101
|
Chang CJ, Bai YC, Jiang H, Ma QW, Hsieh CH, Liu CC, Huang HC, Chen TJ. Microbiome analysis of serum extracellular vesicles in gestational diabetes patients. Acta Diabetol 2025; 62:329-341. [PMID: 39570375 DOI: 10.1007/s00592-024-02358-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/13/2024] [Accepted: 08/07/2024] [Indexed: 11/22/2024]
Abstract
AIM Gestational Diabetes Mellitus (GDM) is among the most common complications during pregnancy, posing serious risks to both the patient's and offspring's health and well-being. Alterations in the maternal microbiome are closely associated with the pathogenesis of GDM, with Extracellular Vesicles (EVs) facilitating communication between microbiota and the host. However, little is known about the relationship between the microbial composition within EVs and the pathogenesis of GDM. Therefore, this study aims to characterize the microbiota within serum EVs of GDM Patients (GDM group) and to identify microbial communities that significantly differ from those in Women With Normal Pregnancies (NonGDM group). METHODS Blood samples were collected from both groups of patients, and EVs derived from serum were isolated via centrifugation. Identification and characterization of EVs were performed using transmission electron microscopy and nanoparticle flow cytometry. Microbiome analysis of serum EVs from both groups was conducted using 16S rRNA sequencing. RESULTS Results indicated altered diversity in microbial communities within serum EVs of GDM patients. Further analysis at the phylum, family, genus, and species levels revealed that Proteobacteria, Actinobacteria, Bacteroidetes, and Firmicutes were the dominant taxa in the EVs of both the NonGDM and GDM groups. Specifically, Actinobacteria and Firmicutes showed increased relative abundance in GDM group EVs compared to NonGDM, leading to a higher Firmicutes/Bacteroidetes ratio, while Proteobacteria and Bacteroidetes exhibited decreased relative abundance. Tax4Fun analysis revealed enrichment of microbial functions related to amino acid metabolism, carbohydrate metabolism, energy metabolism, and metabolism of cofactors and vitamins in both patient groups. CONCLUSION In conclusion, this study reveals a potential correlation between changes in the microbial composition and diversity of serum EVs and the onset and development of GDM. Furthermore, changes in the relative abundance of Actinobacteria, Proteobacteria, Bacteroidetes, and Firmicutes may play an important role in the pathogenesis of GDM.
Collapse
Affiliation(s)
- Chih-Jung Chang
- School of Medicine and Medical Research Center, Xiamen Chang Gung Hospital Huaqiao University, Fujian, China
| | - Yu-Ci Bai
- Department of Obstetrics and Gynecology, Xiamen Chang Gung Hospital Huaqiao University, Fujian, China
| | - Hong Jiang
- Reproductive Medicine Center, The First Affiliated Hospital of Fujian Medical University, Fujian, China
| | - Qi-Wen Ma
- School of Medicine and Medical Research Center, Xiamen Chang Gung Hospital Huaqiao University, Fujian, China
| | - Cheng-Hsien Hsieh
- Department of Emergency Medicine, En Chu Kong Hospital, New Taipei City, Taiwan
| | - Chien-Chun Liu
- Molecular Medicine Research Center, Chang Gung University, Taoyuan, Taiwan
| | - Hung-Chien Huang
- Department of Emergency Medicine, En Chu Kong Hospital, New Taipei City, Taiwan.
| | - Tien-Jui Chen
- Department of Laboratory Medicine, Yeezen General Hospital, Taoyuan, Taiwan.
| |
Collapse
|
102
|
Hu B, Yue K, Zhang D, Feng S, Zhao N, Li G, Gao S, Xing Y, Han S, He H. Association between Capillaria hepatica infection-induced alterations in gut microbiota and estrogen expression in Brandt's voles (Lasiopodomys brandtii). BMC Vet Res 2025; 21:126. [PMID: 40025543 PMCID: PMC11871588 DOI: 10.1186/s12917-025-04524-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2023] [Accepted: 01/25/2025] [Indexed: 03/04/2025] Open
Abstract
BACKGROUND Capillaria hepatica, a zoonotic parasite, is present in the population of Brandt's voles (Lasiopodomys brandtii) and has been a central issue in ecological studies regarding its impact on host populations. Brandt's voles are known for their extremely high reproductive capacity, and the population explosion of Brandt's voles have occurred multiple times in the grasslands of Inner Mongolia over the past few decades. However, the mechanisms underlying the population dynamics of Brandt's voles, particularly in response to C. hepatica infection, remain poorly understood. Given the critical role of the gut microbiota in modulating hormones within the reproductive endocrine system, this study aims to explore how alterations in the gut microbiota influence the host's population dynamics in response to C. hepatica infection. METHODS Female Brandt's voles were inoculated with eggs of infected C. hepatica, and BALB/C mice were used as a control. At the end of the experimental period, cecal contents were collected for 16 S rRNA amplicon sequencing, and the expression levels of reproductive-related hormones were determined using enzyme-linked immunosorbent assay (ELISA). RESULTS C. hepatica infection leads to an increased diversity of gut microbiota in Brandt's voles, with significant changes in microbial composition. The relative abundance of Muribaculaceae and Eubacteriaceae increased significantly, while that of Rikenellaceae and Lachnospiraceae decreased significantly. The expression level of estradiol in the serum of infected Brandt's voles shows a slight decrease without statistical significance. However, the expression of equol is significantly higher in the infected group compared to the uninfected group, and the expression of enterolactone is significantly lower in the infected group than in the uninfected group. CONCLUSIONS This study demonstrates that infection with C. hepatica indirectly affect the abundance of specific gut microbiota in Brandt's voles, which are associated with reproductive hormones. This indirect effect on hormone expression can subsequently impact the reproductive function of the host. By investigating the changes in specific gut microbiota, this study sheds light on the mechanisms through which parasites regulate population fluctuations in Brandt's voles.
Collapse
Affiliation(s)
- Bin Hu
- College of Animal Science and Veterinary Medicine, Henan Institute of Science and Technology, Xinxiang, 453003, China
- Institute of Zoology, Chinese Academy of Sciences, Beijing, 100020, China
- School of Life Sciences, Henan University, Kaifeng, 475004, China
| | - Kening Yue
- Institute of Zoology, Chinese Academy of Sciences, Beijing, 100020, China
| | - Daibao Zhang
- Henan wildlife conservation center, Zhengzhou, 450000, China
| | - Shengyong Feng
- Institute of Zoology, Chinese Academy of Sciences, Beijing, 100020, China
| | - Ning Zhao
- National Institute for Communicable Disease Control and Prevention, Chinese Center for Disease Control and Prevention, 100101, Beijing, China
| | - Gaojian Li
- Institute of Zoology, Chinese Academy of Sciences, Beijing, 100020, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Sichao Gao
- Institute of Zoology, Chinese Academy of Sciences, Beijing, 100020, China
| | - Yanan Xing
- Institute of Zoology, Chinese Academy of Sciences, Beijing, 100020, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shuyi Han
- Institute of Zoology, Chinese Academy of Sciences, Beijing, 100020, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hongxuan He
- Institute of Zoology, Chinese Academy of Sciences, Beijing, 100020, China.
| |
Collapse
|
103
|
Rossouw C, Ryan FJ, Lynn DJ. The role of the gut microbiota in regulating responses to vaccination: current knowledge and future directions. FEBS J 2025; 292:1480-1499. [PMID: 39102299 PMCID: PMC11927049 DOI: 10.1111/febs.17241] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/13/2024] [Accepted: 07/24/2024] [Indexed: 08/07/2024]
Abstract
Antigen-specific B and T cell responses play a critical role in vaccine-mediated protection against infectious diseases, but these responses are highly variable between individuals and vaccine immunogenicity is frequently sub-optimal in infants, the elderly and in people living in low- and middle-income countries. Although many factors such as nutrition, age, sex, genetics, environmental exposures, and infections may all contribute to variable vaccine immunogenicity, mounting evidence indicates that the gut microbiota is an important and targetable factor shaping optimal immune responses to vaccination. In this review, we discuss evidence from human, preclinical and experimental studies supporting a role for a healthy gut microbiota in mediating optimal vaccine immunogenicity, including the immunogenicity of COVID-19 vaccines. Furthermore, we provide an overview of the potential mechanisms through which this could occur and discuss strategies that could be used to target the microbiota to boost vaccine immunogenicity where it is currently sub-optimal.
Collapse
Affiliation(s)
- Charné Rossouw
- Precision MedicineSouth Australian Health and Medical Research Institute (SAHMRI)AdelaideAustralia
- Flinders Health and Medical Research InstituteFlinders UniversityBedford ParkAustralia
| | - Feargal J. Ryan
- Precision MedicineSouth Australian Health and Medical Research Institute (SAHMRI)AdelaideAustralia
- Flinders Health and Medical Research InstituteFlinders UniversityBedford ParkAustralia
| | - David J. Lynn
- Precision MedicineSouth Australian Health and Medical Research Institute (SAHMRI)AdelaideAustralia
- Flinders Health and Medical Research InstituteFlinders UniversityBedford ParkAustralia
| |
Collapse
|
104
|
Ma G, Chen Z, Xie Z, Liu J, Xiao X. Mechanisms underlying changes in intestinal permeability during pregnancy and their implications for maternal and infant health. J Reprod Immunol 2025; 168:104423. [PMID: 39793281 DOI: 10.1016/j.jri.2025.104423] [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: 08/31/2024] [Revised: 12/01/2024] [Accepted: 01/03/2025] [Indexed: 01/13/2025]
Abstract
Proper regulation of intestinal permeability is essential for maintaining the integrity of the intestinal mucosal barrier. An abnormal increase in permeability can significantly contribute to the onset and progression of various diseases, including autoimmune disorders, metabolic conditions, allergies, and inflammatory bowel diseases. The potential connection between intestinal permeability and maternal health during pregnancy is increasingly recognized, yet a comprehensive review remains lacking. Pregnancy triggers a series of physiological structural adaptations and significant hormonal fluctuations that collectively contribute to an increase in intestinal permeability. Although an increase in intestinal permeability is typically a normal physiological response during pregnancy, an abnormal rise is associated with immune dysregulation, metabolic disorders, and various pregnancy-related complications, such as recurrent pregnancy loss, gestational diabetes mellitus, overweight and obesity during pregnancy, intrahepatic cholestasis of pregnancy, and preeclampsia. This paper discusses the components of the intestinal mucosal barrier, the concept of intestinal permeability and its measurement methods, and the mechanisms and physiological significance of increased intestinal permeability during pregnancy. It thoroughly explores the association between abnormal intestinal permeability during pregnancy and maternal diseases, aiming to provide evidence for the pathophysiology of disease development in pregnant women. Additionally, the paper examines intervention methods, such as gut microbiota modulation and nutritional interventions, to regulate intestinal permeability during pregnancy, improve immune and metabolic states, and offer feasible strategies for the prevention and adjuvant treatment of clinical pregnancy complications.
Collapse
Affiliation(s)
- Guangyu Ma
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Zhongsheng Chen
- Department of Colorectal Cancer Surgery, The Second Affiliated Hospital of Harbin Medical University, Harbin Medical University, Harbin, China
| | - Zhuojun Xie
- General Medicine Department, Clinical Medical College & Affiliated Hospital of Chengdu University, Chengdu University, Chengdu, China
| | - JinXiang Liu
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China
| | - Xiaomin Xiao
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Jinan University, Jinan University, Guangzhou, China.
| |
Collapse
|
105
|
Sullivan O, Sie C, Ng KM, Cotton S, Rosete C, Hamden JE, Singh AP, Lee K, Choudhary J, Kim J, Yu H, Clayton CA, Carranza Garcia NA, Voznyuk K, Deng BD, Plett N, Arora S, Ghezzi H, Huan T, Soma KK, Yu JPJ, Tropini C, Ciernia AV. Early-life gut inflammation drives sex-dependent shifts in the microbiome-endocrine-brain axis. Brain Behav Immun 2025; 125:117-139. [PMID: 39674560 DOI: 10.1016/j.bbi.2024.12.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 11/13/2024] [Accepted: 12/09/2024] [Indexed: 12/16/2024] Open
Abstract
Despite recent advances in understanding the connection between the gut microbiota and the adult brain, significant knowledge gaps remain regarding how gut inflammation affects brain development. We hypothesized that gut inflammation during early life would negatively affect neurodevelopment by disrupting microbiota communication to the brain. We therefore developed a novel pediatric chemical model of inflammatory bowel disease (IBD), an incurable condition affecting millions of people worldwide. IBD is characterized by chronic intestinal inflammation, and is associated with comorbid symptoms such as anxiety, depression and cognitive impairment. Notably, 25% of patients with IBD are diagnosed during childhood, and the effects of chronic inflammation during this critical developmental period remain poorly understood. This study investigated the effects of early-life gut inflammation induced by DSS (dextran sulfate sodium) on a range of microbiota, endocrine, and behavioral outcomes, focusing on sex-specific impacts. DSS-treated mice exhibited increased intestinal inflammation and altered microbiota membership, which correlated with changes in microbiota-derived circulating metabolites. The majority of behavioral measures were unaffected, with the exception of impaired mate-seeking behaviors in DSS-treated males. DSS-treated males also showed significantly smaller seminal vesicles, lower circulating androgens, and decreased intestinal hormone-activating enzyme activity compared to vehicle controls. In the brain, DSS treatment led to chronic, sex-specific alterations in microglial morphology. These results suggest that early-life gut inflammation causes changes in gut microbiota composition, affecting short-chain fatty acid (SCFA) producers and glucuronidase (GUS) activity, correlating with altered SCFA and androgen levels. The findings highlight the developmental sensitivity to inflammation-induced changes in endocrine signalling and emphasize the long-lasting physiological and microbiome changes associated with juvenile IBD.
Collapse
Affiliation(s)
- Olivia Sullivan
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Claire Sie
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Katharine M Ng
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Sophie Cotton
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Cal Rosete
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Jordan E Hamden
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Ajay Paul Singh
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Kristen Lee
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada
| | - Jatin Choudhary
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Jennifer Kim
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Huaxu Yu
- Department of Chemistry, University of British Columbia, Vancouver, Canada
| | - Charlotte A Clayton
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | | | - Kateryna Voznyuk
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Brian D Deng
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada
| | - Nadine Plett
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Sana Arora
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada
| | - Hans Ghezzi
- Department of Bioinformatics, University of British Columbia, Vancouver, Canada
| | - Tao Huan
- Department of Chemistry, University of British Columbia, Vancouver, Canada
| | - Kiran K Soma
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Psychology, University of British Columbia, Vancouver Canada
| | - John-Paul J Yu
- Department of Radiology, University of Wisconsin School of Medicine and Public Health, Madison, WI, USA
| | - Carolina Tropini
- Department of Microbiology & Immunology, University of British Columbia, Vancouver, Canada; School of Biomedical Engineering, University of British Columbia, Vancouver, Canada; Humans and the Microbiome Program, Canadian Institute for Advanced Research (CIFAR), Toronto, Canada.
| | - Annie Vogel Ciernia
- Graduate Program in Neuroscience, University of British Columbia, Vancouver, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, Canada; Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, Canada.
| |
Collapse
|
106
|
Sun Y, Yao J, Gao R, Hao J, Liu Y, Liu S. Interactions of non-starch polysaccharides with the gut microbiota and the effect of non-starch polysaccharides with different structures on the metabolism of the gut microbiota: A review. Int J Biol Macromol 2025; 296:139664. [PMID: 39798752 DOI: 10.1016/j.ijbiomac.2025.139664] [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: 08/09/2024] [Revised: 12/19/2024] [Accepted: 01/07/2025] [Indexed: 01/15/2025]
Abstract
Humans consume large amounts of non-starch polysaccharides(NPs) daily. Some NPs, not absorbed by the body, proceed to the intestines. An increasing number of studies reveal a close relationship between NPs and gut microbiota(GM) that impact the human body. This review not only describes in detail the structures of several common NPs and their effects on GM, but also elucidates the degradation mechanisms of NPs in the intestine. The purpose of this review is to elucidate how NPs interact with GM in the intestine, which can provide valuable information for further studies of NPs.
