, and maintenance of gut microbiota. During the recovery process, the local chemiluminescence intensity, which is proportional to the degree of inflammation, was gradually inhibited. The cluster munition of theranostic microgels displayed promising outcomes in monitoring inflammation and precise therapy, and demonstrated the potential for inflammatory disease management.
Collapse
Affiliation(s)
- Zeyuan Jin
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China.
| | - Yaqi Zhang
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Haijun Hu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Qian Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China
| | - Liwen Zhang
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Kefei Zhao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China
| | - Wenxing Liu
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China; Center for Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing, 312099, China.
| | - Lanjuan Li
- State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, National Medical Center for Infectious Diseases, Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, The First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, 310003, China.
| | - Changyou Gao
- MOE Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, 310058, China; Center for Healthcare Materials, Shaoxing Institute, Zhejiang University, Shaoxing, 312099, China; Dr. Li Dak Sum & Yip Yio Chin Center for Stem Cell and Regenerative Medicine, Zhejiang University, Hangzhou, 310058, China.
| |
Collapse
|
28
|
Barreira-Silva P, Lian Y, Kaufmann SHE, Moura-Alves P. The role of the AHR in host-pathogen interactions. Nat Rev Immunol 2025; 25:178-194. [PMID: 39415055 DOI: 10.1038/s41577-024-01088-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/28/2024] [Indexed: 10/18/2024]
Abstract
Host-microorganism encounters take place in many different ways and with different types of outcomes. Three major types of microorganisms need to be distinguished: (1) pathogens that cause harm to the host and must be controlled; (2) environmental microorganisms that can be ignored but must be controlled at higher abundance; and (3) symbiotic microbiota that require support by the host. Recent evidence indicates that the aryl hydrocarbon receptor (AHR) senses and initiates signalling and gene expression in response to a plethora of microorganisms and infectious conditions. It was originally identified as a receptor that binds xenobiotics. However, it was subsequently found to have a critical role in numerous biological processes, including immunity and inflammation and was recently classified as a pattern recognition receptor. Here we review the role of the AHR in host-pathogen interactions, focusing on AHR sensing of different microbial classes, the ligands involved, responses elicited and disease outcomes. Moreover, we explore the therapeutic potential of targeting the AHR in the context of infection.
Collapse
Affiliation(s)
- Palmira Barreira-Silva
- IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal
| | - Yilong Lian
- Ludwig Institute for Cancer Research, Nuffield Department of Clinical Medicine, University of Oxford, Oxford, UK
| | - Stefan H E Kaufmann
- Max Planck Institute for Infection Biology, Berlin, Germany
- Max Planck Institute for Multidisciplinary Sciences, Göttingen, Germany
- Hagler Institute for Advanced Study, Texas A&M University, College Station, TX, USA
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
| | - Pedro Moura-Alves
- IBMC, Instituto de Biologia Molecular e Celular, Universidade do Porto, Porto, Portugal.
- i3S, Instituto de Investigação e Inovação em Saúde, Universidade do Porto, Porto, Portugal.
| |
Collapse
|
29
|
Huchzermeier R, van der Vorst EPC. Aryl hydrocarbon receptor (AHR) and nuclear factor erythroid-derived 2-like 2 (NRF2): An important crosstalk in the gut-liver axis. Biochem Pharmacol 2025; 233:116785. [PMID: 39890034 DOI: 10.1016/j.bcp.2025.116785] [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: 10/28/2024] [Revised: 12/18/2024] [Accepted: 01/27/2025] [Indexed: 02/03/2025]
Abstract
The aryl hydrocarbon receptor (AHR) is a ligand-activated transcription factor, mainly involved in detoxification. However, in the intestine, metabolites derived from the diet, which are converted by a wide range of bacteria can also activate the AHR. This intestinal AHR activation plays a key role in maintaining the gut barrier by, for example, upregulating antimicrobial peptides and anti-inflammatory cytokines. Since the gut barrier influences the gut-liver axis by regulating the leaking of metabolites, bacteria, and endotoxins into circulation and particularly into the liver, the AHR is a key factor in the gut-liver axis. Vice versa, certain liver pathologies also influence the gut microbiome, thereby altering bacteria-derived activation of the AHR. Additionally, bile acids can impact the gut via the liver and thereby also affect the AHR. The aryl hydrocarbon receptor (AHR) interacts with several molecular factors, one of which is the nuclear factor erythroid-derived 2-like 2 (NRF2), a transcription factor primarily associated with regulating antioxidant stress responses. The interplay between AHR and NRF2 has been investigated in the context of various diseases; this review highlights the significance of this interaction within the framework of the gut-liver axis.
Collapse
Affiliation(s)
- Rosanna Huchzermeier
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany; Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany; Department of Internal Medicine I, University Hospital Aachen, RWTH Aachen University, 52074 Aachen, Germany
| | - Emiel P C van der Vorst
- Institute for Molecular Cardiovascular Research (IMCAR), RWTH Aachen University, 52074 Aachen, Germany; Aachen-Maastricht Institute for CardioRenal Disease (AMICARE), RWTH Aachen University, 52074 Aachen, Germany; Department of Internal Medicine I, University Hospital Aachen, RWTH Aachen University, 52074 Aachen, Germany; Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximilians-University Munich, 80336 Munich, Germany.
| |
Collapse
|
30
|
Tian Y, Cheng J, Yang Y, Wang H, Fu Y, Li X, Wang W, Ma S, Xu X, Lu F, Feng P, Han S, Chen H, Hou H, Hu Q, Wu C. A 90-Day Subchronic Exposure to Heated Tobacco Product Aerosol Caused Differences in Intestinal Inflammation and Microbiome Dysregulation in Rats. Nicotine Tob Res 2025; 27:438-446. [PMID: 39028556 DOI: 10.1093/ntr/ntae179] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2023] [Revised: 06/17/2024] [Accepted: 07/14/2024] [Indexed: 07/20/2024]
Abstract
INTRODUCTION Smoking is one of the most important predisposing factors of intestinal inflammatory diseases. Heated tobacco product (HTP) is a novel tobacco category that is claimed to deliver reduced chemicals to humans those reported in combustible cigarette smoke (CS). However, the effect of HTP on the intestine is still unknown. AIMS AND METHODS Our study aims to explore the potential effects of HTP on intestine. In the framework of Organization for Economic Co-operation and Development guidelines 413 guidelines, Sprague-Dawley rats were exposed to HTP aerosol and CS for 13 weeks. The atmosphere was characterized and oxidative stress and inflammation of the intestine were investigated after exposure. Furthermore, the feces we performed with 16S sequencing and metabolomics analysis. RESULTS HTP aerosol and CS led to obvious intestinal damage evidenced by increased intestinal proinflammatory cytokines and oxidative stress in male and female rats After HTP and CS exposure, the abundance that obviously changed were Lactobacillus and Turiciacter in male rats and Lactobacillus and Prevotella in female rats. HTP mainly induces the metabolism of amino acids and fatty acyls such as short-chain fatty acids and tryptophan, while CS is involved in the main metabolism of bile acids, especially indole and derivatives. Although different metabolic pathways in the gut are mediated by HTP and CS, both inflammation and oxidative stress were ultimately induced. CONCLUSIONS HTP aerosol and CS-induced intestinal damage are mediated by different gut microbiota and metabolites, while both lead to inflammation and oxidative stress. IMPLICATIONS The concentration of various harmful components in heated tobacco product aerosol is reported lower than that of traditional cigarette smoke, however, its health risk impact on consumers remains to be studied. Our research findings indicate that heated tobacco products and cigarette smoke inhalation induced intestinal damage through different metabolic pathways mediated by the gut microbiome, indicating the health risk of heated tobacco products in the intestine.
