1
|
Martinez-Medina JN, Ghazisaeedi F, Kramer C, Ziegler JF, McParland V, Mönch PW, Siegmund B, Jarquín-Díaz VH, Fulde M, Forslund-Startceva SK. Mucosal washes are useful for sampling intestinal mucus-associated microbiota despite low biomass. Gut Microbes 2025; 17:2464296. [PMID: 39980334 PMCID: PMC11849919 DOI: 10.1080/19490976.2025.2464296] [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: 12/26/2023] [Revised: 10/24/2024] [Accepted: 02/04/2025] [Indexed: 02/22/2025] Open
Abstract
Understanding the dynamic relationship between mucus-associated microbiota and host health is critical. However, studies predominantly using stool samples may not accurately represent these bacterial communities. Here, we investigated the mucus-associated microbiota in the gastrointestinal tract of mice and the terminal ileum of humans using different sample types: mucosal washes, brushes, scrapings, and intestinal contents in mice and biopsies, brushes and mucosal washes in humans. We used DNA quantification and 16S rRNA amplicon sequencing to evaluate the comparability of the information yielded from the different sample types under a controlled benchmark. In mice, mucosal washes and brushes had comparative bacterial DNA and host DNA contamination than scraping samples. Similarly, in humans, washes outperformed biopsies in bacterial DNA content. Read counts and microbiota alpha diversity remained remarkably similar in mice and between brushes and washes in humans. The composition of the microbiota varied based on the subsegment and sample type in mice and sample type in humans. We conclude that washes and brushes reduce host contamination without inducing substantial compositional bias when sampling mucosal microbiota. Our findings suggest that mucosal washes and brushes are a viable alternative to biopsies in humans and scrapings in mice, thereby improving the transferability of results across hosts. Our study highlights the importance of focusing on mucus-associated microbiota to better capture host-microbiome interactions at their closer interface.
Collapse
Affiliation(s)
- Jennifer N. Martinez-Medina
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin and The Max-Delbrück Center, Berlin, Germany
| | - Fereshteh Ghazisaeedi
- Institute of Microbiology and Epizootics, School of Veterinary Medicine at the Freie Universität Berlin, Berlin, Germany
- Veterinary Centre for Resistance Research (TZR), School of Veterinary Medicine at the Freie Universität Berlin, Berlin, Germany
| | - Catharina Kramer
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany
| | - Jörn F Ziegler
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Junior Clinician Scientist Program, Berlin, Germany
| | - Victoria McParland
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin and The Max-Delbrück Center, Berlin, Germany
| | - Paul W. Mönch
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany
| | - Britta Siegmund
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Department of Gastroenterology, Infectious Diseases and Rheumatology, Berlin, Germany
| | - Víctor Hugo Jarquín-Díaz
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin and The Max-Delbrück Center, Berlin, Germany
| | - Marcus Fulde
- Institute of Microbiology and Epizootics, School of Veterinary Medicine at the Freie Universität Berlin, Berlin, Germany
- Veterinary Centre for Resistance Research (TZR), School of Veterinary Medicine at the Freie Universität Berlin, Berlin, Germany
| | - Sofia K. Forslund-Startceva
- Max-Delbrück-Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- Charité – Universitätsmedizin Berlin, Corporate Member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Experimental and Clinical Research Center, a Cooperation of Charité-Universitätsmedizin and The Max-Delbrück Center, Berlin, Germany
- Berlin Institute of Health at Charité – Universitätsmedizin Berlin, BIH Biomedical Innovation Academy, BIH Charité Junior Clinician Scientist Program, Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
- Structural and Computational Biology, European Molecular Biology Laboratory, Heidelberg, Germany
| |
Collapse
|
2
|
Jamerlan AM, An SSA, Hulme JP. Microbial diversity and fitness in the gut-brain axis: influences on developmental risk for Alzheimer's disease. Gut Microbes 2025; 17:2486518. [PMID: 40207973 PMCID: PMC11988266 DOI: 10.1080/19490976.2025.2486518] [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: 08/28/2024] [Revised: 03/19/2025] [Accepted: 03/25/2025] [Indexed: 04/11/2025] Open
Abstract
The gut-brain axis (GBA) denotes the dynamic and bidirectional communication system that connects the gastrointestinal tract and the central nervous system (CNS). This review explored this axis, focusing on the role of microbial diversity and fitness in maintaining gastrointestinal health and preventing neurodegeneration, particularly in Alzheimer's disease (AD). Gut dysbiosis, characterized by the imbalance in populations of beneficial and harmful bacteria, has been associated with increased systemic inflammation, neuroinflammation, and the progression of AD through pathogenic mechanisms involving amyloid deposition, tauopathy, and increased blood-brain barrier (BBB) permeability. Emerging evidence highlighted the therapeutic potential of probiotics, dietary interventions, and intermittent fasting in restoring microbial balance, reducing inflammation, and minimizing neurodegenerative risks. Probiotics and synbiotics are promising in helping improve cognitive function and metabolic health, while dietary patterns like the Mediterranean diet were linked to decreased neuroinflammation and enhanced gut-brain communication. Despite significant advancement, further research is needed to elucidate the specific microbial strains, metabolites, and mechanisms influencing brain health. Future studies employing longitudinal designs and advanced omics technologies are essential to developing targeted microbiome-based therapies for managing AD-related disorders.
Collapse
Affiliation(s)
- Angelo M. Jamerlan
- Department of Bionanotechnology, Bionano Research Institute, Gachon University, Seongnam-si, Republic of Korea
| | - Seong Soo A. An
- Department of Bionanotechnology, Bionano Research Institute, Gachon University, Seongnam-si, Republic of Korea
| | - John P. Hulme
- Department of Bionanotechnology, Bionano Research Institute, Gachon University, Seongnam-si, Republic of Korea
| |
Collapse
|
3
|
Qu L, Chi Z, Zou ZP, Zhou Y, Ye BC. Development of ultrasound-visualized tumor-targeting engineered bacteria for precise tumor therapy. Synth Syst Biotechnol 2025; 10:774-782. [PMID: 40270642 PMCID: PMC12018036 DOI: 10.1016/j.synbio.2025.03.009] [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: 01/07/2025] [Revised: 03/12/2025] [Accepted: 03/26/2025] [Indexed: 04/25/2025] Open
Abstract
In situ imaging diagnosis and precise treatment of deep tumor tissues are hotspots in life sciences and medical research. In recent years, using focused ultrasound to remotely control engineered bacteria for drug release has become one of the methods for precise in vivo drug delivery. However, non-visualized engineered bacteria pose challenges for precise control within the body. Therefore, there is an urgent need for an engineered bacterial vector capable of deep tissue imaging to precisely locate bacteria in vivo. Acoustic reporter genes (ARGs) are biological elements used for deep tissue imaging, with gene clusters over 8 kb. However, ARGs are often tested on plasmids, which hinders stable expression in vivo and limits the space for inserting components that regulate drug release. Therefore, we used the attenuated Salmonella typhimurium VNP20009, known for its tumor-targeting ability, as the chassis bacteria. By using CRISPR-Cas9 technology, we inserted ARGs into the genome and optimized the promoter strength and copy number for ARG expression, constructing ultrasound-visible engineered bacteria expressing gas vesicles on the genome. Additionally, by knocking out the stress protein gene htrA in VNP20009, we increased the maximum injection dose by tenfold and the tumor specificity by a hundredfold. The constructed ultrasound-visible engineered bacteria can stably synthesize gas vesicles and output ultrasound signals while directly carrying drug plasmids for tumor therapy. Our research provides an effective vector for diagnosis and precise treatment.
Collapse
Affiliation(s)
- Li Qu
- Lab of Biosystems and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Zhou Chi
- Lab of Biosystems and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Zhen-Ping Zou
- Lab of Biosystems and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Ying Zhou
- Lab of Biosystems and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
| | - Bang-Ce Ye
- Lab of Biosystems and Microanalysis, State Key Laboratory of Bioreactor Engineering, East China University of Science and Technology, Shanghai, 200237, China
| |
Collapse
|
4
|
Almonte AA, Thomas S, Zitvogel L. Microbiota-centered interventions to boost immune checkpoint blockade therapies. J Exp Med 2025; 222:e20250378. [PMID: 40261296 PMCID: PMC12013646 DOI: 10.1084/jem.20250378] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2025] [Revised: 04/06/2025] [Accepted: 04/09/2025] [Indexed: 04/24/2025] Open
Abstract
Immune checkpoint blockade therapies have markedly advanced cancer treatment by invigorating antitumor immunity and extending patient survival. However, therapeutic resistance and immune-related toxicities remain major concerns. Emerging evidence indicates that microbial dysbiosis diminishes therapeutic response rates, while a diverse gut ecology and key beneficial taxa correlate with improved treatment outcomes. Therefore, there is a growing understanding that manipulating the gut microbiota could boost therapy efficacy. This review examines burgeoning methods that target the gut microbiome to optimize therapy and innovative diagnostic tools to detect dysbiosis, and highlights challenges that remain to be addressed in the field.
Collapse
Affiliation(s)
- Andrew A. Almonte
- Gustave Roussy Cancer Campus, Clinicobiome, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale Contre le Cancer, Villejuif, France
| | - Simon Thomas
- Gustave Roussy Cancer Campus, Clinicobiome, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Kremlin-Bicêtre, France
| | - Laurence Zitvogel
- Gustave Roussy Cancer Campus, Clinicobiome, Villejuif, France
- Institut National de la Santé et de la Recherche Médicale (INSERM) U1015, Equipe Labellisée-Ligue Nationale Contre le Cancer, Villejuif, France
- Université Paris-Saclay, Kremlin-Bicêtre, France
- Center of Clinical Investigations in Biotherapies of Cancer (BIOTHERIS) 1428, Villejuif, France
| |
Collapse
|
5
|
Yoon H, Gerdes LA, Beigel F, Sun Y, Kövilein J, Wang J, Kuhlmann T, Flierl-Hecht A, Haller D, Hohlfeld R, Baranzini SE, Wekerle H, Peters A. Multiple sclerosis and gut microbiota: Lachnospiraceae from the ileum of MS twins trigger MS-like disease in germfree transgenic mice-An unbiased functional study. Proc Natl Acad Sci U S A 2025; 122:e2419689122. [PMID: 40258140 DOI: 10.1073/pnas.2419689122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2024] [Accepted: 02/25/2025] [Indexed: 04/23/2025] Open
Abstract
We developed a two-tiered strategy aiming to identify gut bacteria functionally linked to the development of multiple sclerosis (MS). First, we compared gut microbial profiles in a cohort of 81 monozygotic twins discordant for MS. This approach allowed to minimize confounding effects by genetic and early environmental factors and identified over 50 differently abundant taxa with the majority of increased taxa within the Firmicutes. These included taxa previously described to be associated with MS (Anaerotruncus colihominis and Eisenbergiella tayi), along with newly identified taxa, such as Copromonas and Acutalibacter. Second, we interrogated the intestinal habitat and functional impact of individual taxa on the development of MS-like disease. In an exploratory approach, we enteroscopically sampled microbiota from different gut segments of selected twin pairs and compared their compositional profiles. To assess their functional potential, samples were orally transferred into germfree transgenic mice prone to develop spontaneous MS-like experimental autoimmune encephalomyelitis (EAE) upon bacterial colonization. We found that MS-derived ileal microbiota induced EAE at substantially higher rates than analogous material from healthy twin donors. Furthermore, female mice were more susceptible to disease development than males. The likely active organisms were identified as Eisenbergiella tayi and Lachnoclostridium, members of the Lachnospiraceae family. Our results identify potentially disease-facilitating bacteria sampled from the ileum of MS affected twins. The experimental strategy may pave the way to functionally understand the role of gut microbiota in initiation of MS.
Collapse
Affiliation(s)
- Hongsup Yoon
- Institute of Clinical Neuroimmunology, University Hospital Ludwig-Maximilians-Universität München, Martinsried 82152, Germany
- Biomedical Center, Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried 82152, Germany
- Max Planck Institute for Biological Intelligence, Martinsried 82152, Germany
| | - Lisa Ann Gerdes
- Institute of Clinical Neuroimmunology, University Hospital Ludwig-Maximilians-Universität München, Martinsried 82152, Germany
- Biomedical Center, Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried 82152, Germany
- Munich Cluster of Systems Neurology, Munich 81377, Germany
| | - Florian Beigel
- Department of Medicine II, University Hospital, Ludwig-Maximilians-Universität München, Munich 81377, Germany
| | - Yihui Sun
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA 94158
| | - Janine Kövilein
- Institute of Clinical Neuroimmunology, University Hospital Ludwig-Maximilians-Universität München, Martinsried 82152, Germany
- Biomedical Center, Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried 82152, Germany
- Max Planck Institute for Biological Intelligence, Martinsried 82152, Germany
| | - Jiancheng Wang
- Institute of Clinical Neuroimmunology, University Hospital Ludwig-Maximilians-Universität München, Martinsried 82152, Germany
- Biomedical Center, Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried 82152, Germany
| | - Tanja Kuhlmann
- Institute of Neuropathology, University Hospital Münster, Münster 48153, Germany
| | - Andrea Flierl-Hecht
- Institute of Clinical Neuroimmunology, University Hospital Ludwig-Maximilians-Universität München, Martinsried 82152, Germany
| | - Dirk Haller
- Zentralinstitut für Ernährungs- und Lebensmittelforschung Institute for Food and Health, Technical University of Munich, Freising 85354, Germany
| | - Reinhard Hohlfeld
- Institute of Clinical Neuroimmunology, University Hospital Ludwig-Maximilians-Universität München, Martinsried 82152, Germany
| | - Sergio E Baranzini
- Weill Institute for Neurosciences, Department of Neurology, University of California San Francisco, San Francisco, CA 94158
| | - Hartmut Wekerle
- Institute of Clinical Neuroimmunology, University Hospital Ludwig-Maximilians-Universität München, Martinsried 82152, Germany
- Max Planck Institute for Biological Intelligence, Martinsried 82152, Germany
| | - Anneli Peters
- Institute of Clinical Neuroimmunology, University Hospital Ludwig-Maximilians-Universität München, Martinsried 82152, Germany
- Biomedical Center, Faculty of Medicine, Ludwig-Maximilians-Universität München, Martinsried 82152, Germany
| |
Collapse
|
6
|
Zwolschen JW, Vos AP, Ariëns RMC, Schols HA. Fermentation characteristics of pectin-derived oligosaccharides from enzyme treated side streams of citrus processing. Carbohydr Polym 2025; 355:123352. [PMID: 40037724 DOI: 10.1016/j.carbpol.2025.123352] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2024] [Revised: 12/28/2024] [Accepted: 02/03/2025] [Indexed: 03/06/2025]
Abstract
This study explored the conversion of citrus juice side streams into fermentable oligosaccharides for potential gut health benefits. Alcohol washed, insoluble lemon peel waste was enzymatically treated using technical pectinolytic enzyme preparations, yielding mixtures of galactose- arabinose- and either methyl-esterified or non-methyl-esterified galacturonic acid oligosaccharides (OS) with a Δ4,5-unsaturated non-reducing end resulting in mixtures of pectin-derived OS: POS and POSNME. Both mixtures were completely fermented during in vitro batch fermentation by proximal and distal microbiota of three healthy adult donors. Fermentation by distal and proximal microbiota resulted in similar methyl-ester-dependent mechanisms of POS utilization, yielding health beneficial acetate, propionate and butyrate in significant amounts. Arabinose-, galactose- and non-methyl-esterified Δ4,5-unsaturated galacturonic acid OS were utilized significantly faster by the distal and proximal microbiota of donors 1 and 2 compared to methyl-esterified Δ4,5-unsaturated galacturonic acid OS, suggesting methyl-esterification of Δ4,5-unsaturated galacturonic acid oligosaccharides as a substantial regulator of POS fermentability. The findings presented in this manuscript suggest that carbohydrate molecular structure and availability, rather than microbiota composition, determine carbohydrate fermentation patterns along the colon, emphasizing that the consumption of differently fermentable fiber is essential to promote gut health along the colon.
