1
|
Huang W, Zhu W, Lin Y, Chan FKL, Xu Z, Ng SC. Roseburia hominis improves host metabolism in diet-induced obesity. Gut Microbes 2025; 17:2467193. [PMID: 39976263 PMCID: PMC11845086 DOI: 10.1080/19490976.2025.2467193] [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: 07/24/2024] [Revised: 02/05/2025] [Accepted: 02/10/2025] [Indexed: 02/21/2025] Open
Abstract
Next-generation live biotherapeutics are promising to aid the treatment of obesity and metabolic diseases. Here, we reported a novel anti-obesity probiotic candidate, Roseburia hominis, that was depleted in stool samples of obese subjects compared with lean controls, and its abundance was negatively correlated with body mass index and serum triglycerides. Supplementation of R. hominis prevented body weight gain and disorders of glucose and lipid metabolism, prevented fatty liver, inhibited white adipose tissue expansion and brown adipose tissue whitening in mice fed with high-fat diet, and boosted the abundance of lean-related species. The effects of R. hominis could be partially attributed to the production of nicotinamide riboside and upregulation of the Sirtuin1/mTOR signaling pathway. These results indicated that R. hominis is a promising candidate for the development of next-generation live biotherapeutics for the prevention of obesity and metabolic diseases.
Collapse
Affiliation(s)
- Wenli Huang
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Wenyi Zhu
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Yu Lin
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Francis K. L. Chan
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhilu Xu
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
| | - Siew C. Ng
- Microbiota I-Center (MagIC), Hong Kong, China
- Department of Medicine and Therapeutics, The Chinese University of Hong Kong, Hong Kong, China
- Institute of Digestive Disease, State Key Laboratory of Digestive Diseases, Li Ka Shing Institute of Health Science, The Chinese University of Hong Kong, Hong Kong, China
- Center for Gut Microbiota Research, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| |
Collapse
|
2
|
Aranda-Carrillo SG, Del Carmen Ramos-Sustaita L, Cárdenas-Castro AP, Gutiérrez-Sarmiento W, Sánchez-Burgos JA, Ruíz-Valdiviezo VM, Sáyago-Ayerdi SG. Microbiota modulation and microbial metabolites produced during the in vitro colonic fermentation of Psidium guajava species. Food Res Int 2025; 208:116228. [PMID: 40263797 DOI: 10.1016/j.foodres.2025.116228] [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: 11/24/2024] [Revised: 02/27/2025] [Accepted: 03/11/2025] [Indexed: 04/24/2025]
Abstract
The interaction between gut microbiota and its metabolites is a growing area of research. Therefore, this study analyzed the bioactive compound profile of the indigestible fraction (IF) from Psidium species and evaluated its effects on microbiota composition during in vitro colonic fermentation. Hydroxycinnamic acids, hydroxybenzoic acids, and ellagitannins were the predominant phenolic compounds, with P. friedrichsthalianum ('Cas') exhibiting the highest concentrations. During in vitro colonic fermentation, a reduction in bacterial genera such as Enterobacteriaceae and Klebsiella was observed, while Faecalibacterium, Oscillibacter, Dialister, and Ruminococcaceae positively correlated with phenolic microbial metabolites. These findings suggest that the IF of Psidium species modulates gut microbiota composition and potentially contributes to the production of beneficial metabolites during human colonic fermentation, reinforcing the role of whole fruit consumption as a comprehensive matrix of nutrients and bioactive compounds beneficial to gut health.
Collapse
Affiliation(s)
- Suecia Grissol Aranda-Carrillo
- Tecnológico Nacional de México/ Instituto Tecnológico de Tepic, Av. Tecnológico No 2595, Col. Lagos del Country, CP 63175 Tepic, Nayarit, Mexico
| | - Lourdes Del Carmen Ramos-Sustaita
- Tecnológico Nacional de México/Instituto Tecnológico de Tuxtla-Gutiérrez, Departamento de Ingeniería Química y Bioquímica, Laboratory of Molecular Biology, Carretera Panamericana km 1080, CP 29050 Tuxtla Gutiérrez, Chiapas, Mexico
| | - Alicia Paulina Cárdenas-Castro
- Tecnológico Nacional de México/ Instituto Tecnológico de Tepic, Av. Tecnológico No 2595, Col. Lagos del Country, CP 63175 Tepic, Nayarit, Mexico
| | - Wilbert Gutiérrez-Sarmiento
- Chiapas Medicinal Plant Research Center, Pharmacobiology Experimental Laboratory, Autonomus University of Chiapas, Tuxtla Gutierrez, Chiapas, Mexico
| | - Jorge Alberto Sánchez-Burgos
- Tecnológico Nacional de México/ Instituto Tecnológico de Tepic, Av. Tecnológico No 2595, Col. Lagos del Country, CP 63175 Tepic, Nayarit, Mexico
| | - Víctor Manuel Ruíz-Valdiviezo
- Tecnológico Nacional de México/Instituto Tecnológico de Tuxtla-Gutiérrez, Departamento de Ingeniería Química y Bioquímica, Laboratory of Molecular Biology, Carretera Panamericana km 1080, CP 29050 Tuxtla Gutiérrez, Chiapas, Mexico
| | - Sonia Guadalupe Sáyago-Ayerdi
- Tecnológico Nacional de México/ Instituto Tecnológico de Tepic, Av. Tecnológico No 2595, Col. Lagos del Country, CP 63175 Tepic, Nayarit, Mexico.
| |
Collapse
|
3
|
Mishra S, Jain S, Agadzi B, Yadav H. A Cascade of Microbiota-Leaky Gut-Inflammation- Is it a Key Player in Metabolic Disorders? Curr Obes Rep 2025; 14:32. [PMID: 40208464 DOI: 10.1007/s13679-025-00624-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/21/2025] [Indexed: 04/11/2025]
Abstract
PURPOSE OF REVIEW This review addresses critical gaps in knowledge and provides a literature overview of the molecular pathways connecting gut microbiota dysbiosis to increased intestinal permeability (commonly referred to as "leaky gut") and its contribution to metabolic disorders. Restoring a healthy gut microbiota holds significant potential for enhancing intestinal barrier function and metabolic health. These interventions offer promising therapeutic avenues for addressing leaky gut and its associated pathologies in metabolic syndrome. RECENT FINDINGS In metabolic disorders such as obesity and type 2 diabetes (T2D), beneficial microbes such as those producing short-chain fatty acids (SCFAs) and other key metabolites like taurine, spermidine, glutamine, and indole derivatives are reduced. Concurrently, microbes that degrade toxic metabolites such as ethanolamine also decline, while proinflammatory, lipopolysaccharide (LPS)-enriched microbes increase. These microbial shifts place a higher burden on intestinal epithelial cells, which are in closest proximity to the gut lumen, inducing detrimental changes that compromise the structural and functional integrity of the intestinal barrier. Such changes include exacerbation of tight junction protein (TJP)s dysfunction, particularly through mechanisms such as destabilization of zona occludens (Zo)-1 mRNA or post-translational modifications. Emerging therapeutic strategies including ketogenic and Mediterranean diets, as well as probiotics, prebiotics, synbiotics, and postbiotics have demonstrated efficacy in restoring beneficial microbial populations, enhancing TJP expression and function, supporting gut barrier integrity, reducing leaky gut and inflammation, and ultimately improving metabolic disorders. This review summarizes the mechanisms by which gut microbiota contribute to the development of leaky gut and inflammation associated with metabolic syndrome. It also explores strategies for restoring gut microbiota balance and functionality by promoting beneficial microbes, increasing the production of beneficial metabolites, clearing toxic metabolites, and reducing the proportion of proinflammatory microbes. These approaches can alleviate the burden on intestinal epithelial cells, reduce leaky gut and inflammation, and improve metabolic health.
Collapse
Affiliation(s)
- Sidharth Mishra
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Center for Excellence of Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Byrd Alzheimer's Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Shalini Jain
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Center for Excellence of Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Byrd Alzheimer's Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Bryan Agadzi
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Hariom Yadav
- USF Center for Microbiome Research, Microbiomes Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
- Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
- Center for Excellence of Aging and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
- Byrd Alzheimer's Institute, University of South Florida Morsani College of Medicine, Tampa, FL, USA.
- Director of USF Center for Microbiome Research, Microbiomes Institute, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, 12901 Bruce B Downs Blvd, MDC78, Tampa, FL, 33612, USA.
| |
Collapse
|
4
|
Zhang G, Lu Y, Wang Z, Ma R, Jin H, Zhang J, Liu F, Ding Y. Causal relationship between gut microbiota and ageing: A multi-omics Mendelian randomization study. Arch Gerontol Geriatr 2025; 131:105765. [PMID: 39988416 DOI: 10.1016/j.archger.2025.105765] [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: 11/08/2024] [Revised: 01/12/2025] [Accepted: 01/18/2025] [Indexed: 02/25/2025]
Abstract
INTRODUCTION Observational studies and clinical trials have suggested a connection between gut microbiota and aging. However, the causal relationship between them remains undetermined. OBJECTIVES This study aimed to use bidirectional two-sample Mendelian randomization (TSMR) analysis to explore the causal relationship between gut microbiota and aging. METHODS Summary statistics from genome-wide association studies (GWAS) on gut microbiota and seven aging-related phenotypes were employed for TSMR analysis. Reverse Mendelian randomization (MR) analysis was performed to assess the potential for reverse causality. Additionally, the relationship between Akkermansia muciniphila and inflammation-related proteins and metabolites was further investigated. The effects of Akkermansia muciniphila on aging were also examined in Caenorhabditis elegans by measuring both lifespan and healthspan. RESULTS MR analysis of 207 microbial taxa and seven aging phenotypes revealed 44 causal relationships between the gut microbiota and aging. Akkermansia muciniphila was found to be causally linked to several aging-related traits, including mvAge, appendicular lean mass, and grip strength (P < 0.05). Reverse MR analysis identified 23 causal relationships, but no bidirectional causality was observed. Moreover, Akkermansia muciniphila is causally related to ST1A1, taurine bile acid, and mannose (P < 0.05). In Caenorhabditis elegans, treatment with Akkermansia muciniphila significantly extended lifespan (P < 0.05) and improved mobility in aging nematodes. CONCLUSION TSMR analysis uncovers multiple potential causal links between gut microbiota and aging, particularly Akkermansia muciniphila. Experimental results support its role in alleviating aging. This study provides a strong foundation for future research on gut microbiota's role in aging.
Collapse
Affiliation(s)
- Guolin Zhang
- Department of Cardiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yuqing Lu
- Department of Ophthalmology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Zhen Wang
- Department of General Surgery, The First Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Ruicong Ma
- Department of Cardiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Hongjin Jin
- Department of Cardiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Jingsi Zhang
- Department of Cardiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Fengyi Liu
- Department of Cardiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China
| | - Yanchun Ding
- Department of Cardiology, The Second Hospital of Dalian Medical University, Dalian, Liaoning, China.
| |
Collapse
|
5
|
Caesar R. The impact of novel probiotics isolated from the human gut on the gut microbiota and health. Diabetes Obes Metab 2025; 27 Suppl 1:3-14. [PMID: 39726216 PMCID: PMC11894790 DOI: 10.1111/dom.16129] [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: 10/25/2024] [Revised: 12/02/2024] [Accepted: 12/02/2024] [Indexed: 12/28/2024]
Abstract
The gut microbiota plays a pivotal role in influencing the metabolism and immune responses of the body. A balanced microbial composition promotes metabolic health through various mechanisms, including the production of beneficial metabolites, which help regulate inflammation and support immune functions. In contrast, imbalance in the gut microbiota, known as dysbiosis, can disrupt metabolic processes and increase the risk of developing diseases, such as obesity, type 2 diabetes, and inflammatory disorders. The composition of the gut microbiota is dynamic and can be influenced by environmental factors such as diet, medication, and the consumption of live bacteria. Since the early 1900s, bacteria isolated from food and have been used as probiotics. However, the human gut also offers an enormous reservoir of bacterial strains, and recent advances in microbiota research have led to the discovery of strains with probiotic potentials. These strains, derived from a broad spectrum of microbial taxa, differ in their ecological properties and how they interact with their hosts. For most probiotics bacterial structural components and metabolites, such as short-chain fatty acids, contribute to the maintenance of metabolic and immunological homeostasis by regulating inflammation and reinforcing gut barrier integrity. Metabolites produced by probiotic strains can also be used for bacterial cross-feeding to promote a balanced microbiota. Despite the challenges related to safety, stability, and strain-specific properties, several newly identified strains offer great potential for personalized probiotic interventions, allowing for targeted health strategies.
Collapse
Affiliation(s)
- Robert Caesar
- The Wallenberg Laboratory, Department of Molecular and Clinical MedicineUniversity of GothenburgGothenburgSweden
| |
Collapse
|
6
|
Day AW, Perez-Lozada J, DiLeo A, Blandino K, Maguire J, Kumamoto CA. Candida albicans Colonization Modulates Murine Ethanol Consumption and Behavioral Responses Through Elevation of Serum Prostaglandin E 2 and Impact on the Striatal Dopamine System. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.25.640044. [PMID: 40060518 PMCID: PMC11888247 DOI: 10.1101/2025.02.25.640044] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
Candida albicans is a commensal yeast that is a common component of the gastrointestinal (GI) microbiome of humans. C. albicans has been shown to bloom in the GI tract of individuals with alcohol use disorder (AUD) and can promote and increase the severity of alcoholic liver disease (ALD). However, the effects of C. albicans blooms on the host in the context of AUD or AUD-related phenotypes, such as ethanol preference, have been unstudied. In this work, we report a reduction in ethanol consumption and preference in mice colonized with C. albicans. C. albicans-colonized mice exhibited elevated levels of serum PGE2 and reduced ethanol preference was reversed by injection with antagonists of PGE2 receptors. Further, injection of mice with a PGE2 derivative decreased their ethanol preference. These results show that PGE2 acting on its receptors EP1 and EP2 drives reduced ethanol preference in C. albicans-colonized mice. We also showed altered transcription of dopamine receptors in the dorsal striatum of C. albicans-colonized mice and more rapid acquisition of ethanol conditioned taste aversion, suggesting alterations to reinforcement or aversion learning. Finally, C. albicans-colonized mice were more susceptible to ethanol-induced motor coordination impairment showing significant alterations to the behavioral effects of ethanol. This study identifies a member of the fungal microbiome that alters ethanol preference and demonstrates a role for PGE2 signaling in these phenotypes.
Collapse
Affiliation(s)
- Andrew W. Day
- Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, 02111, USA
- Department of Molecular Biology and Microbiology, Tufts University, Boston, Massachusetts, 02111, USA
| | - Jeyra Perez-Lozada
- Department of Molecular Biology and Microbiology, Tufts University, Boston, Massachusetts, 02111, USA
- current address: San Juan Bautista School of Medicine, Caguas, Puerto Rico, 00727, USA
| | - Alyssa DiLeo
- Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, 02111, USA
- Department of Neuroscience, Tufts University, Boston, Massachusetts, 02111, USA
| | - Katrina Blandino
- Graduate School of Biomedical Sciences, Tufts University, Boston, Massachusetts, 02111, USA
- Department of Neuroscience, Tufts University, Boston, Massachusetts, 02111, USA
| | - Jamie Maguire
- Department of Neuroscience, Tufts University, Boston, Massachusetts, 02111, USA
| | - Carol A. Kumamoto
- Department of Molecular Biology and Microbiology, Tufts University, Boston, Massachusetts, 02111, USA
| |
Collapse
|
7
|
Zhang L, Su H, Wang S, Fu Y, Wang M. Gut Microbiota and Neurotransmitter Regulation: Functional Effects of Four Traditional Chinese Fermented Soybean (Sojae Semen Praeparatum). Foods 2025; 14:671. [PMID: 40002115 PMCID: PMC11854601 DOI: 10.3390/foods14040671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2025] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 02/27/2025] Open
Abstract
This study aims to evaluate the potential disease prevention and treatment functions of four types of traditional Chinese fermented Sojae Semen Praeparatum (SSP) by analyzing their nutritional active components and their effects on the gut microbiota. Raw soybeans and the four SSPs were administered as dietary supplements to normal SD rats for 6 weeks. Fecal samples were collected at weeks 0, 2, and 6 to assess changes in the gut microbiota. Our results revealed that different fermentation methods resulted in variations in soybean isoflavone content. Fermented soybeans promoted the growth of beneficial microorganisms associated with short-chain fatty acid production in the gut microbiota, such as Christensenellaceae_R_7_group, compared to unfermented soybeans. Supplementation with SSPs fermented with different processes increased the diversity of the rat gut microbiota, except for the fermented group of qingwenjiedu decoction (QW). The dominant gut microbiota in the fermented group of Artemisia Annuae Herba and Mori Folium (QS) exhibited anti-inflammatory effects, while the dominant gut microbiota in the fermented group of Ephedrae Herba and Perillae Folium (MZ) showed antidepressant effects. In the neurotransmitter analysis, MZ reduced gamma-aminobutyric acid (GABA) levels, the fermented group without Chinese medicine (DD) decreased dopamine levels, and both QS and QW increased norepinephrine levels. Correlation analysis highlighted connections between gut microbiota, neurotransmitters, and chemical levels. The results indicate that SSPs may contribute uniquely to health by maintaining intestinal balance and improving neurological disorders while predicting a potential association between neurotransmitters and gut microbiota by correlation analysis.