Collapse
Affiliation(s)
- Yushan Tian
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Jiale Cheng
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Yanan Yang
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Hongjuan Wang
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Yaning Fu
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Xianmei Li
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Wenming Wang
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Shuhao Ma
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Xiaoxiao Xu
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Fengjun Lu
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Pengxia Feng
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Shulei Han
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Huan Chen
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Hongwei Hou
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Qingyuan Hu
- Quality Satety and FCTC Research Team, China National Tobacco Quality Supervision and Test Center, Zhengzhou, China
- Risk Assessment and Awareness Team, Key Laboratory of Tobacco Biological Effects, Zhengzhou, China
- Research Division 1, Beijing Life Science Academy, Beijing, China
- Research Division 1, Key Laboratory of Tobacco Biological Effects and Biosynthesis, Beijing, China
| | - Chongming Wu
- School of Chinese Materia Medica, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
31
|
Chen K, Ying J, Zhu J, Chen L, Liu R, Jing M, Wang Y, Zhou K, Wu L, Wu C, Xiao J, Ni W. Urolithin A alleviates NLRP3 inflammasome activation and pyroptosis by promoting microglial mitophagy following spinal cord injury. Int Immunopharmacol 2025; 148:114057. [PMID: 39827665 DOI: 10.1016/j.intimp.2025.114057] [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: 09/05/2024] [Revised: 01/02/2025] [Accepted: 01/06/2025] [Indexed: 01/22/2025]
Abstract
Spinal cord injury (SCI) is a potentially fatal condition that often results in loss of motor and sensory functions, thereby significantly burdening global health initiatives. Urolithin A (UA), an intestinal microbial metabolite of ellagic acid, is known for its potent anti-inflammatory properties in chronic inflammation contexts. UA treatment in humans induces a molecular signature of improved mitochondrial and cellular health. Yet, its effects on acute inflammation following SCI remain unclear. In this study, we developed an impact-induced mouse model for SCI and treated the injured mice with UA (50 mg/kg/d, till 8 weeks) via intragastric administration. Furthermore, we subjected BV2 cells to lipopolysaccharide and adenosine 5'-triphosphate to simulate the post-injury inflammatory response. Our results demonstrated that pre-treatment with UA (10 μM) effectively inhibited NLRP3 inflammasome activation in LPS-primed BV2 cells. This inhibition was evidenced by reduced cleaved Caspase-1 and mature IL-1β release, diminished ASC speck formation, and decreased gasdermin D (GSDMD)-mediated pyroptosis. Additionally, UA treatment restored mitochondrial activity and ROS production attenuated by NLRP3 activation, increased LC3-II expression, and enhanced LC3 co-localization with mitochondria. 3-Methyladenine (3-MA), an autophagy inhibitor, can partially reverse the stimulatory effect of UA on mitophagy, as well as the inhibitory effect of UA on pyroptosis. This study highlighted the protective role of UA against SCI through its promotion of mitophagy, which in turn inhibits NLRP3 inflammasome activation and pyroptosis.
Collapse
Affiliation(s)
- Kongbin Chen
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000 China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000 China
| | - Jiahao Ying
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000 China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000 China
| | - Jiangwei Zhu
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000 China
| | - Liang Chen
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000 China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000 China
| | - Rongjie Liu
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000 China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000 China
| | - Mengqi Jing
- Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000 China
| | - Yuchao Wang
- Department of Orthopedic, The Second Affiliated Hospital of Dalian Medical University, Dalian, Liaoning 116600, China
| | - Kailiang Zhou
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000 China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000 China
| | - Long Wu
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000 China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000 China.
| | - Chenyu Wu
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000 China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000 China; Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000 China.
| | - Jian Xiao
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000 China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000 China; Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000 China.
| | - Wenfei Ni
- Department of Orthopedic, The Second Affiliated Hospital and Yuying Children's Hospital of Wenzhou Medical University, Wenzhou 325000 China; Key Laboratory of Orthopaedics of Zhejiang Province, Wenzhou 325000 China; Molecular Pharmacology Research Center, School of Pharmaceutical Science, Wenzhou Medical University, Wenzhou 325000 China.
| |
Collapse
|
32
|
Kumar S, Mukherjee R, Gaur P, Leal É, Lyu X, Ahmad S, Puri P, Chang CM, Raj VS, Pandey RP. Unveiling roles of beneficial gut bacteria and optimal diets for health. Front Microbiol 2025; 16:1527755. [PMID: 40041870 PMCID: PMC11877911 DOI: 10.3389/fmicb.2025.1527755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2024] [Accepted: 02/03/2025] [Indexed: 04/02/2025] Open
Abstract
The gut microbiome plays a pivotal role in human health, influencing digestion, immunity, and disease prevention. Beneficial gut bacteria such as Akkermansia muciniphila, Adlercreutzia equolifaciens, and Christensenella minuta contribute to metabolic regulation and immune support through bioactive metabolites like short-chain fatty acids (SCFAs). Dietary patterns rich in prebiotics, fermented foods, and plant-based bioactive compounds, including polyphenols and flavonoids, promote microbiome diversity and stability. However, challenges such as individual variability, bioavailability, dietary adherence, and the dynamic nature of the gut microbiota remain significant. This review synthesizes current insights into gut bacteria's role in health, emphasizing the mechanisms by which dietary interventions modulate microbiota. Additionally, it highlights advancements in microbiome-targeted therapies and the transformative potential of personalized nutrition, leveraging microbiota profiling and artificial intelligence (AI) to develop tailored dietary strategies for optimizing gut health and mitigating chronic inflammatory disorders. Addressing these challenges requires a multidisciplinary approach that integrates scientific innovation, ethical frameworks, and practical implementation strategies.
Collapse
Affiliation(s)
- Suresh Kumar
- National Institute of Biologicals, Ministry of Health & Family Welfare, Govt. of India, Noida, India
| | - Riya Mukherjee
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Taoyuan, Taiwan
| | - Pratibha Gaur
- Centre for Drug Design Discovery and Development (C4D), SRM University Delhi-NCR, Sonepat, India
- Department of Biotechnology and Microbiology, SRM University Delhi-NCR, Sonepat, India
| | - Élcio Leal
- Laboratório de Diversidade Viral, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, Brazil
| | - Xiaoming Lyu
- Department of Laboratory Medicine, The Third Affiliated Hospital, Southern Medical University, Guangzhou, China
| | - Saheem Ahmad
- Department of Medical Laboratory Sciences, College of Applied Medical Sciences, University of Hail, Ha'il, Saudi Arabia
| | - Paridhi Puri
- University Centre for Research and Development, Chandigarh University, Mohali, India
| | - Chung-Ming Chang
- Graduate Institute of Biomedical Sciences, Chang Gung University, Taoyuan, Taiwan
- Department of Medical Biotechnology and Laboratory Science, Chang Gung University, Taoyuan, Taiwan
- Master & Ph.D Program in Biotechnology Industry, Chang Gung University, Taoyuan, Taiwan
| | - V. Samuel Raj
- Department of Biotechnology and Microbiology, SRM University Delhi-NCR, Sonepat, India
- Laboratório de Diversidade Viral, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, Brazil
| | - Ramendra Pati Pandey
- Department of Biotechnology and Microbiology, SRM University Delhi-NCR, Sonepat, India
- Laboratório de Diversidade Viral, Instituto de Ciências Biológicas, Universidade Federal Do Pará, Belém, Brazil
| |
Collapse
|
33
|
Foata F, Duboux S, Herzig S, Sizzano F, Thevenet J, Guy P, Rezzi S, Métairon S, Bourqui B, Montoliu I, Mercenier A, Bosco N. Identification and Biological Characterization of a Novel NRF2 Activator Molecule Released From the Membranes of Heat-Treated Bifidobacterium breve NCC 2950. Mol Nutr Food Res 2025:e202400770. [PMID: 39911038 DOI: 10.1002/mnfr.202400770] [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: 09/20/2024] [Revised: 12/16/2024] [Accepted: 01/14/2025] [Indexed: 02/07/2025]
Abstract
Postbiotics are defined as a "preparation of inanimate microorganisms and/or their components that confers a health benefit on the host". They represent an attractive alternative to probiotics as they could be used in a broader range of applications, where probiotic stability is limiting. To date knowledge on the mechanism of action of inanimate microorganisms is relatively scarce. In this study, we investigated the impact of heat treatment on NRF2 activation by several candidate probiotic strains from the Nestlé Culture Collection (NCC), including species encompassed in the Bifidobacterium genus and the Lactobacillaceae family. We identified an NRF2-activating bioactive molecule, 4-oxo-2-pentenoic acid (OPA), specifically released during heat treatment of Bifidobacterium breve NCC 2950. We explored cellular pathways that can be modulated by OPA, such as antiinflammatory signals and organismal defense against oxidative stress in zebrafish in vivo. We identified a new B. breve NCC 2950-derived postbiotic that, based on the mode of action, may have important applications for nutritional strategies to benefit human health.