Collapse
Affiliation(s)
- Yujiao Sun
- Natural Food Macromolecule Research Center, School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an 710021, PR China; Shaanxi Research Institute of Agricultural Products Processing Technology, Xi'an 710021, PR China.
| | - Jiaxuan Yao
- Natural Food Macromolecule Research Center, School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an 710021, PR China
| | - Running Gao
- Natural Food Macromolecule Research Center, School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an 710021, PR China
| | - Junyu Hao
- Natural Food Macromolecule Research Center, School of Food Science and Engineering, Shaanxi University of Science and Technology, Xi'an 710021, PR China
| | - Yang Liu
- Shaanxi Academy of Traditional Chinese Medicine, Xi'an 710003, China
| | - Shuai Liu
- Shaanxi Academy of Traditional Chinese Medicine, Xi'an 710003, China.
| |
Collapse
|
107
|
Zhang L, Yin Y, Jin S. Gut microbial metabolites: The bridge connecting diet and atherosclerosis, and next-generation targets for dietary interventions. Microbiol Res 2025; 292:128037. [PMID: 39752807 DOI: 10.1016/j.micres.2024.128037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2024] [Revised: 12/05/2024] [Accepted: 12/19/2024] [Indexed: 01/19/2025]
Abstract
Mounting evidence indicates that gut microbial metabolites are central hubs linking the gut microbiota to atherosclerosis (AS). Gut microbiota enriched with pathobiont bacteria responsible for producing metabolites like trimethylamine N-oxide and phenylacetylglutamine are related to an increased risk of cardiovascular events. Furthermore, gut microbiota enriched with bacteria responsible for producing short-chain fatty acids, indole, and its derivatives, such as indole-3-propionic acid, have demonstrated AS-protective effects. This study described AS-related gut microbial composition and how microbial metabolites affect AS. Summary findings revealed gut microbiota and their metabolites-targeted diets could benefit AS treatment. In conclusion, dietary interventions centered on the gut microbiota represent a promising strategy for AS treatment, and understanding diet-microbiota interactions could potentially be devoted to developing novel anti-AS therapies.
Collapse
Affiliation(s)
- Liyin Zhang
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, 39 Lake Road, East Lake Ecological Scenic, Wuhan, Hubei 430077, China
| | - Yao Yin
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, 39 Lake Road, East Lake Ecological Scenic, Wuhan, Hubei 430077, China
| | - Si Jin
- Department of Endocrinology, Institute of Geriatric Medicine, Liyuan Hospital, Tongji Medical College, Huazhong University of Science and Technology, 39 Lake Road, East Lake Ecological Scenic, Wuhan, Hubei 430077, China.
| |
Collapse
|
108
|
Chao J, Coleman RA, Keating DJ, Martin AM. Gut Microbiome Regulation of Gut Hormone Secretion. Endocrinology 2025; 166:bqaf004. [PMID: 40037297 PMCID: PMC11879239 DOI: 10.1210/endocr/bqaf004] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Indexed: 03/06/2025]
Abstract
The gut microbiome, comprising bacteria, viruses, fungi, and bacteriophages, is one of the largest microbial ecosystems in the human body and plays a crucial role in various physiological processes. This review explores the interaction between the gut microbiome and enteroendocrine cells (EECs), specialized hormone-secreting cells within the intestinal epithelium. EECs, which constitute less than 1% of intestinal epithelial cells, are key regulators of gut-brain communication, energy metabolism, gut motility, and satiety. Recent evidence shows that gut microbiota directly influence EEC function, maturation, and hormone secretion. For instance, commensal bacteria regulate the production of hormones like glucagon-like peptide 1 and peptide YY by modulating gene expression and vesicle cycling in EE cells. Additionally, metabolites such as short-chain fatty acids, derived from microbial fermentation, play a central role in regulating EEC signaling pathways that affect metabolism, gut motility, and immune responses. Furthermore, the interplay between gut microbiota, EECs, and metabolic diseases, such as obesity and diabetes, is examined, emphasizing the microbiome's dual role in promoting health and contributing to disease states. This intricate relationship between the gut microbiome and EECs offers new insights into potential therapeutic strategies for metabolic and gut disorders.
Collapse
Affiliation(s)
- Jessica Chao
- Gut Hormones in Health and Disease Lab, Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia
| | - Rosemary A Coleman
- Gut Hormones in Health and Disease Lab, Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia
| | - Damien J Keating
- Gut Sensory Systems Group, Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia
| | - Alyce M Martin
- Gut Hormones in Health and Disease Lab, Flinders Health and Medical Research Institute, College of Medicine and Public Health, Flinders University, Adelaide 5042, Australia
| |
Collapse
|
109
|
Connolly KR, Sweeney T, Ryan MT, Vigors S, O’Doherty JV. Impact of Reduced Dietary Crude Protein and Propionic Acid Preservation on Intestinal Health and Growth Performance in Post-Weaned Pigs. Animals (Basel) 2025; 15:702. [PMID: 40075985 PMCID: PMC11899430 DOI: 10.3390/ani15050702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Revised: 01/10/2025] [Accepted: 02/19/2025] [Indexed: 03/14/2025] Open
Abstract
This study investigated whether organic acid (OA)-preserved grain could mitigate the negative effects of low crude protein (CP) diets on growth performance, intestinal health, and the coefficient of total tract digestibility (CATTD) of nutrients in weaned piglets. The grain was either conventionally dried or preserved post-harvest with 4 kg of OA per tonne. Ninety-six piglets (28 days old) were assigned to one of four diets in a 2 × 2 factorial design: (1) dried standard CP diet, (2) OA-preserved standard CP diet, (3) dried low CP diet, and (4) OA-preserved low CP diet. Standard and low CP diets contained 20% and 19% CP during the first 15 days, reduced to 19% and 17% CP from days 15-35 post-weaning. Faecal scores (FS) were assessed twice a day while microbial composition, inflammatory markers, colonic volatile fatty acid concentrations, and intestinal morphology were measured on the 8th day post-weaning. Performance metrics were measured over the 35-day experimental period. Low CP diets consistently reduced FS (p < 0.05) and increased colonic molar butyrate proportions (p < 0.01) but increased duodenal IL1B expression compared to standard CP diets (p < 0.05). The OA-preserved grain enhanced beneficial microbial populations (Lactobacillus, Roseburia) while lowering pro-inflammatory cytokines (IL1A, IL17) (p < 0.05). While dried grain with low CP diets reduced average daily gain (ADG), colonic short-chain fatty acids (SCFA) concentrations, and nitrogen digestibility, OA-preserved grain with low CP maintained these parameters and improved final body weight (p < 0.05). Overall, OA-preserved grain mitigated the performance decline associated with low CP diets by enhancing gut health and nutrient digestibility and reducing inflammation, thus presenting a promising alternative nutritional strategy for post-weaned piglets.
Collapse
Affiliation(s)
- Kathryn Ruth Connolly
- School of Agriculture and Food Science, University College Dublin, D04 W6F6 Dublin, Ireland; (K.R.C.); (S.V.)
| | - Torres Sweeney
- School of Veterinary Medicine, University College Dublin, D04 W6F6 Dublin, Ireland; (T.S.); (M.T.R.)
| | - Marion T. Ryan
- School of Veterinary Medicine, University College Dublin, D04 W6F6 Dublin, Ireland; (T.S.); (M.T.R.)
| | - Stafford Vigors
- School of Agriculture and Food Science, University College Dublin, D04 W6F6 Dublin, Ireland; (K.R.C.); (S.V.)
| | - John V. O’Doherty
- School of Agriculture and Food Science, University College Dublin, D04 W6F6 Dublin, Ireland; (K.R.C.); (S.V.)
| |
Collapse
|
110
|
García G, Carlin M, Cano RDJ. Holobiome Harmony: Linking Environmental Sustainability, Agriculture, and Human Health for a Thriving Planet and One Health. Microorganisms 2025; 13:514. [PMID: 40142407 PMCID: PMC11945859 DOI: 10.3390/microorganisms13030514] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 02/14/2025] [Accepted: 02/21/2025] [Indexed: 03/28/2025] Open
Abstract
The holobiome is an interconnected network of microbial ecosystems spanning soil, plants, animals, humans, and the environment. Microbial interactions drive nutrient cycling, pathogen suppression, and climate regulation. Soil microbiomes facilitate carbon sequestration and enhance soil fertility, while marine microbiomes contribute to carbon capture and climate stability. However, industrial agriculture, extensive herbicide use, antibiotic overuse, and climate change threaten microbial diversity, leading to ecosystem and health disruptions. Probiotic interventions help to restore microbial balance. In human health, probiotics support gut microbiota diversity, reduce inflammation, and regulate metabolism. In agriculture, soil probiotics enhance microbial diversity, improve nutrient cycling, and degrade contaminants, increasing crop yields and soil health. Case studies show that microbial inoculants effectively remediate degraded soils and enhance nutrient uptake. Artificial intelligence is transforming microbiome research by enabling predictive modeling, precision probiotic design, and microbial consortia optimization. Interdisciplinary collaboration and supportive policies are essential for restoring microbial equilibria, ensuring ecosystem resilience, and promoting long-term sustainability. The integration of artificial intelligence, clinical research, and sustainable practices is crucial for advancing holobiome science. The holobiome framework underscores the need for interdisciplinary collaboration to address global challenges, bridging environmental sustainability, agriculture, and public health for a resilient future.
Collapse
Affiliation(s)
- Gissel García
- Pathology Department, Hospital Hermanos Ameijeiras, La Habana 10400, Cuba;
| | | | - Raul de Jesus Cano
- Biological Sciences Department, California Polytechnic State University, San Luis Obispo, CA 93407, USA
- Chauvell, LLC, San Luis Obispo, CA 93401, USA
| |
Collapse
|
111
|
Chen W, Song T, Li D, Chen M, Wang P, Ye J. Effect of dietary Clostridium butyricum supplementation on growth performance, immune function, and intestinal health of hybrid grouper ( Epinephelus fuscoguttatus ♀ × Epinephelus lanceolatus ♂). Front Immunol 2025; 16:1557256. [PMID: 40078994 PMCID: PMC11897522 DOI: 10.3389/fimmu.2025.1557256] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Accepted: 02/10/2025] [Indexed: 03/14/2025] Open
Abstract
Introduction The aim of this study is to investigate the effects of supplementing Clostridium butyricum (C. butyricum) on hybrid grouper (Epinephelus fuscoguttatus ♀ × Epinephelus lanceolatus ♂), with a particular focus on its impact on growth performance, blood composition, intestinal antioxidant capacity, gut microbiota, tight junction protein (ZO-1) expression, and inflammatory gene expression. The study seeks to uncover the potential health benefits of C. butyricum supplementation for hybrid grouper. Methods The experiment included four groups: a control group (CON) and three experimental groups, each supplemented with different strains of C. butyricum (KM, DZN, and CLH), with a concentration of 1 × 10⁷ colony-forming units per gram. These groups were designated as CB1 (KM), CB2 (DZN), and CB3 (CLH). The study evaluated growth performance, blood composition, intestinal antioxidant capacity, gut microbiota, ZO-1 protein expression, and inflammatory gene expression (IL-1β and Ikk-β). Result The results indicated that supplementation with C. butyricum had no significant effect on body weight gain (WG), feed efficiency (FE), or body composition. However, the CB3 group significantly increased the activity of superoxide dismutase (SOD) and glutathione peroxidase (GSH-Px) in the intestine, as well as the expression of ZO-1. In addition, the CB3 group significantly increased serum lysozyme (LZM) activity, complement 4 (C4) levels, and immunoglobulin M (IgM) concentration, while significantly reducing the expression of pro-inflammatory genes (IL-1β and Ikk-β). After supplementation with C. butyricum, the level of malondialdehyde (MDA) in the intestine was significantly lower than that in the control group, indicating a reduction in intestinal oxidative stress. Supplementation with C. butyricum also altered the composition of the gut microbiota, promoting the growth of beneficial bacteria and inhibiting pathogenic bacteria, thereby further enhancing ZO-1 expression and intestinal barrier function. Discussion This study suggests that supplementing C. butyricum has a significant immunomodulatory effect on hybrid grouper, enhancing serum immune parameters, alleviating intestinal inflammation and oxidative stress, and promoting intestinal health. Although no significant impact was observed on growth performance, the role of C. butyricum in improving intestinal barrier function and modulating the gut microbiota highlights its potential for enhancing fish health.
Collapse
Affiliation(s)
| | | | | | | | | | - Jidan Ye
- Fisheries College of Jimei University, Xiamen Key Laboratory for Feed Quality Testing and Safety Evaluation, Xiamen, China
| |
Collapse
|
112
|
Zhao Z, Hu Z, Li L. Cardiac energy metabolic disorder and gut microbiota imbalance: a study on the therapeutic potential of Shenfu Injection in rats with heart failure. Front Microbiol 2025; 16:1509548. [PMID: 40071211 PMCID: PMC11895768 DOI: 10.3389/fmicb.2025.1509548] [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: 10/11/2024] [Accepted: 02/11/2025] [Indexed: 03/14/2025] Open
Abstract
Objective To investigate the relationship between heart failure (HF) and gut microbiota-mediated energy metabolism, and to explore the role of Shenfu Injection in this process. Materials and methods In this study, Adriamycin-induced chronic heart failure (CHF) rat model was used and randomly divided into the blank control group (Normal, n = 9), HF control group (Model, n = 12), Shenfu Injection treatment group (SFI, n = 9), and positive drug control group (TMZ, n = 9). The changes in gut microbiota structure were analyzed by 16S rRNA high-throughput sequencing, the content of short-chain fatty acids (SCFAs) was detected by targeted metabolomics technology, and cardiac function and energy metabolism-related indicators were evaluated. Results Myocardial energy metabolism in HF rats was disordered, characterized by reduced fatty acid oxidation, enhanced anaerobic glycolysis of glucose, mitochondrial damage, and decreased ATP content; The gut microbiota of HF rats was imbalanced, with a reduction in beneficial bacteria, an increase in conditional pathogenic bacteria, and impaired intestinal barrier function; Both Shenfu Injection and trimetazidine improved myocardial energy metabolism and cardiac function, but Shenfu Injection was more significant in regulating gut microbiota and improving intestinal health; The production of SCFAs from the gut microbiota of HF rats increased, which may be closely related to myocardial energy metabolism; SCFAs-producing bacteria Akkermansia and Blautia played a key role in the development of HF, and their abundance was positively correlated with SCFAs content. Conclusion Shenfu Injection in treating HF may improve myocardial energy metabolism and intestinal health by regulating gut microbiota, especially the abundance of SCFAs-producing bacteria Akkermansia and Blautia, thereby exerting therapeutic effects. This provides theoretical support for treatment strategies based on gut microbiota.
Collapse
Affiliation(s)
- Zhenyu Zhao
- Provincial Key Laboratory of TCM Diagnostics, Hunan University of Chinese Medicine, Changsha, China
| | - Zhixi Hu
- Institute of TCM Diagnosis, Hunan University of Chinese Medicine, Changsha, China
| | - Lin Li
- Institute of TCM Diagnosis, Hunan University of Chinese Medicine, Changsha, China
| |
Collapse
|
113
|
Abdelqader EM, Mahmoud WS, Gebreel HM, Kamel MM, Abu-Elghait M. Correlation between gut microbiota dysbiosis, metabolic syndrome and breast cancer. Sci Rep 2025; 15:6652. [PMID: 39994329 PMCID: PMC11850770 DOI: 10.1038/s41598-025-89801-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2024] [Accepted: 02/07/2025] [Indexed: 02/26/2025] Open
Abstract
Breast cancer is a widespread cancer with a high death rate globally. The incidence of breast cancer is expected to increase, particularly in low and middle-income countries due to environmental factors and lifestyle changes. Several risk factors, such as age, family history, hormonal and reproductive factors, have been identified to influence breast cancer development. Metabolic syndrome, is a metabolic disorder that has also been linked to breast cancer risk. The gut microbiome has been suggested as one of the environmental factors leading to breast cancer. The human microbiome is mainly colonized in the intestine by various bacterial species, including Lactobacillus, Bifidobacterium, and Streptococcus and protect the host against pathogenic microorganisms and regulate the immune system. This study included 50 female breast cancer patients and 50 healthy controls with matched ages. Stool fresh samples were taken from test and control groups and stored at - 20 °C until further investigations. DNA of the bacteria in stool samples was extracted using reverse transcription-quantitative polymerase chain reaction to check for the bacterial 16s rRNA gene. The exclusion criteria included other malignancies, recent intestinal surgery, infectious diarrhea, prolonged use of antibiotics, substance addiction, and pregnancy or lactation. Our findings exhibited that breast cancer patients had a higher incidence of metabolic syndrome (60%) compared to cancer-free controls (40%). Furthermore, breast cancer patients had significantly lower Bifidobacterium and Lactobacillus counts than the controls. No significant difference was found in Streptococcus counts between groups. These findings support the relationship between breast cancer and metabolic syndrome and suggest the potential involvement of Lactobacillus and Bifidobacterium in breast cancer pathophysiology. Our study supports the relation between breast cancer and disorder of metabolic syndrome and suggests the potential involvement of Lactobacillus and Bifidobacterium in breast cancer pathophysiology. Further research is necessary to investigate the complex interactions between genes, the environment, and the gut microbiome in breast cancer development. Understanding these interactions could lead to the progress of novel strategies for breast cancer prevention and treatment.
