1
|
Wang Y, Dou W, Qian X, Chen H, Zhang Y, Yang L, Wu Y, Xu X. Advancements in the study of short-chain fatty acids and their therapeutic effects on atherosclerosis. Life Sci 2025; 369:123528. [PMID: 40049368 DOI: 10.1016/j.lfs.2025.123528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2024] [Revised: 02/15/2025] [Accepted: 03/02/2025] [Indexed: 03/09/2025]
Abstract
Atherosclerosis (AS) remains a leading cause of cardiovascular disease and mortality globally. This chronic condition is characterized by inflammation, lipid accumulation, and the deposition of cellular components within arterial walls. Emerging evidence has highlighted the multifaceted therapeutic potential of short-chain fatty acids (SCFAs) in mitigating AS progression. SCFAs have demonstrated anti-inflammatory properties and the ability to regulate immune responses, metabolic pathways, vascular integrity, and intestinal barrier function in animal models of AS. Consequently, SCFAs have garnered significant attention as a promising approach for the prevention and treatment of AS. However, further clinical trials and studies are necessary to fully elucidate the underlying mechanisms and effects of SCFAs. Additionally, different types of SCFAs may exert distinct impacts, necessitating more in-depth investigation into their specific roles and mechanisms. This review provides an overview of the diverse cellular mechanisms contributing to AS formation, as well as a discussion of the significance of SCFAs in AS pathogenesis and their multifaceted therapeutic potential. Nonetheless, additional research is warranted to comprehensively understand and harness the potential of various SCFAs in the context of AS.
Collapse
Affiliation(s)
- Yongsen Wang
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China; Department of Hepatobiliary Pancreatic and Splcnic Surgery, Luzhou People's Hospital, Luzhou, Sichuan 646000, PR China; Department of Vascular and Breast Surgery, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan 621000, PR China
| | - Wei Dou
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China
| | - Xin Qian
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China
| | - Hao Chen
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China
| | - Yi Zhang
- Department of Vascular and Breast Surgery, The Third Hospital of Mianyang, Sichuan Mental Health Center, Mianyang, Sichuan 621000, PR China
| | - Liu Yang
- Department of Hepatobiliary Pancreatic and Splcnic Surgery, Luzhou People's Hospital, Luzhou, Sichuan 646000, PR China
| | - Ya Wu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China
| | - Xiongfei Xu
- Department of Vascular Surgery, The Affiliated Hospital, Southwest Medical University, Taiping Street 25, Luzhou, Sichuan 646000, PR China.
| |
Collapse
|
2
|
Saadh MJ, Ahmed HH, Kareem RA, Sanghvi G, Ganesan S, Agarwal M, Kaur P, Taher WM, Alwan M, Jawad MJ, Hamad AK. Short-chain fatty acids in Huntington's disease: Mechanisms of action and their therapeutic implications. Pharmacol Biochem Behav 2025; 249:173972. [PMID: 39983928 DOI: 10.1016/j.pbb.2025.173972] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 02/10/2025] [Accepted: 02/14/2025] [Indexed: 02/23/2025]
Abstract
Huntington's disease (HD) is a progressive neurodegenerative disorder characterized by motor dysfunction, cognitive decline, and emotional instability, primarily resulting from the abnormal accumulation of mutant huntingtin protein. Growing research highlights the role of intestinal microbiota and their metabolites, particularly short-chain fatty acids (SCFAs), in modulating HD progression. SCFAs, including acetate, propionate, and butyrate, are produced by gut bacteria through dietary fiber fermentation and are recognized for their neuroprotective properties. Evidence suggests that SCFAs regulate neuroinflammation, neuronal communication, and metabolic functions within the central nervous system (CNS). In HD, these compounds may support neuronal health, reduce oxidative stress, and enhance blood-brain barrier (BBB) integrity. Their mechanisms of action involve binding to G-protein-coupled receptors (GPCRs) and modulating gene expression through epigenetic pathways, underscoring their therapeutic potential. This analysis examines the significance of SCFAs in HD, emphasizing the gut-brain axis and the benefits of dietary interventions aimed at modifying gut microbiota composition and promoting SCFA production. Further research into these pathways may pave the way for novel HD management strategies and improved therapeutic outcomes.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman 11831, Jordan.
| | | | | | - Gaurav Sanghvi
- Marwadi University Research Center, Department of Microbiology, Faculty of Science, Marwadi University, Rajkot 360003, Gujarat, India
| | - Subbulakshmi Ganesan
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to be University), Bangalore, Karnataka, India
| | - Mohit Agarwal
- Department of Pharmaceutical Chemistry, NIMS Institute of Pharmacy, NIMS University, Rajasthan, Jaipur,302131, India
| | - Parjinder Kaur
- Chandigarh Pharmacy College, Chandigarh Group of Colleges-Jhanjeri, Mohali 140307, Punjab, India
| | - Waam Mohammed Taher
- College of Nursing, National University of Science and Technology, Dhi Qar, Iraq
| | | | | | | |
Collapse
|
3
|
Yakut A. Gut microbiota in the development and progression of chronic liver diseases: Gut microbiota-liver axis. World J Hepatol 2025; 17:104167. [DOI: 10.4254/wjh.v17.i3.104167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2024] [Revised: 01/28/2025] [Accepted: 02/25/2025] [Indexed: 03/26/2025] Open
Abstract
The gut microbiota (GM) is a highly dynamic ecology whose density and composition can be influenced by a wide range of internal and external factors. Thus, “How do GM, which can have commensal, pathological, and mutualistic relationships with us, affect human health?” has become the most popular research issue in recent years. Numerous studies have demonstrated that the trillions of microorganisms that inhabit the human body can alter host physiology in a variety of systems, such as metabolism, immunology, cardiovascular health, and neurons. The GM may have a role in the development of a number of clinical disorders by producing bioactive peptides, including neurotransmitters, short-chain fatty acids, branched-chain amino acids, intestinal hormones, and secondary bile acid conversion. These bioactive peptides enter the portal circulatory system through the gut-liver axis and play a role in the development of chronic liver diseases, cirrhosis, and hepatic encephalopathy. This procedure is still unclear and quite complex. In this study, we aim to discuss the contribution of GM to the development of liver diseases, its effects on the progression of existing chronic liver disease, and to address the basic mechanisms of the intestinal microbiota-liver axis in the light of recent publications that may inspire the future.
Collapse
Affiliation(s)
- Aysun Yakut
- Department of Gastroenterology, İstanbul Medipol University Sefakoy Health Practice Research Center, İstanbul 38000, Türkiye
| |
Collapse
|
4
|
Kugawa M, Kawakami K, Kise R, Suomivuori CM, Tsujimura M, Kobayashi K, Kojima A, Inoue WJ, Fukuda M, Matsui TE, Fukunaga A, Koyanagi J, Kim S, Ikeda H, Yamashita K, Saito K, Ishikita H, Dror RO, Inoue A, Kato HE. Structural insights into lipid chain-length selectivity and allosteric regulation of FFA2. Nat Commun 2025; 16:2809. [PMID: 40140663 PMCID: PMC11947310 DOI: 10.1038/s41467-025-57983-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2024] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
The free fatty acid receptor 2 (FFA2) is a G protein-coupled receptor (GPCR) that selectively recognizes short-chain fatty acids to regulate metabolic and immune functions. As a promising therapeutic target, FFA2 has been the focus of intensive development of synthetic ligands. However, the mechanisms by which endogenous and synthetic ligands modulate FFA2 activity remain unclear. Here, we present the structures of the human FFA2-Gi complex activated by the synthetic orthosteric agonist TUG-1375 and the positive allosteric modulator/allosteric agonist 4-CMTB, along with the structure of the inactive FFA2 bound to the antagonist GLPG0974. Structural comparisons with FFA1 and mutational studies reveal how FFA2 selects specific fatty acid chain lengths. Moreover, our structures reveal that GLPG0974 functions as an allosteric antagonist by binding adjacent to the orthosteric pocket to block agonist binding, whereas 4-CMTB binds the outer surface of transmembrane helices 6 and 7 to directly activate the receptor. Supported by computational and functional studies, these insights illuminate diverse mechanisms of ligand action, paving the way for precise GPCR-targeted drug design.
Collapse
Affiliation(s)
- Mai Kugawa
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro, Tokyo, Japan
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Kouki Kawakami
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro, Tokyo, Japan
| | - Ryoji Kise
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Carl-Mikael Suomivuori
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA
| | - Masaki Tsujimura
- Department of Advanced Interdisciplinary Studies, The University of Tokyo, Meguro, Tokyo, Japan
| | - Kazuhiro Kobayashi
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro, Tokyo, Japan
| | - Asato Kojima
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro, Tokyo, Japan
- Department of Life Sciences, School of Arts and Sciences, The University of Tokyo, Meguro, Tokyo, Japan
| | - Wakana J Inoue
- Department of Life Sciences, School of Arts and Sciences, The University of Tokyo, Meguro, Tokyo, Japan
| | - Masahiro Fukuda
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro, Tokyo, Japan
| | - Toshiki E Matsui
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro, Tokyo, Japan
- Department of Life Sciences, School of Arts and Sciences, The University of Tokyo, Meguro, Tokyo, Japan
| | - Ayami Fukunaga
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro, Tokyo, Japan
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Junki Koyanagi
- Department of Life Sciences, School of Arts and Sciences, The University of Tokyo, Meguro, Tokyo, Japan
| | - Suhyang Kim
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro, Tokyo, Japan
| | - Hisako Ikeda
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro, Tokyo, Japan
| | - Keitaro Yamashita
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro, Tokyo, Japan
| | - Keisuke Saito
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro, Tokyo, Japan
- Department of Applied Chemistry, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Hiroshi Ishikita
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro, Tokyo, Japan
- Department of Applied Chemistry, The University of Tokyo, Bunkyo, Tokyo, Japan
| | - Ron O Dror
- Department of Computer Science, Stanford University, Stanford, CA, USA
- Department of Molecular and Cellular Physiology, Stanford University School of Medicine, Stanford, CA, USA
- Department of Structural Biology, Stanford University School of Medicine, Stanford, CA, USA
- Institute for Computational and Mathematical Engineering, Stanford University, Stanford, CA, USA
| | - Asuka Inoue
- Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.
- Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, Japan.
- FOREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan.
| | - Hideaki E Kato
- Research Center for Advanced Science and Technology, The University of Tokyo, Meguro, Tokyo, Japan.
- Department of Biological Sciences, Graduate School of Science, The University of Tokyo, Bunkyo, Tokyo, Japan.
- Department of Life Sciences, School of Arts and Sciences, The University of Tokyo, Meguro, Tokyo, Japan.
- FOREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan.
- CREST, Japan Science and Technology Agency, Kawaguchi, Saitama, Japan.
| |
Collapse
|
5
|
Diesse JM, Jadhav S, Tamekou SL, Simo G, Dzoyem JP, Souopgui J, Kuiate JR, Nema V. Disturbances in the gut microbiota potentially associated with metabolic syndrome among patients living with HIV-1 and on antiretroviral therapy at Bafoussam Regional Hospital, Cameroon. Diabetol Metab Syndr 2025; 17:86. [PMID: 40089790 PMCID: PMC11909933 DOI: 10.1186/s13098-025-01653-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 02/27/2025] [Indexed: 03/17/2025] Open
Abstract
BACKGROUND This study investigates the gut microbiota components associated with metabolic syndrome in patients living with HIV-1 at Bafoussam Regional Hospital, West Cameroon, it focuses on gastrointestinal mucosal barrier disruption and dysbiosis, and their effects on persistent inflammation and metabolic disorders. METHODS A pilot study was conducted involving fourteen patients living with HIV-1. The patients were divided into two groups of seven in each group. One group consisted of patients with metabolic syndrome, and the other group included patients without metabolic syndrome. Gut microbiota was characterized using 16 S rRNA gene-targeted sequencing to analyze microbial diversity and composition. Beta diversity and the relative abundance of bacterial taxa were compared between patients with and without metabolic syndrome. RESULTS Patients living with HIV-1 and metabolic syndrome showed significantly altered beta diversity compared to those without metabolic syndrome. A higher relative abundance of Firmicutes and increased proliferation of Proteobacteria were observed in patients with metabolic syndrome. Additionally, a decrease in metabolically beneficial bacteria, such as Bifidobacterium sp., Lactobacillus sp., Akkermansia sp., and Faecalibacterium sp., was noted. Several beneficial bacterial species were associated with participants' metadata, suggesting potential links between gut microbiota and metabolic syndrome. CONCLUSION This preliminary study highlights that gut microbial balance, rather than the presence of specific bacteria, plays a crucial role in managing metabolic health in patients living with HIV-1. The altered gut microbiota in participants with metabolic syndrome emphasizes the need for further research into the optimal gut microbial structure. Understanding the interaction between gut microbiota changes and the chemical environment in these patients could guide targeted interventions to improve metabolic outcomes.
Collapse
Affiliation(s)
- Joël Martial Diesse
- Department of Biochemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
| | - Sushama Jadhav
- Division of Molecular Biology, Indian Council of Medical Research - National Institute of Translational Virology and AIDS Research , 73 G MIDC Bhosari, Pune, 411026, India
| | | | - Gustave Simo
- Department of Biochemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
| | - Jean Paul Dzoyem
- Department of Biochemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
| | - Jacob Souopgui
- Laboratory of Embryology & Biotechnology DBM-IBMM, "Université Libre de Bruxelles", Brussels, Belgium
| | - Jules-Roger Kuiate
- Department of Biochemistry, Faculty of Science, University of Dschang, Dschang, Cameroon
| | - Vijay Nema
- Division of Molecular Biology, Indian Council of Medical Research - National Institute of Translational Virology and AIDS Research , 73 G MIDC Bhosari, Pune, 411026, India.
| |
Collapse
|
6
|
Song J, Wang C, Zhao T, Zhang Y, Xing J, Zhao X, Zhang Y, Zhang Z. Multi-omics approaches for biomarker discovery and precision diagnosis of prediabetes. Front Endocrinol (Lausanne) 2025; 16:1520436. [PMID: 40162315 PMCID: PMC11949806 DOI: 10.3389/fendo.2025.1520436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 02/24/2025] [Indexed: 04/02/2025] Open
Abstract
Recent advancements in multi-omics technologies have provided unprecedented opportunities to identify biomarkers associated with prediabetes, offering novel insights into its diagnosis and management. This review synthesizes the latest findings on prediabetes from multiple omics domains, including genomics, epigenomics, transcriptomics, proteomics, metabolomics, microbiomics, and radiomics. We explore how these technologies elucidate the molecular and cellular mechanisms underlying prediabetes and analyze potential biomarkers with predictive value in disease progression. Integrating multi-omics data helps address the limitations of traditional diagnostic methods, enabling early detection, personalized interventions, and improved patient outcomes. However, challenges such as data integration, standardization, and clinical validation and translation remain to be resolved. Future research leveraging artificial intelligence and machine learning is expected to further enhance the predictive power of multi-omics technologies, contributing to the precision diagnosis and tailored management of prediabetes.
Collapse
Affiliation(s)
- Jielin Song
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- TCM Institute of Sore and Ulcer, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Institute of Traditional Chinese Medicine Surgery, Tianjin, China
| | - Chuanfu Wang
- Department of Encephalopathy, Liangping District Hospital of Traditional Chinese Medicine, Chongqing, China
| | - Tong Zhao
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- TCM Institute of Sore and Ulcer, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Institute of Traditional Chinese Medicine Surgery, Tianjin, China
| | - Yu Zhang
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- TCM Institute of Sore and Ulcer, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Institute of Traditional Chinese Medicine Surgery, Tianjin, China
| | - Jixiang Xing
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- TCM Institute of Sore and Ulcer, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Institute of Traditional Chinese Medicine Surgery, Tianjin, China
| | - Xuelian Zhao
- Graduate School, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- TCM Institute of Sore and Ulcer, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Institute of Traditional Chinese Medicine Surgery, Tianjin, China
| | - Yunsha Zhang
- TCM Institute of Sore and Ulcer, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Institute of Traditional Chinese Medicine Surgery, Tianjin, China
- School of Integrative Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin, China
| | - Zhaohui Zhang
- TCM Institute of Sore and Ulcer, Tianjin University of Traditional Chinese Medicine, Tianjin, China
- Tianjin Institute of Traditional Chinese Medicine Surgery, Tianjin, China
- Department of Traditional Chinese Medicine Surgery, The Second Affiliated Hospital of Tianjin University of Traditional Chinese Medicine, Tianjin, China
| |
Collapse
|
7
|
Menozzi E, Schapira AHV, Borghammer P. The Gut-Brain Axis in Parkinson disease: Emerging Concepts and Therapeutic Implications. Mov Disord Clin Pract 2025. [PMID: 40079755 DOI: 10.1002/mdc3.70029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 02/20/2025] [Accepted: 02/22/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND The gut-brain axis, i.e. the bidirectional communication system between the gut and the brain, has become of central importance in Parkinson disease (PD) research over the past 20 years. AIMS We aimed to describe the milestones of the gut-brain axis research in PD and the development of theories proposing the involvement of the gastrointestinal tract in PD pathogenesis. METHODS We searched PubMed using the terms 'gut-brain axis' AND 'Parkinson disease', and selected relevant articles to provide the foundation for reconstructing an historical overview of the gut-brain axis research in PD. RESULTS Mounting evidence from preclinical, clinical and post-mortem studies suggests that a subgroup of PD patients present with a range of prodromal symptoms (e.g., autonomic dysfunction, rapid eye movement sleep behaviour disorder) which reflect initial accumulation and later spread of pathological α-synuclein rostrally from the gastrointestinal tract ("body-first" PD). Through neural connections along the gut-brain axis, pathological α-synuclein may spread to the brain, producing clinically manifest disease. Recently, two mechanisms involving the gut-brain axis have attracted increasing attention for their role in PD pathogenesis and progression, namely the perturbation of the composition of the microorganisms living in the gut (the gut microbiome), and the dysfunction of enteroendocrine cells. CONCLUSION Treatments targeting the gut-brain axis, especially the gut microbiome and the enteroendocrine cells pathway, could potentially slow disease progression or even prevent disease onset. Among these, pre/probiotics, faecal microbiota transplantation, and glucagon-like peptide-1 receptor agonists, have entered advanced stages of clinical trials in humans and shown potential symptomatic and disease-modifying effects.