Collapse
Affiliation(s)
- Francis Foata
- Clinical Research Unit, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Stéphane Duboux
- Nestlé Institute of Food Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Sébastien Herzig
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Federico Sizzano
- Nestlé Institute of Food Safety and Analytical Science, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Jonathan Thevenet
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Philippe Guy
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Serge Rezzi
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Sylviane Métairon
- Nestlé Institute of Food Safety and Analytical Science, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Bertrand Bourqui
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Ivan Montoliu
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Annick Mercenier
- Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Nabil Bosco
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| |
Collapse
|
34
|
Xiao Y, Feng Y, Zhao J, Chen W, Lu W. Achieving healthy aging through gut microbiota-directed dietary intervention: Focusing on microbial biomarkers and host mechanisms. J Adv Res 2025; 68:179-200. [PMID: 38462039 PMCID: PMC11785574 DOI: 10.1016/j.jare.2024.03.005] [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: 12/29/2023] [Revised: 02/23/2024] [Accepted: 03/07/2024] [Indexed: 03/12/2024] Open
Abstract
BACKGROUND Population aging has become a primary global public health issue, and the prevention of age-associated diseases and prolonging healthy life expectancies are of particular importance. Gut microbiota has emerged as a novel target in various host physiological disorders including aging. Comprehensive understanding on changes of gut microbiota during aging, in particular gut microbiota characteristics of centenarians, can provide us possibility to achieving healthy aging or intervene pathological aging through gut microbiota-directed strategies. AIM OF REVIEW This review aims to summarize the characteristics of the gut microbiota associated with aging, explore potential biomarkers of aging and address microbiota-associated mechanisms of host aging focusing on intestinal barrier and immune status. By summarizing the existing effective dietary strategies in aging interventions, the probability of developing a diet targeting the gut microbiota in future is provided. KEY SCIENTIFIC CONCEPTS OF REVIEW This review is focused on three key notions: Firstly, gut microbiota has become a new target for regulating health status and lifespan, and its changes are closely related to age. Thus, we summarized aging-associated gut microbiota features at the levels of key genus/species and important metabolites through comparing the microbiota differences among centenarians, elderly people and younger people. Secondly, exploring microbiota biomarkers related to aging and discussing future possibility using dietary regime/components targeted to aging-related microbiota biomarkers promote human healthy lifespan. Thirdly, dietary intervention can effectively improve the imbalance of gut microbiota related to aging, such as probiotics, prebiotics, and postbiotics, but their effects vary among.
Collapse
Affiliation(s)
- Yue Xiao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, PR China.
| | - Yingxuan Feng
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, PR China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, PR China
| | - Wenwei Lu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, PR China; School of Food Science and Technology, Jiangnan University, Wuxi 214122, PR China; National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, PR China.
| |
Collapse
|
35
|
Li X, Duan W, Zhu Y, Ji R, Feng K, Kathuria Y, Xiao H, Yu Y, Cao Y. Transcriptomics and metabolomics reveal the alleviation effect of pectic polysaccharide on dextran sodium sulfate-induced colitis mice. Int J Biol Macromol 2025; 288:138755. [PMID: 39674473 DOI: 10.1016/j.ijbiomac.2024.138755] [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: 10/03/2024] [Revised: 12/04/2024] [Accepted: 12/11/2024] [Indexed: 12/16/2024]
Abstract
Ulcerative colitis (UC) is a relapsing disease with an increasing morbidity and prevalence. Dietary polysaccharides have recently become a research hotspot because of their therapeutic effects and safety on UC. Our previous research elucidated that pectic polysaccharide from Phyllanthus emblica L. (PEP-1) could alleviate dextran sodium sulfate-induced UC mice. Herein, metabolomics and transcriptomics were further applied to disclose the underlying mechanisms behind PEP-1's anti-inflammatory effects. PEP-1 intervention altered the serum metabolite contents and pathways represented by decreasing xanthine and sphinganine levels. Changes in gene expressions correlated with metabolite variations led by the suppression of the expression of the inflammatory factors, colorectal cancer promoter, and NF-κB pathway as well as the enhancement of tight junctions. This study demonstrated that the ameliorating effect of chronic UC was partially ascribed to the alteration of the serum metabolites and changes in gene expression.
Collapse
Affiliation(s)
- Xiaoqing Li
- College of Food Science and Engineering, South China University of Technology, Guangzhou 510006, China; Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China; Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA
| | - Wen Duan
- College of Food Science and Engineering, South China University of Technology, Guangzhou 510006, China
| | - Yi Zhu
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China
| | - Ruya Ji
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA
| | - Konglong Feng
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China; Foshan University, Foshan 528000, China
| | - Yukti Kathuria
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA
| | - Hang Xiao
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA.
| | - Yigang Yu
- College of Food Science and Engineering, South China University of Technology, Guangzhou 510006, China.
| | - Yong Cao
- Guangdong Provincial Key Laboratory of Nutraceuticals and Functional Foods, College of Food Science, South China Agricultural University, Guangzhou 510642, China.
| |
Collapse
|
36
|
Lopez-Rodulfo IM, Stentoft EW, Martinez MM. Comparative assessment of polyphenol bioaccessibility in cold-pressed apple fractions using static and semi-dynamic digestion models. Food Res Int 2025; 202:115743. [PMID: 39967186 DOI: 10.1016/j.foodres.2025.115743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 01/09/2025] [Accepted: 01/10/2025] [Indexed: 02/20/2025]
Abstract
The INFOGEST semi-dynamic digestion model more closely aligns the kinetics of nutrient digestion with structural changes in the food matrix during gastric digestion, which can significantly influence polyphenol bioaccessibility. In this study, the static and semi-dynamic INFOGEST models were compared to assess polyphenol bioaccessibility across various matrix scenarios, using different apple fractions. Each digesta, regardless of the model used, underwent re-solubilization, centrifugal filtration, and UHPLC-ESI-QTOF-MS/MS analysis to approximate transepithelial absorption and facilitate untargeted polyphenol screening and semi-quantification. The semi-dynamic model was initially optimized using whole apple. Overhead stirring with a paddle led to greater browning and degradation of phenolic acids and dihydrochalcones than magnetic stirring, the latter showing bolus stratification and closer physiological conditions for oxygenation and intragastric chyme homogenization. The suitability of a 2 kcal/min gastric emptying rate was tested with pomace, resulting in 8.25 min total gastric emptying time due to low caloric content. Compared to the gastric emptying time of whole apple (139.5 min), the caloric-driven emptying of pomace produced similar polyphenol bioaccessibility but a three-fold higher coefficient of variation (19.5 % vs. 69.4 %). Finally, using several apple fractions, the semi-dynamic setup with magnetic stirring and a fixed gastric emptying rate of 139.5 min showed greater extraction of hydroxybenzoic acids and dihydrochalcones from apple and of hydroxybenzoic and hydroxycinnamic acids from pomace than the static model. However, flavanols in juice degraded more extensively under semi-dynamic conditions. Minimal differences were observed between models for an apple polyphenol extract, indicating that in the absence of matrix, the static setup might be preferred.