Collapse
Affiliation(s)
| | | | | | | | - Manyuan Wang
- Beijing Key Laboratory of TCM Collateral Disease Theory Research, School of Traditional Chinese Medicine, Capital Medical University, Beijing 100069, China
| |
Collapse
|
8
|
Zhang J, Li L, Zhang M, Fang J, Xu Z, Zheng Y, Lin Z, Pan M. Distinct vaginal microbiome and metabolome profiles in women with preterm delivery following cervical cerclage. Front Cell Infect Microbiol 2025; 15:1444028. [PMID: 40007613 PMCID: PMC11850995 DOI: 10.3389/fcimb.2025.1444028] [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: 06/05/2024] [Accepted: 01/07/2025] [Indexed: 02/27/2025] Open
Abstract
Preterm birth (PTB) is a major cause of infant morbidity and mortality. The aim of this study was to investigate the effect of vaginal microbiota and metabolites on the outcome of pregnant women. In this study, a total of 127 pregnant women provided written informed consent prior to enrollment in accordance with the approved institutional guidelines, but only 45 pregnancies met the experimental requirements, and then blood and cervical vaginal fluid (CVF) samples were collected before delivery (at the second week after cervical cerclage). Pregnant women with PTB exhibited high white blood cell and neutrophil contents, high neutrophil-to-lymphocyte ratio (NLR), and high systemic inflammation response index (SIRI) in the blood. Vaginal microbiome revealed that the proportion of beneficial bacteria (including Lactobacillus, [Ruminococcus] gnavus group, and Megamonas) significantly decreased in the PTB group, and the proportion of harmful bacteria (including Desulfovibrionaceae, Helicobacter, and Gardnerella) significantly increased, which is strongly related to the biochemical parameters of blood (white blood cells, neutrophils, NLR, and SIRI). In addition, vaginal metabolomics-based liquid chromatography-Orbitrap-tandem mass spectrometry (LC-Orbitrap-MS/MS) found that the alteration in vaginal metabolites in pregnant women with PTB is involved in starch and sucrose metabolism; arginine and praline metabolism; galactose metabolism; purine metabolism; arginine metabolism; tryptophan metabolism and N-glycan biosynthesis; cysteine and methionine metabolism; taurine and hypotaurine metabolism; amino acid metabolism; propanoate metabolism; valine, leucine, and isoleucine biosynthesis; glycine, serine, and threonine metabolism; and steroid hormone biosynthesis. These results elaborated that distinct vaginal microbiome and metabolome profiles in women with preterm delivery following cervical cerclage provide valuable information for establishing the prediction models for PTB.
Collapse
Affiliation(s)
| | | | | | | | | | | | - Zhi Lin
- Department of Obstetrics and Gynecology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
| | - Mian Pan
- Department of Obstetrics and Gynecology, Fujian Maternity and Child Health Hospital, College of Clinical Medicine for Obstetrics and Gynecology and Pediatrics, Fujian Medical University, Fuzhou, Fujian, China
| |
Collapse
|
9
|
Yang F, Li X, Sun J, Pang X, Sun Q, Lu Y. Regulatory mechanisms of the probiotic-targeted gut-liver axis for the alleviation of alcohol-related liver disease: a review. Crit Rev Food Sci Nutr 2025:1-22. [PMID: 39905925 DOI: 10.1080/10408398.2025.2455954] [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: 02/06/2025]
Abstract
Alcohol abuse-triggered alcohol-related liver disease (ALD) has become as a global public health concern that substantially affects the well-being and clinical status of patients. Although modern medicine provides various treatments for ALD, their effectiveness is limited and can lead to adverse side effects. Probiotics have been employed to prevent, alleviate, and even treat ALD, with promising results. However, few comprehensive reviews are available on how they mitigate ALD by targeting the gut-liver axis. This review systematically clarifies the specific mediators of the gut-liver axis in healthy states. It also describes the alterations observed in ALD. Furthermore, this review thoroughly summarizes the underlying mechanisms through which probiotics act on the gut-liver axis to relieve ALD. It also discusses the current status and challenges faced in clinical research applications. Finally, we discuss the challenges and future prospects of using probiotics to treat ALD. This review improves our understanding of ALD and supports the development and application of probiotics that target the gut-liver axis for therapeutic use.
Collapse
Affiliation(s)
- Feiyu Yang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, China
| | - Xiangfei Li
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Jing Sun
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Xinyi Pang
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| | - Quancai Sun
- Department of Health, Nutrition, and Food Sciences, Florida State University, Tallahassee, FL, USA
| | - Yingjian Lu
- College of Food Science and Engineering, Nanjing University of Finance and Economics/Collaborative Innovation Center for Modern Grain Circulation and Safety, Nanjing, China
| |
Collapse
|
10
|
Nychas E, Marfil-Sánchez A, Chen X, Mirhakkak M, Li H, Jia W, Xu A, Nielsen HB, Nieuwdorp M, Loomba R, Ni Y, Panagiotou G. Discovery of robust and highly specific microbiome signatures of non-alcoholic fatty liver disease. MICROBIOME 2025; 13:10. [PMID: 39810263 PMCID: PMC11730835 DOI: 10.1186/s40168-024-01990-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/03/2024] [Accepted: 11/26/2024] [Indexed: 01/16/2025]
Abstract
BACKGROUND The pathogenesis of non-alcoholic fatty liver disease (NAFLD) with a global prevalence of 30% is multifactorial and the involvement of gut bacteria has been recently proposed. However, finding robust bacterial signatures of NAFLD has been a great challenge, mainly due to its co-occurrence with other metabolic diseases. RESULTS Here, we collected public metagenomic data and integrated the taxonomy profiles with in silico generated community metabolic outputs, and detailed clinical data, of 1206 Chinese subjects w/wo metabolic diseases, including NAFLD (obese and lean), obesity, T2D, hypertension, and atherosclerosis. We identified highly specific microbiome signatures through building accurate machine learning models (accuracy = 0.845-0.917) for NAFLD with high portability (generalizable) and low prediction rate (specific) when applied to other metabolic diseases, as well as through a community approach involving differential co-abundance ecological networks. Moreover, using these signatures coupled with further mediation analysis and metabolic dependency modeling, we propose synergistic defined microbial consortia associated with NAFLD phenotype in overweight and lean individuals, respectively. CONCLUSION Our study reveals robust and highly specific NAFLD signatures and offers a more realistic microbiome-therapeutics approach over individual species for this complex disease. Video Abstract.
Collapse
Affiliation(s)
- Emmanouil Nychas
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Beutenbergstraße 11A, Jena, 07745, Germany
| | - Andrea Marfil-Sánchez
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Beutenbergstraße 11A, Jena, 07745, Germany
| | - Xiuqiang Chen
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Beutenbergstraße 11A, Jena, 07745, Germany
| | - Mohammad Mirhakkak
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Beutenbergstraße 11A, Jena, 07745, Germany
| | - Huating Li
- Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Diabetes Institute, Shanghai, 200233, China
| | - Weiping Jia
- Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Diabetes Institute, Shanghai, 200233, China
| | - Aimin Xu
- The State Key Laboratory of Pharmaceutical Biotechnology, The University of Hong Kong, Hong Kong SAR, China
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China
- Department of Pharmacology and Pharmacy, The University of Hong Kong, Hong Kong SAR, China
| | | | - Max Nieuwdorp
- Amsterdam UMC, Location AMC, Department of Vascular Medicine, University of Amsterdam, Amsterdam, The Netherlands
| | - Rohit Loomba
- Department of Medicine, MASLD Research Center, University of California, San Diego, La Jolla, CA, 92093, USA
| | - Yueqiong Ni
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Beutenbergstraße 11A, Jena, 07745, Germany.
- Department of Endocrinology and Metabolism, Shanghai Clinical Center for Diabetes, Shanghai Key Laboratory of Diabetes Mellitus, Shanghai Jiao Tong University Affiliated Sixth People's Hospital, Shanghai Diabetes Institute, Shanghai, 200233, China.
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany.
| | - Gianni Panagiotou
- Department of Microbiome Dynamics, Leibniz Institute for Natural Product Research and Infection Biology - Hans Knöll Institute, Beutenbergstraße 11A, Jena, 07745, Germany.
- Faculty of Biological Sciences, Friedrich Schiller University, Jena, 07745, Germany.
- Department of Medicine, The University of Hong Kong, Hong Kong SAR, China.
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany.
| |
Collapse
|
11
|
Tang S, Wu S, Zhang W, Ma L, Zuo L, Wang H. Immunology and treatments of fatty liver disease. Arch Toxicol 2025; 99:127-152. [PMID: 39692857 DOI: 10.1007/s00204-024-03920-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Accepted: 11/26/2024] [Indexed: 12/19/2024]
Abstract
Alcoholic liver disease (ALD) and non-alcoholic fatty liver disease (NAFLD) are two major chronic liver diseases worldwide. The triggers for fatty liver can be derived from external sources such as adipose tissue, the gut, personal diet, and genetics, or internal sources, including immune cell responses, lipotoxicity, hepatocyte death, mitochondrial dysfunction, and extracellular vesicles. However, their pathogenesis varies to some extent. This review summarizes various immune mechanisms and therapeutic targets associated with these two types of fatty liver disease. It describes the gut-liver axis and adipose tissue-liver crosstalk, as well as the roles of different immune cells (both innate and adaptive immune cells) in fatty liver disease. Additionally, mitochondrial dysfunction, extracellular vesicles, microRNAs (miRNAs), and gastrointestinal hormones are also related to the pathogenesis of fatty liver. Understanding the pathogenesis of fatty liver and corresponding therapeutic strategies provides a new perspective for developing novel treatments for fatty liver disease.
Collapse
Affiliation(s)
- Sainan Tang
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Science, Anhui Medical University, Hefei, 230022, Anhui, China
| | - Shanshan Wu
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
- Department of Gastroenterology, the First Affiliated Hospital of Anhui Medical University, Hefei, Anhui, China
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, Anhui, China
| | - Wenzhe Zhang
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
- The First College of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Lili Ma
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China
- The First College of Clinical Medicine, Anhui Medical University, Hefei, Anhui, China
| | - Li Zuo
- Innovation and Entrepreneurship Laboratory for College Students, Anhui Medical University, Hefei, Anhui, China.
- Laboratory of Molecular Biology, Department of Biochemistry, School of Basic Medical Science, Anhui Medical University, Hefei, 230022, Anhui, China.
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, 230022, Anhui, China.
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Anhui Medical University, Hefei, Anhui, China.
| |
Collapse
|
12
|
Yin R, Wang T, Sun J, Dai H, Zhang Y, Liu N, Liu H. Postbiotics From Lactobacillus Johnsonii Activates Gut Innate Immunity to Mitigate Alcohol-Associated Liver Disease. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2405781. [PMID: 39574408 PMCID: PMC11727117 DOI: 10.1002/advs.202405781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/26/2024] [Revised: 10/10/2024] [Indexed: 01/14/2025]
Abstract
Prolonged alcohol consumption disrupts the gut microbiota and the immune system, contributing to the pathogenesis of alcohol-associated liver disease (ALD). Probiotic-postbiotic intervention strategies can effectively relieve ALD by maintaining gut homeostasis. Herein, the efficacy of heat-killed Lactobacillus johnsonii (HKLJ) in mitigating alcoholic liver damage is demonstrated in mouse models of ALD. The gut-liver axis is identified as a pivotal pathway for the protective effects of L. johnsonii against ALD. Specifically, HKLJ is found to upregulate the expression of intestinal lysozymes, thereby enhancing the production of immunoregulatory substances from gut bacteria, which subsequently activated the Nucleotide-binding oligomerization domain 2 (NOD2)-interleukin (IL-23)-IL-22 innate immune axis. The elevated IL-22 upregulated the antimicrobial peptide synthesis to maintain intestinal homeostasis and moreover activated the Signal transducer and activator of Transcription3 (STAT3) pathway in the liver to facilitate the repair of hepatic injuries. The heat-killed L. johnsonii provoked immunity helps correct the gut microbiota dysbiosis, specifically by reversing the reduction of butyrate-producing bacteria (such as Faecalibaculum rodentium) and the expansion of opportunistic pathogens (such as Helicobacter sp. and Pichia kudriavzevii) induced by ethanol. The findings provide novel insights into the gut microbiota-liver axis that may be leveraged to enhance the treatment of ALD.
Collapse
Affiliation(s)
- Ruopeng Yin
- State Key Laboratory of MycologyInstitute of MicrobiologyChinese Academy of SciencesBeijing100101China
- Medical SchoolUniversity of Chinese Academy of SciencesBeijing100049China
| | - Tao Wang
- State Key Laboratory of MycologyInstitute of MicrobiologyChinese Academy of SciencesBeijing100101China
| | - Jingzu Sun
- State Key Laboratory of MycologyInstitute of MicrobiologyChinese Academy of SciencesBeijing100101China
| | - Huanqin Dai
- State Key Laboratory of MycologyInstitute of MicrobiologyChinese Academy of SciencesBeijing100101China
- Medical SchoolUniversity of Chinese Academy of SciencesBeijing100049China
| | - Yuting Zhang
- State Key Laboratory of MycologyInstitute of MicrobiologyChinese Academy of SciencesBeijing100101China
- Medical SchoolUniversity of Chinese Academy of SciencesBeijing100049China
| | - Ningning Liu
- CAS Key Laboratory of Pathogenic Microbiology and ImmunologyInstitute of MicrobiologyChinese Academy of SciencesBeijing100101China
| | - Hongwei Liu
- State Key Laboratory of MycologyInstitute of MicrobiologyChinese Academy of SciencesBeijing100101China
- Medical SchoolUniversity of Chinese Academy of SciencesBeijing100049China
| |
Collapse
|
13
|
Wang G, Qiu X, Sun M, Li Y, Chen A, Tong J, Cheng Z, Zhao W, Chang C, Yu G. RegⅢγ promotes the proliferation, and inhibits inflammation response of macrophages by Akt, STAT3 and NF-κB pathways. Int Immunopharmacol 2024; 143:113442. [PMID: 39490142 DOI: 10.1016/j.intimp.2024.113442] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2024] [Revised: 10/16/2024] [Accepted: 10/17/2024] [Indexed: 11/05/2024]
Abstract
As an inflammatory regulator, intestinal regenerating islet-derived 3 gamma (RegⅢγ) contributes to alleviating liver injury in liver diseases and colitis. However, it is unclear whether hepatic RegⅢγ exerts a vital impact on liver regeneration (LR). In this study, the expression profile and localization of RegⅢγ in LR were demonstrated by microarray analysis, qRT-PCR and immunofluorescence staining. Then, RAW264.7 cells with RegⅢγ deficiency and overexpression were obtained by the CRISPR/Cas9 system and lentivirus infection, respectively. MTT, flow cytometry, EdU, transwell, neutral red phagocytosis, and NO assays were performed to detect the functions of RegⅢγ in RAW264.7 cell proliferation and inflammation. Finally, the regulatory mechanism of RegⅢγ was explored by co-immunoprecipitation and Western blot assays. According to our findings, RegⅢγ showed significant expression changes in Kupffer cells during LR, and RegⅢγ overexpression stimulated the viability, proliferation, phagocytosis and migration of RAW264.7 cells, whereas RegⅢγ deficiency reversed these effects. Similarly, RegⅢγ overexpression facilitated the expression of HO-1 and IL-10, while RegⅢγ deficiency promoted NO production and p-Akt, p-STAT3, p-p65 and TNF-α expression. In conclusion, RegⅢγ may facilitate LR by promoting the proliferation of macrophages and inhibiting their inflammatory response through Akt, STAT3 and NF-κB pathways in the priming stage of LR.