Collapse
Affiliation(s)
- Eslam M Abdelqader
- Department of Microbiology, Faculty of Science, Ain Shams University, Cairo, Egypt.
| | - Walaa S Mahmoud
- Biological Anthropology Department National Research Centre, Dokki, Giza, Egypt
| | - Hassan M Gebreel
- Department of Microbiology, Faculty of Science, Ain Shams University, Cairo, Egypt
| | - Mahmoud M Kamel
- Clinical Pathology Development, National Cancer Institute Cairo University, Cairo, Egypt
- Laboratory Development Bahyea Centre for Early Detection and Cancer Treatment, Cairo, Egypt
| | - Mohammed Abu-Elghait
- Department of Botany and Microbiology, Faculty of Science, Al-Azhar University, Cairo, 11884, Egypt
| |
Collapse
|
114
|
Chen X, Wei J, Zhang L, Wang H, Zhang Y, Li Z, Wang X, Liu L, Zhang Y, Zhang T. Association between plasma short-chain fatty acids and inflammation in human immunodeficiency virus-associated neurocognitive disorder: a pilot study. Lipids Health Dis 2025; 24:66. [PMID: 39984934 PMCID: PMC11846350 DOI: 10.1186/s12944-025-02477-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2025] [Accepted: 02/10/2025] [Indexed: 02/23/2025] Open
Abstract
BACKGROUND AND AIMS Short-chain fatty acids (SCFAs), key metabolites produced by gut microbiota, have neuroprotective effects in neurodegenerative diseases by modulating immune responses. However, their role in human immunodeficiency virus (HIV)-associated neurocognitive disorder (HAND) remains largely unexplored. METHODS We recruited HAND patients, HIV Control, and healthy controls (HC). Plasma SCFAs and SCFA-producing gut microbiota were quantified via gas chromatography-mass spectrometry and fecal metagenomic analysis. Inflammatory cytokine levels were measured using liquid chromatography. Receiver operating characteristic (ROC) curves were generated to evaluate the predictive accuracy of SCFAs for HAND. RESULTS Plasma SCFAs were significantly reduced in HAND patients, correlating with a decrease in SCFA-producing gut bacteria, such as Prevotella and its related species. Reduced SCFAs were positively correlated with pro-inflammatory cytokines and cognitive impairment, while being negatively correlated with anti-inflammatory cytokines. ROC curve analysis demonstrated that several SCFAs exhibited strong predictive accuracy for HAND status. CONCLUSIONS SCFAs may influence cognitive function by modulating inflammatory responses, and identifies plasma SCFAs as potential biomarkers and therapeutic targets for HAND. Further investigation is needed to delineate the mechanisms that SCFAs influence HAND pathology.
Collapse
Affiliation(s)
- Xue Chen
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Youan Hospital, Beijing Institute of Hepatology, Capital Medical University, Beijing, China
| | - Jiaqi Wei
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Ling Zhang
- Beijing Youan Hospital, Beijing Institute of Hepatology, Capital Medical University, Beijing, China
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Hu Wang
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yang Zhang
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Zhen Li
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Xia Wang
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Lifeng Liu
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China
| | - Yulin Zhang
- Department of Respiratory and Critical Care Medicine, Beijing Youan Hospital, Capital Medical University, Beijing, China.
| | - Tong Zhang
- Clinical and Research Center for Infectious Diseases, Beijing Youan Hospital, Capital Medical University, Beijing, China.
- Beijing Key Laboratory for HIV/AIDS Research, Beijing Youan Hospital, Capital Medical University, Beijing, China.
| |
Collapse
|
115
|
Blok L, Hanssen N, Nieuwdorp M, Rampanelli E. From Microbes to Metabolites: Advances in Gut Microbiome Research in Type 1 Diabetes. Metabolites 2025; 15:138. [PMID: 39997763 PMCID: PMC11857261 DOI: 10.3390/metabo15020138] [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: 02/01/2025] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/26/2025] Open
Abstract
Background: Type 1 diabetes (T1D) is a severe chronic T-cell mediated autoimmune disease that attacks the insulin-producing beta cells of the pancreas. The multifactorial nature of T1D involves both genetic and environmental components, with recent research focusing on the gut microbiome as a crucial environmental factor in T1D pathogenesis. The gut microbiome and its metabolites play an important role in modulating immunity and autoimmunity. In recent years, studies have revealed significant alterations in the taxonomic and functional composition of the gut microbiome associated with the development of islet autoimmunity and T1D. These changes include reduced production of short-chain fatty acids, altered bile acid and tryptophan metabolism, and increased intestinal permeability with consequent perturbations of host (auto)immune responses. Methods/Results: In this review, we summarize and discuss recent observational, mechanistic and etiological studies investigating the gut microbiome in T1D and elucidating the intricate role of gut microbes in T1D pathogenesis. Moreover, we highlight the recent advances in intervention studies targeting the microbiota for the prevention or treatment of human T1D. Conclusions: A deeper understanding of the evolution of the gut microbiome before and after T1D onset and of the microbial signals conditioning host immunity may provide us with essential insights for exploiting the microbiome as a prognostic and therapeutic tool.
Collapse
Affiliation(s)
- Lente Blok
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, 1105 AZ Amsterdam, The Netherlands; (N.H.); (M.N.)
| | - Nordin Hanssen
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, 1105 AZ Amsterdam, The Netherlands; (N.H.); (M.N.)
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, 1105 AZ Amsterdam, The Netherlands; (N.H.); (M.N.)
| | - Elena Rampanelli
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, 1105 AZ Amsterdam, The Netherlands; (N.H.); (M.N.)
- Amsterdam Institute for Infection and Immunity (AII), Amsterdam, The Netherlands
| |
Collapse
|
116
|
Hsu KY, Majeed A, Ho CT, Pan MH. Bisdemethoxycurcumin and Curcumin Alleviate Inflammatory Bowel Disease by Maintaining Intestinal Epithelial Integrity and Regulating Gut Microbiota in Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:3494-3506. [PMID: 39873626 PMCID: PMC11826975 DOI: 10.1021/acs.jafc.4c11101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 01/12/2025] [Accepted: 01/20/2025] [Indexed: 01/30/2025]
Abstract
Curcuminoids, found in turmeric (Curcuma longa L.), include curcumin (CUR), demethoxycurcumin (DMC), and bisdemethoxycurcumin (BDMC). Although CUR and DMC are well-studied, the anti-inflammatory effects of BDMC remain less explored. Recent studies highlight BDMC's stronger NF-κB inhibition compared to CUR and DMC in cell models, along with its ability to target pathways associated with inflammatory bowel disease (IBD) in DSS-induced colitis mice, reflected by lower disease activity scores and reduced inflammation. This study assessed CUR and BDMC in a DSS-induced colitis mouse model. Dietary administration of CUR or BDMC strengthened tight junction (TJ) proteins, reduced inflammatory cytokine secretion, and attenuated intestinal inflammatory protein expression, thereby alleviating DSS-induced IBD in mice. Furthermore, gut microbiota and short-chain fatty acid analyses revealed that CUR and BDMC effectively regulated gut microbial imbalance and promoted the relative abundance of butyrate-producing bacteria. Furthermore, CUR showed low absorption and was primarily excreted in feces, while BDMC had higher absorption levels. In conclusion, while both BDMC and CUR have potential as adjunct therapies for IBD, BDMC at a concentration of 0.1% showed strong anti-inflammatory effects and enhanced TJ proteins, suggesting that BDMC, even at lower concentrations than CUR, holds promising therapeutic potential and prospects.
Collapse
Affiliation(s)
- Kai-Yu Hsu
- Institute
of Food Sciences and Technology, National
Taiwan University, Taipei 10617, Taiwan
| | - Anju Majeed
- Sami-Sabinsa
Group Limited, Bengaluru, Karnataka 560058, India
| | - Chi-Tang Ho
- Department
of Food Science, Rutgers University, New Brunswick New Jersey 08901 United States
| | - Min-Hsiung Pan
- Institute
of Food Sciences and Technology, National
Taiwan University, Taipei 10617, Taiwan
- Department
of Medical Research, China Medical University Hospital, China Medical University, Taichung 40402, Taiwan
| |
Collapse
|
117
|
Kaimori JY, Sakaguchi Y, Oka T, Isaka Y. Plant-Dominant Low-Protein Diets: A Promising Dietary Strategy for Mitigating Disease Progression in People with Chronic Kidney Disease-A Comprehensive Review. Nutrients 2025; 17:643. [PMID: 40004970 PMCID: PMC11857991 DOI: 10.3390/nu17040643] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Revised: 02/04/2025] [Accepted: 02/10/2025] [Indexed: 02/27/2025] Open
Abstract
Chronic kidney disease (CKD) is a global health crisis affecting over 10% of the population, with mortality rates increasing significantly. Current management strategies, including expensive medications and renal replacement therapies, highlight the need for cost-effective, conservative approaches. This review examines the evidence for plant-dominant low-protein diets (PLADO) in managing non-dialysis-dependent CKD. Existing guidelines for protein restriction in CKD vary considerably, with inconsistencies and a lack of personalization noted in the KDOQI and KDIGO recommendations. While traditional low-protein diet trials show limited success due to poor adherence and marginal benefits, PLADO offers a potentially more sustainable alternative. PLADO's advantages include improved nutrient density, reduced dietary acid load, anti-inflammatory effects, and beneficial modulation of the gut microbiome, potentially reducing uremic toxins and improving cardiovascular health. However, challenges remain, including adherence issues, potential nutrient deficiencies, and potassium management. Although observational studies show promise, further large-scale randomized controlled trials are necessary to validate PLADO's efficacy and establish optimal dietary composition. A personalized, multidisciplinary approach is essential for successful implementation and monitoring to maximize PLADO's benefits in improving outcomes for individuals with NDD-CKD.
Collapse
Affiliation(s)
- Jun-Ya Kaimori
- Department of Health and Nutrition, Otemae University, 2-1-88 Otemae, Chuo-ku, Osaka 540-0008, Japan
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita 565-0871, Japan; (Y.S.); (T.O.); (Y.I.)
| | - Yusuke Sakaguchi
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita 565-0871, Japan; (Y.S.); (T.O.); (Y.I.)
| | - Tatsufumi Oka
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita 565-0871, Japan; (Y.S.); (T.O.); (Y.I.)
| | - Yoshitaka Isaka
- Department of Nephrology, Osaka University Graduate School of Medicine, Suita 565-0871, Japan; (Y.S.); (T.O.); (Y.I.)
| |
Collapse
|
118
|
Galiot L, Audet I, Ouattara B, Bissonnette N, Talbot G, Raymond F, Deschênes T, Lessard M, Lapointe J, Guay F, Matte JJ. Effect of Neonatal Interventions with Specific Micronutrients and Bovine Colostrum on Micronutrient and Oxidative Statuses and on Gut Microbiota in Piglets from Birth to Post-Weaning Period. Vet Sci 2025; 12:151. [PMID: 40005911 PMCID: PMC11860533 DOI: 10.3390/vetsci12020151] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 02/04/2025] [Accepted: 02/07/2025] [Indexed: 02/27/2025] Open
Abstract
This study aimed to determine the impact of supplementations of copper, vitamins A and D (ADCU), and a bovine colostrum extract (BC) on the micronutrient status, antioxidant status, and intestinal microbiota of piglets until the post-weaning period. Twenty-three sows were fed conventional gestation and lactation diets, and twenty-four sows were fed conventional diets supplemented with ADCU. For each litter, all piglets received one of four treatments during lactation: no supplementation; ADCU; BC; and ADCU + BC. Within each litter, one low (LW) and one high birth weight (HW) piglet were euthanized before and after weaning to collect liver and intestinal samples. Serum vitamin D, liver retinol, and liver Cu were greater in ADCU piglets (p < 0.01), mostly before weaning. After weaning, liver Cu decreased markedly with a drop of 75% in all treatments, despite high levels of Cu in their post-weaning diets. The antioxidant status of piglets was not globally altered by treatments (p > 0.05). For microbiota composition, sow supplementation increased (p < 0.01) richness in bacterial species in the piglet colon, either before or shortly after weaning. Short-chain fatty acids in caecal digesta were increased by sow supplementation in LW piglets before weaning at 16 days of age (p < 0.05). In conclusion, oral supplementations to piglets increased postnatal micronutrient statuses during lactation, but this did not generally persist after weaning. Treatments to sows or piglets did not improve the response of piglets to oxidative stress, but supplementation to sows favoured gut microbiota diversity, particularly in LW piglets.
Collapse
Affiliation(s)
- Lucie Galiot
- Département des Sciences Animales, Université Laval, Ville de Québec, QC G1V 0A6, Canada; (L.G.); (F.G.)
| | - Isabelle Audet
- Agriculture et Agroalimentaire Canada, Centre de Recherche et de Développement de Sherbrooke, Sherbrooke, QC J1M 0C8, Canada; (I.A.); (B.O.); (N.B.); (G.T.); (M.L.); (J.L.)
| | - Bazoumana Ouattara
- Agriculture et Agroalimentaire Canada, Centre de Recherche et de Développement de Sherbrooke, Sherbrooke, QC J1M 0C8, Canada; (I.A.); (B.O.); (N.B.); (G.T.); (M.L.); (J.L.)
- Biological Sciences, Animal Biology, Université Peleforo GON COULIBALY, Korhogo 1328, Côte d’Ivoire
| | - Nathalie Bissonnette
- Agriculture et Agroalimentaire Canada, Centre de Recherche et de Développement de Sherbrooke, Sherbrooke, QC J1M 0C8, Canada; (I.A.); (B.O.); (N.B.); (G.T.); (M.L.); (J.L.)
| | - Guylaine Talbot
- Agriculture et Agroalimentaire Canada, Centre de Recherche et de Développement de Sherbrooke, Sherbrooke, QC J1M 0C8, Canada; (I.A.); (B.O.); (N.B.); (G.T.); (M.L.); (J.L.)
| | - Frédéric Raymond
- École de Nutrition, Centre Nutrition, Santé et Société (NUTRISS), et Institut de la Nutrition et des Aliments Fonctionnels (INAF), Université Laval, Ville de Québec, QC G1V 0A6, Canada; (F.R.); (T.D.)
| | - Thomas Deschênes
- École de Nutrition, Centre Nutrition, Santé et Société (NUTRISS), et Institut de la Nutrition et des Aliments Fonctionnels (INAF), Université Laval, Ville de Québec, QC G1V 0A6, Canada; (F.R.); (T.D.)
| | - Martin Lessard
- Agriculture et Agroalimentaire Canada, Centre de Recherche et de Développement de Sherbrooke, Sherbrooke, QC J1M 0C8, Canada; (I.A.); (B.O.); (N.B.); (G.T.); (M.L.); (J.L.)
| | - Jérôme Lapointe
- Agriculture et Agroalimentaire Canada, Centre de Recherche et de Développement de Sherbrooke, Sherbrooke, QC J1M 0C8, Canada; (I.A.); (B.O.); (N.B.); (G.T.); (M.L.); (J.L.)
| | - Frédéric Guay
- Département des Sciences Animales, Université Laval, Ville de Québec, QC G1V 0A6, Canada; (L.G.); (F.G.)
| | - Jean Jacques Matte
- Agriculture et Agroalimentaire Canada, Centre de Recherche et de Développement de Sherbrooke, Sherbrooke, QC J1M 0C8, Canada; (I.A.); (B.O.); (N.B.); (G.T.); (M.L.); (J.L.)
| |
Collapse
|
119
|
Paul JK, Azmal M, Haque ANMSNB, Meem M, Talukder OF, Ghosh A. Unlocking the secrets of the human gut microbiota: Comprehensive review on its role in different diseases. World J Gastroenterol 2025; 31:99913. [PMID: 39926224 PMCID: PMC11718612 DOI: 10.3748/wjg.v31.i5.99913] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Revised: 10/25/2024] [Accepted: 12/05/2024] [Indexed: 12/30/2024] Open
Abstract
The human gut microbiota, a complex and diverse community of microorganisms, plays a crucial role in maintaining overall health by influencing various physiological processes, including digestion, immune function, and disease susceptibility. The balance between beneficial and harmful bacteria is essential for health, with dysbiosis - disruption of this balance - linked to numerous conditions such as metabolic disorders, autoimmune diseases, and cancers. This review highlights key genera such as Enterococcus, Ruminococcus, Bacteroides, Bifidobacterium, Escherichia coli, Akkermansia muciniphila, Firmicutes (including Clostridium and Lactobacillus), and Roseburia due to their well-established roles in immune regulation and metabolic processes, but other bacteria, including Clostridioides difficile, Salmonella, Helicobacter pylori, and Fusobacterium nucleatum, are also implicated in dysbiosis and various diseases. Pathogenic bacteria, including Escherichia coli and Bacteroides fragilis, contribute to inflammation and cancer progression by disrupting immune responses and damaging tissues. The potential for microbiota-based therapies, such as probiotics, prebiotics, fecal microbiota transplantation, and dietary interventions, to improve health outcomes is examined. Future research directions in the integration of multi-omics, the impact of diet and lifestyle on microbiota composition, and advancing microbiota engineering techniques are also discussed. Understanding the gut microbiota's role in health and disease is essential for formulating personalized, efficacious treatments and preventive strategies, thereby enhancing health outcomes and progressing microbiome research.