Collapse
Affiliation(s)
- J W Zwolschen
- Wageningen University & Research, Laboratory of Food Chemistry, Bornse Weilanden 9, 6708 WG Wageningen, the Netherlands
| | - A P Vos
- Wageningen Food & Biobased Research, Wageningen, the Netherlands
| | - R M C Ariëns
- Wageningen Food & Biobased Research, Wageningen, the Netherlands
| | - H A Schols
- Wageningen University & Research, Laboratory of Food Chemistry, Bornse Weilanden 9, 6708 WG Wageningen, the Netherlands.
| |
Collapse
|
7
|
Tanabe M, Kunisawa K, Saito I, Kosuge A, Tezuka H, Kawai T, Kon Y, Yoshidomi K, Kagami A, Hasegawa M, Kubota H, Ojika H, Fujii T, Tochio T, Hirooka Y, Saito K, Nabeshima T, Mouri A. Adolescent social isolation decreases colonic goblet cells and impairs spatial cognition through the reduction of cystine. Mol Psychiatry 2025; 30:2137-2151. [PMID: 39613916 PMCID: PMC12014494 DOI: 10.1038/s41380-024-02826-9] [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: 10/29/2024] [Accepted: 11/04/2024] [Indexed: 12/01/2024]
Abstract
Negative experiences during adolescence, such as social isolation (SI), bullying, and abuse, increase the risk of psychiatric diseases in adulthood. However, the pathogenesis of psychiatric diseases induced by these factors remain poorly understood. In adolescents, stress affects the intestinal homeostasis in the gut-brain axis. This study determined whether adolescent SI induces behavioral abnormalities by disrupting colonic function. Adolescent mice exposed to SI exhibit spatial cognitive deficits and microglial activation in the hippocampus (HIP). SI decreased the differentiation of mucin-producing goblet cells, which was accompanied by alterations in the composition of the gut microbiota, particularly the depletion of mucin-feeding bacteria. Treatment with rebamipide, which promotes goblet cell differentiation in the colon, attenuated SI-induced spatial cognitive deficits and microglial activation in the HIP and decreased cystine, a downstream metabolite of homocysteine. Treatment with cystine ameliorated SI-induced spatial cognitive deficits and increased microglial C-C motif chemokine ligand 7 (CCL7) levels in the HIP. Inhibition of CCL7 receptors by antagonists of CC motif chemokine receptors 2 (CCR2) and 3 (CCR3) in the HIP prevented spatial cognitive deficits induced by SI. Infusion of CCL7 into the HIP following microglial ablation with clodronate liposome induced spatial cognitive deficits. These findings suggest that adolescent SI decreases serum cystine levels by damaging the colonic goblet cells, resulting in spatial cognitive deficits by triggering microglial activation in the HIP. Our results indicate that increased CCL7 expression in hippocampal microglia may contribute to spatial cognitive deficits by activating CCR2 and CCR3.
Collapse
Affiliation(s)
- Moeka Tanabe
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
- Laboratory of Health and Medical Science Innovation, Fujita Health University Graduate School of Medical Science, Toyoake, Aichi, Japan
| | - Kazuo Kunisawa
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan.
- International Center for Brain Science (ICBS), Fujita Health University, Toyoake, Aichi, Japan.
| | - Imari Saito
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
| | - Aika Kosuge
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
| | - Hiroyuki Tezuka
- Department of Cellular Function Analysis, Research Promotion Headquarters, Fujita Health University, Toyoake, Aichi, Japan
| | - Tomoki Kawai
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
| | - Yuki Kon
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
| | - Koyo Yoshidomi
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
| | - Akari Kagami
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
| | - Masaya Hasegawa
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
| | - Hisayoshi Kubota
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
- Division of Behavioral Neuropharmacology, International Center for Brain Science (ICBS), Fujita Health University, Toyoake, Aichi, Japan
| | - Haruto Ojika
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan
| | - Tadashi Fujii
- Department of Medical Research on Prebiotics and Probiotics, Fujita Health University, Toyoake, Aichi, Japan
- Department of Gastroenterology and Hepatology, Fujita Health University, Toyoake, Aichi, Japan
| | - Takumi Tochio
- Department of Medical Research on Prebiotics and Probiotics, Fujita Health University, Toyoake, Aichi, Japan
- Department of Gastroenterology and Hepatology, Fujita Health University, Toyoake, Aichi, Japan
| | - Yoshiki Hirooka
- Department of Medical Research on Prebiotics and Probiotics, Fujita Health University, Toyoake, Aichi, Japan
- Department of Gastroenterology and Hepatology, Fujita Health University, Toyoake, Aichi, Japan
| | - Kuniaki Saito
- Laboratory of Health and Medical Science Innovation, Fujita Health University Graduate School of Medical Science, Toyoake, Aichi, Japan
- Advanced Diagnostic System Research Laboratory, Fujita Health University Graduate School of Medical Science, Toyoake, Aichi, Japan
| | - Toshitaka Nabeshima
- Laboratory of Health and Medical Science Innovation, Fujita Health University Graduate School of Medical Science, Toyoake, Aichi, Japan
- International Center for Brain Science (ICBS), Fujita Health University, Toyoake, Aichi, Japan
- Japanese Drug Organization of Appropriate Use and Research (J-DO), Nagoya, Aichi, Japan
| | - Akihiro Mouri
- Department of Regulatory Science for Evaluation & Development of Pharmaceuticals & Devices, Fujita Health University Graduate School of Medical Sciences, Toyoake, Aichi, Japan.
- International Center for Brain Science (ICBS), Fujita Health University, Toyoake, Aichi, Japan.
- Japanese Drug Organization of Appropriate Use and Research (J-DO), Nagoya, Aichi, Japan.
| |
Collapse
|
8
|
Rukavina Mikusic NL, Prince PD, Choi MR, Chuffa LGA, Simão VA, Castro C, Manucha W, Quesada I. Microbiota, mitochondria, and epigenetics in health and disease: converging pathways to solve the puzzle. Pflugers Arch 2025; 477:635-655. [PMID: 40111427 DOI: 10.1007/s00424-025-03072-w] [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: 07/05/2024] [Revised: 02/23/2025] [Accepted: 02/25/2025] [Indexed: 03/22/2025]
Abstract
Dysbiosis, which refers to an imbalance in the composition of the gut microbiome, has been associated with a range of metabolic disorders, including type 2 diabetes, obesity, and metabolic syndrome. Although the exact mechanisms connecting gut dysbiosis to these conditions are not fully understood, various lines of evidence strongly suggest a substantial role for the interaction between the gut microbiome, mitochondria, and epigenetics. Current studies suggest that the gut microbiome has the potential to affect mitochondrial function and biogenesis through the production of metabolites. A well-balanced microbiota plays a pivotal role in supporting normal mitochondrial and cellular functions by providing metabolites that are essential for mitochondrial bioenergetics and signaling pathways. Conversely, in the context of illnesses, an unbalanced microbiota can impact mitochondrial function, leading to increased aerobic glycolysis, reduced oxidative phosphorylation and fatty acid oxidation, alterations in mitochondrial membrane permeability, and heightened resistance to cellular apoptosis. Mitochondrial activity can also influence the composition and function of the gut microbiota. Because of the intricate interplay between nuclear and mitochondrial communication, the nuclear epigenome can regulate mitochondrial function, and conversely, mitochondria can produce metabolic signals that initiate epigenetic changes within the nucleus. Given the epigenetic modifications triggered by metabolic signals from mitochondria in response to stress or damage, targeting an imbalanced microbiota through interventions could offer a promising strategy to alleviate the epigenetic alterations arising from disrupted mitochondrial signaling.
Collapse
Affiliation(s)
- Natalia Lucia Rukavina Mikusic
- Instituto Alberto C. Taquini de Investigaciones en Medicina Traslacional (IATIMET) CONICET, Universidad de Buenos Aires, 1122, Buenos Aires, Argentina
- Departamento de Ciencias Biológicas, Cátedra de Anatomía E Histología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, 1113, Buenos Aires, Argentina
| | - Paula Denise Prince
- Instituto Alberto C. Taquini de Investigaciones en Medicina Traslacional (IATIMET) CONICET, Universidad de Buenos Aires, 1122, Buenos Aires, Argentina
- Departamento de Ciencias Químicas, Cátedra de Fisicoquímica, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, 1113, Buenos Aires, Argentina
| | - Marcelo Roberto Choi
- Instituto Alberto C. Taquini de Investigaciones en Medicina Traslacional (IATIMET) CONICET, Universidad de Buenos Aires, 1122, Buenos Aires, Argentina.
- Departamento de Ciencias Biológicas, Cátedra de Anatomía E Histología, Facultad de Farmacia y Bioquímica, Universidad de Buenos Aires, 1113, Buenos Aires, Argentina.
| | - Luiz Gustavo A Chuffa
- Department of Structural and Functional Biology, Institute of Biosciences, UNESP - São Paulo State University, P.O. Box 18618-689, Botucatu, São Paulo, Zip Code 510, Brazil
| | - Vinícius Augusto Simão
- Department of Structural and Functional Biology, Institute of Biosciences, UNESP - São Paulo State University, P.O. Box 18618-689, Botucatu, São Paulo, Zip Code 510, Brazil
| | - Claudia Castro
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU) CONICET-Universidad Nacional de Cuyo, Mendoza, Argentina
| | - Walter Manucha
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU) CONICET-Universidad Nacional de Cuyo, Mendoza, Argentina.
- Laboratorio de Farmacología Básica y Traslacional, Área de Farmacología, Departamento de Patología, Facultad de Ciencias Médicas, Universidad Nacional de Cuyo, 5500, Mendoza, Argentina.
| | - Isabel Quesada
- Instituto de Medicina y Biología Experimental de Cuyo (IMBECU) CONICET-Universidad Nacional de Cuyo, Mendoza, Argentina.
| |
Collapse
|
9
|
Lemons JMS, Narrowe AB, Firrman J, Mahalak KK, Liu L, Higgins S, Moustafa AM, Baudot A, Deyaert S, Van den Abbeele P. The food additive butylated hydroxyanisole minimally affects the human gut microbiome ex vivo. Food Chem 2025; 473:143037. [PMID: 39919360 DOI: 10.1016/j.foodchem.2025.143037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 01/15/2025] [Accepted: 01/21/2025] [Indexed: 02/09/2025]
Abstract
Butylated hydroxyanisole (BHA) continues to raise consumer concerns. All previous evaluations of this additive have failed to consider its effect on the gut microbiome, even though it enters the colon. An ex vivo model was used to assess the effect of BHA on microbial communities from 24 donors, aged infants to older adults. A dose of 0.35 g/L BHA elicited no statistically significant changes in the functional outputs or community structure for any age group. Although not large enough to affect community diversity, there were some significant decreases at the phylum level. Among the genes most significantly affected by treatment with BHA across age groups are those involved in lipopolysaccharide synthesis and bacterial electron transport encoded by Bacteroidota, Proteobacteria, and Verrucomicrobiota. Given what is known about the intracellular activity of BHA, these genes may hint at a mechanism behind BHA's evident, but minimally detrimental effect on the gut microbiota.