Collapse
Affiliation(s)
- Elisa Menozzi
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Anthony H V Schapira
- Department of Clinical and Movement Neurosciences, UCL Queen Square Institute of Neurology, London, UK
- Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, Maryland, USA
| | - Per Borghammer
- Department of Clinical Medicine, Aarhus University, Aarhus, Denmark
- Department of Nuclear Medicine and PET, Aarhus University Hospital, Aarhus, Denmark
| |
Collapse
|
8
|
Kang L, Li X, Zhao X, Liu T, Jin Y, Duan Y. Effects of L-arginine supplementation on fat deposition and meat quality in growing lambs: Interactions with gut microbiota and metabolic signalling pathways. Food Chem 2025; 479:143677. [PMID: 40081064 DOI: 10.1016/j.foodchem.2025.143677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 02/23/2025] [Accepted: 02/28/2025] [Indexed: 03/15/2025]
Abstract
Arginine (ARG) improves meat quality and fat deposition; however, its effects on gut microbiota-mediated lipid metabolism in lambs remain unclear. Twenty-four lambs were divided into control (fed a basal diet) and ARG groups (with 1 % ARG added). In the ARG group, backfat thickness, shear force in the longissimus thoracis (LT) muscle, and C16:0 and SFA contents in the subcutaneous adipose tissue (SAT) were reduced, whereas the eye muscle area, a* value, and intramuscular fat, C18:2n-6c, C20:4n-6, C20:5n-3, and PUFA contents in the LT were elevated. Moreover, the ARG group exhibited higher levels of Prevotella, Akkermansia, Faecalibacterium, SCFAs, and GLP-1 in the colon, and lower serum triglyceride and glucose levels. Interestingly, ARG differentially regulated lipid metabolism in the SAT and LT via the GLP-1R/AMPK and triglyceride metabolism signalling pathways. Overall, ARG addition may optimise gut microbiota composition, fat deposition, and meat quality, providing application guidance for regulating fat deposition in lambs.
Collapse
Affiliation(s)
- Letian Kang
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Integrative Research Base of Beef and Lamb Processing Technology, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Hohhot 010018, China
| | - Xuan Li
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Integrative Research Base of Beef and Lamb Processing Technology, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Hohhot 010018, China
| | - Xin Zhao
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Integrative Research Base of Beef and Lamb Processing Technology, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Hohhot 010018, China
| | - Ting Liu
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Integrative Research Base of Beef and Lamb Processing Technology, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Hohhot 010018, China
| | - Ye Jin
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Integrative Research Base of Beef and Lamb Processing Technology, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Hohhot 010018, China
| | - Yan Duan
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Hohhot 010018, China; Integrative Research Base of Beef and Lamb Processing Technology, Ministry of Agriculture and Rural Affairs of the People's Republic of China, Hohhot 010018, China.
| |
Collapse
|
9
|
Hou Y, Gao X, Gong J, Dong X, Hao Y, Zhai Z, Zhang H, Zhang M, Liu R, Wang R, Zhao L. Targeted Sodium Acetate Liposomes for Hepatocytes and Kupffer Cells: An Oral Dual-Targeted Therapeutic Approach for Non-Alcoholic Fatty Liver Disease Alleviation. Nutrients 2025; 17:930. [PMID: 40077800 PMCID: PMC11901740 DOI: 10.3390/nu17050930] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 02/28/2025] [Accepted: 03/05/2025] [Indexed: 03/14/2025] Open
Abstract
Background/Objectives: Sodium acetate (NaA) has demonstrated potential in improving non-alcoholic fatty liver disease (NAFLD) by targeting hepatocytes and Kupffer cells. However, its clinical application is hindered by low oral bioavailability and insufficient liver concentrations. Liposomes, with their capacity to encapsulate water-soluble drugs and be surface-modified, offer a promising solution for targeted oral drug delivery. Methods: We designed NaA-loaded liposomes modified with sodium cholate (SC) and mannose (MAN) (NaA@SC/MAN-LPs) to target hepatocytes and Kupffer cells. Results: The NaA@SC/MAN-LPs had a mean diameter of approximately 100 nm with a positive surface charge. Compared to free NaA, NaA@SC/MAN-LPs significantly extended the serum half-life from 2.85 h to 15.58 h, substantially improving in vivo bioavailability. In vivo distribution studies revealed that NaA@SC/MAN-LPs extended the acetate peak time in the liver from 15 min to 60 min and increased hepatic acetate accumulation to 3.75 times that of free NaA. In in vitro cell experiments, NaA@SC/MAN-LPs significantly reduced the lipid droplet, triglycerides (TG), and total cholesterol (TC) in a fatty acid-induced hepatocyte steatosis model and suppressed proinflammation in a lipopolysaccharide (LPS)-activated Kupffer cell inflammation model. Free NaA effectively improved hepatic lipid deposition in NAFLD mice. Furthermore, NaA@SC/MAN-LPs decreased hepatic TG, TC, and the relative area of lipid droplets by 30.44%, 15.26%, and 55.83%, compared to free NaA. Furthermore, the liposomes reduced macrophage infiltration and pro-inflammatory response. Conclusions: The NaA@SC/MAN-LPs demonstrated effective dual targeting effects on hepatocytes and Kupffer cells, significantly improving the pathogenesis of NAFLD, compared to free NaA. This study provides a new strategy for developing effective and safe oral drugs for NAFLD.
Collapse
Affiliation(s)
- Yichao Hou
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (Y.H.); (X.G.); (Y.H.); (R.L.); (R.W.)
| | - Xilong Gao
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (Y.H.); (X.G.); (Y.H.); (R.L.); (R.W.)
| | - Jiahui Gong
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (J.G.); (X.D.); (Z.Z.); (H.Z.)
| | - Xinrui Dong
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (J.G.); (X.D.); (Z.Z.); (H.Z.)
| | - Yanling Hao
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (Y.H.); (X.G.); (Y.H.); (R.L.); (R.W.)
| | - Zhengyuan Zhai
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (J.G.); (X.D.); (Z.Z.); (H.Z.)
| | - Hao Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (J.G.); (X.D.); (Z.Z.); (H.Z.)
| | - Ming Zhang
- School of Food and Health, Beijing Technology and Business University, Beijing 100048, China;
| | - Rong Liu
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (Y.H.); (X.G.); (Y.H.); (R.L.); (R.W.)
| | - Ran Wang
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (Y.H.); (X.G.); (Y.H.); (R.L.); (R.W.)
- Research Center for Probiotics, China Agricultural University, Beijing 101299, China
| | - Liang Zhao
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (Y.H.); (X.G.); (Y.H.); (R.L.); (R.W.)
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China; (J.G.); (X.D.); (Z.Z.); (H.Z.)
- Research Center for Probiotics, China Agricultural University, Beijing 101299, China
| |
Collapse
|
10
|
Cremer A, Rosewick N, Kelsey M, Trépo E, Libert F, De Vos M, Baert F, Moreels T, Louis E, Rahier JF, Demetter P, Sedivy JM, Vermeire S, Franchimont D. A transcriptomic score to classify the inflammation-dysplasia-cancer sequence lesions in inflammatory bowel disease. J Crohns Colitis 2025; 19:jjaf026. [PMID: 39945142 PMCID: PMC11904305 DOI: 10.1093/ecco-jcc/jjaf026] [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] [Indexed: 03/14/2025]
Abstract
BACKGROUND AND AIMS Inflammatory bowel disease (IBD) is associated with a higher risk of developing colorectal cancer, according to the inflammation-dysplasia-cancer (IDC) sequence from inflammation to colitis-associated colorectal cancer (CAC). The objective of this study was to identify and generate a transcriptomic signature and score, related to the IDC sequence, that could ultimately classify dysplasia and cancer in IBD. METHODS Demographics, clinical parameters, histological characteristics, and RNA-sequencing data were evaluated on 134 formalin-fixed paraffin-embedded lesions from 2 independent cohorts of IBD patients with low- or high-grade dysplasia (LGD, HGD) and/or CAC. An ordinal logistic regression screened for significant IDC sequence-associated genes that were computed in a transcriptomic signature score. RESULTS Principal component analysis and unsupervised clustering on 1% of the most variable genes showed a good clustering between the 4 lesion groups (Normal Mucosa, Inflamed Mucosa, LGD/HGD, and CAC). A gene signature was identified on 27 genes that correlated with the lesion groups in the exploratory cohort. The most weighted gene in this transcriptomic signature was the long non-coding regulatory RNA KCNQ1OT1, a gatekeeper against genomic instability and transposon activation. Based on the expression of these 27 genes, we built and validated a transcriptomic signature score to classify dysplasia and CAC. The overall accuracy of the transcriptomic signature score was 85.71% in the exploratory cohort and 90.91% in the validation cohort. CONCLUSION We identified a tissue-based transcriptomic score to classify IDC lesions in IBD patients and uncovered some of the pivotal genes in carcinogenesis related to inflammation in IBD.
Collapse
Affiliation(s)
- Anneline Cremer
- Department of Gastroenterology, HUB Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| | - Nicolas Rosewick
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| | - Maxfield Kelsey
- Center on the Biology of Aging, and the Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, United States
| | - Eric Trépo
- Department of Gastroenterology, HUB Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| | - Frédérick Libert
- Institut de Recherche Interdisciplinaire en Biologie Humaine et Moléculaire (IRIBHM), Université Libre de Bruxelles, Brussels, Belgium
| | - Martine De Vos
- Department of Gastroenterology, University Hospital Ghent, Ghent, Belgium
| | - Filip Baert
- Department of Gastroenterology, AZ Delta, Roeselare, Belgium
| | - Tom Moreels
- Department of Gastroenterology, University Hospital Antwerp, Edegem, Belgium
| | - Edouard Louis
- Department of Gastroenterology, University Hospital Liège, Liège, Belgium
| | - Jean-François Rahier
- Department of Gastroenterology, CHU UCL Namur site Mont-Godinne, Université Catholique de Louvain, Yvoir, Belgium
| | - Pieter Demetter
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| | - John M Sedivy
- Center on the Biology of Aging, and the Department of Molecular Biology, Cell Biology, and Biochemistry, Brown University, Providence, RI, United States
| | - Séverine Vermeire
- Department of Gastroenterology, University Hospital Leuven, Leuven, Belgium
| | - Denis Franchimont
- Department of Gastroenterology, HUB Erasme University Hospital, Université Libre de Bruxelles, Brussels, Belgium
- Laboratory of Experimental Gastroenterology, Université Libre de Bruxelles, Brussels, Belgium
| |
Collapse
|
11
|
Ng HY, Zhang L, Tan JT, Hui RWH, Yuen MF, Seto WK, Leung WK, Cheung KS. Gut Microbiota Predicts Treatment Response to Empagliflozin Among MASLD Patients Without Diabetes Mellitus. Liver Int 2025; 45:e70023. [PMID: 39950834 PMCID: PMC11827547 DOI: 10.1111/liv.70023] [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] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Revised: 01/11/2025] [Accepted: 01/30/2025] [Indexed: 02/17/2025]
Abstract
BACKGROUND AND AIM We aimed to investigate whether gut microbiota could predict the treatment response to pharmacological agents among metabolic dysfunction-associated steatotic liver disease (MASLD) patients without diabetes mellitus (DM), as data are lacking. METHODS We prospectively followed up non-diabetic MASLD patients who used empagliflozin. Clinical, anthropometric, laboratory assessments and magnetic resonance imaging-proton density fat fraction (MRI-PDFF) were performed from baseline to week 52 (EOT). Baseline stool samples were collected, and shotgun DNA metagenomic sequencing was performed to profile microbiome. The primary outcome was treatment response to empagliflozin at EOT, defined as MRI-PDFF decline ≥ 30% at EOT from baseline. Linear discriminant analysis [LDA] effect size was used to identify putative bacterial species. Multivariable logistic regression was used to derive adjusted odds ratio (aOR) of outcome with bacterial species by adjusting for clinical factors. RESULTS Twenty-two (48.9%) of 45 patients (median age: 56.9 years [IQR: 51.0-63.2]; male: 23 [51.1%]) achieved treatment response at EOT. There was difference in alpha diversity (Shannon index: p < 0.001; Simpson index: p = 0.001) and beta diversity (p = 0.048) in baseline microbiome between treatment response and non-response groups. Faecalibacterium prausnitzii (log10LDAscore = 4.27), Lachnospira pectinoschiza (log10LDAscore = 3.99), Anaerostipes hadrus (log10LDAscore = 3.98), Roseburia faecis (log10LDAscore = 3.97), Roseburia inulinivorans (log10LDAscore = 3.58) and Agathobaculum butyriciproducens (log10LDAscore = 2.77) were enriched in the treatment response group. L. pectinoschiza (aOR: 34.1; p = 0.015), A. hadrus (aOR:35.0; p = 0.032) and A. butyriciproducens (aOR:22.3; p = 0.023) independently predicted treatment response but not clinical factors. These three species collectively predicted treatment response with AUROC of 0.89 (95% CI: 0.80-0.99). CONCLUSIONS Certain gut bacterial species, particularly the combination of A. hadrus, L. pectinoschiza and A. butyriciproducens, may predict treatment response to empagliflozin in MAFLD patients without DM.
Collapse
Affiliation(s)
- Ho Yu Ng
- Li Ka Shing Faculty of MedicineThe University of Hong Kong, Queen Mary HospitalHong Kong
| | - Lina Zhang
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of MedicineThe University of Hong Kong, Queen Mary HospitalHong Kong
| | - Jing Tong Tan
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of MedicineThe University of Hong Kong, Queen Mary HospitalHong Kong
| | - Rex Wan Hin Hui
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of MedicineThe University of Hong Kong, Queen Mary HospitalHong Kong
| | - Man Fung Yuen
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of MedicineThe University of Hong Kong, Queen Mary HospitalHong Kong
- State Key Laboratory of Liver ResearchThe University of Hong KongHong Kong
| | - Wai Kay Seto
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of MedicineThe University of Hong Kong, Queen Mary HospitalHong Kong
- State Key Laboratory of Liver ResearchThe University of Hong KongHong Kong
| | - Wai K. Leung
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of MedicineThe University of Hong Kong, Queen Mary HospitalHong Kong
| | - Ka Shing Cheung
- Department of Medicine, School of Clinical Medicine, Li Ka Shing Faculty of MedicineThe University of Hong Kong, Queen Mary HospitalHong Kong
| |
Collapse
|
12
|
Gawey BJ, Mars RA, Kashyap PC. The role of the gut microbiome in disorders of gut-brain interaction. FEBS J 2025; 292:1357-1377. [PMID: 38922780 PMCID: PMC11664017 DOI: 10.1111/febs.17200] [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: 01/14/2024] [Revised: 04/03/2024] [Accepted: 06/05/2024] [Indexed: 06/28/2024]
Abstract
Disorders of Gut-Brain Interaction (DGBI) are widely prevalent and commonly encountered in gastroenterology practice. While several peripheral and central mechanisms have been implicated in the pathogenesis of DGBI, a recent body of work suggests an important role for the gut microbiome. In this review, we highlight how gut microbiota and their metabolites affect physiologic changes underlying symptoms in DGBI, with a particular focus on their mechanistic influence on GI transit, visceral sensitivity, intestinal barrier function and secretion, and CNS processing. This review emphasizes the complexity of local and distant effects of microbial metabolites on physiological function, influenced by factors such as metabolite concentration, duration of metabolite exposure, receptor location, host genetics, and underlying disease state. Large-scale in vitro work has elucidated interactions between host receptors and the microbial metabolome but there is a need for future research to integrate such preclinical findings with clinical studies. The development of novel, targeted therapeutic strategies for DGBI hinges on a deeper understanding of these metabolite-host interactions, offering exciting possibilities for the future of treatment of DGBI.