Collapse
Affiliation(s)
- Jibon Kumar Paul
- Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Mahir Azmal
- Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - ANM Shah Newaz Been Haque
- Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Meghla Meem
- Faculty of Medicine, Dhaka University, Dhaka 1000, Bangladesh
| | - Omar Faruk Talukder
- Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| | - Ajit Ghosh
- Biochemistry and Molecular Biology, Shahjalal University of Science and Technology, Sylhet 3114, Bangladesh
| |
Collapse
|
120
|
Rubio-Casillas A, Rodríguez-Quintero CM, Hromić-Jahjefendić A, Uversky VN, Redwan EM, Brogna C. The essential role of prebiotics in restoring gut health in long COVID. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2025; 213:385-411. [PMID: 40246350 DOI: 10.1016/bs.pmbts.2025.01.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/19/2025]
Abstract
The gut microbiota (GM) plays an essential role in human health, influencing not only digestive processes but also the immune system´s functionality. The COVID-19 pandemic has highlighted the complex interaction between viral infections and the GM. Emerging evidence has demonstrated that SARS-CoV-2 can disrupt microbial homeostasis, leading to dysbiosis and compromised immune responses. The severity of COVID-19 has been associated with a reduction in the abundance of several beneficial bacteria in the gut. It has been proposed that consuming probiotics may help to re-colonize the GM. Although probiotics are important, prebiotics are essential for their metabolism, growth, and re-colonization capabilities. This chapter delves into the critical role of prebiotics in restoring GM after COVID-19 disease. The mechanisms by which prebiotics enhance the metabolism of beneficial bacteria will be described, and how prebiotics mediate the re-colonization of the gut with beneficial bacteria, thereby restoring microbial diversity and promoting the resilience of the gut-associated immune system. The benefits of consuming prebiotics from natural sources are superior to those from chemically purified commercial products.
Collapse
Affiliation(s)
- Alberto Rubio-Casillas
- Autlan Regional Hospital, Jalisco Health Services, Autlan, Jalisco, Mexico; Biology Laboratory, Autlan Regional Preparatory School, University of Guadalajara, Autlan, Jalisco, Mexico.
| | | | - Altijana Hromić-Jahjefendić
- Department of Genetics and Bioengineering, Faculty of Engineering and Natural Sciences, International University of Sarajevo, Sarajevo, Bosnia and Herzegovina.
| | - Vladimir N Uversky
- Department of Molecular Medicine and USF Health Byrd Alzheimer's Research Institute, Morsani College of Medicine, University of South Florida, Tampa, FL, United States; Laboratory of New Methods in Biology, Institute for Biological Instrumentation of the Russian Academy of Sciences, Federal Research Center "Pushchino Scientific Center for Biological Research of the Russian Academy of Sciences", Pushchino, Russia.
| | - Elrashdy M Redwan
- Department of Biological Sciences, Faculty of Science, King Abdulaziz University, Jeddah, Saudi Arabia; Therapeutic and Protective Proteins Laboratory, Protein Research Department, Genetic Engineering and Biotechnology Research Institute, City for Scientific Research and Technology Applications, New Borg EL-Arab, Alexandria, Egypt
| | - Carlo Brogna
- Craniomed Group Srl, Research Facility, Montemiletto (Av), Italy
| |
Collapse
|
121
|
Molan K, Ambrožič Avguštin J, Likar M, Pongrac Barlovic D, Žgur Bertok D, Starčič Erjavec M. Fecal Short-Chain Fatty Acids Are Associated with Obesity in Gestational Diabetes. Biomedicines 2025; 13:387. [PMID: 40002799 PMCID: PMC11853429 DOI: 10.3390/biomedicines13020387] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/31/2025] [Accepted: 02/03/2025] [Indexed: 02/27/2025] Open
Abstract
Background: Short-chain fatty acids (SCFAs), which are produced by the microbial fermentation of undigested carbohydrates, play an important role in the metabolism and physiology of the host. SCFAs are involved in the regulation of maternal metabolism during pregnancy and influence weight gain, glucose metabolism, and metabolic hormones. Methods: In 2017, women who were treated for gestational diabetes mellitus (GDM) at the University Medical Centre Ljubljana were invited to participate in a longitudinal study. A total of 45 women were included in this study and comprehensively phenotyped. During the second and third trimester of pregnancy, the women with GDM provided fecal samples for SCFA analysis. The samples were analyzed by high-performance liquid chromatography for the simultaneous determination of acetate, propionate, and butyrate. Results: SCFA concentrations in feces differed between overweight/obese and normal-weight women with GDM. Acetate and propionate concentrations were significantly higher in pregnant women who were overweight or obese before pregnancy compared to normal-weight women but butyrate concentrations were not. Butyrate was elevated in the third trimester in the group with excessive gestational weight gain. Conclusions: The relationship between SCFAs and obesity is complex, and the association between SCFAs and GDM remains to be clarified. Regardless of the conflicting publications on the role of SCFAs, our study showed that higher acetate and propionate levels were associated with the weight categories of overweight or obesity before pregnancy and higher butyrate levels were associated with excessive gestational weight gain.
Collapse
Affiliation(s)
- Katja Molan
- Faculty of Health Sciences, University of Novo mesto, 8000 Novo mesto, Slovenia;
| | - Jerneja Ambrožič Avguštin
- Department of Biology, Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (J.A.A.); (M.L.); (D.Ž.B.)
| | - Matevž Likar
- Department of Biology, Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (J.A.A.); (M.L.); (D.Ž.B.)
| | - Drazenka Pongrac Barlovic
- Department of Endocrinology, Diabetes and Metabolic Diseases, University Medical Centre, 1000 Ljubljana, Slovenia;
- Faculty of Medicine, University of Ljubljana, 1000 Ljubljana, Slovenia
| | - Darja Žgur Bertok
- Department of Biology, Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia; (J.A.A.); (M.L.); (D.Ž.B.)
| | - Marjanca Starčič Erjavec
- Department of Microbiology, Biotechnical Faculty, University of Ljubljana, 1000 Ljubljana, Slovenia
- Department of Biology, Faculty of Natural Sciences and Mathematics, University of Maribor, 2000 Maribor, Slovenia
| |
Collapse
|
122
|
Enache RM, Roşu OA, Profir M, Pavelescu LA, Creţoiu SM, Gaspar BS. Correlations Between Gut Microbiota Composition, Medical Nutrition Therapy, and Insulin Resistance in Pregnancy-A Narrative Review. Int J Mol Sci 2025; 26:1372. [PMID: 39941139 PMCID: PMC11818759 DOI: 10.3390/ijms26031372] [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: 12/09/2024] [Revised: 01/31/2025] [Accepted: 02/04/2025] [Indexed: 02/16/2025] Open
Abstract
Many physiological changes accompany pregnancy, most of them involving metabolic perturbations. Alterations in microbiota composition occur both before and during pregnancy and have recently been correlated with an important role in the development of metabolic complications, such as insulin resistance and gestational diabetes mellitus (GDM). These changes may be influenced by physiological adaptations to pregnancy itself, as well as by dietary modifications during gestation. Medical nutritional therapy (MNT) applied to pregnant women at risk stands out as one of the most important factors in increasing the microbiota's diversity at both the species and genus levels. In this review, we discuss the physiological changes during pregnancy and their impact on the composition of the intestinal microbiota, which may contribute to GDM. We also discuss findings from previous studies regarding the effectiveness of MNT in reducing insulin resistance. In the future, additional studies should aim to identify specific gut microbial profiles that serve as early indicators of insulin resistance during gestation. Early diagnosis, achievable through stool analysis or metabolite profiling, may facilitate the timely implementation of dietary or pharmaceutical modifications, thereby mitigating the development of insulin resistance and its associated sequelae.
Collapse
Affiliation(s)
- Robert-Mihai Enache
- Department of Radiology and Medical Imaging, Fundeni Clinical Institute, 022328 Bucharest, Romania;
| | - Oana Alexandra Roşu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (O.A.R.); (M.P.); (L.A.P.)
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania
| | - Monica Profir
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (O.A.R.); (M.P.); (L.A.P.)
- Department of Oncology, Elias University Emergency Hospital, 011461 Bucharest, Romania
| | - Luciana Alexandra Pavelescu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (O.A.R.); (M.P.); (L.A.P.)
| | - Sanda Maria Creţoiu
- Department of Morphological Sciences, Cell and Molecular Biology and Histology, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania; (O.A.R.); (M.P.); (L.A.P.)
| | - Bogdan Severus Gaspar
- Department of Surgery, Carol Davila University of Medicine and Pharmacy, 050474 Bucharest, Romania;
- Surgery Clinic, Bucharest Emergency Clinical Hospital, 014461 Bucharest, Romania
| |
Collapse
|
123
|
Ashiqueali SA, Hayslip N, Chaudhari DS, Schneider A, Zhu X, Rubis B, Seavey CE, Alam MT, Hussein R, Noureddine SA, Golusinska-Kardach E, Pazdrowski P, Yadav H, Masternak MM. Fecal microbiota transplant from long-living Ames dwarf mice alters the microbial composition and biomarkers of liver health in normal mice. GeroScience 2025:10.1007/s11357-025-01539-3. [PMID: 39904968 DOI: 10.1007/s11357-025-01539-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2024] [Accepted: 01/20/2025] [Indexed: 02/06/2025] Open
Abstract
Aging is associated with intestinal dysbiosis, a condition characterized by diminished microbial biodiversity and inflammation. This leads to increased vulnerability to extraintestinal manifestations such as autoimmune, metabolic, and neurodegenerative conditions thereby accelerating mortality. As such, modulation of the gut microbiome is a promising way to extend healthspan. In this study, we explore the effects of fecal microbiota transplant (FMT) from long-living Ames dwarf donors to their normal littermates, and vice versa, on the recipient gut microbiota and liver transcriptome. Importantly, our previous studies highlight differences between the microbiome of Ames dwarf mice relative to their normal siblings, potentially contributing to their extended lifespan and remarkable healthspan. Our findings demonstrate that FMT from Ames dwarf mice to normal mice significantly alters the recipient's gut microbiota, potentially reprogramming bacterial functions related to healthy aging, and changes the liver transcriptome, indicating improved metabolic health. Particularly, the microbiome of Ames dwarf mice, characterized by a higher abundance of beneficial bacterial families such as Peptococcaceae, Oscillospiraceae, and Lachnospiraceae, appears to play a crucial role in modulating these effects. Alongside, our mRNA sequencing and RT-PCR validation reveals that FMT may contribute to the significant downregulation of p21, Elovl3, and Insig2, genes involved with cellular senescence and liver metabolic pathways. Our data suggest a regulatory axis exists between the gut and liver, highlighting the potential of microbiome-targeted therapies in promoting healthy aging. Future research should focus on functional validation of altered microbial communities and explore the underlying biomolecular pathways that confer geroprotection.
Collapse
Affiliation(s)
- Sarah A Ashiqueali
- Robert and Arlene Kogod Center on Aging, Mayo Clinic, 200 First Street SW, Rochester, MN, 55905, USA
- Department of Physical Medicine and Rehabilitation, Mayo Clinic, Rochester, MN, USA
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, USA
| | - Natalie Hayslip
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, USA
- University of South Florida (USF) Morsani College of Medicine, Tampa, FL, USA
| | - Diptaraj S Chaudhari
- Research and Development Service, John D. Dingell VA Medical Center, Detroit, MI, USA
- Department of Physiology, Wayne State University School of Medicine, Detroit, MI, USA
- USF Center for Microbiome Research, Microbiomes Institute, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Augusto Schneider
- Faculdade de Nutrição, Universidade Federal de Pelotas, Pelotas, Brazil
| | - Xiang Zhu
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, USA
| | - Blazej Rubis
- Department of Clinical Chemistry and Molecular Diagnostics, Poznań University of Medical Sciences, Poznań, Poland
| | - Corey E Seavey
- Enteric Neuroscience Program (ENSP), Division of Gastroenterology & Hepatology, Mayo Clinic, Rochester, MN, USA
- Department of Physiology & Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
- Department of Internal Medicine, University of Central Florida College of Medicine, Orlando, FL, USA
| | - Md Tanjim Alam
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, USA
| | - Ridwan Hussein
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, USA
- Sidney Kimmel Medical College, Philadelphia, PA, USA
| | - Sarah A Noureddine
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, USA
| | - Ewelina Golusinska-Kardach
- Department of Dental Surgery, Periodontology and Oral Mucosa Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Pawel Pazdrowski
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, USA
- Poznan University of Medical Sciences, Student Scientific Association, Poznan, Poland
| | - Hariom Yadav
- USF Center for Microbiome Research, Microbiomes Institute, Department of Neurosurgery and Brain Repair, University of South Florida Morsani College of Medicine, Tampa, FL, USA
| | - Michal M Masternak
- Burnett School of Biomedical Sciences, University of Central Florida College of Medicine, Orlando, FL, USA.
- Department of Head and Neck Surgery, Poznan University of Medical Sciences, Poznan, Poland.
| |
Collapse
|
124
|
Zhou M, Luo Y, Qiu J, Wang H, Li X, Zhang K, Li X, Yaqoob MU, Wang M. Effects of dietary supplementation with butyrate glycerides on lipid metabolism, intestinal morphology, and microbiota population in laying hens. Poult Sci 2025; 104:104755. [PMID: 39862486 PMCID: PMC11803851 DOI: 10.1016/j.psj.2024.104755] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Revised: 12/26/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
The present study investigated the impact of butyrate glycerides (BG) on lipid metabolism, intestinal morphology, and microbiota of laying hens. Four hundred eighty 54-week-old Hy-line Brown laying hens were randomly selected and divided into five groups. The control group (ND) was fed a basal diet. Meanwhile, the remaining groups were given a basal supplemented with 0.5, 1, 2, and 4 g/kg of the product containing BG and were designated as BG-0.5, BG-1, BG-2, and BG-4 groups, respectively. The findings showed that: (1) BG supplementation significantly decreased (P < 0.001) the blood Glu levels (BG-0.5, BG-1, BG-2, and BG-4) and increased (P < 0.001) the serum HDL-C levels (BG-2, and BG-4). (2) The BG-2 and BG-4 groups showed an increase (P < 0.01) in abdominal lipid HSL activity. (3) The levels of hepatic TC and TG in all BG groups were significantly decreased (P < 0.05). (4) The addition of BG resulted in a significant reduction in the mRNA expression of the liver X receptor alpha (LXRα) (P < 0.05). (5) All BG groups presented a substantial reduction in duodenal crypt depth and a notable increase in the ratio of villus height to crypt depth (V/C) (P < 0.01). Additionally, all BG groups exhibited a significant increase in villus height in the ileum (P < 0.001). (6) Both the BG-1 and BG-4 groups exhibited a significant reduction in the amounts of n-butyric and n-glutaric acids in the cecum contents (P < 0.05). (7) The inclusion of BG did not substantially impact the diversity of cecal microbiota in laying hens. However, it dramatically boosted the proportion of the beneficial bacterium Alistipes (P < 0.05) and reduced the abundance of the harmful bacterium Verrucomicrobiota (P < 0.05). Overall, incorporating BG with glycerol monobutyrate as the diet's primary active component reduces fat accumulation in laying hens' blood and liver. It potentially regulates lipid metabolism via the PPARγ-LXRα-SREBP1c pathway. Additionally, BG has the potential to enhance the structure of the small intestine's mucous membrane and increase the presence of beneficial bacteria. Under the experimental conditions, late-laying hens supplemented with 4 g/kg BG performed best overall.
Collapse
Affiliation(s)
- Minyao Zhou
- College of Animal Science, Zhejiang University, Hangzhou 310058, China
| | - Yanqiu Luo
- College of Animal Science, Zhejiang University, Hangzhou 310058, China
| | - Ji Qiu
- College of Animal Science, Zhejiang University, Hangzhou 310058, China
| | - Haidong Wang
- College of Animal Science, Zhejiang University, Hangzhou 310058, China
| | - Xinyu Li
- College of Animal Science, Zhejiang University, Hangzhou 310058, China
| | - Kexin Zhang
- College of Animal Science, Zhejiang University, Hangzhou 310058, China
| | - Xiaoteng Li
- College of Animal Science, Zhejiang University, Hangzhou 310058, China
| | | | - Minqi Wang
- College of Animal Science, Zhejiang University, Hangzhou 310058, China.
| |
Collapse
|
125
|
O'Connor G, Velandia AC, Capriles ZH. The impact of an enteral formula with food-derived ingredients on dietetic practice at a specialist children's hospital in the UK: Retrospective study. J Hum Nutr Diet 2025; 38:e13374. [PMID: 39410807 PMCID: PMC11589405 DOI: 10.1111/jhn.13374] [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: 03/08/2024] [Accepted: 09/07/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND Blended tube feeds are reported to be better tolerated in some children compared to standard commercial enteral formulas, allowing children to normalise feeding by having similar foods as the rest of the family. However, a blended tube feed is contraindicated in patients who are immunocompromised or require post-pyloric feeding as a result of a food safety risk. Other contraindications for blended diet include children who require continuous pump feeding via gastrostomy or nasogastric feeding tube (< 12 Fr) and fluid restrictions. To meet the demands of consumers, manufacturers have developed enteral formulas with food-derived ingredients (EFI). Commercially available EFIs are relatively novel in the UK. The present study aimed to monitor the implementation of an EFI by dietitians in a specialist children's hospital. METHODS A single-centre retrospective study was conducted to monitor the dietetic practice of commencing a commercially available EFI (Compleat® paediatric; Nestlé Health Science; 1.2 kcal/ml with 14% food-derived ingredients). Using electronic medical notes, data were collected on all children who commenced an EFI via an enteral feeding tube in a specialist paediatric hospital between August 2022 and December 2023. Data were gathered on demographics (age, sex and primary diagnosis), anthropometric measurements (weight-for-age Z-score and height-for-age Z-score), feed regimens (feed volume, feeding route, mode of feeding [continuous, bolus]), gastrointestinal symptoms (gastro-oesophageal reflux, vomiting, abdominal discomfort, constipation and loose stools) and geographical discharge area for children on home enteral nutrition. RESULTS Seventy children were included in the analysis. The mean ± SD age was 4.7 ± 6 years. The median admission weight-for-age Z-score was -1.50. The most common primary diagnosis was a neurological impairment in 37/70 (47%) children. Most children were fed via a percutaneous endoscopic gastrostomy 31/70 (44%) and 8/70 (11%) of the children fed directly into the jejunum. The most common reason being gastrointestinal symptoms, 58/70 (83%). The most common gastrointestinal symptom reported before commencing an EFI was loose stools in 22/58 (38%) children. Within 7 days of commencing an EFI, there was reported improvement in gastrointestinal symptoms in all categories. In total, 42/70 children were discharged on an EFI. CONCLUSIONS In our specialist children's hospital, EFI is primarily implemented by dietitians in children who are already established on an enteral formula displaying gastrointestinal symptoms. However, dietitians are increasingly implementing an EFI as their first-line whole protein enteral formula. Furthermore, an EFI was also implemented as a compromise to a blended diet.
