1
|
Liu Z, You C. The bile acid profile. Clin Chim Acta 2025; 565:120004. [PMID: 39419312 DOI: 10.1016/j.cca.2024.120004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 10/13/2024] [Accepted: 10/14/2024] [Indexed: 10/19/2024]
Abstract
As a large and structurally diverse family of small molecules, bile acids play a crucial role in regulating lipid, glucose, and energy metabolism. In the human body, bile acids share a similar chemical structure with many isomers, exhibit little difference in polarity, and possess various physiological activities. The types and contents of bile acids present in different diseases vary significantly. Therefore, comprehensive and accurate detection of the content of various types of bile acids in different biological samples can not only provide new insights into the pathogenesis of diseases but also facilitate the exploration of novel strategies for disease diagnosis, treatment, and prognosis. The detection of disease-induced changes in bile acid profiles has emerged as a prominent research focus in recent years. Concurrently, targeted metabolomics methods utilizing high-performance liquid chromatography-mass spectrometry (HPLC-MS) have progressively established themselves as the predominant technology for the separation and detection of bile acids. Bile acid profiles will increasingly play an important role in diagnosis and guidance in the future as the relationship between disease and changes in bile acid profiles becomes clearer. This highlights the growing diagnostic value of bile acid profiles and their potential to guide clinical decision-making. This review aims to explore the significance of bile acid profiles in clinical diagnosis from four perspectives: the synthesis and metabolism of bile acids, techniques for detecting bile acid profiles, changes in bile acid profiles associated with diseases, and the challenges and future prospects of applying bile acid profiles in clinical settings.
Collapse
Affiliation(s)
- Zhenhua Liu
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China
| | - Chongge You
- Laboratory Medicine Center, The Second Hospital & Clinical Medical School, Lanzhou University, Lanzhou 730030, China.
| |
Collapse
|
2
|
Zheng ZL, Zheng QF, Wang LQ, Liu Y. Bowel preparation before colonoscopy: Consequences, mechanisms, and treatment of intestinal dysbiosis. World J Gastroenterol 2025; 31:100589. [DOI: 10.3748/wjg.v31.i2.100589] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2024] [Revised: 10/22/2024] [Accepted: 11/12/2024] [Indexed: 12/18/2024] Open
Abstract
The term “gut microbiota” primarily refers to the ecological community of various microorganisms in the gut, which constitutes the largest microbial community in the human body. Although adequate bowel preparation can improve the results of colonoscopy, it may interfere with the gut microbiota. Bowel preparation for colonoscopy can lead to transient changes in the gut microbiota, potentially affecting an individual’s health, especially in vulnerable populations, such as patients with inflammatory bowel disease. However, measures such as oral probiotics may ameliorate these adverse effects. We focused on the bowel preparation-induced changes in the gut microbiota and host health status, hypothesized the factors influencing these changes, and attempted to identify measures that may reduce dysbiosis, thereby providing more information for individualized bowel preparation for colonoscopy in the future.
Collapse
Affiliation(s)
- Ze-Long Zheng
- Department of Gastroenterology (Endoscopy Center), China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Qing-Fan Zheng
- Department of Gastroenterology (Endoscopy Center), China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Li-Qiang Wang
- Department of Gastroenterology (Endoscopy Center), China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Yi Liu
- Department of Gastroenterology (Endoscopy Center), China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| |
Collapse
|
3
|
Kellogg TD, Ceglia S, Mortzfeld BM, Tanna TM, Zeamer AL, Mancini MR, Foley SE, Ward DV, Bhattarai SK, McCormick BA, Reboldi A, Bucci V. Succinate-producing microbiota drives tuft cell hyperplasia to protect against Clostridioides difficile. J Exp Med 2025; 222:e20232055. [PMID: 39589553 PMCID: PMC11602550 DOI: 10.1084/jem.20232055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2023] [Revised: 09/26/2024] [Accepted: 10/23/2024] [Indexed: 11/27/2024] Open
Abstract
The role of microbes and their metabolites in modulating tuft cell (TC) dynamics in the large intestine and the relevance of this pathway to infections is unknown. Here, we uncover that microbiome-driven colonic TC hyperplasia protects against Clostridioides difficile infection. Using selective antibiotics, we demonstrate increased type 2 cytokines and TC hyperplasia in the colon but not in the ileum. We demonstrate the causal role of the microbiome in modulating this phenotype using fecal matter transplantation and administration of consortia of succinate-producing bacteria. Administration of succinate production-deficient microbes shows a reduced response in a Pou2f3-dependent manner despite similar intestinal colonization. Finally, antibiotic-treated mice prophylactically administered with succinate-producing bacteria show increased protection against C. difficile-induced morbidity and mortality. This effect is nullified in Pou2f3-/- mice, confirming that the protection occurs via the TC pathway. We propose that activation of TCs by the microbiota in the colon is a mechanism evolved by the host to counterbalance microbiome-derived cues that facilitate invasion by pathogens.
Collapse
Affiliation(s)
- Tasia D. Kellogg
- Department of Microbiology, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
| | - Simona Ceglia
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
- Department of Pathology, UMass Chan Medical School, Worcester, MA, USA
| | - Benedikt M. Mortzfeld
- Department of Microbiology, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
| | - Tanvi M. Tanna
- Department of Pathology, UMass Chan Medical School, Worcester, MA, USA
| | - Abigail L. Zeamer
- Department of Microbiology, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
| | - Matthew R. Mancini
- Department of Microbiology, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
| | - Sage E. Foley
- Department of Microbiology, UMass Chan Medical School, Worcester, MA, USA
| | - Doyle V. Ward
- Department of Microbiology, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
| | - Shakti K. Bhattarai
- Department of Microbiology, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
| | - Beth A. McCormick
- Department of Microbiology, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
| | - Andrea Reboldi
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
- Department of Pathology, UMass Chan Medical School, Worcester, MA, USA
| | - Vanni Bucci
- Department of Microbiology, UMass Chan Medical School, Worcester, MA, USA
- Program in Microbiome Dynamics, UMass Chan Medical School, Worcester, MA, USA
- Immunology and Microbial Pathogenesis Program, UMass Chan Medical School, Worcester, MA, USA
| |
Collapse
|
4
|
Yilmaz B, Macpherson AJ. Delving the depths of 'terra incognita' in the human intestine - the small intestinal microbiota. Nat Rev Gastroenterol Hepatol 2025; 22:71-81. [PMID: 39443711 DOI: 10.1038/s41575-024-01000-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 09/25/2024] [Indexed: 10/25/2024]
Abstract
The small intestinal microbiota has a crucial role in gastrointestinal health, affecting digestion, immune function, bile acid homeostasis and nutrient metabolism. The challenges of accessibility at this site mean that our knowledge of the small intestinal microbiota is less developed than of the colonic or faecal microbiota. Here, we summarize the features and fluctuations of the microbiota along the small intestinal tract, focusing on humans, and discuss physicochemical factors and assessment methods, including the technical challenges of investigating the low microbial biomass of the proximal small bowel. We highlight the essential protective mechanisms of the small intestine, including motility, the paracellular barrier and mucus, and secretory immunity, to show their roles in limiting excessive exposure of host tissues to microbial metabolites. We address current knowledge gaps, particularly the variability among individuals, the effects of dysbiosis of the small intestinal microbiota on health and how different taxa in small intestinal microbiota could compensate for each other functionally.
Collapse
Affiliation(s)
- Bahtiyar Yilmaz
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, Bern, Switzerland.
- Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, Bern, Switzerland.
- Bern Center for Precision Medicine (BCPM), University of Bern, Bern, Switzerland.
| | - Andrew J Macpherson
- Department of Visceral Surgery and Medicine, Bern University Hospital, University of Bern, Bern, Switzerland.
- Maurice Müller Laboratories, Department for Biomedical Research, University of Bern, Bern, Switzerland.
- Bern Center for Precision Medicine (BCPM), University of Bern, Bern, Switzerland.
| |
Collapse
|
5
|
Singh S, Verma R. Exploring the Therapeutic Potential of Flavonoids in the Management of Cancer. Curr Pharm Biotechnol 2025; 26:17-47. [PMID: 38591206 DOI: 10.2174/0113892010297456240327062614] [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: 01/11/2024] [Revised: 02/10/2024] [Accepted: 02/21/2024] [Indexed: 04/10/2024]
Abstract
Flavonoids are a class of polyphenolic compounds that can be classified into six distinct categories, namely isoflavonoids, flavanones, flavanols, flavonols, flavones, and anthocyanidins. These compounds are naturally occurring and can be found in a diverse range of plant species. Flavonoids, a class of bioactive compounds, are mostly obtained through the consumption of vegetables, fruits and plant-derived beverages such as wine, cocoa-based products and green tea. Flavonoids have been demonstrated to exhibit a diverse range of anticancer properties. These include the modulation of activities of enzymes involved in scavenging reactive oxygen species, involvement in cell cycle arrest, induction of apoptosis and autophagy, as well as suppression of cancer cell proliferation and invasiveness. Flavonoids exhibit a dual role in maintaining reactive oxygen species balance. They function as antioxidants in regular physiological conditions, while also demonstrating significant pro-oxidant properties in cancer cells. This prooxidant activity induces apoptotic pathways and downregulates pro-inflammatory signalling pathways. The paper explores the biochemical characteristics, bioavailability, anticancer efficacy, and modes of action of flavonoids.
Collapse
Affiliation(s)
- Sonia Singh
- Institute of Pharmaceutical Research GLA University, Chaumuhan, Uttar Pradesh, 281406, India
| | - Riya Verma
- Institute of Pharmaceutical Research GLA University, Chaumuhan, Uttar Pradesh, 281406, India
| |
Collapse
|
6
|
Anand S, Patel TN. Integrating the metabolic and molecular circuits in diabetes, obesity and cancer: a comprehensive review. Discov Oncol 2024; 15:779. [PMID: 39692821 DOI: 10.1007/s12672-024-01662-1] [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: 10/04/2024] [Accepted: 12/02/2024] [Indexed: 12/19/2024] Open
Abstract
The progressive globalization of sedentary lifestyles and diets rich in lipids and processed foods has caused two major public health hazards-diabetes and obesity. The strong interlink between obesity and type 2 diabetes mellitus and their combined burden encompass them into a single term 'Diabesity'. They have also been tagged as the drivers for the onset of cancer. The clinical association between diabetes, obesity, and several types of human cancer demands an assessment of vital junctions correlating the three. This review focuses on revisiting the molecular axis linking diabetes and obesity to cancer through pathways that get imbalanced owing to metabolic upheaval. We also attempt to describe the functional disruptions of DNA repair mechanisms due to overwhelming oxidative DNA damage caused by diabesity. Genomic instability, a known cancer hallmark results when DNA repair does not work optimally, and as will be inferred from this review the obtruded metabolic homeostasis in diabetes and obesity creates a favorable microenvironment supporting metabolic reprogramming and enabling malignancies. Altered molecular and hormonal landscapes in these two morbidities provide a novel connection between metabolomics and oncogenesis. Understanding various aspects of the tumorigenic process in diabesity-induced cancers might help in the discovery of new biomarkers and prompt targeted therapeutic interventions.
Collapse
Affiliation(s)
- Shrikirti Anand
- Department of Integrative Biology, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India
| | - Trupti N Patel
- Department of Integrative Biology, School of Bio-Sciences and Technology, Vellore Institute of Technology, Vellore, Tamil Nadu, 632014, India.
| |
Collapse
|
7
|
Gopal RK, Ganesh PS, Pathoor NN. Synergistic Interplay of Diet, Gut Microbiota, and Insulin Resistance: Unraveling the Molecular Nexus. Mol Nutr Food Res 2024; 68:e2400677. [PMID: 39548908 DOI: 10.1002/mnfr.202400677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2024] [Revised: 10/04/2024] [Indexed: 11/18/2024]
Abstract
This comprehensive review explores the intricate relationship between gut microbiota, diet, and insulin resistance, emphasizing the novel roles of diet-induced microbial changes in influencing metabolic health. It highlights how diet significantly influences gut microbiota composition, with different dietary patterns fostering diverse microbial communities. These diet-induced changes in the microbiome impact human metabolism by affecting inflammation, energy balance, and insulin sensitivity, particularly through microbial metabolites like short-chain fatty acids (SCFAs). Focusing the key mediators like endotoxemia and systemic inflammation, and introduces personalized microbiome-based therapeutic strategies, it also investigates the effects of dietary components-fiber, polyphenols, and lipids-on microbiota and insulin sensitivity, along with the roles of protein intake and amino acid metabolism. The study compares the effects of Western and Mediterranean diets on the microbiota-insulin resistance axis. Therapeutic implications, including probiotics, fecal microbiota transplantation (FMT), and personalized diets, are discussed. Key findings reveal that high-fat diets, especially those rich in saturated fats, contribute to dysbiosis and increased intestinal permeability, while high-fiber diets promote beneficial bacteria and SCFAs. The review underscores the future potential of food and microbiota interventions for preventing or managing insulin resistance.
Collapse
Affiliation(s)
- Rajesh Kanna Gopal
- Department of Microbiology, Centre for Infectious Diseases, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University (Deemed to be University), Chennai, Tamil Nadu, 600077, India
| | - Pitchaipillai Sankar Ganesh
- Department of Microbiology, Centre for Infectious Diseases, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University (Deemed to be University), Chennai, Tamil Nadu, 600077, India
| | - Naji Naseef Pathoor
- Department of Microbiology, Centre for Infectious Diseases, Saveetha Dental College and Hospitals, Saveetha Institute of Medical and Technical Sciences (SIMATS), Saveetha University (Deemed to be University), Chennai, Tamil Nadu, 600077, India
| |
Collapse
|
8
|
Castelnuovo G, Perez-Diaz-Del-Campo N, Guariglia M, Poggiolini I, Armandi A, Rosso C, Caviglia GP, Bugianesi E. Prebiotics targeting gut-liver axis to treat non-alcoholic fatty liver disease. Minerva Gastroenterol (Torino) 2024; 70:446-453. [PMID: 36892817 DOI: 10.23736/s2724-5985.23.03361-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/10/2023]
Abstract
Non-alcoholic steatohepatitis (NASH) is a high-prevalence, rapidly growing form of non-alcoholic fatty liver disease (NAFLD), which is closely linked to obesity and metabolic disorders. Gut microbiota has been increasingly recognized as a key factor in the onset of NAFLD in recent years. The liver can be strongly influenced by changes in the gut microbiota through the portal vein, giving the gut-liver axis a very important role in understanding the pathophysiology of liver diseases. A healthy intestinal barrier is characterized by selective permeability to nutrients, metabolites, water and bacterial products and its impairment may be a predisposing or aggravating condition for the progression of NAFLD. In most cases, NAFLD patients follow a Western diet pattern, which is closely linked to obesity and associated metabolic diseases, promoting inflammation, structural and behavioral changes in the gut microbiota. In fact, factors such as age, gender, genetic or environmental factors may induce a dysbiotic microbiota that promotes epithelial barrier dysfunction and increased intestinal permeability, favoring the progression of NAFLD. In this context, new dietary approaches, such as prebiotics, are emerging to prevent disease and maintain health. In this review, we reported the role of the gut-liver axis in the pathogenesis of NAFLD and investigated the potential therapeutic effect of prebiotics on the enhancement of intestinal barrier dysfunction, hepatic steatosis and, consequently, the progression of NAFLD.