Collapse
Affiliation(s)
- Johanna M S Lemons
- United States Department of Agriculture, Agricultural Research Service, Eastern Regional Research Center, Dairy and Functional Foods Research Unit, 600 East Mermaid Lane, Wyndmoor, PA 19038, USA.
| | - Adrienne B Narrowe
- United States Department of Agriculture, Agricultural Research Service, Eastern Regional Research Center, Dairy and Functional Foods Research Unit, 600 East Mermaid Lane, Wyndmoor, PA 19038, USA
| | - Jenni Firrman
- United States Department of Agriculture, Agricultural Research Service, Eastern Regional Research Center, Dairy and Functional Foods Research Unit, 600 East Mermaid Lane, Wyndmoor, PA 19038, USA
| | - Karley K Mahalak
- United States Department of Agriculture, Agricultural Research Service, Eastern Regional Research Center, Dairy and Functional Foods Research Unit, 600 East Mermaid Lane, Wyndmoor, PA 19038, USA
| | - LinShu Liu
- United States Department of Agriculture, Agricultural Research Service, Eastern Regional Research Center, Dairy and Functional Foods Research Unit, 600 East Mermaid Lane, Wyndmoor, PA 19038, USA
| | - Stephanie Higgins
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA
| | - Ahmed M Moustafa
- Division of Gastroenterology, Hepatology, and Nutrition, The Children's Hospital of Philadelphia, Philadelphia, PA 19104, USA; Department of Pediatrics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Center for Microbial Medicine, Children's Hospital of Philadelphia, Philadelphia, PA, USA
| | - Aurélien Baudot
- Cryptobiotix, Technologiepark-Zwijnaarde 82, Ghent 9052, Belgium
| | - Stef Deyaert
- Cryptobiotix, Technologiepark-Zwijnaarde 82, Ghent 9052, Belgium
| | | |
Collapse
|
10
|
Chen Z, Xiao C, Zhang J, Jian S, Li P, Lin J, He C, Chen Z, Qi Y, Shi J, Chen Q, Chen J, Bo H. The Impact of Diet on the Colonization of Beneficial Microbes from an Ecological Perspective. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025. [PMID: 40234746 DOI: 10.1021/acs.jafc.5c02086] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/17/2025]
Abstract
With growing recognition of the pivotal role of gut microbiota in human health, probiotics have gained widespread attention for their potential to restore microbial homeostasis. However, a critical challenge persists: limited colonization efficiency among most probiotic strains compromises their therapeutic efficacy. This overview synthesizes ecological principles with cutting-edge microbiome research to elucidate the dynamic interplay between dietary components and probiotic colonization within the intestinal niche. This overview systematically analyzes: (1) stage-specific colonization mechanisms spanning microbial introduction, establishment, and proliferation; (2) nutrient-driven modulation of gut microbiota composition and function; and (3) the dual role of common dietary patterns as both facilitators and disruptors of probiotic persistence. Notably, this overview identifies key dietary strategies, including precision delivery of prebiotic fibers and polyphenol-microbiota crosstalk, that enhance niche adaptation through pH optimization, adhesion potentiation, and competitive exclusion of pathogens. Furthermore, this overview critically evaluates current limitations in probiotic research, particularly strain-specific variability and methodological constraints in simulating host-microbe-diet tripartite interactions. To bridge these gaps, this overview proposes an interdisciplinary framework integrating omics-driven strain selection, engineered delivery systems, and personalized nutrition models. Collectively, this work advances a mechanistic understanding of diet-microbiota interactions while providing actionable insights for developing targeted probiotic therapies and evidence-based dietary interventions to optimize gut ecosystem resilience.
Collapse
Affiliation(s)
- Zelin Chen
- School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, 510006 Guangzhou, Guangdong China
| | - Chuntao Xiao
- School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, 510006 Guangzhou, Guangdong China
| | - Jiantang Zhang
- School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, 510006 Guangzhou, Guangdong China
| | - Shiqi Jian
- School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, 510006 Guangzhou, Guangdong China
| | - Pinyue Li
- School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, 510006 Guangzhou, Guangdong China
| | - Jiayi Lin
- School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, 510006 Guangzhou, Guangdong China
| | - Cai He
- School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, 510006 Guangzhou, Guangdong China
| | - Zixia Chen
- School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, 510006 Guangzhou, Guangdong China
| | - Yutong Qi
- School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, 510006 Guangzhou, Guangdong China
| | - Jingwen Shi
- School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, 510006 Guangzhou, Guangdong China
| | - Qizhu Chen
- School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, 510006 Guangzhou, Guangdong China
| | - Jun Chen
- College of Pharmacy, Guangdong Pharmaceutical University, 510006 Guangzhou, Guangdong China
| | - Huaben Bo
- School of Bioscience and Biopharmaceutics, Guangdong Pharmaceutical University, 510006 Guangzhou, Guangdong China
| |
Collapse
|
11
|
Kamp DL, Kerwin AH, McAnulty SJ, Nyholm SV. Organ structure and bacterial microbiogeography in a reproductive organ of the Hawaiian bobtail squid reveal dimensions of a defensive symbiosis. Appl Environ Microbiol 2025:e0216324. [PMID: 40231847 DOI: 10.1128/aem.02163-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2024] [Accepted: 03/09/2025] [Indexed: 04/16/2025] Open
Abstract
Many plants and animals house symbiotic microorganisms in specialized tissues or organs. Here, we used multidimensional in situ imaging techniques to illuminate how host organ structure and bacterial microbiogeography contribute to the symbiotic function of an organ in the Hawaiian bobtail squid, Euprymna scolopes. Along with the well-studied light organ, female E. scolopes harbor a community of bacteria in the accessory nidamental gland (ANG). The ANG is a dense network of epithelium-lined tubules, some of which are dominated by a single bacterial taxon. These bacteria are deposited into squid eggs, where they defend the developing embryos from harmful biofouling. This study used a combination of imaging techniques to visualize different dimensions of the ANG and its bacterial communities. Imaging entire organs with light sheet microscopy revealed that the ANG is a composite tissue of individual, non-intersecting tubules that each harbor their own bacterial population. The organ is bisected, with tubules converging toward two points at the posterior end. At these points, tubules empty into a space where bacteria can mix with squid jelly to be deposited onto eggs. Observations of the symbiotic community correlated bacterial taxa with cell morphology and revealed that tubule populations varied: some tubules contained populations of mixed taxa, whereas others contained only one bacterial genus. Together, these data shed light on how bacterial populations interact within the ANG and how the host uses physical structure to maintain and employ a symbiotic bacterial population in a defensive context.IMPORTANCESequence-based microbiome studies have revealed much about how hosts interact with communities of symbiotic microbiota but often lack a spatial understanding of how microbes relate to each other and the host in which they reside. This study uses a combination of microscopy techniques to reveal how the structure of a symbiotic organ in the female bobtail squid, Euprymna scolopes, houses diverse, beneficial bacterial populations and deploys them for egg defense. These findings suggest that spatial partitioning may be key to harboring a diverse population of antimicrobial-producing bacteria and establishing a foundation for further understanding how host structures mediate symbiotic interactions.
Collapse
Affiliation(s)
- Derrick L Kamp
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut, USA
| | - Allison H Kerwin
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut, USA
- Department of Biology, McDaniel College, Westminster, Maryland, USA
| | - Sarah J McAnulty
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut, USA
- Skype a Scientist, Philadelphia, Pennsylvania, USA
| | - Spencer V Nyholm
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut, USA
| |
Collapse
|
12
|
Wasim R, Sumaiya, Ahmad A, Anwar A, Salman A. Microbial imbalance in the gut: a new frontier in Rheumatoid arthritis research. Inflammopharmacology 2025:10.1007/s10787-025-01737-7. [PMID: 40220199 DOI: 10.1007/s10787-025-01737-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2025] [Accepted: 03/27/2025] [Indexed: 04/14/2025]
Abstract
A chronic autoimmune illness that causes joint destruction and inflammation, rheumatoid arthritis (RA) often results in disability. Genetic, environmental, and immune system variables all have a role in the pathophysiology of RA. The complex community of bacteria that live in the gastrointestinal system, known as the gut microbiota, has been implicated in the onset and progression of RA in recent years, according to mounting data. An imbalance in the gut microbiota's composition, known as dysbiosis, has been noted in RA patients. This imbalance may impact inflammatory pathways and immunological responses, which in turn may contribute to the development and severity of the illness. Research has shown that some bacterial species, including Firmicutes, Bacteroidetes, and Proteobacteria, are either more abundant or less prevalent in RA patients than in healthy people. The gut-immune system axis may be modulated, immunological tolerance may be affected, and pro-inflammatory cytokine production may be enhanced by these microbial changes, all of which may lead to systemic inflammation linked to RA. Moreover, changes in intestinal permeability and a rise in microbial metabolite translocation may make autoimmune reactions worse. Probiotics, antibiotics, and dietary changes have also been investigated as possible treatment approaches to help RA patients regain the balance of their gut microbiota. Still up for debate, however, are the precise ways in which the gut microbiome affects RA. Comprehending the complex connection between gut microbiota and RA may give new perspectives on managing and preventing the condition, as well as future prospects for medicines that target the microbiome.
Collapse
Affiliation(s)
- Rufaida Wasim
- Department of Pharmacology, Faculty of Pharmacy, Integral University, Lucknow, UP, 226022, India.
| | - Sumaiya
- Career Post Graduate Institute of Dental Sciences and Hospital, Lucknow, India
| | - Asad Ahmad
- Department of Pharmacology, Faculty of Pharmacy, Integral University, Lucknow, UP, 226022, India
| | - Aamir Anwar
- Department of Pharmacology, Faculty of Pharmacy, Integral University, Lucknow, UP, 226022, India
| | - Aimen Salman
- Department of Pharmacology, Faculty of Pharmacy, Integral University, Lucknow, UP, 226022, India
| |
Collapse
|
13
|
Chesneau G, Herpell J, Wolf SM, Perin S, Hacquard S. MetaFlowTrain: a highly parallelized and modular fluidic system for studying exometabolite-mediated inter-organismal interactions. Nat Commun 2025; 16:3310. [PMID: 40210863 PMCID: PMC11985495 DOI: 10.1038/s41467-025-58530-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Accepted: 03/25/2025] [Indexed: 04/12/2025] Open
Abstract
Metabolic fluxes between cells, organisms, or communities drive ecosystem assembly and functioning and explain higher-level biological organization. Exometabolite-mediated inter-organismal interactions, however, remain poorly described due to technical challenges in measuring these interactions. Here, we present MetaFlowTrain, an easy-to-assemble, cheap, semi-high-throughput, and modular fluidic system in which multiple media can be flushed at adjustable flow rates into gnotobiotic microchambers accommodating diverse micro-organisms, ranging from bacteria to small eukaryotes. These microchambers can be used alone or connected in series to create microchamber trains within which metabolites, but not organisms, directionally travel between microchambers to modulate organismal growth. Using MetaFlowTrain, we uncover soil conditioning effects on synthetic community structure and plant growth, and reveal microbial antagonism mediated by exometabolite production. Our study highlights MetaFlowTrain as a versatile system for investigating plant-microbe-microbe metabolic interactions. We also discuss the system´s potential to discover metabolites that function as signaling molecules, drugs, or antimicrobials across various systems.
Collapse
Affiliation(s)
- Guillaume Chesneau
- Department of Plant Microbe Interactions, Max Planck Institute for Plant Breeding Research, Cologne, Germany
| | - Johannes Herpell
- Department of Plant Microbe Interactions, Max Planck Institute for Plant Breeding Research, Cologne, Germany
| | - Sarah Marie Wolf
- Department of Plant Microbe Interactions, Max Planck Institute for Plant Breeding Research, Cologne, Germany
| | - Silvina Perin
- Department of Plant Microbe Interactions, Max Planck Institute for Plant Breeding Research, Cologne, Germany
| | - Stéphane Hacquard
- Department of Plant Microbe Interactions, Max Planck Institute for Plant Breeding Research, Cologne, Germany.
- Cluster of Excellence on Plant Sciences (CEPLAS), Max Planck Institute for Plant Breeding Research, Cologne, Germany.
| |
Collapse
|
14
|
Quan X, Miao Z, Han R, Deng R, Cao Y, Tian J, Lu Y, Wang G, Yu X, Wu Y, Dai C. Proteomic analysis reveals that Acalypha australis L. mitigates chronic colitis by modulating the FABP4/PPARγ/NF-κB signaling pathway. JOURNAL OF ETHNOPHARMACOLOGY 2025; 345:119585. [PMID: 40049341 DOI: 10.1016/j.jep.2025.119585] [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: 12/18/2024] [Revised: 02/28/2025] [Accepted: 03/02/2025] [Indexed: 03/14/2025]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Acalypha australis L. (AAL), a traditional medicinal herb from the Euphorbiaceae family, has been widely used in Chinese medicine for its heat-clearing, detoxifying, and diuretic properties, as well as for treating gastrointestinal disorders such as diarrhea and dysentery. Its reported anti-inflammatory and hemostatic effects are closely linked to inflammatory pathways. While previous studies have demonstrated AAL's efficacy in acute colitis, its therapeutic potential in chronic colitis and the underlying mechanisms remain largely unexplored. AIM OF THE STUDY This study aims to investigate the therapeutic efficacy of AAL in dextran sulfate sodium (DSS)-induced chronic colitis and elucidate its anti-inflammatory and barrier-protective mechanisms, with a specific focus on the FABP4/PPARγ/NF-κB signaling pathway. MATERIALS AND METHODS The chemical composition of AAL was characterized using ultra-high-performance liquid chromatography coupled with quadrupole time-of-flight mass spectrometry (UPLC-QTOF-MS). Chronic colitis was induced in mice through three cycles of DSS administration, and the therapeutic effects of AAL were evaluated by assessing body weight, Disease Activity Index (DAI), colon length, and pathological alterations. Enzyme-linked immunosorbent assay (ELISA) was used to quantify inflammatory cytokine levels. Immunohistochemistry and Western blotting were performed to assess mucosal barrier proteins, including Mucin 2 (MUC2), zonula occludens-1 (ZO-1), and Occludin, as well as key signaling proteins such as fatty acid-binding protein 4 (FABP4), peroxisome proliferator-activated receptor gamma (PPARγ), and phosphorylated P65 (p-P65). Proteomic analysis combined with Gene Set Enrichment Analysis (GSEA) was conducted to identify differentially expressed proteins and enriched pathways. The role of the FABP4/PPARγ/NF-κB axis was further validated using the PPARγ antagonist GW9662. Additionally, molecular docking and molecular dynamics simulations were employed to identify bioactive components in AAL and their interactions with FABP4 and PPARγ. RESULTS UPLC-QTOF-MS analysis identified 47 compounds in AAL, including flavonoids, terpenoids, and polyphenols. Bergaptol and corilagin were identified as major constituents with potential anti-inflammatory properties. AAL treatment significantly alleviated chronic colitis symptoms, as evidenced by reduced DAI scores, restoration of body weight, and improved colon length. Pathological and immunohistochemical analyses demonstrated that AAL preserved intestinal mucosal integrity by upregulating MUC2, ZO-1, and Occludin expression. Proteomic and GSEA analyses identified the FABP4/PPARγ/NF-κB pathway as a key target of AAL. Western blotting confirmed that AAL suppressed FABP4 expression, enhanced PPARγ levels, and reduced p-P65 expression, indicating inhibition of NF-κB activation. Notably, the therapeutic effects of AAL were abolished by GW9662, further validating the involvement of PPARγ signaling. Molecular docking and molecular dynamics simulations demonstrated strong binding affinities of bergaptol and corilagin to FABP4 and PPARγ, suggesting their role as active compounds responsible for AAL's therapeutic effects. CONCLUSIONS AAL effectively mitigates chronic colitis by preserving intestinal barrier integrity, suppressing inflammatory responses, and modulating the FABP4/PPARγ/NF-κB pathway. The bioactive compounds bergaptol and corilagin may contribute to these therapeutic effects, highlighting AAL as a promising natural therapeutic agent for ulcerative colitis.
