1
|
Lv LX, Yin JN, Sun YL, Wei MY, Jiang WQ, Gu YC, Yang XP, Shao CL. Marine natural products as potential anti-Pseudomonas aeruginosa agents: challenges and advances. Eur J Med Chem 2025; 292:117670. [PMID: 40305937 DOI: 10.1016/j.ejmech.2025.117670] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/17/2025] [Accepted: 04/19/2025] [Indexed: 05/02/2025]
Abstract
Antimicrobial resistance (AMR) has become a pressing need to address in the major global public health challenges, posing a serious threat to human health. Pseudomonas aeruginosa (PA) is one of the most concerning Gram-negative pathogens and is typically treated with broad-spectrum antibiotics. PA exhibits resistance to multiple antibiotics, multifactorial virulence, and dynamic hyperadaptation, which results in a particularly formidable challenge in eliminating PA from patients. The problem of drug resistance is becoming increasingly serious, and the development of new antibiotics is extremely lagging behind, resulting in no drug with a new structure and mechanism being approved for the treatment of infections caused by drug-resistant Gram-negative bacteria over the past half-century. Consequently, the development of new antibiotics is of utmost urgency and importance. Marine natural products (MNPs) have become an important source for developing new antibiotics due to their unique properties. So far, 44 potential molecules with significant anti-PA activity have been isolated from marine organisms, of which 19 have been reported as quorum-sensing system inhibitors (QSIs) with potential for further development. In this review, we provide a comprehensive summary of the current status of drug resistance, pathogenic mechanisms, and resistance mechanisms associated with PA infections. We also highlight the challenges and opportunities presented by MNPs in the development of anti-PA drugs, and offer recommendations to accelerate the antibiotic development process, thereby providing valuable insights for the study and exploitation of novel antibiotics.
Collapse
Affiliation(s)
- Liu-Xia Lv
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, People's Republic of China
| | - Jun-Na Yin
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, People's Republic of China
| | - Yi-Lin Sun
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, People's Republic of China
| | - Mei-Yan Wei
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, People's Republic of China
| | - Wen-Qing Jiang
- Qingdao Traditional Chinese Medicine Hospital, Qingdao Hiser Hospital Affiliated of Qingdao University, Qingdao, 266100, People's Republic of China
| | - Yu-Cheng Gu
- Syngenta Jealott's Hill International Research Centre, Bracknell, Berkshire, RG42 6EY, United Kingdom
| | - Xiao-Ping Yang
- Qingdao Traditional Chinese Medicine Hospital, Qingdao Hiser Hospital Affiliated of Qingdao University, Qingdao, 266100, People's Republic of China.
| | - Chang-Lun Shao
- Key Laboratory of Marine Drugs, The Ministry of Education of China, School of Medicine and Pharmacy, Ocean University of China, Qingdao, 266003, People's Republic of China; Key Laboratory of Tropical Medicinal Resource Chemistry of Ministry of Education, College of Chemistry and Chemical Engineering, Hainan Normal University, Haikou, 571158, People's Republic of China.
| |
Collapse
|
2
|
Liang C, Jia Y, He RT, Ding B, Yang ZD, Cao XQ, Zhang J. Wheat bran supplementation improved polystyrene degradation efficiency of Zophobas atratus larvae by alleviating intestinal injury caused by polystyrene-intake. JOURNAL OF HAZARDOUS MATERIALS 2025; 492:138148. [PMID: 40184973 DOI: 10.1016/j.jhazmat.2025.138148] [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: 01/21/2025] [Revised: 03/05/2025] [Accepted: 04/01/2025] [Indexed: 04/07/2025]
Abstract
Wheat bran supplementation significantly improves the polystyrene consumption capacity of Zophobas atratus larvae. However, the underlying mechanism remains unclear, limiting further advancements in degradation efficiency. This study seeks to clarify the mechanism by analyzing intestinal morphology, gut microbiota structure, gene transcription, and targeted metabolites. Specifically, histopathology results demonstrated that wheat bran supplementation alleviated polystyrene-induced intestinal injury in larvae. From a genetic perspective, genes related to annexins and tight junction proteins were upregulated in intestinal tissues. These genetic changes positively correlated with increased levels of short-chain fatty acids, eicosanoids, and beneficial bacteria (e.g., Latilactobacillus curvatus). Moreover, wheat bran supplementation alleviated gut microbiota dysbiosis, suppressed pathogenic bacteria, reduced over upregulation of NADPH oxidase activity related genes, and up-regulated genes linked to the intestinal immune system process pathway, thereby mitigating the intestinal injury. It also upregulated genes associated with the aromatase activity pathway, promoting the degradation of polystyrene and its intermediates. In summary, wheat bran supplementation enhanced the polystyrene degradation efficiency of Z. atratus larvae by upregulating genes linked to the aromatase activity pathway and mitigating intestinal injury (through modulation of intestinal gene transcription, microbiota structure, and metabolites). Our findings offer new insights into improving the efficiency of insect-mediated plastic biodegradation.
Collapse
Affiliation(s)
- Cong Liang
- College of safety and environmental engineering, Shandong University of Science and Technology, Qingdao, Shandong 266590, China
| | - Yun Jia
- College of New Energy, China University of Petroleum (East China), Qingdao, Shandong 266580, China
| | - Ren-Tao He
- College of New Energy, China University of Petroleum (East China), Qingdao, Shandong 266580, China
| | - Bin Ding
- College of New Energy, China University of Petroleum (East China), Qingdao, Shandong 266580, China.
| | - Zheng-da Yang
- College of New Energy, China University of Petroleum (East China), Qingdao, Shandong 266580, China
| | - Xiao-Qiang Cao
- College of safety and environmental engineering, Shandong University of Science and Technology, Qingdao, Shandong 266590, China; Institute of Yellow River Delta Earth Surface Processes and Ecological Integrity, Shandong University of Science and Technology, Qingdao, Shandong 266590, China
| | - Jian Zhang
- College of safety and environmental engineering, Shandong University of Science and Technology, Qingdao, Shandong 266590, China; Institute of Yellow River Delta Earth Surface Processes and Ecological Integrity, Shandong University of Science and Technology, Qingdao, Shandong 266590, China; College of Chemistry, Chemical Engineering and Materials Science, Shandong Normal University, Jinan, Shandong 250014, China.
| |
Collapse
|
3
|
Thakur R, Kaur S. Use of postbiotics and parabiotics from lactobacilli in the treatment of infectious diarrhea. Microb Pathog 2025; 204:107580. [PMID: 40222563 DOI: 10.1016/j.micpath.2025.107580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 01/30/2025] [Accepted: 04/11/2025] [Indexed: 04/15/2025]
Abstract
Probiotics are effective in the treatment of diarrheal disease which is the second leading cause of death in children below the age of five years via the production of antimicrobial peptides and lactic acid. These live bacteria are known to benefit the host by modulating their gut microbiome and competitively excluding pathogens from the gut. As probiotics are live microbial cells, their safety evaluation is a concern that shifts the focus from the usage of live cells to parabiotics and postbiotics. In recent years attempts have been made to study the efficacy of postbiotics and parabiotics against enteric pathogens. Enteric pathogens are the major cause of diarrhea resulting in watery stools and electrolyte imbalance. Among various gastrointestinal illnesses, 30 % are caused by bacteria. These gastrointestinal infections in adults have usually mild to moderate symptoms that disappear spontaneously but, in some cases, they can cause chronic diseases such as typhoid, irritable bowel syndrome, ulcerative colitis and bacteremia. The extensive use of antibiotics for the treatment of bacterial-infection-induced diarrhea has led to the emergence of drug resistance among these enteric pathogens. Drug resistance poses a major threat in the treatment of various other diseases as well. Further, the use of antibiotics is known to disrupt the homeostasis of the gut by killing the normal gut flora thereby worsening the situation. Therefore, the urgent need for new interventions to combat these enteric pathogens along with restoration of gut barrier. Lactobacillus-derived parabiotics and postbiotics have emerged as promising approaches for managing and treating diarrheal diseases. Therefore, our research is focused on studying the efficacy and underlying mechanisms of Lactobacillus spp.-derived postbiotics and parabiotics against enteric pathogens. Understanding these mechanisms helps in combatting diarrhea associated with enteric pathogens and results in reducing the morbidity and mortality rates associated with infectious diarrhea and its complications.
Collapse
Affiliation(s)
- Raman Thakur
- Department of Medical Laboratory Sciences, Lovely Professional University, Punjab, 144411, India
| | - Sumanpreet Kaur
- Department of Medical Laboratory Sciences, Lovely Professional University, Punjab, 144411, India.
| |
Collapse
|
4
|
Yuan X, Wu F, Cheng L, Ji T, Zheng C, Ma Y, Jin Y, Dong J, Jin Y, Fang B. Chlorpyrifos Inhibits Intestinal Stem Cell Proliferation and Differentiation at the Acceptable Daily Intake and Disrupts Immune Responses at High Doses. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025. [PMID: 40357541 DOI: 10.1021/acs.jafc.4c13249] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/15/2025]
Abstract
The acceptable daily intake (ADI) and maximum residue limits in food for chlorpyrifos (CPF), a widely used organophosphorus pesticide, may damage the intestine. Here, we evaluated damage to the intestine by CPF at the ADI (0.01 mg/kg bodyweight/day) and at 10 times the ADI (10ADI; 0.1 mg/kg bodyweight/day) in mice after 8 weeks of exposure and evaluated the resulting immune response to an enterotoxigenic Escherichia coli (ETEC) infection. CPF at the ADI dose significantly disrupted the intestinal integrity and intestinal stem cell functionality, which may be associated with reduced indole-3-propionic acid levels. However, mice in the 10ADI group exhibited only elevated pro-inflammatory cell and cytokine levels. During ETEC infection, intestinal mucosal immunity was activated by the 10ADI dose, as indicated by increased regulatory T cells and IL-10 levels, which were associated with decreased fecal butyric acid content. Our study demonstrated that the effects of pesticide residues appear to be dose-specific, bringing attention to the health risk at the ADI level.
Collapse
Affiliation(s)
- Xinlei Yuan
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Fang Wu
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Le Cheng
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Tengteng Ji
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Chenyan Zheng
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Yumeng Ma
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Yutong Jin
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Jianguo Dong
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| | - Yan Jin
- State Key Laboratory of Food Nutrition and Safety, College of Food Science and Engineering, Tianjin University of Science and Technology, Tianjin 300457, China
| | - Bing Fang
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China
| |
Collapse
|
5
|
Ajith TA, Anita B. Impact of Gut Microbiota and Probiotics on Rheumatoid Arthritis: A Potential Treatment Challenge. Int J Rheum Dis 2025; 28:e70266. [PMID: 40329613 DOI: 10.1111/1756-185x.70266] [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: 12/27/2024] [Revised: 04/02/2025] [Accepted: 04/30/2025] [Indexed: 05/08/2025]
Abstract
Over the past few decades, there has been a surge in global study on the relationship between gut microbiota and human health. Numerous human illnesses have been linked to dysbiosis. Gram-positive firmicutes and Gram-negative bacteroidetes are the two leading bacterial phyla that make up 90% of the gut microbiome. Many symbionts in the gut environment establish intricate relationships with host defense to stop both local and non-native dangerous bacteria from colonizing and invading. Dysbiosis alters the paracellular route and damages the epithelium, enabling them to penetrate the epithelium and come into contact with the immune cells. Impaired intestinal barrier function, immune regulation mediated by metabolites derived from the gut microbiota, posttranslational modification of host proteins such as increased citrullination, regulation of the gut microbiota's effect on immune cells, intestinal epithelial cell autophagy, interaction between the microbiome and human leukocyte antigen alleles, and interaction with microRNAs are some of the mechanisms involved in rheumatoid arthritis (RA). The gut microbiota, Prevotella copri, and Collinsella spp. were shown to be higher in the early/preclinical phases of RA, while Bacteroidetes, Bifidobacteria, and Eubacterium rectale were found to be lower. Probiotic-based early dietary intervention may reduce inflammation and slow the rate of joint deterioration, and such intervention can also aid in the restoration of gut microbiota equilibrium. This review article describes the gut microbial dysbiosis and role of probiotics in RA.
Collapse
Affiliation(s)
| | - Bejoy Anita
- Department of General Medicine, Amala Institute of Medical Sciences, Thrissur, Kerala, India
| |
Collapse
|
6
|
Wilson TM. Lung Microbiome in Autoimmune-Associated Interstitial Lung Disease. Rheum Dis Clin North Am 2025; 51:201-212. [PMID: 40246438 DOI: 10.1016/j.rdc.2025.01.003] [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: 04/19/2025]
Abstract
The lung microbiome is a diverse mucosal environment that has been shown to be implicated in the pathogenesis of various chronic lung diseases including insterstitial lung diseases (ILD) such as idiopathic pulmonary fibrosis (IPF). ILD is a well-established manifestation of several types of autoimmune diseases. This review will highlight recent work exploring the role of the lung microbiome in the pathogenesis of autoimmune-related ILD.
Collapse
Affiliation(s)
- Timothy M Wilson
- Division of Rheumatology, Thomas Jefferson University, 211 South 9th Street, Suite 210, Philadelphia, PA 19107, USA.
| |
Collapse
|
7
|
Yin Y, Cheng X, Xie R, Fan D, Li H, Zhong S, Wegner SV, Zeng W, Chen F. Empowering bacteria with light: Optogenetically engineered bacteria for light-controlled disease theranostics and regulation. J Control Release 2025; 383:113787. [PMID: 40311686 DOI: 10.1016/j.jconrel.2025.113787] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2025] [Revised: 04/19/2025] [Accepted: 04/27/2025] [Indexed: 05/03/2025]
Abstract
Bacterial therapy has emerged as a promising approach for disease treatment due to its environmental sensitivity, immunogenicity, and modifiability. However, the clinical application of engineered bacteria is limited by differences of expression levels in patients and possible off-targeting. Optogenetics, which combines optics and genetics, offers key advantages such as remote controllability, non-invasiveness, and precise spatiotemporal control. By utilizing optogenetic tools, the behavior of engineered bacteria can be finely regulated, enabling on-demand control of the dosage and location of their therapeutic products. In this review, we highlight the latest advancements in the optogenetic engineering of bacteria for light-controlled disease theranostics and therapeutic regulation. By constructing a three-dimensional analytical framework of "sense-produce-apply", we begin by discussing the key components of bacterial optogenetic systems, categorizing them based on their photosensitive protein response to blue, green, and red light. Next, we introduce innovative light-producing tools that extend beyond traditional light sources. Then, special emphasis is placed on the biomedical applications of optogenetically engineered bacteria in treating diseases such as cancer, intestinal inflammation and systemic disease regulation. Finally, we address the challenges and future prospects of bacterial optogenetics, outlining potential directions for enhancing the safety and efficacy of light-controlled bacterial therapies. This review aims to provide insights and strategies for researchers working to advance the application of optogenetically engineered bacteria in drug delivery, precision medicine and therapeutic regulation.