Collapse
Affiliation(s)
| | | | - Marta Guariglia
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Irene Poggiolini
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Angelo Armandi
- Department of Medical Sciences, University of Turin, Turin, Italy
- Metabolic Liver Disease Research Program, First Department of Medicine, University Medical Center of the Johannes Gutenberg-University, Mainz, Germany
| | - Chiara Rosso
- Department of Medical Sciences, University of Turin, Turin, Italy
| | - Gian P Caviglia
- Department of Medical Sciences, University of Turin, Turin, Italy -
| | - Elisabetta Bugianesi
- Department of Medical Sciences, University of Turin, Turin, Italy
- Unit of Gastroenterology, Molinette Hospital, Città della Salute e della Scienza, Turin, Italy
| |
Collapse
|
9
|
Fan X, Dong W, Huang Y, Shu Y, Yan Y, Mi J, Lu L, Zeng X, Cao Y. Aqueous Extract of Lycium ruthenicum Murray Attenuates Neuroinflammation in C57BL/6J Mice Induced by High-Fat and High-Fructose Diet Through Regulating Gut Microbiota and Bile Acid Metabolism. Foods 2024; 13:3812. [PMID: 39682885 DOI: 10.3390/foods13233812] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 11/09/2024] [Accepted: 11/21/2024] [Indexed: 12/18/2024] Open
Abstract
The aqueous extract of Lycium ruthenicum Murray (LRE) could attenuate neuroinflammation in mice induced by a high-fat and high-fructose diet (HFFD). Moreover, LRE could adjust bile acid (BA) metabolism and the gut microbiota. Behavioral tests revealed that LRE prevented HFFD-induced cognitive deficits. The treatment of LRE resulted in a decreased expression of inflammation-related mRNA of TNF-α, IL-6, and IL-1β in the cerebral cortex and hippocampus. Furthermore, LRE ameliorated gut microbiota disorder caused by HFFD by markedly elevating the relative abundances of Streptococcus and probiotics such as Lactococcus. Concurrently, it reduced the relative abundances of Helicobacter and Clostridium_XIVa. The levels of tauroursodeoxycholic acid, known for its neuroprotective property, and taurocholic acid, recognized as an anti-inflammatory agent, were significantly enhanced in the hippocampus and cerebral cortex due to the treatment with LRE. In a word, LRE might have the potential to alleviate HFFD-induced cognitive dysfunction by modulating intestinal microbiota and promoting the synthesis of neuroprotective BAs.
Collapse
Affiliation(s)
- Xia Fan
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Wei Dong
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yujie Huang
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yifan Shu
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Yamei Yan
- Institute of Wolfberry Engineering Technology, Ningxia Academy of Agriculture and Forestry Sciences, Yinchuan 750002, China
- National Wolfberry Engineering Research Center, Yinchuan 750002, China
| | - Jia Mi
- Institute of Wolfberry Engineering Technology, Ningxia Academy of Agriculture and Forestry Sciences, Yinchuan 750002, China
- National Wolfberry Engineering Research Center, Yinchuan 750002, China
| | - Lu Lu
- Institute of Wolfberry Engineering Technology, Ningxia Academy of Agriculture and Forestry Sciences, Yinchuan 750002, China
- National Wolfberry Engineering Research Center, Yinchuan 750002, China
| | - Xiaoxiong Zeng
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China
| | - Youlong Cao
- Institute of Wolfberry Engineering Technology, Ningxia Academy of Agriculture and Forestry Sciences, Yinchuan 750002, China
- National Wolfberry Engineering Research Center, Yinchuan 750002, China
| |
Collapse
|
10
|
McCune E, Sharma A, Johnson B, O'Meara T, Theiner S, Campos M, Heditsian D, Brain S, Gilbert JA, Esserman L, Campbell MJ. Gut and oral microbial compositional differences in women with breast cancer, women with ductal carcinoma in situ, and healthy women. mSystems 2024; 9:e0123724. [PMID: 39470189 PMCID: PMC11575313 DOI: 10.1128/msystems.01237-24] [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: 09/10/2024] [Accepted: 10/05/2024] [Indexed: 10/30/2024] Open
Abstract
This study characterized and compared the fecal and oral microbiota from women with early-stage breast cancer (BC), women with ductal carcinoma in situ (DCIS), and healthy women. Fecal and oral samples were collected from newly diagnosed patients prior to any therapy and characterized using 16S rRNA sequencing. Measures of gut microbial alpha diversity were significantly lower in the BC vs healthy cohort. Beta diversity differed significantly between the BC or DCIS and healthy groups, and several differentially abundant taxa were identified. Clustering (non-negative matrix factorization) of the gut microbiota identified five bacterial guilds dominated by Prevotella, Enterobacteriaceae, Akkermansia, Clostridiales, or Bacteroides. The Bacteroides and Enterobacteriaceae guilds were significantly more abundant in the BC cohort compared to healthy controls, whereas the Clostridiales guild was more abundant in the healthy group. Finally, prediction of functional pathways identified 23 pathways that differed between the BC and healthy gut microbiota including lipopolysaccharide biosynthesis, glycan biosynthesis and metabolism, lipid metabolism, and sphingolipid metabolism. In contrast to the gut microbiomes, there were no significant differences in alpha or beta diversity in the oral microbiomes, and very few differentially abundant taxa were observed. Non-negative matrix factorization analysis of the oral microbiota samples identified seven guilds dominated by Veillonella, Prevotella, Gemellaceae, Haemophilus, Neisseria, Propionibacterium, and Streptococcus; however, none of these guilds were differentially associated with the different cohorts. Our results suggest that alterations in the gut microbiota may provide the basis for interventions targeting the gut microbiome to improve treatment outcomes and long-term prognosis. IMPORTANCE Emerging evidence suggests that the gut microbiota may play a role in breast cancer. Few studies have evaluated both the gut and oral microbiomes in women with breast cancer (BC), and none have characterized these microbiomes in women with ductal carcinoma in situ (DCIS). We surveyed the gut and oral microbiomes from women with BC or DCIS and healthy women and identified compositional and functional features of the gut microbiota that differed between these cohorts. In contrast, very few differential features were identified in the oral microbiota. Understanding the role of gut bacteria in BC and DCIS may open up new opportunities for the development of novel markers for early detection (or markers of susceptibility) as well as new strategies for prevention and/or treatment.
Collapse
Affiliation(s)
- Emma McCune
- Department of Surgery, University of California, San Francisco, California, USA
| | - Anukriti Sharma
- Department of Quantitative Health Sciences, Cleveland Clinic, Cleveland, Ohio, USA
| | - Breanna Johnson
- Department of Surgery, University of California, San Francisco, California, USA
| | - Tess O'Meara
- Department of Surgery, University of California, San Francisco, California, USA
| | - Sarah Theiner
- Department of Surgery, University of California, San Francisco, California, USA
| | - Maribel Campos
- Department of Surgery, University of California, San Francisco, California, USA
| | - Diane Heditsian
- Department of Surgery, University of California, San Francisco, California, USA
| | - Susie Brain
- Department of Surgery, University of California, San Francisco, California, USA
| | - Jack A Gilbert
- Department of Pediatrics, University of California, San Diego, California, USA
| | - Laura Esserman
- Department of Surgery, University of California, San Francisco, California, USA
| | - Michael J Campbell
- Department of Surgery, University of California, San Francisco, California, USA
| |
Collapse
|
11
|
Chen L, Yan H, Di S, Guo C, Zhang H, Zhang S, Gold A, Wang Y, Hu M, Wu D, Johnson CH, Wang X, Zhu J. Mapping Pesticide-Induced Metabolic Alterations in Human Gut Bacteria. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.15.623895. [PMID: 39605636 PMCID: PMC11601348 DOI: 10.1101/2024.11.15.623895] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Pesticides can modulate gut microbiota (GM) composition, but their specific effects on GM remain largely elusive. Our study demonstrated that pesticides inhibit or promote growth in various GM species, even at low concentrations, and can accumulate in GM to prolong their presence in the host. Meanwhile, the pesticide induced changes in GM composition are associated with significant alterations in gut bacterial metabolism that reflected by the changes of hundreds of metabolites. We generated a pesticide-GM-metabolites (PMM) network that not only reveals pesticide-sensitive gut bacteria species but also report specific metabolic changes in 306 pesticide-GM pairs (PGPs). Using an in vivo mice model, we further demonstrated a PGP's interactions and verified the inflammation-inducing effects of pesticides on the host through dysregulated lipid metabolism of microbes. Taken together, our findings generate a PMM interactions atlas, and shed light on the molecular level of how pesticides impact host health by modulating GM metabolism.
Collapse
Affiliation(s)
- Li Chen
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
- James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Hong Yan
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
- State Key Laboratory of Environmental and Biological Analysis, Hong Kong Baptist University, Hong Kong, China
| | - Shanshan Di
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products/ Key Laboratory of Detection for Pesticide Residues and Control of Zhejiang, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Chao Guo
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Huan Zhang
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
- James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| | - Shiqi Zhang
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Andrew Gold
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Yu Wang
- Department of Physiology, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | - Ming Hu
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
| | - Dayong Wu
- Department of Cancer Biology and Genetics, The Ohio State University College of Medicine, Columbus, OH 43210, USA
| | - Caroline H. Johnson
- Department of Environmental Health Sciences, Yale School of Public Health, New Haven, CT, USA
| | - Xinquan Wang
- State Key Laboratory for Managing Biotic and Chemical Threats to the Quality and Safety of Agro-products/ Key Laboratory of Detection for Pesticide Residues and Control of Zhejiang, Institute of Agro-product Safety and Nutrition, Zhejiang Academy of Agricultural Sciences, Hangzhou 310021, China
| | - Jiangjiang Zhu
- Human Nutrition Program, Department of Human Sciences, The Ohio State University, Columbus, OH 43210, USA
- James Comprehensive Cancer Center, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
12
|
Fang X, Lee S, Rayalam S, Park HJ. Docosahexaenoic acid supplementation and infant brain development: role of gut microbiome. Nutr Res 2024; 131:1-13. [PMID: 39342808 DOI: 10.1016/j.nutres.2024.08.005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Revised: 08/30/2024] [Accepted: 08/30/2024] [Indexed: 10/01/2024]
Abstract
Perinatal stage represents a critical period for brain development. Docosahexaenoic acid (DHA) is a ω-3 polyunsaturated fatty acid preferentially accumulated in the brain that may benefit neurodevelopment. Microbial colonization and maturation parallel with the rapid development of infant metabolic and brain function that may influence the effects of DHA on neurological development. This review aims to summarize the current literature on the mediating effects of DHA on brain and gut microbiome development and attempts to reevaluate the efficacy of DHA from a gut microbiome-mediated perspective. Specifically, the regulatory roles of DHA on hypothalamic-pituitary-adrenal axis, inflammation, and neuroactive mediators may be partly moderated through gut microbiome. Consideration of the gut microbiome and gut-brain communication, when evaluating the efficacy of DHA, may provide new insights in better understanding the mechanisms of DHA and impart advantages to future development of nutritional therapy based on the nutrient-microbiome interaction.
Collapse
Affiliation(s)
- Xi Fang
- Department of Nutritional Sciences, College of Family and Consumer Sciences, University of Georgia, Athens, GA
| | - Soon Lee
- Department of Nutritional Sciences, College of Family and Consumer Sciences, University of Georgia, Athens, GA
| | - Srujana Rayalam
- Department of Pharmaceutical Sciences, Philadelphia College of Osteopathic Medicine, Philadelphia, PA
| | - Hea Jin Park
- Department of Nutritional Sciences, College of Family and Consumer Sciences, University of Georgia, Athens, GA.
| |
Collapse
|
13
|
Sun Y, Men Q, Ren X, Yan C, Song S, Ai C. Low molecular fucoidan alleviated alcohol-induced liver injury in BALB/c mice by regulating the gut microbiota-bile acid-liver axis. Int J Biol Macromol 2024; 282:136930. [PMID: 39490864 DOI: 10.1016/j.ijbiomac.2024.136930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2024] [Revised: 09/24/2024] [Accepted: 10/24/2024] [Indexed: 11/05/2024]
Abstract
Fucoidan has attracted significant attention owing to its remarkable bioactivities, but the effect of molecular weight (Mw) on its activities in the context of alcoholic liver diseases (ALD) is poorly understood. In this study, low Mw fucoidan (OSLF) was prepared, and its protective effect against alcohol-induced liver injury was assessed in a mouse model. OSLF increased weight gain and colon length, improved lipid disorders, and reduced oxidative stress in mice exposed to alcohol, alleviating liver injury. OSLF alleviated inflammation in the liver by inhibiting alcohol-activated NF-κB and MAPK pathways. The underlying mechanism can be attributed to the improvement of alcohol-induced dysbiosis of the gut microbiota, including a decrease in Proteobacteria and Bacteroidetes and an increase in microbiota diversity, as well as the abundances of Parabacteroides, Bacteroides, and Faecalibaculum. Metabolomics results showed that OSLF improved alcohol-induced abnormalities of microbiota metabolites, primarily involving amino acid metabolism and short chain fatty acids production. In addition, OSLF ameliorated bile acid metabolism in the gut and regulated the expression of bile acid-associated genes in the liver, affecting bile acid synthesis, regulation, and transport. It suggested that OSLF had the potential for the management of ALD by regulating the gut microbiota-bile acid-liver axis.
Collapse
Affiliation(s)
- Yiyun Sun
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Qiuyue Men
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Xiaomeng Ren
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Chunhong Yan
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Shuang Song
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China
| | - Chunqing Ai
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian 116034, PR China; National & Local Joint Engineering Laboratory for Marine Bioactive Polysaccharide Development and Application, Dalian Polytechnic University, Dalian 116034, PR China.
| |
Collapse
|
14
|
Seidler Y, Rimbach G, Lüersen K, Vinderola G, Ipharraguerre IR. The postbiotic potential of Aspergillus oryzae - a narrative review. Front Microbiol 2024; 15:1452725. [PMID: 39507340 PMCID: PMC11538067 DOI: 10.3389/fmicb.2024.1452725] [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: 06/21/2024] [Accepted: 10/07/2024] [Indexed: 11/08/2024] Open
Abstract
The filamentous fungus Aspergillus oryzae has a long tradition in East Asian food processing. It is therefore not surprising that in recent years fermentation products of A. oryzae have attracted attention in the emerging field of postbiotics. This review aims to provide a comprehensive summary of the potential postbiotic effects of fermentation products from A. oryzae, by discussing possible mechanisms of action against the background of the molecular composition determined so far. In particular, cell wall constituents, enzymes, extracellular polymeric substances, and various metabolites found in A. oryzae fermentation preparations are described in detail. With reference to the generally assumed key targets of postbiotics, their putative beneficial bioactivities in modulating the microbiota, improving epithelial barrier function, influencing immune responses, metabolic reactions and signaling through the nervous system are assessed. Drawing on existing literature and case studies, we highlight A. oryzae as a promising source of postbiotics, particularly in the context of animal health and nutrition. Challenges and opportunities in quality control are also addressed, with a focus on the necessity for standardized methods to fully harness the potential of fungal-based postbiotics. Overall, this article sheds light on the emerging field of A. oryzae-derived postbiotics and emphasizes the need for further research to fully realize their therapeutic potential.
Collapse
Affiliation(s)
- Yvonne Seidler
- Institute of Human Nutrition and Food Science, Division of Food Science, Faculty of Agricultural and Nutritional Sciences, University of Kiel, Kiel, Germany
| | - Gerald Rimbach
- Institute of Human Nutrition and Food Science, Division of Food Science, Faculty of Agricultural and Nutritional Sciences, University of Kiel, Kiel, Germany
| | - Kai Lüersen
- Institute of Human Nutrition and Food Science, Division of Food Science, Faculty of Agricultural and Nutritional Sciences, University of Kiel, Kiel, Germany
| | - Gabriel Vinderola
- Instituto de Lactología Industrial (CONICET-UNL), Faculty of Chemical Engineering, National University of Litoral, Santa Fe, Argentina
| | - Ignacio R. Ipharraguerre
- Institute of Human Nutrition and Food Science, Division of Food Science, Faculty of Agricultural and Nutritional Sciences, University of Kiel, Kiel, Germany
| |
Collapse
|
15
|
Habermaass V, Bartoli F, Gori E, Dini R, Cogozzo A, Puccinelli C, Pierini A, Marchetti V. Fecal Bile Acids in Canine Chronic Liver Disease: Results from 46 Dogs. Animals (Basel) 2024; 14:3051. [PMID: 39518774 PMCID: PMC11545594 DOI: 10.3390/ani14213051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 10/15/2024] [Accepted: 10/20/2024] [Indexed: 11/16/2024] Open
Abstract
The concentrations of fecal and serum bile acids (BAs) are known to be altered in human patients with chronic liver diseases (CLDs), especially those with biliary tract involvement (BTD). Scarce literature is available regarding fecal BA modifications during canine CLDs. This study aimed to evaluate fecal BAs in canine CLDs according to different clinical and clinicopathological variables. Forty-six dogs were enrolled. Canine feces were analyzed by HPLC. Cholic Acid (CA), Chenodeoxycholic Acid (CDCA), Ursodeoxycholic Acid (UDCA), Deoxycholic Acid (DCA), and Lithocholic Acid (LCA) were measured, and primary BAs (CA + CDCA), secondary BAs (UDCA + DCA + LCA), and the primary/secondary (P/S) ratio were calculated. Primary BAs (p < 0.0001), CA (p = 0.0003), CDCA (p = 0.003), the P/S ratio (p = 0.002), and total BAs (p = 0.005) were significatively higher in BTD dogs (n = 18) compared to in non-BTD dogs (n = 28). Fecal secondary BAs did not statistically differ between BTD and non-BTD dogs. Gastrointestinal clinical signs (p = 0.028) and diarrhea (p = 0.03) were significantly more prevalent in BTD dogs compared to in non-BTD dogs, supporting the hypothesis of some pathological mechanisms assimilable to bile acid diarrhea (BAD). Our results could reflect imbalances of the fecal BA metabolism in dogs with CLDs. Further studies involving gut microbiome and metabolomic assessment are needed to better understand the possible clinical implications of BA metabolism disruption and their potential role in canine CLDs.