Collapse
Affiliation(s)
- Xiaoyu Quan
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Zhiwei Miao
- Department of Gastroenterology, Zhangjiagang TCM Hospital Affiliated to Nanjing University of Chinese Medicine, Zhangjiagang, 215600, China
| | - Runxi Han
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Rui Deng
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yaqi Cao
- College of Food Science and Technology, Nanjing Agricultural University, Nanjing, 210095, China
| | - Jingshan Tian
- College of Life Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - Yaping Lu
- College of Life Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - Guoxiang Wang
- College of Life Science, Nanjing Agricultural University, Nanjing, 210095, China
| | - Xingjian Yu
- Department of Biochemistry and Molecular Medicine, School of Medicine, University of California, Davis, Sacramento, 95817, CA, USA
| | - Yi Wu
- College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, 210095, China
| | - Chen Dai
- College of Life Science, Nanjing Agricultural University, Nanjing, 210095, China.
| |
Collapse
|
15
|
Lin Y, Lau HCH, Liu C, Ding X, Sun Y, Rong J, Zhang X, Wang L, Yuan K, Miao Y, Wu WKK, Wong SH, Sung JJY, Yu J. Multi-cohort analysis reveals colorectal cancer tumor location-associated fecal microbiota and their clinical impact. Cell Host Microbe 2025; 33:589-601.e3. [PMID: 40209677 DOI: 10.1016/j.chom.2025.03.012] [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: 10/21/2024] [Revised: 02/12/2025] [Accepted: 03/11/2025] [Indexed: 04/12/2025]
Abstract
Microbial alterations in different tumor locations of colorectal cancer (CRC) remain unclear. Here, 1,375 fecal metagenomes from six in-house and published datasets were analyzed, including 128 right-sided CRC (rCRC), 168 left-sided CRC (lCRC), 250 rectal cancer (RC), and 829 controls. Firmicutes progressively increase from rCRC, lCRC, to RC, in contrast to the gradual decrease of Bacteroidetes. Tumor location-associated fecal microbes are identified, including Veillonella parvula for rCRC, Streptococcus angionosus for lCRC, and Peptostreptococcus anaerobius for RC, while Fusobacterium nucleatum is enriched in all tumor locations. Tumor location-associated bacteria correlate with patient survival. Clinically, we establish a microbial biomarker panel for each tumor location that accurately diagnoses rCRC (area under the receiver operating characteristic curve [AUC] = 91.59%), lCRC (AUC = 91.69%), or RC (AUC = 90.53%) from controls. Tumor location-specific biomarkers also have higher diagnostic accuracy (AUC = 91.38%) than location-non-specific biomarkers (AUC = 82.92%). Overall, we characterize fecal microbes associated with different CRC tumor locations, highlighting that tumor location should be considered in non-invasive diagnosis.
Collapse
Affiliation(s)
- Yufeng Lin
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Harry Cheuk-Hay Lau
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Chuanfa Liu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Xiao Ding
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yang Sun
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Yunnan Province Clinical Research Center for Digestive Disease, Yunnan Geriatric Medical Center, Kunming, Yunnan, China
| | - Jiamei Rong
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Yunnan Province Clinical Research Center for Digestive Disease, Yunnan Geriatric Medical Center, Kunming, Yunnan, China
| | - Xiang Zhang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Luyao Wang
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Kai Yuan
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Yinglei Miao
- Department of Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Yunnan Province Clinical Research Center for Digestive Disease, Yunnan Geriatric Medical Center, Kunming, Yunnan, China
| | - William Ka-Kei Wu
- Department of Anaesthesia and Intensive Care and Peter Hung Pain Research Institute, State Key Laboratory of Digestive Disease, The Chinese University of Hong Kong, Hong Kong SAR, China
| | - Sunny Hei Wong
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Joseph Jao-Yiu Sung
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
| | - Jun Yu
- Institute of Digestive Disease and Department of Medicine and Therapeutics, State Key Laboratory of Digestive Disease, Li Ka Shing Institute of Health Sciences, The Chinese University of Hong Kong, Hong Kong SAR, China.
| |
Collapse
|
16
|
Zhang H, Zhou Y, Jiang YH, Hu WP, Huang LL, Lin HX, Zuo ZG, Du JM, Lou YL. Altered microbiota of rectal mucosa in rectal cancer patients. World J Gastroenterol 2025; 31:105248. [PMID: 40248061 PMCID: PMC12001164 DOI: 10.3748/wjg.v31.i13.105248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/16/2025] [Revised: 02/27/2025] [Accepted: 03/21/2025] [Indexed: 04/02/2025] Open
Abstract
BACKGROUND With advances in sequencing techniques, microbiota dysbiosis and pathogenic microbes that accelerate colorectal cancer progression have been identified and widely reported. However, few studies have focused on the microbiota taxa of rectal mucus in rectal cancer (RC) patients. Here, we analyzed the composition and characteristics of the rectal mucosa microbiota of RC patients from Wenzhou city, China, and compared the results with those of healthy controls. AIM To explore the changes in the characteristics of the rectal mucosal flora associated with RC, and identify biomarkers of microbe taxa for RC. METHODS Rectal mucosa samples from a Chinese cohort of 72 recently diagnosed RC patients and 71 healthy controls were obtained. A validation cohort, which included 22 RC patients and 60 healthy controls, was also established. Changes in the rectal mucosal flora were observed by cultivation, 16S ribosomal DNA gene sequencing analysis and quantitative polymerase chain reaction analysis. RESULTS The 16S ribosomal DNA results demonstrated that RC patients presented increased bacterial community richness and alpha diversity as well as an altered rectal mucosal microbiota, with depletion of Proteobacteria and Thermi and enrichment of Bacteroidetes and Fusobacteria in cancerous mucosal tissues (CM) and enrichment of Firmicutes and Cyanobacteria in adjacent noncancerous mucosal tissues (AM). The culture results showed that the mean loads of Escherichia coli, Bifidobacterium, Enterococcus, and Lactobacillus were significantly reduced in RC patients. The ratios of Prevotella to Ruminococcus [areas under the receiver operating curve: 0.795 in AM vs normal control mucosa (NM), 0.77 in CM vs NM] and of Prevotella stercorea to Propionibacterium acnes (areas under the receiver operating curve: 0.808 in AM vs NM, 0.843 in CM vs NM) exhibited excellent abilities to differentiate between healthy controls and RC patients. CONCLUSION RC patients have an altered rectal mucosal microbiota, and the ratio of Prevotella to Ruminococcus or the ratio of Prevotella stercorea to Propionibacterium acnes may serve as a marker for RC diagnosis.
Collapse
Affiliation(s)
- Hao Zhang
- Department of Laboratory Medicine, Hangzhou Geriatric Hospital, Hangzhou 310022, Zhejiang Province, China
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Yan Zhou
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - You-Heng Jiang
- Tomas Lindahl Nobel Laureate Laboratory, The Seventh Affiliated Hospital of Sun Yat-sen University, Shenzhen 518107, Guangdong Province, China
| | - Wan-Ping Hu
- Department of Colorectal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Lu-Lu Huang
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Hai-Xia Lin
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Zhi-Gui Zuo
- Department of Colorectal Surgery, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Ji-Mei Du
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| | - Yong-Liang Lou
- School of Laboratory Medicine and Life Sciences, Wenzhou Medical University, Wenzhou 325035, Zhejiang Province, China
| |
Collapse
|
17
|
Fruet C, Müller EL, Loverdo C, Bitbol AF. Spatial structure facilitates evolutionary rescue by drug resistance. PLoS Comput Biol 2025; 21:e1012861. [PMID: 40179127 PMCID: PMC11967957 DOI: 10.1371/journal.pcbi.1012861] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Accepted: 02/09/2025] [Indexed: 04/05/2025] Open
Abstract
Bacterial populations often have complex spatial structures, which can impact their evolution. Here, we study how spatial structure affects the evolution of antibiotic resistance in a bacterial population. We consider a minimal model of spatially structured populations where all demes (i.e., subpopulations) are identical and connected to each other by identical migration rates. We show that spatial structure can facilitate the survival of a bacterial population to antibiotic treatment, starting from a sensitive inoculum. Specifically, the bacterial population can be rescued if antibiotic resistant mutants appear and are present when drug is added, and spatial structure can impact the fate of these mutants and the probability that they are present. Indeed, the probability of fixation of neutral or deleterious mutations providing drug resistance is increased in smaller populations. This promotes local fixation of resistant mutants in the structured population, which facilitates evolutionary rescue by drug resistance in the rare mutation regime. Once the population is rescued by resistance, migrations allow resistant mutants to spread in all demes. Our main result that spatial structure facilitates evolutionary rescue by antibiotic resistance extends to more complex spatial structures, and to the case where there are resistant mutants in the inoculum.
Collapse
Affiliation(s)
- Cecilia Fruet
- Institute of Bioengineering, School of Life Sciences, ÉcolePolytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- SIB SwissInstitute of Bioinformatics, Lausanne, Switzerland
| | - Ella Linxia Müller
- Institute of Bioengineering, School of Life Sciences, ÉcolePolytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- SIB SwissInstitute of Bioinformatics, Lausanne, Switzerland
| | - Claude Loverdo
- Sorbonne Université, CNRS,Institut de Biologie Paris-Seine (IBPS), Laboratoire Jean Perrin (LJP), Paris,France
| | - Anne-Florence Bitbol
- Institute of Bioengineering, School of Life Sciences, ÉcolePolytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
- SIB SwissInstitute of Bioinformatics, Lausanne, Switzerland
| |
Collapse
|
18
|
Zhang Z, Liu C, Zhao L, Yao J. Systems biology of dry eye: Unraveling molecular mechanisms through multi-omics integration. Ocul Surf 2025; 36:25-40. [PMID: 39746576 DOI: 10.1016/j.jtos.2024.12.010] [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: 10/27/2024] [Revised: 12/15/2024] [Accepted: 12/30/2024] [Indexed: 01/04/2025]
Abstract
Dry eye disease (DED) is a multifactorial condition with complex and incompletely understood molecular mechanisms. Advances in multi-omics technologies, including genomics, transcriptomics, proteomics, metabolomics, and microbiomics, have provided new insights into the pathophysiology of DED. Genomic analyses have identified key genetic variants linked to immune regulation and lacrimal gland function. Transcriptomic studies reveal upregulated inflammatory pathways in ocular surface tissues, implicating these as core drivers of chronic inflammation. Proteomic research highlights significant alterations in tear protein composition, especially proteins involved in inflammation and tissue repair. Metabolomics studies focus on disrupted lipid metabolism and oxidative stress, which are crucial in maintaining tear film stability. Furthermore, microbiome research has demonstrated reduced microbial diversity and increased pathogenic bacteria, exacerbating inflammatory responses. The integration of multi-omics data allows for the identification of novel biomarkers and therapeutic targets, enabling precision diagnostics and personalized treatments. Therefore, this review highlights the critical importance of multi-omics approaches in deepening our understanding of DED's complex molecular mechanisms and their potential to transform clinical management and therapeutic innovations in this challenging field.
Collapse
Affiliation(s)
- Zhirui Zhang
- Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Changxing Liu
- Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Lingying Zhao
- Heilongjiang University of Chinese Medicine, Harbin, 150040, China
| | - Jing Yao
- The First Hospital Affiliated to Heilongjiang University of Chinese Medicine, Harbin, 150040, China.
| |
Collapse
|
19
|
Du Y, Zhou Y, Lin H, Yang C, Wang W. Three-Dimensional Imaging of Native Microbiota in Intact Colon. Anal Chem 2025; 97:6571-6577. [PMID: 40108811 DOI: 10.1021/acs.analchem.4c06059] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
Mapping the spatial locations of gut bacteria in their native environment enhances our understanding of bacteria-host interactions and the physiological and pathological roles these microbes play. However, the intricate composition of bacterial communities in millimeter-scale intestinal tissues presents a great challenge for in situ imaging of their spatial distributions. To address this, we introduce a three-dimensional (3D) imaging strategy that combines a fluorescent tetrapeptide (TetraAA-AcLys) metabolic labeling probe with a tissue clearing protocol. This method enables high-resolution visualization of the microbiota within intact colon, allowing for clear observation of the 3D distribution of gut bacteria across various sections, without interference from host tissues. Moreover, 3D quantitative analysis of the labeled bacteria in a enteritis model reveals their penetration into the mucus layer in colon, highlighting the technique's potential for studying gut microbiota biogeography in health and disease. This 3D imaging method offers valuable spatial insights into the dynamic relationship between the microbial community and its host.