Collapse
Affiliation(s)
- Brent J Gawey
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Ruben A Mars
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| | - Purna C Kashyap
- Division of Gastroenterology and Hepatology, Department of Medicine, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
13
|
Romaní-Pérez M, Líebana-García R, Flor-Duro A, Bonillo-Jiménez D, Bullich-Vilarrubias C, Olivares M, Sanz Y. Obesity and the gut microbiota: implications of neuroendocrine and immune signaling. FEBS J 2025; 292:1397-1420. [PMID: 39159270 PMCID: PMC11927058 DOI: 10.1111/febs.17249] [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: 12/06/2023] [Revised: 06/29/2024] [Accepted: 08/06/2024] [Indexed: 08/21/2024]
Abstract
Obesity is a major health challenge due to its high prevalence and associated comorbidities. The excessive intake of a diet rich in fat and sugars leads to a persistent imbalance between energy intake and energy expenditure, which increases adiposity. Here, we provide an update on relevant diet-microbe-host interactions contributing to or protecting from obesity. In particular, we focus on how unhealthy diets shape the gut microbiota and thus impact crucial intestinal neuroendocrine and immune system functions. We describe how these interactions promote dysfunction in gut-to-brain neuroendocrine pathways involved in food intake control and postprandial metabolism and elevate the intestinal proinflammatory tone, promoting obesity and metabolic complications. In addition, we provide examples of how this knowledge may inspire microbiome-based interventions, such as fecal microbiota transplants, probiotics, and biotherapeutics, to effectively combat obesity-related disorders. We also discuss the current limitations and gaps in knowledge of gut microbiota research in obesity.
Collapse
Affiliation(s)
- Marina Romaní-Pérez
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Rebeca Líebana-García
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Alejandra Flor-Duro
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Daniel Bonillo-Jiménez
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Clara Bullich-Vilarrubias
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Marta Olivares
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| | - Yolanda Sanz
- Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Valencia, Spain
| |
Collapse
|
14
|
Xie C, Qi C, Zhang J, Wang W, Meng X, Aikepaer A, Lin Y, Su C, Liu Y, Feng X, Gao H. When short-chain fatty acids meet type 2 diabetes mellitus: Revealing mechanisms, envisioning therapies. Biochem Pharmacol 2025; 233:116791. [PMID: 39894305 DOI: 10.1016/j.bcp.2025.116791] [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: 10/25/2024] [Revised: 01/19/2025] [Accepted: 01/30/2025] [Indexed: 02/04/2025]
Abstract
Evidence is accumulating that short-chain fatty acids (SCFAs) produced by the gut microbiota play pivotal roles in host metabolism. They contribute to the metabolic regulation and energy homeostasis of the host not only by preserving intestinal health and serving as energy substrates but also by entering the systemic circulation as signaling molecules, affecting the gut-brain axis and neuroendocrine-immune network. This review critically summarizes the current knowledge regarding the effects of SCFAs in the fine-tuning of the pathogenesis of type 2 diabetes mellitus (T2DM) and insulin resistance, with an emphasis on the complex relationships among diet, microbiota-derived metabolites, T2DM inflammation, glucose metabolism, and the underlying mechanisms involved. We hold an optimistic view that elucidating how diet can influence gut bacterial composition and activity, SCFA production, and metabolic functions in the host will advance our understanding of the mutual interactions of the intestinal microbiota with other metabolically active organs, and may pave the way for harnessing these pathways to develop novel personalized therapeutics for glucometabolic disorders.
Collapse
Affiliation(s)
- Cong Xie
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China
| | - Cong Qi
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China
| | - Jianwen Zhang
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China; School of Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617 China
| | - Wei Wang
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China
| | - Xing Meng
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China; School of Chinese Medicine, Tianjin University of Traditional Chinese Medicine, Tianjin 301617 China
| | - Aifeila Aikepaer
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China; Dongzhimen Hospital, the First Clinical Medical School of Beijing University of Chinese Medicine, Beijing 100700 China
| | - Yuhan Lin
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China; Dongzhimen Hospital, the First Clinical Medical School of Beijing University of Chinese Medicine, Beijing 100700 China
| | - Chang Su
- Life Science and Engineering College, Northwest Minzu University, Lanzhou 730124 China
| | - Yunlu Liu
- Experimental Research Center, China Academy of Chinese Medical Sciences, Beijing 100700 China
| | - Xingzhong Feng
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China.
| | - Huijuan Gao
- Department of Endocrinology, Yuquan Hospital, School of Clinical Medicine, Tsinghua University, Beijing 100040 China.
| |
Collapse
|
15
|
Guo X, Su Y, Du Y, Zhang F, Yu W, Ren W, Li S, Kuang H, Wu L. Vinegar-processed Schisandra chinensis polysaccharide ameliorates type 2 diabetes via modulation serum metabolic profiles, gut microbiota, and fecal SCFAs. Int J Biol Macromol 2025; 294:139514. [PMID: 39761882 DOI: 10.1016/j.ijbiomac.2025.139514] [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] [Revised: 12/18/2024] [Accepted: 01/03/2025] [Indexed: 01/21/2025]
Abstract
Numerous studies indicate that Schisandra chinensis (Turcz.) Baill (SC) has anti-type 2 diabetes mellitus (T2DM) effects, and its processed products are commonly used in clinical practice. However, limited reports exist on the mechanisms of polysaccharides from its vinegar products and their role in T2DM. We purified a novel polysaccharide from vinegar-processed Schisandra chinensis (VSC) and used intestinal microbiota 16S rRNA analysis and metabolomics to study changes in T2DM mice after vinegar-processed Schisandra chinensis polysaccharide (VSP) intervention, aiming to elucidate how VSP alleviates T2DM. VSP has shown significant therapeutic effects in T2DM mice, which can regulate the imbalance of glucose and lipid metabolism, alleviate pancreatic and liver damage, restore the integrity of the intestinal barrier, and inhibit the inflammatory response. Serum metabolomics and microbiological analysis showed that VSP could significantly regulate 104 endogenous metabolites and rectify gut microbiota disorders in T2DM mice. Additionally, VSP enhanced the levels of short-chain fatty acids (SCFAs) and the expression of GPR41/43 in the colon of T2DM mice. Correlation analysis revealed significant correlations among specific gut microbiota, serum metabolites, and fecal SCFAs. Overall, these findings will provide a basis for further VSP development.
Collapse
Affiliation(s)
- Xingyu Guo
- School of Pharmacy, Heilongjiang University Of Chinese Medicine, Harbin 150040, China
| | - Yang Su
- School of Pharmacy, Heilongjiang University Of Chinese Medicine, Harbin 150040, China
| | - Yongqiang Du
- Heilongjiang Province Healthcare Security Administration, Harbin 150036, China
| | - Fan Zhang
- School of Pharmacy, Heilongjiang University Of Chinese Medicine, Harbin 150040, China
| | - Wenting Yu
- School of Pharmacy, Heilongjiang University Of Chinese Medicine, Harbin 150040, China
| | - Wenchen Ren
- School of Pharmacy, Heilongjiang University Of Chinese Medicine, Harbin 150040, China
| | - Shanshan Li
- Heilongjiang Province Health Management Service Evaluation Center, Harbin 150030, China
| | - Haixue Kuang
- School of Pharmacy, Heilongjiang University Of Chinese Medicine, Harbin 150040, China
| | - Lun Wu
- Institute of Traditional Chinese Medicine, Heilongjiang University of Chinese Medicine, Harbin 150040, China.
| |
Collapse
|
16
|
Kawamura M, Hariya N, Ishiyama S, Tanaka Y, Ozato K, Mochizuki K. Effects of feeding the medium-chain triacylglycerol on GLP-1 secretion in aged Brd4-heterozygous mice, which are a mouse model of aging. Nutrition 2025; 131:112656. [PMID: 39708424 DOI: 10.1016/j.nut.2024.112656] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Revised: 11/03/2024] [Accepted: 11/19/2024] [Indexed: 12/23/2024]
Abstract
OBJECTIVES Gastrointestinal hormones, such as glucagon-like peptide 1 (GLP-1), gastric inhibitory polypeptide, and peptide YY (PYY) are important for reducing malnutrition at older ages because they are related to assimilation and feeding behavior. Medium-chain triacylglycerol (MCT) ameliorates metabolic symptoms and frailty in adults; however, whether it has the same effect in old age is unknown. To address this, we examined the changes in insulin and gastrointestinal hormones in aged Brd4 (+/-) mice exhibiting symptoms of old age. RESEARCH METHODS AND PROCEDURES Aged male wild-type and Brd4 (+/-) mice were fed a long-chain triacylglycerol (LCT)- or MCT diet. Feeding, blood glucose, and plasma active GLP-1 protein concentrations were determined at 9 weeks using a meal tolerance test, and those gastrointestinal hormone genes were determined at 10 weeks. RESULTS The liver and stomach weights and mRNA expression of Gcg (encodes GLP-1 protein) and Pyy in the colon were lower in LCT-fed Brd4 (+/-) mice than those in LCT-fed wild-type mice; these were restored by the MCT diet. The blood concentration of active GLP-1 protein at 15 and 30 minutes postload was higher in MCT-fed Brd4 (+/-) mice than that in those fed an LCT diet. CONCLUSIONS Aged Brd4 (+/-) mice showed lower mRNA expression of Gcg and Pyy genes, and active GLP-1 protein secretion in the blood, which were as restored and enhanced with MCT feeding.
Collapse
Affiliation(s)
- Musashi Kawamura
- Department of Integrated Applied Life Science, Integrated Graduate School of Medicine, Engineering, and Agricultural Sciences, University of Yamanashi, Kofu, Yamanashi, Japan; Department of Local Produce and Food Sciences, Laboratory of Food and Nutritional Sciences, Faculty of Life and Environmental Sciences, University of Yamanashi, Kofu, Yamanashi, Japan
| | - Natsuyo Hariya
- Department of Nutrition and Dietetics, Faculty of Family and Consumer Sciences, Kamakura Women's University, Kamakura, Kanagawa, Japan
| | - Shiori Ishiyama
- Department of Integrated Applied Life Science, Integrated Graduate School of Medicine, Engineering, and Agricultural Sciences, University of Yamanashi, Kofu, Yamanashi, Japan; Department of Local Produce and Food Sciences, Laboratory of Food and Nutritional Sciences, Faculty of Life and Environmental Sciences, University of Yamanashi, Kofu, Yamanashi, Japan
| | - Yuji Tanaka
- New Industry Creation Hatchery Center, Tohoku University, Sendai, Japan
| | - Keiko Ozato
- Laboratory of Molecular Growth Regulation, NICHD, NIH, Bethesda, Maryland, USA
| | - Kazuki Mochizuki
- Department of Integrated Applied Life Science, Integrated Graduate School of Medicine, Engineering, and Agricultural Sciences, University of Yamanashi, Kofu, Yamanashi, Japan; Department of Local Produce and Food Sciences, Laboratory of Food and Nutritional Sciences, Faculty of Life and Environmental Sciences, University of Yamanashi, Kofu, Yamanashi, Japan.
| |
Collapse
|
17
|
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] [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
|
18
|
Zhou W, He Y, Lv JM, Wang R, He H, Wu M, Zhang R, He J. Preparation technologies, structural characteristics and biological activities of polysaccharides from bee pollen: A review. Int J Biol Macromol 2025; 306:141545. [PMID: 40020838 DOI: 10.1016/j.ijbiomac.2025.141545] [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: 12/12/2024] [Revised: 02/13/2025] [Accepted: 02/25/2025] [Indexed: 03/03/2025]
Abstract
Bee pollen, a natural honeybee product, is hailed as a treasure trove of human nutrition. Among the nourishing substances of bee pollen, the constituents with a low molecular weight (such as phenolic acids and flavonoid glycosides) have been extensively studied in the past decades, whereas the polysaccharides with a relatively high molecular weight have received much less attention. To deepen our understanding of bee pollen polysaccharides, this review summarizes the published findings related to their preparation technologies, structural characteristics and biological activities. Among the preparation technologies, ultrasonic-assisted extraction is currently the most effective technology for the recovery of polysaccharides from bee pollen, because ultrasound can crack the pollen exine into fragments and facilitate the release of polysaccharides present in the pollen intine. The preliminary structures, including the molecular weight and monosaccharide composition, of bee pollen polysaccharides have been widely reported, but their fine structures have not fully elucidated. Moreover, bee pollen polysaccharides have antioxidant, immunomodulatory, and antitumor activities, exhibiting potential application in functional foods. Furthermore, bee pollen polysaccharides can modulate the composition of gut microbiota and promote the production of short-chain fatty acids. It is expected that this review can provide inspiration for the development and utilization of bee pollen polysaccharides.
Collapse
Affiliation(s)
- Wangting Zhou
- National R & D Center for Se-rich Agricultural Products Processing, School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, PR China
| | - Yuzhen He
- National R & D Center for Se-rich Agricultural Products Processing, School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, PR China
| | - Ji-Min Lv
- The State Agriculture Ministry Laboratory of Horticultural Plant Growth, Development and Quality Improvement, Zhejiang University, Hangzhou 310058, PR China; Xianghu Laboratory, Hangzhou 311231, PR China
| | - Runqi Wang
- National R & D Center for Se-rich Agricultural Products Processing, School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, PR China
| | - Huaiye He
- National R & D Center for Se-rich Agricultural Products Processing, School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, PR China
| | - Muci Wu
- National R & D Center for Se-rich Agricultural Products Processing, School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, PR China; Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, Wuhan 430023, PR China
| | - Rui Zhang
- National R & D Center for Se-rich Agricultural Products Processing, School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, PR China; Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, Wuhan 430023, PR China.
| | - Jingren He
- National R & D Center for Se-rich Agricultural Products Processing, School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, PR China; Hubei Key Laboratory for Processing and Transformation of Agricultural Products, Wuhan Polytechnic University, Wuhan 430023, PR China.
| |
Collapse
|
19
|
Jameson KG, Kazmi SA, Ohara TE, Son C, Yu KB, Mazdeyasnan D, Leshan E, Vuong HE, Paramo J, Lopez-Romero A, Yang L, Schweizer FE, Hsiao EY. Select microbial metabolites in the small intestinal lumen regulates vagal activity via receptor-mediated signaling. iScience 2025; 28:111699. [PMID: 39877906 PMCID: PMC11772968 DOI: 10.1016/j.isci.2024.111699] [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: 07/12/2024] [Revised: 09/22/2024] [Accepted: 12/24/2024] [Indexed: 01/31/2025] Open
Abstract
The vagus nerve is proposed to enable communication between the gut microbiome and the brain, but activity-based evidence is lacking. We find that mice reared germ-free exhibit decreased vagal tone relative to colonized controls, which is reversed via microbiota restoration. Perfusing antibiotics into the small intestines of conventional mice, but not germ-free mice, acutely decreases vagal activity which is restored upon re-perfusion with intestinal filtrates from conventional, but not germ-free, mice. Microbiome-dependent short-chain fatty acids, bile acids, and 3-indoxyl sulfate indirectly stimulate vagal activity in a receptor-dependent manner. Serial perfusion of each metabolite class activates both shared and distinct neuronal subsets with varied response kinetics. Metabolite-induced and receptor-dependent increases in vagal activity correspond with the activation of brainstem neurons. Results from this study reveal that the gut microbiome regulates select metabolites in the intestinal lumen that differentially activate vagal afferent neurons, thereby enabling the microbial modulation of chemosensory signals for gut-brain communication.