Collapse
Affiliation(s)
- Verena Habermaass
- Department of Veterinary Sciences, Veterinary Teaching Hospital “Mario Modenato”, University of Pisa, Via Livornese Lato Monte, San Piero a Grado, 56122 Pisa, Italy; (V.H.); (V.M.)
| | - Francesco Bartoli
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Savi 10, 56126 Pisa, Italy
| | - Eleonora Gori
- Department of Veterinary Sciences, Veterinary Teaching Hospital “Mario Modenato”, University of Pisa, Via Livornese Lato Monte, San Piero a Grado, 56122 Pisa, Italy; (V.H.); (V.M.)
| | - Rebecca Dini
- Department of Veterinary Sciences, Veterinary Teaching Hospital “Mario Modenato”, University of Pisa, Via Livornese Lato Monte, San Piero a Grado, 56122 Pisa, Italy; (V.H.); (V.M.)
| | - Aurora Cogozzo
- Department of Veterinary Sciences, Veterinary Teaching Hospital “Mario Modenato”, University of Pisa, Via Livornese Lato Monte, San Piero a Grado, 56122 Pisa, Italy; (V.H.); (V.M.)
| | - Caterina Puccinelli
- Department of Veterinary Sciences, Veterinary Teaching Hospital “Mario Modenato”, University of Pisa, Via Livornese Lato Monte, San Piero a Grado, 56122 Pisa, Italy; (V.H.); (V.M.)
| | - Alessio Pierini
- Department of Veterinary Sciences, Veterinary Teaching Hospital “Mario Modenato”, University of Pisa, Via Livornese Lato Monte, San Piero a Grado, 56122 Pisa, Italy; (V.H.); (V.M.)
| | - Veronica Marchetti
- Department of Veterinary Sciences, Veterinary Teaching Hospital “Mario Modenato”, University of Pisa, Via Livornese Lato Monte, San Piero a Grado, 56122 Pisa, Italy; (V.H.); (V.M.)
| |
Collapse
|
16
|
Catalan EA, Seguel-Fuentes E, Fuentes B, Aranguiz-Varela F, Castillo-Godoy DP, Rivera-Asin E, Bocaz E, Fuentes JA, Bravo D, Schinnerling K, Melo-Gonzalez F. Oral Pathobiont-Derived Outer Membrane Vesicles in the Oral-Gut Axis. Int J Mol Sci 2024; 25:11141. [PMID: 39456922 PMCID: PMC11508520 DOI: 10.3390/ijms252011141] [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: 09/06/2024] [Revised: 10/04/2024] [Accepted: 10/12/2024] [Indexed: 10/28/2024] Open
Abstract
Oral pathobionts are essential in instigating local inflammation within the oral cavity and contribute to the pathogenesis of diseases in the gastrointestinal tract and other distant organs. Among the Gram-negative pathobionts, Porphyromonas gingivalis and Fusobacterium nucleatum emerge as critical drivers of periodontitis, exerting their influence not only locally but also as inducers of gut dysbiosis, intestinal disturbances, and systemic ailments. This dual impact is facilitated by their ectopic colonization of the intestinal mucosa and the subsequent mediation of distal systemic effects by releasing outer membrane vesicles (OMVs) into circulation. This review elucidates the principal components of oral pathobiont-derived OMVs implicated in disease pathogenesis within the oral-gut axis, detailing virulence factors that OMVs carry and their interactions with host epithelial and immune cells, both in vitro and in vivo. Additionally, we shed light on the less acknowledged interplay between oral pathobionts and the gut commensal Akkermansia muciniphila, which can directly impede oral pathobionts' growth and modulate bacterial gene expression. Notably, OMVs derived from A. muciniphila emerge as promoters of anti-inflammatory effects within the gastrointestinal and distant tissues. Consequently, we explore the potential of A. muciniphila-derived OMVs to interact with oral pathobionts and prevent disease in the oral-gut axis.
Collapse
Affiliation(s)
- Eduardo A. Catalan
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Emilio Seguel-Fuentes
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Brandon Fuentes
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Felipe Aranguiz-Varela
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Daniela P. Castillo-Godoy
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Elizabeth Rivera-Asin
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Elisa Bocaz
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Juan A. Fuentes
- Laboratorio de Genética y Patogénesis Bacteriana, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile;
| | - Denisse Bravo
- Cellular Interactions Laboratory, Faculty of Dentistry, Universidad Andrés Bello, Santiago 8370133, Chile;
| | - Katina Schinnerling
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| | - Felipe Melo-Gonzalez
- Laboratorio de Inmunología Traslacional, Facultad de Ciencias de la Vida, Universidad Andrés Bello, República 330, Santiago 8370186, Chile; (E.A.C.); (E.S.-F.); (B.F.); (F.A.-V.); (D.P.C.-G.); (E.R.-A.); (E.B.)
| |
Collapse
|
17
|
Connell E, Blokker B, Kellingray L, Le Gall G, Philo M, Pontifex MG, Narbad A, Müller M, Vauzour D. Refined diet consumption increases neuroinflammatory signalling through bile acid dysmetabolism. Nutr Neurosci 2024; 27:1088-1101. [PMID: 38170169 DOI: 10.1080/1028415x.2023.2301165] [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: 01/05/2024]
Abstract
Over recent decades, dietary patterns have changed significantly due to the increasing availability of convenient, ultra-processed refined foods. Refined foods are commonly depleted of key bioactive compounds, which have been associated with several deleterious health conditions. As the gut microbiome can influence the brain through a bidirectional communication system known as the 'microbiota-gut-brain axis', the consumption of refined foods has the potential to affect cognitive health. In this study, multi-omics approaches were employed to assess the effect of a refined diet on the microbiota-gut-brain axis, with a particular focus on bile acid metabolism. Mice maintained on a refined low-fat diet (rLFD), consisting of high sucrose, processed carbohydrates and low fibre content, for eight weeks displayed significant gut microbial dysbiosis, as indicated by diminished alpha diversity metrics (p < 0.05) and altered beta diversity (p < 0.05) when compared to mice receiving a chow diet. Changes in gut microbiota composition paralleled modulation of the metabolome, including a significant reduction in short-chain fatty acids (acetate, propionate and n-butyrate; p < 0.001) and alterations in bile acid concentrations. Interestingly, the rLFD led to dysregulated bile acid concentrations across both the colon (p < 0.05) and the brain (p < 0.05) which coincided with altered neuroinflammatory gene expression. In particular, the concentration of TCA, TDCA and T-α-MCA was inversely correlated with the expression of NF-κB1, a key transcription factor in neuroinflammation. Overall, our results suggest a novel link between a refined low-fat diet and detrimental neuronal processes, likely in part through modulation of the microbiota-gut-brain axis and bile acid dysmetabolism.
Collapse
Affiliation(s)
- Emily Connell
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Britt Blokker
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - Lee Kellingray
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | | | - Mark Philo
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | | | - Arjan Narbad
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Michael Müller
- Norwich Medical School, University of East Anglia, Norwich, UK
| | - David Vauzour
- Norwich Medical School, University of East Anglia, Norwich, UK
| |
Collapse
|
18
|
Yadav S, Sapra L, Srivastava RK. Polysaccharides to postbiotics: Nurturing bone health via modulating "gut-immune axis". Int J Biol Macromol 2024; 278:134655. [PMID: 39128750 DOI: 10.1016/j.ijbiomac.2024.134655] [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: 01/11/2024] [Revised: 08/06/2024] [Accepted: 08/08/2024] [Indexed: 08/13/2024]
Abstract
The increasing prevalence of individuals affected by bone pathologies globally has sparked catastrophic concerns. Ankylosing spondylitis, osteoporosis, rheumatoid arthritis, osteoarthritis, and fractures alone impact an estimated 1.71 billion people worldwide. The gut microbiota plays a crucial role in interacting with the host through the synthesis of a diverse range of metabolites called gut-associated metabolites (GAMs), which originate from external dietary substrates or endogenous host compounds. Many metabolic disorders have been linked to alterations in the gut microbiota's activity and composition. The development of metabolic illnesses has been linked to certain microbiota-derived metabolites, such as branched-chain amino acids, bile acids, short-chain fatty acids, tryptophan, trimethylamine N-oxide, and indole derivatives. Moreover, the modulation of gut microbiota through biotics (prebiotics, probiotics and postbiotics) presents a promising avenue for therapeutic intervention. Biotics selectively promote the growth of beneficial gut bacteria, thereby enhancing the production of GAMs with potential beneficial effects on bone metabolism. Understanding the intricate interplay between GAMs, and bone-associated genes through molecular informatics holds significant promise for early diagnosis, prognosis, and novel treatment strategies for various bone disorders.
Collapse
Affiliation(s)
- Sumedha Yadav
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Leena Sapra
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India
| | - Rupesh K Srivastava
- Translational Immunology, Osteoimmunology & Immunoporosis Lab (TIOIL), Department of Biotechnology, All India Institute of Medical Sciences (AIIMS), New Delhi 110029, India.
| |
Collapse
|
19
|
Datta S, Pasham S, Inavolu S, Boini KM, Koka S. Role of Gut Microbial Metabolites in Cardiovascular Diseases-Current Insights and the Road Ahead. Int J Mol Sci 2024; 25:10208. [PMID: 39337693 PMCID: PMC11432476 DOI: 10.3390/ijms251810208] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
Cardiovascular diseases (CVDs) are the leading cause of premature morbidity and mortality globally. The identification of novel risk factors contributing to CVD onset and progression has enabled an improved understanding of CVD pathophysiology. In addition to the conventional risk factors like high blood pressure, diabetes, obesity and smoking, the role of gut microbiome and intestinal microbe-derived metabolites in maintaining cardiovascular health has gained recent attention in the field of CVD pathophysiology. The human gastrointestinal tract caters to a highly diverse spectrum of microbes recognized as the gut microbiota, which are central to several physiologically significant cascades such as metabolism, nutrient absorption, and energy balance. The manipulation of the gut microbial subtleties potentially contributes to CVD, inflammation, neurodegeneration, obesity, and diabetic onset. The existing paradigm of studies suggests that the disruption of the gut microbial dynamics contributes towards CVD incidence. However, the exact mechanistic understanding of such a correlation from a signaling perspective remains elusive. This review has focused upon an in-depth characterization of gut microbial metabolites and their role in varied pathophysiological conditions, and highlights the potential molecular and signaling mechanisms governing the gut microbial metabolites in CVDs. In addition, it summarizes the existing courses of therapy in modulating the gut microbiome and its metabolites, limitations and scientific gaps in our current understanding, as well as future directions of studies involving the modulation of the gut microbiome and its metabolites, which can be undertaken to develop CVD-associated treatment options. Clarity in the understanding of the molecular interaction(s) and associations governing the gut microbiome and CVD shall potentially enable the development of novel druggable targets to ameliorate CVD in the years to come.
Collapse
Affiliation(s)
- Sayantap Datta
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | - Sindhura Pasham
- Department of Pharmaceutical Sciences, Irma Lerma College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA
| | - Sriram Inavolu
- Department of Pharmaceutical Sciences, Irma Lerma College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA
| | - Krishna M Boini
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, USA
| | - Saisudha Koka
- Department of Pharmaceutical Sciences, Irma Lerma College of Pharmacy, Texas A&M University, Kingsville, TX 78363, USA
| |
Collapse
|
20
|
Madani WAM, Ramos Y, Cubillos-Ruiz JR, Morales DK. Enterococcal-host interactions in the gastrointestinal tract and beyond. FEMS MICROBES 2024; 5:xtae027. [PMID: 39391373 PMCID: PMC11466040 DOI: 10.1093/femsmc/xtae027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 08/05/2024] [Accepted: 09/06/2024] [Indexed: 10/12/2024] Open
Abstract
The gastrointestinal tract (GIT) is typically considered the natural niche of enterococci. However, these bacteria also inhabit extraintestinal tissues, where they can disrupt organ physiology and cause life-threatening infections. Here, we discuss how enterococci, primarily Enterococcus faecalis, interact with the intestine and other host anatomical locations such as the oral cavity, heart, liver, kidney, and vaginal tract. The metabolic flexibility of these bacteria allows them to quickly adapt to new environments, promoting their persistence in diverse tissues. In transitioning from commensals to pathogens, enterococci must overcome harsh conditions such as nutrient competition, exposure to antimicrobials, and immune pressure. Therefore, enterococci have evolved multiple mechanisms to adhere, colonize, persist, and endure these challenges in the host. This review provides a comprehensive overview of how enterococci interact with diverse host cells and tissues across multiple organ systems, highlighting the key molecular pathways that mediate enterococcal adaptation, persistence, and pathogenic behavior.
Collapse
Affiliation(s)
- Wiam Abdalla Mo Madani
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, NY 10065, United States
| | - Yusibeska Ramos
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, NY 10065, United States
| | - Juan R Cubillos-Ruiz
- Immunology and Microbial Pathogenesis Program, Weill Cornell Graduate School of Medical Sciences, Weill Cornell Medicine, NY 10065, United States
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, NY 10065, United States
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, NY 10065, United States
| | - Diana K Morales
- Department of Obstetrics and Gynecology, Weill Cornell Medicine, NY 10065, United States
| |
Collapse
|
21
|
Wu M, Cheng Y, Zhang R, Han W, Jiang H, Bi C, Zhang Z, Ye M, Lin X, Liu Z. Molecular mechanism and therapeutic strategy of bile acids in Alzheimer's disease from the emerging perspective of the microbiota-gut-brain axis. Biomed Pharmacother 2024; 178:117228. [PMID: 39088965 DOI: 10.1016/j.biopha.2024.117228] [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: 04/21/2024] [Revised: 07/19/2024] [Accepted: 07/28/2024] [Indexed: 08/03/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the accumulation of amyloid-β outside neurons and Tau protein inside neurons. Various pathological mechanisms are implicated in AD, including brain insulin resistance, neuroinflammation, and endocrinal dysregulation of adrenal corticosteroids. These factors collectively contribute to neuronal damage and destruction. Recently, bile acids (BAs), which are metabolites of cholesterol, have shown neuroprotective potential against AD by targeting the above pathological changes. BAs can enter the systematic circulation and cross the blood-brain barrier, subsequently exerting neuroprotective effects by targeting several endogenous receptors. Additionally, BAs interact with the microbiota-gut-brain (MGB) axis to improve immune and neuroendocrine function during AD episodes. Gut microbes impact BA signaling in the brain through their involvement in BA biotransformation. In this review, we summarize the role and molecular mechanisms of BAs in AD while considering the MGB axis and propose novel strategies for preventing the onset and progression of AD.