Collapse
Affiliation(s)
- Graeme O'Connor
- Department of DieteticsGreat Ormond Street Hospital Foundation TrustLondonUK
| | | | | |
Collapse
|
126
|
Fernández-Rodríguez R, Bizzozero-Peroni B, Díaz-Goñi V, Garrido-Miguel M, Bertotti G, Roldán-Ruiz A, López-Moreno M. Plant-based meat alternatives and cardiometabolic health: a systematic review and meta-analysis. Am J Clin Nutr 2025; 121:274-283. [PMID: 39653176 DOI: 10.1016/j.ajcnut.2024.12.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/28/2024] [Accepted: 12/04/2024] [Indexed: 12/26/2024] Open
Abstract
BACKGROUND Plant-based meat alternatives (PBMAs) are emerging in global markets. However, the effects of substituting meat for PBMAs on cardiometabolic health are uncertain. OBJECTIVES This study aimed to determine the effects of replacing meat consumption by PBMAs on cardiometabolic parameters in adults. METHODS Five databases were systematically explored from inception to July 2024, searching for RCTs assessing the effects of replacing meat consumption by PBMAs on cardiometabolic parameters in adults without cardiovascular diseases. Meta-analyses were conducted when ≥4 studies addressed the same outcome (i.e. blood lipids, blood pressure, fasting glucose, and body weight). Pooled raw mean differences (MDs) with their 95% CIs were estimated using a random-effects method. Sensitivity analyses were conducted to assess the robustness of our estimates. RESULTS Eight publications from 7 RCTs comprising 369 adults (60% females; mean age range: 24-61 y) were included. The substitution of PBMAs for meat was associated with significant reductions in LDL-cholesterol: -0.25 mmol/L (95% CI: -0.42, -0.08 mmol/L; I2 = 65.8%; n = 7), total cholesterol (TC): -0.29 mmol/L (95% CI: -0.52, -0.06 mmol/L; I2 = 64.8.%; n = 6), and body weight: -0.72 kg (95% CI: -1.02, -0.42 kg; I2 = 0%; n = 5). No significant changes were shown in HDL-cholesterol, triglycerides, blood pressure, or fasting glucose concentrations. Sensitivity analyses considering mycoprotein-based alternatives showed a significant reduction in LDL-cholesterol (MD: -0.37 mmol/L; 95% CI: -0.61, -0.13 mmol/L; I2 = 52.5%; n = 4), and TC (MD: -0.39 mmol/L; 95% CI: -0.56, -0.21 mmol/L; I2 = 0%; n = 4). CONCLUSIONS Our findings suggest substituting PBMAs for meat for ≤8 wk lowered TC (6%), LDL-cholesterol (12%), and body weight (1%) in adults without cardiovascular diseases. PBMAs may facilitate the transition to a plant-based diet, but long-term studies are needed to evaluate their cardiometabolic effects. This trial was registered at PROSPERO as CRD42024556191.
Collapse
Affiliation(s)
- Rubén Fernández-Rodríguez
- Department of Physical Education and Sports, Faculty of Sports Science, Sport and Health University Research Institute (iMUDS), University of Granada, Granada, Spain
| | - Bruno Bizzozero-Peroni
- Health and Social Research Center, Faculty of Nursing, Universidad de Castilla-La Mancha, Cuenca, Spain; Instituto Superior de Educación Física, Universidad de la República, Rivera, Uruguay.
| | - Valentina Díaz-Goñi
- Health and Social Research Center, Faculty of Nursing, Universidad de Castilla-La Mancha, Cuenca, Spain
| | - Miriam Garrido-Miguel
- Health and Social Research Center, Faculty of Nursing, Universidad de Castilla-La Mancha, Cuenca, Spain; Research Network on Chronicity, Primary Care and Health Promotion (RICAPPS), Cuenca, Spain; Faculty of Nursing, Universidad de Castilla-La Mancha, Albacete, Spain
| | - Gabriele Bertotti
- Diet, Planetary Health and Performance, Faculty of Health Sciences, Universidad Francisco de Vitoria, Pozuelo, Madrid, Spain; School of Physiotherapy, Faculty of Health Sciences, Universidad Francisco de Vitoria, Madrid, Spain
| | - Alberto Roldán-Ruiz
- Diet, Planetary Health and Performance, Faculty of Health Sciences, Universidad Francisco de Vitoria, Pozuelo, Madrid, Spain; School of Physiotherapy, Faculty of Health Sciences, Universidad Francisco de Vitoria, Madrid, Spain
| | - Miguel López-Moreno
- Diet, Planetary Health and Performance, Faculty of Health Sciences, Universidad Francisco de Vitoria, Pozuelo, Madrid, Spain; School of Physiotherapy, Faculty of Health Sciences, Universidad Francisco de Vitoria, Madrid, Spain
| |
Collapse
|
127
|
Li S, Duan Y, Luo S, Zhou F, Wu Q, Lu Z. Short-chain fatty acids and cancer. Trends Cancer 2025; 11:154-168. [PMID: 39638744 DOI: 10.1016/j.trecan.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Revised: 11/04/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024]
Abstract
Short-chain fatty acids (SCFAs), derived from the diet and the microbiota, serve as crucial links between the diet, gut microbiota, metabolism, immunity, and cancer. They function as energy sources through β-oxidation and regulate macromolecular synthesis, G protein-coupled receptor (GPCR) and histone deacetylase (HDAC) activities, protein modifications, signaling pathways, and gene expression in cells within the tumor microenvironment, particularly in tumor and immune cells. The critical role of SCFAs in maintaining normal homeostasis and influencing tumor progression highlights the potential of targeting SCFA-mediated cellular processes for cancer prevention and treatment.
Collapse
Affiliation(s)
- Shan Li
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310029, China; Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310029, China
| | - Yixin Duan
- Department of Oncology, Cancer Institute of The Affiliated Hospital of Qingdao University, Qingdao University, Qingdao Cancer Institute, Qingdao, Shandong 266071, China
| | - Shudi Luo
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310029, China; Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310029, China
| | - Fangxin Zhou
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310029, China; Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310029, China
| | - Qingang Wu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310029, China; Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310029, China
| | - Zhimin Lu
- Zhejiang Key Laboratory of Pancreatic Disease, The First Affiliated Hospital, Zhejiang Key Laboratory of Frontier Medical Research on Cancer Metabolism, and Institute of Translational Medicine, Zhejiang University School of Medicine, Hangzhou, Zhejiang 310029, China; Institute of Fundamental and Transdisciplinary Research, Cancer Center, Zhejiang University, Hangzhou, Zhejiang 310029, China.
| |
Collapse
|
128
|
Zhao HJ, Chen Y, Liu T, McArthur K, Mueller NT. Short-Chain Fatty Acids and Preeclampsia: A Scoping Review. Nutr Rev 2025; 83:e683-e693. [PMID: 38796843 PMCID: PMC11723139 DOI: 10.1093/nutrit/nuae057] [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: 05/29/2024] Open
Abstract
BACKGROUND Preeclampsia (PE) is a pregnancy-associated hypertension disorder with high morbidity and mortality. Short-chain fatty acids (SCFAs)-molecules produced by gut microbes-have been associated with hypertension, yet their relation to PE remains uncertain. OBJECTIVES The aim was to review existing human studies that examined associations of the major SCFAs (acetate, propionate, butyrate) in pregnancy with PE development. METHODS Two reviewers independently searched online databases (EMBASE, PubMed, Web of Science, and Cochrane Database of Systematic Reviews) in January 2024 using the following terms: "short-chain fatty acids," "acetic acid," "butyric acid," "propionic acid," and "preeclampsia." The final set of included studies had to report associations of SCFAs with PE, be peer-reviewed, be written in English, and be conducted in humans. RESULTS The abstracts of 907 studies were screened; 43 underwent full-text screening and 11 (1318 total participants, 352 with PE) were included in the final review. All studies used a case-control design. SCFAs were measured in a range of biospecimens (eg, serum, plasma, feces, placentas, and amniotic fluid) that were collected at distinct time points in pregnancy. All 7 studies that investigated butyrate found that it was lower in PE cases than in controls, with 6 of these showing statistical significance (P < .05). Five studies showed that acetate was significantly lower in individuals with PE compared with healthy individuals, while 1 study found that acetate was significantly higher in PE cases. One study reported significantly higher propionate among PE cases vs controls, while 2 studies reported significantly lower propionate levels in PE cases. The nuance in results for acetate and propionate may owe to reasons such as differences in distributions of population characteristics associated with SCFA level and PE or type of PE (early vs late). CONCLUSION Current epidemiologic evidence, which derives only from case-control studies, suggests that SCFAs, particularly butyrate (protective), in pregnancy are related to the development of PE. Large-cohort studies are warranted to investigate the temporality and potential causality of these associations.
Collapse
Affiliation(s)
- Heather J Zhao
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, United States
- Temerty School of Medicine, University of Toronto, Toronto, ON M5S1A8, Canada
| | - Yingan Chen
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO 80045, United States
| | - Tiange Liu
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, United States
| | - Kristen McArthur
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, United States
| | - Noel T Mueller
- Department of Epidemiology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD 21205, United States
- Lifecourse Epidemiology of Adiposity and Diabetes (LEAD) Center, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
- Department of Epidemiology, Colorado School of Public Health, Aurora, CO 80045, United States
- Department of Pediatrics, Section of Nutrition, University of Colorado Anschutz Medical Campus, Aurora, CO 80045, United States
| |
Collapse
|
129
|
Azcona JA, Wacker AS, Lee CH, Fung EK, Jeitner TM, Manzo OL, Di Lorenzo A, Babich JW, Amor-Coarasa A, Kelly JM. 2-[ 18F]Fluoropropionic Acid PET Imaging of Doxorubicin-Induced Cardiotoxicity. Mol Imaging Biol 2025; 27:109-119. [PMID: 39810069 PMCID: PMC11805620 DOI: 10.1007/s11307-024-01978-y] [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: 08/07/2024] [Revised: 10/21/2024] [Accepted: 12/10/2024] [Indexed: 01/16/2025]
Abstract
PURPOSE Treatment of pediatric cancers with doxorubicin is a common and predictable cause of cardiomyopathy. Early diagnosis of treatment-induced cardiotoxicity and intervention are major determinants for the prevention of advanced disease. The onset of cardiomyopathies is often accompanied by profound changes in lipid metabolism, including an enhanced uptake of short-chain fatty acids (SCFA). Therefore, we explored the utility of 2-[18F]fluoropropionic acid ([18F]FPA), an SCFA analog, as an imaging biomarker of cardiac injury in mice exposed to doxorubicin. PROCEDURES Cardiotoxicity and cardiac dysfunction were induced in mice by an 8-dose regimen of doxorubicin (cumulative dose 24 mg/kg) administered over 14 days. The effects of doxorubicin exposure were assessed by measurement of heart weights, left ventricular ejection fractions, and blood cardiac troponin levels. Whole body and cardiac [18F]FPA uptakes were determined by PET and tissue gamma counting in the presence or absence of AZD3965, a pharmacological inhibitor of monocarboxylate transporter 1 (MCT1). Radiation absorbed doses were estimated using tissue time-activity concentrations. RESULTS Significantly higher cardiac [18F]FPA uptake was observed in doxorubicin-treated animals. This uptake remained constant from 30 to 120 min post-injection. Pharmacological inhibition of MCT1-mediated transport by AZD3965 selectively decreased the uptake of [18F]FPA in tissues other than the heart. Co-administration of [18F]FPA and AZD3965 enhanced the imaging contrast of the diseased heart while reducing overall exposure to radioactivity. CONCLUSIONS [18F]FPA, especially when co-administered with AZD3965, is a new tool for imaging changes in fatty acid metabolism occurring in response to doxorubicin-induced cardiomyopathy by PET.
Collapse
Affiliation(s)
- Juan A Azcona
- Department of Radiology, Weill Cornell Medicine, 413 E 69th Street, Room BB-1604, New York, NY, 10021, USA
| | - Anja S Wacker
- Department of Radiology, Weill Cornell Medicine, 413 E 69th Street, Room BB-1604, New York, NY, 10021, USA
| | - Chul-Hee Lee
- Department of Radiology, Weill Cornell Medicine, 413 E 69th Street, Room BB-1604, New York, NY, 10021, USA
| | - Edward K Fung
- Department of Radiology, Weill Cornell Medicine, 413 E 69th Street, Room BB-1604, New York, NY, 10021, USA
- Citigroup Biomedical Imaging Center, Weill Cornell Medicine, New York, NY, USA
| | - Thomas M Jeitner
- Department of Radiology, Weill Cornell Medicine, 413 E 69th Street, Room BB-1604, New York, NY, 10021, USA
| | - Onorina L Manzo
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Annarita Di Lorenzo
- Department of Pathology and Laboratory Medicine, Weill Cornell Medicine, New York, NY, USA
- Cardiovascular Research Institute, Weill Cornell Medicine, New York, NY, USA
- Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - John W Babich
- Department of Radiology, Weill Cornell Medicine, 413 E 69th Street, Room BB-1604, New York, NY, 10021, USA
- Citigroup Biomedical Imaging Center, Weill Cornell Medicine, New York, NY, USA
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA
- Ratio Therapeutics, Boston, MA, USA
| | - Alejandro Amor-Coarasa
- Department of Radiology, Albert Einstein College of Medicine of Yeshiva University, New York, NY, USA
- Ratio Therapeutics, Boston, MA, USA
| | - James M Kelly
- Department of Radiology, Weill Cornell Medicine, 413 E 69th Street, Room BB-1604, New York, NY, 10021, USA.
- Citigroup Biomedical Imaging Center, Weill Cornell Medicine, New York, NY, USA.
- Sandra and Edward Meyer Cancer Center, Weill Cornell Medicine, New York, NY, USA.
| |
Collapse
|
130
|
Hogenelst K, Krone T, Eveleens Maarse B, Warnke I, Snabel J, van den Broek TJ, Schuren F, Moerland M, Hoevenaars FP. A prebiotic intervention improves mood in everyday life in healthy women but not in men: Exploratory results from a larger double-blind placebo controlled cross-over study. Brain Behav Immun Health 2025; 43:100918. [PMID: 39717875 PMCID: PMC11665422 DOI: 10.1016/j.bbih.2024.100918] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Revised: 11/15/2024] [Accepted: 11/27/2024] [Indexed: 12/25/2024] Open
Abstract
Prebiotic dietary fiber (PDF) may reduce feelings of stress or improve mood in healthy individuals. Yet gut intervention studies that focus on mood in daily life are lacking and few studies include extensive biological sample analyses to gain mechanistic insights. As part of a larger randomized placebo-controlled crossover study including healthy individuals, we explored the effects of 12 weeks of PDF (acacia gum and carrot powder) on everyday mood, as measured with ecological momentary assessment (EMA). Microbiome composition and levels of microbial metabolites, endocrine, and inflammatory markers were determined prior to and after both intervention phases. Fifty-four participants completed the study. The intervention significantly increased daily positive affect (PA) and reduced daily negative affect (NA) in female but not male participants. The intervention-induced reduction in NA was associated with an increase in microbial diversity in female participants. The intervention did not significantly affect levels of fecal short chain fatty acids, cortisol, and inflammatory markers. This is one of the first studies to show that a dietary fiber intervention can positively alter mood as it is experienced in everyday life. Overall, our findings may stimulate more targeted gut-microbiome interventions and detection of its mental health effects in real life.