Collapse
Affiliation(s)
- Kelly G. Jameson
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Sabeen A. Kazmi
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Takahiro E. Ohara
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Celine Son
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Kristie B. Yu
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Donya Mazdeyasnan
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Emma Leshan
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Helen E. Vuong
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Jorge Paramo
- UCLA Goodman-Luskin Microbiome Center, Department of Medicine, Division of Digestive Diseases, David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Arlene Lopez-Romero
- UCLA Goodman-Luskin Microbiome Center, Department of Medicine, Division of Digestive Diseases, David Geffen School of Medicine, Los Angeles, CA 90095, USA
| | - Long Yang
- Department of Neurobiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Felix E. Schweizer
- Department of Neurobiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
| | - Elaine Y. Hsiao
- Department of Integrative Biology & Physiology, University of California, Los Angeles, Los Angeles, CA 90095, USA
- UCLA Goodman-Luskin Microbiome Center, Department of Medicine, Division of Digestive Diseases, David Geffen School of Medicine, Los Angeles, CA 90095, USA
| |
Collapse
|
20
|
Peña-Durán E, García-Galindo JJ, López-Murillo LD, Huerta-Huerta A, Balleza-Alejandri LR, Beltrán-Ramírez A, Anaya-Ambriz EJ, Suárez-Rico DO. Microbiota and Inflammatory Markers: A Review of Their Interplay, Clinical Implications, and Metabolic Disorders. Int J Mol Sci 2025; 26:1773. [PMID: 40004236 PMCID: PMC11854938 DOI: 10.3390/ijms26041773] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2025] [Revised: 02/12/2025] [Accepted: 02/14/2025] [Indexed: 02/27/2025] Open
Abstract
The human microbiota, a complex ecosystem of microorganisms, plays a pivotal role in regulating host immunity and metabolism. This review investigates the interplay between microbiota and inflammatory markers, emphasizing their impact on metabolic and autoimmune disorders. Key inflammatory biomarkers, such as C-reactive protein (CRP), interleukin-6 (IL-6), lipopolysaccharides (LPS), zonulin (ZO-1), and netrin-1 (Ntn1), are discussed in the context of intestinal barrier integrity and chronic inflammation. Dysbiosis, characterized by alterations in microbial composition and function, directly modulates the levels and activity of these biomarkers, exacerbating inflammatory responses and compromising epithelial barriers. The disruption of microbiota is further correlated with increased intestinal permeability and chronic inflammation, serving as a precursor to conditions like type 2 diabetes (T2D), obesity, and non-alcoholic fatty liver disease. Additionally, this review examines therapeutic strategies, including probiotics and prebiotics, designed to restore microbial balance, mitigate inflammation, and enhance metabolic homeostasis. Emerging evidence positions microbiota-targeted interventions as critical components in the advancement of precision medicine, offering promising avenues for diagnosing and treating inflammatory and metabolic disorders.
Collapse
Affiliation(s)
- Emiliano Peña-Durán
- Licenciatura en Médico Cirujano y Partero, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Jesús Jonathan García-Galindo
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Calle Sierra Mojada 950, Independencia Oriente, Guadalajara 44340, Mexico
- Departamento Académico Aparatos y Sistemas II, Decanato de Ciencias de la Salud, Universidad Autónoma de Guadalajara, Zapopan 44670, Mexico
| | - Luis Daniel López-Murillo
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Calle Sierra Mojada 950, Independencia Oriente, Guadalajara 44340, Mexico
- Departamento Académico Aparatos y Sistemas I, Decanato de Ciencias de la Salud, Universidad Autónoma de Guadalajara, Zapopan 44670, Mexico
| | - Alfredo Huerta-Huerta
- Hospital Medica de la Ciudad, Santa Catalina, Calle. Pablo Valdez 719, La Perla, Guadalajara 44360, Mexico
| | - Luis Ricardo Balleza-Alejandri
- Doctorado en Farmacología, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Mexico
| | - Alberto Beltrán-Ramírez
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Calle Sierra Mojada 950, Independencia Oriente, Guadalajara 44340, Mexico
- Departamento Académico Aparatos y Sistemas I, Decanato de Ciencias de la Salud, Universidad Autónoma de Guadalajara, Zapopan 44670, Mexico
| | - Elsa Janneth Anaya-Ambriz
- Departamento de Ciencias de la Salud, Centro Universitario de los Valles, Universidad de Guadalajara, Ameca 46708, Mexico
| | - Daniel Osmar Suárez-Rico
- Departamento de Fisiología, Centro Universitario de Ciencias de la Salud (CUCS), Universidad de Guadalajara, Calle Sierra Mojada 950, Independencia Oriente, Guadalajara 44340, Mexico
- Departamento Académico Aparatos y Sistemas II, Decanato de Ciencias de la Salud, Universidad Autónoma de Guadalajara, Zapopan 44670, Mexico
- Departamento de Farmacobiología, Centro Universitario de Ciencias Exactas e Ingenierías (CUCEI), Universidad de Guadalajara, Guadalajara 44430, Mexico
| |
Collapse
|
21
|
Movahednasab M, Dianat-Moghadam H, Khodadad S, Nedaeinia R, Safabakhsh S, Ferns G, Salehi R. GLP-1-based therapies for type 2 diabetes: from single, dual and triple agonists to endogenous GLP-1 production and L-cell differentiation. Diabetol Metab Syndr 2025; 17:60. [PMID: 39962520 PMCID: PMC11834518 DOI: 10.1186/s13098-025-01623-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Accepted: 02/02/2025] [Indexed: 02/20/2025] Open
Abstract
Glucagon-like peptide-1 (GLP-1) is an incretin peptide hormone mainly secreted by enteroendocrine intestinal L-cells. GLP-1 is also secreted by α-cells of the pancreas and the central nervous system (CNS). GLP-1 secretion is stimulated by nutrient intake and exerts its effects on glucose homeostasis by stimulating insulin secretion, gastric emptying confiding the food intake, and β-cell proliferation. The insulinotropic effects of GLP-1, and the reduction of its effects in type 2 diabetes mellitus (T2DM), have made GLP-1 an attractive option for the treatment of T2DM. Furthermore, GLP-1-based medications such as GLP-1 receptor agonists and dipeptidyl peptidase-4 inhibitors, have been shown to improve diabetes control in preclinical and clinical trials with human subjects. Importantly, increasing the endogenous production of GLP-1 by different mechanisms or by increasing the number of intestinal L-cells that tend to produce this hormone may be another effective therapeutic approach to managing T2DM. Herein, we briefly describe therapeutic agents/compounds that enhance GLP-1 function. Then, we will discuss the approaches that can increase the endogenous production of GLP-1 through various stimuli. Finally, we introduce the potential of L-cell differentiation as an attractive future therapeutic approach to increase GLP-1 production as an attractive therapeutic alternative for T2DM.
Collapse
Affiliation(s)
- Maedeh Movahednasab
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Hassan Dianat-Moghadam
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Sana Khodadad
- Department of Genetics and Molecular Biology, School of Medicine, Shahrekord University of Medical Sciences, Shahrekord, Iran
| | - Reza Nedaeinia
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Saeid Safabakhsh
- Micronesian Institute for Disease Prevention and Research, 736 Route 4, Suite 103, Sinajana, GU, 96910, USA
| | - Gordon Ferns
- Division of Medical Education, Brighton & Sussex Medical School, Falmer, Brighton, Sussex, BN1 9PH, UK
| | - Rasoul Salehi
- Department of Genetics and Molecular Biology, School of Medicine, Isfahan University of Medical Sciences, Isfahan, Iran.
- Pediatric Inherited Diseases Research Center, Research Institute for Primordial Prevention of Non-Communicable Disease, Isfahan University of Medical Sciences, Isfahan, Iran.
| |
Collapse
|
22
|
Cho YY, Kim S, Kim P, Jo MJ, Park SE, Choi Y, Jung SM, Kang HJ. G-Protein-Coupled Receptor (GPCR) Signaling and Pharmacology in Metabolism: Physiology, Mechanisms, and Therapeutic Potential. Biomolecules 2025; 15:291. [PMID: 40001594 PMCID: PMC11852853 DOI: 10.3390/biom15020291] [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: 01/19/2025] [Revised: 02/10/2025] [Accepted: 02/11/2025] [Indexed: 02/27/2025] Open
Abstract
G-protein coupled receptors (GPCRs), the largest family of integral membrane proteins, enable cells to sense and appropriately respond to the environment through mediating extracellular signaling to intercellular messenger molecules. GPCRs' pairing with a diverse array of G protein subunits and related downstream secondary messengers, combined with their ligand versatility-from conventional peptide hormone to numerous bioactive metabolites, allow GPCRs to comprehensively regulate metabolism and physiology. Consequently, GPCRs have garnered significant attention for their therapeutic potential in metabolic diseases. This review focuses on six GPCRs, GPR40, GPR120, GLP-1R, and ß-adrenergic receptors (ADRB1, ADRB2, and ADRB3), with GLP-1R recognized as a prominent regulator of system-level metabolism, while the roles of GPR40, GPR120 and ß-adrenergic receptors in central carbon metabolism and energy homeostasis are increasingly appreciated. Here, we discuss their physiological functions in metabolism, the current pharmacological landscape, and the intricacies of their signaling pathways via G protein and ß-arrestin activation. Additionally, we discuss the limitations of existing GPCR-targeted strategies for treating metabolic diseases and offer insights into future perspectives for advancing GPCR pharmacology.
Collapse
Affiliation(s)
- Yun Yeong Cho
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; (Y.Y.C.); (P.K.); (M.J.J.); (S.-E.P.)
| | - Soyeon Kim
- Department of Biological Sciences, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea; (S.K.); (Y.C.)
| | - Pankyung Kim
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; (Y.Y.C.); (P.K.); (M.J.J.); (S.-E.P.)
| | - Min Jeong Jo
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; (Y.Y.C.); (P.K.); (M.J.J.); (S.-E.P.)
| | - Song-E Park
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; (Y.Y.C.); (P.K.); (M.J.J.); (S.-E.P.)
| | - Yiju Choi
- Department of Biological Sciences, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea; (S.K.); (Y.C.)
| | - Su Myung Jung
- Department of Biological Sciences, Sungkyunkwan University (SKKU), Suwon 16419, Republic of Korea; (S.K.); (Y.C.)
| | - Hye Jin Kang
- Department of Biotechnology, College of Life Science and Biotechnology, Yonsei University, Seoul 03722, Republic of Korea; (Y.Y.C.); (P.K.); (M.J.J.); (S.-E.P.)
| |
Collapse
|
23
|
Rondanelli M, Borromeo S, Cavioni A, Gasparri C, Gattone I, Genovese E, Lazzarotti A, Minonne L, Moroni A, Patelli Z, Razza C, Sivieri C, Valentini EM, Barrile GC. Therapeutic Strategies to Modulate Gut Microbial Health: Approaches for Chronic Metabolic Disorder Management. Metabolites 2025; 15:127. [PMID: 39997751 PMCID: PMC11857149 DOI: 10.3390/metabo15020127] [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: 12/02/2024] [Revised: 01/17/2025] [Accepted: 01/28/2025] [Indexed: 02/26/2025] Open
Abstract
Numerous recent studies have suggested that the composition of the intestinal microbiota can trigger metabolic disorders, such as diabetes, prediabetes, obesity, metabolic syndrome, sarcopenia, dyslipidemia, hyperhomocysteinemia, and non-alcoholic fatty liver disease. Since then, considerable effort has been made to understand the link between the composition of intestinal microbiota and metabolic disorders, as well as the role of probiotics in the modulation of the intestinal microbiota. The aim of this review was to summarize the reviews and individual articles on the state of the art regarding ideal therapy with probiotics and prebiotics in order to obtain the reversion of dysbiosis (alteration in microbiota) to eubiosis during metabolic diseases, such as diabetes, prediabetes, obesity, hyperhomocysteinemia, dyslipidemia, sarcopenia, and non-alcoholic fatty liver diseases. This review includes 245 eligible studies. In conclusion, a condition of dysbiosis, or in general, alteration of the intestinal microbiota, could be implicated in the development of metabolic disorders through different mechanisms, mainly linked to the release of pro-inflammatory factors. Several studies have already demonstrated the potential of using probiotics and prebiotics in the treatment of this condition, detecting significant improvements in the specific symptoms of metabolic diseases. These findings reinforce the hypothesis that a condition of dysbiosis can lead to a generalized inflammatory picture with negative consequences on different organs and systems. Moreover, this review confirms that the beneficial effects of probiotics on metabolic diseases are promising, but more research is needed to determine the optimal probiotic strains, doses, and administration forms for specific metabolic conditions.
Collapse
Affiliation(s)
- Mariangela Rondanelli
- Department of Public Health, Experimental and Forensic Medicine, University of Pavia, 27100 Pavia, Italy;
| | - Sara Borromeo
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Alessandro Cavioni
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Clara Gasparri
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Ilaria Gattone
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Elisa Genovese
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Alessandro Lazzarotti
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Leonardo Minonne
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Alessia Moroni
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Zaira Patelli
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Claudia Razza
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Claudia Sivieri
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Eugenio Marzio Valentini
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| | - Gaetan Claude Barrile
- Endocrinology and Nutrition Unit, Azienda di Servizi alla Persona “Istituto Santa Margherita”, University of Pavia, 27100 Pavia, Italy; (S.B.); (A.C.); (C.G.); (I.G.); (E.G.); (A.L.); (L.M.); (A.M.); (Z.P.); (C.R.); (C.S.); (E.M.V.)
| |
Collapse
|
24
|
Shallangwa SM, Ross AW, Morgan PJ. Single, but not mixed dietary fibers suppress body weight gain and adiposity in high fat-fed mice. Front Microbiol 2025; 16:1544433. [PMID: 40012787 PMCID: PMC11861375 DOI: 10.3389/fmicb.2025.1544433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2024] [Accepted: 01/13/2025] [Indexed: 02/28/2025] Open
Abstract
Dietary fiber can suppress excess adipose tissue and weight gain in rodents and humans when fed high fat diets. The gut microbiome is thought to have a key role, although exactly how remains unclear. In a tightly controlled murine study, we explored how different types of dietary fiber and doses affect the gut microbiota and gut epithelial gene expression. We show that 10% pectin and 10% FOS suppress high fat diet (HFD)-induced weight gain, effects not seen at 2% doses. Furthermore, 2 and 10% mixtures of dietary fiber were also without effect. Each fiber treatment stimulated a distinct gut microbiota profile at the family and operational taxonomic unit (OTU) level. Mechanistically it is likely that the single 10% fiber dose shifted selected bacteria above some threshold abundance, required to suppress body weight, which was not achieved by the 10% Mix, composed of 4 fibers each at 2.5%. Plasma levels of the gut hormone PYY were elevated by 10% pectin and FOS, but not 10% mixed fibers, and similarly RNA seq revealed some distinct effects of the 10% single fibers on gut epithelial gene expression. These data show how the ability of dietary fiber to suppress HFD-induced weight gain is dependent upon both fiber type and dose. It also shows that the microbial response to dietary fiber is distinct and that there is not a single microbial response associated with the inhibition of adiposity and weight gain. PYY seems key to the latter response, although the role of other factors such as Reg3γ and CCK needs to be explored.
Collapse
Affiliation(s)
| | | | - Peter J. Morgan
- Rowett Institute, University of Aberdeen, Aberdeen, United Kingdom
| |
Collapse
|
25
|
Li C, Yao J, Yang C, Yu S, Yang Z, Wang L, Li S, He N. Gut microbiota-derived short chain fatty acids act as mediators of the gut-liver-brain axis. Metab Brain Dis 2025; 40:122. [PMID: 39921774 DOI: 10.1007/s11011-025-01554-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2024] [Accepted: 02/03/2025] [Indexed: 02/10/2025]
Abstract
The gut microbiota plays a crucial role in the communication between the gut, liver, and brain through the production of short chain fatty acids (SCFAs). SCFAs serve as key mediators in the Gut-Liver-Brain Axis, influencing various physiological processes and contributing to overall health. SCFAs are produced by bacterial fermentation of dietary fiber in the gut, and they exert systemic effects by signaling through various pathways. In the Gut-Liver axis, SCFAs regulate liver metabolism through peroxisome proliferator-activated receptor-γ (PPAR-γ), AMP-activated protein kinase (AMPK) and other pathways, promotes fat oxidation, modulate inflammation through mTOR pathway, and impact metabolic health. In the Gut-Brain axis, SCFAs influence brain function, behavior, and may have implications for neurological disorders, in which G-protein coupled receptors (GPCRs) play an essential role, along with other pathways such as hypothalamic-pituitary-adrenal (HPA) pathway. Understanding the mechanisms by which SCFAs mediate communication between the gut, liver, and brain is crucial for elucidating the complex interplay of the Gut-Liver-Brain Axis. This review aims to provide insight into the role of gut microbiota-derived SCFAs as mediators of the Gut-Liver-Brain Axis and their potential therapeutic implications. Further research in this area will be instrumental in developing novel strategies to target the Gut-Liver-Brain Axis for the prevention and treatment of various health conditions.
Collapse
Affiliation(s)
- Cunyin Li
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, 266071, China
- Department of Obstetrics and Gynecology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, 266000, China
| | - Jingtong Yao
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, 266071, China
| | - Chang Yang
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, 266071, China
| | - Shengnan Yu
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, 266071, China
- Affiliated Hospital of Inner Mongolia University for Nationalities, TongLiao, 028005, China
| | - Zizhen Yang
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, 266071, China
| | - Lijing Wang
- Department of Obstetrics and Gynecology, The Affiliated Qingdao Municipal Hospital of Qingdao University, Qingdao, 266000, China.
- Department of Obstetrics, Qingdao Municipal Hospital, Qingdao, 266000, China.
| | - Shangyong Li
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, 266071, China.