Collapse
Affiliation(s)
- Menglu Wu
- Clinical Laboratory, Shaoxing Seventh People's Hospital (Affiliated Mental Health Center, Medical College of Shaoxing University), Shaoxing, Zhejiang, China; Department of Behavioral Neurosciences, Science Research Center of Medical School, Shaoxing University, Shaoxing, Zhejiang, China
| | - Yongyi Cheng
- Department of Behavioral Neurosciences, Science Research Center of Medical School, Shaoxing University, Shaoxing, Zhejiang, China
| | - Ruolin Zhang
- Department of Behavioral Neurosciences, Science Research Center of Medical School, Shaoxing University, Shaoxing, Zhejiang, China
| | - Wenwen Han
- Department of Behavioral Neurosciences, Science Research Center of Medical School, Shaoxing University, Shaoxing, Zhejiang, China
| | - Hanqi Jiang
- Department of Behavioral Neurosciences, Science Research Center of Medical School, Shaoxing University, Shaoxing, Zhejiang, China
| | - Chenchen Bi
- Department of Behavioral Neurosciences, Science Research Center of Medical School, Shaoxing University, Shaoxing, Zhejiang, China
| | - Ziyi Zhang
- Department of Behavioral Neurosciences, Science Research Center of Medical School, Shaoxing University, Shaoxing, Zhejiang, China
| | - Mengfei Ye
- Department of Psychiatry, Shaoxing Seventh People's Hospital (Affiliated Mental Health Center, Medical College of Shaoxing University), Shaoxing, Zhejiang, China
| | - Xiuqin Lin
- Clinical Laboratory, Shaoxing Seventh People's Hospital (Affiliated Mental Health Center, Medical College of Shaoxing University), Shaoxing, Zhejiang, China.
| | - Zheng Liu
- Department of Behavioral Neurosciences, Science Research Center of Medical School, Shaoxing University, Shaoxing, Zhejiang, China; Department of Pharmacology, School of Medicine, Shaoxing University, Shaoxing, Zhejiang, China.
| |
Collapse
|
22
|
Palmiotti A, Berk KA, Koehorst M, Hovingh MV, Pranger AT, van Faassen M, de Boer JF, van der Valk ES, van Rossum EFC, Mulder MT, Kuipers F. Reversal of insulin resistance in people with obesity by lifestyle-induced weight loss does not impact the proportion of circulating 12α-hydroxylated bile acids. Diabetes Obes Metab 2024; 26:4019-4029. [PMID: 38957937 DOI: 10.1111/dom.15754] [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: 03/07/2024] [Revised: 06/06/2024] [Accepted: 06/15/2024] [Indexed: 07/04/2024]
Abstract
AIM Bile acids (BAs) are implicated in the pathogenesis of several metabolic syndrome-related diseases, including insulin resistance (IR) and type 2 diabetes (T2D). It has been reported that IR and T2D are associated with an increased ratio of 12α/non-12α-hydroxylated BAs in the circulating BA pool. It is, however, unknown whether the improvement of insulin sensitivity inversely affects BA composition in humans. Therefore, we assessed whether lifestyle-induced weight loss induces changes in BA metabolism in people with obesity, with or without T2D, and if these changes are associated with metabolic parameters. MATERIALS AND METHODS Individual BAs and C4 were quantified by ultra-high-performance liquid chromatography-tandem mass spectrometry in plasma samples collected from two cohorts of people with obesity (OB) and with T2D and obesity (T2D), before and after a lifestyle intervention. RESULTS Lifestyle-induced weight loss improved glycaemic control in both cohorts, with plasma BA concentrations not affected by the lifestyle interventions. The ratio of 12α/non-12α-hydroxylated BAs remained unchanged in OB (p = .178) and even slightly increased upon intervention in T2D (p = .0147). Plasma C4 levels were unaffected in OB participants (p = .20) but significantly reduced in T2D after intervention (p = .0003). There were no significant correlations between the ratio of 12α/non-12α-hydroxylated BAs and glucose, insulin, or homeostatic model assessment-IR, nor in plasma triglycerides, low-density lipoprotein cholesterol, lipoprotein (a) in the T2D cohort. CONCLUSIONS Lifestyle-induced weight loss did improve glycaemic control but did not affect BA concentrations. Improvements in insulin sensitivity were not associated with changes in BA parameters in people with obesity, with or without T2D.
Collapse
Affiliation(s)
- Anna Palmiotti
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Kirsten A Berk
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Martijn Koehorst
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Milaine V Hovingh
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Alle T Pranger
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Martijn van Faassen
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Jan Freark de Boer
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - Eline S van der Valk
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Internal Medicine, Obesity Centre CGG, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Elisabeth F C van Rossum
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
- Department of Internal Medicine, Obesity Centre CGG, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Monique T Mulder
- Department of Internal Medicine, Division of Endocrinology, Erasmus MC, University Medical Center Rotterdam, Rotterdam, The Netherlands
| | - Folkert Kuipers
- Department of Pediatrics, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Laboratory Medicine, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department for the Biology of Ageing, European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
23
|
Bharti A, Sharma I, Mahajan R, Langer S, Kapoor N. From Cirrhosis to the Dysbiosis (A Loop of Cure or Complications?). Indian J Microbiol 2024; 64:810-820. [PMID: 39282182 PMCID: PMC11399373 DOI: 10.1007/s12088-024-01267-w] [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: 09/28/2023] [Accepted: 03/18/2024] [Indexed: 09/18/2024] Open
Abstract
Gut dysbiosis and liver cirrhosis are two corelated complications that highly disturbs the metabolism of a normal human body. Liver cirrhosis is scarring of the hepatic tissue and gut dysbiosis is the imbalance in the microbiome of the gut. Gut dysbiosis in cirrhosis occurs due to increased permeability of the intestinal membrane which might induce immune responses and damage the normal functioning of the body. Dysbiosis can cause liver damage from cirrhosis and can further lead to liver failure by hepatocellular carcinoma. In this review we discuss if eubiosis can revert the poorly functioning cirrhotic liver to normal functioning state? A normal microbiome converts various liver products into usable forms that regulates the overgrowth of microbiome in the gut. The imbalance caused by dysbiosis retards the normal functioning of liver and increases the complications. To correct this dysbiosis, measures like use of antibiotics with probiotics and prebiotics are used. This correction of the gut microbiome serves as a ray of hope to recover from this chronic illness. In case of alcohol induced liver cirrhosis, intervention of microbes can possibly be helpful in modulating the addiction as well as associated complications like depression as microbes are known to produce and consume neurotransmitters that are involved in alcohol addiction. Hence a correction of gut liver brain axis using microbiome can be a milestone achieved not only for treatment of liver cirrhosis but also for helping alcohol addicts quit and live a healthy or at least a near healthy life.
Collapse
Affiliation(s)
- Aanchal Bharti
- School of Biotechnology, University of Jammu, Jammu, Jammu and Kashmir 180006 India
| | - Isar Sharma
- School of Biotechnology, University of Jammu, Jammu, Jammu and Kashmir 180006 India
| | - Ritu Mahajan
- School of Biotechnology, University of Jammu, Jammu, Jammu and Kashmir 180006 India
| | - Seema Langer
- Department of Zoology, University of Jammu, Jammu, Jammu and Kashmir 180006 India
| | - Nisha Kapoor
- School of Biotechnology, University of Jammu, Jammu, Jammu and Kashmir 180006 India
| |
Collapse
|
24
|
Guo X, Xu J, Lu X, Zheng X, Chen X, Sun Z, Shen B, Tang H, Duan Y, Zhou Z, Feng X, Chen Y, Wang J, Pang J, Jiang Q, Huang B, Gu N, Li J. Chenodeoxycholic Acid-Modified Polyethyleneimine Nano-Composites Deliver Low-Density Lipoprotein Receptor Genes for Lipid-Lowering Therapy by Targeting the Liver. Adv Healthc Mater 2024; 13:e2400254. [PMID: 38857027 DOI: 10.1002/adhm.202400254] [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: 01/25/2024] [Revised: 06/03/2024] [Indexed: 06/11/2024]
Abstract
Lipid-lowering drugs, especially statins, are extensively utilized in clinical settings for the prevention of hyperlipidemia. Nevertheless, prolonged usage of current lipid-lowering medications is associated with significant adverse reactions. Therefore, it is imperative to develop novel therapeutic agents for lipid-lowering therapy. In this study, a chenodeoxycholic acid and lactobionic acid double-modified polyethyleneimine (PDL) nanocomposite as a gene delivery vehicle for lipid-lowering therapy by targeting the liver, are synthesized. Results from the in vitro experiments demonstrate that PDL exhibits superior transfection efficiency compared to polyethyleneimine in alpha mouse liver 12 (AML12) cells and effectively carries plasmids. Moreover, PDL can be internalized by AML12 cells and rapidly escape lysosomal entrapment. Intravenous administration of cyanine5.5 (Cy5.5)-conjugated PDL nanocomposites reveals their preferential accumulation in the liver compared to polyethyleneimine counterparts. Systemic delivery of low-density lipoprotein receptor plasmid-loaded PDL nanocomposites into mice leads to reduced levels of low-density lipoprotein cholesterol (LDL-C) and triglycerides (TC) in the bloodstream without any observed adverse effects on mouse health or well-being. Collectively, these findings suggest that low-density lipoprotein receptor plasmid-loaded PDL nanocomposites hold promise as potential therapeutics for lipid-lowering therapy.
Collapse
Affiliation(s)
- Xiaotang Guo
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, P. R. China
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Jiming Xu
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Xiyuan Lu
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, P. R. China
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Xiaoyan Zheng
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Xi Chen
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210029, P. R. China
- Department of Ophthalmology, Northern Jiangsu People's Hospital, Yangzhou, 225001, P. R. China
| | - Zhenning Sun
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
| | - Beilei Shen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, P. R. China
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Hao Tang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, P. R. China
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Yiman Duan
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, P. R. China
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Zhengwei Zhou
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, P. R. China
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Xu Feng
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, P. R. China
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Yang Chen
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, P. R. China
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Junjie Wang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, P. R. China
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Jing Pang
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, P. R. China
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Qin Jiang
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210029, P. R. China
| | - Bin Huang
- School of Biomedical Engineering and Informatics, Nanjing Medical University, Nanjing, 211166, P. R. China
| | - Ning Gu
- Medical School, Nanjing University, Nanjing, 210093, P. R. China
| | - Juxue Li
- State Key Laboratory of Reproductive Medicine and Offspring Health, Nanjing Medical University, Nanjing, 211166, P. R. China
- Key Laboratory of Human Functional Genomics of Jiangsu Province, Nanjing Medical University, Nanjing, 211166, P. R. China
- School of Biological Science and Medical Engineering, Southeast University, Nanjing, 210096, P. R. China
- The Affiliated Eye Hospital, Nanjing Medical University, Nanjing, 210029, P. R. China
- The Second Affiliated Hospital of Nanjing Medical University, Nanjing, 210011, P. R. China
| |
Collapse
|
25
|
Cao C, Yue S, Lu A, Liang C. Host-Gut Microbiota Metabolic Interactions and Their Role in Precision Diagnosis and Treatment of Gastrointestinal Cancers. Pharmacol Res 2024; 207:107321. [PMID: 39038631 DOI: 10.1016/j.phrs.2024.107321] [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: 02/24/2024] [Revised: 06/30/2024] [Accepted: 07/17/2024] [Indexed: 07/24/2024]
Abstract
The critical role of the gut microbiome in gastrointestinal cancers is becoming increasingly clear. Imbalances in the gut microbial community, referred to as dysbiosis, are linked to increased risks for various forms of gastrointestinal cancers. Pathogens like Fusobacterium and Helicobacter pylori relate to the onset of esophageal and gastric cancers, respectively, while microbes such as Porphyromonas gingivalis and Clostridium species have been associated with a higher risk of pancreatic cancer. In colorectal cancer, bacteria such as Fusobacterium nucleatum are known to stimulate the growth of tumor cells and trigger cancer-promoting pathways. On the other hand, beneficial microbes like Bifidobacteria offer a protective effect, potentially inhibiting the development of gastrointestinal cancers. The potential for therapeutic interventions that manipulate the gut microbiome is substantial, including strategies to engineer anti-tumor metabolites and employ microbiota-based treatments. Despite the progress in understanding the influence of the microbiome on gastrointestinal cancers, significant challenges remain in identifying and understanding the precise contributions of specific microbial species and their metabolic products. This knowledge is essential for leveraging the role of the gut microbiome in the development of precise diagnostics and targeted therapies for gastrointestinal cancers.
Collapse
Affiliation(s)
- Chunhao Cao
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, 999077, Hong Kong Special Administrative Region of China
| | - Siran Yue
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, 999077, Hong Kong Special Administrative Region of China
| | - Aiping Lu
- Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, 999077, Hong Kong Special Administrative Region of China; Guangdong-Hong Kong-Macau Joint Lab on Chinese Medicine and Immune Disease Research, Guangzhou 510006, China; Shanghai University of Traditional Chinese Medicine, Shanghai 200032, China.
| | - Chao Liang
- Department of Systems Biology, School of Life Sciences, Southern University of Science and Technology, Shenzhen 518055, China; Institute of Integrated Bioinfomedicine and Translational Science (IBTS), School of Chinese Medicine, Hong Kong Baptist University, 999077, Hong Kong Special Administrative Region of China; State Key Laboratory of Proteomics, National Center for Protein Sciences (Beijing), Beijing Institute of Lifeomics, Beijing 100850, China.
| |
Collapse
|
26
|
Shealy NG, Baltagulov M, Byndloss MX. A long journey to the colon: The role of the small intestine microbiota in intestinal disease. Mol Microbiol 2024; 122:304-312. [PMID: 38690771 PMCID: PMC11499051 DOI: 10.1111/mmi.15270] [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: 03/03/2024] [Revised: 04/09/2024] [Accepted: 04/17/2024] [Indexed: 05/03/2024]
Abstract
The small intestine represents a complex and understudied gut niche with significant implications for human health. Indeed, many infectious and non-infectious diseases center within the small intestine and present similar clinical manifestations to large intestinal disease, complicating non-invasive diagnosis and treatment. One major neglected aspect of small intestinal diseases is the feedback relationship with the resident collection of commensal organisms, the gut microbiota. Studies focused on microbiota-host interactions in the small intestine in the context of infectious and non-infectious diseases are required to identify potential therapeutic targets dissimilar from those used for large bowel diseases. While sparsely populated, the small intestine represents a stringent commensal bacterial microenvironment the host relies upon for nutrient acquisition and protection against invading pathogens (colonization resistance). Indeed, recent evidence suggests that disruptions to host-microbiota interactions in the small intestine impact enteric bacterial pathogenesis and susceptibility to non-infectious enteric diseases. In this review, we focus on the microbiota's impact on small intestine function and the pathogenesis of infectious and non-infectious diseases of the gastrointestinal (GI) tract. We also discuss gaps in knowledge on the role of commensal microorganisms in proximal GI tract function during health and disease.
Collapse
Affiliation(s)
- Nicolas G. Shealy
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, U.S.A
| | - Madi Baltagulov
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, U.S.A
| | - Mariana X. Byndloss
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN 37232, U.S.A
- Howard Hughes Medical Institute, Vanderbilt University Medical Center, Nashville, TN 37232, U.S.A
- Vanderbilt Institute of Infection, Immunology, and Inflammation, Vanderbilt University Medical Center, Nashville, TN 37232, U.S.A
- Vanderbilt Microbiome Innovation Center, Vanderbilt University, Nashville, TN 37235, U.S.A
| |
Collapse
|
27
|
He S, Li L, Yao Y, Su J, Lei S, Zhang Y, Zeng H. Bile acid and its bidirectional interactions with gut microbiota: a review. Crit Rev Microbiol 2024; 50:684-701. [PMID: 37766478 DOI: 10.1080/1040841x.2023.2262020] [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: 06/27/2023] [Revised: 09/14/2023] [Accepted: 09/18/2023] [Indexed: 09/29/2023]
Abstract
Bile acids (BAs) are an important metabolite produced by cholesterol catabolism. It serves important roles in glucose and lipid metabolism and host-microbe interaction. Recent research has shown that different gut-microbiota can secrete different metabolic-enzymes to mediate the deconjugation, dehydroxylation and epimerization of BAs. In addition, microbes mediate BAs transformation and exert physiological functions in metabolic diseases may have a potentially close relationship with diet. Therefore, elaborating the pathways by which gut microbes mediate the transformation of BAs through enzymatic reactions involved are principal to understand the mechanism of effects between dietary patterns, gut microbes and BAs, and to provide theoretical knowledge for the development of functional foods to regulate metabolic diseases. In the present review, we summarized works on the physiological function of BAs, as well as the classification and composition of BAs in different animal models and its organs. In addition, we mainly focus on the bidirectional interactions of gut microbes with BAs transformation, and discuss the effects of diet on microbial transformation of BAs. Finally, we raised the question of further in-depth investigation of the food-gut microbial-BAs relationship, which might contribute to the improvement of metabolic diseases through dietary interventions in the future.