Collapse
Affiliation(s)
- Koen Hogenelst
- Department of Human Performance, The Netherlands Organisation for Applied Scientific Research (TNO), Soesterberg, the Netherlands
| | - Tanja Krone
- Department of Risk Analysis for Products in Development, The Netherlands Organisation for Applied Scientific Research (TNO), Utrecht, the Netherlands
| | - Boukje Eveleens Maarse
- Centre for Human Drug Research, Leiden, the Netherlands
- Leiden University Medical Center, Leiden, the Netherlands
| | | | - Jessica Snabel
- Department of Metabolic Health Research, The Netherlands Organisation for Applied Scientific Research (TNO), Leiden, the Netherlands
| | - Tim J. van den Broek
- Department of Microbiology and Systems Biology, The Netherlands Organisation for Applied Scientific Research (TNO), Leiden, the Netherlands
| | - Frank Schuren
- Department of Microbiology and Systems Biology, The Netherlands Organisation for Applied Scientific Research (TNO), Leiden, the Netherlands
| | - Matthijs Moerland
- Centre for Human Drug Research, Leiden, the Netherlands
- Leiden University Medical Center, Leiden, the Netherlands
| | - Femke P.M. Hoevenaars
- Department of Microbiology and Systems Biology, The Netherlands Organisation for Applied Scientific Research (TNO), Leiden, the Netherlands
| |
Collapse
|
131
|
Wishna-Kadawarage RN, Połtowicz K, Hickey RM, Siwek M. Modulation of gene expression in immune-related organs by in ovo stimulation with probiotics and prophybiotics in broiler chickens. J Appl Genet 2025; 66:195-205. [PMID: 38987456 PMCID: PMC11761985 DOI: 10.1007/s13353-024-00891-y] [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: 05/17/2024] [Revised: 06/20/2024] [Accepted: 06/28/2024] [Indexed: 07/12/2024]
Abstract
In ovo stimulation has been studied intensively as an alternative to antibiotic use in poultry production. We investigated the potential use of a probiotic in combination with a phytobiotic as a prophybiotic for in ovo stimulation and reported its beneficial effects on the gut microbiome of broiler chickens. The current study further investigates the gene expression in the immune-related organs of these chickens to understand the tissue-specific immunomodulatory effects of the treatments. The selected prophybiotic (Leuconostoc mesenteroides with garlic aqueous extract) and its probiotic component alone were injected into ROSS308 chicken eggs on the 12th day of incubation, and gene expression in cecal tonsils, spleen, and liver at 35 days of age was determined using qPCR method. The relative expression of each treatment was compared to the positive control, chickens injected with physiological saline in ovo. The results displayed a downregulation of pro- and anti-inflammatory cytokines in the cecal tonsils of the probiotic group and the liver of the prophybiotic group. The spleen displayed upregulated AVBD1 in both groups and upregulated IL1-β in the probiotic group. The probiotic group displayed increased expression of genes related to metabolism of energy (COX16), protein (mTOR), and lipids (CYP46A1) whereas the prophybiotic group displayed reduced expression of genes related to cholesterol synthesis (SREBP1) and glucose transportation (SLC2A2) in the liver. In conclusion, Leuconostoc mesenteroides differentially modulated gene expression in chickens when administered in ovo in combination with garlic aqueous extract. Further in ovo studies with different prophybiotic combinations are required to optimize the benefits in broiler chickens.
Collapse
Affiliation(s)
- Ramesha N Wishna-Kadawarage
- Department of Animal Biotechnology and Genetics, Faculty of Animal Breeding and Biology, Bydgoszcz University of Science and Technology, Mazowiecka 28, 85-084, Bydgoszcz, Poland.
| | - Katarzyna Połtowicz
- Department of Poultry Breeding, National Research Institute of Animal Production, Krakowska 1, 32-083, Balice, Poland
| | - Rita M Hickey
- Teagasc Food Research Centre, Moorepark, P61 C996, Fermoy, Co. Cork, Ireland
| | - Maria Siwek
- Department of Animal Biotechnology and Genetics, Faculty of Animal Breeding and Biology, Bydgoszcz University of Science and Technology, Mazowiecka 28, 85-084, Bydgoszcz, Poland
| |
Collapse
|
132
|
Doghish AS, Elazazy O, Mohamed HH, Mansour RM, Ghanem A, Faraag AHI, Elballal MS, Elrebehy MA, Elesawy AE, Abdel Mageed SS, Saber S, Nassar YA, Abulsoud AI, Abdel-Reheim MA, Elawady AS, Ali MA, Basiouny MS, Hemdan M, Lutfy RH, Awad FA, El-Sayed SA, Ashour MM, El-Sayyad GS, Mohammed OA. A Review on miRNAs in Enteric Bacteria-mediated Host Pathophysiology: Mechanisms and Implications. J Biochem Mol Toxicol 2025; 39:e70160. [PMID: 39907181 DOI: 10.1002/jbt.70160] [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: 09/15/2024] [Revised: 12/22/2024] [Accepted: 01/16/2025] [Indexed: 02/06/2025]
Abstract
Recently, many studies focused on the billions of native bacteria found inside and all over the human body, commonly known as the microbiota, and its interactions with the eukaryotic host. One of the niches for such microbiota is the gastrointestinal tract (GIT), which harbors hundreds to thousands of bacterial species commonly known as enteric bacteria. Changes in the enteric bacterial populations were linked to various pathologies such as irritable bowel syndrome and obesity. The gut microbiome could affect the health status of individuals. MicroRNAs (miRNAs) are one of the extensively studied small-sized noncoding RNAs (ncRNAs) over the past decade to explore their multiple roles in health and disease. It was proven that miRNAs circulate in almost all body fluids and tissues, showing signature patterns of dysregulation associated with pathologies. Both cellular and circulating miRNAs participate in the posttranscriptional regulation of genes and are considered the potential key regulators of genes and participate in cellular communication. This manuscript explores the unique interplay between miRNAs and enteric bacteria in the gastrointestinal tract, emphasizing their dual role in shaping host-microbiota dynamics. It delves into the molecular mechanisms by which miRNAs influence bacterial colonization and host immune responses, linking these findings to gut-related diseases. The review highlights innovative therapeutic and diagnostic opportunities, offering insights for targeted treatments of dysbiosis-associated pathologies.
Collapse
Affiliation(s)
- Ahmed S Doghish
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Egypt
- Biochemistry and Molecular Biology Department, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Egypt
| | - Ola Elazazy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Egypt
| | - Hend H Mohamed
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Egypt
- Biochemistry Department, Faculty of Science, Cairo University, Giza, Egypt
| | - Reda M Mansour
- Zoology and Entomology Department, Faculty of Science, Helwan University, Helwan, Egypt
- Molecular Biology and Biotechnology Department, School of Biotechnology, Badr University in Cairo (BUC), Badr City, Egypt
| | - Aml Ghanem
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Egypt
| | - Ahmed H I Faraag
- Botany and Microbiology Department, Faculty of Science, Helwan University, Helwan, Egypt
- Medical Department, School of Biotechnology, Badr University in Cairo, Badr City, Egypt
| | - Mohammed S Elballal
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Egypt
- BK21 FOUR Team and Integrated Research Institute for Drug Development, College of Pharmacy, Dongguk University-Seoul, Goyang, Republic of Korea
| | - Mahmoud A Elrebehy
- Department of Biochemistry, Faculty of Pharmacy, Galala University, New Galala City, Egypt
| | - Ahmed E Elesawy
- Department of Biochemistry, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Egypt
| | - Sherif S Abdel Mageed
- Pharmacology and Toxicology Department, Faculty of Pharmacy, Badr University in Cairo (BUC), Badr City, Egypt
| | - Sameh Saber
- Department of Pharmacology, Faculty of Pharmacy, Delta University for Science and Technology, Gamasa, Egypt
| | - Yara A Nassar
- Department of Botany, Faculty of Science, Biotechnology and Its Application Program, Mansoura University, Mansoura, Egypt
| | - Ahmed I Abulsoud
- Biochemistry Department, Faculty of Pharmacy, Heliopolis University, Cairo, Egypt
- Department of Biochemistry, Faculty of Pharmacy (Boys), Al-Azhar University, Nasr City, Egypt
| | | | - Alaa S Elawady
- Department of Biochemistry, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Mohamed A Ali
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Egypt
| | | | - Mohamed Hemdan
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Egypt
| | - Radwa H Lutfy
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Egypt
| | - Farah A Awad
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Egypt
| | - Salma A El-Sayed
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Egypt
| | - Mohamed M Ashour
- School of Biotechnology, Badr University in Cairo (BUC), Badr City, Egypt
| | - Gharieb S El-Sayyad
- Medical Laboratory Technology Department, Faculty of Applied Health Sciences Technology, Badr University in Cairo (BUC), Cairo, Egypt
- Microbiology and Immunology Department, Faculty of Pharmacy, Galala University, New Galala city, Egypt
| | - Osama A Mohammed
- Department of Pharmacology, College of Medicine, University of Bisha, Bisha, Saudi Arabia
| |
Collapse
|
133
|
Wang M, Monaco MH, Daniels VC, Hirvonen J, Jensen HM, Ouwehand AC, Mukherjea R, Christensen N, Lehtinen MJ, Hibberd AA, Dilger RN, Donovan SM. Individual and Combined Effects of 2'-Fucosyllactose and Bifidobacterium longum subsp. infantis on the Gut Microbiota Composition of Piglets. J Nutr 2025; 155:509-522. [PMID: 40354037 PMCID: PMC11867128 DOI: 10.1016/j.tjnut.2024.12.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2024] [Revised: 11/27/2024] [Accepted: 12/18/2024] [Indexed: 05/14/2025] Open
Abstract
BACKGROUND Human milk is a source of oligosaccharides that promote the growth of beneficial bacteria, including Bifidobacterium longum subsp. infantis, which can utilize human milk oligosaccharides. OBJECTIVES To evaluate the individual and combined effects of 2'-fucosyllactose (2'-FL), B. infantis Bi-26 (Bi-26) on piglet gut microbiota composition, and short-chain fatty acid (SCFA) concentrations. METHODS Intact male pigs (n = 63) had ad libitum access to milk replacer without (control; CON) or with 1.0 g/L 2'-FL (FL) from postnatal day 2 to 34/35. Pigs were further stratified to receive either 12% glycerol or 109 CFU/d Bi-26 in glycerol (BI and FLBI). Gut microbiota and SCFA concentrations were determined in ascending colon contents (AC) and rectal contents (RC) by 16S ribosomal ribonucleic acid gene sequencing and gas chromatography, respectively. Microbiota composition and functional profiles were analyzed using QIIME 2 and Phylogenetic Investigation of Communities by Reconstruction of Unobserved States-2 (PICRUSt2). RESULTS Supplementation of 2'-FL increased valerate concentration in AC (P = 0.03) and tended to modulate the overall bacterial composition in RC (P = 0.06). Compared with CON, 2'-FL alone increased the acetate concentration in AC (P < 0.05). The addition of Bi-26 decreased Shannon indices and reduced propionate and butyrate concentrations in AC (P < 0.05). Bi-26 alone affected the relative abundances of several bacterial amplicon sequence variants (ASVs) in AC and RC, including the ASVs identified as Phocaeicola (Bacteroides) vulgatus and Faecalibacterium prausnitzii. Additionally, 2'-FL and Bi-26 individually increased the relative abundances of 9 PICRUSt2-predicted metabolic pathways related to fatty acid and lipid biosynthesis or carboxylate degradation/secondary metabolite degradation in the RC; however, these effects were negated, and the values were identical to the CON group when 2'-FL and Bi-26 were supplemented together. CONCLUSIONS 2'-FL and Bi-26 added to milk replacer exerted distinct influences on gut bacterial composition and metabolic function, and 2'-FL alone increased specific SCFA concentrations, demonstrating its prebiotic potential.
Collapse
Affiliation(s)
- Mei Wang
- Department of Food Science & Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Marcia H Monaco
- Department of Food Science & Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Victoria C Daniels
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | | | - Henrik Max Jensen
- IFF R&D - Enabling Technologies, Advanced Analytical, Brabrand, Denmark
| | | | | | - Niels Christensen
- IFF R&D - Enabling Technologies, Advanced Analytical, Brabrand, Denmark
| | | | | | - Ryan N Dilger
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States; Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States
| | - Sharon M Donovan
- Department of Food Science & Human Nutrition, University of Illinois at Urbana-Champaign, Urbana, IL, United States; Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, IL, United States.
| |
Collapse
|
134
|
Croisier E, Brown T, Grigg A, Chan P, Bauer J. Dietary counselling to increase soluble fibre in patients with gynaecological cancers undergoing pelvic radiotherapy: A feasibility study. J Hum Nutr Diet 2025; 38:e13402. [PMID: 39587777 DOI: 10.1111/jhn.13402] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Revised: 10/30/2024] [Accepted: 11/07/2024] [Indexed: 11/27/2024]
Abstract
BACKGROUND This study aimed to determine the feasibility of increasing soluble fibre intake via dietary counselling to improve gastrointestinal toxicity and quality of life in patients with gynaecological cancers undergoing pelvic radiotherapy without adverse consequences on radiation treatment (RT) delivery accuracy. METHODS A single-arm, single-centre intervention feasibility trial included patients with gynaecological cancers undergoing pelvic RT ± chemotherapy at a tertiary hospital. Participants were provided weekly dietary counselling over the duration of their RT (5-6 weeks) to increase soluble fibre intake incrementally each week. Stakeholder surveys were also completed. RESULTS In total, 9 of 14 eligible patients participated (55 years old [SD 13.2], diagnosis: cervical [n = 3], endometrial/uterine [n = 5] and vaginal [n = 1]), with the majority categorised as low fibre consumers at baseline (n = 6). On average, soluble fibre intake increased by 150% throughout treatment. There were no adverse events or major adjustments required for RT delivery. There were improving trends in the functional subset identified. Results may be confounded by the sample size resulting from limited eligibility (n = 14) and a high attrition rate (n = 4). CONCLUSIONS Most participants successfully increased their soluble fibre intake throughout treatment, without significant adverse events noted for RT delivery accuracy. These results provide preliminary data to calculate the sample size required to produce meaningful effect sizes. However, this study highlighted challenges in participant recruitment and retention, with limited organisational support and perceived compatibility.
Collapse
Affiliation(s)
- Emilie Croisier
- School of Human Movement & Nutrition Sciences, University of Queensland, Brisbane, Queensland, Australia
- Dietetics & Food Services, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Teresa Brown
- Dietetics & Food Services, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Alice Grigg
- Radiation Therapy, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
| | - Philip Chan
- Radiation Oncology, Royal Brisbane and Women's Hospital, Brisbane, Queensland, Australia
- School of Medicine, University of Queensland, Brisbane, Queensland, Australia
| | - Judy Bauer
- Nutrition, Dietetics and Food, Monash University, Notting Hill, Victoria, Australia
| |
Collapse
|
135
|
Zhao C, Pan J, Wang Y, Zhao J, Huang J. Differential Analysis of Fecal SCFAs and Their Contribution to Adipogenesis in UCP1 Knock-In Pigs. Vet Sci 2025; 12:102. [PMID: 40005862 PMCID: PMC11860427 DOI: 10.3390/vetsci12020102] [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: 11/07/2024] [Revised: 01/21/2025] [Accepted: 01/30/2025] [Indexed: 02/27/2025] Open
Abstract
This study aimed to investigate the changes in fecal short-chain fatty acids (SCFAs) content in UCP1 knock-in pigs (KI pigs) and their effect on adipogenesis. Fecal samples from five 6-month-old wild-type (WT) and KI pigs were collected for targeted metabolomics and 16s rRNA sequencing analyses to identify differences in SCFAs and gut microbiota that may contribute to regulating fat deposition in pigs. The metabolome of pig fecal samples targeted for an analysis of SCFAs identified seven SCFAs, with caproic acid (except isovaleric acid) being the significantly different one. The results of the fecal 16s rRNA analysis demonstrated a notable reduction in the abundance of Streptococcus spp. in the KI pigs in comparison to the WT pigs, with a statistically significant difference. Correlation analyses demonstrated a statistically significant positive correlation between the abundance of Streptococcus spp. and SCFAs, as well as pig body weight and fatness. It was postulated that the reduction in SCFAs in the intestinal tracts of KI pigs may be associated with a reduction in Streptococcus spp. abundance. Compared to WT pigs, the concentration of fecal SCFAs in KI pigs was significantly reduced, which may be related to the decreased abundance of Streptococcus. The in vitro experiments showed that caproic acid could significantly enhance the differentiation efficiency of porcine SVF cells into mature adipocytes by activating the FFAR4 gene.