- Department of Ultrasound, the Affiliated Hospital of Qingdao University, Qingdao, 266003, China.
| | - Ningning He
- School of Basic Medicine, Qingdao Medical College, Qingdao University, Qingdao, 266071, China.
| |
Collapse
|
26
|
Yu W, Sun S, Yan Y, Zhou H, Liu Z, Fu Q. The role of short-chain fatty acid in metabolic syndrome and its complications: focusing on immunity and inflammation. Front Immunol 2025; 16:1519925. [PMID: 39991152 PMCID: PMC11842938 DOI: 10.3389/fimmu.2025.1519925] [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: 10/30/2024] [Accepted: 01/09/2025] [Indexed: 02/25/2025] Open
Abstract
Metabolic syndrome (Mets) is an important contributor to morbidity and mortality in cardiovascular, liver, neurological, and reproductive diseases. Short-chain fatty acid (SCFA), an organismal energy donor, has recently been demonstrated in an increasing number of studies to be an important molecule in ameliorating immuno-inflammation, an important causative factor of Mets, and to improve lipid distribution, blood glucose, and body weight levels in animal models of Mets. This study reviews recent research advances on SCFA in Mets from an immune-inflammatory perspective, including complications dominated by chronic inflammation, as well as the fact that these findings also contribute to the understanding of the specific mechanisms by which gut flora metabolites contribute to metabolic processes in humans. This review proposes an emerging role for SCFA in the inflammatory Mets, followed by the identification of major ambiguities to further understand the anti-inflammatory potential of this substance in Mets. In addition, this study proposes novel strategies to modulate SCFA for the treatment of Mets that may help to mitigate the prognosis of Mets and its complications.
Collapse
Affiliation(s)
- Wenqian Yu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Siyuan Sun
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Yutong Yan
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Hong Zhou
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Ziyi Liu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
- First Clinical Medical College, Beijing University of Chinese Medicine, Beijing, China
| | - Qiang Fu
- Dongzhimen Hospital, Beijing University of Chinese Medicine, Beijing, China
| |
Collapse
|
27
|
Gürbüzer N, Tozoğlu EÖ, Ozkaya AL, Mercantepe F. Serum Asprosin and Peptide Tyrosine Tyrosine (PYY) Levels in Bipolar Disorder. J Clin Med 2025; 14:1012. [PMID: 39941681 PMCID: PMC11818612 DOI: 10.3390/jcm14031012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/29/2025] [Accepted: 02/03/2025] [Indexed: 02/16/2025] Open
Abstract
Objective: In our study, we aimed to investigate the differences in metabolic parameters, serum asprosin and peptide tyrosine tyrosine (PYY) levels in a bipolar disorder manic (BD-M) group, a euthymic group and in healthy controls; we also aimed to evaluate the relationship of asprosin and PYY levels with metabolic parameters and psychopathology in patients. Methods: The study included 54 manic patients, 40 euthymic patients and 39 healthy controls. The sociodemographic characteristics of the participants were recorded, and biochemical parameters and asprosin and PYY levels were measured. The Young Mania Rating Scale (YMRS) and the Hamilton Depression Rating Scale (HAM-D) were completed. Results: Body mass index (BMI) showed significant differences between the three groups (p < 0.001); the lowest was found in the control group and the highest in the euthymic group. Triglyceride levels were significantly higher in the euthymic group compared with the BD-M group and controls (p = 0.003). Glucose levels were significantly higher in the BD-M group compared with euthymic (pmanic-euthymic = 0.008) and controls (pmanic-control < 0.001). Asprosin (pmanic-control < 0.001, peuthymic-control = 0.046, pmanic-euthymic = 0.015) and PYY (pmanic-control < 0.001, peuthymic-control = 0.037, pmanic-euthymic = 0.002) levels were significantly different between the three groups, with the lowest levels in the BD-M group and the highest levels in the control group. The eta squared = 0.18 for asprosin and 0.21 for PYY. In the BD-M group, a moderate negative correlation was found between YMRS and asprosin (r = -0.345; p = 0.011) and PYY (r = -0.376; p = 0.005) levels. ROC analysis results showed that asprosin and PYY could be used to predict the manic period in BD-I (AUCasprosin:0.775; AUCPYY:0.760). After adjusting for asprosin as a covariate using ANCOVA, the difference in PYY between groups remained significant (manic-euthymic groups, p = 0.040; manic-control groups, p = 0.013). Conclusions: The study results revealed that asprosin and PYY levels were low, and metabolic parameters were impaired in the patients. Low asprosin and PYY levels may be indicators of impaired energy homeostasis in BD-I. PYY may be a state marker for manic episodes.
Collapse
Affiliation(s)
- Nilifer Gürbüzer
- Department of Psychiatry, Erzurum Faculty of Medicine, University of Health Sciences, Erzurum 25240, Türkiye; (N.G.); (E.Ö.T.)
| | - Elif Özcan Tozoğlu
- Department of Psychiatry, Erzurum Faculty of Medicine, University of Health Sciences, Erzurum 25240, Türkiye; (N.G.); (E.Ö.T.)
| | | | - Filiz Mercantepe
- Department of Endocrinology and Metabolism, Faculty of Medicine, Recep Tayyip Erdogan University, Rize 53100, Türkiye
| |
Collapse
|
28
|
Lee J, Jang HR, Lee D, Lee Y, Lee HY. Gut Bacteria-Derived Tryptamine Ameliorates Diet-Induced Obesity and Insulin Resistance in Mice. Int J Mol Sci 2025; 26:1327. [PMID: 39941095 PMCID: PMC11818187 DOI: 10.3390/ijms26031327] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2024] [Revised: 02/01/2025] [Accepted: 02/03/2025] [Indexed: 02/16/2025] Open
Abstract
Tryptophan is an essential amino acid that is metabolized in the intestine by gut bacteria into indole derivatives, including tryptamine. However, little is known about which bacterial tryptophan metabolites directly influence obesity. In this study, we identified tryptamine as a bacterial metabolite that significantly reduced fat mass following the intraperitoneal injection of five bacterial tryptophan end-products in a diet-induced obese mouse model. Interestingly, tryptamine, a serotonin analog, inhibited both lipogenesis and lipolysis in adipose tissue, which was further confirmed in a 3T3-L1 adipocyte cell culture study. Moreover, oral tryptamine supplementation markedly reduced fat mass and improved insulin sensitivity in a long-term, high-fat-diet, pair-feeding model. These studies demonstrate the therapeutic potential of tryptamine, a bacterial tryptophan metabolite, in ameliorating obesity and insulin resistance by directly regulating lipogenesis and lipolysis in white adipose tissue.
Collapse
Affiliation(s)
- Jongjun Lee
- Laboratory of Mitochondria and Metabolic Diseases, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea; (J.L.); (H.-R.J.)
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences & Technology, Gachon University, Incheon 21999, Republic of Korea
| | - Hye-Rim Jang
- Laboratory of Mitochondria and Metabolic Diseases, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea; (J.L.); (H.-R.J.)
| | - Dongjin Lee
- Laboratory of Mitochondria and Metabolic Diseases, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea; (J.L.); (H.-R.J.)
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences & Technology, Gachon University, Incheon 21999, Republic of Korea
| | - Yeonmi Lee
- Laboratory of Mitochondria and Metabolic Diseases, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea; (J.L.); (H.-R.J.)
| | - Hui-Young Lee
- Laboratory of Mitochondria and Metabolic Diseases, Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea; (J.L.); (H.-R.J.)
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences & Technology, Gachon University, Incheon 21999, Republic of Korea
- Department of Molecular Medicine, Gachon University College of Medicine, Incheon 21936, Republic of Korea
| |
Collapse
|
29
|
Abavisani M, Faraji S, Ebadpour N, Karav S, Sahebkar A. Beyond the Hayflick limit: How microbes influence cellular aging. Ageing Res Rev 2025; 104:102657. [PMID: 39788433 DOI: 10.1016/j.arr.2025.102657] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 12/23/2024] [Accepted: 01/05/2025] [Indexed: 01/12/2025]
Abstract
Cellular senescence, a complex biological process resulting in permanent cell-cycle arrest, is central to aging and age-related diseases. A key concept in understanding cellular senescence is the Hayflick Limit, which refers to the limited capacity of normal human cells to divide, after which they become senescent. Senescent cells (SC) accumulate with age, releasing pro-inflammatory and tissue-remodeling factors collectively known as the senescence-associated secretory phenotype (SASP). The causes of senescence are multifaceted, including telomere attrition, oxidative stress, and genotoxic damage, and they extend to influences from microbial sources. Research increasingly emphasizes the role of the microbiome, especially gut microbiota (GM), in modulating host senescence processes. Beneficial microbial metabolites, such as short-chain fatty acids (SCFAs), support host health by maintaining antioxidant defenses and reducing inflammation, potentially mitigating senescence onset. Conversely, pathogenic bacteria like Pseudomonas aeruginosa and Helicobacter pylori introduce factors that damage host DNA or increase ROS, accelerating senescence via pathways such as NF-κB and p53-p21. This review explores the impact of bacterial factors on cellular senescence, highlighting the role of specific bacterial toxins in promoting senescence. Additionally, it discusses how dysbiosis and the loss of beneficial microbial species further contribute to age-related cellular deterioration. Modulating the gut microbiome to delay cellular senescence opens a path toward targeted anti-aging strategies. This work underscores the need for deeper investigation into microbial influence on aging, supporting innovative interventions to manage and potentially reverse cellular senescence.
Collapse
Affiliation(s)
- Mohammad Abavisani
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Saba Faraji
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negar Ebadpour
- Student research committee, Mashhad University of Medical Sciences, Mashhad, Iran; Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Canakkale Onsekiz Mart University, Canakkale 17100, Turkey
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Iran.
| |
Collapse
|
30
|
Linder L. Naturopathic Management to Taper Off Glucagon-Like Peptide-1 Receptor Agonist Therapy in Type 2 Diabetes: A Case Report. Integr Med (Encinitas) 2025; 24:26-30. [PMID: 39896831 PMCID: PMC11778319] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
This case report presents a 46-year-old woman with type 2 diabetes mellitus (T2DM) who sought to discontinue glucagon-like peptide-1 (GLP-1) receptor agonists (RAs) therapy while maintaining her weight and metabolic health. Her desire to reduce medication dependency, combined with her apprehension about potential metabolic regression following discontinuation of the drug, highlights a challenge often faced by patients managing chronic conditions, and the value of naturopathic adjunctive therapies to support these individuals in their wellness goals. This report details a comprehensive intervention strategy combining probiotics, nutrient supplementation, and lifestyle modifications to address her concerns. Over an 8-week period post-GLP-1 RAs therapy discontinuation, she maintained weight loss, improved glycemic control and lipid profile, demonstrated improvements in micronutrient levels, and experienced improvements in her quality of life. This case contributes to the growing body of evidence suggesting that targeted naturopathic interventions can play a supportive role in managing T2DM and mitigating the reliance on pharmacotherapy without compromising health outcomes.
Collapse
Affiliation(s)
- Leah Linder
- Corresponding author: Leah Linder, ND E-mail:
| |
Collapse
|
31
|
Guo Q, Wang X, Ke J, Hou X, Shen G, Li S, Chen H, Cui Q, Yu J, Luo Q, Liu H, Chen A, Liu Y, Zhang Z. Chayote pectin regulates blood glucose through the gut-liver axis: Gut microbes/SCFAs/FoxO1 signaling pathways. Food Res Int 2025; 202:115706. [PMID: 39967162 DOI: 10.1016/j.foodres.2025.115706] [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: 10/16/2024] [Revised: 12/31/2024] [Accepted: 01/05/2025] [Indexed: 02/20/2025]
Abstract
Despite significant evidence on the anti-diabetic effect of chayote fruit and phenolic compounds, research on the mechanism of chayote (Sechium edule) pectin (CP) regulating blood glucose in type 2 diabetes mellitus (T2DM) is scarce. Therefore, this study aims to explore the potential mechanisms by which CP modulates blood glucose levels through an 8-week administration in db/db mice. The results showed that the CP treatment in db/db mice resulted in an elevation in glucagon-like peptide (GLP-1) secretion, an increase in hepatic glycogen storage, and a decrease in homeostasis model assessment-insulin resistance (HOMA-IR). Western blotting results showed that CP intervention significantly upregulated the expression of phosphatidylinositol 3 kinase (PI3K), phosphorylated protein kinase B (P-AKT) and downregulated the expression of fork-head transcription factor O1(FoxO1), glucose-6-phosphatase (G6Pase) and phosphoenolpyruvate carboxykinase (PEPCK). Moreover, CP effectively upregulated the protein expression of hepatic G protein-coupled receptor 43 (GPR43) and phosphorylated adenosine 5'-monophosphate (AMP)-activated protein kinase (P-AMPK). Furthermore, CP rearranged the gut microbiota structure by increasing beneficial bacteria (unclassified_Ruminococcaceae, Muribaculaceae, Alloprevotella, Rikenella, and Parabacteroides) and reducing the Firmicutes/Bacteroidetes ratio. Additionally, CP improved the gut barrier by increasing the number and area of goblet cells and significantly upregulating the expression of Claudin-1 and Mucin-2. Overall, these findings suggest that CP regulated blood glucose by activating the gut-liver axis signaling pathway: gut microbiota/ SCFAs/ GLP-1, PI3K/AKT/FoxO1, and GPR43/AMPK/FoxO1. This study provides a scientific basis for the development and application of pectin-based functional foods.
Collapse
Affiliation(s)
- Qing Guo
- College of Food Science, Sichuan Agricultural University, Ya'an, Sichuan 625014, China.
| | - Xiaoxue Wang
- College of Food Science, Sichuan Agricultural University, Ya'an, Sichuan 625014, China.
| | - Jingxuan Ke
- Zhang Zhongjing School of Chinese Medicine, Nanyang Institute of Technology, Nanyang 473004, China.
| | - Xiaoyan Hou
- College of Food Science, Sichuan Agricultural University, Ya'an, Sichuan 625014, China.
| | - Guanghui Shen
- College of Food Science, Sichuan Agricultural University, Ya'an, Sichuan 625014, China.
| | - Shanshan Li
- College of Food Science, Sichuan Agricultural University, Ya'an, Sichuan 625014, China.
| | - Hong Chen
- College of Food Science, Sichuan Agricultural University, Ya'an, Sichuan 625014, China.
| | - Qiang Cui
- College of Food Science, Sichuan Agricultural University, Ya'an, Sichuan 625014, China.
| | - Jie Yu
- College of Food Science, Sichuan Agricultural University, Ya'an, Sichuan 625014, China.
| | - Qingying Luo
- College of Food Science, Sichuan Agricultural University, Ya'an, Sichuan 625014, China.
| | - Hong Liu
- Library of Sichuan Agricultural University, Ya'an, Sichuan 625014, China.
| | - Anjun Chen
- College of Food Science, Sichuan Agricultural University, Ya'an, Sichuan 625014, China.
| | - Yuntao Liu
- College of Food Science, Sichuan Agricultural University, Ya'an, Sichuan 625014, China.
| | - Zhiqing Zhang
- College of Food Science, Sichuan Agricultural University, Ya'an, Sichuan 625014, China.
| |
Collapse
|
32
|
Iida A, Takahashi E, Kuranuki S, Shimamoto S, Nakamura T, Kitagaki H. Water-Soluble Cellulose Acetate Changes the Intestinal Microbiota in Mice with Non-Alcoholic Steatohepatitis. Nutrients 2025; 17:500. [PMID: 39940357 PMCID: PMC11820315 DOI: 10.3390/nu17030500] [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: 01/05/2025] [Revised: 01/24/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
Objectives: Non-alcoholic fatty liver disease (NAFLD) is a prevalent chronic disorder of the liver and affects many people worldwide. Intestinal bacteria are thought to be involved in the pathological progression of NAFLD; therefore, improving the intestinal microbiota may be important in controlling NAFLD. In this study, we assessed the effects of water-soluble cellulose acetate (WSCA) on the intestinal microbiota in a non-alcoholic steatohepatitis (NASH) mouse model. Methods: NASH model (STAM mice) was created by streptozotocin injection and feeding the mice a high-fat diet. The serum biochemical parameters were analyzed. Intestinal bacterial populations were analyzed using paired-end sequencing of 16S rRNA, 18S rRNA, and internal transcribed spacer gene. Results: Our findings indicated that WSCA administration tends to improve the serum alanine aminotransferase and glucose levels in STAM mice and decreased the alpha diversity and altered the beta diversity of their intestinal microbiota. Additionally, WSCA intake resulted in an increase in the abundance of Coriobacteriaceae_UCG-002 and a decrease in the abundance of Enterobacter. Conclusions: WSCA intake can alter specific microbial compositions to improve blood glucose levels and liver functions and may improve the pathogenesis of NAFLD.