Collapse
Affiliation(s)
- Gaiping Wang
- State Key Laboratory of Cell Differentiation and Regulation, Henan Normal University, Xinxiang 453007, Henan, China; Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Henan Normal University, Xinxiang 453007, Henan, China; Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Normal University, Xinxiang 453007, Henan, China; Institute of Biomedical Science, Henan Normal University, Xinxiang 453007, Henan, China; College of Life Science, Henan Normal University, Xinxiang 453007, Henan, China.
| | - Xianglei Qiu
- State Key Laboratory of Cell Differentiation and Regulation, Henan Normal University, Xinxiang 453007, Henan, China; College of Life Science, Henan Normal University, Xinxiang 453007, Henan, China
| | - Meiqing Sun
- State Key Laboratory of Cell Differentiation and Regulation, Henan Normal University, Xinxiang 453007, Henan, China; College of Life Science, Henan Normal University, Xinxiang 453007, Henan, China
| | - Yingle Li
- State Key Laboratory of Cell Differentiation and Regulation, Henan Normal University, Xinxiang 453007, Henan, China; College of Life Science, Henan Normal University, Xinxiang 453007, Henan, China
| | - Anqi Chen
- State Key Laboratory of Cell Differentiation and Regulation, Henan Normal University, Xinxiang 453007, Henan, China; College of Life Science, Henan Normal University, Xinxiang 453007, Henan, China
| | - Jiahui Tong
- State Key Laboratory of Cell Differentiation and Regulation, Henan Normal University, Xinxiang 453007, Henan, China; College of Life Science, Henan Normal University, Xinxiang 453007, Henan, China
| | - Zhipeng Cheng
- State Key Laboratory of Cell Differentiation and Regulation, Henan Normal University, Xinxiang 453007, Henan, China; College of Life Science, Henan Normal University, Xinxiang 453007, Henan, China
| | - Weiming Zhao
- Institute of Biomedical Sciences, Henan Academy of Sciences, Zhengzhou 450046, Henan, China
| | - Cuifang Chang
- State Key Laboratory of Cell Differentiation and Regulation, Henan Normal University, Xinxiang 453007, Henan, China; Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Henan Normal University, Xinxiang 453007, Henan, China; Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Normal University, Xinxiang 453007, Henan, China; Institute of Biomedical Science, Henan Normal University, Xinxiang 453007, Henan, China; College of Life Science, Henan Normal University, Xinxiang 453007, Henan, China
| | - Guoying Yu
- State Key Laboratory of Cell Differentiation and Regulation, Henan Normal University, Xinxiang 453007, Henan, China; Henan Center for Outstanding Overseas Scientists of Organ Fibrosis, Henan Normal University, Xinxiang 453007, Henan, China; Henan International Joint Laboratory of Pulmonary Fibrosis, Henan Normal University, Xinxiang 453007, Henan, China; Institute of Biomedical Science, Henan Normal University, Xinxiang 453007, Henan, China; College of Life Science, Henan Normal University, Xinxiang 453007, Henan, China.
| |
Collapse
|
14
|
Li K, Wei W, Xu C, Lian X, Bao J, Yang S, Wang S, Zhang X, Zheng X, Wang Y, Zhong S. Prebiotic inulin alleviates anxiety and depression-like behavior in alcohol withdrawal mice by modulating the gut microbiota and 5-HT metabolism. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2024; 135:156181. [PMID: 39488100 DOI: 10.1016/j.phymed.2024.156181] [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: 06/30/2024] [Revised: 10/02/2024] [Accepted: 10/21/2024] [Indexed: 11/04/2024]
Abstract
BACKGROUND Alcohol dependence (AD) is a common psychiatric disorder, often accompanied by anxiety and depression. These comorbidities are linked to disturbances in serotonin (5-HT) metabolism and gut microbiota dysbiosis. Clinical studies suggest that inulin, a prebiotic, can alleviate anxiety and depression in AD patients by affecting the gut microbiota, although the mechanisms remain unclear. PURPOSE The purpose of this study is to investigate the potential mechanisms by which inulin, a prebiotic, improves anxiety and depression-like behaviors in AD withdrawal mice. This research is based on the drug and food homology and intestinal treatment of encephalopathy, with the goal of developing new clinical strategies for AD treatment. STUDY DESIGN For this purpose, fecal samples from AD patients were analyzed to identify microorganisms associated with AD. An AD withdrawal mouse model was created, with inulin as the intervention and fluvoxamine maleate as the control. Techniques such as 16S microbiome sequencing and UPLC-TQMS-targeted metabolomics were used to assess gut microbiota, short-chain fatty acids (SCFAs) levels, and 5-HT metabolism. METHODS The AD withdrawal model was built using the "Drinking-in-the-dark" protocol over 6 weeks. Inulin (2 g/kg/day) and fluvoxamine maleate (30 mg/kg/day) were administered for 4 weeks. The open field test, forced swim test, and tail suspension test were used to evaluate anxiety and depression-like behaviors in mice. ELISA and qRT-PCR assessed 5-HT metabolism in the colon, blood, and prefrontal cortex, while 16S microbiome sequencing analyzed changes in gut microbiota and UPLC-TQMS examined SCFAs levels. Immunohistochemistry was used to study intestinal barrier integrity. RESULTS AD patients showed reduced SCFA-producing bacteria such as Faecalibacterium and Roseburia. In mice, AD withdrawal led to anxiety and depression-like behaviors, disrupted 5-HT metabolism, and gut microbiota dysbiosis. Inulin supplementation alleviated these behaviors, increased 5-HT and 5-hydroxytryptophan (5-HTP) levels, upregulated colonic tryptophan hydroxylase 1 (TPH1) expression, and promoted the growth of beneficial bacteria such as Faecalibacterium and Roseburia, while also increasing SCFAs levels. CONCLUSION Inulin increases the abundance of Faecalibacterium and Roseburia, enhances SCFAs production, and regulates 5-HT metabolism, improving anxiety and depression-like behaviors in AD withdrawal mice. These findings suggest that inulin may serve as a nutritional intervention for mental health in AD patients by targeting the microbiome-gut-brain axis.
Collapse
Affiliation(s)
- Kuan Li
- School of Forensic Medicine, Kunming Medical University, Kunming, 650500, China
| | - Wei Wei
- School of Forensic Medicine, Kunming Medical University, Kunming, 650500, China
| | - Chongchong Xu
- School of Forensic Medicine, Kunming Medical University, Kunming, 650500, China
| | - Xinqing Lian
- School of Forensic Medicine, Kunming Medical University, Kunming, 650500, China
| | - Jianjun Bao
- Department of Geriatric Psychiatry, The Mental Hospital of Yunnan Province, Kunming, 650224, China
| | - Shuo Yang
- School of Forensic Medicine, Kunming Medical University, Kunming, 650500, China
| | - Shixu Wang
- School of Forensic Medicine, Kunming Medical University, Kunming, 650500, China
| | - Xulan Zhang
- Department of Psychiatry/Alcohol Dependence Treatment, The Mental Hospital of Yunnan Province, Kunming, 650224, China
| | - Xinjian Zheng
- School of Forensic Medicine, Kunming Medical University, Kunming, 650500, China
| | - Yue Wang
- School of Forensic Medicine, Kunming Medical University, Kunming, 650500, China
| | - Shurong Zhong
- School of Forensic Medicine, Kunming Medical University, Kunming, 650500, China; NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, Kunming Medical University, Kunming, 650500, China; Forensic Biology Identification Laboratory, Judicial Identification Center of Kunming Medical University, Kunming, 650500, China.
| |
Collapse
|
15
|
Huang YE, Chen SY, Li TJ, Tsai YS, Chen CC, Yen GC. Gastroprotective effects of Pediococcus acidilactici GKA4 and Lactobacillus brevis GKL93 against ethanol-induced gastric ulcers via regulation of the immune response and gut microbiota in mice. Food Funct 2024; 15:11491-11507. [PMID: 39480654 DOI: 10.1039/d4fo04106b] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2024]
Abstract
Excessive alcohol consumption is a significant pathogenic factor involved in the initiation of noninfectious gastric ulcers. Probiotics based on a specific strain can mitigate gastric damage. However, the protective effects of Pediococcus acidilactici (GKA4) and Lactobacillus brevis (GKL93) against alcohol-induced gastric mucosal damage remain unclear. Hence, the gastroprotective effects of these probiotic strains were investigated in BALB/c mice with gastric mucosa damage induced by absolute alcohol. The results revealed that preadministration of GKA4 and GKL93 increased the expression of antioxidative enzymes (SOD, catalase, GPx), anti-inflammatory cytokines (IL-4 and IL-10), and heat shock protein genes (HSP70 and HSP90) and decreased the expression of apoptosis-related genes (Bax, cytochrome c, and caspase-3), MDA, and pro-inflammatory cytokines (IL-1β, IL-6, and TNF-α). Mechanistically, GKA4 and GKL93 increased the relative abundance of beneficial flora (Coriobacteriia, Lachnospiraceae_NK4A136_group, Roseburia, f__Oscillospiraceae unclassified, Ruminococcus, Adlercreutzia, and [Eubacterium]_xylanophilum_group) that may promote antioxidant and anti-inflammatory effects via upregulation of the expression of proteins in the Nrf2/HO-1 pathway and downregulation of the expression of proteins in the NF-κB/iNOS/COX-2 signaling pathway, subsequently attenuating gastrointestinal permeability and ulcer symptoms. Furthermore, correlation analysis revealed that [Eubacterium]_xylanophilum_group and f_Oscillospiraceae_unclassified were two significant beneficial flora associated with ethanol-induced gastric ulcers after preadministration of GKA4 and GKL93. In summary, the gastroprotective effects of P. acidilactici GKA4 and L. brevis GKL93 against ethanol-induced gastric ulcers in mice include suppressing oxidative- and inflammatory-related pathways and modulation of the gut microbiota. This novel finding highlights the potential of these probiotics as functional materials in preventing alcohol-induced gastric mucosal damage.
Collapse
Affiliation(s)
- Yun-En Huang
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung 40227, Taiwan.
| | - Sheng-Yi Chen
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung 40227, Taiwan.
| | - Tsung-Ju Li
- Biotech Research Institute, Grape King Bio Ltd, Taoyuan 32542, Taiwan
| | - You-Shan Tsai
- Biotech Research Institute, Grape King Bio Ltd, Taoyuan 32542, Taiwan
| | - Chin-Chu Chen
- Biotech Research Institute, Grape King Bio Ltd, Taoyuan 32542, Taiwan
| | - Gow-Chin Yen
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung 40227, Taiwan.
- Advanced Plant and Food Crop Biotechnology Center, National Chung Hsing University, Taichung 40227, Taiwan
| |
Collapse
|
16
|
Geng X, Zhuang M, Tian W, Shang H, Gong Z, Lv Y, Li J. Green Radish Polysaccharide Prevents Alcoholic Liver Injury by Interfering with Intestinal Bacteria and Short-Chain Fatty Acids in Mice. Foods 2024; 13:3733. [PMID: 39682806 DOI: 10.3390/foods13233733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2024] [Revised: 11/12/2024] [Accepted: 11/19/2024] [Indexed: 12/18/2024] Open
Abstract
This study aimed to ascertain the potential benefits of green radish polysaccharide (GRP) in treating alcoholic liver disease (ALD) in mice and explore its mechanism of action. Using biochemical analysis, high-throughput sequencing of gut microbiota, and gas chromatography-mass spectrometry to measure short-chain fatty acids (SCFAs) in feces, we found that GRP intervention significantly improved lipid metabolism and hepatic function in mice subjected to excessive alcohol intake. The GRP intervention reduced malondialdehyde levels by 66% and increased total superoxide dismutase levels by 22%, thereby mitigating alcohol-induced oxidative stress. Furthermore, GRP intervention in mice with alcohol consumption resulted in a reduction in tumor necrosis factor, interleukin 6, and lipopolysaccharide levels by 12%, 9%, and 25%, respectively, effectively attenuating alcoholic liver inflammation. 16S rRNA amplicon sequencing demonstrated that excessive alcohol consumption markedly altered the gut microbiota composition in mice. The GRP treatment resulted in a significant reduction in the number of beneficial bacteria (Lactobacillus and Lachnospiraceae_NK4A136_group) and an increase in the proportion of harmful bacteria (Muribaculaceae and Verrucomicrobiota). The metabolomic analyses of the SCFAs demonstrated an increase in the contents of SCFAs, acetic acid, propionic acid, and butyric acid, following GRP supplementation. Furthermore, the metabolic levels of cholinergic synapses and glycolysis/gluconeogenesis were found to be modulated. In conclusion, these findings suggest that GRP may attenuate alcohol-induced oxidative damage in the liver by modulating the gut microbiota and hepatic metabolic pathways. This may position GRP as a potential functional component for ALD prevention.
Collapse
Affiliation(s)
- Xiong Geng
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Miaomiao Zhuang
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Weina Tian
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Huayan Shang
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Ziyi Gong
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Yanfang Lv
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| | - Jianrong Li
- College of Food Science and Engineering, Bohai University, Jinzhou 121013, China
| |
Collapse
|
17
|
Pan X, Song Y, Liang Y, Feng G, Wang Z. Roseburia intestinalis: A possible target for vascular calcification. Heliyon 2024; 10:e39865. [PMID: 39524709 PMCID: PMC11550659 DOI: 10.1016/j.heliyon.2024.e39865] [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: 01/21/2024] [Revised: 10/22/2024] [Accepted: 10/25/2024] [Indexed: 11/16/2024] Open
Abstract
With the advancement of metagenomics and metabolomics techniques, the crucial role of the gut microbiome in intestinal, cardiovascular, and metabolic disorders has been extensively explored. Vascular calcification (VC) is common in atherosclerosis, hypertension, diabetes mellitus, and chronic kidney disease. Moreover, it is a significant cause of cardiovascular diseases and mortality. Roseburia intestinalis, as a promising candidate for the next generation of probiotics, plays a substantial role in inhibiting the systemic inflammatory response and holds great potential in the treatment of intestinal diseases, cardiovascular diseases, and metabolic disorders. Its primary metabolite, butyrate, acts on specific receptors (GPR43, GPR41, GPR109a). It enters cells via transporters (MCT1, SMCT1), affecting gene expression through HDACs, PPARγ and Nrf2, promoting energy metabolism and changing the concentration of other metabolites (including AGEs, LPS, BHB) in the circulation to affect the body's life activities. In this paper, we focus on the possible mechanism of the primary metabolite butyrate of Roseburia intestinalis in inhibiting VC, which may become a potential therapeutic target for the treatment of VC and the ways to enhance its effect.
Collapse
Affiliation(s)
- Xinyun Pan
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
- Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang, 21200, China
| | - Yunjian Song
- Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang, 21200, China
| | - Yapeng Liang
- Department of Emergency, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Guoquan Feng
- Department of Imaging, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
| | - Zhongqun Wang
- Department of Cardiology, Affiliated Hospital of Jiangsu University, Zhenjiang, 212001, China
- Institue of Cardiovascular Diseases, Jiangsu University, Zhenjiang, 21200, China
| |
Collapse
|
18
|
Yang Y, Schnabl B. Gut Bacteria in Alcohol-Associated Liver Disease. Clin Liver Dis 2024; 28:663-679. [PMID: 39362714 PMCID: PMC11450261 DOI: 10.1016/j.cld.2024.06.008] [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] [Indexed: 10/05/2024]
Abstract
Alcohol-associated liver disease (ALD) poses a significant global public health challenge, with high patient mortality rates and economic burden. The gut microbiome plays an important role in the onset and progression of alcohol-associated liver disease. Excessive alcohol consumption disrupts the intestinal barrier, facilitating the entry of harmful microbes and their products into the liver, exacerbating liver damage. Dysbiosis, marked by imbalance in gut bacteria, correlates with ALD severity. Promising microbiota-centered therapies include probiotics, phages, and fecal microbiota transplantation. Clinical trials demonstrate the potential of these interventions to improve liver function and patient outcomes, offering a new frontier in ALD treatment.
Collapse
Affiliation(s)
- Yongqiang Yang
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | - Bernd Schnabl
- Department of Medicine, University of California San Diego, La Jolla, CA 92093, USA; Department of Medicine, VA San Diego Healthcare System, San Diego, CA 92161, USA.
| |
Collapse
|
19
|
Romero-Rodríguez A, Ruíz-Villafán B, Sánchez S, Paredes-Sabja D. Is there a role for intestinal sporobiota in the antimicrobial resistance crisis? Microbiol Res 2024; 288:127870. [PMID: 39173554 DOI: 10.1016/j.micres.2024.127870] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 07/23/2024] [Accepted: 08/06/2024] [Indexed: 08/24/2024]
Abstract
Antimicrobial resistance (AMR) is a complex issue requiring specific, multi-sectoral measures to slow its spread. When people are exposed to antimicrobial agents, it can cause resistant bacteria to increase. This means that the use, misuse, and excessive use of antimicrobial agents exert selective pressure on bacteria, which can lead to the development of "silent" reservoirs of antimicrobial resistance genes. These genes can later be mobilized into pathogenic bacteria and contribute to the spread of AMR. Many socioeconomic and environmental factors influence the transmission and dissemination of resistance genes, such as the quality of healthcare systems, water sanitation, hygiene infrastructure, and pollution. The sporobiota is an essential part of the gut microbiota that plays a role in maintaining gut homeostasis. However, because spores are highly transmissible and can spread easily, they can be a vector for AMR. The sporobiota resistome, particularly the mobile resistome, is important for tracking, managing, and limiting the spread of antimicrobial resistance genes among pathogenic and commensal bacterial species.