Collapse
Affiliation(s)
- Chengyu Zhao
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China;
- College of Life Science, Qingdao Agricultural University, Qingdao 266109, China
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (J.P.); (Y.W.)
| | - Jianfei Pan
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (J.P.); (Y.W.)
| | - Yanfang Wang
- State Key Laboratory of Animal Biotech Breeding, Institute of Animal Science, Chinese Academy of Agricultural Sciences, Beijing 100193, China; (J.P.); (Y.W.)
| | - Jianguo Zhao
- State Key Laboratory of Stem Cell and Reproductive Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing 100101, China;
| | - Jiaojiao Huang
- College of Animal Science and Technology, Qingdao Agricultural University, Qingdao 266109, China;
- College of Life Science, Qingdao Agricultural University, Qingdao 266109, China
| |
Collapse
|
136
|
Kanemaru E, Ichinose F. Essential role of sulfide oxidation in brain health and neurological disorders. Pharmacol Ther 2025; 266:108787. [PMID: 39719173 PMCID: PMC11806942 DOI: 10.1016/j.pharmthera.2024.108787] [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: 07/11/2024] [Revised: 11/21/2024] [Accepted: 12/19/2024] [Indexed: 12/26/2024]
Abstract
Hydrogen sulfide (H2S) is an environmental hazard well known for its neurotoxicity. In mammalian cells, H2S is predominantly generated by transsulfuration pathway enzymes. In addition, H2S produced by gut microbiome significantly contributes to the total sulfide burden in the body. Although low levels of H2S is believed to exert various physiological functions such as neurotransmission and vasomotor control, elevated levels of H2S inhibit the activity of cytochrome c oxidase (i.e., mitochondrial complex IV), thereby impairing oxidative phosphorylation. To protect the electron transport chain from respiratory poisoning by H2S, the compound is actively oxidized to form persulfides and polysulfides by a mitochondrial resident sulfide oxidation pathway. The reaction, catalyzed by sulfide:quinone oxidoreductase (SQOR), is the initial and critical step in sulfide oxidation. The persulfide species are subsequently oxidized to sulfite, thiosulfate, and sulfate by persulfide dioxygenase (ETHE1 or SDO), thiosulfate sulfurtransferase (TST), and sulfite oxidase (SUOX). While SQOR is abundantly expressed in the colon, liver, lung, and skeletal muscle, its expression is notably low in the brains of most mammals. Consequently, the brain's limited capacity to oxidize H2S renders it particularly sensitive to the deleterious effects of H2S accumulation. Impaired sulfide oxidation can lead to fatal encephalopathy, and the overproduction of H2S has been implicated in the developmental delays observed in Down syndrome. Our recent findings indicate that the brain's limited capacity to oxidize sulfide exacerbates its sensitivity to oxygen deprivation. The beneficial effects of sulfide oxidation are likely to be mediated not only by the detoxification of H2S but also by the formation of persulfide, which exerts cytoprotective effects through multiple mechanisms. Therefore, pharmacological agents designed to scavenge H2S and/or enhance persulfide levels may offer therapeutic potential against neurological disorders characterized by impaired or insufficient sulfide oxidation or excessive H2S production.
Collapse
Affiliation(s)
- Eiki Kanemaru
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| | - Fumito Ichinose
- Anesthesia Center for Critical Care Research, Department of Anesthesia, Critical Care and Pain Medicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA.
| |
Collapse
|
137
|
Jankowski WM, Fichna J, Tarasiuk-Zawadzka A. A systematic review of the relationship between gut microbiota and prevalence of pancreatic diseases. Microb Pathog 2025; 199:107214. [PMID: 39653281 DOI: 10.1016/j.micpath.2024.107214] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2024] [Revised: 11/24/2024] [Accepted: 12/06/2024] [Indexed: 12/12/2024]
Abstract
Acute pancreatitis (AP) represents one of the most common gastrointestinal (GI) diseases; it can manifest in varying degrees of severity, sometimes leading to a life-threatening condition for the patient. Pancreatic ductal adenocarcinoma (PDAC), due to its high malignancy and uncertain prognosis, is widely regarded as one of the most fatal diseases. The increasing prevalence of AP and PDAC represents a major burden on public health and the healthcare system worldwide. The aim of this systematic review was to discuss the current state of knowledge regarding the relationship between the gut microbiota and the incidence, prognosis, diagnosis and treatment of AP and PDAC. To identify studies that analyzed the relationship between the gut microbiota and the occurrence/development of pancreatic diseases or PDAC, the online databases PubMed, Scopus and Google Scholar were searched between November 2023 and January 2024. Finally, 14 publications met the inclusion criteria (1. were conducted exclusively in humans and/or animals; 2. original, published in English in peer-reviewed journals after 2019; 3. described the relationship between gut microbiota and the occurrence of AP or PDAC). The collected studies indicated significant changes in the gut microbiota of patients with AP and PDAC. Moreover, they highlighted the presence of a relationship between the gut microbiota and the occurrence, course, treatment efficiency and prognosis of the disease in question. Further research is needed to understand precisely the relationship between the gut microbiota and the occurrence of pancreatic diseases and whether it may be a starting point for the development of modern forms of therapy based on the use of prebiotics and/or diet to restore the normal composition of the intestinal bacteria.
Collapse
Affiliation(s)
- Wojciech Michał Jankowski
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215, Lodz, Poland
| | - Jakub Fichna
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215, Lodz, Poland
| | - Aleksandra Tarasiuk-Zawadzka
- Department of Biochemistry, Faculty of Medicine, Medical University of Lodz, Mazowiecka 5, 92-215, Lodz, Poland.
| |
Collapse
|
138
|
Kumar P, Kumar A, Kumar V. Role of Microbiota-Derived Metabolites in Prostate Cancer Inflammation and Progression. Cell Biochem Funct 2025; 43:e70050. [PMID: 39891389 DOI: 10.1002/cbf.70050] [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: 07/11/2024] [Revised: 12/25/2024] [Accepted: 01/20/2025] [Indexed: 02/03/2025]
Abstract
Prostate cancer (PCa) is the most commonly detected malignancy in men worldwide. PCa is a slow-growing cancer with the absence of symptoms at early stages. The pathogenesis has not been entirely understood including the key risk factors related to PCa development like diet and microbiota derived metabolites. Microbiota may influence the host's immunological responses, inflammatory responses, and metabolic pathways, which may be crucial for the development and metastasis. Similarly, short-chain fatty acids, methylamines, hippurate, bile acids, and other metabolites generated by microbiota may have potential roles in cancer inflammation and progression of cancer. Most studies have focused on the role of metabolites and their pathways involved in chronic inflammation, tumor initiation, proliferation, and progression. In summary, the review discusses the role of microbiota and microbial-derived metabolite-built strategies in inflammation and progression of the PCa.
Collapse
Affiliation(s)
- Pradeep Kumar
- Department of NMR, All India Institute of Medical Sciences, New Delhi, India
| | - Anil Kumar
- Gene Regulation Laboratory, National Institute of Immunology, New Delhi, India
| | - Virendra Kumar
- Department of NMR, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
139
|
Salahlou M, Hajimohammadi A, Nazifi S, Rowshan-Ghasrodashti A, Nikzad M, Mirzaei A. Effects of probiotic and yeast extract supplement on liver functionality index and metabolic parameters in transition period of dairy cattle. Vet J 2025; 309:106280. [PMID: 39617095 DOI: 10.1016/j.tvjl.2024.106280] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 07/09/2024] [Accepted: 11/26/2024] [Indexed: 12/06/2024]
Abstract
This research sought to evaluate the potential effects of probiotics and yeast cell wall (YCW) supplements on the liver functionality index (LFI) and metabolic parameters of dairy cattle throughout the transitional period. A cohort of forty dry cows was randomly divided into four groups, namely the probiotic group (Pr) receiving a basal diet combined with a blend of Bacillus subtilis, Bacillus lechiniformis, Streptococcus Thermophilis, and Enterococcus faecium; the YCW group receiving a basal diet enriched with Saccharomyces cerevisiae; the probiotic and yeast cell wall extract group (P & Y) receiving a basal diet supplemented with a mixture of probiotic and yeast cell wall extract; and the control group adhering to the basal diet. The intervention was initiated 21 days before calving and persisted until 28 days post-calving, except for the control group. The study entailed the collection of blood samples at four sampling times, encompassing 21 days preceding calving, seven days before calving, seven days post-calving, and four weeks post-calving. Multiple biochemical parameters were assessed, including urea, blood urea nitrogen (BUN), Gamma-glutamyl transferase (GGT), total bilirubin (TB), albumin, total protein (TP), globulin, glucose, triglyceride, cholesterol, and liver functionality index. The results showed that the Pr group exhibited reduced average levels of GGT and glucose compared to the control group (P < 0.05). Similarly, the P & Y group demonstrated lower average BUN, TB, and cholesterol levels than the control (P < 0.05). Notably, the LFI exhibited a discernible trend towards elevation in the Pr group compared to the control group (P = 0.007) and the P & Y group (P = 0.007). In essence, supplementation of YCW and probiotics is associated with advantageous effects on metabolic parameters and liver function.
Collapse
Affiliation(s)
- Mohammad Salahlou
- Resident of Large Animal Internal Medicine, Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Ali Hajimohammadi
- Associate Professor of Large Animal Internal Medicine, Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran.
| | - Saeed Nazifi
- Professor of Clinical Pathology, Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Abbas Rowshan-Ghasrodashti
- Assistant Professor of Large Animal Internal Medicine, Department of Clinical Studies, School of Veterinary Medicine, Islamic Azad University, Kazerun Branch, Kazerun, Iran
| | - Mohammad Nikzad
- Resident of Large Animal Internal Medicine, Department of Clinical Sciences, School of Veterinary Medicine, Shiraz University, Shiraz, Iran
| | - Ahmadreza Mirzaei
- Post-doctoral associate at College of Veterinary Medicine, University of Florida, FL 32610, USA
| |
Collapse
|
140
|
Hassib L, Kanashiro A, Pedrazzi JFC, Vercesi BF, Higa S, Arruda Í, Soares Y, de Jesus de Souza A, Jordão AA, Guimarães FS, Ferreira FR. Should we consider microbiota-based interventions as a novel therapeutic strategy for schizophrenia? A systematic review and meta-analysis. Brain Behav Immun Health 2025; 43:100923. [PMID: 39839986 PMCID: PMC11745983 DOI: 10.1016/j.bbih.2024.100923] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2024] [Revised: 12/04/2024] [Accepted: 12/10/2024] [Indexed: 01/06/2025] Open
Abstract
Schizophrenia is a chronic psychiatric disorder characterized by a variety of symptoms broadly categorized into positive, negative, and cognitive domains. Its etiology is multifactorial, involving a complex interplay of genetic, neurobiological, and environmental factors, and its neurobiology is associated with abnormalities in different neurotransmitter systems. Due to this multifactorial etiology and neurobiology, leading to a wide heterogeneity of symptoms and clinical presentations, current antipsychotic treatments face challenges, underscoring the need for novel therapeutic approaches. Recent studies have revealed differences in the gut microbiome of individuals with schizophrenia compared to healthy controls, establishing an intricate link between this disorder and gastrointestinal health, and suggesting that microbiota-targeted interventions could help alleviate clinical symptoms. Therefore, this meta-analysis investigates whether gut microbiota manipulation can ameliorate psychotic outcomes in patients with schizophrenia receiving pharmacological treatment. Nine studies (n = 417 participants) were selected from 81 records, comprising seven randomized controlled trials and two open-label studies, all with a low risk of bias, included in this systematic review and meta-analysis. The overall combined effect size indicated significant symptom improvement following microbiota treatment (Hedges' g = 0.48, 95% CI = 0.09 to 0.88, p = 0.004, I2 = 62.35%). However, according to Hedges' g criteria, the effect size was small (approaching moderate), and study heterogeneity was moderate based on I2 criteria. This review also discusses clinical and preclinical studies to elucidate the neural, immune, and metabolic pathways by which microbiota manipulation, particularly with Lactobacillus and Bifidobacterium genera, may exert beneficial effects on schizophrenia symptoms via the gut-brain axis. Finally, we address the main confounding factors identified in our systematic review, highlight key limitations, and offer recommendations to guide future high-quality trials with larger participant cohorts to explore microbiome-based therapies as a primary or adjunctive treatment for schizophrenia.
Collapse
Affiliation(s)
- Lucas Hassib
- Department of Pharmacology, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, SP, Brazil
- Oswaldo Cruz Foundation, Institute Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Alexandre Kanashiro
- Faillace Department of Psychiatry and Behavioral Sciences, Translational Psychiatry Program, McGovern Medical School, The University of Texas Health Science Center at Houston (UTHealth), Houston, TX, USA
| | | | - Bárbara Ferreira Vercesi
- Department of Health Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Sayuri Higa
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of Sao Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Íris Arruda
- Oswaldo Cruz Foundation, Institute Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Yago Soares
- Oswaldo Cruz Foundation, Institute Oswaldo Cruz, Rio de Janeiro, RJ, Brazil
| | - Adriana de Jesus de Souza
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of Sao Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | - Alceu Afonso Jordão
- Department of Health Sciences, Ribeirão Preto Medical School, University of São Paulo, Ribeirão Preto, Brazil
| | - Francisco Silveira Guimarães
- Department of Pharmacology, School of Medicine of Ribeirão Preto, University of Sao Paulo, Ribeirão Preto, SP, 14049-900, Brazil
| | | |
Collapse
|
141
|
Baskaran K, Moshkovich M, Hart L, Shah N, Chowdhury F, Shanmuganathan M, Britz-McKibbin P, Pai N. The role of urine metabolomics in the diagnosis and management of adult and pediatric Crohn's disease and ulcerative colitis. Biomarkers 2025; 30:104-113. [PMID: 39642943 DOI: 10.1080/1354750x.2024.2438734] [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: 07/03/2024] [Accepted: 12/02/2024] [Indexed: 12/09/2024]
Abstract
INTRODUCTION Urine metabolomics offers a non-invasive approach to diagnose and manage inflammatory bowel disease (IBD), including Crohn's disease (CD) and ulcerative colitis (UC), by identifying distinct metabolic signatures. OBJECTIVES This narrative review summarizes current findings on urinary metabolites in IBD, evaluating their roles in disease differentiation, assessment of activity, and monitoring therapeutic response. METHODS A comprehensive literature search of PubMed and MEDLINE up to October 2023 was conducted using keywords, such as 'urine metabolomics', 'inflammatory bowel disease', 'Crohn's disease', 'ulcerative colitis', and 'urinary biomarkers'. Studies were included that described alterations to metabolic pathways, including those related to the urea cycle, central energy metabolism (Krebs cycle), amino acid metabolism, and neurotransmitters. RESULTS Specific urinary metabolites differentiate IBD patients from healthy controls and between CD and UC. Decreased urinary levels of hippurate, acetate, methanol, formate, and methylamine are observed in IBD, indicating altered gut microbiota. In CD patients, urea cycle alterations include reduced urinary urea and ornithine with increased arginine. Changes in Krebs cycle intermediates show decreased citrate and succinate in adults, but increased fumarate and isocitrate in pediatric patients, reflecting energy metabolism differences. Amino acid metabolism differs by age: Adults exhibit decreased urinary asparagine, lysine, and histidine, while pediatric patients show increased methionine, proline, aspartic acid, and isoleucine. Elevated urinary neurotransmitters like dopamine are noted in pediatric IBD patients. Urine metabolomics also can monitor treatment efficacy by distinguishing responders from non-responders to therapies and differentiating active disease from remission. CONCLUSION Urine metabolomics provides promising, non-invasive biomarkers to enhance IBD diagnostics by distinguishing CD from UC and offering insights into underlying metabolic disturbances, paving the way for more precise, accessible patient care.
Collapse
Affiliation(s)
- Kanish Baskaran
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Michal Moshkovich
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
- Faculty of Medicine, University of Toronto, Toronto, Ontario, Canada
| | - Lara Hart
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, McMaster Children's Hospital, Hamilton, Ontario, Canada
| | - Nyah Shah
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Fariha Chowdhury
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
- Department of Health Research Methods, Evidence and Impact, McMaster University, Hamilton, Ontario, Canada
| | - Meera Shanmuganathan
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario, Canada
| | - Philip Britz-McKibbin
- Department of Chemistry and Chemical Biology, McMaster University, Hamilton, Ontario, Canada
| | - Nikhil Pai
- Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
- Division of Pediatric Gastroenterology, Hepatology and Nutrition, McMaster Children's Hospital, Hamilton, Ontario, Canada
- Division of Gastroenterology, Hepatology and Nutrition, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
142
|
Okoro FO, Markus V. Artificial sweeteners and Type 2 Diabetes Mellitus: A review of current developments and future research directions. J Diabetes Complications 2025; 39:108954. [PMID: 39854925 DOI: 10.1016/j.jdiacomp.2025.108954] [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: 09/05/2024] [Revised: 11/12/2024] [Accepted: 01/20/2025] [Indexed: 01/27/2025]
Abstract
While artificial sweeteners are Generally Regarded as Safe (GRAS), the scientific community remains divided on their safety status. The previous assumption that artificial sweeteners are inert within the body is no longer valid. Artificial sweeteners, known for their high intense sweetness and low or zero calories, are extensively used today in food and beverage products as sugar substitutes and are sometimes recommended for weight management and Type 2 Diabetes Mellitus (T2DM) patients. The general omission of information about the concentration of artificial sweeteners on market product labels makes it challenging to determine the amounts of artificial sweeteners consumed by people. Despite regulatory authorization for their usage, such as from the United States Food and Drug Administration (FDA), concerns remain about their potential association with metabolic diseases, such as T2DM, which the artificial sweeteners were supposed to reduce. This review discusses the relationship between artificial sweetener consumption and the risk of developing T2DM. With the increasing number of recent scientific studies adding to the debate on this subject matter, we assessed recent literature and up-to-date evidence. Importantly, we highlight future research directions toward furthering knowledge in this field of study.