Collapse
Affiliation(s)
- Ayaka Iida
- School of Nutrition and Dietetics, Faculty of Health and Social Services, Kanagawa University of Human Services, 1-10-1 Heisei-cho, Yokosuka 238-8522, Japan; (E.T.); (S.K.)
| | - Ena Takahashi
- School of Nutrition and Dietetics, Faculty of Health and Social Services, Kanagawa University of Human Services, 1-10-1 Heisei-cho, Yokosuka 238-8522, Japan; (E.T.); (S.K.)
| | - Sachi Kuranuki
- School of Nutrition and Dietetics, Faculty of Health and Social Services, Kanagawa University of Human Services, 1-10-1 Heisei-cho, Yokosuka 238-8522, Japan; (E.T.); (S.K.)
| | - Shu Shimamoto
- Daicel Corporation, Konan 2-18-1, Minatoku, Tokyo 108-8230, Japan;
| | - Tsuyoshi Nakamura
- Department of Food and Health Sciences, International College of Arts and Sciences, Fukuoka Women’s University, 1-1-1, Kasumigaoka, Higashi-ku, Fukuoka 813-8529, Japan;
| | - Hiroshi Kitagaki
- Faculty of Agriculture, Saga University, Honjo-cho, 1, Saga 840-8502, Japan;
| |
Collapse
|
33
|
Kacena C. Effects of the Curcuminoid and Non-Curcuminoid Compounds of Turmeric on the Gut Microbiome and Inflammation: Potential Use in the Treatment and Prevention of Disease. Nutr Rev 2025:nuae221. [PMID: 39873671 DOI: 10.1093/nutrit/nuae221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2025] Open
Abstract
The gut microbiome is a complex system that directly interacts with and influences many systems in the body. This delicate balance of microbiota plays an important role in health and disease and is highly influenced by lifestyle factors and the surrounding environment. As further research emerges, understanding the full potential of the gut microbiome and the impact of using nutraceuticals to positively influence its function may open the door to greater therapeutic outcomes in the treatment and prevention of disease. Curcumin, a bioactive compound derived from the turmeric rhizome, has been studied in depth for its influence on human health as a potent anti-inflammatory and antioxidant properties. However, the therapeutic activity of curcumin is limited by its low oral bioavailability. While most available research has primarily focused on the curcuminoid compounds of turmeric, the non-curcuminoid compounds hold promise to offer therapeutic benefits while synergistically enhancing the bioavailability of curcumin and supporting the gut microbiome. This review summarizes current knowledge of the relationship between the gut and the various systems within the body, and how dysbiosis, or disruption in the gut microbial balance, leads to inflammation and increased risk of chronic disease. The review also summarizes recent research that focuses on the bioactivity of both the curcuminoid and non-curcuminoid compounds that comprise the whole turmeric root and their synergistic role in enhancing bioavailability to support a healthy gut microbiome and promising use in the treatment and prevention of disease.
Collapse
|
34
|
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
|
35
|
Abavisani M, Faraji N, Ebadpour N, Kesharwani P, Sahebkar A. Beyond digestion: Exploring how the gut microbiota modulates human social behaviors. Neuroscience 2025; 565:52-62. [PMID: 39615647 DOI: 10.1016/j.neuroscience.2024.11.068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2024] [Revised: 11/23/2024] [Accepted: 11/25/2024] [Indexed: 01/07/2025]
Abstract
For a long time, traditional medicine has acknowledged the gut's impact on general health. Contemporary science substantiates this association through investigations of the gut microbiota, the extensive community of microorganisms inhabiting our gastrointestinal system. These microscopic residents considerably improve digestive processes, nutritional absorption, immunological function, and pathogen defense. Nevertheless, a variety of gastrointestinal and extra-intestinal disorders can result from dysbiosis, an imbalance of the microbial composition of the gut microbiota. A groundbreaking discovery is the gut-brain axis, a complex communication network that links the enteric and central nervous system (CNS). This bidirectional communication allows the brain to influence gut activities and vice versa, impacting mental health and mood disorders like anxiety and depression. The gut microbiota can influence this communication by creating neurotransmitters and short-chain fatty acids, among other biochemical processes. These factors may affect our mental state, our ability to regulate our emotions, and the hypothalamic-pituitary-adrenal (HPA) axis. This study aimed to explore the complex interrelationships between the brain and the gut microbiota. We also conducted a thorough examination of the existing understanding in the area of how microbiota affects social behaviors, including emotions, stress responses, and cognitive functions. We also explored the potential of interventions that focus on the connection between the gut and the brain, such as using probiotics to treat diseases of the CNS. This research opens up new possibilities for addressing mental health and neurological conditions in an innovative manner.
Collapse
Affiliation(s)
- Mohammad Abavisani
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Navid Faraji
- Student Research Committee, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Negar Ebadpour
- Immunology Research Center, Mashhad University of Medical Sciences, Mashhad, Iran
| | - Prashant Kesharwani
- Department of Pharmaceutics, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi 110062, India
| | - Amirhossein Sahebkar
- Center for Global Health Research, Saveetha Medical College and Hospitals, Saveetha Institute of Medical and Technical Sciences, Saveetha University, Chennai, India; Biotechnology Research Center, Pharmaceutical Technology Institute, Mashhad University of Medical Sciences, Mashhad, Islamic Republic of Iran; Applied Biomedical Research Center, Mashhad University of Medical Sciences, Mashhad, Islamic Republic of Iran.
| |
Collapse
|
36
|
Yang J, Li G, Wang S, He M, Dong S, Wang T, Shi B, Rensen PCN, Wang Y. Butyrate Prevents Obesity Accompanied by HDAC9-Mediated Browning of White Adipose Tissue. Biomedicines 2025; 13:260. [PMID: 40002674 PMCID: PMC11852213 DOI: 10.3390/biomedicines13020260] [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: 11/15/2024] [Revised: 01/14/2025] [Accepted: 01/17/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Mounting evidence indicates that the short-chain fatty acid butyrate protects against obesity and associated comorbidities, partially through the induction of adipose tissue thermogenesis. However, the effects of butyrate on white adipose tissue (WAT) browning and its molecular mechanism are still elusive. The objective of this study was to investigate butyrate-induced thermogenesis in white adipose tissue and its underlying mechanism. METHODS We studied the effects of butyrate on diet-induced obesity in the humanized APOE*3-Leiden.CETP transgenic mouse model and explored factors related to white adipose browning. Specifically, mice were challenged with a high-fat diet supplemented with butyrate. Adiposity was measured to assess obesity development. Energy metabolism was detected using an indirect calorimetry system. RNA-seq analysis was conducted to analyze the transcription landscape of WAT and responsible targets. Furthermore, the revealed molecular mechanism was verified in vitro. RESULTS Butyrate alleviated high-fat diet-induced obesity and promoted energy expenditure accompanied by brown adipose tissue activation and WAT browning. Mechanistically, RNA-seq analysis revealed that butyrate downregulated HDAC9 in WAT. Additionally, butyrate decreased HDAC9 while increasing thermogenesis in vitro. Inhibition of HDAC9 with TMP269 promoted thermogenic gene expression, mimicking the effects of butyrate. CONCLUSIONS Butyrate protects against diet-induced obesity accompanied by decreasing the expression of HDAC9 in white adipose tissue and inducing browning. This study reveals a new mechanism whereby butyrate activates adaptive thermogenesis and provides new insights for the development of weight-loss drugs targeting adipose HDAC9.
Collapse
Affiliation(s)
- Jing Yang
- Department of Endocrinology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China; (J.Y.)
- Med-X Institute, Center for Immunological and Metabolic Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Guoli Li
- Med-X Institute, Center for Immunological and Metabolic Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Shan Wang
- Med-X Institute, Center for Immunological and Metabolic Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Mingqian He
- Department of Endocrinology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China; (J.Y.)
- Med-X Institute, Center for Immunological and Metabolic Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Sijing Dong
- Med-X Institute, Center for Immunological and Metabolic Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Ting Wang
- Department of Cardiovascular Medicine, Shaanxi Provincial People’s Hospital, Xi’an 710061, China
| | - Binyin Shi
- Department of Endocrinology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China; (J.Y.)
- Med-X Institute, Center for Immunological and Metabolic Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
| | - Patrick C. N. Rensen
- Department of Endocrinology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China; (J.Y.)
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 Leiden, The Netherlands
| | - Yanan Wang
- Department of Endocrinology, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China; (J.Y.)
- Med-X Institute, Center for Immunological and Metabolic Diseases, First Affiliated Hospital of Xi’an Jiaotong University, Xi’an 710061, China
- Department of Medicine, Division of Endocrinology, and Einthoven Laboratory for Experimental Vascular Medicine, Leiden University Medical Center, P.O. Box 9600, 2300 Leiden, The Netherlands
| |
Collapse
|
37
|
Ahmad F, Ahmed SH, Choucair F, Chouliaras S, Awwad J, Terranegra A. A disturbed communication between hypothalamic-pituitary-ovary axis and gut microbiota in female infertility: is diet to blame? J Transl Med 2025; 23:92. [PMID: 39838491 PMCID: PMC11749209 DOI: 10.1186/s12967-025-06117-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] [Received: 11/28/2024] [Accepted: 01/08/2025] [Indexed: 01/23/2025] Open
Abstract
Female infertility is a multifactorial condition influenced by various genetic, environmental, and lifestyle factors. Recent research has investigated the significant impact of gut microbiome dysbiosis on systemic inflammation, metabolic dysfunction, and hormonal imbalances, which can potentially impair fertility. The gut-brain axis, a bidirectional communication system between the gut and the brain, also plays a significant role in regulating reproductive functions. Emerging evidence suggests that the gut microbiome can influence brain functions and behavior, further emphasizing the importance of the microbiota-gut-brain axis in reproduction. Given their role as a major modulator of the gut microbiome, diet and dietary factors, including dietary patterns and nutrient intake, have been implicated in the development and management of female infertility. Hence, this review aims to highlight the impact of dietary patterns, such as the Western diet (WD) and Mediterranean diet (MD), and to decipher their modulatory action on the microbiota-gut-brain axis in infertile women. By contrasting the detrimental effects of WD with the therapeutic potential of MD, we emphasize the pivotal role of a balanced diet rich in nutrients in promoting a healthy gut microbiome. These insights underscore the potential of targeted dietary interventions and lifestyle modifications as promising strategies to enhance reproductive outcomes in subfertile women.
Collapse
Affiliation(s)
- Fatima Ahmad
- Translational Medicine Department, Sidra Medicine, Doha, Qatar
- College of Health and Life Sciences, Hamad bin Khalifa University, Doha, Qatar
| | - Salma H Ahmed
- Translational Medicine Department, Sidra Medicine, Doha, Qatar
| | - Fadi Choucair
- Reproductive Medicine Unit, Sidra Medicine, Doha, Qatar
| | - Spyridon Chouliaras
- Reproductive Medicine Unit, Sidra Medicine, Doha, Qatar
- Weill Cornell Medicine, Ar-Rayyan, Qatar
| | - Johnny Awwad
- Reproductive Medicine Unit, Sidra Medicine, Doha, Qatar
- Vincent Memorial Obstetrics and Gynecology Service, Massachusetts General Hospital, Boston, MA, USA
| | - Annalisa Terranegra
- Translational Medicine Department, Sidra Medicine, Doha, Qatar.
- College of Health and Life Sciences, Hamad bin Khalifa University, Doha, Qatar.
| |
Collapse
|
38
|
Cheng CK, Ye L, Wang Y, Wang YL, Xia Y, Wong SHS, Chen S, Huang Y. Exercised gut microbiota improves vascular and metabolic abnormalities in sedentary diabetic mice through gut‒vascular connection. JOURNAL OF SPORT AND HEALTH SCIENCE 2025; 14:101026. [PMID: 39827989 PMCID: PMC11937666 DOI: 10.1016/j.jshs.2025.101026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 01/22/2025]
Abstract
BACKGROUND Exercise elicits cardiometabolic benefits, reducing the risks of cardiovascular diseases and type 2 diabetes. This study aimed to investigate the vascular and metabolic effects of gut microbiota from exercise-trained donors on sedentary mice with type 2 diabetes and the potential mechanism. METHODS Leptin receptor-deficient diabetic (db/db) and nondiabetic (db/m+) mice underwent running treadmill exercise for 8 weeks, during which fecal microbiota transplantation (FMT) was parallelly performed from exercise-trained to sedentary diabetic (db/db) mice. Endothelial function, glucose homeostasis, physical performance, and vascular signaling of recipient mice were assessed. Vascular and intestinal stresses, including inflammation, oxidative stress, and endoplasmic reticulum (ER) stress, were investigated. RNA sequencing analysis on mouse aortic and intestinal tissues was performed. Gut microbiota profiles of recipient mice were evaluated by metagenomic sequencing. RESULTS Chronic exercise improved vascular and metabolic abnormalities in donor mice. Likewise, FMT from exercised donors retarded body weight gain and slightly improved grip strength and rotarod performance in recipient mice. Exercise-associated FMT enhanced endothelial function in different arteries, suppressed vascular and intestinal stresses, and improved glucose homeostasis in recipient mice, with noted microRNA-181b upregulation in aortas and intestines. Altered gut microbiota profiles and gut-derived factors (e.g., short-chain fatty acids and glucagon-like peptide-1) as well as improved intestinal integrity shall contribute to the cardiometabolic benefits, implying a gut‒vascular connection. CONCLUSION This proof-of-concept study indicates that exercised microbiota confers cardiometabolic benefits on sedentary db/db mice, extending the beneficial mechanism of exercise through gut‒vascular communication. The findings open up new therapeutic opportunities for cardiometabolic diseases and shed light on the development of exercise mimetics by targeting the gut microbiota.
Collapse
Affiliation(s)
- Chak Kwong Cheng
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR 999077, China; School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Lianwei Ye
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong SAR 999077, China
| | - Yu Wang
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR 999077, China; Department of Endocrinology and Metabolism, Shenzhen University General Hospital, Shenzhen 518071, China
| | - Ya-Ling Wang
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong SAR 999077, China
| | - Yin Xia
- School of Biomedical Sciences, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Stephen Heung-Sang Wong
- Department of Sports Science and Physical Education, The Chinese University of Hong Kong, Hong Kong SAR 999077, China
| | - Sheng Chen
- Department of Food Science and Nutrition, The Hong Kong Polytechnic University, Hong Kong SAR 999077, China.
| | - Yu Huang
- Department of Biomedical Sciences, City University of Hong Kong, Hong Kong SAR 999077, China; Tung Biomedical Sciences Centre, City University of Hong Kong, Hong Kong SAR 999077, China.
| |
Collapse
|
39
|
Fernández-Pinteño A, Pilla R, Suchodolski J, Apper E, Torre C, Salas-Mani A, Manteca X. Age-Related Changes in Gut Health and Behavioral Biomarkers in a Beagle Dog Population. Animals (Basel) 2025; 15:234. [PMID: 39858234 PMCID: PMC11758293 DOI: 10.3390/ani15020234] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2024] [Revised: 01/11/2025] [Accepted: 01/14/2025] [Indexed: 01/27/2025] Open
Abstract
The gut and the gut microbiome communicate with the nervous system through the gut-brain axis via neuroimmune and neuroendocrine mechanisms. Despite existing research, studies exploring this link in aging dogs are limited. This study aims to examine multiple blood and fecal biomarkers of intestinal health, along with various behavioral indicators based on saliva, blood, observations, and activity, in different age populations (junior: <2 y.o.; adult: 2-7 y.o.; senior: >7 y.o.) of thirty-seven Beagle dogs. In our study, Bacteroides were significantly higher in senior dogs. The relative abundance of Faecalibacterium and Blautia showed age-related trends, higher in senior and junior dogs, respectively. Fecal short-chain fatty acid concentration, especially acetate, increased with age, while propionate was higher in junior dogs. For the behavioral indicators we considered, blood thyroxine concentration, playing, exploring, and total activity were higher in junior dogs. The differences observed between the biomarkers of gut health and behavior, particularly those significant for the age correlations, emphasize the importance of considering age-related factors when studying the gut microbiome and behavior. However, further research is needed to better understand the mechanisms and specific pathways involved in the relationship between the studied biomarkers and age.