Collapse
Affiliation(s)
- A Romero-Rodríguez
- Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México, Ciudad de México 04510, Mexico.
| | - B Ruíz-Villafán
- Laboratorio de Microbiología Industrial. Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - S Sánchez
- Laboratorio de Microbiología Industrial. Departamento de Biología Molecular y Biotecnología, Instituto de Investigaciones Biomédicas, Universidad Nacional Autónoma de México, Ciudad de México 04510, Mexico
| | - D Paredes-Sabja
- Department of Biology, Texas A&M University, College Station, TX 77843, USA
| |
Collapse
|
20
|
Sharma SP, Gupta H, Kwon GH, Lee SY, Song SH, Kim JS, Park JH, Kim MJ, Yang DH, Park H, Won SM, Jeong JJ, Oh KK, Eom JA, Lee KJ, Yoon SJ, Ham YL, Baik GH, Kim DJ, Suk KT. Gut microbiome and metabolome signatures in liver cirrhosis-related complications. Clin Mol Hepatol 2024; 30:845-862. [PMID: 39048520 PMCID: PMC11540350 DOI: 10.3350/cmh.2024.0349] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 07/24/2024] [Accepted: 07/24/2024] [Indexed: 07/27/2024] Open
Abstract
BACKGROUND/AIMS Shifts in the gut microbiota and metabolites are interrelated with liver cirrhosis progression and complications. However, causal relationships have not been evaluated comprehensively. Here, we identified complication-dependent gut microbiota and metabolic signatures in patients with liver cirrhosis. METHODS Microbiome taxonomic profiling was performed on 194 stool samples (52 controls and 142 cirrhosis patients) via V3-V4 16S rRNA sequencing. Next, 51 samples (17 controls and 34 cirrhosis patients) were selected for fecal metabolite profiling via gas chromatography mass spectrometry and liquid chromatography coupled to time-of-flight mass spectrometry. Correlation analyses were performed targeting the gut-microbiota, metabolites, clinical parameters, and presence of complications (varices, ascites, peritonitis, encephalopathy, hepatorenal syndrome, hepatocellular carcinoma, and deceased). RESULTS Veillonella bacteria, Ruminococcus gnavus, and Streptococcus pneumoniae are cirrhosis-related microbiotas compared with control group. Bacteroides ovatus, Clostridium symbiosum, Emergencia timonensis, Fusobacterium varium, and Hungatella_uc were associated with complications in the cirrhosis group. The areas under the receiver operating characteristic curve (AUROCs) for the diagnosis of cirrhosis, encephalopathy, hepatorenal syndrome, and deceased were 0.863, 0.733, 0.71, and 0.69, respectively. The AUROCs of mixed microbial species for the diagnosis of cirrhosis and complication were 0.808 and 0.847, respectively. According to the metabolic profile, 5 increased fecal metabolites in patients with cirrhosis were biomarkers (AUROC >0.880) for the diagnosis of cirrhosis and complications. Clinical markers were significantly correlated with the gut microbiota and metabolites. CONCLUSION Cirrhosis-dependent gut microbiota and metabolites present unique signatures that can be used as noninvasive biomarkers for the diagnosis of cirrhosis and its complications.
Collapse
Affiliation(s)
- Satya Priya Sharma
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Korea
| | - Haripriya Gupta
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Korea
| | - Goo-Hyun Kwon
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Korea
| | - Sang Yoon Lee
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Korea
| | - Seol Hee Song
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Korea
| | - Jeoung Su Kim
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Korea
| | - Jeong Ha Park
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Korea
| | - Min Ju Kim
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Korea
| | - Dong-Hoon Yang
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Korea
| | - Hyunjoon Park
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Korea
| | - Sung-Min Won
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Korea
| | - Jin-Ju Jeong
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Korea
| | - Ki-Kwang Oh
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Korea
| | - Jung A Eom
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Korea
| | - Kyeong Jin Lee
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Korea
| | - Sang Jun Yoon
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Korea
| | - Young Lim Ham
- Department of Nursing Daewon University College Jecheon, Korea
| | - Gwang Ho Baik
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Korea
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Korea
| | - Dong Joon Kim
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Korea
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Korea
| | - Ki Tae Suk
- Institute for Liver and Digestive Diseases, Hallym University, Chuncheon, Korea
- Department of Internal Medicine, Hallym University College of Medicine, Chuncheon, Korea
| |
Collapse
|
21
|
Yin C, Liu L, Xu D, Li M, Li M, Qin Y, Zhang B, Sun Y, Liu Y, Xiao Y. Integrative metagenomic and lipidomic analyses reveal alterations in children with obesity and after lifestyle intervention. Front Nutr 2024; 11:1423724. [PMID: 39318384 PMCID: PMC11420138 DOI: 10.3389/fnut.2024.1423724] [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: 04/26/2024] [Accepted: 08/26/2024] [Indexed: 09/26/2024] Open
Abstract
Background Despite emerging evidence linking alterations in gut microbiota to childhood obesity, the metabolic mechanisms linking gut microbiota to the lipid profile during childhood obesity and weight loss remain poorly understood. Methodology In this study, children with obesity were treated with lifestyle weight loss therapy. Metagenomics association studies and serum untargeted lipidomics analyses were performed in children with obesity and healthy controls before and after weight loss. Main findings We identified alterations in gut microbiota associated with childhood obesity, as well as variations in circulating metabolite concentrations. Children with obesity showed significant decreases in the levels of s-Rothia_kristinae and s-Enterobacter_roggenkampii, alongsige elevated levels of s-Clostridiales_bacterium_Marseille-P5551. Following weight loss, the levels of s-Streptococcus_infantarius and s-Leuconostoc_citreum increased by factors of 3.354 and 1.505, respectively, in comparison to their pre-weight loss levels. Correlation analyses indicated a significant positive relationship between ChE(2:0) levels and both with s-Lachnospiraceae_bacterium_TF09-5 and fasting glucose levels. CoQ8 levels were significantly negatively correlated with s-Rothia_kristinae and HOMA-IR. Conclusion We linked altered gut microbiota and serum lipid levels in children with obesity to clinical indicators, indicating a potential impact on glucose metabolism via lipids. This study contributes to understanding the mechanistic relationship between altered gut microbiota and childhood obesity and weight loss, suggesting gut microbiome as a promising target for intervention. Clinical trial registration https://www.chictr.org.cn/showproj.html?proj=178971, ChiCTR2300072179.
Collapse
Affiliation(s)
- Chunyan Yin
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Lujie Liu
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Dong Xu
- Department of Pediatrics, Tongji Medical College, Tongji Hospital, Huazhong University of Science and Technology, Wuhan, China
| | - Meng Li
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Min Li
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yujie Qin
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Bei Zhang
- Department of Pediatrics, Luoyang Central Hospital, Luoyang, China
| | - Yongfa Sun
- Department of Pediatrics, Luoyang Central Hospital, Luoyang, China
| | - Yuesheng Liu
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| | - Yanfeng Xiao
- Department of Pediatrics, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi’an, China
| |
Collapse
|
22
|
Huang J, Liu S, Li P, Wei L, Lin G, Lin J, Luo Y, Liu Y, Mao Y, Ruan H, Qin B, Fan P, Lu T, Cai W, Yi H, Mou X, Lu Z, Zhao W, Wu A. Multi-omics analysis of gut-brain axis reveals novel microbial and neurotransmitter signatures in patients with arteriosclerotic cerebral small vessel disease. Pharmacol Res 2024; 208:107385. [PMID: 39245190 DOI: 10.1016/j.phrs.2024.107385] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/11/2024] [Revised: 08/27/2024] [Accepted: 08/28/2024] [Indexed: 09/10/2024]
Abstract
Arteriosclerotic cerebral small vessel disease (aCSVD) is a major cause of stroke and dementia. Although its underlying pathogenesis remains poorly understood, both inflammaging and gut microbiota dysbiosis have been hypothesized to play significant roles. This study investigated the role of gut microbiota in the pathogenesis of aCSVD through a comparative analysis of the gut microbiome and metabolome between CSVD patients and healthy controls. The results showed that patients with aCSVD exhibited a marked reduction in potentially beneficial bacterial species, such as Faecalibacterium prausnitzli and Roseburia intestinalis, alongside an increase in taxa from Bacteroides and Proteobacteria. Integrated metagenomic and metabolomic analyses revealed that alterations in microbial metabolic pathways, including LPS biosynthesis and phenylalanine-tyrosine metabolism, were associated with the status of aCSVD. Our findings indicated that microbial LPS biosynthesis and phenylalanine-tyrosine metabolism potentially influenced the symptoms and progression of aCSVD via pro-inflammatory effect and modulation of systemic neurotransmitters, respectively. These results imply that gut microbiota characteristics may serve as indicators for early detection of aCSVD and as potential gut-directed therapeutic intervention target.
Collapse
Affiliation(s)
- Jiayuan Huang
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Sanxin Liu
- Department of Neurology, Center for the Study of Mental and Neurological Disorders, the Third Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Peijie Li
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Lei Wei
- Department of Neurology, Center for the Study of Mental and Neurological Disorders, the Third Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Gan Lin
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Jiahao Lin
- Department of Neurology, Center for the Study of Mental and Neurological Disorders, the Third Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Yuting Luo
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Yixin Liu
- Department of Neurology, Center for the Study of Mental and Neurological Disorders, the Third Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Yudan Mao
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China
| | - Hengfang Ruan
- Department of Neurology, Center for the Study of Mental and Neurological Disorders, the Third Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Bing Qin
- Department of Neurology, Center for the Study of Mental and Neurological Disorders, the Third Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Ping Fan
- Department of Neurology, Center for the Study of Mental and Neurological Disorders, the Third Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Tingting Lu
- Department of Neurology, Center for the Study of Mental and Neurological Disorders, the Third Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Wei Cai
- Department of Neurology, Center for the Study of Mental and Neurological Disorders, the Third Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Haotong Yi
- Department of Neurology, Center for the Study of Mental and Neurological Disorders, the Third Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, Guangdong 510630, China
| | - Xiangyu Mou
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| | - Zhengqi Lu
- Department of Neurology, Center for the Study of Mental and Neurological Disorders, the Third Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, Guangdong 510630, China.
| | - Wenjing Zhao
- Shenzhen Key Laboratory of Systems Medicine for Inflammatory Diseases, School of Medicine, Shenzhen Campus of Sun Yat-Sen University, Shenzhen, Guangdong 518107, China.
| | - Aimin Wu
- Department of Neurology, Center for the Study of Mental and Neurological Disorders, the Third Affiliated Hospital of Sun Yat-Sen University, Sun Yat-Sen University, Guangzhou, Guangdong 510630, China.
| |
Collapse
|
23
|
He Y, Nong Y, Qin J, Feng L, Qin J, Wang Q, Deng L, Tang S, Zhang M, Fan X, Dong M, Wei J, Pan S, Su Z. Protective effects of oyster polypeptide on cyclophosphamide-induced immunosuppressed rats. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2024; 104:7143-7158. [PMID: 38629663 DOI: 10.1002/jsfa.13537] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Revised: 02/26/2024] [Accepted: 04/17/2024] [Indexed: 05/01/2024]
Abstract
BACKGROUND Oyster polypeptide (OP) is a mixture of oligopeptides extracted from oysters through enzyme lysis, separation, and purification. It is associated with immunomodulatory effects, but the underlying mechanisms are not known. This study therefore combined proton nuclear magnetic resonance (1H-NMR) urinary metabolomics and 16S rRNA gene sequencing of the gut microbiome to determine the immunoprotective mechanisms of OP in rats subjected to cyclophosphamide-induced immunosuppression. RESULTS Oyster polypeptide restored the body weight and the structure of spleen and thymus in rats with cyclophosphamide-induced immunosuppression. It upregulated the levels of white blood cells (WBCs), hemoglobin (HGB), platelets (PLT), red blood cells (RBCs), immunoglobulin G (IgG), immunoglobulin M (IgM), cytokines such as interleukin‑6 (IL-6) and tumor necrosis factor-α (TNF-α), and increased the numbers of CD3+ and CD4+ T cells in the immunosuppressed rats. The 1H-NMR metabolomics results showed that OP significantly reversed the levels of ten metabolites in urine, including 2-oxoglutarate, citrate, dimethylamine, taurine, N-phenylacetylglycine, alanine, betaine, creatinine, uracil, and benzoate. The 16S rRNA gene sequencing results showed that OP restored the gut microbiome homeostasis by increasing the abundance of beneficial bacteria and reducing the abundance of pathogenic bacteria. Finally, a combination of metabolomics and microbiomics found that the metabolism of taurine and hypotaurine, and the metabolism of alanine, aspartate, and glutamate were disturbed, but these metabolic pathways were restored by OP. CONCLUSION This study demonstrated that OP had immunoprotective effects in rats with cyclophosphamide-induced immunosuppression by restoring key metabolic pathways and the gut microbiome homeostasis. Our findings provide a framework for further research into the immunoregulatory mechanisms of OP and its potential use in drugs and nutritional supplements. © 2024 Society of Chemical Industry.
Collapse
Affiliation(s)
- Ying He
- First clinical medical college, Guangxi Medical University, Nanning, China
| | - Yunyuan Nong
- Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Junliang Qin
- Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Linlin Feng
- Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Jinghua Qin
- Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Qianyi Wang
- Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Lijun Deng
- Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Siqi Tang
- First clinical medical college, Guangxi Medical University, Nanning, China
| | - Meiling Zhang
- Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Xiaofeng Fan
- Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Min Dong
- Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Jinbin Wei
- Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Shihan Pan
- Pharmaceutical College, Guangxi Medical University, Nanning, China
| | - Zhiheng Su
- Pharmaceutical College, Guangxi Medical University, Nanning, China
- Guangxi Key Laboratory of Bioactive Molecules Research and Evaluation, Nanning, China
- Guangxi Beibu Gulf Marine Biomedicine Precision Development and High-value Utilization Engineering Research Center, Nanning, China
- Guangxi Health Commission Key Laboratory of Basic Research on Antigeriatric Drugs, Nanning, China
| |
Collapse
|
24
|
Chen K, Geng H, Ye C, Liu J. Dysbiotic alteration in the fecal microbiota of patients with polycystic ovary syndrome. Microbiol Spectr 2024; 12:e0429123. [PMID: 38990031 PMCID: PMC11302149 DOI: 10.1128/spectrum.04291-23] [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: 12/25/2023] [Accepted: 02/13/2024] [Indexed: 07/12/2024] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common disease associated with high androgen and infertility. The gut microbiota plays an important role in metabolic diseases including obesity, hyperglycemia, and fatty liver. Although the gut microbiota has been associated with PCOS, little is known about the gut microbial structure and function in individuals with PCOS from Northeast China. In this study, 17 PCOS individuals and 17 age-matched healthy individuals were recruited for community structure and function analysis of the gut microbiota. The results showed that PCOS individuals have reduced diversity and richness of the gut microbiota compared with healthy individuals. Beta diversity analysis showed that the community structure of the gut microbiota of individuals with PCOS was significantly separated from healthy individuals. At the phylum level, PCOS individuals have reduced Firmicutes and Bacteroidota and increased Actinobacteriota and Proteobacteria compared with healthy individuals. At the family and genus levels, the composition of the gut microbiota between PCOS patients and healthy individuals was also significantly different. In addition, PICRUSt2 showed that individuals with PCOS have different microbial functions in the gut compared with healthy individuals. We finally confirmed that Bifidobacterium was enriched in the fecal samples of PCOS patients, while other 11 genera including Bacteroides, UCG_002, Eubacterium__coprostanoligenes_group_unclassified, Dialister, Firmicutes_unclassified, Ruminococcus, Alistipes, Christensenellaceae_R_7_group, Clostridia_UCG_014_unclassified, Roseburia, and Lachnospiraceae_unclassified were depleted compared with healthy individuals. These results indicate that individuals with PCOS have altered community structure and functions of the gut microbiota, which suggests that targeting the gut microbiota might be a potential strategy for PCOS intervention. IMPORTANCE Gut microbiota plays a critical role in the development of PCOS. There is a complex and close interaction between PCOS and gut microbiota. The relationship between the pathogenesis and pathophysiological processes of PCOS and the structure and function of the gut microbiota needs further investigation.
Collapse
Affiliation(s)
- Ke Chen
- Department of Gynecology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Huafeng Geng
- Department of Gynecology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Cong Ye
- Department of Gynecology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| | - Junbao Liu
- Department of Gynecology, China-Japan Union Hospital of Jilin University, Changchun, Jilin, China
| |
Collapse
|
25
|
Choi YJ, Kim Y, Hwang S. Role of Neutrophils in the Development of Steatotic Liver Disease. Semin Liver Dis 2024; 44:300-318. [PMID: 39117322 DOI: 10.1055/s-0044-1789207] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/10/2024]
Abstract
This review explores the biological aspects of neutrophils, their contributions to the development of steatotic liver disease, and their potential as therapeutic targets for the disease. Although alcohol-associated and metabolic dysfunction-associated liver diseases originate from distinct etiological factors, the two diseases frequently share excessive lipid accumulation as a common contributor to their pathogenesis, thereby classifying them as types of steatotic liver disease. Dysregulated lipid deposition in the liver induces hepatic injury, triggering the activation of the innate immunity, partially through neutrophil recruitment. Traditionally recognized for their role in microbial clearance, neutrophils have recently garnered attention for their involvement in sterile inflammation, a pivotal component of steatotic liver disease pathogenesis. In conclusion, technological innovations, including single-cell RNA sequencing, have gradually disclosed the existence of various neutrophil subsets; however, how the distinct subsets of neutrophil population contribute differentially to the development of steatotic liver disease remains unclear.
Collapse
Affiliation(s)
- You-Jin Choi
- College of Pharmacy, Daegu Catholic University, Gyeongsan, Republic of Korea
| | - Yeonsoo Kim
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| | - Seonghwan Hwang
- College of Pharmacy and Research Institute for Drug Development, Pusan National University, Busan, Republic of Korea
| |
Collapse
|
26
|
Han HS, Hwang S, Choi SY, Hitayezu E, Humphrey MA, Enkhbayar A, Song D, Kim M, Park J, Park Y, Park J, Cha KH, Choi KY. Roseburia intestinalis-derived extracellular vesicles ameliorate colitis by modulating intestinal barrier, microbiome, and inflammatory responses. J Extracell Vesicles 2024; 13:e12487. [PMID: 39166405 PMCID: PMC11336657 DOI: 10.1002/jev2.12487] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2024] [Revised: 05/16/2024] [Accepted: 06/29/2024] [Indexed: 08/22/2024] Open
Abstract
Inflammatory bowel disease (IBD) is a chronic disorder characterized by recurrent gastrointestinal inflammation, lacking a precise aetiology and definitive cure. The gut microbiome is vital in preventing and treating IBD due to its various physiological functions. In the interplay between the gut microbiome and human health, extracellular vesicles secreted by gut bacteria (BEVs) are key mediators. Herein, we explore the role of Roseburia intestinalis (R)-derived EVs (R-EVs) as potent anti-inflammatory mediators in treating dextran sulfate sodium-induced colitis. R was selected as an optimal BEV producer for IBD treatment through ANCOM analysis. R-EVs with a 76 nm diameter were isolated from R using a tangential flow filtration system. Orally administered R-EVs effectively accumulated in inflamed colonic tissues and increased the abundance of Bifidobacterium on microbial changes, inhibiting colonic inflammation and prompting intestinal recovery. Due to the presence of Ile-Pro-Ile in the vesicular structure, R-EVs reduced the DPP4 activity in inflamed colonic tissue and increased the active GLP-1, thereby downregulating the NFκB and STAT3 via the PI3K pathway. Our results shed light on the impact of BEVs on intestinal recovery and gut microbiome alteration in treating IBD.