Collapse
Affiliation(s)
- Ying Yin
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Xiang Cheng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Ruyan Xie
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Duoyang Fan
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Haohan Li
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China
| | - Shibo Zhong
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster 48149, Germany
| | - Seraphine V Wegner
- Institute of Physiological Chemistry and Pathobiochemistry, University of Münster, Münster 48149, Germany
| | - Wenbin Zeng
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China.
| | - Fei Chen
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410013, China.
| |
Collapse
|
8
|
Romani FE, Luvira V, Chancharoenthana W, Albanese M, Maddaloni L, Branda F, D'Amelio S, Gabrielli S, Scagnolari C, Mastroianni CM, Ceccarelli G, d'Ettorre G. Human microbiota in dengue infection: A narrative review. Microb Pathog 2025; 205:107643. [PMID: 40306589 DOI: 10.1016/j.micpath.2025.107643] [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: 11/24/2024] [Revised: 04/24/2025] [Accepted: 04/28/2025] [Indexed: 05/02/2025]
Abstract
Dengue fever, a widespread mosquito-borne viral infection in tropical regions, typically manifests fever and gastrointestinal symptoms, including nausea, vomiting, and diarrhea. However, the human gut microbiota's role in dengue pathogenesis remains incompletely understood. Studies have demonstrated dysbiosis during dengue virus infection, characterized by increased abundance of potentially pathogenic bacteria like Bacteroidaceae and Proteobacteria, particularly during the critical phase. Furthermore, microbial translocation and leaky gut syndrome, characterized by the translocation of intestinal microbial products, have been observed in dengue patients and are associated with hypercytokinemia, plasma leakage, and disease severity. These findings underscore the necessity for an in-depth investigation into the role of human intestinal microbiota as a potential contributing factor in the pathogenesis and progression of dengue. Further research focusing on human intestinal microbiota, leaky gut syndrome, and the potential implications of treatment with oral bacteriotherapy, as previously observed in other viral diseases, is essential to clarify dengue pathology and evaluate new therapeutic strategies.
Collapse
Affiliation(s)
- Francesco Eugenio Romani
- Department of Public Health and Infectious Diseases, University of Rome Sapienza, Rome, Italy; Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Viravarn Luvira
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Wiwat Chancharoenthana
- Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Mattia Albanese
- Department of Public Health and Infectious Diseases, University of Rome Sapienza, Rome, Italy; Department of Clinical Tropical Medicine, Faculty of Tropical Medicine, Mahidol University, Bangkok, 10400, Thailand
| | - Luca Maddaloni
- Department of Public Health and Infectious Diseases, University of Rome Sapienza, Rome, Italy
| | - Francesco Branda
- Unit of Medical Statistics and Molecular Epidemiology, University Campus Bio-Medico of Rome, 00128, Rome, Italy
| | - Stefano D'Amelio
- Department of Public Health and Infectious Diseases, University of Rome Sapienza, Rome, Italy
| | - Simona Gabrielli
- Department of Public Health and Infectious Diseases, University of Rome Sapienza, Rome, Italy
| | - Carolina Scagnolari
- Department of Molecular Medicine, Laboratory of Virology, University of Rome Sapienza, Rome, Italy
| | | | - Giancarlo Ceccarelli
- Department of Public Health and Infectious Diseases, University of Rome Sapienza, Rome, Italy; Azienda Ospedaliero Universitaria Policlinico Umberto I, Rome, Italy; Migrant and Global Health Research Organization (Mi-HeRO), Rome, Italy.
| | - Gabriella d'Ettorre
- Department of Public Health and Infectious Diseases, University of Rome Sapienza, Rome, Italy
| |
Collapse
|
9
|
Dong Y, Wu X, Zhang Y, Hu A, Zhou Q, Yue X, Liu Z, Li M. The Role of Probiotics in Modulating the Gut Microbiome in Alzheimer's Disease: A Review. Foods 2025; 14:1531. [PMID: 40361614 DOI: 10.3390/foods14091531] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/20/2025] [Accepted: 04/24/2025] [Indexed: 05/15/2025] Open
Abstract
Alzheimer's disease (AD) has emerged as a global public health priority characterized by escalating prevalence and the limited efficacy of current therapeutic approaches. Although the pathological complexity of AD is well-recognized, its underlying etiology remains incompletely elucidated. Current research highlights a bidirectional gut-brain axis (GBA) interaction, wherein gut microbiome perturbations may impair intestinal barrier stability, influence immune responses, and blood-brain barrier permeability through microbial metabolite-mediated pathways, thereby contributing to AD pathophysiology. Notably, probiotics demonstrate therapeutic potential by restoring gut microbiome homeostasis, reinforcing intestinal barrier integrity, and mitigating neuroinflammatory responses via GBA. This review focuses on investigating the gut microbiome alterations in AD pathogenesis, the interaction of probiotics with GBA, and its significance in AD pathogenesis. By synthesizing current clinical evidence, this review aims to establish a scientific foundation for probiotic-based interventions as a novel therapeutic strategy in AD management.
Collapse
Affiliation(s)
- Yushi Dong
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Xilin Wu
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Yumeng Zhang
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Adi Hu
- Liaoning Industrial and Information Technology Development Research Institute, Shenyang 110180, China
| | - Qian Zhou
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Xiqing Yue
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
| | - Zhenmin Liu
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Shanghai 200436, China
| | - Mohan Li
- State Key Laboratory of Dairy Biotechnology, Shanghai Engineering Research Center of Dairy Biotechnology, Shanghai 200436, China
- College of Food Science, Shenyang Agricultural University, Shenyang 110866, China
- School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai 200240, China
| |
Collapse
|
10
|
Buchynskyi M, Kamyshna I, Halabitska I, Petakh P, Kunduzova O, Oksenych V, Kamyshnyi O. Unlocking the gut-liver axis: microbial contributions to the pathogenesis of metabolic-associated fatty liver disease. Front Microbiol 2025; 16:1577724. [PMID: 40351307 PMCID: PMC12061941 DOI: 10.3389/fmicb.2025.1577724] [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: 02/16/2025] [Accepted: 04/07/2025] [Indexed: 05/14/2025] Open
Abstract
Metabolic dysfunction-associated fatty liver disease (MAFLD) is a complex metabolic disorder characterized by hepatic lipid accumulation and subsequent inflammation. This condition is closely linked to metabolic syndrome and obesity, with its prevalence rising due to sedentary lifestyles and high-calorie diets. The pathogenesis of MAFLD involves multiple factors, including insulin resistance, lipotoxicity, oxidative stress, and inflammatory responses. The gut microbiota plays a crucial role in MAFLD development, with dysbiosis contributing to liver inflammation through various mechanisms, such as enhanced intestinal permeability and the translocation of bacterial products like lipopolysaccharide (LPS). Microbial metabolites, including short-chain fatty acids (SCFAs) and bile acids, influence hepatic function and immune responses, with potential implications for disease progression. Specific gut microbiome signatures have been identified in MAFLD patients, offering potential diagnostic and therapeutic targets. Moreover, gut-derived toxins, such as endotoxins, lipopolysaccharides, trimethylamine-N-oxide and bacterial metabolites, significantly influence liver damage and inflammation, highlighting the complex interplay between the gut microbiome and hepatic health. This review comprehensively examines the complex interplay between the gut microbiota and MAFLD, focusing on underlying pathogenic mechanisms, potential biomarkers, and emerging microbiome-targeted therapeutic strategies for disease management.
Collapse
Affiliation(s)
- Mykhailo Buchynskyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Iryna Kamyshna
- Department of Medical Rehabilitation, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Iryna Halabitska
- Department of Therapy and Family Medicine, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| | - Pavlo Petakh
- Department of Biochemistry and Pharmacology, Uzhhorod National University, Uzhhorod, Ukraine
| | - Oksana Kunduzova
- Institute of Metabolic and Cardiovascular Diseases (I2MC), National Institute of Health and Medical Research (INSERM) 1297, Toulouse III University, Toulouse, France
| | - Valentyn Oksenych
- Department of Clinical Science, University of Bergen, Bergen, Norway
| | - Oleksandr Kamyshnyi
- Department of Microbiology, Virology, and Immunology, I. Horbachevsky Ternopil National Medical University, Ternopil, Ukraine
| |
Collapse
|
11
|
Bao M, Wu R, Li J, Tang R, Song C. Research summary, possible mechanisms and perspectives of gut microbiota changes causing precocious puberty. Front Nutr 2025; 12:1596654. [PMID: 40352262 PMCID: PMC12061974 DOI: 10.3389/fnut.2025.1596654] [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: 03/20/2025] [Accepted: 04/08/2025] [Indexed: 05/14/2025] Open
Abstract
The increasing global incidence of precocious puberty, linked to environmental, metabolic, and genetic factors, necessitates innovative therapies beyond gonadotropin-releasing hormone (GnRH) analogs. Accumulating evidence implicates gut microbiota dysbiosis as a pivotal regulator of pubertal timing via interactions with hormone metabolism (e.g., estrogen reactivation via β-glucuronidase), neuroendocrine pathways (nitric oxide signaling), and immune-inflammatory responses. This review delineates taxonomic alterations in central precocious puberty (CPP) and obesity-related subtypes, including Streptococcus enrichment and Alistipes depletion, alongside functional shifts in microbial metabolite production. Mechanistic insights highlight microbiota-driven modulation of the hypothalamic-pituitary-gonadal (HPG) axis, leptin/insulin dynamics, and epigenetic regulation. Emerging interventions-probiotics, fecal microbiota transplantation (FMT), and dietary modifications-demonstrate efficacy in preclinical models and early clinical studies for delaying puberty onset and restoring hormonal balance. Translational efforts to validate these strategies are critical for addressing the clinical and psychosocial challenges posed by precocious puberty, positioning gut microbiota modulation as a novel therapeutic frontier in pediatric endocrinology.
Collapse
Affiliation(s)
- Maorong Bao
- Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Rui Wu
- Ba’nan Hospital Affiliated to Chongqing Medical University, Chongqing, China
| | - Jingwei Li
- Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Runan Tang
- Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| | - Cui Song
- Chongqing Key Laboratory of Pediatrics, Ministry of Education Key Laboratory of Child Development and Disorders, National Clinical Research Center for Child Health and Disorders, Children’s Hospital of Chongqing Medical University, Chongqing, China
| |
Collapse
|
12
|
Mirab F, Pirhaghi M, Otzen DE, Saboury AA. Parkinson's disease and gut microbiota metabolites: The dual impact of vitamins and functional amyloids. Biochim Biophys Acta Mol Basis Dis 2025; 1871:167862. [PMID: 40254265 DOI: 10.1016/j.bbadis.2025.167862] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2025] [Revised: 04/08/2025] [Accepted: 04/17/2025] [Indexed: 04/22/2025]
Abstract
Parkinson's disease (PD) is a neurodegenerative disorder characterized by the abnormal accumulation of alpha-synuclein (α-Syn). Recent research emphasizes the significant role of the gut microbiota, the diverse community of microbes living in the intestines, in modulating α-Syn pathology. This review explores the bi-directional communication along the microbiota-gut-brain axis, highlighting the paradoxical impact of two gut microbiota metabolites-functional bacterial amyloids (FuBA) and vitamins-on neurodegenerative diseases, particularly PD. FuBA contributes to PD pathogenesis by promoting α-Syn aggregation, while vitamins offer neuroprotection through their anti-amyloidogenic, antioxidant, and anti-inflammatory properties. Understanding these processes could lead to precision clinical approaches and novel strategies for managing and preventing PD.
Collapse
Affiliation(s)
- Fatemeh Mirab
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran 1417614335, Iran
| | - Mitra Pirhaghi
- Department of Biological Sciences, Institute for Advanced Studies in Basic Sciences (IASBS), Zanjan 6673145137, Iran
| | - Daniel E Otzen
- Interdisciplinary Nanoscience Center (iNANO), Aarhus University, 8000 Aarhus, C 1592-224, Denmark
| | - Ali Akbar Saboury
- Institute of Biochemistry and Biophysics, University of Tehran, Tehran 1417614335, Iran.
| |
Collapse
|
13
|
Rybicka I, Kaźmierczak Z. The human phageome: niche-specific distribution of bacteriophages and their clinical implications. Appl Environ Microbiol 2025:e0178824. [PMID: 40237489 DOI: 10.1128/aem.01788-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 04/18/2025] Open
Abstract
Bacteriophages (phages) play a crucial role in shaping the composition and diversity of the human microbiome across various body niches. Recent advancements in high-throughput sequencing technologies have enabled comprehensive analysis of the human phageome in different body sites. This review comprehensively analyzes phage populations across major human body niches, examining their distribution and dynamics through recent metagenomic discoveries. We explore how phage-bacteria interactions within different body sites contribute to homeostasis and disease development. Emerging evidence demonstrates that phageome perturbations can serve as early indicators of various disorders, particularly in the gut microbiome. Understanding these complex microbial interactions offers promising opportunities for developing novel diagnostic markers and therapeutic approaches. However, the causal relationship between phages, bacteria, and disease development remains unclear. Further research is needed to elucidate the role of phages in human health and disease and to explore their potential as diagnostic or therapeutic tools. Understanding the intricate interactions between phages, bacteria, and the human host is crucial for unraveling the complexities of the human microbiome and its impact on health and disease.
Collapse
Affiliation(s)
- Izabela Rybicka
- Laboratory of Phage Molecular Biology, Hirszfeld Institute of Immunology and Experimental Therapy, Wrocław, Poland
| | - Zuzanna Kaźmierczak
- Laboratory of Phage Molecular Biology, Hirszfeld Institute of Immunology and Experimental Therapy, Wrocław, Poland
- Research and Development Center, Regional Specialist Hospital in Wrocław, Wrocław, Poland
- Faculty of Medicine, Department of Preclinical Sciences, Pharmacology and Medical Diagnostics, Wrocław University of Science and Technology, Wrocław, Poland
| |
Collapse
|
14
|
Piotin A, Poirot A, Wurm M, Lutz C, Khayath N, de Blay F, Metz-Favre C. A case report of carboxymethylcellulose allergy: exploring tolerance based on administration route. Immunopharmacol Immunotoxicol 2025; 47:234-238. [PMID: 39980337 DOI: 10.1080/08923973.2025.2469214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Accepted: 02/13/2025] [Indexed: 02/22/2025]
Abstract
BACKGROUND The management of hypersensitivity to excipients and food additives remains a significant issue for healthcare professionals and patients. Avoiding carboxymethylcellulose (CMC) can be a considerable challenge for patients allergic to CMC due to its widespread use. We assessed the tolerance of CMC through different route of administration in a patient with a confirmed CMC allergy. We conducted a literature review to analyze all relevant cases of patients allergic to CMC, focusing on tolerance through non-injectable routes. METHODS Skin tests, basophil activation tests, oral and nasal provocation tests with CMC were performed to evaluate patient's hypersensitivity. RESULTS Skin tests and basophil activation tests with CMC were positive and confirmed IgE-mediated hypersensitivity to CMC in the patient. While the patient tolerated oral administration of CMC and CMC-containing eye drops, nasal provocation test resulted in asthma exacerbation and rhinitis. CONCLUSION Tolerance of CMC appears to be route-dependent. Provocation tests with CMC through various routes of administration are essential for assessing tolerance and providing appropriate recommendations for patients with CMC allergy.