Collapse
Affiliation(s)
- Ivan M Lopez-Rodulfo
- Centre for Innovative Food (CiFOOD), Department of Food Science, Aarhus University, Agro Food Park 48, Aarhus N 8200 Denmark
| | - Emil W Stentoft
- Centre for Innovative Food (CiFOOD), Department of Food Science, Aarhus University, Agro Food Park 48, Aarhus N 8200 Denmark
| | - Mario M Martinez
- Centre for Innovative Food (CiFOOD), Department of Food Science, Aarhus University, Agro Food Park 48, Aarhus N 8200 Denmark; Food Technology Area, Department of Agricultural Engineering, University of Valladolid, Spain.
| |
Collapse
|
37
|
Yue N, Zhao H, Hu P, Zhang Y, Tian C, Kong C, Mai Z, Huang L, Luo Q, Wei D, Shi R, Tang S, Nie Y, Liang Y, Yao J, Wang L, Li D. Real-world of Limosilactobacillus reuteri in mitigation of acute experimental colitis. J Nanobiotechnology 2025; 23:65. [PMID: 39891249 PMCID: PMC11783912 DOI: 10.1186/s12951-025-03158-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2024] [Accepted: 01/22/2025] [Indexed: 02/03/2025] Open
Abstract
Probiotics have been proposed as a potential strategy for managing ulcerative colitis (UC). However, the underlying mechanisms mediating microbiota-host crosstalk remain largely elusive. Here, we report that Limosilactobacillus reuteri (L. reuteri), as a probiotic, secretes cytoplasmic membrane vesicles (CMVs) that communicate with host cells, alter host physiology, and alleviate dextran sulfate sodium (DSS)-induced colitis. First, L. reuteri-CMVs selectively promoted the proliferation of the beneficial bacterium Akkermansia muciniphila (AKK) by upregulating the expression of glycosidases (beta-N-acetylhexosaminidase and alpha-N-acetylglucosaminidase) involved in glycan degradation and metabolic pathways and restored the disrupted gut microbiota balance. Second, L. reuteri-CMVs were taken up by intestinal epithelial cells (IECs), elevated the expression of ZO-1, E-cadherin (Cdh1), and Occludin (Ocln), decreased intestinal permeability, and exerted protective effects on epithelial tight junction functionality. RNA sequencing analysis demonstrated that L. reuteri-CMVs repaired intestinal barrier by activating the HIF-1 signaling pathway and upregulating HMOX1 expression. Third, L. reuteri-CMVs increased the population of double positive (DP) CD4+CD8+ T cells in the intestinal epithelial layer, suppressing gut inflammation and maintaining gut mucosal homeostasis. Finally, L. reuteri-CMVs exhibited satisfactory stability and safety in the gastrointestinal tract and specifically targeted the desired sites in colitis mice. Collectively, these findings shed light on how L. reuteri interact with the host in colitis, and provide new insights into potential strategies for alleviating colitis.
Collapse
Affiliation(s)
- Ningning Yue
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518020, China
| | - Hailan Zhao
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518020, China
- Department of Gastroenterology, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, 510630, China
| | - Peng Hu
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai, 200093, China
| | - Yuan Zhang
- Department of Medical Administration, Huizhou Institute of Occupational Diseases Control and Prevention, Huizhou, Guangdong, 516008, China
| | - Chengmei Tian
- Department of Emergency, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, 518020, China
| | - Chen Kong
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518020, China
| | - Zhiliang Mai
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518020, China
| | - Longbin Huang
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518020, China
| | - Qianjun Luo
- Department of Endocrine, Shenzhen Qianhai Shekou Free Trade Zone Hospital, Shenzhen, Guangdong, 518067, China
| | - Daoru Wei
- Department of Rehabilitation, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, 518020, China
| | - Ruiyue Shi
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518020, China
| | - Shaohui Tang
- Department of Gastroenterology, The First Affiliated Hospital, Jinan University, Guangzhou, Guangdong, 510630, China
| | - Yuqiang Nie
- Department of Gastroenterology and Hepatology, Guangzhou Digestive Disease Center, Guangzhou First People's Hospital, School of Medicine, South China University of Technology, Guangzhou, Guangdong, 510180, China
| | - Yujie Liang
- Department of Child and Adolescent Psychiatry, Shenzhen Kangning Hospital, Shenzhen Mental Health Center, Shenzhen, Guangdong, 518020, China.
| | - Jun Yao
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518020, China.
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, 518020, China.
| | - Lisheng Wang
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518020, China.
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, 518020, China.
| | - Defeng Li
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University), Shenzhen, Guangdong, 518020, China.
- Department of Gastroenterology, Shenzhen People's Hospital (The Second Clinical Medical College, Jinan University; The First Affiliated Hospital, Southern University of Science and Technology), Shenzhen, Guangdong, 518020, China.
| |
Collapse
|
38
|
Pidgeon R, Mitchell S, Shamash M, Suleiman L, Dridi L, Maurice CF, Castagner B. Diet-derived urolithin A is produced by a dehydroxylase encoded by human gut Enterocloster species. Nat Commun 2025; 16:999. [PMID: 39856097 PMCID: PMC11760930 DOI: 10.1038/s41467-025-56266-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2024] [Accepted: 01/13/2025] [Indexed: 01/27/2025] Open
Abstract
Urolithin A (uroA) is a polyphenol derived from the multi-step metabolism of dietary ellagitannins by the human gut microbiota. Once absorbed, uroA can trigger mitophagy and aryl hydrocarbon receptor signaling pathways, altering host immune function, mitochondrial health, and intestinal barrier integrity. Most individuals harbor a microbiota capable of uroA production; however, the mechanisms underlying the dehydroxylation of its catechol-containing precursor (uroC) are unknown. Here, we use a combination of untargeted bacterial transcriptomics, proteomics, and comparative genomics to uncover an inducible uroC dehydroxylase (ucd) operon in Enterocloster species. We show that the ucd operon encodes a predicted molybdopterin-dependent enzyme complex that dehydroxylates urolithins at a specific position (9-OH). By interrogating publicly available metagenomics datasets, we observed that uroC-metabolizing Enterocloster species and ucd operon genes are prevalent in human feces. In ex vivo experiments with human fecal samples, only samples actively transcribing ucd could produce uroA, possibly explaining differences in urolithin metabolism between individuals. Collectively, this work identifies Enterocloster species and the ucd operon as important contributors to uroA production and establishes a multi-omics framework to further our mechanistic understanding of polyphenol metabolism by the human gut microbiota.
Collapse
Affiliation(s)
- Reilly Pidgeon
- Department of Pharmacology & Therapeutics, McGill University, 3655 Prom. Sir-William-Osler, Montreal, Quebec, H3G 1Y6, Canada
| | - Sacha Mitchell
- Department of Pharmacology & Therapeutics, McGill University, 3655 Prom. Sir-William-Osler, Montreal, Quebec, H3G 1Y6, Canada
| | - Michael Shamash
- Department of Microbiology & Immunology, McGill University, 3775 University Street, Montreal, Quebec, H3A 2B4, Canada
| | - Layan Suleiman
- Department of Pharmacology & Therapeutics, McGill University, 3655 Prom. Sir-William-Osler, Montreal, Quebec, H3G 1Y6, Canada
| | - Lharbi Dridi
- Department of Pharmacology & Therapeutics, McGill University, 3655 Prom. Sir-William-Osler, Montreal, Quebec, H3G 1Y6, Canada
| | - Corinne F Maurice
- Department of Microbiology & Immunology, McGill University, 3775 University Street, Montreal, Quebec, H3A 2B4, Canada
- McGill Centre for Microbiome Research, Montreal, Quebec, Canada
| | - Bastien Castagner
- Department of Pharmacology & Therapeutics, McGill University, 3655 Prom. Sir-William-Osler, Montreal, Quebec, H3G 1Y6, Canada.