Collapse
Affiliation(s)
- Hwa Seung Han
- Department of Marine Bio‐Food ScienceGangneung‐Wonju National UniversityGangneungRepublic of Korea
- Natural Product Informatics Research CenterKorea Institute of Science and Technology (KIST)GangneungRepublic of Korea
| | - Soonjae Hwang
- Natural Product Informatics Research CenterKorea Institute of Science and Technology (KIST)GangneungRepublic of Korea
| | | | - Emmanuel Hitayezu
- Natural Product Informatics Research CenterKorea Institute of Science and Technology (KIST)GangneungRepublic of Korea
| | - Mabwi A. Humphrey
- Natural Product Informatics Research CenterKorea Institute of Science and Technology (KIST)GangneungRepublic of Korea
| | - Altai Enkhbayar
- Natural Product Informatics Research CenterKorea Institute of Science and Technology (KIST)GangneungRepublic of Korea
| | - Dae‐Geun Song
- Natural Product Informatics Research CenterKorea Institute of Science and Technology (KIST)GangneungRepublic of Korea
| | - Myungsuk Kim
- Natural Product Informatics Research CenterKorea Institute of Science and Technology (KIST)GangneungRepublic of Korea
| | | | - Young‐Tae Park
- Natural Product Research CenterKorea Institute of Science and Technology (KIST)GangneungRepublic of Korea
| | - Jin‐Soo Park
- Natural Product Informatics Research CenterKorea Institute of Science and Technology (KIST)GangneungRepublic of Korea
| | - Kwang Hyun Cha
- Natural Product Informatics Research CenterKorea Institute of Science and Technology (KIST)GangneungRepublic of Korea
| | - Ki Young Choi
- Department of Marine Bio‐Food ScienceGangneung‐Wonju National UniversityGangneungRepublic of Korea
- Natural Product Informatics Research CenterKorea Institute of Science and Technology (KIST)GangneungRepublic of Korea
- NVience Inc.SeoulRepublic of Korea
| |
Collapse
|
27
|
Liu L, Zhao Z, Liu H, Xia X, Ai C, Song S, Yan C. Haematococcus pluvialis polysaccharides improve microbiota-driven gut epithelial and vascular barrier and prevent alcoholic steatohepatitis development. Int J Biol Macromol 2024; 274:133014. [PMID: 38852729 DOI: 10.1016/j.ijbiomac.2024.133014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/22/2024] [Accepted: 06/06/2024] [Indexed: 06/11/2024]
Abstract
Algal polysaccharides possess many biological activities and health benefits, such as antioxidant, anti-tumor, anti-coagulant, and immunomodulatory potential. Gut microbiota has emerged as one of the major contributor in mediating the health benefits of algal polysaccharides. In this study we showed that Haematococcus pluvialis polysaccharides (HPP) decreased serum transaminase levels and hepatic triglyceride content, alleviated inflammation and oxidative stress in the liver of chronic and binge ethanol diet-fed mice. Furthermore, HPP reduced endotoxemia, improved gut microbiota dysbiosis, inhibited epithelial barrier disruption and gut vascular barrier (GVB) damage in ethanol diet-fed mice. Co-housing vehicle-fed mice with HPP-fed mice alleviated ethanol-induced liver damage and endotoxemia. Moreover, fecal microbiota transplantation from HPP-fed mice into antibiotic-induced microbiota-depleted recipients also alleviated ethanol-induced liver injury and improved gut epithelial and vascular barrier. Our study demonstrated that HPP ameliorated ethanol-induced gut epithelial and vascular barrier dysfunction through alteration of gut microbiota, therefore preventing alcoholic liver damage.
Collapse
Affiliation(s)
- Lingzhi Liu
- Liaoning Key Laboratory of Food Nutrition and Health, Collaborative Innovation Center of Seafood Deep Processing, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Zhikun Zhao
- Liaoning Key Laboratory of Food Nutrition and Health, Collaborative Innovation Center of Seafood Deep Processing, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Huanhuan Liu
- Liaoning Key Laboratory of Food Nutrition and Health, Collaborative Innovation Center of Seafood Deep Processing, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Xiaodong Xia
- Liaoning Key Laboratory of Food Nutrition and Health, Collaborative Innovation Center of Seafood Deep Processing, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Chunqing Ai
- Liaoning Key Laboratory of Food Nutrition and Health, Collaborative Innovation Center of Seafood Deep Processing, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Shuang Song
- Liaoning Key Laboratory of Food Nutrition and Health, Collaborative Innovation Center of Seafood Deep Processing, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Chunhong Yan
- Liaoning Key Laboratory of Food Nutrition and Health, Collaborative Innovation Center of Seafood Deep Processing, National Engineering Research Center of Seafood, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China.
| |
Collapse
|
28
|
Yu D, Fan Q, Yang J, Jin M, Shi L, Zhou B, Zhao L, Zhang J, Lin Z, Zhang T, Liu H. Gut Roseburia is a protective marker for peritoneal metastasis of gastric cancer. Cancer Med 2024; 13:e70037. [PMID: 39109683 PMCID: PMC11304227 DOI: 10.1002/cam4.70037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2024] [Revised: 07/02/2024] [Accepted: 07/10/2024] [Indexed: 08/10/2024] Open
Abstract
BACKGROUND Gastric cancer (GC), particularly for advanced stage of GC, commonly undergoes peritoneal metastasis (PM), which is the leading cause of GC-related death. However, there currently has no reliable biomarker to predict the onset of GCPM. It is well known that the imbalance of gut microbiota contributes to the development and metastasis of gastrointestinal tumors. Unfortunately, little is known about how the alternation in gut microbiota is associated with the onset of GCPM. METHODS Our current study analyzed structural characteristics and functional prediction of gut microbiota in GC patients with PM (PM group) and without PM (non-PM group). Fresh fecal samples were collected from a discovery cohort (PM = 38, non-PM = 54) and a validation cohort (PM = 15, non-PM = 21) of GC patients and their 16S ribosomal RNA (16s rRNA) gene amplicons were sequenced, followed by bioinformatics. RESULTS The results indicated an increase in the biodiversity of gut microbiota in the non-PM group of the discovery cohort, compared with the PM group. Moreover, LEfSe analysis found 31 significantly different microorganisms, of which the Roseburia ranked the fifth in the random forest (RF) model. The characteristics of intestinal microbiota in GCPM patients were changed, and the abundance of Roseburia in gut microbiota from the GCPM patients was reduced and receiver operating characteristic (ROC) analysis revealed that the reduced abundance of gut Roseburia effectively predicted the onset of GCPM. CONCLUSION This signature was also observed in the validation cohort. Therefore, Roseburia is a protective microbial marker and the reduced abundance of Roseburia in gut microbiota may help early diagnosis of GCPM.
Collapse
Affiliation(s)
- Dandan Yu
- Cancer Center, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanPeople's Republic of China
- Hubei Key Laboratory of Precision Radiation OncologyWuhanPeople's Republic of China
| | - Qilin Fan
- Department of Gastroenterology, General Hospital of Central Theater CommandWuhanPeople's Republic of China
| | - Jinru Yang
- Cancer Center, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanPeople's Republic of China
- Hubei Key Laboratory of Precision Radiation OncologyWuhanPeople's Republic of China
| | - Min Jin
- Cancer Center, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanPeople's Republic of China
- Hubei Key Laboratory of Precision Radiation OncologyWuhanPeople's Republic of China
| | - Linli Shi
- Cancer Center, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanPeople's Republic of China
- Hubei Key Laboratory of Precision Radiation OncologyWuhanPeople's Republic of China
| | - Bin Zhou
- Cancer Center, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanPeople's Republic of China
- Hubei Key Laboratory of Precision Radiation OncologyWuhanPeople's Republic of China
| | - Lei Zhao
- Cancer Center, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanPeople's Republic of China
- Hubei Key Laboratory of Precision Radiation OncologyWuhanPeople's Republic of China
| | - Jieying Zhang
- Cancer Center, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanPeople's Republic of China
- Hubei Key Laboratory of Precision Radiation OncologyWuhanPeople's Republic of China
| | - Zhenyu Lin
- Cancer Center, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanPeople's Republic of China
- Hubei Key Laboratory of Precision Radiation OncologyWuhanPeople's Republic of China
| | - Tao Zhang
- Cancer Center, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanPeople's Republic of China
- Hubei Key Laboratory of Precision Radiation OncologyWuhanPeople's Republic of China
| | - Hongli Liu
- Cancer Center, Union Hospital, Tongji Medical CollegeHuazhong University of Science and TechnologyWuhanPeople's Republic of China
- Institute of Radiation Oncology, Union Hospital, Tongji Medical College, Huazhong University of Science and TechnologyWuhanPeople's Republic of China
- Hubei Key Laboratory of Precision Radiation OncologyWuhanPeople's Republic of China
| |
Collapse
|
29
|
Sun L, Yue Z, Wang L. Research on the function of epigenetic regulation in the inflammation of non-alcoholic fatty liver disease. LIFE MEDICINE 2024; 3:lnae030. [PMID: 39872862 PMCID: PMC11749620 DOI: 10.1093/lifemedi/lnae030] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 04/01/2024] [Accepted: 08/30/2024] [Indexed: 01/30/2025]
Abstract
Nonalcoholic fatty liver disease (NAFLD) is the most prevalent chronic liver condition, characterized by a spectrum that progresses from simple hepatic steatosis to nonalcoholic steatohepatitis, which may eventually lead to cirrhosis and hepatocellular carcinoma. The precise pathogenic mechanisms underlying NAFLD and its related metabolic disturbances remain elusive. Epigenetic modifications, which entail stable transcriptional changes without altering the DNA sequence, are increasingly recognized as pivotal. The principal forms of epigenetic modifications include DNA methylation, histone modifications, chromatin remodeling, and noncoding RNAs. These alterations participate in the regulation of hepatic lipid metabolism, insulin resistance, mitochondrial injury, oxidative stress response, and release of inflammatory cytokines, all of which are associated with the onset and progression of NAFLD. This review discussed recent advances in understanding the potential epigenetic regulation of inflammation in NAFLD. Unraveling these epigenetic mechanisms may facilitate the identification of early diagnostic biomarkers and the development of targeted therapeutic strategies for NAFLD.
Collapse
Affiliation(s)
- Lin Sun
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Changle West Road, Xincheng District, Xi’an, Shaanxi 710032, China
| | - Zhensheng Yue
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Changle West Road, Xincheng District, Xi’an, Shaanxi 710032, China
- Department of Ophthalmology, Xi-Jing Hospital, Fourth Military Medical University, Changle West Road, Xincheng District, Xi’an, Shaanxi 710032, China
| | - Lin Wang
- Department of Hepatobiliary Surgery, Xi-Jing Hospital, Fourth Military Medical University, Changle West Road, Xincheng District, Xi’an, Shaanxi 710032, China
| |
Collapse
|
30
|
Cheng Z, Yang L, Chu H. The role of gut microbiota, exosomes, and their interaction in the pathogenesis of ALD. J Adv Res 2024:S2090-1232(24)00268-6. [PMID: 38969094 DOI: 10.1016/j.jare.2024.07.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Revised: 06/30/2024] [Accepted: 07/01/2024] [Indexed: 07/07/2024] Open
Abstract
BACKGROUND The liver disorders caused by alcohol abuse are termed alcoholic-related liver disease (ALD), including alcoholic steatosis, alcoholic steatohepatitis, alcoholic hepatitis, and alcoholic cirrhosis, posing a significant threat to human health. Currently, ALD pathogenesis has not been completely clarified, which is likely to be related to the direct damage caused by alcohol and its metabolic products, oxidative stress, gut dysbiosis, and exosomes. AIMS The existing studies suggest that both the gut microbiota and exosomes contribute to the development of ALD. Moreover, there exists an interaction between the gut microbiota and exosomes. We discuss whether this interaction plays a role in the pathogenesis of ALD and whether it can be a potential therapeutic target for ALD treatment. KEY SCIENTIFIC CONCEPTS OF REVIEW Chronic alcohol intake alters the diversity and composition of gut microbiota, which greatly contributes to ALD's progression. Some approaches targeting the gut microbiota, including probiotics, fecal microbiota transplantation, and phage therapy, have been confirmed to effectively ameliorate ALD in many animal experiments and/or several clinical trials. In ALD, the levels of exosomes and the expression profile of microRNA have also changed, which affects the pathogenesis of ALD. Moreover, there is an interplay between exosomes and the gut microbiota, which also putatively acts as a pathogenic factor of ALD.
Collapse
Affiliation(s)
- Zilu Cheng
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China
| | - Ling Yang
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China.
| | - Huikuan Chu
- Division of Gastroenterology, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, 1277 Jiefang Avenue, Wuhan, Hubei Province 430022, China.
| |
Collapse
|
31
|
Song Y, Zhang J, Li Y, Wang Y, Wan Y. Potential Protective Effect of Selenium-Enriched Lactobacillus plantarum on Cadmium-Induced Liver Injury in Mice. J Microbiol Biotechnol 2024; 34:1328-1339. [PMID: 38754999 PMCID: PMC11239402 DOI: 10.4014/jmb.2312.12051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/25/2024] [Accepted: 04/05/2024] [Indexed: 05/18/2024]
Abstract
Cadmium (Cd) is a prevalent environmental contaminant that poses a potential hazard to the health of both humans and animals. In this study, biosynthesized selenium-enriched Lactobacillus plantarum and selenium nanoparticles (SeNPs) were developed and evaluated for their protective effects against Cd-induced hepatic injury in mice through oral administration for 4 weeks. Cadmium exposure resulted in severe impairment of liver function, as evidenced by increased levels of serum markers of liver injury and, oxidative stress and significant damage to liver tissue, and a notable decrease in the diversity of the intestinal microbiota. Oral administration of Se-enriched L. plantarum (LS) reduced cadmium accumulation in the liver by 49.5% and, restored other cadmium-induced damage markers to normal levels. A comparison of the effects with those of L. plantarum (L) and SeNPs isolated from LS revealed that LS could more effectively alleviate hepatic oxidative stress and reduce the intrahepatic inflammatory responses of the liver, further protecting against cadmium-induced liver injury. These findings suggest that the development of LS may be effective at protecting the liver and intestinal tract from cadmium-induced damage.
Collapse
Affiliation(s)
- Yanyan Song
- College of Biochemical Engineering, Beijing Union University, Beijing 100023, P.R. China
| | - Jing Zhang
- College of Biochemical Engineering, Beijing Union University, Beijing 100023, P.R. China
| | - Yidan Li
- College of Biochemical Engineering, Beijing Union University, Beijing 100023, P.R. China
| | - Yuxuan Wang
- College of Biochemical Engineering, Beijing Union University, Beijing 100023, P.R. China
| | - Yingxin Wan
- College of Biochemical Engineering, Beijing Union University, Beijing 100023, P.R. China
| |
Collapse
|
32
|
Celano G, Calabrese FM, Riezzo G, D’Attoma B, Ignazzi A, Di Chito M, Sila A, De Nucci S, Rinaldi R, Linsalata M, Apa CA, Mancini L, De Angelis M, Giannelli G, De Pergola G, Russo F. A Multi-Omics Approach to Disclose Metabolic Pathways Impacting Intestinal Permeability in Obese Patients Undergoing Very Low Calorie Ketogenic Diet. Nutrients 2024; 16:2079. [PMID: 38999827 PMCID: PMC11243313 DOI: 10.3390/nu16132079] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 06/25/2024] [Accepted: 06/26/2024] [Indexed: 07/14/2024] Open
Abstract
A very low calorie ketogenic diet (VLCKD) impacts host metabolism in people marked by an excess of visceral adiposity, and it affects the microbiota composition in terms of taxa presence and relative abundances. As a matter of fact, there is little available literature dealing with microbiota differences in obese patients marked by altered intestinal permeability. With the aim of inspecting consortium members and their related metabolic pathways, we inspected the microbial community profile, together with the set of volatile organic compounds (VOCs) from untargeted fecal and urine metabolomics, in a cohort made of obese patients, stratified based on both normal and altered intestinal permeability, before and after VLCKD administration. Based on the taxa relative abundances, we predicted microbiota-derived metabolic pathways whose variations were explained in light of our cohort symptom picture. A totally different number of statistically significant pathways marked samples with altered permeability, reflecting an important shift in microbiota taxa. A combined analysis of taxa, metabolic pathways, and metabolomic compounds delineates a set of markers that is useful in describing obesity dysfunctions and comorbidities.