Collapse
Affiliation(s)
- Anays Piotin
- Division of Asthma and Allergy, Chest Diseases Department, Strasbourg University Hospital, Strasbourg, France
- Physiology and Functional Exploration Service, University Hospital of Strasbourg, Strasbourg, France
| | - Anh Poirot
- Division of Asthma and Allergy, Chest Diseases Department, Strasbourg University Hospital, Strasbourg, France
| | - Maxence Wurm
- Division of Asthma and Allergy, Chest Diseases Department, Strasbourg University Hospital, Strasbourg, France
| | - Celine Lutz
- Division of Asthma and Allergy, Chest Diseases Department, Strasbourg University Hospital, Strasbourg, France
| | - Naji Khayath
- Division of Asthma and Allergy, Chest Diseases Department, Strasbourg University Hospital, Strasbourg, France
- EA 3070 Federation of Translational Medicine, FHU Homicare, University of Strasbourg, Strasbourg, France
| | - Frédéric de Blay
- Division of Asthma and Allergy, Chest Diseases Department, Strasbourg University Hospital, Strasbourg, France
- EA 3070 Federation of Translational Medicine, FHU Homicare, University of Strasbourg, Strasbourg, France
| | - Carine Metz-Favre
- Division of Asthma and Allergy, Chest Diseases Department, Strasbourg University Hospital, Strasbourg, France
| |
Collapse
|
15
|
Saadh MJ, Ahmed HH, Kareem RA, Sanghvi G, Ganesan S, Agarwal M, Kaur P, Taher WM, Alwan M, Jawad MJ, Hamad AK. Short-chain fatty acids in Huntington's disease: Mechanisms of action and their therapeutic implications. Pharmacol Biochem Behav 2025; 249:173972. [PMID: 39983928 DOI: 10.1016/j.pbb.2025.173972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 02/23/2025]
Abstract
Huntington's disease (HD) is a progressive neurodegenerative disorder characterized by motor dysfunction, cognitive decline, and emotional instability, primarily resulting from the abnormal accumulation of mutant huntingtin protein. Growing research highlights the role of intestinal microbiota and their metabolites, particularly short-chain fatty acids (SCFAs), in modulating HD progression. SCFAs, including acetate, propionate, and butyrate, are produced by gut bacteria through dietary fiber fermentation and are recognized for their neuroprotective properties. Evidence suggests that SCFAs regulate neuroinflammation, neuronal communication, and metabolic functions within the central nervous system (CNS). In HD, these compounds may support neuronal health, reduce oxidative stress, and enhance blood-brain barrier (BBB) integrity. Their mechanisms of action involve binding to G-protein-coupled receptors (GPCRs) and modulating gene expression through epigenetic pathways, underscoring their therapeutic potential. This analysis examines the significance of SCFAs in HD, emphasizing the gut-brain axis and the benefits of dietary interventions aimed at modifying gut microbiota composition and promoting SCFA production. Further research into these pathways may pave the way for novel HD management strategies and improved therapeutic outcomes.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan.
| | | | | | - Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot 360003, Gujarat, India
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Mohit Agarwal
- Department of Pharmaceutical Chemistry, NIMS Institute of Pharmacy, NIMS University, Rajasthan, Jaipur,302131, India
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India
| | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | | | | | | |
Collapse
|
16
|
Dei H, Yokoyama Y, Mizuno T, Asahara T, Igami T, Natsume S, Shimizu Y, Ebata T. Impact of the ratio of fecal short-chain fatty acids to lactic acid concentration on postoperative infectious complications after pancreaticoduodenectomy. Surgery 2025; 180:109040. [PMID: 39756337 DOI: 10.1016/j.surg.2024.109040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/19/2024] [Accepted: 11/26/2024] [Indexed: 01/07/2025]
Abstract
BACKGROUND We previously reported that the balance of short-chain fatty acids and lactic acid in feces affects postoperative infectious complications after major hepatectomy. However, the effect remains unclear in pancreaticoduodenectomy. METHODS Preoperative fecal samples were collected from 210 patients who underwent pancreaticoduodenectomy at 2 institutions between January 2019 and June 2021. Organic acid concentrations were measured per 1 g of feces by high-performance liquid chromatography; the ratio, defined as the sum of acetic, propionic, and butyric acid divided by lactic acid, was calculated. The correlation between the acetic, propionic, and butyric acid divided by lactic acid ratio and postoperative infectious complications was determined using univariate and multivariate analyses. RESULTS Sixty-one patients (29%) had postoperative infectious complications, represented by intra-abdominal abscess, cholangitis, and surgical-site infection. Fecal lactic acid levels ranged from 0.13 to 36.98, with a median of 0.69; the median level was 1.10 μmol/g in the postoperative infectious complications group and 0.36 μmol/g in the nonpostoperative infectious complications group (P < .001). The acetic, propionic, and butyric acid divided by lactic acid ratio ranged from 1.65 to 1,753.4, with a median of 105.8; the median ratio was 59.1 in the postoperative infectious complications group and 198.6 in the nonpostoperative infectious complications group (P = .002). Multivariate analysis revealed that a low acetic, propionic, and butyric acid divided by lactic acid ratio (<75) in the preoperative feces was an independent risk factor for postoperative infectious complications, with an odds ratio of 3.5 and a 95% confidence interval of 1.8-7.0 (P < .001). Preoperative biliary drainage was significantly associated with a low acetic, propionic, and butyric acid divided by lactic acid ratio. CONCLUSION The preoperative fecal organic acid profile determined using the acetic, propionic, and butyric acid divided by lactic acid ratio clinically impacted the incidence of postoperative infectious complications in patients who underwent pancreaticoduodenectomy.
Collapse
Affiliation(s)
- Hideyuki Dei
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yukihiro Yokoyama
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan; Division of Perioperative Medicine, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan.
| | - Takashi Mizuno
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Takashi Asahara
- Yakult Central Institute, Yakult Honsha Co Ltd, Tokyo, Japan
| | - Tsuyoshi Igami
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Seiji Natsume
- Department of Gastroenterological Surgery, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Yasuhiro Shimizu
- Department of Gastroenterological Surgery, Aichi Cancer Center Hospital, Nagoya, Japan
| | - Tomoki Ebata
- Division of Surgical Oncology, Department of Surgery, Nagoya University Graduate School of Medicine, Nagoya, Japan
| |
Collapse
|
17
|
Hromić-Jahjefendić A, Mahmutović L, Sezer A, Bećirević T, Rubio-Casillas A, Redwan EM, Uversky VN. The intersection of microbiome and autoimmunity in long COVID-19: Current insights and future directions. Cytokine Growth Factor Rev 2025; 82:43-54. [PMID: 39179487 DOI: 10.1016/j.cytogfr.2024.08.002] [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: 07/15/2024] [Revised: 08/08/2024] [Accepted: 08/13/2024] [Indexed: 08/26/2024]
Abstract
Long COVID-19 affects a significant percentage of patients and is characterized by a wide range of symptoms, including weariness and mental fog as well as emotional symptoms like worry and sadness. COVID-19 is closely linked to the autoimmune disorders that are becoming more prevalent worldwide and are linked to immune system hyperactivation, neutrophil extracellular trap (NET) development, and molecular mimicry pathways. Long-term COVID-related autoimmune responses include a watchful immune system referring to the ability of immune system to constantly monitor the body for signs of infection, disease, or abnormal cells; altered innate and adaptive immune cells, autoantigens secreted by living or dead neutrophils, and high concentrations of autoantibodies directed against different proteins. The microbiome, which consists of billions of bacteria living in the human body, is essential for controlling immune responses and supporting overall health. The microbiome can affect the course of long COVID-associated autoimmunity, including the degree of illness, the rate of recovery, and the onset of autoimmune reactions. Although the precise role of the microbiome in long COVID autoimmunity is still being investigated, new studies indicate that probiotics, prebiotics, and dietary changes-interventions that target the microbiome-may be able to reduce autoimmune reactions and enhance long-term outcomes for COVID-19 survivors. More research is required to precisely understand how the microbiome affects COVID-19-related autoimmunity and to create tailored treatment plans.
Collapse
Affiliation(s)
- Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka cesta 15, Sarajevo 71000, Bosnia and Herzegovina.
| | - Lejla Mahmutović
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka cesta 15, Sarajevo 71000, Bosnia and Herzegovina.
| | - Abas Sezer
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Hrasnicka cesta 15, Sarajevo 71000, Bosnia and Herzegovina.
| | - Tea Bećirević
- Atrijum Polyclinic, Sarajevo, Bosnia and Herzegovina
| | - Alberto Rubio-Casillas
- Autlan Regional Hospital, Health Secretariat, Autlan, Jalisco 48900, Mexico; Biology Laboratory, Autlan Regional Preparatory School, University of Guadalajara, Autlan, Jalisco 48900, Mexico.
| | - Elrashdy M Redwan
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah 21589, Saudi Arabia; Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City of Scientific Research and Technological Applications, New Borg EL-Arab 21934, Alexandria, Egypt.
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, 12901 Bruce B. Downs Blvd., MDC07, Tampa, FL, USA.
| |
Collapse
|
18
|
Yu M, Xu M, Wang G, Feng J, Zhang M. Effects of different photoperiods on melatonin level, cecal microbiota and breast muscle morphology of broiler chickens. Front Microbiol 2025; 16:1504264. [PMID: 40201434 PMCID: PMC11975912 DOI: 10.3389/fmicb.2025.1504264] [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: 09/30/2024] [Accepted: 03/10/2025] [Indexed: 04/10/2025] Open
Abstract
Long photoperiods are often characterized by enhanced oxidative stress-induced damage to skeletal muscle, reduced melatonin (MT) levels and intestinal microbiota dysfunction in broilers. In this study, we aimed to investigate the association of breast muscle morphology with melatonin levels and the cecal microbiota of broilers under different photoperiods. A total of 216 healthy 5-day-old Arbor Acres (AA) male broilers were randomly assigned to 12 L:12D, 18 L:6D and 24 L:0D photoperiods for 4 weeks (L = hours of light, D = hours of darkness). The concentration of inflammatory factors and MT concentrations was measured using ELISA kits, whereas breast muscle morphology was examined through the hematoxylin (H) and eosin (E) staining, and microbiota composition was identified through 16 s rRNA analysis. Extended light exposure significantly improved the growth rate of broilers, but significantly decreased feed efficiency (FE). Furthermore, it upregulated the concentration of IL-1β, IL-6 and TNF-α and induced an abnormal breast muscle morphology. Extended light exposure significantly decreased MT levels in the hypothalamus, cecum and breast muscle, while triggering the cecal microbiota composition disorder. Specifically, there was significant alteration to the dominant bacterial phylum, following exposure to long photoperiods, with the abundance of Firmicutes decreasing and the abundance of Bacteroidota increasing. Notably, the relative abundance of Lactobacillus showed a positive correlation with MT levels and a negative correlation with inflammatory cytokines. In conclusion, the present findings indicated that extended light exposure reduced the MT levels, which were related to disturbed cecal microbiota, damaging breast muscle morphology and inducing breast muscle inflammation in broilers.
Collapse
Affiliation(s)
- Miao Yu
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Mengjie Xu
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Guangju Wang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
- Adaptation Physiology Group, Wageningen University and Research, Wageningen, Netherlands
| | - Jinghai Feng
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| | - Minhong Zhang
- State Key Laboratory of Animal Nutrition and Feeding, Institute of Animal Sciences, Chinese Academy of Agricultural Sciences, Beijing, China
| |
Collapse
|
19
|
Gohain BB, Mazumder B, Rajkhowa S, Al-Hussain SA, Zaki MEA. Subtractive genomics and drug repurposing strategies for targeting Streptococcus pneumoniae: insights from molecular docking and dynamics simulations. Front Microbiol 2025; 16:1534659. [PMID: 40170924 PMCID: PMC11958985 DOI: 10.3389/fmicb.2025.1534659] [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: 11/26/2024] [Accepted: 02/28/2025] [Indexed: 04/03/2025] Open
Abstract
Introduction Streptococcus pneumoniae is a Gram-positive bacterium responsible for severe infections such as meningitis and pneumonia. The increasing prevalence of antibiotic resistance necessitates the identification of new therapeutic targets. This study aimed to discover potential drug targets against S. pneumoniae using an in silico subtractive genomics approach. Methods The S. pneumoniae genome was compared to the human genome to identify non-homologous sequences using CD-HIT and BLASTp. Essential genes were identified using the Database of Essential Genes (DEG), with consideration for human gut microflora. Protein-protein interaction analyses were conducted to identify key hub genes, and gene ontology (GO) studies were performed to explore associated pathways. Due to the lack of crystal structure data, a potential target was modeled in silico and subjected to structure-based virtual screening. Results Approximately 2,000 of the 2,027 proteins from the S. pneumoniae genome were identified as non-homologous to humans. The DEG identified 48 essential genes, which was reduced to 21 after considering human gut microflora. Key hub genes included gpi, fba, rpoD, and trpS, associated with 20 pathways. Virtual screening of 2,509 FDA-approved compounds identified Bromfenac as a leading candidate, exhibiting a binding energy of -26.335 ± 29.105 kJ/mol. Discussion Bromfenac, particularly when conjugated with AuAgCu2O nanoparticles, has demonstrated antibacterial and anti-inflammatory properties against Staphylococcus aureus. This suggests that Bromfenac could be repurposed as a potential therapeutic agent against S. pneumoniae, pending further experimental validation. The approach highlights the potential for drug repurposing by targeting proteins essential in pathogens but absent in the host.
Collapse
Affiliation(s)
- Borakha Bura Gohain
- Centre for Biotechnology and Bioinformatics, Dibrugarh University, Dibrugarh, Assam, India
| | - Bhaskar Mazumder
- Department of Pharmaceutical Sciences, Dibrugarh University, Dibrugarh, Assam, India
| | - Sanchaita Rajkhowa
- Centre for Biotechnology and Bioinformatics, Dibrugarh University, Dibrugarh, Assam, India
| | - Sami A. Al-Hussain
- Department of Chemistry, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| | - Magdi E. A. Zaki
- Department of Chemistry, Imam Mohammad Ibn Saud Islamic University (IMSIU), Riyadh, Saudi Arabia
| |
Collapse
|
20
|
Luu QQ, Kim T, Cao TBT, Choi I, Yang SY, An BS, Hwang DY, Choi Y, Park HS. Therapeutic Potential of Arginine-Loaded Red Blood Cell Nanovesicles Targeting Obese Asthma. Mediators Inflamm 2025; 2025:8248722. [PMID: 40134943 PMCID: PMC11936518 DOI: 10.1155/mi/8248722] [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: 11/20/2024] [Accepted: 02/12/2025] [Indexed: 03/27/2025] Open
Abstract
Purpose: The role of the gut microbiomes has been emphasized in the pathogenesis of obese asthma (OA). However, the molecular mechanism of airway dysfunction underlying OA has not yet been fully elucidated. The effects of microbiomes on arginine metabolism in relation to lung functions and a novel method for delivering arginine to lung tissue based on arginine-loaded red blood cell (RBC)-derived nanovesicles (NVs) (NVArg) will be investigated. Materials and Methods: Inflammatory status, amino acid profiles, and microbial diversity were evaluated in 20 adult patients with OA compared to 30 adult patients with non-OA (NOA) and 10 healthy control (HC) groups. Changes in gut or lung microbial composition that altered arginine metabolism in relation to airway inflammation were investigated in an OA mouse model in vivo. Additionally, this study evaluated the delivery of arginine to lung tissue utilizing NVArg in vivo and in vitro. Results: Significantly increased Bacteroides abundance but decreased serum arginine concentration with lower forced exhaled volume at 1 s (FEV1) (%) was noted in the OA group compared to the NOA and HC groups. In mouse experiments, when OA mice were given living bacteria from normal control (NC) mice, lung arginine concentration and airway resistance were restored. However, the administration of arginine or its metabolite (citrulline) did not increase the arginine levels in the lung tissues. We therefore created NVArg, which successfully delivered arginine into the cytoplasm of the airway epithelial cell line in vitro. Oral administration of NVArg for OA mice significantly induced the AMP-activated protein kinase (AMPK) and endothelial nitric oxide synthase (eNOS) pathways in airway epithelial cells, which reduced airway resistance and inflammation. Conclusion: These findings suggest that microbiomes contribute to airway dysfunction by regulating arginine metabolism, whereas NVArg treatment may be a potential option for managing OA.