- McGill Centre for Microbiome Research, Montreal, Quebec, Canada.
| |
Collapse
|
39
|
Huang J, Li J, Geng Z, Yin L, Niu M, Li Q, Liu X, Cheng X, Zhang X, Song X, Wang Y, Wang L, Zuo L, Hu J. Cynaroside ameliorates TNBS-induced colitis by inhibiting intestinal epithelial cell apoptosis via the PI3K/AKT signalling pathway. Front Pharmacol 2025; 15:1496068. [PMID: 39902073 PMCID: PMC11788346 DOI: 10.3389/fphar.2024.1496068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Accepted: 12/30/2024] [Indexed: 02/05/2025] Open
Abstract
Background and aims Patients with Crohn's disease (CD) exhibit excessive apoptosis of intestinal epithelial cells (IECs), which contributes to damage to the intestinal barrier structure and function, thereby playing a role in the progression of colitis. Preventing IEC apoptosis and protecting the intestinal barrier are critical to alleviating colitis. Natural plant monomers have been reported to possess multiple pharmacological properties, particularly with the potential to treat CD. This study focuses on Cynaroside (Cyn) to explore its effect on IEC apoptosis and evaluate its pharmacological impact on the intestinal barrier and colitis. Methods The 2,4,6-trinitrobenzenesulfonic acid (TNBS)-induced CD-like colitis mice model was employed in this study. We assessed the therapeutic effect of Cyn on CD-like colitis by evaluating the disease activity index (DAI), body weight changes, intestinal tissue pathological damage, and inflammatory factor levels. Immunofluorescence and Western blotting were used to detect the expression and localization of tight junction (TJ) proteins, allowing us to analyze the intestinal barrier structure. The function of the intestinal barrier was examined using FITC-dextran (FD4), TEER values, and bacterial translocation. Network pharmacology enrichment analysis revealed that Cyn could inhibit cell apoptosis. We also explored the effect and underlying mechanism of Cyn in inhibiting IEC apoptosis on intestinal barrier function and colitis using both the TNF-α-induced colonic organoid model and the TNBS-induced mouse model. Results Our findings show that Cyn significantly alleviates TNBS-induced colitis symptoms in mice, as evidenced by reduced body weight loss, colon shortening, DAI score, colon histopathology score, and lower levels of inflammatory factors (IL-1β, TNF-α, and IL-6) compared to the model group. Additionally, the Cyn intervention group showed significant improvements in both the intestinal barrier structure (elevated tight junction protein levels and proper localization) and function (reduced serum FD4 levels, increased intestinal TEER, and decreased bacterial translocation rates in mesenteric lymph nodes [MLNs] and livers). Combining network pharmacology prediction analysis with our validation data from animal models and colonic organoids, we demonstrated that Cyn significantly inhibits IEC apoptosis, as indicated by a decrease in the proportion of TUNEL-positive cells and changes in apoptosis-related protein levels. KEGG enrichment analysis and signaling pathway intervention experiments confirmed that Cyn inhibits the activation of PI3K/AKT signaling. Conclusion Cyn inhibits IEC apoptosis by blocking the PI3K/AKT signaling pathway, which is the primary mechanism underlying its protective effects on the intestinal barrier and its ability to improve CD-like colitis. This study also supports the potential of the Chinese medicine monomer Cyn as a promising therapeutic agent for the treatment of CD.
Collapse
Affiliation(s)
- Ju Huang
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Jing Li
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Zhijun Geng
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Lixia Yin
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Minzhu Niu
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Qingqing Li
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Department of Clinical Laboratory, The Third the People’s Hospital of Bengbu, Bengbu, Anhui, China
| | - Xinyue Liu
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Xinke Cheng
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Xiaofeng Zhang
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Xue Song
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Department of Central Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Yueyue Wang
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Lian Wang
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Lugen Zuo
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| | - Jianguo Hu
- Department of Clinical Laboratory, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
- Anhui Province Key Laboratory of Basic and Translational Research of Inflammation-Related Diseases, First Affiliated Hospital of Bengbu Medical University, Bengbu, Anhui, China
| |
Collapse
|
40
|
Alfei S, Zuccari G. Ellagic Acid: A Green Multi-Target Weapon That Reduces Oxidative Stress and Inflammation to Prevent and Improve the Condition of Alzheimer's Disease. Int J Mol Sci 2025; 26:844. [PMID: 39859559 PMCID: PMC11766176 DOI: 10.3390/ijms26020844] [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: 12/15/2024] [Revised: 01/08/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
Oxidative stress (OS), generated by the overrun of reactive species of oxygen and nitrogen (RONS), is the key cause of several human diseases. With inflammation, OS is responsible for the onset and development of clinical signs and the pathological hallmarks of Alzheimer's disease (AD). AD is a multifactorial chronic neurodegenerative syndrome indicated by a form of progressive dementia associated with aging. While one-target drugs only soften its symptoms while generating drug resistance, multi-target polyphenols from fruits and vegetables, such as ellagitannins (ETs), ellagic acid (EA), and urolithins (UROs), having potent antioxidant and radical scavenging effects capable of counteracting OS, could be new green options to treat human degenerative diseases, thus representing hopeful alternatives and/or adjuvants to one-target drugs to ameliorate AD. Unfortunately, in vivo ETs are not absorbed, while providing mainly ellagic acid (EA), which, due to its trivial water-solubility and first-pass effect, metabolizes in the intestine to yield UROs, or irreversible binding to cellular DNA and proteins, which have very low bioavailability, thus failing as a therapeutic in vivo. Currently, only UROs have confirmed the beneficial effect demonstrated in vitro by reaching tissues to the extent necessary for therapeutic outcomes. Unfortunately, upon the administration of food rich in ETs or ETs and EA, URO formation is affected by extreme interindividual variability that renders them unreliable as novel clinically usable drugs. Significant attention has therefore been paid specifically to multitarget EA, which is incessantly investigated as such or nanotechnologically manipulated to be a potential "lead compound" with protective action toward AD. An overview of the multi-factorial and multi-target aspects that characterize AD and polyphenol activity, respectively, as well as the traditional and/or innovative clinical treatments available to treat AD, constitutes the opening of this work. Upon focus on the pathophysiology of OS and on EA's chemical features and mechanisms leading to its antioxidant activity, an all-around updated analysis of the current EA-rich foods and EA involvement in the field of AD is provided. The possible clinical usage of EA to treat AD is discussed, reporting results of its applications in vitro, in vivo, and during clinical trials. A critical view of the need for more extensive use of the most rapid diagnostic methods to detect AD from its early symptoms is also included in this work.