Collapse
Affiliation(s)
- Giuseppe Celano
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, 70126 Bari, Italy; (G.C.); (C.A.A.); (L.M.); (M.D.A.)
| | - Francesco Maria Calabrese
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, 70126 Bari, Italy; (G.C.); (C.A.A.); (L.M.); (M.D.A.)
| | - Giuseppe Riezzo
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy; (G.R.); (B.D.); (A.I.); (M.L.)
| | - Benedetta D’Attoma
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy; (G.R.); (B.D.); (A.I.); (M.L.)
| | - Antonia Ignazzi
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy; (G.R.); (B.D.); (A.I.); (M.L.)
| | - Martina Di Chito
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy; (M.D.C.); (A.S.); (S.D.N.); (R.R.); (G.D.P.)
| | - Annamaria Sila
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy; (M.D.C.); (A.S.); (S.D.N.); (R.R.); (G.D.P.)
| | - Sara De Nucci
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy; (M.D.C.); (A.S.); (S.D.N.); (R.R.); (G.D.P.)
| | - Roberta Rinaldi
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy; (M.D.C.); (A.S.); (S.D.N.); (R.R.); (G.D.P.)
| | - Michele Linsalata
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy; (G.R.); (B.D.); (A.I.); (M.L.)
| | - Carmen Aurora Apa
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, 70126 Bari, Italy; (G.C.); (C.A.A.); (L.M.); (M.D.A.)
| | - Leonardo Mancini
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, 70126 Bari, Italy; (G.C.); (C.A.A.); (L.M.); (M.D.A.)
| | - Maria De Angelis
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, 70126 Bari, Italy; (G.C.); (C.A.A.); (L.M.); (M.D.A.)
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy;
| | - Giovanni De Pergola
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy; (M.D.C.); (A.S.); (S.D.N.); (R.R.); (G.D.P.)
| | - Francesco Russo
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, 70013 Castellana Grotte, Italy; (G.R.); (B.D.); (A.I.); (M.L.)
| |
Collapse
|
33
|
Zhang J, Zhang C, Yu L, Tian F, Chen W, Zhai Q. Analysis of the key genes of Lactobacillus reuteri strains involved in the protection against alcohol-induced intestinal barrier damage. Food Funct 2024; 15:6629-6641. [PMID: 38812427 DOI: 10.1039/d4fo01796j] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/31/2024]
Abstract
Gastrointestinal inflammation and intestinal barrier function have important effects on human health. Alcohol, an important foodborne hazard factor, damages the intestinal barrier, increasing the risk of disease. Lactobacillus reuteri strains have been reported to reduce gastrointestinal inflammation and strengthen the intestinal barrier. In this study, we selected three anti-inflammatory L. reuteri strains to evaluate their role in the protection of the intestinal barrier and their immunomodulatory activity in a mouse model of gradient alcohol intake. Among the three strains tested (FSCDJY33M3, FGSZY33L6, and FCQHCL8L6), L. reuteri FSCDJY33M3 was found to protect the intestinal barrier most effectively, possibly due to its ability to reduce the expression of interleukin (IL)-1β, IL-6, and tumor necrosis factor-alpha (TNF-α) and increase the expression of tight junction proteins (occludin, claudin-3). Genomic analysis suggested that the protective effects of L. reuteri FSCDJY33M3 may be related to functional genes and glycoside hydrolases associated with energy production and conversion, amino acid transport and metabolism, carbohydrate transport and metabolism, and DNA replication, recombination, and repair. These genes include COG2856, COG1804, COG2071, and COG1061, which encode adenine deaminase, acyl-CoA transferases, glutamine amidotransferase, RNA helicase, and glycoside hydrolases, including GH13_20, GH53, and GH70. Our results identified functional genes that may be related to protection against alcohol-induced intestinal barrier damage, which might be useful for screening lactic acid bacterial strains that can protect the intestinal barrier.
Collapse
Affiliation(s)
- Jiayi Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Chengcheng Zhang
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Leilei Yu
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Fengwei Tian
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Qixiao Zhai
- State Key Laboratory of Food Science and Resources, Jiangnan University, Wuxi 214122, China.
- School of Food Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|
34
|
Zhang Y, Bai B, Huang K, Li S, Cao H, Guan X. Bound Polyphenols of Oat Bran Released by Gut Microbiota Mitigate High Fat Diet-Induced Oxidative Stress and Strengthen the Gut Barrier via the Colonic ROS/Akt/Nrf2 Pathway. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2024; 72:13099-13110. [PMID: 38807079 DOI: 10.1021/acs.jafc.4c01666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/30/2024]
Abstract
Whole-grain foods are rich in bound polyphenols (BPs) whose health benefits were largely underestimated compared with free polyphenols. We first found that DFBP (dietary fiber with BPs from oat bran) exhibited stronger colonic antioxidant activities than DF. 16S rRNA sequencing showed that DFBP selectively changed gut microbial composition, which reciprocally released BPs from DFBP. Released polyphenols from DFBP reduced excessive colonic ROS and exhibited colonic antioxidant activities via the ROS/Akt/Nrf2 pathway revealed by transcriptome and western blot analysis. Colonic antioxidant activities of DFBP mediated by gut microbiota were next proven by treating mice with broad-spectrum antibiotics. Next, Clostridium butyricum, as a distinguished bacterium after DFBP intervention, improved colonic antioxidant capacities synergistically with DFBP in HFD-fed mice. This was explained by the upregulated mRNA expression of esterase, and cellulase of Clostridium butyricum participated in releasing BPs. Our results would provide a solid basis for explaining the health benefits of whole grains.
Collapse
Affiliation(s)
- Yu Zhang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, Shanghai 200093, China
| | - Bing Bai
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
| | - Kai Huang
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, Shanghai 200093, China
| | - Sen Li
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, Shanghai 200093, China
| | - Hongwei Cao
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, Shanghai 200093, China
| | - Xiao Guan
- School of Health Science and Engineering, University of Shanghai for Science and Technology, Shanghai 200093, China
- National Grain Industry (Urban Grain and Oil Security) Technology Innovation Center, Shanghai 200093, China
| |
Collapse
|
35
|
Huo Z, Li J, Li X, Xiao H, Lin Y, Ma Y, Li J, Yang H, Zhang C. Functional fractions of Astragalus polysaccharides as a potential prebiotic to alleviate ulcerative colitis. Int J Biol Macromol 2024; 271:132580. [PMID: 38788871 DOI: 10.1016/j.ijbiomac.2024.132580] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2024] [Revised: 05/02/2024] [Accepted: 05/20/2024] [Indexed: 05/26/2024]
Abstract
Ulcerative colitis (UC) is a chronic inflammatory disease of the intestine that is significantly influenced by an imbalance in the gut microbiota. Astragalus membranaceus, particularly its polysaccharide components, has shown therapeutic potential for the treatment of UC, although the specific active constituents and their mechanistic pathways remain to be fully elucidated. In this study, we investigated two molecular weight fractions of Astragalus polysaccharides (APS), APS1 (Mw < 10 kDa) and APS2 (10 kDa < Mw < 50 kDa), isolated by ultrafiltration, focusing on their prebiotic effects, effects on UC, and the underlying mechanism. Our results showed that both APS1 and APS2 exhibit prebiotic properties, with APS1 significantly outperforming APS2 in ameliorating UC symptoms. APS1 significantly attenuated weight loss and UC manifestations, reduced colonic pathology, and improved intestinal mucosal barrier integrity. In addition, APS1 significantly reduced the levels of inflammatory cytokines in the serum and colonic tissue, and downregulated colonic chemokines. Furthermore, APS1 ameliorated dextran sulfate sodium salt (DSS)-induced intestinal dysbiosis by promoting the growth of beneficial microbes and inhibiting the proliferation of potential pathogens, leading to a significant increase in short-chain fatty acids. In conclusion, this study highlights the potential of APS1 as a novel prebiotic for the prevention and treatment of UC.
Collapse
Affiliation(s)
- Zeqi Huo
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China; Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou 730000, China; Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou 730000, China
| | - Junxiang Li
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China; Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou 730000, China; Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou 730000, China
| | - Xiaofeng Li
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China; Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou 730000, China; Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou 730000, China
| | - Han Xiao
- Lanzhou University Second Hospital, Lanzhou, Gansu 730030, China
| | - Yang Lin
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China; Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou 730000, China; Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou 730000, China
| | - Yuchan Ma
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Jiaru Li
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Hui Yang
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Chunjiang Zhang
- School of Life Sciences, Lanzhou University, Lanzhou 730000, China; Key Laboratory of Cell Activities and Stress Adaptations, Ministry of Education, Lanzhou University, Lanzhou 730000, China; Gansu Key Laboratory of Biomonitoring and Bioremediation for Environmental Pollution, Lanzhou University, Lanzhou 730000, China.
| |
Collapse
|
36
|
Schöler D, Schnabl B. The role of the microbiome in liver disease. Curr Opin Gastroenterol 2024; 40:134-142. [PMID: 38362864 PMCID: PMC10990783 DOI: 10.1097/mog.0000000000001013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/17/2024]
Abstract
PURPOSE OF REVIEW The intestinal microbiome and the gut-liver axis play a major role in health and disease. The human gut harbors trillions of microbes and a disruption of the gut homeostasis can contribute to liver disease. In this review, the progress in the field within the last 3 years is summarized, focusing on metabolic dysfunction-associated steatotic liver disease (MASLD), alcohol-associated liver disease (ALD), autoimmune liver disease (AILD), and hepatocellular carcinoma (HCC). RECENT FINDINGS Changes in the fecal virome and fungal mycobiome have been described in patients with various liver diseases. Several microbial derived metabolites including endogenous ethanol produced by bacteria, have been mechanistically linked to liver disease such as MASLD. Virulence factors encoded by gut bacteria contribute to ALD, AILD and HCC. Novel therapeutic approaches focused on the microbiome including phages, pre- and postbiotics have been successfully used in preclinical models. Fecal microbiota transplantation has been effective in attenuating liver disease. Probiotics are safe in patients with alcohol-associated hepatitis and improve liver disease and alcohol addiction. SUMMARY The gut-liver axis plays a key role in the pathophysiology of liver diseases. Understanding the microbiota in liver disease can help to develop precise microbiota centered therapies.
Collapse
Affiliation(s)
- David Schöler
- Department of Medicine, University of California, San Diego
| | - Bernd Schnabl
- Department of Medicine, University of California, San Diego
- Department of Medicine, VA San Diego Healthcare System, San Diego, California, USA
| |
Collapse
|
37
|
Sun X, Zhou C, Ju M, Feng W, Guo Z, Qi C, Yang K, Xiao R. Roseburia intestinalis Supplementation Could Reverse the Learning and Memory Impairment and m6A Methylation Modification Decrease Caused by 27-Hydroxycholesterol in Mice. Nutrients 2024; 16:1288. [PMID: 38732535 PMCID: PMC11085097 DOI: 10.3390/nu16091288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2024] [Revised: 04/05/2024] [Accepted: 04/09/2024] [Indexed: 05/13/2024] Open
Abstract
The abnormality in N6-methyladenosine (m6A) methylation is involved in the course of Alzheimer's disease (AD), while the intervention of 27-Hydroxycholesterol (27-OHC) can affect the m6A methylation modification in the brain cortex. Disordered gut microbiota is a key link in 27-OHC leading to cognitive impairment, and further studies have found that the abundance of Roseburia intestinalis in the gut is significantly reduced under the intervention of 27-OHC. This study aims to investigate the association of 27-OHC, Roseburia intestinalis in the gut, and brain m6A modification in the learning and memory ability injury. In this study, 9-month-old male C57BL/6J mice were treated with antibiotic cocktails for 6 weeks to sweep the intestinal flora, followed by 27-OHC or normal saline subcutaneous injection, and then Roseburia intestinalis or normal saline gavage were applied to the mouse. The 27-OHC level in the brain, the gut barrier function, the m6A modification in the brain, and the memory ability were measured. From the results, we observed that 27-OHC impairs the gut barrier function, causing a disturbance in the expression of m6A methylation-related enzymes and reducing the m6A methylation modification level in the brain cortex, and finally leads to learning and memory impairment. However, Roseburia intestinalis supplementation could reverse the negative effects mentioned above. This study suggests that 27-OHC-induced learning and memory impairment might be linked to brain m6A methylation modification disturbance, while Roseburia intestinalis, as a probiotic with great potential, could reverse the damage caused by 27-OHC. This research could help reveal the mechanism of 27-OHC-induced neural damage and provide important scientific evidence for the future use of Roseburia intestinalis in neuroprotection.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Rong Xiao
- Beijing Key Laboratory of Environmental Toxicology, School of Public Health, Capital Medical University, Beijing 100069, China; (X.S.); (C.Z.); (M.J.); (W.F.); (Z.G.); (C.Q.); (K.Y.)
| |
Collapse
|
38
|
Al-Fakhrany OM, Elekhnawy E. Next-generation probiotics: the upcoming biotherapeutics. Mol Biol Rep 2024; 51:505. [PMID: 38619680 PMCID: PMC11018693 DOI: 10.1007/s11033-024-09398-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2023] [Accepted: 02/28/2024] [Indexed: 04/16/2024]
Abstract
Recent and continuing advances in gut microbiome research have pointed out the role of the gut microbiota as an unexplored source of potentially beneficial probiotic microbes. Along the lines of these advances, both public awareness and acceptance of probiotics are increasing. That's why; academic and industrial research is dedicated to identifying and investigating new microbial strains for the development of next-generation probiotics (NGPs). At this time, there is a growing interest in NGPs as biotherapeutics that alter the gut microbiome and affect various diseases development. In this work, we have focused on some emergent and promising NGPs, specifically Eubacterium hallii, Faecalibacterium prausnitzii, Roseburia spp., Akkermansia muciniphila, and Bacteroides fragilis, as their presence in the gut can have an impact on the development of various diseases. Emerging studies point out the beneficial roles of these NGPs and open up novel promising therapeutic options. Interestingly, these NGPs were found to enhance gastrointestinal immunity, enhance immunotherapy efficacy in cancer patients, retain the intestinal barrier integrity, generate valuable metabolites, especially short-chain fatty acids, and decrease complications of chemotherapy and radiotherapy. Although many of these NGPs are considered promising for the prevention and treatment of several chronic diseases, research on humans is still lacking. Therefore, approval of these microbes from regulatory agencies is rare. Besides, some issues limit their wide use in the market, such as suitable methods for the culture and storage of these oxygen-sensitive microbes. The present review goes over the main points related to NGPs and gives a viewpoint on the key issues that still hinder their wide application. Furthermore, we have focused on the advancement in NGPs and human healthiness investigations by clarifying the limitations of traditional probiotic microorganisms, discussing the characteristics of emerging NGPs and defining their role in the management of certain ailments. Future research should emphasize the isolation, mechanisms of action of these probiotics, safety, and clinical efficacy in humans.
Collapse
Affiliation(s)
- Omnia Momtaz Al-Fakhrany
- Pharmaceutical Microbiology Department, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt.
| | - Engy Elekhnawy
- Pharmaceutical Microbiology Department, Faculty of Pharmacy, Tanta University, Tanta, 31527, Egypt.
| |
Collapse
|
39
|
Ganesan R, Gupta H, Jeong JJ, Sharma SP, Won SM, Oh KK, Yoon SJ, Han SH, Yang YJ, Baik GH, Bang CS, Kim DJ, Suk KT. Characteristics of microbiome-derived metabolomics according to the progression of alcoholic liver disease. Hepatol Int 2024; 18:486-499. [PMID: 37000389 DOI: 10.1007/s12072-023-10518-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2022] [Accepted: 03/07/2023] [Indexed: 04/01/2023]
Abstract
BACKGROUND AND AIM The prevalence and severity of alcoholic liver disease (ALD) are increasing. The incidence of alcohol-related cirrhosis has risen up to 2.5%. This study aimed to identify novel metabolite mechanisms involved in the development of ALD in patients. The use of gut microbiome-derived metabolites is increasing in targeted therapies. Identifying metabolic compounds is challenging due to the complex patterns that have long-term effects on ALD. We investigated the specific metabolite signatures in ALD patients. METHODS This study included 247 patients (heathy control, HC: n = 62, alcoholic fatty liver, AFL; n = 25, alcoholic hepatitis, AH; n = 80, and alcoholic cirrhosis, AC, n = 80) identified, and stool samples were collected. 16S rRNA sequencing and metabolomics were performed with MiSeq sequencer and liquid chromatography coupled to time-of-flight-mass spectrometry (LC-TOF-MS), respectively. The untargeted metabolites in AFL, AH, and AC samples were evaluated by multivariate statistical analysis and metabolic pathotypic expression. Metabolic network classifiers were used to predict the pathway expression of the AFL, AH, and AC stages. RESULTS The relative abundance of Proteobacteria was increased and the abundance of Bacteroides was decreased in ALD samples (p = 0.001) compared with that in HC samples. Fusobacteria levels were higher in AH samples (p = 0.0001) than in HC samples. Untargeted metabolomics was applied to quantitatively screen 103 metabolites from each stool sample. Indole-3-propionic acid levels are significantly lower in AH and AC (vs. HC, p = 0.001). Indole-3-lactic acid (ILA: p = 0.04) levels were increased in AC samples. AC group showed an increase in indole-3-lactic acid (vs. HC, p = 0.040) level. Compared with that in HC samples, the levels of short-chain fatty acids (SCFAs: acetic acid, butyric acid, propionic acid, iso-butyric acid, and iso-valeric acid) and bile acids (lithocholic acids) were significantly decreased in AC. The pathways of linoleic acid metabolism, indole compounds, histidine metabolism, fatty acid degradation, and glutamate metabolism were closely associated with ALD metabolism. CONCLUSIONS This study identified that microbial metabolic dysbiosis is associated with ALD-related metabolic dysfunction. The SCFAs, bile acids, and indole compounds were depleted during ALD progression. CLINICAL TRIAL Clinicaltrials.gov, number NCT04339725.