Collapse
Affiliation(s)
- Quoc Quang Luu
- Department of Oral and Maxillofacial Surgery, Loma Linda University School of Dentistry, Loma Linda, California, USA
| | - Taejune Kim
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science, Pusan National University, Miryang, Republic of Korea
| | - Thi Bich Tra Cao
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Republic of Korea
| | - Injung Choi
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science, Pusan National University, Miryang, Republic of Korea
| | - Seung Yun Yang
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science, Pusan National University, Miryang, Republic of Korea
| | - Beum-Soo An
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science, Pusan National University, Miryang, Republic of Korea
| | - Dae Youn Hwang
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science, Pusan National University, Miryang, Republic of Korea
| | - Youngwoo Choi
- Department of Biomaterials Science (BK21 FOUR Program), College of Natural Resources and Life Science, Pusan National University, Miryang, Republic of Korea
| | - Hae-Sim Park
- Department of Allergy and Clinical Immunology, Ajou University School of Medicine, Suwon, Republic of Korea
| |
Collapse
|
21
|
Bai Y, Hu Y, Chen X, Hu L, Wu K, Liang S, Zheng J, Gänzle MG, Chen C. Comparative metagenome-associated analysis of gut microbiota and antibiotic resistance genes in acute gastrointestinal injury patients with the risk of in-hospital mortality. mSystems 2025; 10:e0144424. [PMID: 40013797 PMCID: PMC11915821 DOI: 10.1128/msystems.01444-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Accepted: 02/03/2025] [Indexed: 02/28/2025] Open
Abstract
Acute gastrointestinal injury (AGI) is known for its poor long-term prognosis and the associated increase in mortality among intensive care unit (ICU) patients. As the role of the gut microbiome and resistome in AGI remains unclear, the present study aimed to explore the possible associations between dysbacteriosis and in-hospital mortality in ICU patients with gastrointestinal dysfunction. Fecal samples were collected from a prospective cohort of 210 ICU patients with AGI, and shotgun metagenomic sequencing was used to determine the taxonomic composition of gut microbiota and the differences of antibiotic resistance genes (ARGs) between the Death and Survival groups. Compared to the Survival group, patients in the Death group shifted from strict anaerobes to facultative anaerobes in the fecal microbial community, with more Klebsiella but less Prevotella. The co-occurrence patterns revealed that more ARG subtypes were enriched in microbial taxa in the Death group, especially for Clostridium and Methanobrevibacter. Furthermore, the ARG type had large area under the curve (AUCs) in receiver operating characteristic for predicting the disease severity, and a combined gut microbiota-ARG subtype classifiers showed better performance than either of them. Thus, comparative metagenome-associated analysis can help to obtain valuable information about gut microbiota and gene coding for antibiotic resistance in AGI patients. IMPORTANCE A metagenomic-related strategy was conducted to obtain a highly valuable resource to improve understanding of intestinal microbiota dysbiosis and antibiotic resistance genes (ARGs) profiles. The results indicate that intestinal microbiota, including Klebsiella and Prevotella, changed dramatically in intensive care unit (ICU) patients with acute gastrointestinal injury (AGI). Due to longer ICU stays and receiving more antibiotic treatment, the types and correlations of ARGs in the Death group were significantly higher than those in the Survival group. The findings of this study are expected to expand our knowledge of gut microbiota and resistome profiles reflecting gastrointestinal status, accelerate the identification of disease biomarkers, and provide new insights into the prevention and treatment of AGI-related diseases.
Collapse
Affiliation(s)
- Yunpeng Bai
- Department of Pharmacy, Shenzhen People’s Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Yali Hu
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Xiangyin Chen
- Department of Critical Care Medicine, Shenzhen People’s Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
- Department of Surgery Intensive Care Medicine, Maoming People’s Hospital, Maoming, China
| | - Linhui Hu
- Department of Critical Care Medicine, Maoming People’s Hospital, Maoming, China
| | - Kunyong Wu
- Center of Scientific Research, Maoming People’s Hospital, Maoming, China
- Biological Resource Center of Maoming People’s Hospital, Maoming, China
| | - Silin Liang
- Department of Critical Care Medicine, Shenzhen People’s Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| | - Jinshui Zheng
- State Key Laboratory of Agricultural Microbiology, Hubei Hongshan Laboratory, Huazhong Agricultural University, Wuhan, China
- Hubei Key Laboratory of Agricultural Bioinformatics, College of Informatics, Huazhong Agricultural University, Wuhan, China
| | - Michael G. Gänzle
- Department of Agricultural, Food, and Nutritional Science, University of Alberta, 4-10 Ag/For Center, Edmonton, Canada
- Hubei University of Technology, College of Bioengineering and Food Science, Wuhan, China
| | - Chunbo Chen
- Department of Critical Care Medicine, Shenzhen People’s Hospital, The Second Clinical Medical College of Jinan University, The First Affiliated Hospital of Southern University of Science and Technology, Shenzhen, China
| |
Collapse
|
22
|
Attiq A. Early-life antibiotic exposures: Paving the pathway for dysbiosis-induced disorders. Eur J Pharmacol 2025; 991:177298. [PMID: 39864578 DOI: 10.1016/j.ejphar.2025.177298] [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: 11/17/2024] [Revised: 01/06/2025] [Accepted: 01/22/2025] [Indexed: 01/28/2025]
Abstract
Microbiota encompasses a diverse array of microorganisms inhabiting specific ecological niches. Gut microbiota significantly influences physiological processes, including gastrointestinal motor function, neuroendocrine signalling, and immune regulation. They play a crucial role in modulating the central nervous system and bolstering body defence mechanisms by influencing the proliferation and differentiation of innate and adaptive immune cells. Given the potential consequences of antibiotic therapy on gut microbiota equilibrium, there is a need for prudent antibiotic use to mitigate associated risks. Observational studies have linked increased antibiotic usage to various pathogenic conditions, including obesity, inflammatory bowel disease, anxiety-like effects, asthma, and pulmonary carcinogenesis. Addressing dysbiosis incidence requires proactive measures, including prophylactic use of β-lactamase drugs (SYN-004, SYN-006, and SYN-007), hydrolysing the β-lactam in the proximal GIT for maintaining intestinal flora homeostasis. Prebiotic and probiotic supplementations are crucial in restoring intestinal flora equilibrium by competing with pathogenic bacteria for nutritional resources and adhesion sites, reducing luminal pH, neutralising toxins, and producing antimicrobial agents. Faecal microbiota transplantation (FMT) shows promise in restoring gut microbiota composition. Rational antibiotic use is essential to preserve microflora and improve patient compliance with antibiotic regimens by mitigating associated side effects. Given the significant implications on gut microbiota composition, concerted intervention strategies must be pursued to rectify and reverse the occurrence of antibiotic-induced dysbiosis. Here, antibiotics-induced microbiota dysbiosis mechanisms and their systemic implications are reviewed. Moreover, proposed interventions to mitigate the impact on gut microflora are also discussed herein.
Collapse
Affiliation(s)
- Ali Attiq
- School of Pharmaceutical Sciences, Universiti Sains Malaysia, Gelugor, 11800, Penang, Malaysia.
| |
Collapse
|
23
|
Yu J, Li Y, Zhu B, Shen J, Miao L. Research progress on the kidney-gut-brain axis in brain dysfunction in maintenance hemodialysis patients. Front Med (Lausanne) 2025; 12:1538048. [PMID: 40115780 PMCID: PMC11922870 DOI: 10.3389/fmed.2025.1538048] [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: 12/02/2024] [Accepted: 02/14/2025] [Indexed: 03/23/2025] Open
Abstract
Maintenance hemodialysis (MHD) has become the primary renal replacement therapy for patients with end-stage renal disease. The kidney-gut-brain axis represents a communication network connecting the kidney, intestine and brain. In MHD patients, factors such as uremic toxins, hemodynamic changes, vascular damage, inflammation, oxidative stress, and intestinal dysbiosis in MHD patients refers to a range of clinical syndromes, including brain injury, and is manifested by conditions such as white matter disease, brain atrophy, cerebrovascular disease, cognitive impairment, depression, anxiety, and other behavioral or consciousness abnormalities. Numerous studies have demonstrated the prevalence of these brain disorders in MHD patients. Understanding the mechanisms of brain disorders in MHD patients, particularly through the lens of kidney-gut-brain axis dysfunction, offers valuable insights for future research and the development of targeted therapies. This article reviews the brain dysfunction associated with MHD, the impact of the kidney-brain axis, intestinal barrier damage, gut microbiota dysbiosis caused by MHD, and the role of the gut-brain axis in brain dysfunction.
Collapse
Affiliation(s)
- Jie Yu
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Yulu Li
- Department of Nephrology, Taicang Loujiang New City Hospital, Suzhou, China
| | - Bin Zhu
- Department of Critical Care Medicine, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Jianqin Shen
- Department of Blood Purification Center, The Third Affiliated Hospital of Soochow University, Changzhou, China
| | - Liying Miao
- Department of Nephrology, The Third Affiliated Hospital of Soochow University, Changzhou, China
| |
Collapse
|
24
|
Zattoni F, Novara G, Dal Moro F, Tandogdu Z. Prostate Biopsy: The Transperineal Approach Is Better! Eur Urol Focus 2025:S2405-4569(25)00054-9. [PMID: 40037972 DOI: 10.1016/j.euf.2025.02.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 01/31/2025] [Accepted: 02/21/2025] [Indexed: 03/06/2025]
Abstract
Transperineal prostate biopsy offers advantages over transrectal biopsy in terms of cancer detection in specific prostate areas, potential use of larger-gauge needles for tissue sampling, a lower risk of severe infectious complications such as sepsis, and lower use of antibiotic prophylaxis, aligning with antibiotic stewardship policies and reducing the risk of antimicrobial resistance and gut microbiome changes.
Collapse
Affiliation(s)
- Fabio Zattoni
- Urology Clinic, Department of Surgery, Oncology, and Gastroenterology, University of Padua, Padua, Italy; Department of Medicine, University of Padua, Padua, Italy.
| | - Giacomo Novara
- Urology Clinic, Department of Surgery, Oncology, and Gastroenterology, University of Padua, Padua, Italy
| | - Fabrizio Dal Moro
- Urology Clinic, Department of Surgery, Oncology, and Gastroenterology, University of Padua, Padua, Italy
| | - Zafer Tandogdu
- Department of Urology, University College London Hospitals, London, UK; Division of Surgery and Interventional Science, University College London, London, UK
| |
Collapse
|
25
|
Zattoni F, Novara G, Dal Moro F, Tandogdu Z. Prostate Biopsy: The Transperineal Approach is Better! Eur Urol Focus 2025:S2405-4569(25)00050-1. [PMID: 40037974 DOI: 10.1016/j.euf.2025.02.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/13/2025] [Accepted: 02/20/2025] [Indexed: 03/06/2025]
Abstract
Transperineal prostate biopsy offers advantages over transrectal biopsy in terms of cancer detection in specific prostate areas, potential use of larger-gauge needles for tissue sampling, a lower risk of severe infectious complications such as sepsis, and lower use of antibiotic prophylaxis, aligning with antibiotic stewardship policies and reducing the risk of antimicrobial resistance and gut microbiome changes.
Collapse
Affiliation(s)
- Fabio Zattoni
- Urology Clinic, Department of Surgery, Oncology, and Gastroenterology, University of Padua, Padua, Italy; Department of Medicine, University of Padua, Padua, Italy.
| | - Giacomo Novara
- Urology Clinic, Department of Surgery, Oncology, and Gastroenterology, University of Padua, Padua, Italy
| | - Fabrizio Dal Moro
- Urology Clinic, Department of Surgery, Oncology, and Gastroenterology, University of Padua, Padua, Italy
| | - Zafer Tandogdu
- Department of Urology, University College London Hospitals, London, UK; Division of Surgery and Interventional Science, University College London, London, UK
| |
Collapse
|
26
|
Mak JWY, Lo ATW, Ng SC. Early life factors, diet and microbiome, and risk of inflammatory bowel disease. J Can Assoc Gastroenterol 2025; 8:S44-S50. [PMID: 39990509 PMCID: PMC11842909 DOI: 10.1093/jcag/gwae039] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/25/2025] Open
Abstract
Inflammatory bowel diseases (IBD), including Crohn's disease (CD) and ulcerative colitis (UC), result from a loss of immune tolerance to gut microbiota, leading to inflammation. Their incidence is increasing, especially in newly industrialized countries. The etiology is multifactorial, involving genetic, immune, microbiota, and environmental factors. Maternal microbiome changes during pregnancy can elevate IBD risk in offspring, influenced by diet, smoking, and antibiotic exposure. Early life microbiota manipulation shows promise for preventing IBD. Epidemiological and pre-clinical studies highlight diet's significant role in IBD development. High-inflammatory dietary patterns correlate with increased CD risk, while Mediterranean-like diets promote beneficial gut microbiome changes and reduce inflammation. Certain food additives, such as emulsifiers and artificial sweeteners, may exacerbate IBD by altering gut microbiota. A systematic review indicates that higher ultra-processed food consumption significantly increases CD risk. Lifestyle modifications, including healthy dietary adherence, could substantially reduce IBD risk, with studies showing that favorable choices can halve the risk in genetically predisposed individuals. Additionally, maternal diet impacts offspring IBD risk, as seen in mouse models where high-fat diets led to increased inflammation. Evidence suggests that maternal probiotics and specific dietary patterns may mitigate these risks. Overall, these findings emphasize the potential for dietary interventions to modulate gut microbiota and immune responses, offering promising avenues for IBD prevention and management. Further large-scale studies are needed to explore the impact of dietary strategies on IBD risk and gut health.
Collapse
Affiliation(s)
- Joyce Wing Yan Mak
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Aaron Tsz Wang Lo
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong
| | - Siew Chien Ng
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Shatin, Hong Kong
- Microbiota I-Center (MagIC), Shatin, Hong Kong
- Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Shatin, Hong Kong
- State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Shatin, Hong Kong
| |
Collapse
|
27
|
Makkar H, Sriram G. Advances in modeling periodontal host-microbe interactions: insights from organotypic and organ-on-chip systems. LAB ON A CHIP 2025; 25:1342-1371. [PMID: 39963082 PMCID: PMC11833442 DOI: 10.1039/d4lc00871e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 01/27/2025] [Indexed: 02/20/2025]
Abstract
Periodontal disease, a chronic inflammatory condition affecting the supporting structures of teeth, is driven by an imbalanced interaction between the periodontal microbiota and the host inflammatory response. Beyond its local impact, periodontal disease is associated with systemic conditions such as diabetes mellitus, cardiovascular disease, and inflammatory bowel disease, emphasizing the importance of understanding its mechanisms. Traditional pre-clinical models, such as monolayer cultures and animal studies, have provided foundational insights but are limited by their physiological relevance and ethical concerns. Recent advancements in tissue engineering and microfluidic technologies have led to the development of three-dimensional (3D) organotypic culture models and organ-on-chip systems that more closely mimic native tissue microenvironments. This review provides an overview of the evolution of methods to study periodontal host-microbe interactions, from simple 2D monolayer cultures to complex 3D organotypic and microfluidic organ-on-chip (OoC) models. We discuss various fabrication strategies, host-microbe co-culture techniques, and methods for evaluating outcomes in these advanced models. Additionally, we highlight insights gained from gut-on-chip platforms and their potential applications in periodontal research and understanding oral-systemic links of periodontal disease. Through a comprehensive overview of current advancements and future directions, this review provides insights on the transformative potential of OoC technology in periodontal research, offering new avenues for studying disease mechanisms and developing therapeutic strategies.
Collapse
Affiliation(s)
- Hardik Makkar
- Faculty of Dentistry, National University of Singapore, 119085, Singapore.
- Center for Innovation & Precision Dentistry, School of Dental Medicine and School of Engineering, University of Pennsylvania, Philadelphia, PA, 19104, USA.
| | - Gopu Sriram
- Faculty of Dentistry, National University of Singapore, 119085, Singapore.