Collapse
Affiliation(s)
- Yahui Du
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Yingjun Zhou
- State Key Laboratory of Genetic Engineering, Department of Microbiology, Fudan Microbiome Center, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Huibin Lin
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Chaoyong Yang
- The MOE Key Laboratory of Spectrochemical Analysis and Instrumentation, Key Laboratory for Chemical Biology of Fujian Province, State Key Laboratory of Physical Chemistry of Solid Surfaces, Department of Chemical Biology, College of Chemistry and Chemical Engineering, Xiamen University, Xiamen 361005, China
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200127, China
| | - Wei Wang
- State Key Laboratory of Genetic Engineering, Department of Microbiology, Fudan Microbiome Center, School of Life Sciences, Fudan University, Shanghai 200438, China
| |
Collapse
|
20
|
Fan X, Zhou Y, Bai W, Li X, Lin L, Lin H, Yang M, Yu X, Wang J, Lin L, Wang W. Intravital imaging of translocated bacteria via fluorogenic labeling of gut microbiota in situ. Proc Natl Acad Sci U S A 2025; 122:e2415845122. [PMID: 40153461 PMCID: PMC12002288 DOI: 10.1073/pnas.2415845122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2024] [Accepted: 02/19/2025] [Indexed: 03/30/2025] Open
Abstract
The translocation of bacteria from intestinal tracts into blood vessels and distal organs plays pivotal roles in the pathogenesis of numerous severe diseases. Intravital monitoring of bacterial translocation, however, is not yet feasible, which greatly hinders us from comprehending this spatially and temporally dynamic process. Here we report an in vivo fluorogenic labeling method, which enables in situ imaging of mouse gut microbiota and real-time tracking of the translocated bacteria. By mimicking the peptidoglycan stem peptide in bacteria, a tetrapeptide probe composed of alternating D- and L-amino acids and separately equipped with a fluorophore and a quencher on the N- and C-terminal amino acid, is designed. Because of its resistance to host proteases, it can be directly used in gavage and achieves fluorogenic labeling of the microbiota in the gut via the functioning of the L,D-transpeptidases of the labeled bacteria. Using intravital two-photon microscopy, we then successfully visualize the translocation of gut bacteria into the bloodstream and liver in obesity mouse models. This technique can help further exploration into the spatiotemporal activities of gut microbiota in vivo, and be valuable in investigating the less understood pathogenicity of bacterial translocation in many severe diseases.
Collapse
Affiliation(s)
- Xinqi Fan
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai200032, China
| | - Yingjun Zhou
- State Key Laboratory of Genetic Engineering, Department of Microbiology, Fudan Microbiome Center, School of Life Sciences, Fudan University, Shanghai200438, China
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200127, China
| | - Wenjuan Bai
- Shanghai Institute of Immunology, Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai200025, China
| | - Xue Li
- State Key Laboratory of Genetic Engineering, Department of Microbiology, Fudan Microbiome Center, School of Life Sciences, Fudan University, Shanghai200438, China
| | - Liyuan Lin
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200127, China
| | - Huibin Lin
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200127, China
| | - Ming Yang
- State Key Laboratory of Genetic Engineering, Department of Microbiology, Fudan Microbiome Center, School of Life Sciences, Fudan University, Shanghai200438, China
| | - Xiaofei Yu
- State Key Laboratory of Genetic Engineering, Department of Microbiology, Fudan Microbiome Center, School of Life Sciences, Fudan University, Shanghai200438, China
| | - Jing Wang
- Shanghai Institute of Immunology, Department of Microbiology and Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai200025, China
| | - Liang Lin
- State Key Laboratory of Chemical Biology, Shanghai Institute of Organic Chemistry, Chinese Academy of Sciences, Shanghai200032, China
| | - Wei Wang
- State Key Laboratory of Genetic Engineering, Department of Microbiology, Fudan Microbiome Center, School of Life Sciences, Fudan University, Shanghai200438, China
- Institute of Molecular Medicine, Shanghai Key Laboratory for Nucleic Acid Chemistry and Nanomedicine, Renji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai200127, China
| |
Collapse
|
21
|
Tufail MA, Schmitz RA. Exploring the Probiotic Potential of Bacteroides spp. Within One Health Paradigm. Probiotics Antimicrob Proteins 2025; 17:681-704. [PMID: 39377977 PMCID: PMC11925995 DOI: 10.1007/s12602-024-10370-9] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/25/2024] [Indexed: 03/21/2025]
Abstract
Probiotics are pivotal in maintaining or restoring the balance of human intestinal microbiota, a crucial factor in mitigating diseases and preserving the host's health. Exploration into Bacteroides spp. reveals substantial promise in their development as next-generation probiotics due to their profound interaction with host immune cells and capability to regulate the microbiome's metabolism by significantly impacting metabolite production. These beneficial bacteria exhibit potential in ameliorating various health issues such as intestinal disorders, cardiovascular diseases, behavioral disorders, and even cancer. Though it's important to note that a high percentage of them are as well opportunistic pathogens, posing risks under certain conditions. Studies highlight their role in modifying immune responses and improving health conditions by regulating lymphocytes, controlling metabolism, and preventing inflammation and cancer. The safety and efficacy of Bacteroides strains are currently under scrutiny by the European Commission for authorization in food processing, marking a significant step towards their commercialization. The recent advancements in bacterial isolation and sequencing methodologies, coupled with the integration of Metagenome-Assembled Genomes (MAGs) binning from metagenomics data, continue to unveil the potential of Bacteroides spp., aiding in the broader understanding and application of these novel probiotics in health and disease management.
Collapse
Affiliation(s)
- Muhammad Aammar Tufail
- Institut für Allgemeine Mikrobiologie, Christian-Albrechts-Universität zu Kiel, 24118, Kiel, Germany.
| | - Ruth A Schmitz
- Institut für Allgemeine Mikrobiologie, Christian-Albrechts-Universität zu Kiel, 24118, Kiel, Germany.
| |
Collapse
|
22
|
Tan X, Wu J, Zhang H, Li Y, Huang Y, Zheng P, Xie P. Biogeography of intestinal mucus-associated microbiome: Depletion of genus Pseudomonas is associated with depressive-like behaviors in female cynomolgus macaques. J Adv Res 2025; 70:393-404. [PMID: 38735389 PMCID: PMC11976423 DOI: 10.1016/j.jare.2024.05.013] [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: 10/25/2023] [Revised: 05/07/2024] [Accepted: 05/09/2024] [Indexed: 05/14/2024] Open
Abstract
INTRODUCTION Depression is a debilitating and poorly understood mental disorder. There is an urgency to explore new potential biological mechanisms of depression and the gut microbiota is a promising research area. OBJECTIVES Our study was aim to understand regional heterogeneity and potential molecular mechanisms underlying depression induced by dysbiosis of mucus-associated microbiota. METHODS Here, we only selected female macaques because they are more likely to form a natural social hierarchy in a harem-like environment. Because high-ranking macaques rarely displayed depressive-like behaviors, we selected seven monkeys from high-ranking individuals as control group (HC) and the same number of low-ranking ones as depressive-like group (DL), which displayed significant depressive-like behaviors. Then, we collected mucus from the duodenum, jejunum, ileum, cecum and colon of DL and HC monkeys for shotgun metagenomic sequencing, to profile the biogeography of mucus-associated microbiota along duodenum to colon. RESULTS Compared with HC, DL macaques displayed noticeable depressive-like behaviors such as longer duration of huddle and sit alone behaviors (negative emotion behaviors), and fewer duration of locomotion, amicable and ingestion activities (positive emotion behaviors). Moreover, the alpha diversity index (Chao) could predict aforementioned depressive-like behaviors along duodenum to colon. Further, we identified that genus Pseudomonas was consistently decreased in DL group throughout the entire intestinal tract except for the jejunum. Specifically, there were 10, 18 and 28 decreased Pseudomonas spp. identified in ileum, cecum and colon, respectively. Moreover, a bacterial module mainly composed of Pseudomonas spp. was positively associated with three positive emotion behaviors. Functionally, Pseudomonaswas mainly involved in microbiota derived lipid metabolisms such as PPAR signaling pathway, cholesterol metabolism, and fat digestion and absorption. CONCLUSION Different regions of intestinal mucus-associated microbiota revealed that depletion of genus Pseudomonas is associated with depressive-like behaviors in female macaques, which might induce depressive phenotypes through regulating lipid metabolism.
Collapse
Affiliation(s)
- Xunmin Tan
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Disease, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; The Jin Feng Laboratory, Chongqing, China
| | - Jing Wu
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Disease, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; The Jin Feng Laboratory, Chongqing, China
| | - Hanping Zhang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Disease, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; The Jin Feng Laboratory, Chongqing, China
| | - Yifan Li
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Disease, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; The Jin Feng Laboratory, Chongqing, China
| | - Yu Huang
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Disease, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; The Jin Feng Laboratory, Chongqing, China
| | - Peng Zheng
- Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Institute for Brain Science and Disease, Chongqing Medical University, Chongqing, China.
| | - Peng Xie
- NHC Key Laboratory of Diagnosis and Treatment on Brain Functional Disease, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; Department of Neurology, The First Affiliated Hospital of Chongqing Medical University, Chongqing, China; The Jin Feng Laboratory, Chongqing, China.
| |
Collapse
|
23
|
Unverdorben LV, Pirani A, Gontjes K, Moricz B, Holmes CL, Snitkin ES, Bachman MA. Klebsiella pneumoniae evolution in the gut leads to spontaneous capsule loss and decreased virulence potential. mBio 2025:e0236224. [PMID: 40162782 DOI: 10.1128/mbio.02362-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 02/26/2025] [Indexed: 04/02/2025] Open
Abstract
Klebsiella pneumoniae (Kp) is an opportunistic pathogen that poses a major threat in healthcare settings. The gut is a primary Kp reservoir in hospitalized patients, and colonization is a major risk factor for Kp infection. The stability of virulence determinants such as capsule and lipopolysaccharide during gut colonization is largely unexplored. In a murine gut colonization model, we demonstrated that spontaneous capsule loss occurs rapidly but varies by Kp pathotype. A classical Kp strain and a carbapenem-resistant strain of the epidemic sequence type 258 lineage had significant levels (median of 25% and 9.5%, respectively) of capsule loss. In contrast, a hypervirulent strain did not lose capsule to a significant degree (median 0.1%), despite readily losing capsule during in vitro passage. Insertion sequences (ISs) or mutations were found disrupting capsule operon genes of all isolates and in O-antigen encoding genes in a subset of isolates. Mouse-derived acapsular isolates from two pathotypes had significant fitness defects in a murine pneumonia model. Removal of the IS in the capsule operon in a mouse-derived acapsular classical isolate restored capsule production to wild-type levels. Genomic analysis of Klebsiella rectal isolates from hospitalized patients found that 18 of 245 strains (7%) had at least one IS disrupting the capsule operon. Combined, these data indicate that Kp capsule loss can occur during gut colonization in a strain-dependent manner, not only impacting strain virulence but also potentially altering patient infection risk. IMPORTANCE In hospitalized patients, gut colonization by the bacterial pathogen Klebsiella pneumoniae (Kp) is a major risk factor for the development of infections. The genome of Kp varies across isolates, and the presence of certain virulence genes is associated with the ability to progress from colonization to infection. Here, we identified that virulence genes encoding capsule and lipopolysaccharide, which normally protect bacteria from the immune system, are disrupted by mutations during murine gut colonization. These mutations occurred frequently in some isolates of Kp but not others, and these virulence gene mutants from the gut were defective in causing infections. An analysis of 245 human gut isolates demonstrated that this capsule loss also occurred in patients. This work highlights that mutations that decrease virulence occur during gut colonization, the propensity for these mutations differs by isolate, and that stability of virulence genes is important to consider when assessing infection risk in patients.
Collapse
Affiliation(s)
- Lavinia V Unverdorben
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Ali Pirani
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Kyle Gontjes
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Bridget Moricz
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Caitlyn L Holmes
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Evan S Snitkin
- Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| | - Michael A Bachman
- Department of Pathology, University of Michigan Medical School, Ann Arbor, Michigan, USA
| |
Collapse
|
24
|
Wang X, Zhao D, Bi D, Li L, Tian H, Yin F, Zuo T, Ianiro G, Li N, Chen Q, Qin H. Fecal microbiota transplantation: transitioning from chaos and controversial realm to scientific precision era. Sci Bull (Beijing) 2025; 70:970-985. [PMID: 39855927 DOI: 10.1016/j.scib.2025.01.029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2024] [Revised: 12/05/2024] [Accepted: 12/13/2024] [Indexed: 01/27/2025]
Abstract
With the popularization of modern lifestyles, the spectrum of intestinal diseases has become increasingly diverse, presenting significant challenges in its management. Traditional pharmaceutical interventions have struggled to keep pace with these changes, leaving many patients refractory to conventional pharmaceutical treatments. Fecal microbiota transplantation (FMT) has emerged as a promising therapeutic approach for enterogenic diseases. Still, controversies persist regarding its active constituents, mechanism of action, scheme of treatment evaluation, indications, and contraindications. In this review, we investigated the efficacy of FMT in addressing gastrointestinal and extraintestinal conditions, drawing from follow-up data on over 8000 patients. We systematically addressed the controversies surrounding FMT's clinical application. We delved into key issues such as its technical nature, evaluation methods, microbial restoration mechanisms, and impact on the host-microbiota interactions. Additionally, we explored the potential colonization patterns of FMT-engrafted new microbiota throughout the entire intestine and elucidated the specific pathways through which the new microbiota modulates host immunity, metabolism, and genome.