Collapse
Affiliation(s)
- Shuqi He
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
- Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Lanxin Li
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
- Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yingning Yao
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
- Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Jinhan Su
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
- Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Suzhen Lei
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
- Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Yi Zhang
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
- Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou, China
- Engineering Research Centre of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fujian Agriculture and Forestry University, Fuzhou, China
| | - Hongliang Zeng
- College of Food Science, Fujian Agriculture and Forestry University, Fuzhou, China
- Fujian Provincial Key Laboratory of Quality Science and Processing Technology in Special Starch, Fujian Agriculture and Forestry University, Fuzhou, China
- Engineering Research Centre of Fujian-Taiwan Special Marine Food Processing and Nutrition, Ministry of Education, Fujian Agriculture and Forestry University, Fuzhou, China
| |
Collapse
|
28
|
Gharib E, Robichaud GA. From Crypts to Cancer: A Holistic Perspective on Colorectal Carcinogenesis and Therapeutic Strategies. Int J Mol Sci 2024; 25:9463. [PMID: 39273409 PMCID: PMC11395697 DOI: 10.3390/ijms25179463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/19/2024] [Accepted: 08/24/2024] [Indexed: 09/15/2024] Open
Abstract
Colorectal cancer (CRC) represents a significant global health burden, with high incidence and mortality rates worldwide. Recent progress in research highlights the distinct clinical and molecular characteristics of colon versus rectal cancers, underscoring tumor location's importance in treatment approaches. This article provides a comprehensive review of our current understanding of CRC epidemiology, risk factors, molecular pathogenesis, and management strategies. We also present the intricate cellular architecture of colonic crypts and their roles in intestinal homeostasis. Colorectal carcinogenesis multistep processes are also described, covering the conventional adenoma-carcinoma sequence, alternative serrated pathways, and the influential Vogelstein model, which proposes sequential APC, KRAS, and TP53 alterations as drivers. The consensus molecular CRC subtypes (CMS1-CMS4) are examined, shedding light on disease heterogeneity and personalized therapy implications.
Collapse
Affiliation(s)
- Ehsan Gharib
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| | - Gilles A Robichaud
- Département de Chimie et Biochimie, Université de Moncton, Moncton, NB E1A 3E9, Canada
- Atlantic Cancer Research Institute, Moncton, NB E1C 8X3, Canada
| |
Collapse
|
29
|
Dong X, Xiao B, Vu H, Lin H, Sitti M. Millimeter-scale soft capsules for sampling liquids in fluid-filled confined spaces. SCIENCE ADVANCES 2024; 10:eadp2758. [PMID: 39196937 PMCID: PMC11352903 DOI: 10.1126/sciadv.adp2758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/16/2024] [Accepted: 07/23/2024] [Indexed: 08/30/2024]
Abstract
Sampling liquids in small and confined spaces to retrieve chemicals and microbiomes could enable minimally invasive monitoring human physiological conditions for understanding disease development and allowing early screening. However, existing tools are either invasive or too large for sampling liquids in tortuous and narrow spaces. Here we report a fundamental liquid sampling mechanism that enables millimeter-scale soft capsules for sampling liquids in confined spaces. The miniature capsule is enabled by flexible magnetic valves and superabsorbent polymer, fully wirelessly controlled for on-demand fluid sampling. A group of miniature capsules could navigate in fluid-filled and confined spaces safely using a rolling locomotion. The integration of on-demand triggering, sampling, and sealing mechanism and the agile group locomotion allows us to demonstrate precise control of the soft capsules, navigating and sampling body fluids in a phantom and animal organ ex vivo, guided by ultrasound and x-ray medical imaging. The proposed mechanism and wirelessly controlled devices spur the next-generation technologies for minimally invasive disease diagnosis.
Collapse
Affiliation(s)
- Xiaoguang Dong
- Department of Mechanical Engineering, Vanderbilt University, Nashville, TN 37235, USA
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569 Stuttgart, Germany
| | - Boyang Xiao
- Department of Mechanical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Hieu Vu
- Department of Mechanical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Honglu Lin
- Department of Mechanical Engineering, Vanderbilt University, Nashville, TN 37235, USA
| | - Metin Sitti
- Physical Intelligence Department, Max Planck Institute for Intelligent Systems, 70569 Stuttgart, Germany
- School of Medicine and College of Engineering, Koç University, 34450 Istanbul, Turkey
| |
Collapse
|
30
|
Beekman CN, Penumutchu S, Peterson R, Han G, Belenky M, Hasan MH, Belenky A, Beura LK, Belenky P. Spatial analysis of murine microbiota and bile acid metabolism during amoxicillin treatment. Cell Rep 2024; 43:114572. [PMID: 39116202 DOI: 10.1016/j.celrep.2024.114572] [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: 01/30/2024] [Revised: 06/05/2024] [Accepted: 07/17/2024] [Indexed: 08/10/2024] Open
Abstract
Antibiotics cause collateral damage to resident microbes that is associated with various health risks. To date, studies have largely focused on the impacts of antibiotics on large intestinal and fecal microbiota. Here, we employ a gastrointestinal (GI) tract-wide integrated multiomic approach to show that amoxicillin (AMX) treatment reduces bacterial abundance, bile salt hydrolase activity, and unconjugated bile acids in the small intestine (SI). Losses of fatty acids (FAs) and increases in acylcarnitines in the large intestine (LI) correspond with spatially distinct expansions of Proteobacteria. Parasutterella excrementihominis engage in FA biosynthesis in the SI, while multiple Klebsiella species employ FA oxidation during expansion in the LI. We subsequently demonstrate that restoration of unconjugated bile acids can mitigate losses of commensals in the LI while also inhibiting the expansion of Proteobacteria during AMX treatment. These results suggest that the depletion of bile acids and lipids may contribute to AMX-induced dysbiosis in the lower GI tract.
Collapse
Affiliation(s)
- Chapman N Beekman
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA.
| | - Swathi Penumutchu
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Rachel Peterson
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Geongoo Han
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Marina Belenky
- Felicitex Therapeutics Inc., 27 Strathmore Road, Natick, MA 01760, USA
| | - Mohammad H Hasan
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Alexei Belenky
- Felicitex Therapeutics Inc., 27 Strathmore Road, Natick, MA 01760, USA
| | - Lalit K Beura
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Peter Belenky
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
31
|
Wang W, Gu W, Schweitzer R, Koren O, Khatib S, Tseng G, Konnikova L. In utero human intestine contains maternally derived bacterial metabolites. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.20.608888. [PMID: 39229010 PMCID: PMC11370329 DOI: 10.1101/2024.08.20.608888] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/05/2024]
Abstract
Understanding when host-microbiome interactions are first established is crucial for comprehending normal development and identifying disease prevention strategies. Furthermore, bacterially derived metabolites play critical roles in shaping the intestinal immune system. Recent studies have demonstrated that memory T cells infiltrate human intestinal tissue early in the second trimester, suggesting that intestinal immune education begins in utero. Our previous study reported a unique fetal intestinal metabolomic profile with an abundance of several bacterially derived metabolites and aryl hydrocarbon receptor (AHR) ligands implicated in mucosal immune regulation. To follow up on this work, in the current study, we demonstrate that a number of microbial byproducts present in fetal intestines in utero are maternally derived and vertically transmitted to the fetus. Notably, these bacterially derived metabolites, particularly short chain fatty acids and secondary bile acids, are likely biologically active and functional in regulating the fetal immune system and preparing the gastrointestinal tract for postnatal microbial encounters, as the transcripts for their various receptors and carrier proteins are present in second trimester intestinal tissue through single-cell transcriptomic data.
Collapse
Affiliation(s)
- Wenjia Wang
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Weihong Gu
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
| | - Ron Schweitzer
- Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
- Department of Biotechnology, Tel-Hai College, Upper Galilee, Israel
- Department of Natural Compounds and Analytical Chemistry, Migal Galilee Research Institute, Kiryat Shmona, Israel
| | - Omry Koren
- Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Soliman Khatib
- Department of Biotechnology, Tel-Hai College, Upper Galilee, Israel
- Department of Natural Compounds and Analytical Chemistry, Migal Galilee Research Institute, Kiryat Shmona, Israel
| | - George Tseng
- Department of Biostatistics, University of Pittsburgh, Pittsburgh, PA, USA
| | - Liza Konnikova
- Department of Pediatrics, Yale School of Medicine, New Haven, CT, USA
- Departments of Immunobiology, Yale School of Medicine, New Haven, CT, USA
- Departments of Obstetrics, Gynecology, and Reproductive Sciences, Yale School of Medicine, New Haven, CT, USA
- Program in Translational Biomedicine, Yale School of Medicine, New Haven, CT, USA
- Human Translational Immunology Program, Yale School of Medicine, New Haven, CT, USA
- Center for Systems and Engineering Immunology, Yale School of Medicine, New Haven, CT, USA
| |
Collapse
|
32
|
Attaye I, Witjes JJ, Koopen AM, van der Vossen EW, Zwirs D, Wortelboer K, Collard D, Kemper EM, Winkelmeijer M, Holst JJ, Hazen SL, Kuipers F, Stroes ES, Groen AK, de Vos WM, Nieuwdorp M, Herrema H. Oral Anaerobutyricum soehngenii augments glycemic control in type 2 diabetes. iScience 2024; 27:110455. [PMID: 39139405 PMCID: PMC11321313 DOI: 10.1016/j.isci.2024.110455] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2023] [Revised: 11/21/2023] [Accepted: 07/02/2024] [Indexed: 08/15/2024] Open
Abstract
This randomized, double-blind, placebo-controlled trial investigated the impact of 14-day Anaerobutyricum soehngenii L2-7 supplementation on postprandial glucose levels in 25 White Dutch males with type 2 diabetes (T2D) on stable metformin therapy. The primary endpoint was the effect of A. soehngenii versus placebo on glucose excursions and variability as determined by continuous glucose monitoring. Secondary endpoints were changes in ambulatory 24-h blood pressure, incretins, circulating metabolites and excursions of plasma short-chain fatty acids (SCFAs) and bile acids upon a standardized meal. Results showed that A. soehngenii supplementation for 14 days significantly improved glycemic variability and mean arterial blood pressure, without notable changes in SCFAs, bile acids, incretin levels, or anthropometric parameters as compared to placebo-treated controls. Although well-tolerated and effective in improving glycemic control in the intervention group, further research in larger and more diverse populations is needed to generalize these findings.
Collapse
Affiliation(s)
- Ilias Attaye
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Diabetes & Metabolism, Amsterdam, the Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Endocrinology, Metabolism and Nutrition, Amsterdam, the Netherlands
| | - Julia J. Witjes
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Diabetes & Metabolism, Amsterdam, the Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Endocrinology, Metabolism and Nutrition, Amsterdam, the Netherlands
| | - Annefleur M. Koopen
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Diabetes & Metabolism, Amsterdam, the Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Endocrinology, Metabolism and Nutrition, Amsterdam, the Netherlands
| | | | - Diona Zwirs
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Koen Wortelboer
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Diabetes & Metabolism, Amsterdam, the Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Endocrinology, Metabolism and Nutrition, Amsterdam, the Netherlands
| | - Didier Collard
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Elles Marleen Kemper
- Department of Pharmacy and Clinical Pharmacology, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Maaike Winkelmeijer
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Jens J. Holst
- NNF Center for Basic Metabolic Research and Department of Biomedical Sciences, Copenhagen University, Copenhagen, Denmark
| | - Stanley L. Hazen
- Department of Cardiovascular and Metabolic Sciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Folkert Kuipers
- Department of Pediatrics and European Research Institute for the Biology of Ageing (ERIBA), University of Groningen, University Medical Center Groningen, Groningen, the Netherlands
| | - Erik S.G. Stroes
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Albert K. Groen
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Willem M. de Vos
- Wageningen University, Wageningen, the Netherlands
- Human Microbiome Research Program, Faculty of Medicine, University of Helsinki, Helsinki, Finland
| | - Max Nieuwdorp
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| | - Hilde Herrema
- Department of Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands
- Amsterdam Cardiovascular Sciences, Diabetes & Metabolism, Amsterdam, the Netherlands
- Amsterdam Gastroenterology Endocrinology Metabolism, Endocrinology, Metabolism and Nutrition, Amsterdam, the Netherlands
- Department of Experimental Vascular Medicine, Amsterdam University Medical Centers, Amsterdam, the Netherlands
| |
Collapse
|
33
|
Hossain A, Gnanagobal H, Cao T, Chakraborty S, Chukwu-Osazuwa J, Soto-Dávila M, Vasquez I, Santander J. Role of cold shock proteins B and D in Aeromonas salmonicida subsp. salmonicida physiology and virulence in lumpfish ( Cyclopterus lumpus). Infect Immun 2024; 92:e0001124. [PMID: 38920386 PMCID: PMC11320987 DOI: 10.1128/iai.00011-24] [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: 01/08/2024] [Accepted: 06/05/2024] [Indexed: 06/27/2024] Open
Abstract
Cold shock proteins (Csp) are pivotal nucleic acid binding proteins known for their crucial roles in the physiology and virulence of various bacterial pathogens affecting plant, insect, and mammalian hosts. However, their significance in bacterial pathogens of teleost fish remains unexplored. Aeromonas salmonicida subsp. salmonicida (hereafter A. salmonicida) is a psychrotrophic pathogen and the causative agent of furunculosis in marine and freshwater fish. Four csp genes (cspB, cspD, cspA, and cspC) have been identified in the genome of A. salmonicida J223 (wild type). Here, we evaluated the role of DNA binding proteins, CspB and CspD, in A. salmonicida physiology and virulence in lumpfish (Cyclopterus lumpus). A. salmonicida ΔcspB, ΔcspD, and the double ΔcspBΔcspD mutants were constructed and characterized. A. salmonicida ΔcspB and ΔcspBΔcspD mutants showed a faster growth at 28°C, and reduced virulence in lumpfish. A. salmonicida ΔcspD showed a slower growth at 28°C, biofilm formation, lower survival in low temperatures and freezing conditions (-20°C, 0°C, and 4°C), deficient in lipopolysaccharide synthesis, and low virulence in lumpfish. Additionally, ΔcspBΔcspD mutants showed less survival in the presence of bile compared to the wild type. Transcriptome analysis revealed that 200, 37, and 921 genes were differentially expressed in ΔcspB, ΔcspD, and ΔcspBΔcspD, respectively. In ΔcspB and ΔcspBΔcspD virulence genes in the chromosome and virulence plasmid were downregulated. Our analysis indicates that CspB and CspD mostly act as a transcriptional activator, influencing cell division (e.g., treB), virulence factors (e.g., aexT), and ultimately virulence.