- Department of Biomedical Engineering, College of Design and Engineering, National University of Singapore, 117583, Singapore
| |
Collapse
|
28
|
Beretta S, de Araújo RA, Bianchini MO, Bonavina JT, Rocha-Júnior JD, Campos NC, Pizauro LJL, Rodrigues-Silva FA, Toniollo GH, Cardozo MV, Apparício M. Vaginal Seeding: Is There Any Positive Effect in Canine C-Sections? Animals (Basel) 2025; 15:416. [PMID: 39943186 PMCID: PMC11816153 DOI: 10.3390/ani15030416] [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: 09/30/2024] [Revised: 12/19/2024] [Accepted: 01/02/2025] [Indexed: 02/16/2025] Open
Abstract
This study aimed to scrutinize variations in the intestinal microbiota of neonatal dogs born through natural birth versus elective cesarean section, focusing on evaluating the influence of vaginal seeding on the microbiota of cesarean-born neonates. Samples were collected from cesarean-sectioned females before anesthesia and from naturally birthing females during prodrome signs, along with neonates at eight time points from birth to 15 days of age. In the cesarean section group, seeding was performed in half of the neonates (cesarean section seeding group; seeding consisted of gently rubbing the gauze, obtained from the mother's vagina, onto the mouths, faces, and bodies of the newborns), while the other half underwent microbiological sample collection without seeding (cesarean section group). Another group (normal birth group) consisted of naturally born neonates. Microbiota analysis included counting for enterobacteria, Staphylococcus spp., and Streptococcus spp. The results suggested that vertical transmission played a crucial role, but the method of birth did not emerge as the primary determinant of observed differences. Under study conditions, vaginal seeding failed to effectively modulate the microbiota of neonates born through elective cesarean section. Further investigations into the gut-brain axis are suggested for understanding factors influencing the initial development of the canine intestinal microbiota in neonates born through different delivery routes.
Collapse
Affiliation(s)
- Samara Beretta
- Department of Pathology, Reproduction and One Health, School of Agricultural and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal 14884-900, SP, Brazil; (S.B.); (R.A.d.A.); (M.O.B.); (J.D.R.-J.); (N.C.C.); (G.H.T.); (M.V.C.)
| | - Renatha Almeida de Araújo
- Department of Pathology, Reproduction and One Health, School of Agricultural and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal 14884-900, SP, Brazil; (S.B.); (R.A.d.A.); (M.O.B.); (J.D.R.-J.); (N.C.C.); (G.H.T.); (M.V.C.)
| | - Melissa Oliveira Bianchini
- Department of Pathology, Reproduction and One Health, School of Agricultural and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal 14884-900, SP, Brazil; (S.B.); (R.A.d.A.); (M.O.B.); (J.D.R.-J.); (N.C.C.); (G.H.T.); (M.V.C.)
| | - Jaqueline Tamara Bonavina
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu 18618-681, SP, Brazil; (J.T.B.); (F.A.R.-S.)
| | - João Domingos Rocha-Júnior
- Department of Pathology, Reproduction and One Health, School of Agricultural and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal 14884-900, SP, Brazil; (S.B.); (R.A.d.A.); (M.O.B.); (J.D.R.-J.); (N.C.C.); (G.H.T.); (M.V.C.)
| | - Nayara Camatta Campos
- Department of Pathology, Reproduction and One Health, School of Agricultural and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal 14884-900, SP, Brazil; (S.B.); (R.A.d.A.); (M.O.B.); (J.D.R.-J.); (N.C.C.); (G.H.T.); (M.V.C.)
| | - Lucas José Luduverio Pizauro
- Department of Agricultural and Environmental Sciences, State University of Santa Cruz (UESC), Ilhéus 45662-900, BA, Brazil;
| | - Fernanda Andreza Rodrigues-Silva
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu 18618-681, SP, Brazil; (J.T.B.); (F.A.R.-S.)
| | - Gilson Hélio Toniollo
- Department of Pathology, Reproduction and One Health, School of Agricultural and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal 14884-900, SP, Brazil; (S.B.); (R.A.d.A.); (M.O.B.); (J.D.R.-J.); (N.C.C.); (G.H.T.); (M.V.C.)
| | - Marita Vedovelli Cardozo
- Department of Pathology, Reproduction and One Health, School of Agricultural and Veterinary Sciences, São Paulo State University (UNESP), Jaboticabal 14884-900, SP, Brazil; (S.B.); (R.A.d.A.); (M.O.B.); (J.D.R.-J.); (N.C.C.); (G.H.T.); (M.V.C.)
| | - Maricy Apparício
- Department of Veterinary Surgery and Animal Reproduction, School of Veterinary Medicine and Animal Science, São Paulo State University (UNESP), Botucatu 18618-681, SP, Brazil; (J.T.B.); (F.A.R.-S.)
| |
Collapse
|
29
|
Lista S, Munafò A, Caraci F, Imbimbo C, Emanuele E, Minoretti P, Pinto-Fraga J, Merino-País M, Crespo-Escobar P, López-Ortiz S, Monteleone G, Imbimbo BP, Santos-Lozano A. Gut microbiota in Alzheimer's disease: Understanding molecular pathways and potential therapeutic perspectives. Ageing Res Rev 2025; 104:102659. [PMID: 39800223 DOI: 10.1016/j.arr.2025.102659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Revised: 11/29/2024] [Accepted: 01/09/2025] [Indexed: 01/15/2025]
Abstract
Accumulating evidence suggests that gut microbiota (GM) plays a crucial role in Alzheimer's disease (AD) pathogenesis and progression. This narrative review explores the complex interplay between GM, the immune system, and the central nervous system in AD. We discuss mechanisms through which GM dysbiosis can compromise intestinal barrier integrity, enabling pro-inflammatory molecules and metabolites to enter systemic circulation and the brain, potentially contributing to AD hallmarks. Additionally, we examine other pathophysiological mechanisms by which GM may influence AD risk, including the production of short-chain fatty acids, secondary bile acids, and tryptophan metabolites. The role of the vagus nerve in gut-brain communication is also addressed. We highlight potential therapeutic implications of targeting GM in AD, focusing on antibiotics, probiotics, prebiotics, postbiotics, phytochemicals, and fecal microbiota transplantation. While preclinical studies showed promise, clinical evidence remains limited and inconsistent. We critically assess clinical trials, emphasizing challenges in translating GM-based therapies to AD patients. The reviewed evidence underscores the need for further research to elucidate precise molecular mechanisms linking GM to AD and determine whether GM dysbiosis is a contributing factor or consequence of AD pathology. Future studies should focus on large-scale clinical trials to validate GM-based interventions' efficacy and safety in AD.
Collapse
Affiliation(s)
- Simone Lista
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain.
| | - Antonio Munafò
- Department of Neurosciences, Psychology, Drug Research and Child Health, University of Florence, Florence 50139, Italy.
| | - Filippo Caraci
- Department of Drug and Health Sciences, University of Catania, Catania 95125, Italy; Oasi Research Institute-IRCCS, Troina 94018, Italy.
| | - Camillo Imbimbo
- Department of Brain and Behavioral Sciences, University of Pavia, Pavia 27100, Italy.
| | | | | | - José Pinto-Fraga
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain.
| | - María Merino-País
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain.
| | - Paula Crespo-Escobar
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain.
| | - Susana López-Ortiz
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain.
| | - Giovanni Monteleone
- Department of Systems Medicine, University of Rome Tor Vergata, Rome 00133, Italy; Unit of Gastroenterology, Policlinico Tor Vergata University Hospital, Rome 00133, Italy.
| | - Bruno P Imbimbo
- Department of Research and Development, Chiesi Farmaceutici, Parma 43122, Italy.
| | - Alejandro Santos-Lozano
- i+HeALTH Strategic Research Group, Department of Health Sciences, Miguel de Cervantes European University (UEMC), Valladolid 47012, Spain; Physical Activity and Health Research Group (PaHerg), Research Institute of the Hospital 12 de Octubre ('imas12'), Madrid 28041, Spain.
| |
Collapse
|
30
|
Wang Y, Wu W, Zeng F, Meng X, Peng M, Wang J, Chen Z, Liu W. The role of kynurenine pathway metabolism mediated by exercise in the microbial-gut-brain axis in Alzheimer's disease. Exp Neurol 2025; 384:115070. [PMID: 39603488 DOI: 10.1016/j.expneurol.2024.115070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 11/29/2024]
Abstract
In recent years, the role of the microbiome-gut-brain axis in the pathogenesis of Alzheimer's disease (AD) has garnered increasing attention. Specifically, tryptophan metabolism via the kynurenine pathway (KP) plays a crucial regulatory role in this axis. This study reviews how exercise regulates the microbiome-gut-brain axis by influencing kynurenine pathway metabolism, thereby exerting resistance against AD. This paper also discusses how exercise positively impacts AD via the microbiome-gut-brain axis by modulating the endocrine, autonomic nervous, and immune systems. Although the specific mechanisms are not fully understood, research indicates that exercise may optimize tryptophan metabolism by promoting the growth of beneficial microbiota and inhibiting harmful microbiota, producing substances that are beneficial to the nervous system and combating AD. The aim of this review is to provide new perspectives and potential intervention strategies for the prevention and treatment of AD by exploring the links between exercise, KP and the gut-brain axis.
Collapse
Affiliation(s)
- Yiyang Wang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Weijia Wu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Fanqi Zeng
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Xiangyuan Meng
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Mei Peng
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Juan Wang
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Zeyu Chen
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China
| | - Wenfeng Liu
- Hunan Provincial Key Laboratory of Physical Fitness and Sports Rehabilitation, Hunan Normal University, Changsha 410012, China; Key Laboratory of Protein Chemistry and Developmental Biology of Ministry of Education, Hunan Normal University, Changsha 410081, China.
| |
Collapse
|
31
|
Jiao F, Zhou L, Wu Z. The microbiota-gut-brain axis: a potential target in the small-molecule compounds and gene therapeutic strategies for Parkinson's disease. Neurol Sci 2025; 46:561-578. [PMID: 39546084 PMCID: PMC11772541 DOI: 10.1007/s10072-024-07878-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 11/05/2024] [Indexed: 11/17/2024]
Abstract
BACKGROUNDS Parkinson's disease (PD) is a common neurodegenerative disorder characterized by motor symptoms and non-motor symptoms. It has been found that intestinal issues usually precede motor symptoms. Microorganisms in the gastrointestinal tract can affect central nervous system through the microbiota-gut-brain axis. Accumulating evidence has shown that disturbances in the microbiota-gut-brain axis are linked with PD. Thus, this pathway appears to be a promising therapeutic target for treatment of PD. OBJECTIVES In this review, we mainly described gut dysbiosis in PD and their underlying mechanisms for mediating neuroinflammation and peripheral immune response in PD pathology and futher discussed the potential small-molecule compounds and genic therapeutic strategies targeting the microbiota-gut-brain axis and their applications in PD. CONCLUSIONS Studies have found that some small molecule compounds and alterations of inflammation-related genes can improve the motor and non-motor symptoms of PD by improving the microbiota-gut-brain axis, which may provide potentially beneficial drugs and molecular targets for the therapies of PD.
Collapse
Affiliation(s)
- Fengjuan Jiao
- School of Mental Health, Jining Medical University, No. 45, Jianshe South Road, Jining City, Shandong Province, 272067, P. R. China.
- Shandong Collaborative Innovation Center for Diagnosis, Treatment and Behavioral Interventions of Mental Disorders, Institute of Mental Health, Jining Medical University, Jining, Shandong, 272067, P. R. China.
| | - Lincong Zhou
- School of Clinical Medicine, Jining Medical University, Jining, Shandong, 272067, PR China
| | - Zaixin Wu
- School of Clinical Medicine, Jining Medical University, Jining, Shandong, 272067, PR China
| |
Collapse
|
32
|
Pacher-Deutsch C, Schweighofer N, Hanemaaijer M, Marut W, Žukauskaitė K, Horvath A, Stadlbauer V. The microplastic-crisis: Role of bacteria in fighting microplastic-effects in the digestive system. ENVIRONMENTAL POLLUTION (BARKING, ESSEX : 1987) 2025; 366:125437. [PMID: 39631654 DOI: 10.1016/j.envpol.2024.125437] [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: 09/17/2024] [Revised: 11/13/2024] [Accepted: 12/01/2024] [Indexed: 12/07/2024]
Abstract
Plastic particles smaller than 5 mm, referred to as Microplastics, pose health risks, like metabolic, immunological, neurological, reproductive, and carcinogenic effects, after being ingested. Smaller plastic particles are more likely to be absorbed by human cells, with nanoplastics showing higher potential for cellular damage, including DNA fragmentation and altered protein functions. Micro- and nanoplastics (MNPs) affect the gastrointestinal tract by altering the microbial composition, they could influence digestive enzymes, and possibly disrupt mucus layers. In the stomach, they potentially interfere with digestion and barrier functions, while in the intestines, they could increase permeability via inflammation and tissue disruption. MNPs can lead to microbial dysbiosis, leading to gastrointestinal symptoms. By activating inflammatory pathways, altering T cell functions and affecting dendritic cells and macrophages, immune system homeostasis could possibly be disrupted. Probiotics offer potential strategies to alleviate plastic effects, by either degrading plastic particles or directly countering health effects. We compared genetic sequences of probiotics to the genome of known plastic degraders and concluded that no probiotic bacteria could serve the role of plastic degradation. However, probiotics could directly mitigate MNP-health effects. They can restore microbial diversity, enhance the gut barrier, regulate bile acid metabolism, reduce inflammation, regulate insulin balance, and counteract metabolic disruptions. Antioxidative properties protect against lipid peroxidation and MNP-related reproductive system damage. Probiotics can also bind and degrade toxins, like heavy metals and bisphenol A. Additionally, bacteria could be used to aggregate MNPs and reduce their impact. Therefore, probiotics offer a variety of strategies to counter MNP-induced health effects.
Collapse
Affiliation(s)
- Christian Pacher-Deutsch
- Division of Gastroenterology and Hepatology, Medical University of Graz, Graz, Austria; Center for Biomarker Research in Medicine (CBmed), Graz, Austria.
| | | | | | | | - Kristina Žukauskaitė
- Division of Gastroenterology and Hepatology, Medical University of Graz, Graz, Austria; Center for Biomarker Research in Medicine (CBmed), Graz, Austria; Life Sciences Center, Vilnius University, Vilnius, Lithuania
| | - Angela Horvath
- Division of Gastroenterology and Hepatology, Medical University of Graz, Graz, Austria; Center for Biomarker Research in Medicine (CBmed), Graz, Austria
| | - Vanessa Stadlbauer
- Division of Gastroenterology and Hepatology, Medical University of Graz, Graz, Austria; Center for Biomarker Research in Medicine (CBmed), Graz, Austria; BioTechMed-Graz, Graz, Austria
| |
Collapse
|
33
|
D’Ambrosio R, Cavallo S, Brunetti R, Pellicanò R, Vaccaro E, Borriello G, Paradiso R, Serpe FP, Lambiase S, Bruzzese F, Palma G, Rea D, Barbieri A, D’Amore M, Dimatteo M, degli Uberti B, Paciello O, Baldi L. The Use of Antimicrobials in Animal Husbandry as a Potential Factor for the Increased Incidence of Colorectal Cancer: Food Safety and Kinetics in a Murine Model. Animals (Basel) 2025; 15:315. [PMID: 39943084 PMCID: PMC11815752 DOI: 10.3390/ani15030315] [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: 12/24/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 02/16/2025] Open
Abstract
The aim of this research was to investigate the effects of the prolonged use of the broad-spectrum antimicrobial widely used in animal husbandry. By means of a mouse model, a translational study was carried out on immunocompetent mice (with a complete immune system). This study highlighted the effect of antimicrobial residues taken in with food on the growth time of cancer and on alterations to the gut microbiota. This project considered the fight against antimicrobial resistance from a One Health perspectivethrough collaboration between human medicine and veterinary medicine. Regarding food safety, antimicrobial residues in products of animal origin are rarely detected; they therefore constitute a negligible factor in determining colorectal cancer.