Collapse
Affiliation(s)
- Xinjun Wang
- Tenth People's Hospital of Tongji University, Shanghai 200072, China; Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215000, China; Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Shanghai Gastrointestinal Microecology Research Center, Shanghai 200072, China; Shanghai Institution of Gut Microbiota Research and Engineering Development, Shanghai 200072, China; Clinical Research Center for Digestive Diseases, Tongji University School of Medicine, Shanghai 200072, China.
| | - Di Zhao
- Tenth People's Hospital of Tongji University, Shanghai 200072, China; Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Shanghai Gastrointestinal Microecology Research Center, Shanghai 200072, China; Shanghai Institution of Gut Microbiota Research and Engineering Development, Shanghai 200072, China; Clinical Research Center for Digestive Diseases, Tongji University School of Medicine, Shanghai 200072, China
| | - Dexi Bi
- Department of Pathology, Tenth People's Hospital of Tongji University, Shanghai 200072, China
| | - Long Li
- Tenth People's Hospital of Tongji University, Shanghai 200072, China; Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Shanghai Gastrointestinal Microecology Research Center, Shanghai 200072, China; Shanghai Institution of Gut Microbiota Research and Engineering Development, Shanghai 200072, China; Clinical Research Center for Digestive Diseases, Tongji University School of Medicine, Shanghai 200072, China
| | - Hongliang Tian
- Tenth People's Hospital of Tongji University, Shanghai 200072, China; Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Shanghai Gastrointestinal Microecology Research Center, Shanghai 200072, China; Shanghai Institution of Gut Microbiota Research and Engineering Development, Shanghai 200072, China; Clinical Research Center for Digestive Diseases, Tongji University School of Medicine, Shanghai 200072, China
| | - Fang Yin
- Tenth People's Hospital of Tongji University, Shanghai 200072, China; Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Shanghai Gastrointestinal Microecology Research Center, Shanghai 200072, China; Shanghai Institution of Gut Microbiota Research and Engineering Development, Shanghai 200072, China; Clinical Research Center for Digestive Diseases, Tongji University School of Medicine, Shanghai 200072, China
| | - Tao Zuo
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Sun Yat-sen University, Guangzhou 510655, China
| | - Gianluca Ianiro
- Department of Translational Medicine and Surgery, Università Cattolica del Sacro Cuore, Rome, 00168, Italy; Department of Medical and Surgical Sciences, UOC Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, 00168, Italy; Department of Medical and Surgical Sciences, UOC CEMAD Centro Malattie dell'Apparato, Rome, 00168, Italy
| | - Ning Li
- Tenth People's Hospital of Tongji University, Shanghai 200072, China; Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Shanghai Gastrointestinal Microecology Research Center, Shanghai 200072, China; Shanghai Institution of Gut Microbiota Research and Engineering Development, Shanghai 200072, China; Clinical Research Center for Digestive Diseases, Tongji University School of Medicine, Shanghai 200072, China
| | - Qiyi Chen
- Tenth People's Hospital of Tongji University, Shanghai 200072, China; Department of Functional Intestinal Diseases, General Surgery of Shanghai Tenth People's Hospital, Tongji University School of Medicine, Shanghai 200072, China; Shanghai Gastrointestinal Microecology Research Center, Shanghai 200072, China; Shanghai Institution of Gut Microbiota Research and Engineering Development, Shanghai 200072, China; Clinical Research Center for Digestive Diseases, Tongji University School of Medicine, Shanghai 200072, China.
| | - Huanlong Qin
- Tenth People's Hospital of Tongji University, Shanghai 200072, China; Affiliated Suzhou Hospital of Nanjing Medical University, Suzhou Municipal Hospital, Gusu School, Nanjing Medical University, Suzhou 215000, China.
| |
Collapse
|
25
|
Espinoza Miranda SS, Abbaszade G, Hess WR, Drescher K, Saliba AE, Zaburdaev V, Chai L, Dreisewerd K, Grünberger A, Westendorf C, Müller S, Mascher T. Resolving spatiotemporal dynamics in bacterial multicellular populations: approaches and challenges. Microbiol Mol Biol Rev 2025; 89:e0013824. [PMID: 39853129 PMCID: PMC11948493 DOI: 10.1128/mmbr.00138-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2025] Open
Abstract
SUMMARYThe development of multicellularity represents a key evolutionary transition that is crucial for the emergence of complex life forms. Although multicellularity has traditionally been studied in eukaryotes, it originates in prokaryotes. Coordinated aggregation of individual cells within the confines of a colony results in emerging, higher-level functions that benefit the population as a whole. During colony differentiation, an almost infinite number of ecological and physiological population-forming forces are at work, creating complex, intricate colony structures with divergent functions. Understanding the assembly and dynamics of such populations requires resolving individual cells or cell groups within such macroscopic structures. Addressing how each cell contributes to the collective action requires pushing the resolution boundaries of key technologies that will be presented in this review. In particular, single-cell techniques provide powerful tools for studying bacterial multicellularity with unprecedented spatial and temporal resolution. These advancements include novel microscopic techniques, mass spectrometry imaging, flow cytometry, spatial transcriptomics, single-bacteria RNA sequencing, and the integration of spatiotemporal transcriptomics with microscopy, alongside advanced microfluidic cultivation systems. This review encourages exploring the synergistic potential of the new technologies in the study of bacterial multicellularity, with a particular focus on individuals in differentiated bacterial biofilms (colonies). It highlights how resolving population structures at the single-cell level and understanding their respective functions can elucidate the overarching functions of bacterial multicellular populations.
Collapse
Affiliation(s)
| | | | - Wolfgang R. Hess
- Faculty of Biology, Genetics and Experimental Bioinformatics, University of Freiburg, Freiburg, Germany
| | | | - Antoine-Emmanuel Saliba
- Institute for Molecular Infection Biology (IMIB), University of Würzburg, Würzburg, Germany
- Helmholtz Institute for RNA-based Infection Research (HIRI), Helmholtz Center for Infection Research (HZI), Würzburg, Germany
| | - Vasily Zaburdaev
- Department of Biology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
- Max-Planck-Zentrum für Physik und Medizin, Erlangen, Germany
| | - Liraz Chai
- Institute of Chemistry, The Hebrew University of Jerusalem, Jerusalem, Israel
- The Harvey M. Krueger Family Center for Nanoscience and Nanotechnology, The Hebrew University of Jerusalem, Jerusalem, Israel
| | | | - Alexander Grünberger
- Microsystems in Bioprocess Engineering (μBVT), Institute of Process Engineering in Life Sciences (BLT), Karlsruhe Institute of Technology (KIT), Karlsruhe, Germany
| | - Christian Westendorf
- Peter Debye Institute for Soft Matter Physics, Leipzig University, Leipzig, Germany
| | - Susann Müller
- Helmholtz Centre for Environmental Research–UFZ, Leipzig, Germany
| | - Thorsten Mascher
- General Microbiology, Technische Universität Dresden, Dresden, Germany
| |
Collapse
|
26
|
Hu W, Wang Y, Han J, Zhang W, Chen J, Li X, Wang L. Microfluidic organ-on-a-chip models for the gut-liver axis: from structural mimicry to functional insights. Biomater Sci 2025; 13:1624-1656. [PMID: 40019226 DOI: 10.1039/d4bm01273a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/01/2025]
Abstract
The gut-liver axis plays a crucial role in maintaining metabolic balance and overall human health. It orchestrates various processes, such as blood flow, nutrient transfer, metabolite processing, and immune cell communication between the two organs. Traditional methods, such as animal models and two-dimensional (2D) cell cultures, are insufficient in fully replicating the intricate functions of the gut-liver axis. The emergence of microfluidic technology has revolutionized this field, facilitating the development of organ-on-a-chip (OOC) systems. These systems are capable of mimicking the complex structures and dynamic environments of the gut and liver in vitro and incorporating sensors for real-time monitoring. In this article, we review the latest progress in gut-on-a-chip (GOC) and liver-on-a-chip (LOC) systems, as well as the integrated gut-liver-on-a-chip (GLOC) models. Our focus lies in the simulation of physiological parameters, three-dimensional (3D) structural mimicry, microbiome integration, and multicellular co-culture. All these aspects are essential for constructing accurate in vitro models of the gut and liver. Furthermore, we explore the current applications of OOC technology in the study of the gut and liver, including its use in disease modeling, toxicity testing, and drug screening. Finally, we discuss the challenges that remain and outline potential future directions for advancing GOC and LOC development in vitro.
Collapse
Affiliation(s)
- Wanlin Hu
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Yushen Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Junlei Han
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Wenhong Zhang
- College of Mechanical Engineering, Donghua University, Shanghai 201620, China
| | - Jun Chen
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| | - Xinyu Li
- Department of Minimally Invasive Comprehensive Treatment of Cancer, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, 250021, China.
| | - Li Wang
- School of Mechanical Engineering, Qilu University of Technology (Shandong Academy of Sciences), Jinan 250353, China.
- Shandong Institute of Mechanical Design and Research, Jinan 250353, China
| |
Collapse
|
27
|
Rivera-Rodriguez DE, Busby C, Cervantes-Barragan L, Weiss DS. Widespread heteroresistance to antibiotics in Lactobacillus species. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.24.644958. [PMID: 40196655 PMCID: PMC11974758 DOI: 10.1101/2025.03.24.644958] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Lactobacilli are prevalent members of the intestinal and reproductive tract microbiota of humans and other species. They are commonly used in probiotics and various food products due to their beneficial effects on human health. For example, these beneficial microbes are used to treat diarrhea caused by antibiotic therapy and are commonly given during antibiotic treatment. Despite the many studies conducted to understand the beneficial effects of Lactobacilli, less is known about their resistance and heteroresistance to antibiotics. In this study, we evaluated the resistance heterogeneity in eight Lactobacillus species. Our results demonstrate that several Lactobacilli species, including Lactobacillus rhamnosus, are heteroresistant to antibiotics, a recently discovered phenotype commonly seen in multidrug-resistant organisms that cause clinical failures but understudied in commensals and probiotics.
Collapse
Affiliation(s)
- Dormarie E. Rivera-Rodriguez
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| | - Chayse Busby
- Department of Microbiology and Immunology, Emory University School of Medicine, Atlanta, GA
| | | | - David S. Weiss
- Emory Vaccine Center, Emory University School of Medicine, Atlanta, GA
- Division of Infectious Diseases, Department of Medicine, Emory University School of Medicine, Atlanta, GA
| |
Collapse
|
28
|
McDonnell KJ. Operationalizing Team Science at the Academic Cancer Center Network to Unveil the Structure and Function of the Gut Microbiome. J Clin Med 2025; 14:2040. [PMID: 40142848 PMCID: PMC11943358 DOI: 10.3390/jcm14062040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 02/28/2025] [Accepted: 03/05/2025] [Indexed: 03/28/2025] Open
Abstract
Oncologists increasingly recognize the microbiome as an important facilitator of health as well as a contributor to disease, including, specifically, cancer. Our knowledge of the etiologies, mechanisms, and modulation of microbiome states that ameliorate or promote cancer continues to evolve. The progressive refinement and adoption of "omic" technologies (genomics, transcriptomics, proteomics, and metabolomics) and utilization of advanced computational methods accelerate this evolution. The academic cancer center network, with its immediate access to extensive, multidisciplinary expertise and scientific resources, has the potential to catalyze microbiome research. Here, we review our current understanding of the role of the gut microbiome in cancer prevention, predisposition, and response to therapy. We underscore the promise of operationalizing the academic cancer center network to uncover the structure and function of the gut microbiome; we highlight the unique microbiome-related expert resources available at the City of Hope of Comprehensive Cancer Center as an example of the potential of team science to achieve novel scientific and clinical discovery.
Collapse
Affiliation(s)
- Kevin J McDonnell
- Center for Precision Medicine, Department of Medical Oncology & Therapeutics Research, City of Hope Comprehensive Cancer Center, Duarte, CA 91010, USA
| |
Collapse
|
29
|
Vogtmann E, Yano Y, Zouiouich S, Hua X, Wan Y, Purandare V, Li S, Dagnall CL, Jones K, Hicks BD, Hutchinson A, Caporaso JG, Wheeler W, Huang W, Freedman ND, Sandler DP, Beane Freeman LE, Liao LM, Gail MH, Shi J, Abnet CC, Sinha R. The human oral microbiome and risk of colorectal cancer within three prospective cohort studies in the United States. Cancer 2025; 131:e35802. [PMID: 40069139 PMCID: PMC11896928 DOI: 10.1002/cncr.35802] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2025] [Revised: 01/27/2025] [Accepted: 02/05/2025] [Indexed: 03/15/2025]
Abstract
BACKGROUND Oral microbes detected in feces have been associated with colorectal cancer (CRC) in cross-sectional studies. This study investigated the prospective associations between the oral microbiome and incident CRC in the Agricultural Health Study (AHS), National Institutes of Health-AARP (NIH-AARP) Diet and Health Study, and Prostate, Lung, Colorectal, and Ovarian (PLCO) Cancer Screening Trial. METHODS Individuals with oral samples collected before incident CRC diagnoses were identified in the AHS (N = 331), NIH-AARP (N = 249), and PLCO (N = 446) and compared with referent subcohorts (N = 3431). The V4 region of the 16S ribosomal RNA gene was sequenced from oral wash DNA, and the data were processed with QIIME2. Hazard ratios (HRs) and 95% confidence intervals (CIs) for overall CRC and by anatomic subsite (i.e., proximal colon, distal colon, and rectum) were estimated with Cox proportional hazards models with adjustment for potential confounders by cohort and then meta-analyzed. RESULTS Overall, no associations were found between microbial characteristics and CRC risk. However, associations were observed with alpha and beta diversity indices and individual genera in analyses stratified by anatomic subsite. For instance, the presence of Olsenella was strongly positively associated with distal colon cancer risk (HR, 2.16; 95% CI, 1.59-2.95), whereas the presence of Prevotella 2 was positively associated with rectal cancer risk (HR, 1.68; 95% CI, 1.14-2.46). CONCLUSIONS This large study of the prospective association between the oral microbiome and CRC risk showed numerous site-specific associations, including multiple associations with distal colon and rectal cancer risk.