Collapse
Affiliation(s)
- Raja Ganesan
- Institute for Liver and Digestive Diseases, Hallym University College of Medicine, Chuncheon, 24253, Republic of Korea
| | - Haripriya Gupta
- Institute for Liver and Digestive Diseases, Hallym University College of Medicine, Chuncheon, 24253, Republic of Korea
| | - Jin-Ju Jeong
- Institute for Liver and Digestive Diseases, Hallym University College of Medicine, Chuncheon, 24253, Republic of Korea
| | - Satya Priya Sharma
- Institute for Liver and Digestive Diseases, Hallym University College of Medicine, Chuncheon, 24253, Republic of Korea
| | - Sung-Min Won
- Institute for Liver and Digestive Diseases, Hallym University College of Medicine, Chuncheon, 24253, Republic of Korea
| | - Ki-Kwang Oh
- Institute for Liver and Digestive Diseases, Hallym University College of Medicine, Chuncheon, 24253, Republic of Korea
| | - Sang Jun Yoon
- Institute for Liver and Digestive Diseases, Hallym University College of Medicine, Chuncheon, 24253, Republic of Korea
| | - Sang Hak Han
- Department of Pathology, Hallym University College of Medicine, Chuncheon, Republic of Korea
| | - Young Joo Yang
- Institute for Liver and Digestive Diseases, Hallym University College of Medicine, Chuncheon, 24253, Republic of Korea
| | - Gwang Ho Baik
- Institute for Liver and Digestive Diseases, Hallym University College of Medicine, Chuncheon, 24253, Republic of Korea
| | - Chang Seok Bang
- Institute for Liver and Digestive Diseases, Hallym University College of Medicine, Chuncheon, 24253, Republic of Korea
| | - Dong Joon Kim
- Institute for Liver and Digestive Diseases, Hallym University College of Medicine, Chuncheon, 24253, Republic of Korea
| | - Ki Tae Suk
- Institute for Liver and Digestive Diseases, Hallym University College of Medicine, Chuncheon, 24253, Republic of Korea.
| |
Collapse
|
40
|
Dong J, Wang B, Xiao Y, Liu J, Wang Q, Xiao H, Jin Y, Liu Z, Chen Z, Li Y, Fan S, Li Y, Cui M. Roseburia intestinalis sensitizes colorectal cancer to radiotherapy through the butyrate/OR51E1/RALB axis. Cell Rep 2024; 43:113846. [PMID: 38412097 DOI: 10.1016/j.celrep.2024.113846] [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: 05/22/2023] [Revised: 10/30/2023] [Accepted: 02/06/2024] [Indexed: 02/29/2024] Open
Abstract
The radioresistant signature of colorectal cancer (CRC) hampers the clinical utility of radiotherapy. Here, we find that fecal microbiota transplantation (FMT) potentiates the tumoricidal effects of radiation and degrades the intertwined adverse events in azoxymethane (AOM)/dextran sodium sulfate (DSS)-induced CRC mice. FMT cumulates Roseburia intestinalis (R. intestinalis) in the gastrointestinal tract. Oral gavage of R. intestinalis assembles at the CRC site and synthetizes butyrate, sensitizing CRC to radiation and alleviating intestinal toxicity in primary and CRC hepatic metastasis mouse models. R. intestinalis-derived butyrate activates OR51E1, a G-protein-coupled receptor overexpressing in patients with rectal cancer, facilitating radiogenic autophagy in CRC cells. OR51E1 shows a positive correlation with RALB in clinical rectal cancer tissues and CRC mouse model. Blockage of OR51E1/RALB signaling restrains butyrate-elicited autophagy in irradiated CRC cells. Our findings highlight that the gut commensal bacteria R. intestinalis motivates radiation-induced autophagy to accelerate CRC cell death through the butyrate/OR51E1/RALB axis and provide a promising radiosensitizer for CRC in a pre-clinical setting.
Collapse
Affiliation(s)
- Jiali Dong
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Bin Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Yunong Xiao
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Jia Liu
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Qi Wang
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Huiwen Xiao
- Department of Microbiology, College of Life Sciences, Nankai University, Tianjin 300071, China
| | - Yuxiao Jin
- Department of Anesthesiology, Changshu No. 2 People's Hospital, Changshu, Jiangsu Province 215501, China
| | - Zhihong Liu
- Department of General Surgery, The Second Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province 215004, China
| | - Zhiyuan Chen
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Yiliang Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Saijun Fan
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China
| | - Yuan Li
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| | - Ming Cui
- Tianjin Key Laboratory of Radiation Medicine and Molecular Nuclear Medicine, Institute of Radiation Medicine, Chinese Academy of Medical Sciences and Peking Union Medical College, Tianjin 300192, China.
| |
Collapse
|
41
|
Liu Y, Zhu Q, Guo G, Xie Z, Li S, Lai C, Wu Y, Wang L, Zhong S. Causal associations of genetically predicted gut microbiota and blood metabolites with inflammatory states and risk of infections: a Mendelian randomization analysis. Front Microbiol 2024; 15:1342653. [PMID: 38585702 PMCID: PMC10995310 DOI: 10.3389/fmicb.2024.1342653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Accepted: 02/28/2024] [Indexed: 04/09/2024] Open
Abstract
Background Inflammation serves as a key pathologic mediator in the progression of infections and various diseases, involving significant alterations in the gut microbiome and metabolism. This study aims to probe into the potential causal relationships between gut microbial taxa and human blood metabolites with various serum inflammatory markers (CRP, SAA1, IL-6, TNF-α, WBC, and GlycA) and the risks of seven common infections (gastrointestinal infections, dysentery, pneumonia, bacterial pneumonia, bronchopneumonia and lung abscess, pneumococcal pneumonia, and urinary tract infections). Methods Two-sample Mendelian randomization (MR) analysis was performed using inverse variance weighted (IVW), maximum likelihood, MR-Egger, weighted median, and MR-PRESSO. Results After adding other MR models and sensitivity analyses, genus Roseburia was simultaneously associated adversely with CRP (Beta IVW = -0.040) and SAA1 (Beta IVW = -0.280), and family Bifidobacteriaceae was negatively associated with both CRP (Beta IVW = -0.034) and pneumonia risk (Beta IVW = -0.391). After correction by FDR, only glutaroyl carnitine remained significantly associated with elevated CRP levels (Beta IVW = 0.112). Additionally, threonine (Beta IVW = 0.200) and 1-heptadecanoylglycerophosphocholine (Beta IVW = -0.246) were found to be significantly associated with WBC levels. Three metabolites showed similar causal effects on different inflammatory markers or infectious phenotypes, stearidonate (18:4n3) was negatively related to SAA1 and urinary tract infections, and 5-oxoproline contributed to elevated IL-6 and SAA1 levels. In addition, 7-methylguanine showed a positive correlation with dysentery and bacterial pneumonia. Conclusion This study provides novel evidence confirming the causal effects of the gut microbiome and the plasma metabolite profile on inflammation and the risk of infection. These potential molecular alterations may aid in the development of new targets for the intervention and management of disorders associated with inflammation and infections.
Collapse
Affiliation(s)
- Yingjian Liu
- Department of Pharmacy, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
| | - Qian Zhu
- Department of Neurosurgery, Huizhou Third People's Hospital, Guangzhou Medical University, Huizhou, Guangdong, China
| | - Gongjie Guo
- Department of Pharmacy, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Zhipeng Xie
- Department of Pharmacy, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Senlin Li
- Department of Pharmacy, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Chengyang Lai
- Department of Pharmacy, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Yonglin Wu
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
| | - Liansheng Wang
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| | - Shilong Zhong
- Department of Pharmacy, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Southern Medical University, Guangzhou, Guangdong, China
- School of Medicine, South China University of Technology, Guangzhou, Guangdong, China
- Guangdong Cardiovascular Institute, Guangdong Provincial People's Hospital (Guangdong Academy of Medical Sciences), Southern Medical University, Guangzhou, China
| |
Collapse
|
42
|
Shen M, Zhao H, Han M, Su L, Cui X, Li D, Liu L, Wang C, Yang F. Alcohol-induced gut microbiome dysbiosis enhances the colonization of Klebsiella pneumoniae on the mouse intestinal tract. mSystems 2024; 9:e0005224. [PMID: 38345382 PMCID: PMC10949497 DOI: 10.1128/msystems.00052-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Accepted: 01/12/2024] [Indexed: 03/20/2024] Open
Abstract
Chronic alcohol consumption, an important risk factor for diseases and deaths, can cause intestinal microbiota dysbiosis and increase the infection of some opportunistic pathogens. However, the current studies on the effects of alcohol-induced intestinal microbiota dysbiosis on gut colonization of Klebsiella pneumoniae are still scarce. In the present study, we established a binge-on-chronic alcohol model in mice to identify the characteristics of alcohol-induced intestinal microbiome and metabolite dysbiosis using multi-omics and explored the effects and potential mechanisms of these dysbioses on the intestinal colonization of K. pneumoniae. The results show that chronic alcohol consumption alters the diversity and composition of gut microbiota (including bacteria and fungi), decreases the complexity of the interaction between intestinal bacteria and fungi, disturbs the gut metabolites, and promotes the colonization of K. pneumoniae on the gut of mice. The relevance analyses find that alcohol-induced gut microbiome dysbiosis has a strong correlation with the alteration of secondary bile acids. In vitro results suggest that the high concentration of lithocholic acid, a secondary bile acid, could significantly inhibit the proliferation of K. pneumoniae, and the adhesion of K. pneumoniae to Caco-2 cells. Our results indicate that alcohol-induced microbiome dysbiosis contributes to decreased levels of secondary bile acids, which was one of the main reasons affecting the colonization of K. pneumoniae in mice's intestines. Some secondary bile acids (e.g., lithocholic acid) might be a potential drug to prevent the colonization and spread of K. pneumoniae.IMPORTANCEAlcohol is one of the most commonly misused substances in our lives. However, long-term heavy drinking will increase the colonization of some opportunistic pathogens (e.g., Klebsiella pneumoniae) in the body. Here, we revealed that binge-on-chronic alcohol consumption disrupted the balance between gut bacteria and fungi, induced the gut microbiome and metabolites dysbiosis, and promoted the colonization of K. pneumoniae in the intestine of mice. In particular, alcohol-taking disrupted intestinal bile acid metabolism and reduced the lithocholic acid concentration. However, a high concentration of lithocholic acid can protect against intestinal colonization of K. pneumoniae by inhabiting the bacterial growth and adhesion to the host cell. Hence, regulating the balance of gut microbiota and intestinal bile acid metabolism may be a potential strategy for reducing the risk of K. pneumoniae infection and spread.
Collapse
Affiliation(s)
- Mengke Shen
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China
- Department of Pathogenic Biology and Immunology, Sanquan College of Xinxiang Medical University, Xinxiang, China
| | - Huajie Zhao
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China
| | - Meiqing Han
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China
| | - Lin Su
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China
| | - Xiaojian Cui
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China
| | - Duan Li
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China
| | - Liang Liu
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China
| | - Chuansheng Wang
- The Second Affiliated Hospital of Xinxiang Medical University, Henan Key Laboratory of Biological Psychiatry, Xinxiang Medical University, Xinxiang, China
| | - Fan Yang
- Department of Pathogenic Biology, School of Basic Medical Science, Xinxiang Medical University, Xinxiang, China
| |
Collapse
|
43
|
Wei B, Peng Z, Zheng W, Yang S, Wu M, Liu K, Xiao M, Huang T, Xie M, Xiong T. Probiotic-fermented tomato alleviates high-fat diet-induced obesity in mice: Insights from microbiome and metabolomics. Food Chem 2024; 436:137719. [PMID: 37839120 DOI: 10.1016/j.foodchem.2023.137719] [Citation(s) in RCA: 23] [Impact Index Per Article: 23.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2023] [Revised: 10/02/2023] [Accepted: 10/08/2023] [Indexed: 10/17/2023]
Abstract
Probiotic-fermented plant-based foods are associated with weight loss. Here, we hypothesized probiotic-fermented tomato (FT) as a functional food with potential to alleviate obesity, thus the obesity-alleviating effects and mechanisms of FT on high-fat diet-induced obese mice were explored via biochemical, gut microbiome, and serum metabolomics analysis. The results showed that FT performed better than unfermented tomato in reducing body weight gain and fat accumulation, improving dyslipidemia and glucose homeostasis, and relieving inflammation and adipocytokine dysregulation. Particularly, live probiotic-fermented tomato (LFT) was associated with improved diversity, composition, and structure of gut microbiota, suppressed obesity-related genera growth (e.g., Clostridium, Olsenella, and Mucispirillum), and promoted beneficial genera growth (e.g., Roseburia, Coprococcus, and Oscillospira), which were associated negatively with body weight, TC, TG, and TNF-α levels. Additionally, LFT was associated with positive changes in glycerophospholipids, sphingolipids, unsaturated fatty acids, and amino acids levels. Collectively, as a functional food, LFT possessed potential for obesity alleviation.
Collapse
Affiliation(s)
- Benliang Wei
- State Key Laboratory of Food Science and Resources, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China
| | - Zhen Peng
- State Key Laboratory of Food Science and Resources, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China
| | - Wendi Zheng
- State Key Laboratory of Food Science and Resources, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China
| | - Shiyu Yang
- State Key Laboratory of Food Science and Resources, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China
| | - Min Wu
- State Key Laboratory of Food Science and Resources, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China
| | - Kui Liu
- State Key Laboratory of Food Science and Resources, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China
| | - Muyan Xiao
- State Key Laboratory of Food Science and Resources, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China; International Institute of Food Innovation, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China
| | - Tao Huang
- State Key Laboratory of Food Science and Resources, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China; International Institute of Food Innovation, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China
| | - Mingyong Xie
- State Key Laboratory of Food Science and Resources, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China
| | - Tao Xiong
- State Key Laboratory of Food Science and Resources, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China; School of Food Science and Technology, Nanchang University, No. 235 Nanjing East Road, Nanchang, Jiangxi 330047, PR China.
| |
Collapse
|
44
|
Hua Z, Zhang X, Xing S, Li J, Liang D, Chen Y, Abd El-Aty A, Zhu BW, Liu D, Tan M. Design and preparation of multifunctional astaxanthin nanoparticles with good acid stability and hepatocyte-targeting ability for alcoholic liver injury alleviation. MATERIALS TODAY NANO 2024; 25:100436. [DOI: 10.1016/j.mtnano.2023.100436] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
45
|
Seo B, Jeon K, Kim WK, Jang YJ, Cha KH, Ko G. Strain-Specific Anti-Inflammatory Effects of Faecalibacterium prausnitzii Strain KBL1027 in Koreans. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10213-7. [PMID: 38411865 DOI: 10.1007/s12602-024-10213-7] [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] [Accepted: 01/04/2024] [Indexed: 02/28/2024]
Abstract
Faecalibacterium prausnitzii is one of the most dominant commensal bacteria in the human gut, and certain anti-inflammatory functions have been attributed to a single microbial anti-inflammatory molecule (MAM). Simultaneously, substantial diversity among F. prausnitzii strains is acknowledged, emphasizing the need for strain-level functional studies aimed at developing innovative probiotics. Here, two distinct F. prausnitzii strains, KBL1026 and KBL1027, were isolated from Korean donors, exhibiting notable differences in the relative abundance of F. prausnitzii. Both strains were identified as the core Faecalibacterium amplicon sequence variant (ASV) within the healthy Korean cohort, and their MAM sequences showed a high similarity of 98.6%. However, when a single strain was introduced to mice with dextran sulfate sodium (DSS)-induced colitis, KBL1027 showed the most significant ameliorative effects, including alleviation of colonic inflammation and restoration of gut microbial dysbiosis. Moreover, the supernatant from KBL1027 elevated the secretion of IL-10 cytokine more than that of KBL1026 in mouse bone marrow-derived macrophage (BMDM) cells, suggesting that the strain-specific, anti-inflammatory efficacy of KBL1027 might involve effector compounds other than MAM. Through analysis of the Faecalibacterium pan-genome and comparative genomics, strain-specific functions related to extracellular polysaccharide biosynthesis were identified in KBL1027, which could contribute to the observed morphological disparities. Collectively, our findings highlight the strain-specific, anti-inflammatory functions of F. prausnitzii, even within the same core ASV, emphasizing the influence of their human origin.