Collapse
Affiliation(s)
- Anna Fernández-Pinteño
- Department of Research and Development, Affinity Petcare, 08902 L’Hospitalet de Llobregat, Spain; (E.A.); (C.T.); (A.S.-M.)
| | - Rachel Pilla
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX 77843, USA; (R.P.); (J.S.)
| | - Jan Suchodolski
- Gastrointestinal Laboratory, Department of Small Animal Clinical Sciences, Texas A&M University, College Station, TX 77843, USA; (R.P.); (J.S.)
| | - Emmanuelle Apper
- Department of Research and Development, Affinity Petcare, 08902 L’Hospitalet de Llobregat, Spain; (E.A.); (C.T.); (A.S.-M.)
| | - Celina Torre
- Department of Research and Development, Affinity Petcare, 08902 L’Hospitalet de Llobregat, Spain; (E.A.); (C.T.); (A.S.-M.)
| | - Anna Salas-Mani
- Department of Research and Development, Affinity Petcare, 08902 L’Hospitalet de Llobregat, Spain; (E.A.); (C.T.); (A.S.-M.)
| | - Xavier Manteca
- School of Veterinary Science, Universitat Autònoma de Barcelona, 08193 Bellaterra, Spain;
| |
Collapse
|
40
|
Saadh MJ, Allela OQB, Kareem RA, Sanghvi G, Menon SV, Sharma P, Tomar BS, Sharma A, Sameer HN, Hamad AK, Athab ZH, Adil M. From Gut to Brain: The Impact of Short-Chain Fatty Acids on Brain Cancer. Neuromolecular Med 2025; 27:10. [PMID: 39821841 DOI: 10.1007/s12017-025-08830-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 01/06/2025] [Indexed: 01/19/2025]
Abstract
The primary source of short-chain fatty acids (SCFAs), now recognized as critical mediators of host health, particularly in the context of neurobiology and cancer development, is the gut microbiota's fermentation of dietary fibers. Recent research highlights the complex influence of SCFAs, such as acetate, propionate, and butyrate, on brain cancer progression. These SCFAs impact immune modulation and the tumor microenvironment, particularly in brain tumors like glioma. They play a critical role in regulating cellular processes, including apoptosis, cell differentiation, and inflammation. Moreover, studies have linked SCFAs to maintaining the integrity of the blood-brain barrier (BBB), suggesting a protective role in preventing tumor infiltration and enhancing anti-tumor immunity. As our understanding of the gut-brain axis deepens, it becomes increasingly important to investigate SCFAs' therapeutic potential in brain cancer management. Looking into how SCFAs affect brain tumor cells and the environment around them could lead to new ways to prevent and treat these diseases, which could lead to better outcomes for people who are dealing with these challenging cancers.
Collapse
Affiliation(s)
- Mohamed J Saadh
- Faculty of Pharmacy, Middle East University, Amman, 11831, Jordan.
| | | | | | - Gaurav Sanghvi
- Department of Microbiology, Faculty of Science, Marwadi University Research Center, Marwadi University, Rajkot, Gujarat, 360003, India
| | - Soumya V Menon
- Department of Chemistry and Biochemistry, School of Sciences, JAIN (Deemed to Be University), Bangalore, Karnataka, India
| | - Pawan Sharma
- Department of Sciences, Vivekananda Global University, Jaipur, Rajasthan, 303012, India
| | - Balvir S Tomar
- Institute of Pediatric Gastroenterology and Hepatology, National Institute of Medical Sciences, NIMS University Rajasthan, Jaipur, India
| | - Aanchal Sharma
- Department of Medical Lab Sciences, Chandigarh Group of Colleges-Jhanjeri, Mohali, Punjab, 140307, India
| | - Hayder Naji Sameer
- Collage of Pharmacy, National University of Science and Technology, Dhi Qar, 64001, Iraq
| | | | - Zainab H Athab
- Department of Pharmacy, Al-Zahrawi University College, Karbala, Iraq
| | - Mohaned Adil
- Pharmacy College, Al-Farahidi University, Baghdad, Iraq
| |
Collapse
|
41
|
Fang H, Rodrigues e-Lacerda R, Barra NG, Kukje Zada D, Robin N, Mehra A, Schertzer JD. Postbiotic Impact on Host Metabolism and Immunity Provides Therapeutic Potential in Metabolic Disease. Endocr Rev 2025; 46:60-79. [PMID: 39235984 PMCID: PMC11720174 DOI: 10.1210/endrev/bnae025] [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: 04/22/2024] [Revised: 07/18/2024] [Accepted: 09/04/2024] [Indexed: 09/07/2024]
Abstract
The gut microbiota influences aspects of metabolic disease, including tissue inflammation, adiposity, blood glucose, insulin, and endocrine control of metabolism. Prebiotics or probiotics are often sought to combat metabolic disease. However, prebiotics lack specificity and can have deleterious bacterial community effects. Probiotics require live bacteria to find a colonization niche sufficient to influence host immunity or metabolism. Postbiotics encompass bacterial-derived components and molecules, which are well-positioned to alter host immunometabolism without relying on colonization efficiency or causing widespread effects on the existing microbiota. Here, we summarize the potential for beneficial and detrimental effects of specific postbiotics related to metabolic disease and the underlying mechanisms of action. Bacterial cell wall components, such as lipopolysaccharides, muropeptides, lipoteichoic acids and flagellin, have context-dependent effects on host metabolism by engaging specific immune responses. Specific types of postbiotics within broad classes of compounds, such as lipopolysaccharides and muropeptides, can have opposing effects on endocrine control of host metabolism, where certain postbiotics are insulin sensitizers and others promote insulin resistance. Bacterial metabolites, such as short-chain fatty acids, bile acids, lactate, glycerol, succinate, ethanolamine, and ethanol, can be substrates for host metabolism. Postbiotics can fuel host metabolic pathways directly or influence endocrine control of metabolism through immunomodulation or mimicking host-derived hormones. The interaction of postbiotics in the host-microbe relationship should be considered during metabolic inflammation and metabolic disease.
Collapse
Affiliation(s)
- Han Fang
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, and Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| | - Rodrigo Rodrigues e-Lacerda
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, and Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| | - Nicole G Barra
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, and Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| | - Dana Kukje Zada
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, and Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| | - Nazli Robin
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, and Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| | - Alina Mehra
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, and Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| | - Jonathan D Schertzer
- Department of Biochemistry and Biomedical Sciences, Farncombe Family Digestive Health Research Institute, and Centre for Metabolism, Obesity and Diabetes Research, McMaster University, Hamilton, Ontario, Canada, L8N 3Z5
| |
Collapse
|
42
|
Valitutti F, Mennini M, Monacelli G, Fagiolari G, Piccirillo M, Di Nardo G, Di Cara G. Intestinal permeability, food antigens and the microbiome: a multifaceted perspective. FRONTIERS IN ALLERGY 2025; 5:1505834. [PMID: 39850945 PMCID: PMC11754301 DOI: 10.3389/falgy.2024.1505834] [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: 10/03/2024] [Accepted: 12/16/2024] [Indexed: 01/25/2025] Open
Abstract
The gut barrier encompasses several interactive, physical, and functional components, such as the gut microbiota, the mucus layer, the epithelial layer and the gut mucosal immunity. All these contribute to homeostasis in a well-regulated manner. Nevertheless, this frail balance might be disrupted for instance by westernized dietary habits, infections, pollution or exposure to antibiotics, thus diminishing protective immunity and leading to the onset of chronic diseases. Several gaps of knowledge still exist as regards this multi-level interaction. In this review we aim to summarize current evidence linking food antigens, microbiota and gut permeability interference in diverse disease conditions such as celiac disease (CeD), non-celiac wheat sensitivity (NCWS), food allergies (FA), eosinophilic gastrointestinal disorder (EOGID) and irritable bowel syndrome (IBS). Specific food elimination diets are recommended for CeD, NCWS, FA and in some cases for EOGID. Undoubtfully, each of these conditions is very different and quite unique, albeit food antigens/compounds, intestinal permeability and specific microbiota signatures orchestrate immune response and decide clinical outcomes for all of them.
Collapse
Affiliation(s)
- Francesco Valitutti
- Department of Medicine and Surgery, Pediatric Unit, University of Perugia, Perugia, Italy
- European Biomedical Research Institute of Salerno (EBRIS), Salerno, Italy
| | - Maurizio Mennini
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Pediatric Unit, Sant'Andrea University Hospital, Rome, Italy
| | - Gianluca Monacelli
- Department of Medicine and Surgery, Pediatric Unit, University of Perugia, Perugia, Italy
| | - Giulia Fagiolari
- Department of Medicine and Surgery, Pediatric Unit, University of Perugia, Perugia, Italy
| | - Marisa Piccirillo
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Pediatric Unit, Sant'Andrea University Hospital, Rome, Italy
| | - Giovanni Di Nardo
- Department of Neurosciences, Mental Health and Sensory Organs (NESMOS), Faculty of Medicine and Psychology, Sapienza University of Rome, Pediatric Unit, Sant'Andrea University Hospital, Rome, Italy
| | - Giuseppe Di Cara
- Department of Medicine and Surgery, Pediatric Unit, University of Perugia, Perugia, Italy
| |
Collapse
|
43
|
Szymczak-Pajor I, Drzewoski J, Kozłowska M, Krekora J, Śliwińska A. The Gut Microbiota-Related Antihyperglycemic Effect of Metformin. Pharmaceuticals (Basel) 2025; 18:55. [PMID: 39861118 PMCID: PMC11768994 DOI: 10.3390/ph18010055] [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: 12/01/2024] [Revised: 12/26/2024] [Accepted: 12/30/2024] [Indexed: 01/27/2025] Open
Abstract
It is critical to sustain the diversity of the microbiota to maintain host homeostasis and health. Growing evidence indicates that changes in gut microbial biodiversity may be associated with the development of several pathologies, including type 2 diabetes mellitus (T2DM). Metformin is still the first-line drug for treatment of T2DM unless there are contra-indications. The drug primarily inhibits hepatic gluconeogenesis and increases the sensitivity of target cells (hepatocytes, adipocytes and myocytes) to insulin; however, increasing evidence suggests that it may also influence the gut. As T2DM patients exhibit gut dysbiosis, the intestinal microbiome has gained interest as a key target for metabolic diseases. Interestingly, changes in the gut microbiome were also observed in T2DM patients treated with metformin compared to those who were not. Therefore, the aim of this review is to present the current state of knowledge regarding the association of the gut microbiome with the antihyperglycemic effect of metformin. Numerous studies indicate that the reduction in glucose concentration observed in T2DM patients treated with metformin is due in part to changes in the biodiversity of the gut microbiota. These changes contribute to improved intestinal barrier integrity, increased production of short-chain fatty acids (SCFAs), regulation of bile acid metabolism, and enhanced glucose absorption. Therefore, in addition to the well-recognized reduction of gluconeogenesis, metformin also appears to exert its glucose-lowering effect by influencing gut microbiome biodiversity. However, we are only beginning to understand how metformin acts on specific microorganisms in the intestine, and further research is needed to understand its role in regulating glucose metabolism, including the impact of this remarkable drug on specific microorganisms in the gut.
Collapse
Affiliation(s)
- Izabela Szymczak-Pajor
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland;
| | - Józef Drzewoski
- Central Teaching Hospital of the Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland; (J.D.); (J.K.)
| | - Małgorzata Kozłowska
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland;
| | - Jan Krekora
- Central Teaching Hospital of the Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland; (J.D.); (J.K.)
| | - Agnieszka Śliwińska
- Department of Nucleic Acid Biochemistry, Medical University of Lodz, 251 Pomorska Str., 92-213 Lodz, Poland;
| |
Collapse
|
44
|
Yue Z, Xiang W, Duping D, Yuanyuan G, Xuanyi C, Juan L, Xiaojuan H. Integrating 16S rDNA and metabolomics to uncover the therapeutic mechanism of electroacupuncture in type 2 diabetic rats. Front Microbiol 2025; 15:1436911. [PMID: 39834366 PMCID: PMC11743489 DOI: 10.3389/fmicb.2024.1436911] [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: 05/22/2024] [Accepted: 11/25/2024] [Indexed: 01/22/2025] Open
Abstract
Objective This study aimed to investigate the impact of electroacupuncture (EA) on blood glucose levels, gut microbiota, short-chain fatty acids (SCFAs), and glucagon-like peptide-1 (GLP-1) in a rat model of type 2 diabetes mellitus (T2DM). Methods Forty Sprague-Dawley (SD) rats were randomly assigned to five groups (n = 8/group) using a random number table: normal control, T2DM model, electroacupuncture (EA), EA + antibiotics (EA + A), and antibiotics (A). The normal rats received a standard diet and saline gavage, while the other groups were fed a high-fat diet and emulsion. The EA + A and A groups received additional antibiotic solution gavage. The normal, model, and A groups were immobilized and restrained for 30 min, six times per week, for 4 weeks. The EA and EA + A groups received EA treatment at specific acupoints for 30 min, six times per week, for 4 weeks. EA parameters were continuous waves at 10 Hz and 1-2 mA. During the intervention, water and food consumption, body weight, fasting blood glucose (FBG), and oral glucose tolerance test (OGTT) were monitored. Pancreatic tissue was examined using hematoxylin and eosin (H&E) staining. Fecal microbial communities were analyzed by 16S rDNA sequencing, and short-chain fatty acids (SCFAs) were measured using gas chromatography-mass spectrometry (GC-MS). Serum levels of fasting insulin (FINS), glycated hemoglobin (HbA1c), and glucagon-like peptide-1 (GLP-1) were determined using enzyme-linked immunosorbent assay (ELISA). Results EA significantly improved daily water intake, food consumption, and body weight in T2DM rats (p < 0.01). EA also reduced FBG, the area under the curve of the OGTT, FINS, and homeostasis model assessment of insulin resistance (HOMA-IR) in T2DM rats (p < 0.05). The ELISA results showed a lower concentration of HbA1c in the EA group (p < 0.05). EA improved the overall morphology and area of pancreatic islets, increased the number of β-cell nuclei, and alleviated β-cell hypertrophy. The abundance of operational taxonomic units (OTUs) in the EA group increased than the model group (p < 0.05), and EA upregulated the Shannon, Chao1, and Ace indices (p < 0.05). EA increased the concentrations of acetic acid, butyric acid, and GLP-1 (p < 0.05). Correlation analysis revealed negative associations between Lactobacillaceae (R = -0.81, p = 0.015) and Lactobacillus (R = -0.759, p = 0.029) with FBG. Peptostreptococcaceae and Romboutsia were negatively correlated with HbA1c (R = -0.81, p = 0.015), while Enterobacteriaceae was positively correlated with OGTT (R = 0.762, p = 0.028). GLP-1 was positively correlated with acetic acid (R = 0.487, p = 0.001), butyric acid (R = 0.586, p = 0.000), isovaleric acid (R = 0.374, p = 0.017), valeric acid (R = 0.535, p = 0.000), and caproic acid (R = 0.371, p = 0.018). Antibiotics disrupted the intestinal microbiota structure and weakened the therapeutic effects of EA. Conclusion EA effectively improved glucose metabolism in T2DM rats. The hypoglycemic effects of EA were associated with the regulation of gut microbiota, SCFAs, and GLP-1.
Collapse
Affiliation(s)
- Zhang Yue
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wang Xiang
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Deng Duping
- Department of Rehabilitation Medicine, Meishan Hospital of Traditional Chinese Medicine, Meishan, China
| | - Gong Yuanyuan
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Chen Xuanyi
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Li Juan
- School of Health Preservation and Rehabilitation, Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Affiliated Sichuan Provincial Rehabilitation Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Hong Xiaojuan
- School of Acupuncture-Moxibustion and Tuina, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
45
|
Sternini C, Rozengurt E. Bitter taste receptors as sensors of gut luminal contents. Nat Rev Gastroenterol Hepatol 2025; 22:39-53. [PMID: 39468215 DOI: 10.1038/s41575-024-01005-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 10/03/2024] [Indexed: 10/30/2024]
Abstract
Taste is important in the selection of food and is orchestrated by a group of distinct receptors, the taste G protein-coupled receptors (GPCRs). Taste 1 receptors (Tas1rs in mice and TAS1Rs in humans; also known as T1Rs) detect sweet and umami tastes, and taste 2 receptors (Tas2rs in mice and TAS2Rs in humans; also known as T2Rs) detect bitterness. These receptors are also expressed in extraoral sites, including the gastrointestinal mucosa. Tas2rs/TAS2Rs have gained interest as potential targets to prevent or treat metabolic disorders. These bitter taste receptors are expressed in functionally distinct types of gastrointestinal mucosal cells, including enteroendocrine cells, which, upon stimulation, increase intracellular Ca2+ and release signalling molecules that regulate gut chemosensory processes critical for digestion and absorption of nutrients, for neutralization and expulsion of harmful substances, and for metabolic regulation. Expression of Tas2rs/TAS2Rs in gut mucosa is upregulated by high-fat diets, and intraluminal bitter 'tastants' affect gastrointestinal functions and ingestive behaviour through local and gut-brain axis signalling. Tas2rs/TAS2Rs are also found in Paneth and goblet cells, which release antimicrobial peptides and glycoproteins, and in tuft cells, which trigger type 2 immune response against parasites, thus providing a direct line of defence against pathogens. This Review will focus on gut Tas2r/TAS2R distribution, signalling and regulation in enteroendocrine cells, supporting their role as chemosensors of luminal content that serve distinct functions as regulators of body homeostasis and immune response.
Collapse
Affiliation(s)
- Catia Sternini
- Division of Digestive Diseases, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA.
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA.