Collapse
Affiliation(s)
- Silvana Alfei
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano, 4, 16148 Genova, Italy
| | - Guendalina Zuccari
- Department of Pharmacy (DIFAR), University of Genoa, Viale Cembrano, 4, 16148 Genova, Italy
- Laboratory of Experimental Therapies in Oncology, IRCCS Istituto Giannina Gaslini, Via G. Gaslini 5, 16147 Genoa, Italy
| |
Collapse
|
41
|
Fernández-Fígares Jiménez MDC. Role of a Whole Plant Foods Diet in Breast Cancer Prevention and Survival. JOURNAL OF THE AMERICAN NUTRITION ASSOCIATION 2025:1-17. [PMID: 39784140 DOI: 10.1080/27697061.2024.2442631] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/30/2024] [Revised: 11/25/2024] [Accepted: 12/10/2024] [Indexed: 01/12/2025]
Abstract
Breast cancer (BC) is one of the leading causes of death and morbidity among women worldwide. Epidemiologic evidence shows that the risk of BC and other chronic diseases decreases as the proportion of whole plant foods increases, while the proportion of animal foods (fish, meat, poultry, eggs, seafood, and dairy products) and non-whole plant foods (e.g., refined grains, added sugars, French fries) in the diet decreases. Whole plant foods include fruits, vegetables, roots, tubers, whole grains, legumes, nuts, and seeds from which no edible part has been removed and to which no non-whole food been added. A whole plant foods diet lowers insulin resistance, inflammation, excess body fat, cholesterol, and insulin-like growth factor 1 and sex hormone bioavailability; it also increases estrogen excretion, induces favorable changes in the gut microbiota, and may also favorably affect mammary microbiota composition and decrease the risk of early menarche, all contributing to reduced BC incidence, recurrence, and mortality. This review explores the connection between a whole plant foods diet and BC risk and mortality as well as the potential mechanisms involved. Additionally, this diet is compared with other dietary approaches recommended for BC. A whole plant foods diet seems the optimal dietary pattern for BC and overall disease prevention as it exclusively consists of whole plant foods which, based on existing evidence, lead to the best health outcomes.
Collapse
|
42
|
Liu S, Li S, Lu S, Yang M, Liu M, Li J, Li S, Jian F. Effects of fermented Artemisia annua on the intestinal microbiota and metabolites of Hu lambs with naturally infected with Eimeria spp. Front Cell Infect Microbiol 2025; 14:1448516. [PMID: 39839259 PMCID: PMC11747653 DOI: 10.3389/fcimb.2024.1448516] [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: 06/13/2024] [Accepted: 12/03/2024] [Indexed: 01/23/2025] Open
Abstract
Background Sheep coccidiosis could disturb the balance of intestinal microbiota, causing diarrhea, and even death in lambs. Chemical drugs are the primary method of treating sheep coccidiosis, but their use will bring drug resistance, toxic side effects, drug residues, and other problems. Chinese herbal medicines are investigated as alternative methods for controlling coccidian infections. Methods In this study, the effect of fermented Artemisia annua (FA) on oocysts per gram (OPG), average daily gain (ADG), and expression of inflammatory factors were investigated in lambs that were naturally infected with coccidia. Results The results showed that the FA had similar anti-coccidiosis effect to the original drug, while the FA demonstrated a more significant effect on weight gain, and a better ability to reduce the inflammatory response compared to the unfermented drug during the treatment period (P < 0.05). Furthermore, High-throughput sequencing technology was used to study the effects of FA on intestinal microbiota, and fecal metabolites of naturally infected lambs. The species richness of intestinal microbiota of lambs was significantly improved by FA. The abundance of bacteria unclassified_Muribaculaceae, and UCG_005 were increased by fermentation of A. annua. The abundance of bacteria Escherichia_Shigella, unclassified_Clostridia_UCG_014, and Alistipes was reduced. The prevention, and treatment of coccidiosis by fermentation of A. annua may also be related to a series of metabolites affected by intestinal microbiota, including artemisinin, Lysyl-Proline, and TRP-tyrosine. Conclusion FA was found to have superior anti-coccidiosis, anti-inflammatory, and weight gain effects compared to the original Artemisia annua. Intestinal microbes and metabolites such as unclassified_Muribaculaceae, UCG-005, and Artemisinin were identified, suggesting their potential significance. Alistipes was proposed as a biomarker for predicting intestinal coccidia outbreak risk in lambs, pending further validation. The correlation between microbiota, and metabolites may provide new insights into pathogenic changes associated with Eimeria spp.
Collapse
Affiliation(s)
- Shuaiqi Liu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, China
- Key Laboratory of Quality and Safety Control of Poultry Products (Zhengzhou), Ministry of Agriculture and Rural Affairs, Zhengzhou, China
| | - Shiheng Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, China
- Key Laboratory of Quality and Safety Control of Poultry Products (Zhengzhou), Ministry of Agriculture and Rural Affairs, Zhengzhou, China
| | - Shunli Lu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, China
- Key Laboratory of Quality and Safety Control of Poultry Products (Zhengzhou), Ministry of Agriculture and Rural Affairs, Zhengzhou, China
| | - Mingfan Yang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Manyu Liu
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, China
- Key Laboratory of Quality and Safety Control of Poultry Products (Zhengzhou), Ministry of Agriculture and Rural Affairs, Zhengzhou, China
| | - Juanfeng Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, China
- Key Laboratory of Quality and Safety Control of Poultry Products (Zhengzhou), Ministry of Agriculture and Rural Affairs, Zhengzhou, China
| | - Senyang Li
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, China
- Key Laboratory of Quality and Safety Control of Poultry Products (Zhengzhou), Ministry of Agriculture and Rural Affairs, Zhengzhou, China
| | - Fuchun Jian
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- International Joint Research Laboratory for Zoonotic Diseases of Henan, Zhengzhou, China
- Key Laboratory of Quality and Safety Control of Poultry Products (Zhengzhou), Ministry of Agriculture and Rural Affairs, Zhengzhou, China
| |
Collapse
|
43
|
Arumugam P, Saha K, Nighot P. Intestinal Epithelial Tight Junction Barrier Regulation by Novel Pathways. Inflamm Bowel Dis 2025; 31:259-271. [PMID: 39321109 DOI: 10.1093/ibd/izae232] [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: 06/28/2024] [Indexed: 09/27/2024]
Abstract
Intestinal epithelial tight junctions (TJs), a dynamically regulated barrier structure composed of occludin and claudin family of proteins, mediate the interaction between the host and the external environment by allowing selective paracellular permeability between the luminal and serosal compartments of the intestine. TJs are highly dynamic structures and can undergo constant architectural remodeling in response to various external stimuli. This is mediated by an array of intracellular signaling pathways that alters TJ protein expression and localization. Dysfunctional regulation of TJ components compromising the barrier homeostasis is an important pathogenic factor for pathological conditions including inflammatory bowel disease (IBD). Previous studies have elucidated the significance of TJ barrier integrity and key regulatory mechanisms through various in vitro and in vivo models. In recent years, considerable efforts have been made to understand the crosstalk between various signaling pathways that regulate formation and disassembly of TJs. This review provides a comprehensive view on the novel mechanisms that regulate the TJ barrier and permeability. We discuss the latest evidence on how ion transport, cytoskeleton and extracellular matrix proteins, signaling pathways, and cell survival mechanism of autophagy regulate intestinal TJ barrier function. We also provide a perspective on the context-specific outcomes of the TJ barrier modulation. The knowledge on the diverse TJ barrier regulatory mechanisms will provide further insights on the relevance of the TJ barrier defects and potential target molecules/pathways for IBD.
Collapse
Affiliation(s)
- Priya Arumugam
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA, USA
| | - Kushal Saha
- Department of Pathology, Brigham and Women's Hospital, Harvard Medical School, Boston, MA, USA
| | - Prashant Nighot
- Division of Gastroenterology and Hepatology, Department of Medicine, Pennsylvania State College of Medicine, Hershey, PA, USA
| |
Collapse
|
44
|
Wang F, Li Q, Xu T, Li Z, Jiang Y, Ma Y, Li X, Wang W, Qian H. An orally administered gold nanocluster with ROS scavenging for inflammatory bowel disease treatment. FUNDAMENTAL RESEARCH 2025; 5:381-390. [PMID: 40166122 PMCID: PMC11955046 DOI: 10.1016/j.fmre.2022.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2022] [Revised: 07/06/2022] [Accepted: 07/10/2022] [Indexed: 11/30/2022] Open
Abstract
Oxidative stress that is induced by excessive reactive oxygen species (ROS) is considered to be a key pathophysiological mechanism of inflammatory bowel disease (IBD), and restoring redox homeostasis in the inflammatory region by eliminating ROS is an effective way to treat IBD. Herein, ultrasmall Au25 nanoclusters (Au25 NCs) were synthesized using a simple improved protocol, which has good physiological stability and biosafety and can be noninvasively monitored by clinical computed tomography (CT) after oral administration. Au25 NCs can eliminate ROS such as ABTS radicals, superoxide free radicals (•O2 -), and hydroxyl free radicals (•OH), upregulate the expression level of antioxidant enzymes, inhibit the expression of proinflammatory cytokines, and finally interrupt the inflammatory circuit of IBD to achieve the effective prevention and delayed treatment of IBD. This work will demonstrate the protective effect of Au25 NCs on IBD in living animals, which suggests a new nanomedicine strategy for IBD treatment.