Collapse
Affiliation(s)
- Ahmed Hossain
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, Ocean Science Center, St. John's, Newfoundland, Canada
| | - Hajarooba Gnanagobal
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, Ocean Science Center, St. John's, Newfoundland, Canada
| | - Trung Cao
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, Ocean Science Center, St. John's, Newfoundland, Canada
| | - Setu Chakraborty
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, Ocean Science Center, St. John's, Newfoundland, Canada
| | - Joy Chukwu-Osazuwa
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, Ocean Science Center, St. John's, Newfoundland, Canada
| | - Manuel Soto-Dávila
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, Ocean Science Center, St. John's, Newfoundland, Canada
| | - Ignacio Vasquez
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, Ocean Science Center, St. John's, Newfoundland, Canada
| | - Javier Santander
- Marine Microbial Pathogenesis and Vaccinology Laboratory, Department of Ocean Sciences, Memorial University of Newfoundland, Ocean Science Center, St. John's, Newfoundland, Canada
| |
Collapse
|
34
|
Dinat S, Orchard A, Van Vuuren S. Antimicrobial activity of Southern African medicinal plants on Helicobacter pylori and Lactobacillus species. JOURNAL OF ETHNOPHARMACOLOGY 2024; 330:118238. [PMID: 38663780 DOI: 10.1016/j.jep.2024.118238] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 04/07/2024] [Accepted: 04/20/2024] [Indexed: 05/02/2024]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Numerous medicinal plants have been used traditionally in South Africa for gastric ulcer treatment. Helicobacter pylori is known for causing inflammation and the onset of gastric ulcers. While several studies explored medicinal plants against H. pylori, investigation of medicinal plants used for gastric ulcers has been neglected, as well as the effects these plants would have on bacteria occurring naturally in the gut microbiome. AIM OF THE STUDY This study aimed to investigate Southern African medicinal plants used traditionally for treating gastric ulcers against H. pylori , as well as the effects that these plants have when combined with Lactobacillus species and tested against H. pylori. METHODOLOGY Based on evidence from the ethnobotanical literature, 21 plants were collected. Their antimicrobial activity was assessed against five clinical H. pylori strains, and in combination with each of three Lactobacillus species, using the minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) broth microdilution assays. Toxicity was assessed using the brine shrimp lethality assay. RESULTS Noteworthy activity was observed against at least one H. pylori strain for 12 plant species. The lowest mean MICs were from organic extracts of Carissa edulis Vahl (0.18 mg/mL) and Chironia baccifera L. (0.20 mg/mL), and aqueous extracts of Sansevieria hyacinthoides (L.) Druce (0.26 mg/mL) and Dodonaea viscosa Jacq. (0.30 mg/mL). Aqueous extracts of the investigated plants were combined with Lactobacillus species, and the majority of combinations showed increased antimicrobial activity compared with the extracts alone. Combinations of Lactobacillus rhamnosus with 18 of the 21 aqueous plant extracts showed at least a two-fold decrease in the mean MBC against all H. pylori strains tested. Lactobacillus acidophilus combined with either Protea repens L., Carpobrotus edulis (L.) L. Bolus or Warburgia salutaris (Bertol.f.) Chiov. aqueous extracts had the best anti-H. pylori activity (mean MBCs of 0.10 mg/mL for each combination). Only four organic and one aqueous extract(s) were considered toxic. CONCLUSION These results highlight the potential of medicinal plants to inhibit H. pylori growth and their role in traditional treatments for the management of ulcers. The results also indicate that aqueous extracts of these plants do not hinder the growth of bacteria that occur naturally in the gut microbiome and play a role in maintaining gut health, as well as show the potential benefit of including Lactobacillus species as potentiators of H. pylori activity.
Collapse
Affiliation(s)
- S Dinat
- Department of Pharmacy and Pharmacology, University of the Witwatersrand, Johannesburg, South Africa
| | - A Orchard
- Department of Pharmacy and Pharmacology, University of the Witwatersrand, Johannesburg, South Africa
| | - S Van Vuuren
- Department of Pharmacy and Pharmacology, University of the Witwatersrand, Johannesburg, South Africa.
| |
Collapse
|
35
|
Sum R, Lim SJM, Sundaresan A, Samanta S, Swaminathan M, Low W, Ayyappan M, Lim TW, Choo MD, Huang GJ, Cheong I. Clostridium septicum manifests a bile salt germinant response mediated by Clostridioides difficile csp gene orthologs. Commun Biol 2024; 7:947. [PMID: 39103440 PMCID: PMC11300598 DOI: 10.1038/s42003-024-06617-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2023] [Accepted: 07/23/2024] [Indexed: 08/07/2024] Open
Abstract
Clostridium septicum infections are highly predictive of certain malignancies in human patients. To initiate infections, C. septicum spores must first germinate and regain vegetative growth. Yet, what triggers the germination of C. septicum spores is still unknown. Here, we observe that C. septicum germinates in response to specific bile salts. Putative bile salt recognition genes are identified in C. septicum based on their similarity in sequence and organization to bile salt-responsive csp genes in Clostridioides difficile. Inactivating two of these csp orthologs (cspC-82 and cspC-1718) results in mutant spores that no longer germinate in the presence of their respective cognate bile salts. Additionally, inactivating the putative cspBA or sleC genes in C. septicum abrogates the germination response to all bile salt germinants, suggesting that both act at a convergent point downstream of cspC-82 and cspC-1718. Molecular dynamics simulations show that both CspC-82 and CspC-1718 bear a strong structural congruence with C. difficile's CspC. The existence of functional bile salt germination sensors in C. septicum may be relevant to the association between infection and malignancy.
Collapse
Affiliation(s)
- Rongji Sum
- Temasek Life Sciences Laboratory, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Sylvester Jian Ming Lim
- Temasek Life Sciences Laboratory, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | - Ajitha Sundaresan
- Temasek Life Sciences Laboratory, Singapore, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore
| | | | | | - Wayne Low
- Temasek Life Sciences Laboratory, Singapore, Singapore
| | - Madhumitha Ayyappan
- Temasek Life Sciences Laboratory, Singapore, Singapore
- NUS High School of Mathematics and Sciences, Singapore, Singapore
| | - Ting Wei Lim
- Temasek Life Sciences Laboratory, Singapore, Singapore
- NUS High School of Mathematics and Sciences, Singapore, Singapore
| | - Marvin Dragon Choo
- Temasek Life Sciences Laboratory, Singapore, Singapore
- NUS High School of Mathematics and Sciences, Singapore, Singapore
| | | | - Ian Cheong
- Temasek Life Sciences Laboratory, Singapore, Singapore.
- Department of Biological Sciences, National University of Singapore, Singapore, Singapore.
| |
Collapse
|
36
|
Hsiao YC, Yang Y, Liu CW, Peng J, Feng J, Zhao H, Teitelbaum T, Lu K. Multiomics to Characterize the Molecular Events Underlying Impaired Glucose Tolerance in FXR-Knockout Mice. J Proteome Res 2024; 23:3332-3341. [PMID: 38967328 DOI: 10.1021/acs.jproteome.3c00475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 07/06/2024]
Abstract
The prevalence of different metabolic syndromes has grown globally, and the farnesoid X receptor (FXR), a metabolic homeostat for glucose, lipid, and bile acid metabolisms, may serve an important role in the progression of metabolic disorders. Glucose intolerance by FXR deficiency was previously reported and observed in our study, but the underlying biology remained unclear. To investigate the ambiguity, we collected the nontargeted profiles of the fecal metaproteome, serum metabolome, and liver proteome in Fxr-null (Fxr-/-) and wild-type (WT) mice with LC-HRMS. FXR deficiency showed a global impact on the different molecular levels we monitored, suggesting its serious disruption in the gut microbiota, hepatic metabolism, and circulating biomolecules. The network and enrichment analyses of the dysregulated metabolites and proteins suggested the perturbation of carbohydrate and lipid metabolism by FXR deficiency. Fxr-/- mice presented lower levels of hepatic proteins involved in glycogenesis. The impairment of glycogenesis by an FXR deficiency may leave glucose to accumulate in the circulation, which may deteriorate glucose tolerance. Lipid metabolism was dysregulated by FXR deficiency in a structural-dependent manner. Fatty acid β-oxidations were alleviated, but cholesterol metabolism was promoted by an FXR deficiency. Together, we explored the molecular events associated with glucose intolerance by impaired FXR with integrated novel multiomic data.
Collapse
Affiliation(s)
- Yun-Chung Hsiao
- Department of Environmental Sciences and Engineering, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Yifei Yang
- Department of Environmental Sciences and Engineering, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Chih-Wei Liu
- Department of Environmental Sciences and Engineering, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Jingya Peng
- Department of Environmental Sciences and Engineering, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Jiahao Feng
- Department of Environmental Sciences and Engineering, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Haoduo Zhao
- Department of Environmental Sciences and Engineering, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Taylor Teitelbaum
- Department of Environmental Sciences and Engineering, University of North Carolina, Chapel Hill, North Carolina 27599, United States
- Department of Chemistry, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| | - Kun Lu
- Department of Environmental Sciences and Engineering, University of North Carolina, Chapel Hill, North Carolina 27599, United States
| |
Collapse
|
37
|
Machado F, Gómez-Domínguez I, Hurtado-Ribeira R, Martin D, Coimbra MA, Del Castillo MD, Coreta-Gomes F. In vitro human colonic fermentation of coffee arabinogalactan and melanoidin-rich fractions. Int J Biol Macromol 2024; 275:133740. [PMID: 38986986 DOI: 10.1016/j.ijbiomac.2024.133740] [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: 05/24/2024] [Revised: 06/28/2024] [Accepted: 07/06/2024] [Indexed: 07/12/2024]
Abstract
Coffee beverage is a source of dietary fiber composed by arabinogalactans, which can also be associated to proteins and phenolic compounds, originating melanoidins. Human colonic in vitro fermentations of coffee fractions, one rich in melanoidins (Mel) and the other in its parental polysaccharide arabinogalactans (AG), were performed in order to evaluate the metabolites produced by microbiota, namely short-chain fatty acids (SCFA), phenolic compounds, and bile acids. After 48 h of fermentation, a higher fermentability of the carbohydrate fraction of AG (62 %) than that of Mel (27 %) was observed, resulting in a SCFA content of 63 mM and 22 mM, respectively. Supplementation with AG and Mel fractions decreased the acetate:propionate ratio from 4.7 (in the absence of coffee fractions) to 2.5 and 3.5, respectively, suggesting a potential inhibition of HMG-CoA reductase, a rate-limiting enzyme for cholesterol synthesis. The fermentation of coffee fractions yielded dihydroferulic and dihydrocaffeic acids, known to have antioxidant properties. In the presence of Mel, it was observed a decrease (from 0.25 to 0.16 mg/mL) in the production of secondary bile acids, whose high content is associated to the development of several diseases, such as colorectal cancer, neurodegenerative and cardiovascular.
Collapse
Affiliation(s)
- Fernanda Machado
- LAQV-REQUIMTE, Chemistry Department, University of Aveiro, 3810-193 Aveiro, Portugal
| | - Irene Gómez-Domínguez
- Institute of Food Science Research (CIAL) (CSIC-UAM), Calle Nicolás Cabrera Madrid, Spain
| | - Raul Hurtado-Ribeira
- Institute of Food Science Research (CIAL) (CSIC-UAM), Calle Nicolás Cabrera Madrid, Spain
| | - Diana Martin
- Institute of Food Science Research (CIAL) (CSIC-UAM), Calle Nicolás Cabrera Madrid, Spain
| | - Manuel A Coimbra
- LAQV-REQUIMTE, Chemistry Department, University of Aveiro, 3810-193 Aveiro, Portugal
| | | | - Filipe Coreta-Gomes
- LAQV-REQUIMTE, Chemistry Department, University of Aveiro, 3810-193 Aveiro, Portugal; Coimbra Chemistry Centre - Institute of Molecular Sciences (CQC-IMS), University of Coimbra, 3004-535 Coimbra, Portugal.
| |
Collapse
|
38
|
Ismail HM, Perera D, Mandal R, DiMeglio LA, Evans-Molina C, Hannon T, Petrosino J, Javornik Cregeen S, Schmidt NW. Gut microbial changes associated with obesity in youth with type 1 diabetes: a Pilot Study. J Clin Endocrinol Metab 2024:dgae529. [PMID: 39078977 DOI: 10.1210/clinem/dgae529] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 07/11/2024] [Accepted: 07/29/2024] [Indexed: 08/07/2024]
Abstract
CONTEXT Obesity is prevalent in type 1 diabetes (T1D) and is problematic with higher risk for diabetes complications. It is unknown to what extent gut microbiome changes are associated with obesity and T1D. OBJECTIVE To describe the gut microbiome and microbial metabolite changes associated with obesity in T1D. We hypothesized significant gut microbial and metabolite differences in lean T1D youth (BMI: 5-<85%) vs. those with obesity (BMI: ≥95%). METHODS We analyzed stool samples for gut microbial (using metagenomic shotgun sequencing) and short-chain fatty acid (SCFA) differences in lean (n=27) and obese (n=21) T1D youth in a pilot study. The mean±SD age was 15.3±2.2yrs, A1c 7.8±1.3%, diabetes duration 5.1±4.4yrs, 42.0% females, and 94.0% were White. RESULTS Bacterial community composition showed between sample diversity differences (β-diversity) by BMI group (p=0.013). There was a higher ratio of Prevotella to Bacteroides in the obese group (p=0.0058). There was a differential distribution of significantly abundant taxa in either the lean or obese groups, including increased relative abundance of Prevotella copri, among other taxa in the obese group. Functional profiling showed an upregulation of branched chain amino acid (BCAA) biosynthesis in the obese group and upregulation of BCAA degradation, tyrosine metabolism and secondary bile acid biosynthesis in the lean group. Stool SCFAs were higher in the obese versus the lean group (p<0.05 for all). CONCLUSIONS Our findings identify a gut microbiome and microbial metabolite signature associated with obesity in T1D. These findings could help identify gut microbiome targeted therapies to manage obesity in T1D.
Collapse
Affiliation(s)
- Heba M Ismail
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - Dimuthu Perera
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX
| | - Rabindra Mandal
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - Linda A DiMeglio
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | | | - Tamara Hannon
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - Joseph Petrosino
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX
| | - Sara Javornik Cregeen
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX
| | - Nathan W Schmidt
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| |
Collapse
|
39
|
Chakravarty K, Gaur S, Kumar R, Jha NK, Gupta PK. Exploring the Multifaceted Therapeutic Potential of Probiotics: A Review of Current Insights and Applications. Probiotics Antimicrob Proteins 2024:10.1007/s12602-024-10328-x. [PMID: 39069588 DOI: 10.1007/s12602-024-10328-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2024] [Indexed: 07/30/2024]
Abstract
The interplay between human health and the microbiome has gained extensive attention, with probiotics emerging as pivotal therapeutic agents due to their vast potential in treating various health issues. As significant modulators of the gut microbiota, probiotics are crucial in maintaining intestinal homeostasis and enhancing the synthesis of short-chain fatty acids. Despite extensive research over the past decades, there remains an urgent need for a comprehensive and detailed review that encapsulates probiotics' latest insights and applications. This review focusses on the multifaceted roles of probiotics in promoting health and preventing disease, highlighting the complex mechanisms through which these beneficial bacteria influence both gut flora and the human body at large. This paper also explores probiotics' neurological and gastrointestinal applications, focussing on their significant impact on the gut-brain axis and their therapeutic potential in a broad spectrum of pathological conditions. Current innovations in probiotic formulations, mainly focusing on integrating genomics and biotechnological advancements, have also been comprehensively discussed herein. This paper also critically examines the regulatory landscape that governs probiotic use, ensuring safety and efficacy in clinical and dietary settings. By presenting a comprehensive overview of recent studies and emerging trends, this review aims to illuminate probiotics' extensive therapeutic capabilities, leading to future research and clinical applications. However, besides extensive research, further advanced explorations into probiotic interactions and mechanisms will be essential for developing more targeted and effective therapeutic strategies, potentially revolutionizing health care practices for consumers.
Collapse
Affiliation(s)
- Kashyapi Chakravarty
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, Uttar Pradesh, 201309, India
| | - Smriti Gaur
- Department of Biotechnology, Jaypee Institute of Information Technology, Noida, Uttar Pradesh, 201309, India.
| | - Rohit Kumar
- Centre for Development of Biomaterials and Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
| | - Niraj Kumar Jha
- Department of Biotechnology, Sharda School of Engineering and Technology, Sharda University, Greater Noida, Uttar Pradesh, 201310, India
- Centre for Global Health Research, Saveetha Medical College, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, Tamil Nadu, 602105, India
- School of Bioengineering & Biosciences, Lovely Professional University, Phagwara, Punjab, 144411, India
- Centre of Research Impact and Outcome, Chitkara University, Rajpura, Punjab, 140401, India
| | - Piyush Kumar Gupta
- Centre for Development of Biomaterials and Department of Life Sciences, Sharda School of Basic Sciences and Research, Sharda University, Greater Noida, Uttar Pradesh, 201310, India.