Collapse
Affiliation(s)
- Rosa D’Ambrosio
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Stefania Cavallo
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Roberta Brunetti
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Roberta Pellicanò
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Emanuela Vaccaro
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Napoli, Italy; (E.V.); (O.P.)
| | - Giorgia Borriello
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Rubina Paradiso
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Francesco Paolo Serpe
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Sara Lambiase
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Francesca Bruzzese
- Experimental Animal Unit, Istituto Nazionale Tumori-IRCCS-Fondazione “G. Pascale”, 80131 Naples, Italy; (F.B.); (G.P.)
| | - Giuseppe Palma
- Experimental Animal Unit, Istituto Nazionale Tumori-IRCCS-Fondazione “G. Pascale”, 80131 Naples, Italy; (F.B.); (G.P.)
| | - Domenica Rea
- Laboratory Medicine Unit, Istituto Nazionale Tumori- IRCCS- Fondazione “G. Pascale”, 80131 Naples, Italy;
| | - Antonio Barbieri
- ASL Salerno UOC Laboratorio d’Analisi, Vallo della Lucania, 84078 Salerno, Italy
| | - Marianna D’Amore
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Maria Dimatteo
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Barbara degli Uberti
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| | - Orlando Paciello
- Department of Veterinary Medicine and Animal Production, University of Naples Federico II, 80137 Napoli, Italy; (E.V.); (O.P.)
| | - Loredana Baldi
- Istituto Zooprofilattico Sperimentale del Mezzogiorno (IZSM), 80055 Portici, Italy; (R.D.); (S.C.); (R.P.); (G.B.); (R.P.); (F.P.S.); (S.L.); (M.D.); (B.d.U.); (L.B.)
| |
Collapse
|
34
|
Ji L, Shangguan Y, Chen C, Wei C, Zhu J, Hong X, Liu X, Zhu X, Li W. Dietary Tannic Acid Promotes Growth Performance and Resistance Against Aeromonas hydrophila Infection by Improving the Antioxidative Capacity and Intestinal Health in the Chinese Soft-Shelled Turtle ( Pelodiscus sinensis). Antioxidants (Basel) 2025; 14:112. [PMID: 39857447 PMCID: PMC11759827 DOI: 10.3390/antiox14010112] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2024] [Revised: 01/15/2025] [Accepted: 01/16/2025] [Indexed: 01/27/2025] Open
Abstract
To investigate the effect of tannic acid (TA) on the growth, disease resistance, and intestinal health of Chinese soft-shelled turtles, individual turtles were fed with 0 g/kg (CG), 0.5 g/kg, 1 g/kg, 2 g/kg, and 4 g/kg TA diets for 98 days. Afterwards, the turtles' disease resistance was tested using Aeromonas hydrophila. The results showed that 0.5-4 g/kg of dietary TA increased the growth performance and feed utilization (p < 0.05), with 2.38 g/kg being the optimal level for the specific growth rate (SGR). The addition of 0.5-4 g/kg of TA in diets increased the mucosal fold height and submucosa thickness of the small intestine, which reached a maximum of 2 g/kg. The addition of 0.5-2 g/kg of TA effectively reduced the cumulative mortality that had been induced by A. hydrophila, with the 2 g/kg dosage leading to the lowest mortality. Additionally, 1-4 g/kg of TA improved the T-SOD, CAT, and GSH-Px activities during infection, while 2 g/kg of dietary TA enhanced the richness and diversity of the microbiota, for example, by increasing Actinobacteria but inhibiting Firmicutes. The transcriptome demonstrated that the predominant differentially expressed genes (DEGs) in TA2 were mainly enriched in the PPAR signaling pathway (Acsl5, Apoa2, Apoa5, Fabp1, Fabp2, and Fabp6); in glycine, serine, and threonine metabolism (Chdh, Gatm, and Shmt1); and in steroid biosynthesis (Cel, Hsd17b7, Soat2, and Sqle). The main differentially expressed metabolites (DEMs) that were discovered by means of metabolome analysis included cholylhistidine, calcipotriol, 13-O-tetradecanoylphorbol 12-acetate, and hexahomomethionine in CG vs. TA2. Integrative analyses of two omics revealed that 2 g/kg of TA mitigated inflammation by activating the PPAR signaling pathway and regulating the lipid metabolism via multiple pathways, such as steroid biosynthesis and α-linolenic acid metabolism. In general, the inclusion of 2 g/kg of TA in turtle diets can optimally promote growth and bacterial resistance by maintaining intestinal health and improving antioxidant capacity.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xinping Zhu
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510380, China; (L.J.)
| | - Wei Li
- Key Laboratory of Tropical and Subtropical Fishery Resources Application and Cultivation, Ministry of Agriculture and Rural Affairs, Pearl River Fisheries Research Institute, Chinese Academy of Fishery Sciences, Guangzhou 510380, China; (L.J.)
| |
Collapse
|
35
|
Koppula S, Wankhede N, Kyada A, Ballal S, Arya R, Singh AK, Gulati M, Sute A, Sarode S, Polshettiwar S, Marde V, Taksande B, Upaganlawar A, Fareed M, Umekar M, Kopalli SR, Kale M. The gut-brain axis: Unveiling the impact of xenobiotics on neurological health and disorders. Prog Neuropsychopharmacol Biol Psychiatry 2025; 136:111237. [PMID: 39732317 DOI: 10.1016/j.pnpbp.2024.111237] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Revised: 11/12/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
The Gut-Brain Axis (GBA) is a crucial link between the gut microbiota and the central nervous system. Xenobiotics, originating from diverse sources, play a significant role in shaping this interaction. This review examines how these compounds influence neurotransmitter dynamics within the GBA. Environmental pollutants can disrupt microbial populations, impacting neurotransmitter synthesis-especially serotonin, gamma-aminobutyric acid (GABA), and dopamine pathways. Such disruptions affect mood regulation, cognition, and overall neurological function. Xenobiotics also contribute to the pathophysiology of neurological disorders, with changes in serotonin levels linked to mood disorders and imbalances in GABA and dopamine associated with anxiety, stress, and reward pathway disorders. These alterations extend beyond the GBA, leading to complications in neurological health, including increased risk of neurodegenerative diseases due to neuroinflammation triggered by neurotransmitter imbalances. This review provides a comprehensive overview of how xenobiotics influence the GBA and their implications for neurological well-being.
Collapse
Affiliation(s)
- Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea
| | - Nitu Wankhede
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Ashishkumar Kyada
- Marwadi University Research Center, Department of Pharmaceutical Sciences, Faculty of Health Sciences, Marwadi University, Rajkot, -360003, Gujarat, India
| | - Suhas Ballal
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Renu Arya
- Department of Pharmacy, Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India
| | | | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 1444411, India; ARCCIM, Faculty of Health, University of Technology Sydney, Ultimo, NSW 20227, Australia
| | - Astha Sute
- National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Sanskruti Sarode
- National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Shruti Polshettiwar
- National Institute of Pharmaceutical Education and Research (NIPER), Hyderabad, Telangana, India
| | - Vaibhav Marde
- Indian Institute of Technology (IIT), Hyderabad, Telangana 502284, India
| | - Brijesh Taksande
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Aman Upaganlawar
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nashik, Maharashtra, India
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, Riyadh 11597, Saudi Arabia
| | - Milind Umekar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Mayur Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India.
| |
Collapse
|
36
|
Xu Y, Lv S, Li X, Zhai C, Shi Y, Li X, Feng Z, Luo G, Wang Y, Gao X. Photoaffinity probe-enabled discovery of sennoside A reductase in Bifidobacterium pseudocatenulatum. J Pharm Anal 2025; 15:101108. [PMID: 39902460 PMCID: PMC11788863 DOI: 10.1016/j.jpha.2024.101108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 08/29/2024] [Accepted: 09/18/2024] [Indexed: 02/05/2025] Open
Abstract
Sennoside A (SA), a typical prodrug, exerts its laxative effect only after its transformation into rheinanthrone catalyzed by gut microbial hydrolases and reductases. Hydrolases have been identified, but reductases remain unknown. By linking a photoreactive group to the SA scaffold, we synthesized a photoaffinity probe to covalently label SA reductases and identified SA reductases using activity-based protein profiling (ABPP). From lysates of an active strain, Bifidobacterium pseudocatenulatum (B. pseudocatenulatum), 397 proteins were enriched and subsequently identified using mass spectrometry (MS). Among these proteins, chromate reductase/nicotinamide adenine dinucleotide (NADH) phosphate (NADPH)-dependent flavin mononucleotide (FMN) reductase/oxygen-insensitive NADPH nitroreductase (nfrA) was identified as a potent SA reductase through further bioinformatic analysis and The Universal Protein Resource (UniProt) database screening. We also determined that recombinant nfrA could reduce SA. Our study contributes to further illuminating mechanisms of SA transformation to rheinanthrone and simultaneously offers an effective method to identify gut bacterial reductases.
Collapse
Affiliation(s)
| | | | | | - Chuanjia Zhai
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Yulian Shi
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Xuejiao Li
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Zhiyang Feng
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Gan Luo
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Ying Wang
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| | - Xiaoyan Gao
- School of Chinese Materia Medica, Beijing University of Chinese Medicine, Beijing, 102488, China
| |
Collapse
|
37
|
Li W, Gao W, Yan S, Yang L, Zhu Q, Chu H. Gut Microbiota as Emerging Players in the Development of Alcohol-Related Liver Disease. Biomedicines 2024; 13:74. [PMID: 39857657 PMCID: PMC11761646 DOI: 10.3390/biomedicines13010074] [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: 12/07/2024] [Revised: 12/23/2024] [Accepted: 12/29/2024] [Indexed: 01/27/2025] Open
Abstract
The global incidence and mortality rates of alcohol-related liver disease are on the rise, reflecting a growing health concern worldwide. Alcohol-related liver disease develops due to a complex interplay of multiple reasons, including oxidative stress generated during the metabolism of ethanol, immune response activated by immunogenic substances, and subsequent inflammatory processes. Recent research highlights the gut microbiota's significant role in the progression of alcohol-related liver disease. In patients with alcohol-related liver disease, the relative abundance of pathogenic bacteria, including Enterococcus faecalis, increases and is positively correlated with the level of severity exhibited by alcohol-related liver disease. Supplement probiotics like Lactobacillus, as well as Bifidobacterium, have been found to alleviate alcohol-related liver disease. The gut microbiota is speculated to trigger specific signaling pathways, influence metabolite profiles, and modulate immune responses in the gut and liver. This research aimed to investigate the role of gut microorganisms in the onset and advancement of alcohol-related liver disease, as well as to uncover the underlying mechanisms by which the gut microbiota may contribute to its development. This review outlines current treatments for reversing gut dysbiosis, including probiotics, fecal microbiota transplantation, and targeted phage therapy. Particularly, targeted therapy will be a vital aspect of future alcohol-related liver disease treatment. It is to be hoped that this article will prove beneficial for the treatment of alcohol-related liver disease.
Collapse
Affiliation(s)
- Wei Li
- Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Hubei Clinical Research Center for Infectious Diseases, Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Wuhan 430023, China;
| | - Wenkang Gao
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (W.G.); (S.Y.); (L.Y.)
| | - Shengqi Yan
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (W.G.); (S.Y.); (L.Y.)
| | - Ling Yang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (W.G.); (S.Y.); (L.Y.)
| | - Qingjing Zhu
- Wuhan Jinyintan Hospital, Tongji Medical College of Huazhong University of Science and Technology, Hubei Clinical Research Center for Infectious Diseases, Wuhan Research Center for Communicable Disease Diagnosis and Treatment, Chinese Academy of Medical Sciences, Joint Laboratory of Infectious Diseases and Health, Wuhan Institute of Virology and Wuhan Jinyintan Hospital, Chinese Academy of Sciences, Wuhan 430023, China;
| | - Huikuan Chu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022, China; (W.G.); (S.Y.); (L.Y.)
| |
Collapse
|
38
|
Qu J, Meng F, Wang Z, Xu W. Unlocking Cardioprotective Potential of Gut Microbiome: Exploring Therapeutic Strategies. J Microbiol Biotechnol 2024; 34:2413-2424. [PMID: 39467697 PMCID: PMC11729380 DOI: 10.4014/jmb.2405.05019] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2024] [Revised: 06/05/2024] [Accepted: 06/15/2024] [Indexed: 10/30/2024]
Abstract
The microbial community inhabiting the human gut resembles a bustling metropolis, wherein beneficial bacteria play pivotal roles in regulating our bodily functions. These microorganisms adeptly break down resilient dietary fibers to fuel our energy, synthesize essential vitamins crucial for our well-being, and maintain the delicate balance of our immune system. Recent research indicates a potential correlation between alterations in the composition and activities of these gut microbes and the development of coronary artery disease (CAD). Consequently, scientists are delving into the intriguing realm of manipulating these gut inhabitants to potentially mitigate disease risks. Various promising strategies have emerged in this endeavor. Studies have evidenced that probiotics can mitigate inflammation and enhance the endothelial health of our blood vessels. Notably, strains such as Lactobacilli and Bifidobacteria have garnered substantial attention in both laboratory settings and clinical trials. Conversely, prebiotics exhibit anti-inflammatory properties and hold potential in managing conditions like hypertension and hypercholesterolemia. Synbiotics, which synergistically combine probiotics and prebiotics, show promise in regulating glucose metabolism and abnormal lipid profiles. However, uncertainties persist regarding postbiotics, while antibiotics are deemed unsuitable due to their potential adverse effects. On the other hand, TMAO blockers, such as 3,3-dimethyl-1-butanol, demonstrate encouraging outcomes in laboratory experiments owing to their anti-inflammatory and tissue-protective properties. Moreover, fecal transplantation, despite yielding mixed results, warrants further exploration and refinement. In this comprehensive review, we delve into the intricate interplay between the gut microbiota and CAD, shedding light on the multifaceted approaches researchers are employing to leverage this understanding for therapeutic advancements.
Collapse
Affiliation(s)
- Jun Qu
- Department of Internal Medicine-Cardiovascular, YanTai YuHuangDing Hospital, Yantai, Shandong, P.R. China
| | - Fantao Meng
- Department of Internal Medicine-Cardiovascular, LinYi Central Hospital, LinYi, Shandong, P.R. China
| | - Zhen Wang
- Department of Internal Medicine-Cardiovascular, YanTai YuHuangDing Hospital, Yantai, Shandong, P.R. China
| | - Wenhao Xu
- Department of Internal Medicine-Cardiovascular, YanTai YuHuangDing Hospital, Yantai, Shandong, P.R. China
| |
Collapse
|
39
|
Bircan H, Demircan T, Yulek F, Yuksel N. PCR-based characterization of nasal and ocular surface flora and functional analysis of bacterial metabolic pathways in congenital nasolacrimal duct obstruction. Int Ophthalmol 2024; 45:17. [PMID: 39704869 DOI: 10.1007/s10792-024-03386-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Accepted: 12/10/2024] [Indexed: 12/21/2024]
Abstract
PURPOSE To evaluate the role of Streptococcus pneumoniae, Staphylococcus aureus, Haemophilus influenzae, Serratia marcescens, and Moraxella catarrhalis in the nasal and ocular surface flora, along with their metabolic activities in children with unilateral congenital nasolacrimal duct obstruction (CNLDO). METHODS Swabs were taken from the bilateral inferior meatus and ocular surface of 26 children with unilateral CNLDO before probing. Nasal and ocular surface swabs from non-operated eyes of children who underwent unilateral blepharoptosis or strabismus surgery formed the control group. The quantitative polymerase chain reaction (qPCR) method was used to analyze the samples. RESULTS The detection of Streptococcus pneumoniae in the nasal flora, Staphylococcus aureus in the ocular surface flora, and Haemophilus influenzae and Serratia marcescens in both the nasal and ocular surface flora was higher on the obstructed sides of children with CNLDO compared to the controls (p < 0.05). Obstructed sides in children with CNLDO showed a significant difference for Moraxella catarrhalis in the ocular surface and Streptococcus pneumoniae, Staphylococcus aureus, Serratia marcescens, and Moraxella catarrhalis in the nasal flora compared to the patent sides (p < 0.05). Metabolite analysis revealed that betaine, L-carnitine, L-leucine, and L-alanine were negatively regulated in the presence of these bacteria. CONCLUSIONS Significant changes in bacterial composition in the nasal and ocular surface flora may be a cause or a result of CNLDO. More comprehensive microbiological studies that evaluate the entire microbiome would be helpful in understanding CNLDO.