Collapse
Affiliation(s)
- Emily Vogtmann
- Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMarylandUSA
| | - Yukiko Yano
- Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMarylandUSA
| | - Semi Zouiouich
- Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMarylandUSA
| | - Xing Hua
- Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMarylandUSA
- Frederick National Laboratory for Cancer Research/Leidos Biomedical Research LaboratoryFrederickMarylandUSA
| | - Yunhu Wan
- Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMarylandUSA
- Frederick National Laboratory for Cancer Research/Leidos Biomedical Research LaboratoryFrederickMarylandUSA
| | - Vaishnavi Purandare
- Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMarylandUSA
| | - Shilan Li
- Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMarylandUSA
- Bloomberg School of Public HealthJohns Hopkins UniversityBaltimoreMarylandUSA
| | - Casey L. Dagnall
- Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMarylandUSA
- Frederick National Laboratory for Cancer Research/Leidos Biomedical Research LaboratoryFrederickMarylandUSA
| | - Kristine Jones
- Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMarylandUSA
- Frederick National Laboratory for Cancer Research/Leidos Biomedical Research LaboratoryFrederickMarylandUSA
| | - Belynda D. Hicks
- Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMarylandUSA
- Frederick National Laboratory for Cancer Research/Leidos Biomedical Research LaboratoryFrederickMarylandUSA
| | - Amy Hutchinson
- Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMarylandUSA
- Frederick National Laboratory for Cancer Research/Leidos Biomedical Research LaboratoryFrederickMarylandUSA
| | - J. Gregory Caporaso
- Center for Applied Microbiome SciencePathogen and Microbiome InstituteNorthern Arizona UniversityFlagstaffArizonaUSA
| | | | - Wen‐Yi Huang
- Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMarylandUSA
| | - Neal D. Freedman
- Division of Cancer Control and Population ScienceNational Cancer InstituteBethesdaMarylandUSA
| | - Dale P. Sandler
- Chronic Disease Epidemiology GroupEpidemiology BranchNational Institute of Environmental Health SciencesResearch Triangle ParkNorth CarolinaUSA
| | - Laura E. Beane Freeman
- Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMarylandUSA
| | - Linda M. Liao
- Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMarylandUSA
| | - Mitchell H. Gail
- Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMarylandUSA
| | - Jianxin Shi
- Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMarylandUSA
| | - Christian C. Abnet
- Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMarylandUSA
| | - Rashmi Sinha
- Division of Cancer Epidemiology and GeneticsNational Cancer InstituteBethesdaMarylandUSA
| |
Collapse
|
30
|
Gurow K, Joshi DC, Gwasikoti J, Joshi N. Gut Microbial Control of Neurotransmitters and Their Relation to Neurological Disorders: A Comprehensive Review. Horm Metab Res 2025. [PMID: 40073909 DOI: 10.1055/a-2536-1421] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/14/2025]
Abstract
The study explores the vital role of gut microbiota in regulating neurotransmitters and its subsequent effects on brain function and mental health. It aims to unravel the mechanisms by which microbial metabolites influence neurotransmitter synthesis and signaling. The ultimate goal is to identify potential therapeutic strategies targeting gut microbiota for the management and treatment of neurological disorders, such as depression, autism spectrum disorder (ASD), anxiety, and Parkinson's disease. The review synthesizes current research on the gut-brain axis, focusing on the influence of gut microbial metabolites on key neurotransmitters, including dopamine, serotonin, and gamma-aminobutyric acid (GABA). It incorporates a multidisciplinary approach, linking microbiology, neurobiology, and clinical research. Each section presents an in-depth review of scientific studies, clinical trials, and emerging therapeutic strategies. The findings highlight the intricate interplay between gut microbiota and the central nervous system. Gut microbes significantly impact the synthesis and signaling of crucial neurotransmitters, which play a pivotal role in neurological health. Evidence supports the hypothesis that modulating gut microbiota can alter neurotransmitter output and alleviate symptoms associated with neurological disorders. Notable therapeutic potentials include microbiota-targeted interventions for managing depression, ASD, anxiety, and Parkinson's disease. This comprehensive analysis underscores the critical connection between gut microbiota and neurological health. By bridging gaps between microbiology, neurobiology, and clinical practice, the study opens avenues for innovative therapeutic approaches. It provides a valuable resource for researchers, clinicians, and students, emphasizing the need for continued investigation into gut microbiota's role in neurological disorders and its therapeutic potential.
Collapse
Affiliation(s)
- Kajal Gurow
- Gurukul Pharmacy College IPB-13, RIICO Industrial Area, Ranpur, Kota, Rajasthan, India
| | - Deepak Chandra Joshi
- Department of Pharmacy, School of Chemical Sciences and Pharmacy, Central University of Rajasthan, Bandar Sindri, Dist. Ajmer, Rajasthan, India
| | - Jyoti Gwasikoti
- Department of Pharmacy, Graphic Era Hill University, Bhimtal, India
| | - Nirmal Joshi
- Faculty of Pharmaceutical Sciences, Amrapali University, Haldwani, India
| |
Collapse
|
31
|
Ghosh AN, Walsh CJ, Maiden MJ, Stinear TP, Deane AM. Effect of dietary fibre on the gastrointestinal microbiota during critical illness: A scoping review. World J Crit Care Med 2025; 14:98241. [DOI: 10.5492/wjccm.v14.i1.98241] [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: 06/21/2024] [Revised: 09/27/2024] [Accepted: 10/28/2024] [Indexed: 12/11/2024] Open
Abstract
The systemic effects of gastrointestinal (GI) microbiota in health and during chronic diseases is increasingly recognised. Dietary strategies to modulate the GI microbiota during chronic diseases have demonstrated promise. While changes in dietary intake can rapidly change the GI microbiota, the impact of dietary changes during acute critical illness on the microbiota remain uncertain. Dietary fibre is metabolised by carbohydrate-active enzymes and, in health, can alter GI microbiota. The aim of this scoping review was to describe the effects of dietary fibre supplementation in health and disease states, specifically during critical illness. Randomised controlled trials and prospective cohort studies that include adults (> 18 years age) and reported changes to GI microbiota as one of the study outcomes using non-culture methods, were identified. Studies show dietary fibres have an impact on faecal microbiota in health and disease. The fibre, inulin, has a marked and specific effect on increasing the abundance of faecal Bifidobacteria. Short chain fatty acids produced by Bifidobacteria have been shown to be beneficial in other patient populations. Very few trials have evaluated the effect of dietary fibre on the GI microbiota during critical illness. More research is necessary to establish optimal fibre type, doses, duration of intervention in critical illness.
Collapse
Affiliation(s)
- Angajendra N Ghosh
- Department of Intensive Care, The Northern Hospital, Epping 3076, Victoria, Australia
| | - Calum J Walsh
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, Melbourne 3052, Victoria, Australia
| | - Matthew J Maiden
- Department of Intensive Care, The Royal Melbourne Hospital, The University of Melbourne, Parkville 3050, Victoria, Australia
| | - Tim P Stinear
- Department of Microbiology and Immunology, The Peter Doherty Institute for Infection and Immunity, Melbourne 3052, Victoria, Australia
| | - Adam M Deane
- Department of Intensive Care Medicine, The Royal Melbourne Hospital, Parkville 3050, Victoria, Australia
| |
Collapse
|
32
|
Cao L, Guo W, Yang S, Ahmad AA, Dong Y, Gong C, Wang S, Yang X, Cheng Z, Yan Z, Wang W. Survey of gut microbial biogeography and their functional niche in the grow-finishing swine of ordinary feeding. Front Microbiol 2025; 16:1530553. [PMID: 40124893 PMCID: PMC11925874 DOI: 10.3389/fmicb.2025.1530553] [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: 11/19/2024] [Accepted: 02/17/2025] [Indexed: 03/25/2025] Open
Abstract
Background Swine represent one of the most economically significant livestock worldwide, and their intestinal microbial communities are crucial for maintaining physiological development and regulating host metabolism. While extensive research has focused on the fecal microbiota of swine, investigations into microbial communities across different intestinal segments remain limited. Objective This study aims to elucidate the intestinal microbiota of swine by analyzing luminal contents from different intestinal segments, including the duodenum, jejunum, ileum, cecum, and colon. Methods We employed 16S rRNA sequencing to explore the diversity and structure of gut microbial biogeography, microbial functional niches, and their associated pathways. Results Our findings reveal significantly lower microbial richness and diversity in the small intestine (duodenum, jejunum, and ileum) compared to the large intestine (cecum and colon) (p < 0.05). At the phylum level, Firmicutes, Actinobacteria, Proteobacteria, and Bacteroidetes were the dominant phyla, collectively accounting for over 90% of the total sequences. In the small intestine, Proteobacteria (4.76-34.2%), Actinobacteria, and Fusobacteriota were more abundant, whereas in the large intestine, Firmicutes (89.8-90.4%) was predominated. At the genus level, Fusobacterium, Corynebacterium, Rothia, Bradyrhizobium, and Brevundimonas were predominant in duodenum. Romboutsia, Clostridium_sensu_stricto_1, Terrisporobacter, and Jeotgalicoccus demonstrated greater abundances in the jejunum and ileum. Oscillospiraceae_UCG-005 in the cecum and Christensenellaceae_R-7_group in the colon were more abundant with 16.4 and 20.2% relative abundances, respectively. The specialists detected from the duodenum to the colon were all the predominant genera in each intestinal segment with relatively higher relative abundance. For instance, Romboutsia (3.06-36.1%), Clostridium_sensu_stricto_1 (5.31-18.6%), and Terrisporobacter (0.849-5.72%) were dominant genera and specialists in the small intestine, associated with enriched pathways of Amino acid metabolism and Lipid metabolism. Conversely, Oscillospiraceae_UCG-005 (16.4%, 4.06%) and Christensenellaceae_R-7_group (5.44%, 20.2%) are predominant genera and specialists within the large intestine, linked to pathways involved in Glycan biosynthesis and metabolism pathway, as well as the Biosynthesis of other secondary metabolites. Conclusion These highlight the importance of genus specialists compared to genus generalists. The findings provide essential data for assessing the role of the intestinal microbiome in maintaining and enhancing swine health and productivity, offering fundamental guidance for further exploration of host-microbe interaction mechanisms and regulatory pathways.
Collapse
Affiliation(s)
- Lili Cao
- Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, China
- Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province, College of Animal Science, Guizhou University, Guiyang, China
| | - Wei Guo
- Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, China
| | - Shiyu Yang
- College of Resources and Environmental Engineering, Guizhou University, Guiyang, China
- Guizhou Yuhong Biotechnology Co., Ltd., Guiyang, China
| | - Anum Ali Ahmad
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Edinburgh, United Kingdom
| | - Yuntao Dong
- Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, China
| | - Cen Gong
- Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, China
| | - Shuoqi Wang
- Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, China
| | - Xuemin Yang
- Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, China
| | - Zhentao Cheng
- Key Laboratory of Animal Diseases and Veterinary Public Health of Guizhou Province, College of Animal Science, Guizhou University, Guiyang, China
| | - Zhihong Yan
- Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, China
- Institute of New Rural Development, Guizhou University, Guiyang, China
| | - Weiwei Wang
- Laboratory of Animal Genetics, Breeding and Reproduction in the Plateau Mountainous Region, Ministry of Education, College of Animal Science, Guizhou University, Guiyang, China
| |
Collapse
|
33
|
Tardiolo G, La Fauci D, Riggio V, Daghio M, Di Salvo E, Zumbo A, Sutera AM. Gut Microbiota of Ruminants and Monogastric Livestock: An Overview. Animals (Basel) 2025; 15:758. [PMID: 40076043 PMCID: PMC11899476 DOI: 10.3390/ani15050758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2025] [Revised: 03/02/2025] [Accepted: 03/04/2025] [Indexed: 03/14/2025] Open
Abstract
The diversity and composition of the gut microbiota are widely recognized as fundamental factors influencing the well-being and productivity of domestic animals. Advancements in sequencing technologies have revolutionized studies in this research field, allowing for deeper insights into the composition and functionality of microbiota in livestock. Ruminants and monogastric animals exhibit distinct digestive systems and microbiota characteristics: ruminants rely on fermentation, while monogastrics use enzymatic digestion, and monogastric animals have simpler stomach structures, except for horses and rabbits, where both processes coexist. Understanding the gut microbiota's impact and composition in both animal types is essential for optimizing production efficiency and promoting animal health. Following this perspective, the present manuscript review aims to provide a comprehensive overview of the gut microbiota in ruminants (such as cattle, sheep, and goats) and monogastric animals (including horses, pigs, rabbits, and chickens).
Collapse
Affiliation(s)
- Giuseppe Tardiolo
- Department of Veterinary Sciences, University of Messina, Viale Giovanni Palatucci 13, 98168 Messina, Italy; (G.T.); (D.L.F.)
| | - Deborah La Fauci
- Department of Veterinary Sciences, University of Messina, Viale Giovanni Palatucci 13, 98168 Messina, Italy; (G.T.); (D.L.F.)
| | - Valentina Riggio
- The Roslin Institute and Royal (Dick) School of Veterinary Studies, University of Edinburgh, Easter Bush Campus, Edinburgh EH25 9RG, UK;
| | - Matteo Daghio
- Department of Agriculture, Food, Environment and Forestry, University of Florence, Piazzale delle Cascine 18, 50144 Florence, Italy;
| | - Eleonora Di Salvo
- Department of Biomedical, Dental Sciences, Morphological and Functional Imaging, University of Messina, Via Consolare Valeria 1, 98125 Messina, Italy;
| | - Alessandro Zumbo
- Department of Veterinary Sciences, University of Messina, Viale Giovanni Palatucci 13, 98168 Messina, Italy; (G.T.); (D.L.F.)
| | - Anna Maria Sutera
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, Viale Ferdinando Stagno d’Alcontres 31, 98166 Messina, Italy;
| |
Collapse
|
34
|
Li J, Wei W, Ma X, Ji J, Ling X, Xu Z, Guan Y, Zhou L, Wu Q, Huang W, Liu F, Zhao M. Antihypertensive effects of rice peptides involve intestinal microbiome alterations and intestinal inflammation alleviation in spontaneously hypertensive rats. Food Funct 2025; 16:1731-1759. [PMID: 39752320 DOI: 10.1039/d4fo04251d] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/04/2025]
Abstract
Gut dysbiosis serves as an underlying risk factor for the development of hypertension. The resolution of this dysbiosis has emerged as a promising strategy in improving hypertension. Food-derived bioactive protein peptides have become increasingly more attractive in ameliorating hypertension, primarily due to their anti-inflammatory and anti-oxidant activities. However, the regulatory mechanisms linking rice peptides (RP), gut dysbiosis, and hypertension remain to be fully elucidated. In our study, male spontaneously hypertensive rats (SHR) were fed with chow diet and concomitantly treated with ddH2O (Ctrl) or varying doses of rice peptides (20, 100, or 500 mg (kg bw day)-1 designated as low-dose RP, LRP; medium-dose RP, MRP; high-dose RP, HAP) or captopril (Cap) by intragastric administration. Wistar-Kyoto (WKY) rats served as the normotensive control group and were orally administered with ddH2O. We observed beneficial effects of RP in lowering blood pressure and ameliorating cardiovascular risk profiles, as evidenced by improvements in glucolipid metabolic disorders, hepatic and renal damage, left ventricular hypertrophy and endothelial dysfunction in hypertensive rats. More importantly, we found that RP attenuated intestinal pathological damage, improved impaired intestinal barrier, and reduced intestinal inflammation by inhibiting the HMGB1-TLR4-NF-κB pathway. Notably, multi-omics integrative analyses have revealed that RP altered the composition and function of the gut microbiota. This is exemplified by the observed enrichment of beneficial bacterial constituents, such as g_Lactobacillus, g_Lactococcus, s_Lactobacillus_intestinalis, and Lactococcus lactis, and elevated production of microbiota-derived short-chain fatty acid metabolites. Collectively, these studies suggest that the hypotensive effects of RP may be associated with modulation of the gut microbiota and its short-chain fatty acids metabolites. This implicates the microbiota-gut-HMGB1-TLR4-NF-κB axis as a novel venue for the amelioration of hypertension and its complications.