Collapse
Affiliation(s)
- Fei Wang
- Guangdong Provincial Key Laboratory of Digestive Cancer Research and The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, China
| | - Qianhui Li
- Guangdong Provincial Key Laboratory of Digestive Cancer Research and The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, China
| | - Tingting Xu
- School of Basic Medical Sciences Anhui Medical University, Hefei 230032, China
| | - Zhu Li
- Guangdong Provincial Key Laboratory of Digestive Cancer Research and The Seventh Affiliated Hospital of Sun Yat-Sen University, Shenzhen 518107, China
| | - Yongxin Jiang
- School of Basic Medical Sciences Anhui Medical University, Hefei 230032, China
| | - Yan Ma
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, China
| | - Xiaohu Li
- Department of Radiology, The First Affiliated Hospital of Anhui Medical University, Anhui Medical University, Hefei 230032, China
| | - Wanni Wang
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, China
| | - Haisheng Qian
- School of Biomedical Engineering, Research and Engineering Center of Biomedical Materials, Anhui Provincial Institute of Translational Medicine, Anhui Medical University, Hefei 230032, China
| |
Collapse
|
45
|
Mytilinaiou E, Kitopoulou K, Palikaras K. Caenorhabditis elegans as a Screening Platform for Anti-aging Compounds. Methods Mol Biol 2025; 2906:301-313. [PMID: 40082364 DOI: 10.1007/978-1-0716-4426-3_18] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/16/2025]
Abstract
As global life expectancy continues to rise, the need to understand and mitigate the effects of aging on human physiology has become increasingly important. Aging is characterized by cellular and functional decline, resulting in a higher prevalence of chronic diseases. Model organisms, such as Caenorhabditis elegans, provide valuable insights into the molecular mechanisms underlying aging and serve as platforms for developing potential therapeutic interventions. This chapter highlights the utility of C. elegans in aging research by presenting three straightforward protocols: the lifespan assay, thrashing assay, and lipofuscin accumulation assay. These assays are designed to effectively assess key physiological aspects of organismal health and provide a reliable framework for monitoring the aging process and evaluating anti-aging compounds. Here, we demonstrate the application of these protocols using Urolithin A as an example, underscoring their efficacy in advancing our understanding of aging and contributing to the development of potential interventions.
Collapse
Affiliation(s)
- Eirini Mytilinaiou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Katerina Kitopoulou
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece
| | - Konstantinos Palikaras
- Department of Physiology, Medical School, National and Kapodistrian University of Athens, Athens, Greece.
| |
Collapse
|
46
|
Wang J, Zhou T. Unveiling gut microbiota's role: Bidirectional regulation of drug transport for improved safety. Med Res Rev 2025; 45:311-343. [PMID: 39180410 DOI: 10.1002/med.22077] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Revised: 07/20/2024] [Accepted: 08/04/2024] [Indexed: 08/26/2024]
Abstract
Drug safety is a paramount concern in the field of drug development, with researchers increasingly focusing on the bidirectional regulation of gut microbiota in this context. The gut microbiota plays a crucial role in maintaining drug safety. It can influence drug transport processes in the body through various mechanisms, thereby modulating their efficacy and toxicity. The main mechanisms include: (1) The gut microbiota directly interacts with drugs, altering their chemical structure to reduce toxicity and enhance efficacy, thereby impacting drug transport mechanisms, drugs can also change the structure and abundance of gut bacteria; (2) bidirectional regulation of intestinal barrier permeability by gut microbiota, promoting the absorption of nontoxic drugs and inhibiting the absorption of toxic components; (3) bidirectional regulation of the expression and activity of transport proteins by gut microbiota, selectively promoting the absorption of effective components or inhibiting the absorption of toxic components. This bidirectional regulatory role enables the gut microbiota to play a key role in maintaining drug balance in the body and reducing adverse reactions. Understanding these regulatory mechanisms sheds light on novel approaches to minimize toxic side effects, enhance drug efficacy, and ultimately improve drug safety. This review systematically examines the bidirectional regulation of gut microbiota in drug transportation from the aforementioned aspects, emphasizing their significance in ensuring drug safety. Furthermore, it offers a prospective outlook from the standpoint of enhancing therapeutic efficacy and reducing drug toxicity, underscoring the importance of further exploration in this research domain. It aims to provide more effective strategies for drug development and treatment.
Collapse
Affiliation(s)
- Jinyi Wang
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Second Military Medical University, Shanghai, China
| | - Tingting Zhou
- Department of Pharmaceutical Analysis, School of Pharmacy, Second Military Medical University, Shanghai, China
- Shanghai Key Laboratory for Pharmaceutical Metabolite Research, School of Pharmacy, Second Military Medical University, Shanghai, China
| |
Collapse
|
47
|
Kuatov R, Takano J, Arie H, Kominami M, Tateishi N, Wakabayashi KI, Takemoto D, Izumo T, Nakao Y, Nakamura W, Shinohara K, Nakahata Y. Urolithin A Modulates PER2 Degradation via SIRT1 and Enhances the Amplitude of Circadian Clocks in Human Senescent Cells. Nutrients 2024; 17:20. [PMID: 39796454 PMCID: PMC11722880 DOI: 10.3390/nu17010020] [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: 11/19/2024] [Revised: 12/23/2024] [Accepted: 12/23/2024] [Indexed: 01/13/2025] Open
Abstract
BACKGROUND/OBJECTIVES Circadian clocks are endogenous systems that regulate numerous biological, physiological, and behavioral events in living organisms. Aging attenuates the precision and robustness of circadian clocks, leading to prolonged and dampened circadian gene oscillation rhythms and amplitudes. This study investigated the effects of food-derived polyphenols such as ellagic acid and its metabolites (urolithin A, B, and C) on the aging clock at the cellular level using senescent human fibroblast cells, TIG-3 cells. METHODS Lentivirus-infected TIG-3 cells expressing Bmal1-luciferase were used for real-time luciferase monitoring assays. RESULTS We revealed that urolithins boosted the amplitude of circadian gene oscillations at different potentials; urolithin A (UA) amplified the best. Furthermore, we discovered that UA unstabilizes PER2 protein while stabilizing SIRT1 protein, which provably enhances BMAL1 oscillation. CONCLUSIONS The findings suggest that urolithins, particularly UA, have the potential to modulate the aging clock and may serve as therapeutic nutraceuticals for age-related disorders associated with circadian dysfunction.