Collapse
Affiliation(s)
- Hazal Bircan
- Department of Ophthalmology, Sanliurfa Training and Research Hospital, Sanliurfa, Turkey
| | - Turan Demircan
- Department of Medical Biology, Medical Faculty, Mugla Sitki Kocman University, Mugla, Turkey
| | | | - Nilay Yuksel
- Department of Ophthalmology, University of Health Sciences, Ankara Bilkent City Hospital, Ankara, Turkey.
| |
Collapse
|
40
|
Tejedor Vaquero S, Neuman H, Comerma L, Marcos-Fa X, Corral-Vazquez C, Uzzan M, Pybus M, Segura-Garzón D, Guerra J, Perruzza L, Tachó-Piñot R, Sintes J, Rosenstein A, Grasset EK, Iglesias M, Gonzalez Farré M, Lop J, Patriaca-Amiano ME, Larrubia-Loring M, Santiago-Diaz P, Perera-Bel J, Berenguer-Molins P, Martinez Gallo M, Martin-Nalda A, Varela E, Garrido-Pontnou M, Grassi F, Guarner F, Mehandru S, Márquez-Mosquera L, Mehr R, Cerutti A, Magri G. Immunomolecular and reactivity landscapes of gut IgA subclasses in homeostasis and inflammatory bowel disease. J Exp Med 2024; 221:e20230079. [PMID: 39560666 PMCID: PMC11577441 DOI: 10.1084/jem.20230079] [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: 01/12/2023] [Revised: 06/13/2024] [Accepted: 09/24/2024] [Indexed: 11/20/2024] Open
Abstract
The human gut includes plasma cells (PCs) expressing immunoglobulin A1 (IgA1) or IgA2, two structurally distinct IgA subclasses with elusive regulation, function, and reactivity. We show here that intestinal IgA1+ and IgA2+ PCs co-emerged early in life, comparably accumulated somatic mutations, and were enriched within short-lived CD19+ and long-lived CD19- PC subsets, respectively. IgA2+ PCs were extensively clonally related to IgA1+ PCs and a subset of them presumably emerged from IgA1+ precursors. Of note, secretory IgA1 (SIgA1) and SIgA2 dually coated a large fraction of mucus-embedded bacteria, including Akkermansia muciniphila. Disruption of homeostasis by inflammatory bowel disease (IBD) was associated with an increase in actively proliferating IgA1+ plasmablasts, a depletion in long-lived IgA2+ PCs, and increased SIgA1+SIgA2+ gut microbiota. Such increase featured enhanced IgA1 reactivity to pathobionts, including Escherichia coli, combined with depletion of beneficial A. muciniphila. Thus, gut IgA1 and IgA2 emerge from clonally related PCs and show unique changes in both frequency and reactivity in IBD.
Collapse
Affiliation(s)
- Sonia Tejedor Vaquero
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Hadas Neuman
- Computational Immunology Laboratory, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Laura Comerma
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona, Spain
- Pathology Department, Hospital del Mar, Barcelona, Spain
| | - Xavi Marcos-Fa
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Celia Corral-Vazquez
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Mathieu Uzzan
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Immunology Institute, New York, NY, USA
| | - Marc Pybus
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Daniel Segura-Garzón
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Joana Guerra
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Lisa Perruzza
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Roser Tachó-Piñot
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Jordi Sintes
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Adam Rosenstein
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Immunology Institute, New York, NY, USA
| | - Emilie K. Grasset
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Immunology Institute, New York, NY, USA
| | - Mar Iglesias
- Pathology Department, Hospital del Mar, Barcelona, Spain
| | | | - Joan Lop
- Pathology Department, Hospital del Mar, Barcelona, Spain
| | | | | | | | - Júlia Perera-Bel
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Pau Berenguer-Molins
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| | - Monica Martinez Gallo
- Immunology Division, Vall d’Hebron University Hospital and Translational Immunology Research Group, Vall d’Hebron Research Institute (VHIR), Department of Cell Biology, Physiology and Immunology, Autonomous University of Barcelona (UAB), Barcelona, Spain
| | - Andrea Martin-Nalda
- Pediatric Infectious Diseases and Immunodeficiencies Unit, Vall d’Hebron University Hospital, Jeffrey Modell Diagnostic and Research Center for Primary Immunodeficiencies, Barcelona, Spain
| | - Encarna Varela
- Department of Gastroenterology, Vall d’Hebron Research Institute, Barcelona, Spain
- Biomedical Research Networking Center in Hepatic and Digestive Diseases, Instituto Carlos III, Madrid, Spain
| | | | - Fabio Grassi
- Institute for Research in Biomedicine, Faculty of Biomedical Sciences, Università della Svizzera Italiana, Bellinzona, Switzerland
| | - Francisco Guarner
- Department of Gastroenterology, Vall d’Hebron Research Institute, Barcelona, Spain
- Biomedical Research Networking Center in Hepatic and Digestive Diseases, Instituto Carlos III, Madrid, Spain
| | - Saurabh Mehandru
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Immunology Institute, New York, NY, USA
| | - Lucia Márquez-Mosquera
- Department of Gastroenterology, Hospital del Mar Medical Research Institute Barcelona, Barcelona, Spain
| | - Ramit Mehr
- Computational Immunology Laboratory, The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat-Gan, Israel
| | - Andrea Cerutti
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona, Spain
- Department of Medicine, Icahn School of Medicine at Mount Sinai, Immunology Institute, New York, NY, USA
- Catalan Institute for Research and Advanced Studies, Barcelona, Spain
| | - Giuliana Magri
- Translational Clinical Research Program, Hospital del Mar Research Institute, Barcelona, Spain
| |
Collapse
|
41
|
Dong J, Wang L, Bai Y, Huang X, Chen C, Liu Y. Study on the physicochemical properties and immune regulatory mechanism of polysaccharide fraction from Aronia Melanocarpa fruit. Int J Biol Macromol 2024; 283:137696. [PMID: 39557257 DOI: 10.1016/j.ijbiomac.2024.137696] [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: 09/23/2024] [Revised: 11/04/2024] [Accepted: 11/13/2024] [Indexed: 11/20/2024]
Abstract
Aronia Melanocarpa (Michx.) Elliott fruit has been extensively used in the food and medicinal fields. This study aimed to analyze the physicochemical properties of a polysaccharide fraction (AMP2) isolated from this fruit for the first time and investigated its immune regulatory mechanism. The physicochemical properties of AMP2 were determined using high-performance gel permeation chromatography, PMP derivatization-high performance liquid chromatography, Ultraviolet spectroscopy, and infrared spectroscopy. The metagenomic technology was applied to investigate the regulatory effects and mechanisms of AMP2 on the gut microbiota of immunosuppressed mice. The results showed that molecular weight of AMP2 was 83,444 Da, which was mainly composed of D-arabinose, D-xylose, D-mannose, D-rhamnose and D-glucose, and both β-type and α-type glycosidic bonds contained in its structure. AMP2 changed the composition of gut microbiota by increasing the number of beneficial and probiotic bacteria, thereby regulated the intestinal mucosal immune system of host. AMP2 improved intestinal immune system response and antimicrobial capacity through positive regulation of the NOD-like receptor signaling pathway and neutrophil extracellular trap formation. The results demonstrate the potential of AMP2 in immune regulation, providing a new perspective for its subsequent development and contributing to the development and application of related health foods.
Collapse
Affiliation(s)
- Jinxv Dong
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China
| | - Lei Wang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China; Jinuo Biological Engineering Co. Ltd, Changchun, Jilin 130600, China
| | - Yutao Bai
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China
| | - Xin Huang
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China.
| | - Changbao Chen
- Jilin Ginseng Academy, Changchun University of Chinese Medicine, Changchun, Jilin 130117, China
| | - Yan Liu
- Institute of Special Animal and Plant Sciences, Chinese Academy of Agricultural Sciences, Changchun, Jilin 130112, China.
| |
Collapse
|
42
|
Thapa R, Magar AT, Shrestha J, Panth N, Idrees S, Sadaf T, Bashyal S, Elwakil BH, Sugandhi VV, Rojekar S, Nikhate R, Gupta G, Singh SK, Dua K, Hansbro PM, Paudel KR. Influence of gut and lung dysbiosis on lung cancer progression and their modulation as promising therapeutic targets: a comprehensive review. MedComm (Beijing) 2024; 5:e70018. [PMID: 39584048 PMCID: PMC11586092 DOI: 10.1002/mco2.70018] [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: 07/06/2024] [Revised: 10/23/2024] [Accepted: 10/24/2024] [Indexed: 11/26/2024] Open
Abstract
Lung cancer (LC) continues to pose the highest mortality and exhibits a common prevalence among all types of cancer. The genetic interaction between human eukaryotes and microbial cells plays a vital role in orchestrating every physiological activity of the host. The dynamic crosstalk between gut and lung microbiomes and the gut-lung axis communication network has been widely accepted as promising factors influencing LC progression. The advent of the 16s rDNA sequencing technique has opened new horizons for elucidating the lung microbiome and its potential pathophysiological role in LC and other infectious lung diseases using a molecular approach. Numerous studies have reported the direct involvement of the host microbiome in lung tumorigenesis processes and their impact on current treatment strategies such as radiotherapy, chemotherapy, or immunotherapy. The genetic and metabolomic cross-interaction, microbiome-dependent host immune modulation, and the close association between microbiota composition and treatment outcomes strongly suggest that designing microbiome-based treatment strategies and investigating new molecules targeting the common holobiome could offer potential alternatives to develop effective therapeutic principles for LC treatment. This review aims to highlight the interaction between the host and microbiome in LC progression and the possibility of manipulating altered microbiome ecology as therapeutic targets.
Collapse
Affiliation(s)
- Rajan Thapa
- Department of Pharmacy, Universal college of medical sciencesTribhuvan UniversityBhairahawaRupendehiNepal
| | - Anjana Thapa Magar
- Department of MedicineKathmandu Medical College Teaching Hospital, SinamangalKathmanduNepal
| | - Jesus Shrestha
- School of Biomedical EngineeringUniversity of Technology SydneySydneyNew South WalesAustralia
| | - Nisha Panth
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Sobia Idrees
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Tayyaba Sadaf
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Saroj Bashyal
- Department of Pharmacy, Manmohan Memorial Institute of Health SciencesTribhuvan University, SoalteemodeKathmanduNepal
| | - Bassma H. Elwakil
- Department of Medical Laboratory Technology, Faculty of Applied Health Sciences TechnologyPharos University in AlexandriaAlexandriaEgypt
| | - Vrashabh V. Sugandhi
- Department of pharmaceutical sciences, College of Pharmacy & Health SciencesSt. John's UniversityQueensNew YorkUSA
| | - Satish Rojekar
- Department of Pharmacological SciencesIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Ram Nikhate
- Department of PharmaceuticsDattakala Shikshan Sanstha, Dattakala college of pharmacy (Affiliated to Savitribai Phule Pune universityPuneMaharashtraIndia
| | - Gaurav Gupta
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical SciencesSaveetha UniversityChennaiIndia
- Centre of Medical and Bio‐allied Health Sciences ResearchAjman UniversityAjmanUAE
| | - Sachin Kumar Singh
- School of Pharmaceutical SciencesLovely Professional UniversityPhagwaraIndia
- Faculty of Health, Australian Research Centre in Complementary and Integrative MedicineUniversity of Technology SydneyUltimoNew South WalesAustralia
| | - Kamal Dua
- Faculty of Health, Australian Research Centre in Complementary and Integrative MedicineUniversity of Technology SydneyUltimoNew South WalesAustralia
- Discipline of Pharmacy, Graduate School of HealthUniversity of Technology SydneyUltimoNew South WalesAustralia
| | - Philip M Hansbro
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| | - Keshav Raj Paudel
- Centre for Inflammation, Faculty of Science, School of Life SciencesCentenary Institute and University of Technology SydneySydneyNew South WalesAustralia
| |
Collapse
|
43
|
Jiang H, Ye Y, Wang M, Sun X, Sun T, Chen Y, Li P, Zhang M, Wang T. The progress on the relationship between gut microbiota and immune checkpoint blockade in tumors. Biotechnol Genet Eng Rev 2024; 40:4446-4465. [PMID: 37191003 DOI: 10.1080/02648725.2023.2212526] [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: 03/19/2023] [Accepted: 05/05/2023] [Indexed: 05/17/2023]
Abstract
Immune checkpoint blockade (ICB) has emerged as a promising immunotherapeutic approach for the treatment of various tumors. However, the efficacy of this therapy is limited in a subset of patients, and it is important to develop strategies to enhance immune responses. Studies have demonstrated a critical role of gut microbiota in regulating the therapeutic response to ICB. Gut microbiota composition, diversity, and function are mediated by metabolites, such as short-chain fatty acids and secondary bile acids, that interact with host immune cells through specific receptors. In addition, gut bacteria may translocate to the tumor site and stimulate antitumor immune responses. Therefore, maintaining a healthy gut microbiota composition, for instance through avoiding the use of antibiotics or probiotic interventions, can be an effective approach to optimize ICB therapy. This review summarizes the current understanding of the microbiota-immunity interactions in the context of ICB therapy, and discusses potential clinical implications of these findings.
Collapse
Affiliation(s)
- Haili Jiang
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yingquan Ye
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Mingqi Wang
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Xin Sun
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ting Sun
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Yang Chen
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ping Li
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Mei Zhang
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| | - Ting Wang
- Oncology Department of Integrated Traditional Chinese and Western Medicine, The First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
| |
Collapse
|
44
|
Lee JY, Kim Y, Kim J, Kim JK. Fecal Microbiota Transplantation: Indications, Methods, and Challenges. J Microbiol 2024; 62:1057-1074. [PMID: 39557804 DOI: 10.1007/s12275-024-00184-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Revised: 09/25/2024] [Accepted: 10/16/2024] [Indexed: 11/20/2024]
Abstract
Over the past two decades, as the importance of gut microbiota to human health has become widely known, attempts have been made to treat diseases by correcting dysbiosis of gut microbiota through fecal microbiota transplantation (FMT). Apart from current knowledge of gut microbiota, FMT to treat disease has a long history, from the treatment of food poisoning in the fourth century to the treatment of Clostridioides difficile infections in the twentieth century. In 2013, FMT was recognized as a standard treatment for recurrent C. difficile because it consistently showed high efficacy. Though recurrent C. difficile is the only disease internationally recognized for FMT efficacy, FMT has been tested for other diseases and shown some promising preliminary results. Different FMT methods have been developed using various formulations and administration routes. Despite advances in FMT, some issues remain to be resolved, such as donor screening, manufacturing protocols, and unknown components in the fecal microbiota. In this review, we discuss the mechanisms, clinical indications, methods, and challenges of current FMT. We also discuss the development of alternative therapies to overcome the challenges of FMT.