- Department of Biotechnology, Graphic Era (Deemed to be University), Dehradun, Uttarakhand, 248002, India.
| |
Collapse
|
40
|
Wang X, Wen Q, Wu H, Peng W, Cai K, Tan Z, Na W, Wu K. Effect of Sex on Intestinal Microbial Metabolites of Hainan Special Wild Boars. Animals (Basel) 2024; 14:2164. [PMID: 39123691 PMCID: PMC11310994 DOI: 10.3390/ani14152164] [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: 05/18/2024] [Revised: 07/01/2024] [Accepted: 07/19/2024] [Indexed: 08/12/2024] Open
Abstract
The intestinal microbiota and its metabolites are essential for the health and growth development of animals. Current research indicates that sex has a certain impact on the structure and function of the intestinal microbiota, but there are few reports on sex differences in intestinal microbiota metabolites, including those of castrated male animals. This study aimed to explore the impact of sex on the intestinal microbial metabolites of Hainan special wild boars (10 entire male pigs, 10 female pigs, and 10 castrated male pigs, denoted EM, FE, and CM, respectively) by employing non-targeted metabolomics and gas chromatography. A total of 1086 metabolites were detected, with the greatest number of differential metabolites observed between EM and FE (54 differential metabolites, including 18 upregulated and 36 downregulated metabolites), the fewest between CM and FE (7 differential metabolites, including 1 upregulated and 6 downregulated metabolites), and an intermediate number between CM and EM (47 differential metabolites, including 35 upregulated and 12 downregulated metabolites). Differential metabolites were involved in more pathways between EM and FE and between CM and EM, including amino acid metabolism and digestive system pathways, whereas differential metabolites were involved in the fewest pathways between CM and FE. Correlation analysis showed Ruminococcaceae UCG-009, uncultured_bacterium_o_SAR324_cladeMarine_group_B, and Candidatus Saccharimonas contributed to the production of metabolites such as trehalose, docosatrienoic acid, D(-)-beta-hydroxy butyric acid, and acetyl-DL-leucine. The levels of acetic acid, propionic acid, butyric acid, isobutyric acid, valeric acid, and isovaleric acid were significantly higher in EM than in FE, with CM falling between the two. Streptococcus, Lachnospiraceae_NK4A136_group and Rikenellaceae_RC9_gut_group showed a significant positive correlation with the production of short-chain fatty acids (SCFAs), while [Eubacterium]_coprostanoligenes_group, uncultured_bacterium_f_p-251-o5 and Ruminococcaceae_UCG-005 showed a significant negative correlation with the generation of SCFAs. This study provides foundational data and significant insights into precision feeding strategies for Hainan special wild boars of different sexes, as well as the study of sex differences in intestinal microbial metabolites in animals.
Collapse
Affiliation(s)
- Xiaozhe Wang
- School of Tropical Agriculture and Forestry, Hainan University, Danzhou 571737, China; (X.W.); (K.W.)
- Sanya Institute, China Agricultural University, Sanya 572024, China
- State Key Laboratory of Animal Nutrition, Department of Companion Animal Science, China Agricultural University, Beijing 100193, China
| | - Qiong Wen
- School of Tropical Agriculture and Forestry, Hainan University, Danzhou 571737, China; (X.W.); (K.W.)
- Wuhan Xiangda Feedstuff Co., Ltd., Wuhan 430045, China
| | - Hongfen Wu
- School of Tropical Agriculture and Forestry, Hainan University, Danzhou 571737, China; (X.W.); (K.W.)
| | - Wenchuan Peng
- School of Tropical Agriculture and Forestry, Hainan University, Danzhou 571737, China; (X.W.); (K.W.)
- Key Laboratory of Animal Genetics, Breeding and Reproduction of Shaanxi Province, College of Animal Science and Technology, Northwest A&F University, Yangling 712100, China
| | - Keqi Cai
- School of Tropical Agriculture and Forestry, Hainan University, Danzhou 571737, China; (X.W.); (K.W.)
- Shenzhen Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Genome Analysis Laboratory of the Ministry of Agriculture and Rural Affairs, Agricultural Genomics Institute at Shenzhen, Chinese Academy of Agricultural Sciences, Shenzhen 518124, China
| | - Zhen Tan
- School of Tropical Agriculture and Forestry, Hainan University, Danzhou 571737, China; (X.W.); (K.W.)
| | - Wei Na
- School of Tropical Agriculture and Forestry, Hainan University, Danzhou 571737, China; (X.W.); (K.W.)
| | - Kebang Wu
- School of Tropical Agriculture and Forestry, Hainan University, Danzhou 571737, China; (X.W.); (K.W.)
| |
Collapse
|
41
|
Gupta S, Biswas P, Das B, Mondal S, Gupta P, Das D, Mallick AI. Selective depletion of Campylobacter jejuni via T6SS dependent functionality: an approach for improving chickens gut health. Gut Pathog 2024; 16:38. [PMID: 38997758 PMCID: PMC11245787 DOI: 10.1186/s13099-024-00628-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 06/28/2024] [Indexed: 07/14/2024] Open
Abstract
The targeted depletion of potential gut pathogens is often challenging because of their intrinsic ability to thrive in harsh gut environments. Earlier, we showed that Campylobacter jejuni (C. jejuni) exclusively uses the Type-VI Secretion System (T6SS) to target its prey such as Escherichia coli (E. coli), and phenotypic differences between T6SS-negative and T6SS-positive C. jejuni isolates toward bile salt sensitivity. However, it remains unclear how the target-driven T6SS functionality prevails in a polymicrobial gut environment. Here, we investigated the fate of microbial competition in an altered gut environment via bacterial T6SS using a T6SS-negative and -positive C. jejuni or its isogenic mutant of the hemolysin-coregulated protein (hcp). We showed that in the presence of bile salt and prey bacteria (E. coli), T6SS-positive C. jejuni experiences enhanced intracellular stress leading to cell death. Intracellular tracking of fluorophore-conjugated bile salts confirmed that T6SS-mediated bile salt influx into C. jejuni can enhance intracellular oxidative stress, affecting C. jejuni viability. We further investigated whether the T6SS activity in the presence of prey (E. coli) perturbs the in vivo colonization of C. jejuni. Using chickens as primary hosts of C. jejuni and non-pathogenic E. coli as prey, we showed a marked reduction of C. jejuni load in chickens cecum when bile salt solution was administered orally. Analysis of local antibody responses and pro-inflammatory gene expression showed a reduced risk of tissue damage, indicating that T6SS activity in the complex gut environment can be exploited as a possible measure to clear the persistent colonization of C. jejuni in chickens.
Collapse
Affiliation(s)
- Subhadeep Gupta
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal, 741246, India
| | - Prakash Biswas
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal, 741246, India
| | - Bishnu Das
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal, 741246, India
| | - Samiran Mondal
- Department of Veterinary Pathology, West Bengal University of Animal and Fishery Sciences, Kolkata, West Bengal, 700037, India
| | - Parna Gupta
- Department of Chemical Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal, 741246, India
| | - Dipjyoti Das
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal, 741246, India.
| | - Amirul Islam Mallick
- Department of Biological Sciences, Indian Institute of Science Education and Research Kolkata, Mohanpur, Nadia, West Bengal, 741246, India.
| |
Collapse
|
42
|
Tabrizi E, Pourteymour Fard Tabrizi F, Mahmoud Khaled G, Sestito MP, Jamie S, Boone BA. Unraveling the gut microbiome's contribution to pancreatic ductal adenocarcinoma: mechanistic insights and therapeutic perspectives. Front Immunol 2024; 15:1434771. [PMID: 39044834 PMCID: PMC11263025 DOI: 10.3389/fimmu.2024.1434771] [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: 05/18/2024] [Accepted: 06/25/2024] [Indexed: 07/25/2024] Open
Abstract
The gut microbiome plays a significant role in the pathogenesis of pancreatic ductal adenocarcinoma (PDAC), influencing oncogenesis, immune responses, and treatment outcomes. Studies have identified microbial species like Porphyromonas gingivalis and Fusobacterium nucleatum, that promote PDAC progression through various mechanisms. Additionally, the gut microbiome affects immune cell activation and response to immunotherapy, including immune checkpoint inhibitors and CAR-T therapy. Specific microbes and their metabolites play a significant role in the effectiveness of immune checkpoint inhibitors (ICIs). Alterations in the gut microbiome can either enhance or diminish responses to PD-1/PD-L1 and CTLA-4 blockade therapy. Additionally, bacterial metabolites like trimethylamine N-oxide (TMAO) and lipopolysaccharide (LPS) impact antitumor immunity, offering potential targets to augment immunotherapy responses. Modulating the microbiome through fecal microbiota transplantation, probiotics, prebiotics, dietary changes, and antibiotics shows promise in PDAC treatment, although outcomes are highly variable. Dietary modifications, particularly high-fiber diets and specific fat consumption, influence microbiome composition and impact cancer risk. Combining microbiome-based therapies with existing treatments holds potential for improving PDAC therapy outcomes, but further research is needed to optimize their effectiveness.
Collapse
Affiliation(s)
- Eileen Tabrizi
- Department of Microbiology, Immunology and Cell Biology, School of Medicine, West Virginia University, Morgantown, WV, United States
- Cancer Institute, West Virginia University, Morgantown, WV, United States
| | - Fatemeh Pourteymour Fard Tabrizi
- Department of Community Nutrition, Faculty of Nutrition and Food Sciences, Tabriz University of Medical Sciences, Tabriz, East Azerbaijan, Iran
| | - Gehad Mahmoud Khaled
- Department of Biotechnology, School of Sciences and Engineering, American University in Cairo, New Cairo, Cairo, Egypt
| | - Michael P. Sestito
- Department of Surgery, West Virginia University School of Medicine, Morgantown, WV, United States
| | - Saeid Jamie
- Department of Neurobiology, David Geffen School of Medicine, University of California, Los Angeles, Los Angeles, CA, United States
| | - Brian A. Boone
- Department of Microbiology, Immunology and Cell Biology, School of Medicine, West Virginia University, Morgantown, WV, United States
- Department of Surgery, West Virginia University School of Medicine, Morgantown, WV, United States
| |
Collapse
|
43
|
Weng H, Deng L, Wang T, Xu H, Wu J, Zhou Q, Yu L, Chen B, Huang L, Qu Y, Zhou L, Chen X. Humid heat environment causes anxiety-like disorder via impairing gut microbiota and bile acid metabolism in mice. Nat Commun 2024; 15:5697. [PMID: 38972900 PMCID: PMC11228019 DOI: 10.1038/s41467-024-49972-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2023] [Accepted: 06/25/2024] [Indexed: 07/09/2024] Open
Abstract
Climate and environmental changes threaten human mental health, but the impacts of specific environmental conditions on neuropsychiatric disorders remain largely unclear. Here, we show the impact of a humid heat environment on the brain and the gut microbiota using a conditioned housing male mouse model. We demonstrate that a humid heat environment can cause anxiety-like behaviour in male mice. Microbial 16 S rRNA sequencing analysis reveals that a humid heat environment caused gut microbiota dysbiosis (e.g., decreased abundance of Lactobacillus murinus), and metabolomics reveals an increase in serum levels of secondary bile acids (e.g., lithocholic acid). Moreover, increased neuroinflammation is indicated by the elevated expression of proinflammatory cytokines in the serum and cortex, activated PI3K/AKT/NF-κB signalling and a microglial response in the cortex. Strikingly, transplantation of the microbiota from mice reared in a humid heat environment readily recapitulates these abnormalities in germ-free mice, and these abnormalities are markedly reversed by Lactobacillus murinus administration. Human samples collected during the humid heat season also show a decrease in Lactobacillus murinus abundance and an increase in the serum lithocholic acid concentration. In conclusion, gut microbiota dysbiosis induced by a humid heat environment drives the progression of anxiety disorders by impairing bile acid metabolism and enhancing neuroinflammation, and probiotic administration is a potential therapeutic strategy for these disorders.
Collapse
Affiliation(s)
- Huandi Weng
- Department of Neurology and Stroke Center, The First Affiliated Hospital & Clinical Neuroscience Institute of Jinan University, Guangzhou, 510632, PR China
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, PR China
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, PR China
| | - Li Deng
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, PR China
| | - Tianyuan Wang
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, PR China
| | - Huachong Xu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, PR China
| | - Jialin Wu
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, PR China
| | - Qinji Zhou
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, PR China
| | - Lingtai Yu
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, PR China
| | - Boli Chen
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, PR China
| | - Li'an Huang
- Department of Neurology and Stroke Center, The First Affiliated Hospital & Clinical Neuroscience Institute of Jinan University, Guangzhou, 510632, PR China
| | - Yibo Qu
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, PR China
| | - Libing Zhou
- Department of Neurology and Stroke Center, The First Affiliated Hospital & Clinical Neuroscience Institute of Jinan University, Guangzhou, 510632, PR China.
- Guangdong-Hongkong-Macau CNS Regeneration Institute of Jinan University, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong Key Laboratory of Non-human Primate Research, Guangzhou, 510632, PR China.
- Co-innovation Center of Neuroregeneration, Nantong University, Nantong, Jiangsu, PR China.
- Neuroscience and Neurorehabilitation Institute, University of Health and Rehabilitation Sciences, Qingdao, 266071, Shandong, PR China.
- Center for Exercise and Brain Science, School of Psychology, Shanghai University of Sport, Shanghai, 200438, PR China.
| | - Xiaoyin Chen
- School of Traditional Chinese Medicine, Jinan University, Guangzhou, 510632, PR China.
| |
Collapse
|
44
|
Vesković M, Pejović M, Šutulović N, Hrnčić D, Rašić-Marković A, Stanojlović O, Mladenović D. Exploring Fibrosis Pathophysiology in Lean and Obese Metabolic-Associated Fatty Liver Disease: An In-Depth Comparison. Int J Mol Sci 2024; 25:7405. [PMID: 39000518 PMCID: PMC11242866 DOI: 10.3390/ijms25137405] [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: 05/30/2024] [Revised: 06/21/2024] [Accepted: 06/28/2024] [Indexed: 07/16/2024] Open
Abstract
While obesity-related nonalcoholic fatty liver disease (NAFLD) is linked with metabolic dysfunctions such as insulin resistance and adipose tissue inflammation, lean NAFLD more often progresses to liver fibrosis even in the absence of metabolic syndrome. This review aims to summarize the current knowledge regarding the mechanisms of liver fibrosis in lean NAFLD. The most commonly used lean NAFLD models include a methionine/choline-deficient (MCD) diet, a high-fat diet with carbon tetrachloride (CCl4), and a high-fructose and high-cholesterol diet. The major pro-fibrogenic mechanisms in lean NAFLD models include increased activation of the extracellular signal-regulated kinase (ERK) pathway, elevated expression of α-smooth muscle actin (α-SMA), collagen type I, and TGF-β, and modulation of fibrogenic markers such as tenascin-X and metalloproteinase inhibitors. Additionally, activation of macrophage signaling pathways promoting hepatic stellate cell (HSC) activation further contributes to fibrosis development. Animal models cannot cover all clinical features that are evident in patients with lean or obese NAFLD, implicating the need for novel models, as well as for deeper comparisons of clinical and experimental studies. Having in mind the prevalence of fibrosis in lean NAFLD patients, by addressing specific pathways, clinical studies can reveal new targeted therapies along with novel biomarkers for early detection and enhancement of clinical management for lean NAFLD patients.