Collapse
Affiliation(s)
- Boram Seo
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
- Personalized Diet Research Group, Food Functionality Research Division, Korea Food Research Institute, Jeollabuk-do, Republic of Korea
| | - Kyungchan Jeon
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Woon-Ki Kim
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - You Jin Jang
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea
| | - Kwang Hyun Cha
- Natural Product Informatics Research Center, Korea Institute of Science and Technology (KIST), Gangneung, Republic of Korea
| | - GwangPyo Ko
- Department of Environmental Health Sciences, Graduate School of Public Health, Seoul National University, Seoul, Republic of Korea.
- N-Bio, Seoul National University, Seoul, Republic of Korea.
- Center for Human and Environmental Microbiome, Institute of Health and Environment, Seoul National University, Seoul, Republic of Korea.
- KoBioLabs Inc., Seoul, Republic of Korea.
| |
Collapse
|
46
|
Paiano L, Mastronardi M, Campisciano G, Rosso N, Casagranda B, Comar M, de Manzini N, Palmisano S. Liver Bacterial Colonization in Patients with Obesity and Gut Dysbiosis. Obes Surg 2024; 34:402-408. [PMID: 38102371 DOI: 10.1007/s11695-023-06989-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2023] [Revised: 11/27/2023] [Accepted: 12/06/2023] [Indexed: 12/17/2023]
Abstract
PURPOSE Recently, the link between gut microbiota, liver inflammation, and obesity has become an interesting focus of research. The aim of this study is to show the possible relation between gut microbiota dysbiosis in patients with obesity and the presence of bacterial genomes in their liver biopsies. MATERIALS AND METHODS A prospective study on patients undergoing bariatric surgery was carried out. Anthropometric and metabolic data, comorbidities, stool samples, and hepatic biopsies were collected and analyzed at the time of surgery. The V3-16S rRNA region was sequenced using the Ion Torrent new-generation sequencing platform. RESULTS In each of the 23 patients enrolled, the bacterial population was analyzed both in the stools and liver. In eight patients (34.7%), Prevotella (62.5%), Bacteroides (50%), Streptococcus (12.5%), and Dalister (12.5%) were found in both samples, simultaneously; in 15 cases, the liver was free from colonization. The statistically significant difference between groups was a Roseburia intestinalis reduction in fecal samples of patients with liver biopsies colonized by bacteria (1% vs 3%; p = 0.0339). CONCLUSION To the best of our knowledge, this is the first study reporting the presence of bacterial genome in a liver biopsy on bariatric patients, instead of the microbe-associated molecular patterns. Notably, in literature, the presence of Roseburia intestinalis in stool samples has been shown to prevent intestinal inflammation playing its role in the gut barrier integrity. In our population, the Roseburia reduction was associated with the presence of bacterial genome in the liver, probably related to a greater permeability of the gut and vascular barriers.
Collapse
Affiliation(s)
- Lucia Paiano
- Department of Medical, Surgical and Health Sciences, University of Trieste, Strada di Fiume, 447, 34149, Trieste, Italy
- Surgical Clinic Unit, Cattinara Hospital, ASUGI, Strada di Fiume, 447, 34149, Trieste, Italy
| | - Manuela Mastronardi
- Department of Medical, Surgical and Health Sciences, University of Trieste, Strada di Fiume, 447, 34149, Trieste, Italy.
- Surgical Clinic Unit, Cattinara Hospital, ASUGI, Strada di Fiume, 447, 34149, Trieste, Italy.
| | - Giuseppina Campisciano
- Institute for Maternal and Child Health IRCCS, Burlo Garofolo, Via dell' Istria 65/1, 34149, Trieste, Italy
| | - Natalia Rosso
- Fondazione Italiana Fegato, Centro Studi Fegato, Area Science Park Basovizza Bldg.Q SS14 Km, 163.5, 34149, Trieste, Italy
| | - Biagio Casagranda
- Surgical Clinic Unit, Cattinara Hospital, ASUGI, Strada di Fiume, 447, 34149, Trieste, Italy
| | - Manola Comar
- Department of Medical, Surgical and Health Sciences, University of Trieste, Strada di Fiume, 447, 34149, Trieste, Italy
- Institute for Maternal and Child Health IRCCS, Burlo Garofolo, Via dell' Istria 65/1, 34149, Trieste, Italy
| | - Nicolò de Manzini
- Department of Medical, Surgical and Health Sciences, University of Trieste, Strada di Fiume, 447, 34149, Trieste, Italy
- Surgical Clinic Unit, Cattinara Hospital, ASUGI, Strada di Fiume, 447, 34149, Trieste, Italy
| | - Silvia Palmisano
- Department of Medical, Surgical and Health Sciences, University of Trieste, Strada di Fiume, 447, 34149, Trieste, Italy
- Surgical Clinic Unit, Cattinara Hospital, ASUGI, Strada di Fiume, 447, 34149, Trieste, Italy
| |
Collapse
|
47
|
Getachew B, Hauser SR, Bennani S, El Kouhen N, Sari Y, Tizabi Y. Adolescent alcohol drinking interaction with the gut microbiome: implications for adult alcohol use disorder. ADVANCES IN DRUG AND ALCOHOL RESEARCH 2024; 4:11881. [PMID: 38322648 PMCID: PMC10846679 DOI: 10.3389/adar.2024.11881] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Accepted: 01/04/2024] [Indexed: 02/08/2024]
Abstract
Reciprocal communication between the gut microbiota and the brain, commonly referred to as the "gut-brain-axis" is crucial in maintaining overall physiological homeostasis. Gut microbiota development and brain maturation (neuronal connectivity and plasticity) appear to be synchronized and to follow the same timeline during childhood (immature), adolescence (expansion) and adulthood (completion). It is important to note that the mesolimbic reward circuitry develops early on, whereas the maturation of the inhibitory frontal cortical neurons is delayed. This imbalance can lead to increased acquirement of reward-seeking and risk-taking behaviors during adolescence, and consequently eventuate in heightened risk for substance abuse. Thus, there is high initiation of alcohol drinking in early adolescence that significantly increases the risk of alcohol use disorder (AUD) in adulthood. The underlying causes for heightened AUD risk are not well understood. It is suggested that alcohol-associated gut microbiota impairment during adolescence plays a key role in AUD neurodevelopment in adulthood. Furthermore, alcohol-induced dysregulation of microglia, either directly or indirectly through interaction with gut microbiota, may be a critical neuroinflammatory pathway leading to neurodevelopmental impairments and AUD. In this review article, we highlight the influence of adolescent alcohol drinking on gut microbiota, gut-brain axis and microglia, and eventual manifestation of AUD. Furthermore, novel therapeutic interventions via gut microbiota manipulations are discussed briefly.
Collapse
Affiliation(s)
- Bruk Getachew
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, United States
| | - Sheketha R. Hauser
- Department of Psychiatry, Indiana University School of Medicine, Indianapolis, IN, United States
| | - Samia Bennani
- Faculty of Medicine and Pharmacy of Casablanca, Hassan II University, Casablanca, Morocco
| | - Nacer El Kouhen
- Faculty of Medicine and Pharmacy of Casablanca, Hassan II University, Casablanca, Morocco
| | - Youssef Sari
- Department of Pharmacology and Experimental Therapeutics, College of Pharmacy and Pharmaceutical Sciences, University of Toledo, Toledo, OH, United States
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC, United States
| |
Collapse
|
48
|
Ge J, Li G, Chen Z, Xu W, Lei X, Zhu S. Kaempferol and nicotiflorin ameliorated alcohol-induced liver injury in mice by miR-138-5p/SIRT1/FXR and gut microbiota. Heliyon 2024; 10:e23336. [PMID: 38205320 PMCID: PMC10777378 DOI: 10.1016/j.heliyon.2023.e23336] [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: 08/31/2023] [Revised: 11/30/2023] [Accepted: 11/30/2023] [Indexed: 01/12/2024] Open
Abstract
Aims Excessive alcohol consumption can lead to alcoholic liver diseases (ALDs). Tetrastigma hemsleyanum Diels et Gilg is a rare Chinese medicinal herb. Tetrastigma hemsleyanum Diels et Gilg has been validated to be highly effective for treating hepatitis. Kaempferol and nicotiflorin are two highly representative flavonoids, which have exhibit therapeutic effects on liver disease. Therefore, the protective mechanism of kaempferol and nicotiflorin on alcohol-induced liver injury were investigated. Main methods Forty mice were used in this study. After treatment of Kaempferol and nicotiflorin, serum and liver were collected and used for determination of biochemical indicators, H&E staining, and molecular detection. The interaction of miRNAs from serum extracellular vehicles (EVs) with mRNAs and 16S rRNA sequencing of gut microbiota were also investigated. Key findings The results showed that kaempferol and nicotiflorins significantly ameliorated alcohol-induced liver damage and observably regulated gut microbiota. Specifically, the levels of malondialdehyde (MDA) and CYP2E1 in the liver significantly reduced, and the activity of superoxide dismutase (SOD) and glutathione (GSH) in the liver evidently increased. They also significantly relieved liver oxidative stress and lipid accumulation by suppressing miR-138-5p expression, inversely enhancing deacetylase silencing information regulator 2 related enzyme-1 (SIRT1) levels and then decreasing farnesoid X receptor (FXR) acetylation, which then modulated Nrf2 and SREBP-1c signaling pathways to regulate oxidative stress and lipid metabolism induced by alcohol. Significance Kaempferol and nicotiflorin reduced alcohol-induced liver damage by enhancing alcohol metabolism and reducing oxidative stress and lipid metabolism. The intestinal microorganism disorder was also ameliorated after oral kaempferol and nicotiflorin.
Collapse
Affiliation(s)
| | | | | | - Weijia Xu
- College of Life Sciences, China Jiliang University, 258 XueYuan Street, XiaSha Higher Education Zone. Hangzhou, 310018, Zhejiang Province, People's Republic of China
| | - Xuanhao Lei
- College of Life Sciences, China Jiliang University, 258 XueYuan Street, XiaSha Higher Education Zone. Hangzhou, 310018, Zhejiang Province, People's Republic of China
| | - Shengnan Zhu
- College of Life Sciences, China Jiliang University, 258 XueYuan Street, XiaSha Higher Education Zone. Hangzhou, 310018, Zhejiang Province, People's Republic of China
| |
Collapse
|
49
|
Chi X, Sun X, Cheng D, Liu S, Q. Pan C, Xing H. Intestinal microbiome-targeted therapies improve liver function in alcohol-related liver disease by restoring bifidobacteria: a systematic review and meta-analysis. Front Pharmacol 2024; 14:1274261. [PMID: 38259268 PMCID: PMC10800551 DOI: 10.3389/fphar.2023.1274261] [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: 08/08/2023] [Accepted: 12/05/2023] [Indexed: 01/24/2024] Open
Abstract
Objective: To systematically evaluate the efficacy of intestinal microbiome-targeted therapies (MTTs) in alcohol-related liver disease (ALD). Methods: With pre-specified keywords and strategies, we searched databases including Cochrane Library, PubMed, EMBASE, CNKI, Wanfang Data, and Weipu for RCTs on intestinal MTTs in ALD patients from January 2000 to May 2021. Two researchers independently conducted literature screening, data extraction, and quality evaluation according to the eligible criteria. Outcomes of interest included the effects of intestinal MTTs on ALT, AST, GGT, TBIL, TNF-α, IL-6, intestinal Escherichia coli, and Bifidobacteria when compared to the control group. Pooled data were compiled and analyzed with Revman 5.4 software. Results: Among 5 RCTs included with 456 ALD patients who received probiotics, the therapeutic pooled effects in the experimental group were the followings: ALT (MD = -7.16.95% CI: 10.71∼-3.60; p < 0.0001)、AST (MD = -25.11.95% CI: 30.57∼-19.47; p < 0.00001)、GGT (MD = -6.72.95% CI: 11.91∼-1.53; p = 0.01)、IL-6(SMD = -0.82.95% CI: 1.10∼-0.54; p < 0.00001), which were significantly better than those in the placebo or standard treatment group respectively, while the difference of TBIL (SMD = -0.06, 95%CI: 0.29-0.16; p = 0.59), TNF-α(SMD = -0.53.95% CI: 1.57-0.50; p = 0.31)in the two groups was not significant. After intestinal MTT treatment, the number of intestinal Bifidobacteria increased significantly (MD = 0.79.95% CI: 0.00-1.58; p = 0.05)in the experimental group. However, there were no significant changes in the number of E. coli in both groups (SMD = -0.29.95% CI: 0.92-0.34; p = 0.36). Conclusion: Intestinal MTTs can significantly improve liver function, associated with the increase of intestinal Bifidobacteria, which may be beneficial to ALD. Systematic Review Registration: https://www.crd.york.ac.uk/prospero/display_record.php?ID=CRD42021246067, Identifier CRD42021246067.
Collapse
Affiliation(s)
- Xin Chi
- Center of Liver Diseases Division, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Center for Infectious Diseases, Beijing, China
| | - Xiu Sun
- Center of Liver Diseases Division, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Center for Infectious Diseases, Beijing, China
| | - Danying Cheng
- Center of Liver Diseases Division, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Center for Infectious Diseases, Beijing, China
- Peking University Ditan Teaching Hospital, Beijing, China
| | - Shunai Liu
- Beijing Key Laboratory of Emerging Infectious Diseases, Institute of Infectious Disease, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Center for Infectious Diseases, Beijing, China
| | - Calvin Q. Pan
- Center of Liver Diseases Division, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Center for Infectious Diseases, Beijing, China
- Division of Gastroenterology and Hepatology, NYU Langone Health, New York University School of Medicine, New York, NY, United States
| | - Huichun Xing
- Center of Liver Diseases Division, Beijing Ditan Hospital, Capital Medical University, Beijing, China
- National Center for Infectious Diseases, Beijing, China
- Peking University Ditan Teaching Hospital, Beijing, China
| |
Collapse
|
50
|
Zhang Q, Li G, Zhao W, Wang X, He J, Zhou L, Zhang X, An P, Liu Y, Zhang C, Zhang Y, Liu S, Zhao L, Liu R, Li Y, Jiang W, Wang X, Wang Q, Fang B, Zhao Y, Ren Y, Niu X, Li D, Shi S, Hung WL, Wang R, Liu X, Ren F. Efficacy of Bifidobacterium animalis subsp. lactis BL-99 in the treatment of functional dyspepsia: a randomized placebo-controlled clinical trial. Nat Commun 2024; 15:227. [PMID: 38172093 PMCID: PMC10764899 DOI: 10.1038/s41467-023-44292-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2023] [Accepted: 12/07/2023] [Indexed: 01/05/2024] Open
Abstract
Current treatment for functional dyspepsia (FD) has limited and unsustainable efficacy. Probiotics have the sustainable potential to alleviate FD. This randomized controlled clinical trial (Chinese Clinical Trial Registry, ChiCTR2000041430) assigned 200 FD patients to receive placebo, positive-drug (rabeprazole), or Bifidobacterium animalis subsp. lactis BL-99 (BL-99; low, high doses) for 8-week. The primary outcome was the clinical response rate (CRR) of FD score after 8-week treatment. The secondary outcomes were CRR of FD score at other periods, and PDS, EPS, serum indicators, fecal microbiota and metabolites. The CRR in FD score for the BL-99_high group [45 (90.0%)] was significantly higher than that for placebo [29 (58.0%), p = 0.001], BL-99_low [37 (74.0%), p = 0.044] and positive_control [35 (70.0%), p = 0.017] groups after 8-week treatment. This effect was sustained until 2-week after treatment but disappeared 8-week after treatment. Further metagenomic and metabolomics revealed that BL-99 promoted the accumulation of SCFA-producing microbiota and the increase of SCFA levels in stool and serum, which may account for the increase of serum gastrin level. This study supports the potential use of BL-99 for the treatment of FD.
Collapse
Affiliation(s)
- Qi Zhang
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Guang Li
- Department of Gastroenterology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China
| | - Wen Zhao
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Xifan Wang
- Department of Obstetrics and Gynecology, Columbia University, New York, NY, USA
| | - Jingjing He
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Limian Zhou
- School of Food and Biological Engineering, Hefei University of Technology, Hefei, China
| | - Xiaoxu Zhang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Peng An
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yinghua Liu
- Department of Nutrition, Chinese PLA General Hospital, Beijing, China
| | - Chengying Zhang
- Department of General Practice, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yong Zhang
- Department of Nutrition, Chinese PLA General Hospital, Beijing, China
| | - Simin Liu
- Center for Global Cardiometabolic Health, Departments of Epidemiology, Medicine, and Surgery, Brown University, Providence, RI, USA
| | - Liang Zhao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Rong Liu
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yixuan Li
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Wenjian Jiang
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Xiaoyu Wang
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Qingyu Wang
- Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, China
| | - Bing Fang
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Yuyang Zhao
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Yimei Ren
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China
| | - Xiaokang Niu
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Dongjie Li
- Department of Cardiovascular Surgery, Beijing Anzhen Hospital, Capital Medical University, Beijing, China
| | - Shaoqi Shi
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, China
| | - Wei-Lian Hung
- National Center of Technology Innovation for Dairy, Inner Mongolia Dairy Technology Research Institute Co. Ltd., Hohhot, China.
| | - Ran Wang
- Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing, China.
| | - Xinjuan Liu
- Department of Gastroenterology, Beijing Chaoyang Hospital, Capital Medical University, Beijing, China.
| | - Fazheng Ren
- Key Laboratory of Functional Dairy, Co-constructed by Ministry of Education and Beijing Government, Department of Nutrition and Health, China Agricultural University, Beijing, China.
| |
Collapse
|