Collapse
Affiliation(s)
- Jee Young Lee
- Department of Microbiology, Kosin University College of Medicine, Busan, 49267, Republic of Korea
| | - Yehwon Kim
- Department of Medicine, Kosin University College of Medicine, Busan, 49267, Republic of Korea
| | - Jiyoun Kim
- Department of Medicine, Kosin University College of Medicine, Busan, 49267, Republic of Korea
| | - Jiyeun Kate Kim
- Department of Microbiology, Kosin University College of Medicine, Busan, 49267, Republic of Korea.
| |
Collapse
|
45
|
Lucaciu SR, Domokos B, Puiu R, Ruta V, Motoc SN, Rajnoveanu R, Todea D, Stoia AM, Man AM. Lung Microbiome in Lung Cancer: A Systematic Review. Microorganisms 2024; 12:2439. [PMID: 39770642 PMCID: PMC11676289 DOI: 10.3390/microorganisms12122439] [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: 09/12/2024] [Revised: 11/18/2024] [Accepted: 11/22/2024] [Indexed: 01/11/2025] Open
Abstract
To date, the percentage composition of the lung microbiome in bronchopulmonary cancer has not been summarized. Existing studies on identifying the lung microbiome in bronchopulmonary cancer through 16S rRNA sequencing have shown variable results regarding the abundance of bacterial taxa. OBJECTIVE To identify the predominant bacterial taxa at the phylum and genus levels in bronchopulmonary cancer using samples collected through bronchoalveolar lavage and to determine a potential proportional pattern that could contribute to the diagnosis of bronchopulmonary cancer. DATA SOURCES A systematic review of English articles using MEDLINE, Embase, and Web of Science. Search terms included lung microbiome, lung cancer, and bronchoalveolar lavage. STUDY SELECTION Studies that investigated the lung microbiome in bronchopulmonary cancer with samples collected via bronchoalveolar lavage. DATA EXTRACTION Independent extraction of articles using predefined data fields, including study quality indicators. DATA SYNTHESIS Nine studies met the inclusion criteria, focusing on those that utilized a percentage expression of the microbiome at the phylum or genus level. There was noted heterogeneity between studies, both in terms of phylum and genus, with a relatively constant percentage of the Firmicutes phylum and the genera Streptococcus and Veillonella being mentioned. Significant differences were also observed regarding the inclusion criteria for study participants, the method of sample collection, and data processing. CONCLUSIONS To date, there is no consistent percentage pattern at the phylum or genus level in bronchopulmonary cancer, with the predominance of a phylum or genus varying across different patient cohorts, resulting in non-overlapping outcomes.
Collapse
Affiliation(s)
| | - Bianca Domokos
- Department of Pneumology, Iuliu Hațieganu University of Medicine and Pharmacy, 400012 Cluj-Napoca, Romania; (S.-R.L.); (R.P.); (V.R.); (S.N.M.); (R.R.); (D.T.); (A.M.S.); (A.M.M.)
| | | | | | | | | | | | | | | |
Collapse
|
46
|
Yan J, Yu J, Bu C, Yang L, Chen J, Ding X, Yuan P. Antibiotic-Augmented Chemodynamic Therapy for Treatment of Helicobacter pylori Infection in the Dynamic Stomach Environment. NANO LETTERS 2024; 24:14983-14992. [PMID: 39541155 DOI: 10.1021/acs.nanolett.4c03692] [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: 11/16/2024]
Abstract
Helicobacter pylori (H. pylori) is one of the main causes of peptic ulcer disease and gastric cancer. The overuse of antibiotics leads to bacterial drug resistance and disruption to the gut microbiome. Herein, a nanoparticle (TA-FeHMSN@Amox) was developed, comprising amoxicillin (Amox)-loaded iron-engineered hollow mesoporous silica as the core and a metal-polyphenol shell formed by tannic acid (TA) and Fe3+. In acidic stomach conditions, TA-FeHMSN@Amox generates bactericidal ·OH through Fenton/Fenton-like reactions of the degraded product Fe2+ and hydrogen peroxide (H2O2) at the infection site, achieving chemodynamic therapy (CDT). Moreover, released amoxicillin enhances therapeutic efficacy by impeding the self-repair of the bacterial cell wall damaged by CDT, overcoming the limitations of ineffective CDT under conditions lacking sufficient acidity and H2O2. The acidity-responsive CDT combined with reduced antibiotic usage ensures superior in vivo therapeutic efficacy and biocompatibility with intestinal flora, providing a highly potent strategy for treating H. pylori infections in the dynamic stomach environment.
Collapse
Affiliation(s)
- Jiachang Yan
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Jiayin Yu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Changxin Bu
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Li Yang
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Jiaoyu Chen
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| | - Xin Ding
- School of Medicine, Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
- State Key Laboratory of Anti-Infective Drug Discovery and Development, School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, 510006, China
| | - Peiyan Yuan
- School of Pharmaceutical Sciences (Shenzhen), Shenzhen Campus of Sun Yat-sen University, Shenzhen, 518107, China
| |
Collapse
|
47
|
Yang Q, Wang L, Wang P, Yan Z, Chen Q, Zhang P, Li J, Jia R, Li Y, Yin X, Gun S. Effect of the diet level of whole-plant corn silage on the colonic microflora of Hezuo pigs. PeerJ 2024; 12:e18630. [PMID: 39611014 PMCID: PMC11604041 DOI: 10.7717/peerj.18630] [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: 06/11/2024] [Accepted: 11/12/2024] [Indexed: 11/30/2024] Open
Abstract
Background Whole-plant corn silage (WPCS) is an important roughage source for livestock, and have critical influences on rumen or intestinal microbiota, thus affecting the growth performance and feed efficiency. Our previous studies showed that adding WPCS to the diet of Hezuo pigs could promote the growth and fiber digestibility. While the aim of this study is to understand the effect of dietary WPCS on the colonic microflora in Hezuo pigs, which is essential for improving the roughage exploitation of pigs. Methods Thirty-two Hezuo pigs with similar body weight (7.88 ± 0.81) kg were selected and randomly divided into four groups with eight pigs in each group. Pigs in the control group were fed a basal diet, pigs in the experimental groups (Groups I, II, and III) were fed basal diet supplemented with 5%, 10%, and 15% WPCS, respectively, under 120 d experimental period. Six pigs from each group were picked for collecting colonic contents samples. 16S rRNA sequencing was performed to analyze the colonic microbiota of experimental pigs. Results The results showed that community richness indexes Chao1 and Observed_species in group III of Hezuo pig were significantly lower than that of the other three groups, community diversity indexes Shannon and Simpson were significantly higher in group I and II in comparison to the control group, and significantly lower in group III in comparison to the control group, group I and II. Adding WPCS to the diet of Hezuo pigs has no influence on the colonic dominant phylum, Clostridium sensu stricto 1 and Rikenellaceae RC9 gut group were most prevalent in the colon of Hezuo pig. When compared with the control group, the relative abundance of Streptococcus was significantly decreased in three experimental groups, while p-251-o5, Parabacteroides, Prevotellaceae UCG-003, Prevotellaceae UCG-001, and F082 exhibited significantly higher relative abundances in at least two experimental groups. Fibrobacter, Rikenellaceae RC9 gut group in group I, UCG-010 in group II, Bacteroides in group III exhibited increased relative abundance as compared with the control group. PICRUSt functional annotation indicated that the functions of cellular process and signaling were significantly increased in all WPCS-rationed groups, cancers, nervous system, immune system and environmental adaptation were all differed from groups I and II; three predominant pathways of translation, nucleotide metabolism and signal were only differed from the group II. Conclusions Feeding with 5% and 10% WPCS for Hezuo pigs could improve their colonic microflora diversity, and increase the relative abundance of fiber-digesting bacteria, which may potentially help to improve the fibre digestibility of Hezuo pigs by regulating the microbial function of cellular process and signaling, nucleotide metabolism, translation.
Collapse
Affiliation(s)
- Qiaoli Yang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Longlong Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Pengfei Wang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Zunqiang Yan
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Qiong Chen
- Gansu Bailu Grass Industry Development Co., Ltd., Lanzhou, Gansu, China
| | - Pengxia Zhang
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Jie Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Rui Jia
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Yao Li
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Xitong Yin
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
| | - Shuangbao Gun
- College of Animal Science and Technology, Gansu Agricultural University, Lanzhou, Gansu, China
- Gansu Diebu Juema Pig Science and Technology Backyard, Diebu, China
- Gansu Research Center for Swine Production Engineering and Technology, Lanzhou, China
| |
Collapse
|
48
|
Lapenna L, Di Cola S, Merli M. The crucial role of risk factors when dealing with hepatic Encephalopathy. Metab Brain Dis 2024; 40:29. [PMID: 39570425 DOI: 10.1007/s11011-024-01446-0] [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: 06/24/2024] [Accepted: 10/11/2024] [Indexed: 11/22/2024]
Abstract
Hepatic encephalopathy (HE) is a common condition in patients with cirrhosis, representing the second most frequent cause of decompensation. Approximately 30-40% of patients with cirrhosis will experience overt HE during the clinical course of their illness. In most cases, it is possible to identify a precipitating or risk factor for HE. These are distinct concepts that play different roles in the development of this condition. While precipitating factors act acutely, risk factors are generally present over an extended period and contribute to the overall likelihood of developing HE. The two types of factors require different approaches, with risk factors being more susceptible to prevention. The aim of this review is to describe the most important risk factors (such as severity of liver disease, previous episode of HE, minimal/covert HE, spontaneous and iatrogenic shunt, malnutrition, chronic therapies, metabolic diseases) for the development of HE and how to prevent it.
Collapse
Affiliation(s)
- Lucia Lapenna
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Simone Di Cola
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy
| | - Manuela Merli
- Department of Translational and Precision Medicine, Sapienza University of Rome, Rome, Italy.
| |
Collapse
|
49
|
Senaprom S, Namjud N, Ondee T, Bumrungpert A, Pongpirul K. Sugar Composition of Thai Desserts and Their Impact on the Gut Microbiome in Healthy Volunteers: A Randomized Controlled Trial. Nutrients 2024; 16:3933. [PMID: 39599719 PMCID: PMC11597037 DOI: 10.3390/nu16223933] [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: 10/02/2024] [Revised: 11/03/2024] [Accepted: 11/14/2024] [Indexed: 11/29/2024] Open
Abstract
BACKGROUND The relationship between consuming Thai desserts-predominantly composed of carbohydrates-and gut microbiome profiles remains unclear. This study aimed to evaluate the effects of consuming various Thai desserts with different GI values on the gut microbiomes of healthy volunteers. METHODS This open-label, parallel randomized clinical trial involved 30 healthy individuals aged 18 to 45 years. Participants were randomly assigned to one of three groups: Phetchaburi's Custard Cake (192 g, low-GI group, n = 10), Saraburi's Curry Puff (98 g, medium-GI group, n = 10), and Lampang's Crispy Rice Cracker (68 g, high-GI group, n = 10), each consumed alongside their standard breakfast. Fecal samples were collected at baseline and 24 h post-intervention for metagenomic analysis of gut microbiome profiles using 16S rRNA gene sequencing. RESULTS After 24 h, distinct trends in the relative abundance of various gut microbiota were observed among the dessert groups. In the high-GI dessert group, the abundance of Collinsella and Bifidobacterium decreased compared to the low- and medium-GI groups, while Roseburia and Ruminococcus showed slight increases. Correlation analysis revealed a significant negative relationship between sugar intake and Lactobacillus abundance in the medium- and high-GI groups, but not in the low-GI group. Additionally, a moderately negative association was observed between Akkermansia abundance and sugar intake in the high-GI group. These bacteria are implicated in energy metabolism and insulin regulation. LEfSe analysis identified Porphyromonadaceae and Porphyromonas as core microbiota in the low-GI group, whereas Klebsiella was enriched in the high-GI group, with no predominant bacteria identified in the medium-GI group. CONCLUSIONS The findings suggest that Thai desserts with varying GI levels can influence specific gut bacteria, though these effects may be temporary.
Collapse
Affiliation(s)
- Sayamon Senaprom
- Department of Preventive and Social Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (S.S.); (N.N.); (T.O.)
| | - Nuttaphat Namjud
- Department of Preventive and Social Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (S.S.); (N.N.); (T.O.)
| | - Thunnicha Ondee
- Department of Preventive and Social Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (S.S.); (N.N.); (T.O.)
- Center of Excellence in Preventive and Integrative Medicine (CE-PIM), Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
| | - Akkarach Bumrungpert
- College of Integrative Medicine, Dhurakij Pundit University, Bangkok 10210, Thailand;
| | - Krit Pongpirul
- Department of Preventive and Social Medicine, Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand; (S.S.); (N.N.); (T.O.)
- Center of Excellence in Preventive and Integrative Medicine (CE-PIM), Faculty of Medicine, Chulalongkorn University, Bangkok 10330, Thailand
- Bumrungrad International Hospital, Bangkok 10110, Thailand
- Department of International Health, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, USA
- Department of Infection Biology & Microbiomes, Faculty of Health and Life Sciences, University of Liverpool, Liverpool L69 3GB, UK
| |
Collapse
|
50
|
Khan IM, Nassar N, Chang H, Khan S, Cheng M, Wang Z, Xiang X. The microbiota: a key regulator of health, productivity, and reproductive success in mammals. Front Microbiol 2024; 15:1480811. [PMID: 39633815 PMCID: PMC11616035 DOI: 10.3389/fmicb.2024.1480811] [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: 09/10/2024] [Accepted: 10/15/2024] [Indexed: 12/07/2024] Open
Abstract
The microbiota, intensely intertwined with mammalian physiology, significantly impacts health, productivity, and reproductive functions. The normal microbiota interacts with the host through the following key mechanisms: acting as a protective barrier against pathogens, maintain mucosal barrier integrity, assisting in nutrient metabolism, and modulating of the immune response. Therefore, supporting growth and development of host, and providing protection against pathogens and toxic substances. The microbiota significantly influences brain development and behavior, as demonstrated by comprehensive findings from controlled laboratory experiments and human clinical studies. The prospects suggested that gut microbiome influence neurodevelopmental processes, modulate stress responses, and affect cognitive function through the gut-brain axis. Microbiota in the gastrointestinal tract of farm animals break down and ferment the ingested feed into nutrients, utilize to produce meat and milk. Among the beneficial by-products of gut microbiota, short-chain fatty acids (SCFAs) are particularly noteworthy for their substantial role in disease prevention and the promotion of various productive aspects in mammals. The microbiota plays a pivotal role in the reproductive hormonal systems of mammals, boosting reproductive performance in both sexes and fostering the maternal-infant connection, thereby becoming a crucial factor in sustaining mammalian existence. The microbiota is a critical factor influencing reproductive success and production traits in mammals. A well-balanced microbiome improves nutrient absorption and metabolic efficiency, leading to better growth rates, increased milk production, and enhanced overall health. Additionally, it regulates key reproductive hormones like estrogen and progesterone, which are essential for successful conception and pregnancy. Understanding the role of gut microbiota offers valuable insights for optimizing breeding and improving production outcomes, contributing to advancements in agriculture and veterinary medicine. This study emphasizes the critical ecological roles of mammalian microbiota, highlighting their essential contributions to health, productivity, and reproductive success. By integrating human and veterinary perspectives, it demonstrates how microbial communities enhance immune function, metabolic processes, and hormonal regulation across species, offering insights that benefit both clinical and agricultural advancements.
Collapse
Affiliation(s)
| | - Nourhan Nassar
- College of Life Science, Anhui Agricultural University, Hefei, China
- Department of Clinical Pathology, Faculty of Veterinary Medicine, Benha University, Moshtohor, Egypt
| | - Hua Chang
- College of Veterinary Medicine, Yunnan Agricultural University, Kunming, China
| | - Samiullah Khan
- The Scientific Observing and Experimental Station of Crop Pest in Guiyang, Ministry of Agriculture, Institute of Entomology, Guizhou University, Guiyang, China
| | - Maoji Cheng
- Fisugarpeptide Biology Engineering Co. Ltd., Lu’an, China
| | - Zaigui Wang
- College of Life Science, Anhui Agricultural University, Hefei, China
| | - Xun Xiang
- College of Animal Science and Technology, Yunnan Agricultural University, Kunming, China
| |
Collapse
|