Collapse
Affiliation(s)
- Francisca Obianuju Okoro
- Department of Medical Biochemistry, Faculty of Medicine, Near East University, Nicosia, 99138 TRNC, Mersin 10, Turkey
| | - Victor Markus
- Department of Medical Biochemistry, Faculty of Medicine, Near East University, Nicosia, 99138 TRNC, Mersin 10, Turkey.
| |
Collapse
|
143
|
Kaur S, Patel BCK, Collen A, Malhotra R. The microbiome and the eye: a new era in ophthalmology. Eye (Lond) 2025; 39:436-448. [PMID: 39702789 PMCID: PMC11794629 DOI: 10.1038/s41433-024-03517-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 11/10/2024] [Accepted: 11/26/2024] [Indexed: 12/21/2024] Open
Abstract
The human microbiome has progressively been recognised for its role in various disease processes. In ophthalmology, complex interactions between the gut and distinct ocular microbiota within each structure and microenvironment of the eye has advanced our knowledge on the multi-directional relationships of these ecosystems. Increasingly, studies have shown that modulation of the microbiome can be achieved through faecal microbiota transplantation and synbiotics producing favourable outcomes for ophthalmic diseases. As ophthalmologists, we are obliged to educate our patients on measures to cultivate a healthy gut microbiome through a range of holistic measures. Further integrative studies combining microbial metagenomics, metatranscriptomics and metabolomics are necessary to fully characterise the human microbiome and enable targeted therapeutic interventions.
Collapse
Affiliation(s)
- Simerdip Kaur
- Department of Ophthalmology, University Hospitals Sussex NHS Foundation Trust, Sussex Eye Hospital, Eastern Road, Brighton, BN2 5BF, UK.
- Corneoplastic Unit, Queen Victoria Hospital, East Grinstead, RH19 3DZ, UK.
| | - Bhupendra C K Patel
- Department of Ophthalmology and Visual Sciences, John A. Moran Eye Center, University of Utah, Salt Lake City, Utah, 84132, USA
| | - Alanna Collen
- Unaffiliated officially. Independent author, London, UK
| | - Raman Malhotra
- Corneoplastic Unit, Queen Victoria Hospital, East Grinstead, RH19 3DZ, UK
| |
Collapse
|
144
|
Ayyanar MP, Vijayan M. A review on gut microbiota and miRNA crosstalk: implications for Alzheimer's disease. GeroScience 2025; 47:339-385. [PMID: 39562408 PMCID: PMC11872870 DOI: 10.1007/s11357-024-01432-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Accepted: 11/07/2024] [Indexed: 11/21/2024] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by cognitive decline and progressive neuronal damage. Recent research has highlighted the significant roles of the gut microbiota and microRNAs (miRNAs) in the pathogenesis of AD. This review explores the intricate interaction between gut microbiota and miRNAs, emphasizing their combined impact on Alzheimer's progression. First, we discuss the bidirectional communication within the gut-brain axis and how gut dysbiosis contributes to neuroinflammation and neurodegeneration in AD. Changes in gut microbiota composition in Alzheimer's patients have been linked to inflammation, which exacerbates disease progression. Next, we delve into the biology of miRNAs, focusing on their roles in gene regulation, neurodevelopment, and neurodegeneration. Dysregulated miRNAs are implicated in AD pathogenesis, influencing key processes like inflammation, tau pathology, and amyloid deposition. We then examine how the gut microbiota modulates miRNA expression, particularly in the brain, potentially altering neuroinflammatory responses and synaptic plasticity. The interplay between gut microbiota and miRNAs also affects blood-brain barrier integrity, further contributing to Alzheimer's pathology. Lastly, we explore therapeutic strategies targeting this gut microbiota-miRNA axis, including probiotics, prebiotics, and dietary interventions, aiming to modulate miRNA expression and improve AD outcomes. While promising, challenges remain in fully elucidating these interactions and translating them into effective therapies. This review highlights the importance of understanding the gut microbiota-miRNA relationship in AD, offering potential pathways for novel therapeutic approaches aimed at mitigating the disease's progression.
Collapse
Affiliation(s)
- Maruthu Pandian Ayyanar
- Department of Biology, The Gandhigram Rural Institute (Deemed to be University), Gandhigram, 624302, Tamil Nadu, India
| | - Murali Vijayan
- Department of Internal Medicine, Texas Tech University Health Sciences Center, Lubbock, TX, 79430, USA.
| |
Collapse
|
145
|
Dawson SL, Todd E, Ward AC. The Interplay of Nutrition, the Gut Microbiota and Immunity and Its Contribution to Human Disease. Biomedicines 2025; 13:329. [PMID: 40002741 PMCID: PMC11853302 DOI: 10.3390/biomedicines13020329] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/27/2025] Open
Abstract
Nutrition, the gut microbiota and immunity are all important factors in the maintenance of health. However, there is a growing realization of the complex interplay between these elements coalescing in a nutrition-gut microbiota-immunity axis. This regulatory axis is critical for health with disruption being implicated in a broad range of diseases, including autoimmune disorders, allergies and mental health disorders. This new perspective continues to underpin a growing number of innovative therapeutic strategies targeting different elements of this axis to treat relevant diseases. This review describes the inter-relationships between nutrition, the gut microbiota and immunity. It then details several human diseases where disruption of the nutrition-gut microbiota-immunity axis has been identified and presents examples of how the various elements may be targeted therapeutically as alternate treatment strategies for these diseases.
Collapse
Affiliation(s)
- Samantha L. Dawson
- School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia; (S.L.D.); (E.T.)
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Waurn Ponds, VIC 3216, Australia
| | - Emma Todd
- School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia; (S.L.D.); (E.T.)
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Waurn Ponds, VIC 3216, Australia
| | - Alister C. Ward
- School of Medicine, Deakin University, Waurn Ponds, VIC 3216, Australia; (S.L.D.); (E.T.)
- Institute for Mental and Physical Health and Clinical Translation (IMPACT), Deakin University, Waurn Ponds, VIC 3216, Australia
| |
Collapse
|
146
|
Wang X, Zhang B, Jiang R. Microbiome interplays in the gut-liver axis: implications for liver cancer pathogenesis and therapeutic insights. Front Cell Infect Microbiol 2025; 15:1467197. [PMID: 39936163 PMCID: PMC11810975 DOI: 10.3389/fcimb.2025.1467197] [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: 07/19/2024] [Accepted: 01/07/2025] [Indexed: 02/13/2025] Open
Abstract
Globally, primary liver cancer (PLC) ranks the most fatal malignancy. Most of the patients are in advanced stage of PLC at the very time they are diagnosed with it, accounting much for its poor prognosis. With the advancement of modern medical research and care system, the main etiology of PLC more and more switches from hepatitis viruses such as HAV, HBV, HCV, HEV to other causes like metabolism-associated steatohepatitis (MASH) and metabolic-associated fatty liver disease (MAFLD). As a result, it is of great necessity to find out new ways for treatment and early diagnosis to cope with this problem. Nowadays, as the mechanism of the Gut-Liver Axis in the formation of MAFLD, MASH and PLC has been gradually elucidated. The association between gut microbiome and the formation of PLC is of great significance to take an insight into. In this review, we present the concept of Gut-Liver Axis and its function in the mutual influence between gut microbiota and PLC from several aspects in which we will focus on the structure of gut barrier and the functional influences the gut microbiota have on the immune response and metabolic changes on human liver. Furthermore, we conclude the potential association of gut microbiota constitution with the PLC. Eventually, we hope this review can offer novel instructions for early diagnosis and treatment for liver cancer.
Collapse
Affiliation(s)
- Xuran Wang
- Medical School of Nanjing University, Nanjing, Jiangsu, China
| | - Bin Zhang
- Department of Gastroenterology, Affiliated Nanjing Drum Tower Hospital, Medical School of Nanjing University, Nanjing, China
| | - Runqiu Jiang
- Department of Clinical Laboratory, The First Affiliated Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
147
|
Demirci M, Afolabi JM, Kirabo A. Aging and sex differences in salt sensitivity of blood pressure. Clin Sci (Lond) 2025; 139:CS20240788. [PMID: 39873323 DOI: 10.1042/cs20240788] [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: 08/22/2024] [Revised: 01/04/2025] [Accepted: 01/09/2025] [Indexed: 01/30/2025]
Abstract
Salt sensitivity of blood pressure (SSBP) is a complex physiological trait characterized by changes in blood pressure in response to dietary salt intake. Aging introduces an additional layer of complexity to the pathophysiology of SSBP, with mitochondrial dysfunction, epigenetic modifications, and alterations in gut microbiota emerging as critical factors. Despite advancements in understanding these mechanisms, the processes driving increased salt sensitivity with age and their differential impacts across sexes remain unclear. This review explores the current understanding of salt sensitivity, delving into its underlying mechanisms, the role of inflammation, and the influence of aging and sex differences on these processes. We also aim to provide insights into the multifaceted nature of salt sensitivity and its implications for personalized treatment strategies in hypertension management.
Collapse
Affiliation(s)
- Mert Demirci
- Department of Medicine, Division of Nephrology and Hypertension, Vanderbilt University Medical Center, Nashville, TN, U.S.A
| | - Jeremiah M Afolabi
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, U.S.A
| | - Annet Kirabo
- Department of Medicine, Division of Clinical Pharmacology, Vanderbilt University Medical Center, Nashville, TN, U.S.A
- Department of Molecular Physiology & Biophysics, Vanderbilt University, Nashville, TN, U.S.A
- Vanderbilt Center for Immunobiology, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Infection, Immunology and Inflammation, Vanderbilt University Medical Center, Nashville, TN, USA
- Vanderbilt Institute for Global Health, Vanderbilt University Medical Center, Nashville, TN, USA
| |
Collapse
|
148
|
Liang M, Dong Q, Wu W, Fan J. Short-Chain Fatty Acids: Promising Therapeutic Targets for Respiratory Syncytial Virus Infection. Clin Rev Allergy Immunol 2025; 68:8. [PMID: 39873814 DOI: 10.1007/s12016-024-09018-x] [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] [Accepted: 11/02/2024] [Indexed: 01/30/2025]
Abstract
The intestinal microbiota is a complex community of organisms present in the human gastrointestinal tract, some of which can produce short-chain fatty acids (SCFAs) through the fermentation of dietary fiber. SCFAs play a major role in mediating the intestinal microbiota's regulation of host immunity and intestinal homeostasis. Respiratory syncytial virus (RSV) can cause an imbalance between anti-inflammatory and proinflammatory responses in the host. In addition, changes in SCFA levels and the structure of the intestinal microbiota have been observed after RSV infection. Therefore, there may be a link between SCFAs and RSV infection, and SCFAs are expected to be therapeutic targets for RSV infection.
Collapse
Affiliation(s)
- Mingxin Liang
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, China
- Department of Pediatrics, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, Sichuan, China
| | - Qinqin Dong
- School of Medical and Life Sciences, Chengdu University of Traditional Chinese Medicine, Chengdu, 610075, Sichuan, China
- Department of Pediatrics, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, Sichuan, China
| | - Weiyi Wu
- School of Medicine, University of Electronic Science and Technology of China, Chengdu, 610054, Sichuan, China
- Department of Pediatrics, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, Sichuan, China
| | - Juan Fan
- Department of Pediatrics, Sichuan Academy of Medical Sciences and Sichuan Provincial People's Hospital, Chengdu, 610072, Sichuan, China.
| |
Collapse
|
149
|
Kim CH. Functional regulation of cytotoxic T cells by gut microbial metabolites. GUT MICROBES REPORTS 2025; 2:1-16. [PMID: 40115123 PMCID: PMC11922538 DOI: 10.1080/29933935.2025.2454002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/30/2024] [Revised: 12/21/2024] [Accepted: 01/09/2025] [Indexed: 03/23/2025]
Abstract
Metabolites from gut microbes have a wide range of functions within the host body. One important function of these metabolites is to either positively or negatively control CD8+ cytotoxic T lymphocytes (CTLs), which can kill cancer and virus-infected cells. In healthy conditions, gut microbes produce a mixture of metabolites that promote CTL activity but also suppress excessive inflammatory responses. However, gut microbial dysbiosis occurs in patients with cancer, and this leads to changes in the production of gut microbial metabolites that can suppress CTL activity, promote inflammatory responses, and/or aid cancer growth. Decreased levels of CTL-promoting metabolites such as short-chain fatty acids, indole metabolites and polyamines but increased levels of CTL-suppressing metabolites, such as certain bile acids along with oncogenic metabolites, have been observed in patients with cancer. This review summarizes the altered production of major microbial metabolites in patients with cancer and discusses the impact of these changes on anti-cancer CTL responses.
Collapse
Affiliation(s)
- Chang H Kim
- Department of Pathology, University of Michigan School of Medicine, Ann Arbor, MI 48109; Mary H. Weiser Food Allergy Center, Center for Gastrointestinal Research, and Rogel Center for Cancer Research, University of Michigan School of Medicine, Ann Arbor, MI 48109
| |
Collapse
|
150
|
Wankhede NL, Kale MB, Kyada A, M RM, Chaudhary K, Naidu KS, Rahangdale S, Shende PV, Taksande BG, Khalid M, Gulati M, Umekar MJ, Fareed M, Kopalli SR, Koppula S. Sleep deprivation-induced shifts in gut microbiota: Implications for neurological disorders. Neuroscience 2025; 565:99-116. [PMID: 39622383 DOI: 10.1016/j.neuroscience.2024.11.070] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2024] [Revised: 11/26/2024] [Accepted: 11/27/2024] [Indexed: 12/07/2024]
Abstract
Sleep deprivation is a prevalent issue in contemporary society, with significant ramifications for both physical and mental well-being. Emerging scientific evidence illuminates its intricate interplay with the gut-brain axis, a vital determinant of neurological function. Disruptions in sleep patterns disturb the delicate equilibrium of the gut microbiota, resulting in dysbiosis characterized by alterations in microbial composition and function. This dysbiosis contributes to the exacerbation of neurological disorders such as depression, anxiety, and cognitive decline through multifaceted mechanisms, including heightened neuroinflammation, disturbances in neurotransmitter signalling, and compromised integrity of the gut barrier. In response to these challenges, there is a burgeoning interest in therapeutic interventions aimed at restoring gut microbial balance and alleviating neurological symptoms precipitated by sleep deprivation. Probiotics, dietary modifications, and behavioural strategies represent promising avenues for modulating the gut microbiota and mitigating the adverse effects of sleep disturbances on neurological health. Moreover, the advent of personalized interventions guided by advanced omics technologies holds considerable potential for tailoring treatments to individualized needs and optimizing therapeutic outcomes. Interdisciplinary collaboration and concerted research efforts are imperative for elucidating the underlying mechanisms linking sleep, gut microbiota, and neurological function. Longitudinal studies, translational research endeavours, and advancements in technology are pivotal for unravelling the complex interplay between these intricate systems.
Collapse
Affiliation(s)
- Nitu L Wankhede
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Mayur B Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Ashishkumar Kyada
- Marwadi University Research Center, Department of Pharmacy, Faculty of Health Sciences Marwadi University, Rajkot 360003, Gujarat, India
| | - Rekha M M
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Kamlesh Chaudhary
- Department of Neurology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - K Satyam Naidu
- Department of Chemistry, Raghu Engineering College, Visakhapatnam, Andhra Pradesh, India
| | - Sandip Rahangdale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Prajwali V Shende
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Brijesh G Taksande
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Mohammad Khalid
- Department of Pharmacognosy, College of Pharmacy Prince Sattam Bin Abdulaziz University Alkharj, Saudi Arabia
| | - Monica Gulati
- School of Pharmaceutical Sciences, Lovely Professional University, Phagwara, Punjab 1444411, India; ARCCIM, Faculty of Health, University of Technology Sydney, Ultimo, NSW 20227, Australia
| | - Milind J Umekar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra 441002, India
| | - Mohammad Fareed
- Department of Basic Medical Sciences, College of Medicine, AlMaarefa University, P.O. Box 71666, Riyadh 11597, Saudi Arabia
| | - Spandana Rajendra Kopalli
- Department of Bioscience and Biotechnology, Sejong University, Gwangjin-gu, Seoul 05006, Republic of Korea
| | - Sushruta Koppula
- College of Biomedical and Health Sciences, Konkuk University, Chungju-Si, Chungcheongbuk Do 27478, Republic of Korea.
| |
Collapse
|