- Department of Neurobiology, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA.
| | - Enrique Rozengurt
- Division of Digestive Diseases, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
- Department of Medicine, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
- Molecular Biology Institute, David Geffen School of Medicine, University of California at Los Angeles, Los Angeles, CA, USA
| |
Collapse
|
46
|
Saban Güler M, Arslan S, Ağagündüz D, Cerqua I, Pagano E, Berni Canani R, Capasso R. Butyrate: A potential mediator of obesity and microbiome via different mechanisms of actions. Food Res Int 2025; 199:115420. [PMID: 39658184 DOI: 10.1016/j.foodres.2024.115420] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Revised: 11/08/2024] [Accepted: 11/19/2024] [Indexed: 12/12/2024]
Abstract
Butyrate, a short-chain fatty acid, is a crucial product of gut microbial fermentation with significant implications for various metabolic and physiological processes. Dietary sources of butyrate are limited, primarily derived from the fermentation of dietary fibers by butyrate-producing gut bacteria. Butyrate exerts its effects primarily as a histone deacetylase (HDAC) inhibitor and through signaling pathways involving G protein-coupled receptors (GPCRs). Its diverse benefits include promoting gut health, enhancing energy metabolism, and potentially alleviating complications associated with obesity. However, the exact role of butyrate in obesity is still under investigation, with a limited number of human trials necessitating further research to determine its efficacy and safety profile. Moreover, butyrate impact on the gut-brain axis and its modulation of microbiome effect on behavior highlight its broader importance in regulating host physiology. A thorough understanding of the metabolic pathways and mechanisms of butyrate is essential for developing targeted interventions for metabolic disorders. Continued research is crucial to fully realize its therapeutic potential and optimize its clinical applications in human health. In summary, this review illuminates the multifaceted role of butyrate as a potential mediator of obesity and related metabolic changes.
Collapse
Affiliation(s)
- Meryem Saban Güler
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, 06490 Ankara, Turkey
| | - Sabriye Arslan
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, 06490 Ankara, Turkey
| | - Duygu Ağagündüz
- Department of Nutrition and Dietetics, Faculty of Health Sciences, Gazi University, 06490 Ankara, Turkey.
| | - Ida Cerqua
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Ester Pagano
- Department of Pharmacy, School of Medicine and Surgery, University of Naples Federico II, 80131 Naples, Italy
| | - Roberto Berni Canani
- Department of Translational Medical Science and ImmunoNutritionLab at CEINGE Biotechnologies Research Center and Task Force for Microbiome Studies, University of Naples Federico II, Naples, Italy
| | - Raffaele Capasso
- Department of Agricultural Sciences, University of Naples Federico II, Portici, 80055 Naples, Italy.
| |
Collapse
|
47
|
Xia L, Li C, Zhao J, Sun Q, Mao X. Rebalancing immune homeostasis in combating disease: The impact of medicine food homology plants and gut microbiome. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2025; 136:156150. [PMID: 39740376 DOI: 10.1016/j.phymed.2024.156150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 09/27/2024] [Accepted: 10/10/2024] [Indexed: 01/02/2025]
Abstract
BACKGROUND Gut microbiota plays an important role in multiple human physiological processes and an imbalance in it, including the species, abundance, and metabolites can lead to diseases. These enteric microorganisms modulate immune homeostasis by presenting a myriad of antigenic determinants and microbial metabolites. Medicinal and food homologous (MFH) plants, edible herbal materials for both medicine and food, are important parts of Traditional Chinese Medicine (TCM). MFH plants have drawn much attention due to their strong biological activity and low toxicity. However, the interplay of MFH and gut microbiota in rebalancing the immune homeostasis in combating diseases needs systematic illumination. PURPOSE The review discusses the interaction between MFH and gut microbiota, including the effect of MFH on the major group of gut microbiota and the metabolic effect of gut microbiota on MFH. Moreover, how gut microbiota influences the immune system in terms of innate and adaptive immunity is addressed. Finally, the immunoregulatory mechanisms of MFH in regulation of host pathophysiology via gut microbiota are summarized. METHODS Literature was searched, analyzed, and collected using databases, including PubMed, Web of Science, and Google Scholar using relevant keywords. The obtained articles were screened and summarized by the research content of MFH and gut microbiota in immune regulation. RESULTS The review demonstrates the interaction between MFH and gut microbiota in disease prevention and treatment. Not only do the intestinal microorganisms and intestinal mucosa constitute an important immune barrier of the human body, but also lymphoid tissue and diffused immune cells within the mucosa participate in the response of innate immunity and adaptive immunity. MFH modulates immune regulation by affecting intestinal flora, helps maintain the balance of the immune system and interfere with the occurrence and development of a broad category of diseases. CONCLUSION Being absorbed from the gastrointestinal tract, MFH can have profound effects on gut microbiota. In turn, the gut microbiota also actively participate in the bioconversion of complex constituents from MFH, which could further influence their physiological and pharmacological properties. The review deepens the understanding of the relationship among MFH, gut microbiota, immune system, and human diseases and further promotes the progression of additional relevant research.
Collapse
Affiliation(s)
- Lu Xia
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Chuangen Li
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China
| | - Jia Zhao
- School of Chinese Medicine, Li Ka Shing Faculty of Medicine, the University of Hong Kong, Hong Kong SAR, China
| | - Quancai Sun
- Department of Health, Nutrition, and Food sciences, Florida State University, Tallahassee, USA
| | - Xiaowen Mao
- State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macau SAR, China.
| |
Collapse
|
48
|
Kamath S, Sokolenko E, Collins K, Chan NSL, Mills N, Clark SR, Marques FZ, Joyce P. IUPHAR themed review: The gut microbiome in schizophrenia. Pharmacol Res 2025; 211:107561. [PMID: 39732352 DOI: 10.1016/j.phrs.2024.107561] [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: 11/25/2024] [Revised: 12/11/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
Gut microbial dysbiosis or altered gut microbial consortium, in schizophrenia suggests a pathogenic role through the gut-brain axis, influencing neuroinflammatory and neurotransmitter pathways critical to psychotic, affective, and cognitive symptoms. Paradoxically, conventional psychotropic interventions may exacerbate this dysbiosis, with antipsychotics, particularly olanzapine, demonstrating profound effects on microbial architecture through disruption of bacterial phyla ratios, diminished taxonomic diversity, and attenuated short-chain fatty acid synthesis. To address these challenges, novel therapeutic strategies targeting the gut microbiome, encompassing probiotic supplementation, prebiotic compounds, faecal microbiota transplantation, and rationalised co-pharmacotherapy, show promise in attenuating antipsychotic-induced metabolic disruptions while enhancing therapeutic efficacy. Harnessing such insights, precision medicine approaches promise to transform antipsychotic prescribing practices by identifying patients at risk of metabolic side effects based on their microbial profiles. This IUPHAR review collates the current literature landscape of the gut-brain axis and its intricate relationship with schizophrenia while advocating for integrating microbiome assessments and therapeutic management. Such a fundamental shift in proposing microbiome-informed psychotropic prescriptions to optimise therapeutic efficacy and reduce adverse metabolic impacts would align antipsychotic treatments with microbiome safety, prioritising 'gut-neutral' or gut-favourable drugs to safeguard long-term patient outcomes in schizophrenia therapy.
Collapse
Affiliation(s)
- Srinivas Kamath
- UniSA Clinical & Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Elysia Sokolenko
- Discipline of Anatomy and Pathology, School of Biomedicine, University of Adelaide, Adelaide, South Australia 5005, Australia
| | - Kate Collins
- UniSA Clinical & Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Nicole S L Chan
- UniSA Clinical & Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia
| | - Natalie Mills
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Scott R Clark
- Discipline of Psychiatry, Adelaide Medical School, University of Adelaide, Adelaide, South Australia 5000, Australia
| | - Francine Z Marques
- Baker Heart and Diabetes Institute, Melbourne, VIC, Australia; Hypertension Research Laboratory, School of Biological Sciences and Victorian Heart Institute, Monash University, Melbourne, VIC, Australia
| | - Paul Joyce
- UniSA Clinical & Health Sciences, University of South Australia, Adelaide, South Australia 5000, Australia.
| |
Collapse
|
49
|
Harada K, Wada E, Osuga Y, Shimizu K, Uenoyama R, Hirai MY, Maekawa F, Miyazaki M, Hayashi YK, Nakamura K, Tsuboi T. Intestinal butyric acid-mediated disruption of gut hormone secretion and lipid metabolism in vasopressin receptor-deficient mice. Mol Metab 2025; 91:102072. [PMID: 39668067 PMCID: PMC11728074 DOI: 10.1016/j.molmet.2024.102072] [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: 09/18/2024] [Revised: 11/14/2024] [Accepted: 11/18/2024] [Indexed: 12/14/2024] Open
Abstract
OBJECTIVES Arginine vasopressin (AVP), known as an antidiuretic hormone, is also crucial in metabolic homeostasis. Although AVP receptor-deficient mice exhibit various abnormalities in glucose and lipid metabolism, the mechanism underlying these symptoms remains unclear. This study aimed to explore the involvement of the gut hormones including glucagon-like peptide-1 (GLP-1) and microbiota as essential mediators. METHODS We used the mouse GLP-1-secreting cell line, GLUTag, and performed live cell imaging to examine the contribution of V1a and V1b vasopressin receptors (V1aR and V1bR, respectively) to GLP-1 secretion. We next investigated the hormone dynamics of V1aR-deficient mice (V1aR-/- mice), V1bR-deficient mice (V1bR-/- mice), and V1aR/V1bR-double deficient mice (V1aR-/-V1bR-/-mice). RESULTS AVP induced the increase in intracellular Ca2+ levels and GLP-1 secretion from GLUTag cells in a V1aR and V1bR-dependent manner. AVP receptor-deficient mice, particularly V1aR-/-V1bR-/- mice, demonstrated impaired secretion of GLP-1 and peptide YY secreted by enteroendocrine L cells. V1aR-/-V1bR-/-mice also exhibited abnormal lipid accumulation in the brown adipose tissue and skeletal muscle. We discovered that V1aR-/-V1bR-/- mice showed increased Paneth cell-related gene expression in the small intestine, which was attributed to increased fecal butyric acid levels. Exposure to butyric acid reduced GLP-1 secretion in L cell line. Additionally, human Paneth cell-related gene expressions negatively correlated with that of V1 receptor genes. CONCLUSIONS The deficiency in V1 receptor genes may increase gut butyric acid levels and impair the function of L cells, thus dysregulating lipid homeostasis in the brown adipose tissue and skeletal muscle. This study highlights the importance of appropriate control of the gut microbiota and its metabolites, including butyric acid, for the optimum functioning of enteroendocrine cells.
Collapse
Affiliation(s)
- Kazuki Harada
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro, Tokyo 153-8902, Japan; Health and Environmental Risk Division, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba, Ibaraki 305-8506, Japan
| | - Eiji Wada
- Department of Pathophysiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku, Tokyo 160-8402, Japan
| | - Yuri Osuga
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro, Tokyo 153-8902, Japan
| | - Kie Shimizu
- Department of Pharmacology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya, Tokyo 157-8535, Japan; Division of Life Sciences, Graduate School of Science and Engineering, Saitama University, 255 Shimo-Okubo, Sakura, Saitama 338-8570, Japan
| | - Reiko Uenoyama
- The United Graduate School of Agricultural Sciences, Iwate University, 3-18-8 Ueda, Morioka, Iwate 020-8550, Japan
| | - Masami Yokota Hirai
- RIKEN Center for Sustainable Resource Science, 1-7-22 Suehiro-cho, Tsurumi-ku, Yokohama-city, Kanagawa 230-0045, Japan
| | - Fumihiko Maekawa
- Health and Environmental Risk Division, National Institute for Environmental Studies, 16-2 Onogawa, Tsukuba, Ibaraki 305-8506, Japan
| | - Masao Miyazaki
- The United Graduate School of Agricultural Sciences, Iwate University, 3-18-8 Ueda, Morioka, Iwate 020-8550, Japan
| | - Yukiko K Hayashi
- Department of Pathophysiology, Tokyo Medical University, 6-1-1 Shinjuku, Shinjuku, Tokyo 160-8402, Japan
| | - Kazuaki Nakamura
- Department of Pharmacology, National Research Institute for Child Health and Development, 2-10-1 Okura, Setagaya, Tokyo 157-8535, Japan; Division of Life Sciences, Graduate School of Science and Engineering, Saitama University, 255 Shimo-Okubo, Sakura, Saitama 338-8570, Japan
| | - Takashi Tsuboi
- Department of Life Sciences, Graduate School of Arts and Sciences, The University of Tokyo, 3-8-1 Komaba, Meguro, Tokyo 153-8902, Japan.
| |
Collapse
|
50
|
Maqoud F, Calabrese FM, Celano G, Mallardi D, Goscilo F, D’Attoma B, Ignazzi A, Linsalata M, Bitetto G, Di Chito M, Pesole PL, Diciolla A, Apa CA, De Pergola G, Giannelli G, De Angelis M, Russo F. Role of Increasing Body Mass Index in Gut Barrier Dysfunction, Systemic Inflammation, and Metabolic Dysregulation in Obesity. Nutrients 2024; 17:72. [PMID: 39796506 PMCID: PMC11723324 DOI: 10.3390/nu17010072] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 12/24/2024] [Accepted: 12/26/2024] [Indexed: 01/30/2025] Open
Abstract
AIMS This study explores the link between body mass index (BMI), intestinal permeability, and associated changes in anthropometric and impedance parameters, lipid profiles, inflammatory markers, fecal metabolites, and gut microbiota taxa composition in participants having excessive body mass. METHODS A cohort of 58 obese individuals with comparable diet, age, and height was divided into three groups based on a priori clustering analyses that fit with BMI class ranges: Group I (25-29.9), Group II (30-39.9), and Group III (>40). Anthropometric and clinical parameters were assessed, including plasma C-reactive protein and cytokine profiles as inflammation markers. Intestinal permeability was measured using a multisaccharide assay, with fecal/serum zonulin and serum claudin-5 and claudin-15 levels. Fecal microbiota composition and metabolomic profiles were analyzed using a phylogenetic microarray and GC-MS techniques. RESULTS The statistical analyses of the clinical parameters were based on the full sample set, whereas a subset composed of 37 randomized patients was inspected for the GC/MS metabolite profiling of fecal specimens. An increase in potentially pro-inflammatory bacterial genera (e.g., Slackia, Dorea, Granulicatella) and a reduction in beneficial genera (e.g., Adlercreutzia, Clostridia UCG-014, Roseburia) were measured. The gas chromatography/mass spectrometry analysis of urine samples evidenced a statistically significant increase in m-cymen-8-ol, 1,3,5-Undecatriene, (E, Z) and a decreased concentration of p-cresol, carvone, p-cresol, and nonane. CONCLUSIONS Together, these data demonstrated how an increased BMI led to significant changes in inflammatory markers, intestinal barrier metabolites, glucose metabolism, endocrine indicators, and fecal metabolomic profiles that can indicate a different metabolite production from gut microbiota. Our findings suggest that targeting intestinal permeability may offer a therapeutic approach to prevent and manage obesity and related metabolic complications, reinforcing the link between gut barrier function and obesity.
Collapse
Affiliation(s)
- Fatima Maqoud
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (F.M.); (D.M.); (F.G.); (B.D.); (A.I.); (M.L.)
| | - Francesco Maria Calabrese
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, 70126 Bari, Italy; (G.C.); (C.A.A.); (M.D.A.)
| | - Giuseppe Celano
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, 70126 Bari, Italy; (G.C.); (C.A.A.); (M.D.A.)
| | - Domenica Mallardi
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (F.M.); (D.M.); (F.G.); (B.D.); (A.I.); (M.L.)
| | - Francesco Goscilo
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (F.M.); (D.M.); (F.G.); (B.D.); (A.I.); (M.L.)
| | - Benedetta D’Attoma
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (F.M.); (D.M.); (F.G.); (B.D.); (A.I.); (M.L.)
| | - Antonia Ignazzi
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (F.M.); (D.M.); (F.G.); (B.D.); (A.I.); (M.L.)
| | - Michele Linsalata
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (F.M.); (D.M.); (F.G.); (B.D.); (A.I.); (M.L.)
| | - Gabriele Bitetto
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (G.B.); (M.D.C.); (G.D.P.)
| | - Martina Di Chito
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (G.B.); (M.D.C.); (G.D.P.)
| | - Pasqua Letizia Pesole
- Core Facility Biobank, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy;
| | - Arianna Diciolla
- Laboratory of Clinical Pathology, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy;
| | - Carmen Aurora Apa
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, 70126 Bari, Italy; (G.C.); (C.A.A.); (M.D.A.)
| | - Giovanni De Pergola
- Center of Nutrition for the Research and the Care of Obesity and Metabolic Diseases, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (G.B.); (M.D.C.); (G.D.P.)
| | - Gianluigi Giannelli
- Scientific Direction, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy;
| | - Maria De Angelis
- Department of Soil, Plant and Food Science, University of Bari Aldo Moro, 70126 Bari, Italy; (G.C.); (C.A.A.); (M.D.A.)
| | - Francesco Russo
- Functional Gastrointestinal Disorders Research Group, National Institute of Gastroenterology IRCCS “Saverio de Bellis”, Castellana Grotte, 70013 Bari, Italy; (F.M.); (D.M.); (F.G.); (B.D.); (A.I.); (M.L.)
| |
Collapse
|