Collapse
Affiliation(s)
- Milena Vesković
- Institute of Pathophysiology, Faculty of Medicine, University of Belgrade, Dr Subotića 9, 11000 Belgrade, Serbia
| | - Milka Pejović
- Primary Health Center “Vračar”, Velimira Bate Živojinovića 16, 11000 Belgrade, Serbia
| | - Nikola Šutulović
- Institute of Medical Physiology, Faculty of Medicine, University of Belgrade, Višegradska 26, 11000 Belgrade, Serbia
| | - Dragan Hrnčić
- Institute of Medical Physiology, Faculty of Medicine, University of Belgrade, Višegradska 26, 11000 Belgrade, Serbia
| | - Aleksandra Rašić-Marković
- Institute of Medical Physiology, Faculty of Medicine, University of Belgrade, Višegradska 26, 11000 Belgrade, Serbia
| | - Olivera Stanojlović
- Institute of Medical Physiology, Faculty of Medicine, University of Belgrade, Višegradska 26, 11000 Belgrade, Serbia
| | - Dušan Mladenović
- Institute of Pathophysiology, Faculty of Medicine, University of Belgrade, Dr Subotića 9, 11000 Belgrade, Serbia
| |
Collapse
|
45
|
Zhou Y, Zhang D, Cheng H, Wu J, Liu J, Feng W, Peng C. Repairing gut barrier by traditional Chinese medicine: roles of gut microbiota. Front Cell Infect Microbiol 2024; 14:1389925. [PMID: 39027133 PMCID: PMC11254640 DOI: 10.3389/fcimb.2024.1389925] [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: 02/22/2024] [Accepted: 06/14/2024] [Indexed: 07/20/2024] Open
Abstract
Gut barrier is not only part of the digestive organ but also an important immunological organ for the hosts. The disruption of gut barrier can lead to various diseases such as obesity and colitis. In recent years, traditional Chinese medicine (TCM) has gained much attention for its rich clinical experiences enriched in thousands of years. After orally taken, TCM can interplay with gut microbiota. On one hand, TCM can modulate the composition and function of gut microbiota. On the other hand, gut microbiota can transform TCM compounds. The gut microbiota metabolites produced during the actions of these interplays exert noticeable pharmacological effects on the host especially gut barrier. Recently, a large number of studies have investigated the repairing and fortifying effects of TCM on gut barriers from the perspective of gut microbiota and its metabolites. However, no review has summarized the mechanism behand this beneficiary effects of TCM. In this review, we first briefly introduce the unique structure and specific function of gut barrier. Then, we summarize the interactions and relationship amidst gut microbiota, gut microbiota metabolites and TCM. Further, we summarize the regulative effects and mechanisms of TCM on gut barrier including physical barrier, chemical barrier, immunological barrier, and microbial barrier. At last, we discuss the effects of TCM on diseases that are associated gut barrier destruction such as ulcerative colitis and type 2 diabetes. Our review can provide insights into TCM, gut barrier and gut microbiota.
Collapse
Affiliation(s)
- Yaochuan Zhou
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Dandan Zhang
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hao Cheng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Jinlu Wu
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Juan Liu
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wuwen Feng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Cheng Peng
- State Key Laboratory of Southwestern Chinese Medicine Resources, School of Pharmacy and School of Basic Medical Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Key Laboratory of the Ministry of Education for Standardization of Chinese Medicine, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
46
|
Santiago P, Quinn KP, Chen J, Friton JJ, Rypstra CR, Kashyap PC, Raffals LE. Altered Bile Acid and Pouch Microbiota Composition in Patients With Chronic Pouchitis. Inflamm Bowel Dis 2024; 30:1062-1070. [PMID: 38037191 PMCID: PMC11219471 DOI: 10.1093/ibd/izad288] [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: 03/17/2023] [Indexed: 12/02/2023]
Abstract
BACKGROUND Patients with ulcerative colitis and total abdominal proctocolectomy with ileal pouch-anal anastomosis have a 50% risk of pouchitis and a 5% to 10% risk of chronic pouchitis. AIMS The goal of the study was to compare pouch microbiota and stool bile acid composition in patients with chronic pouchitis, chronic pouchitis and primary sclerosing cholangitis, and normal pouch. METHODS Patients with ulcerative colitis and ileal pouch-anal anastomosis were recruited from March 20, 2014, to August 6, 2019, and categorized into normal pouch, chronic pouchitis, and chronic pouchitis/primary sclerosing cholangitis groups. Stool samples were subjected to bile acid quantification and 16S rRNA gene sequencing. Statistical comparisons of absolute bile acid abundance and pouch microbiota α-diversity, β-diversity, and taxa abundance were performed among the patient groups. RESULTS A total of 51 samples were analyzed. Both α-diversity (P = .01, species richness) and β-diversity (P = .001) significantly differed among groups. Lithocholic acid was significantly lower in patients with chronic pouchitis/primary sclerosing cholangitis than in those with chronic pouchitis (P = .01) or normal pouch (P = .03). Decreased α-diversity was associated with an increased primary to secondary bile acid ratio (P = .002), which was also associated with changes in β-diversity (P = .006). CONCLUSIONS Pouch microbiota α- and β-diversity differed among patients with normal pouch, chronic pouchitis, and chronic pouchitis/primary sclerosing cholangitis. Lithocholic acid level and primary to secondary bile acid ratio were highly associated with pouch microbiota richness, structure, and composition. These findings emphasize the associations between pouch microbiota and bile acid composition in dysbiosis and altered metabolism, suggesting that secondary bile acids are decreased in chronic pouchitis.
Collapse
Affiliation(s)
- Priscila Santiago
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, United States
| | - Kevin P Quinn
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, United States
| | - Jun Chen
- Division of Computational Biology, Mayo Clinic, Rochester, United States
| | - Jessica J Friton
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, United States
| | - Chad R Rypstra
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, United States
| | - Purna C Kashyap
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, United States
| | - Laura E Raffals
- Division of Gastroenterology and Hepatology, Mayo Clinic, Rochester, United States
| |
Collapse
|
47
|
Fiorucci S, Marchianò S, Urbani G, Di Giorgio C, Distrutti E, Zampella A, Biagioli M. Immunology of bile acids regulated receptors. Prog Lipid Res 2024; 95:101291. [PMID: 39122016 DOI: 10.1016/j.plipres.2024.101291] [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: 06/12/2024] [Revised: 07/30/2024] [Accepted: 08/02/2024] [Indexed: 08/12/2024]
Abstract
Bile acids are steroids formed at the interface of host metabolism and intestinal microbiota. While primary bile acids are generated in the liver from cholesterol metabolism, secondary bile acids represent the products of microbial enzymes. Close to 100 different enzymatic modifications of bile acids structures occur in the human intestine and clinically guided metagenomic and metabolomic analyses have led to the identification of an extraordinary number of novel metabolites. These chemical mediators make an essential contribution to the composition and function of the postbiota, participating to the bidirectional communications of the intestinal microbiota with the host and contributing to the architecture of intestinal-liver and -brain and -endocrine axes. Bile acids exert their function by binding to a group of cell membrane and nuclear receptors collectively known as bile acid-regulated receptors (BARRs), expressed in monocytes, tissue-resident macrophages, CD4+ T effector cells, including Th17, T regulatory cells, dendritic cells and type 3 of intestinal lymphoid cells and NKT cells, highlighting their role in immune regulation. In this review we report on how bile acids and their metabolitesmodulate the immune system in inflammations and cancers and could be exploiting for developing novel therapeutic approaches in these disorders.
Collapse
Affiliation(s)
- Stefano Fiorucci
- Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Italy.
| | - Silvia Marchianò
- Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Italy
| | - Ginevra Urbani
- Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Italy
| | | | - Eleonora Distrutti
- SC di Gastroenterologia ed Epatologia, Azienda Ospedaliera di Perugia, Perugia, Italy
| | - Angela Zampella
- Department of Pharmacy, University of Napoli Federico II, Napoli, Italy
| | - Michele Biagioli
- Dipartimento di Medicina e Chirurgia, Università di Perugia, Perugia, Italy
| |
Collapse
|
48
|
Yang Y, Chi L, Hsiao YC, Lu K. Sex-specific effects of gut microbiome on shaping bile acid metabolism. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.06.27.601003. [PMID: 38979196 PMCID: PMC11230406 DOI: 10.1101/2024.06.27.601003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/10/2024]
Abstract
Gut microbiome is a group of microorganisms that plays important roles in contributing to health and diseases. These bacterial compositions have been demonstrated to impact bile acids (BAs) profiles, either by directly metabolizing primary BAs to secondary BAs or indirect ways through host metabolism by influencing BAs synthesis, transportation and conjugation in liver. It has been observed sexually dimorphic gut microbiome and bile acids composition, with variations in expression levels of bile acid metabolizing genes in the liver. However, associations between sex-specific differences in gut microbiome and BAs profiles are not well understood. This study aimed to investigate whether gut microbiome could influence BAs profiles in host in a sexspecific manner. We transplanted cecum feces of male and female C57BL/6 mice to male mice and measured BAs concentrations in feces, serum and liver samples 7 days after fecal transplantation. We found different BAs profiles between mice with male and female gut microbiome, including altering levels and proportions of secondary BAs. We also observed varied expression levels of genes related to bile acid metabolism in the liver and distal ileum. Our results highlight sex-specific effects of gut microbiome on shaping bile acid metabolism through gut bacteria and regulation of host genes.
Collapse
Affiliation(s)
- Yifei Yang
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, NC, 27599, United States
| | - Liang Chi
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, NC, 27599, United States
| | - Yun-Chung Hsiao
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, NC, 27599, United States
| | - Kun Lu
- Department of Environmental Sciences and Engineering, University of North Carolina at Chapel Hill, NC, 27599, United States
| |
Collapse
|
49
|
Li X, Guo X, Liu Y, Ren F, Li S, Yang X, Liu J, Zhang Z. Antibiotics affect the pharmacokinetics of n-butylphthalide in vivo by altering the intestinal microbiota. PLoS One 2024; 19:e0297713. [PMID: 38917098 PMCID: PMC11198832 DOI: 10.1371/journal.pone.0297713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2024] [Accepted: 05/31/2024] [Indexed: 06/27/2024] Open
Abstract
OBJECTIVE N-butylphthalide (NBP) is a monomeric compound extracted from natural plant celery seeds, whether intestinal microbiota alteration can modify its pharmacokinetics is still unclear. The purpose of this study is to investigate the effect of intestinal microbiota alteration on the pharmacokinetics of NBP and its related mechanisms. METHODS After treatment with antibiotics and probiotics, plasma NBP concentrations in SD rats were determined by high-performance liquid chromatography-tandem mass spectrometry (HPLC-MS/MS). The effect of intestinal microbiota changes on NBP pharmacokinetics was compared. Intestinal microbiota changes after NBP treatment were analyzed by 16S rRNA sequencing. Expressions of CYP3A1 mRNA and protein in the liver and small intestine tissues under different intestinal flora conditions were determined by qRT-PCR and Western Blot. KEGG analysis was used to analyze the effect of intestinal microbiota changes on metabolic pathways. RESULTS Compared to the control group, the values of Cmax, AUC0-8, AUC0-∞, t1/2 in the antibiotic group increased by 56.1% (P<0.001), 56.4% (P<0.001), 53.2% (P<0.001), and 24.4% (P<0.05), respectively. In contrast, the CL and Tmax values decreased by 57.1% (P<0.001) and 28.6% (P<0.05), respectively. Treatment with antibiotics could reduce the richness and diversity of the intestinal microbiota. CYP3A1 mRNA and protein expressions in the small intestine of the antibiotic group were 61.2% and 66.1% of those of the control group, respectively. CYP3A1 mRNA and protein expressions in the liver were 44.6% and 63.9% of those in the control group, respectively. There was no significant change in the probiotic group. KEGG analysis showed that multiple metabolic pathways were significantly down-regulated in the antibiotic group. Among them, the pathways of drug metabolism, bile acid biosynthesis and decomposition, and fatty acid synthesis and decomposition were related to NBP biological metabolism. CONCLUSION Antibiotic treatment could affect the intestinal microbiota, decrease CYP3A1 mRNA and protein expressions and increase NBP exposure in vivo by inhibiting pathways related to NBP metabolism.
Collapse
Affiliation(s)
- Xiangchen Li
- Department of Pharmacy, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Xiaoli Guo
- Department of Pharmacy, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Yixin Liu
- Department of Pharmacy, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Feifei Ren
- Department of Pharmacy, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Shan Li
- Department of Pharmacy, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Xiuling Yang
- Department of Pharmacy, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Jian Liu
- Department of Pharmacy, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| | - Zhiqing Zhang
- Department of Pharmacy, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei Province, China
| |
Collapse
|
50
|
Tang Z, Yang Y, Yang M, Jiang D, Ge Y, Zhang X, Liu H, Fu Q, Liu X, Yang Y, Wu Z, Ji Y. Elucidating the modulatory role of dietary hydroxyproline on the integrity and functional performance of the intestinal barrier in early-weaned piglets: A comprehensive analysis of its interplay with the gut microbiota and metabolites. Int Immunopharmacol 2024; 134:112268. [PMID: 38759371 DOI: 10.1016/j.intimp.2024.112268] [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: 04/11/2024] [Revised: 05/06/2024] [Accepted: 05/13/2024] [Indexed: 05/19/2024]
Abstract
Piglets receive far less hydroxyproline (Hyp) from a diet after weaning than they obtained from sow's milk prior to weaning, suggesting that Hyp may play a protective role in preserving intestinal mucosal homeostasis. This study aimed to evaluate the effect of Hyp on intestinal barrier function and its associated gut microbiota and metabolites in early-weaned piglets. Eighty weaned piglets were divided into four groups and fed diets containing different Hyp levels (0 %, 0.5 %, 1 %, or 2 %) for 21 days. Samples, including intestinal contents, tissues, and blood, were collected on day 7 for analysis of microbial composition, intestinal barrier function, and metabolites. We demonstrated that dietary supplementation with 2 % Hyp improved the feed conversion ratio and reduced the incidence of diarrhea in early-weaned piglets compared to the control group. Concurrently, Hyp enhanced intestinal barrier function by facilitating tight junction protein (zonula occludens (ZO)-1 and occludin) expression and mucin production in the jejunal, ileal, and colonic mucosas. It also improved mucosal immunity (by increasing the amount of secretory IgA (sIgA) and the ratio of CD4+/CD8+ T lymphocytes and decreasing NF-κB phosphorylation) and increased antioxidant capacity (by raising total antioxidant capacity (T-AOC) and glutathione levels) in the intestinal mucosa. In addition, Hyp supplementation resulted in an increase in the levels of glycine, glutathione, and glycine-conjugated bile acids, while decreasing the concentrations of cortisol and methionine sulfoxide in plasma. Intriguingly, piglets fed diet containing Hyp exhibited a remarkable increase in the abundance of probiotic Enterococcus faecium within their colonic contents. This elevation occurred alongside an attenuation of pro-inflammatory responses and an enhancement in intestinal barrier integrity. Further, these changes were accompanied by a rise in anti-inflammatory metabolites, specifically glycochenodeoxycholic acid and guanosine, along with a suppression of pro-inflammatory lipid peroxidation products, including (12Z)-9,10-dihydroxyoctadec-12-enoic acid (9,10-DHOME) and 13-L-hydroperoxylinoleic acid (13(S)-HPODE). In summary, Hyp holds the capacity to enhance the intestinal barrier function in weaned piglets; this effect is correlated with changes in the gut microbiota and metabolites. Our findings provide novel insights into the role of Hyp in maintaining gut homeostasis, highlighting its potential as a dietary supplement for promoting intestinal health in early-weaned piglets.
Collapse
Affiliation(s)
- Zhining Tang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China
| | - Yang Yang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China
| | - Mingrui Yang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China
| | - Da Jiang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China
| | - Yao Ge
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China
| | - Xinyu Zhang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China
| | - Haozhen Liu
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China
| | - Qingyao Fu
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China
| | - Xiyuan Liu
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China
| | - Ying Yang
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China
| | - Zhenlong Wu
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China; Beijing Advanced Innovation Center for Food Nutrition and Human Health, China Agricultural University, Beijing 100193, China
| | - Yun Ji
- State Key Laboratory of Animal Nutrition and Feeding, China Agricultural University, Beijing 100193, China.
| |
Collapse
|