Collapse
Affiliation(s)
- Rassul Kuatov
- Department of Neurobiology & Behavior, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8523, Japan
| | - Jiro Takano
- Institute for Science of Life, Suntory Wellness Limited, Kyoto 619-0284, Japan (N.T.); (T.I.)
| | - Hideyuki Arie
- Institute for Science of Life, Suntory Wellness Limited, Kyoto 619-0284, Japan (N.T.); (T.I.)
| | - Masaru Kominami
- Institute for Science of Life, Suntory Wellness Limited, Kyoto 619-0284, Japan (N.T.); (T.I.)
| | - Norifumi Tateishi
- Institute for Science of Life, Suntory Wellness Limited, Kyoto 619-0284, Japan (N.T.); (T.I.)
| | - Ken-ichi Wakabayashi
- Institute for Science of Life, Suntory Wellness Limited, Kyoto 619-0284, Japan (N.T.); (T.I.)
| | - Daisuke Takemoto
- Institute for Science of Life, Suntory Wellness Limited, Kyoto 619-0284, Japan (N.T.); (T.I.)
| | - Takayuki Izumo
- Institute for Science of Life, Suntory Wellness Limited, Kyoto 619-0284, Japan (N.T.); (T.I.)
| | - Yoshihiro Nakao
- Institute for Science of Life, Suntory Wellness Limited, Kyoto 619-0284, Japan (N.T.); (T.I.)
| | - Wataru Nakamura
- Department of Oral-Chrono Physiology, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8588, Japan
| | - Kazuyuki Shinohara
- Department of Neurobiology & Behavior, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8523, Japan
| | - Yasukazu Nakahata
- Department of Neurobiology & Behavior, Graduate School of Biomedical Sciences, Nagasaki University, Nagasaki 852-8523, Japan
| |
Collapse
|
48
|
Yuan Y, Wang X, Ling N, Zhou J, Zhao L, Ji B, Zhou F, Zhao L. Identification of Protein Hydrolysates from Sesame Meal and In Vivo Study of Their Gastric Mucosal Protective Effects. Foods 2024; 13:4178. [PMID: 39767120 PMCID: PMC11675995 DOI: 10.3390/foods13244178] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 12/12/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
This study aimed to investigate the protective effects and defense mechanisms of a sesame meal protein hydrolysate against ethanol-induced acute gastric mucosal injury in mice. The target peptides in the hydrolysate were identified by LC-MS/MS, the activity was predicted by PeptideRanker, and the KM mice were orally administered distilled water, a sesame peptide, and omeprazole for 24 consecutive days. Acute gastric mucosal injury was then induced in mice with 70% ethanol, except for the CK group. The sesame peptide significantly inhibited the over-accumulation of ALT, AST, MDA, TNF-α, IL-1β, and MPO, while promoting the reduction in GSH, T-AOC, GSSG, and EGF expression. In addition, a Western blotting analysis showed that sesame peptide significantly up-regulated the expression of HO-1 and NQO1 proteins in the Nrf2/Keap1 signaling pathway, and down-regulated Keap1 protein. The defense effect of a sesame peptide on gastric mucosa may be achieved by alleviating the overproduction of lipid peroxides and improving the antioxidant activity.
Collapse
Affiliation(s)
- Yutong Yuan
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Y.Y.); (X.W.); (J.Z.); (B.J.)
| | - Xinyi Wang
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Y.Y.); (X.W.); (J.Z.); (B.J.)
| | - Nan Ling
- Nanjing Weigang Dairy Co., Ltd., No. 366 Lantian Road, Nanjing 210095, China;
| | - Jingxuan Zhou
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Y.Y.); (X.W.); (J.Z.); (B.J.)
| | - Lei Zhao
- Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China;
| | - Baoping Ji
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Y.Y.); (X.W.); (J.Z.); (B.J.)
| | - Feng Zhou
- Beijing Key Laboratory of Functional Food from Plant Resources, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (Y.Y.); (X.W.); (J.Z.); (B.J.)
| | - Liang Zhao
- Beijing Engineering and Technology Research Center of Food Additives, School of Food and Health, Beijing Technology and Business University, Beijing 100048, China;
| |
Collapse
|
49
|
Zhu C, Tang L, Zhou F, Tang Y, Hu Q, Wang C, Feng X, Zhuang Z. Design, Synthesis, and Anti-Infective Effect Against Candida Albicans of a New Urolithin Derivative. Chem Biodivers 2024:e202402966. [PMID: 39714977 DOI: 10.1002/cbdv.202402966] [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: 11/12/2024] [Revised: 12/11/2024] [Accepted: 12/16/2024] [Indexed: 12/25/2024]
Abstract
Deep mucosal and organ infections caused by the infestation of Candida albicans in immunocompromised patients represent a significant cause of mortality in hospitalized patients. The rise in fungal resistance is a consequence of the overuse of antibiotics. Therefore, innovative immunostimulants must be developed to combat pathogenic fungal infections. We used urolithin A (UA), an intestinal metabolite rich in the naturally occurring polyphenolic antioxidants ellagic acid (EA) or ellagitannin (ET), as a lead compound for structural modification. Through liquid screening of 17 synthesized compounds, we discovered compound 1e effectively inhibited C. albicans biofilm formation, thereby reducing its virulence. Furthermore, it protects animals from severe infections by enhancing tolerance to infection by intestinal pathogens and reducing oxidative stress. Moreover, our findings indicate that compound 1e exerts its effects through the p38 mitogen-activated protein kinase (MAPK) innate immune pathway, which is evolutionarily conserved. These observations not only enhance our comprehension of immune mechanisms but also provide a crucial foundation for the development of immune activators with the potential to resist pathogenic bacterial infections.
Collapse
Affiliation(s)
- Chenyan Zhu
- School of Pharmacy & School of Biological and Food Engineering, Changzhou University, Changzhou, China
| | - Long Tang
- School of Pharmacy & School of Biological and Food Engineering, Changzhou University, Changzhou, China
| | - Feng Zhou
- School of Pharmacy & School of Biological and Food Engineering, Changzhou University, Changzhou, China
| | - Yingmao Tang
- School of Pharmacy & School of Biological and Food Engineering, Changzhou University, Changzhou, China
| | - Qiulin Hu
- School of Pharmacy & School of Biological and Food Engineering, Changzhou University, Changzhou, China
| | - Chenchen Wang
- School of Pharmacy & School of Biological and Food Engineering, Changzhou University, Changzhou, China
| | - Xiaoqing Feng
- School of Pharmacy & School of Biological and Food Engineering, Changzhou University, Changzhou, China
| | - Ziheng Zhuang
- School of Pharmacy & School of Biological and Food Engineering, Changzhou University, Changzhou, China
| |
Collapse
|
50
|
Huang FC. Therapeutic Potential of Nutritional Aryl Hydrocarbon Receptor Ligands in Gut-Related Inflammation and Diseases. Biomedicines 2024; 12:2912. [PMID: 39767818 PMCID: PMC11673835 DOI: 10.3390/biomedicines12122912] [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: 10/19/2024] [Revised: 12/13/2024] [Accepted: 12/18/2024] [Indexed: 01/11/2025] Open
Abstract
A solid scientific foundation is required to build the concept of personalized nutrition developed to promote health and a vision of disease prevention. Growing evidence indicates that nutrition can modulate the immune system through metabolites, which are either generated via microbiota metabolism or host digestion. The aryl hydrocarbon receptor (AhR) plays a crucial role in regulating immune responses, particularly in the gut, and has emerged as a key modulator of gut-mediated inflammation and related diseases. AhR is a ligand-activated transcription factor that responds to environmental, dietary, and microbial-derived signals, influencing immune balance and maintaining intestinal homeostasis. Nutritional AhR ligands play a significant role in modulating intestinal immunity and the function of mucosal immune cells, thereby exerting clinical effects on colitis and innate immunity. Additionally, they have the capacity to orchestrate autophagy, phagocytic cell function, and intestinal epithelial tight junctions. Therapeutic strategies aimed at enhancing AhR activity, restoring gut integrity, and optimizing immune responses hold promise as avenues for future research and potential treatments for critically ill patients.
Collapse
Affiliation(s)
- Fu-Chen Huang
- Department of Pediatrics, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung 833401, Taiwan
| |
Collapse
|