Collapse
Affiliation(s)
- Juan Li
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
- Department of Epidemiology, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Wei Wei
- Zhong Shi Du Qing (Shandong) Biotechnology Company, Heze, 274108, China.
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| | - Xiaomin Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
- Center for Experimental Public Health and Preventive Medicine Education, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Jing Ji
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Xiaomeng Ling
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Zhuyan Xu
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Yutong Guan
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Leyan Zhou
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| | - Qiming Wu
- Nutrilite Health Institute, Shanghai, 201203, China.
| | - Wenhua Huang
- AMWAY (China) R&D Center, Guangzhou, 510730, China.
| | - Fuguo Liu
- College of Food Science and Engineering, Northwest A&F University, Yangling, Shaanxi, 712100, China.
| | - Min Zhao
- Department of Nutrition and Food Hygiene, School of Public Health, Cheeloo College of Medicine, Shandong University, Jinan, 250012, China.
| |
Collapse
|
35
|
Zhang S, Peng L, Goswami S, Li Y, Dang H, Xing S, Feng P, Nigro G, Liu Y, Ma Y, Liu T, Yang J, Jiang T, Yang Y, Barker N, Sansonetti P, Kundu P. Intestinal crypt microbiota modulates intestinal stem cell turnover and tumorigenesis via indole acetic acid. Nat Microbiol 2025; 10:765-783. [PMID: 39972061 DOI: 10.1038/s41564-025-01937-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2024] [Accepted: 01/14/2025] [Indexed: 02/21/2025]
Abstract
Intestinal crypts harbour a specific microbiota but whether and how these bacteria regulate intestinal stem cells (ISCs) or influence colorectal cancer (CRC) development is unclear. Here we screened crypt-resident bacteria in organoids and found that indole acetic acid (IAA) secreted by Acinetobacter radioresistens inhibits ISC turnover. A. radioresistens inhibited cellular proliferation in tumour slices from CRC patients and inhibited intestinal tumorigenesis and spheroid initiation in APCMin/+ mice. Targeted clearance of A. radioresistens from colonic crypts using bacteriophage increased EphB2 expression and consequently promoted cellular proliferation, ISC turnover and tumorigenesis in mouse models of CRC. The protective effects of A. radioresistens were abrogated upon deletion of trpC to prevent IAA production, or upon intestine-specific aryl hydrocarbon receptor (AhR) knockout, identifying an IAA-AhR-Wnt-β-catenin signalling axis that promotes ISC homeostasis. Our findings reveal a protective role for an intestinal crypt-resident microbiota member in tumorigenesis.
Collapse
Affiliation(s)
- Shuning Zhang
- Laboratory for Microbiota-Host Interactions, The Center for Microbes, Development and Health, Shanghai Institute of Immunity and Infection-Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Lihua Peng
- Laboratory for Microbiota-Host Interactions, The Center for Microbes, Development and Health, Shanghai Institute of Immunity and Infection-Chinese Academy of Sciences, Shanghai, China
| | - Shyamal Goswami
- Laboratory for Microbiota-Host Interactions, The Center for Microbes, Development and Health, Shanghai Institute of Immunity and Infection-Chinese Academy of Sciences, Shanghai, China
| | - Yuchen Li
- Laboratory for Microbiota-Host Interactions, The Center for Microbes, Development and Health, Shanghai Institute of Immunity and Infection-Chinese Academy of Sciences, Shanghai, China
| | - Haiyue Dang
- Laboratory for Microbiota-Host Interactions, The Center for Microbes, Development and Health, Shanghai Institute of Immunity and Infection-Chinese Academy of Sciences, Shanghai, China
- University of Chinese Academy of Sciences, Beijing, China
| | - Shuli Xing
- Laboratory for Microbiota-Host Interactions, The Center for Microbes, Development and Health, Shanghai Institute of Immunity and Infection-Chinese Academy of Sciences, Shanghai, China
| | - Panpan Feng
- Laboratory for Microbiota-Host Interactions, The Center for Microbes, Development and Health, Shanghai Institute of Immunity and Infection-Chinese Academy of Sciences, Shanghai, China
| | - Giulia Nigro
- Microenvironment and Immunity Unit, INSERM U1224, Institut Pasteur, Paris, France
| | - Yingying Liu
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, China
| | - Yingfei Ma
- Shenzhen Institutes of Advanced Technology, Chinese Academy of Sciences, Shenzhen, China
| | - Tianhao Liu
- Affiliated Hospital of Jiangnan University and Wuxi Medical College, Jiangnan University, Wuxi, China
| | - Jiahua Yang
- Department of General Surgery, Putuo Hospital, Shanghai, China
| | - Tinglei Jiang
- Jilin Provincial Key Laboratory of Animal Resource Conservation and Utilization, Northeast Normal University, Changchun, China
| | - Yingnan Yang
- Luodian Hospital in Baoshan District, Shanghai, China
| | - Nick Barker
- Institute of Molecular and Cell Biology, Singapore and Department of Physiology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | | | - Parag Kundu
- Laboratory for Microbiota-Host Interactions, The Center for Microbes, Development and Health, Shanghai Institute of Immunity and Infection-Chinese Academy of Sciences, Shanghai, China.
- University of Chinese Academy of Sciences, Beijing, China.
| |
Collapse
|
36
|
Liu S, Tao Z, Qiao M, Shi L. The Functions of Major Gut Microbiota in Obesity and Type 2 Diabetes. Metabolites 2025; 15:167. [PMID: 40137132 PMCID: PMC11943573 DOI: 10.3390/metabo15030167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2025] [Revised: 02/20/2025] [Accepted: 02/27/2025] [Indexed: 03/27/2025] Open
Abstract
Background: Gut microbiomes play a vital role in maintaining whole-body metabolic homeostasis. It has gained significant attention in recent years due to advancements in genome sequencing technologies and a deeper understanding of its relationship with obesity. However, the specific ways in which different microorganisms directly or indirectly influence host obesity, as well as the underlying mechanisms, remain uncertain because of the complexity of gut microbiota composition. Methods: In this review, we summarize the roles of the major gut microbiota phyla such as Bacteroidetes, Firmicutes, Proteobacteria, and Verrucomicrobia in obesity and type 2 diabetes based on studies published in the past five years on PubMed and Google Scholar. The current therapeutic strategies associated with gut microbiota are also explored from clinical trials, and challenges and future directions are discussed. Results and Conclusions: This review will provide a deeper understanding of the functions of major gut microbiota in obesity and type 2 diabetes, which could lead to more individualized and effective treatments for metabolic diseases.
Collapse
Affiliation(s)
- Siman Liu
- Departments of Nutritional Science, University of Connecticut, Storrs, CT 06269, USA
| | - Zhipeng Tao
- Department of Nutrition and Food Sciences, Texas Woman’s University, Denton, TX 76204, USA
| | - Mingyu Qiao
- Departments of Nutritional Science, University of Connecticut, Storrs, CT 06269, USA
| | - Limin Shi
- Departments of Medicine and Biological Chemistry, Johns Hopkins University School of Medicine, Institute for Fundamental Biomedical Research, Johns Hopkins All Children’s Hospital, St. Petersburg, FL 33701, USA
| |
Collapse
|
37
|
Zhang B, Xu N, Bian ZR, Zhang C, Li X, Ren XX, Jiang Z, Wu Z, Yu Q, Zheng KY, Chen MX, Yan C. Clonorchis sinensis Infection prevents DSS-induced Colitis Via Lithocholic Acid in a Gut Microbiota-Dependent Manner. Inflammation 2025:10.1007/s10753-025-02241-4. [PMID: 40014218 DOI: 10.1007/s10753-025-02241-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2024] [Revised: 01/02/2025] [Accepted: 01/07/2025] [Indexed: 02/28/2025]
Abstract
Increasing evidence demonstrates that helminth and its components can ameliorate ulcerative colitis. Clonorchis sinensis (C. sinensis) is a kind of helminth that dwells in the bile ducts for many years, but the roles and underlying mechanisms of C. sinensis-induced protection from colitis are not elucidated. In the present study, the mice were infected with 50 C. sinensis metacercariae and further administrated with 4% Dextran Sodium Sulfate (DSS) in drinking water for 7 days on days 49 post-infection. The disease severity and the integrity of gut barriers were evaluated. Gut microbiota was measured using 16sRNA sequencing, and bile acids in the colon were detected by Liquid Chromatography Mass Spectrometry (LC/MS). The Co-housing approach or microbiota deletion with additional supplies of secondary bile acids (SBAs) was employed to investigate the roles of gut microbiota in the protection from colitis. C. sinensis infection moderated the dysbiosis of the intestinal microbiota and increased the levels of SBAs and bile acid receptor Takeda G protein-coupled receptor 5 (TGR5), which finally benefited anti-inflammation and ameliorated the severity of DSS-induced colitis. Co-housing with C. sinensis-infected mice, and non-infected mice with colitis also showed an increase of TGR5, decreased pro-inflammatory cytokines, and a reduction in the severity of colitis, compared to those mice suffering from colitis without co-housing. Furthermore, C. sinensis-induced protective effects on colitis were attenuated by microbiota deletion, while SBAs (lithocholic acid, LCA) supplementation reversed the colitis. The present study demonstrates that C. sinensis infection ameliorates DSS-induced ulcerative colitis in mice, which is dependent on gut microbiota-associated SBAs.
Collapse
Affiliation(s)
- Beibei Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, National Demonstration Center for Experimental Basic Medical Science Education, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Na Xu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, National Demonstration Center for Experimental Basic Medical Science Education, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Zheng-Rui Bian
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, National Demonstration Center for Experimental Basic Medical Science Education, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Chen Zhang
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, National Demonstration Center for Experimental Basic Medical Science Education, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Xing Li
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, National Demonstration Center for Experimental Basic Medical Science Education, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Xin-Xin Ren
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, National Demonstration Center for Experimental Basic Medical Science Education, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Zhihua Jiang
- Guangxi Key Laboratory for the Prevention and Control of Viral Hepatitis, Guangxi Zhuang Autonomous Region Center for Disease Control and Prevention, Institute of Parasitic Disease Control and Prevention, Nanning, China
| | - Zhongdao Wu
- Department of Parasitology, Key Laboratory of Tropical Disease Control (Ministry of Education), Zhongshan School of Medicine, Sun Yat-Sen University, Guangzhou, 510080, People's Republic of China
| | - Qian Yu
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, National Demonstration Center for Experimental Basic Medical Science Education, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Kui-Yang Zheng
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, National Demonstration Center for Experimental Basic Medical Science Education, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, People's Republic of China
| | - Mu-Xin Chen
- National Key Laboratory of Intelligent Tracking and Forecasting for Infectious Diseases, National Institute of Parasitic Diseases, Chinese Center for Disease Control and Prevention (Chinese Center for Tropical Diseases Research), Key Laboratory of Parasite and Vector Biology, Ministry of Health, WHO Collaborating Center of Malaria, Schistosomiasis and Filariasis, Shanghai, 200025, People's Republic of China.
- Hainan Tropical Diseases Research Center (Hainan Sub-Center, Chinese Center for Tropical Diseases Research), Haikou, 571199, People's Republic of China.
| | - Chao Yan
- Jiangsu Key Laboratory of Immunity and Metabolism, Department of Pathogenic Biology and Immunology, National Demonstration Center for Experimental Basic Medical Science Education, Laboratory of Infection and Immunity, Xuzhou Medical University, Xuzhou, People's Republic of China.
| |
Collapse
|
38
|
Kiouri DP, Batsis GC, Mavromoustakos T, Giuliani A, Chasapis CT. Structure-Based Modeling of the Gut Bacteria-Host Interactome Through Statistical Analysis of Domain-Domain Associations Using Machine Learning. BIOTECH 2025; 14:13. [PMID: 40227324 PMCID: PMC11940256 DOI: 10.3390/biotech14010013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2025] [Revised: 02/16/2025] [Accepted: 02/21/2025] [Indexed: 04/15/2025] Open
Abstract
The gut microbiome, a complex ecosystem of microorganisms, plays a pivotal role in human health and disease. The gut microbiome's influence extends beyond the digestive system to various organs, and its imbalance is linked to a wide range of diseases, including cancer and neurodevelopmental, inflammatory, metabolic, cardiovascular, autoimmune, and psychiatric diseases. Despite its significance, the interactions between gut bacteria and human proteins remain understudied, with less than 20,000 experimentally validated protein interactions between the host and any bacteria species. This study addresses this knowledge gap by predicting a protein-protein interaction network between gut bacterial and human proteins. Using statistical associations between Pfam domains, a comprehensive dataset of over one million experimentally validated pan-bacterial-human protein interactions, as well as inter- and intra-species protein interactions from various organisms, were used for the development of a machine learning-based prediction method to uncover key regulatory molecules in this dynamic system. This study's findings contribute to the understanding of the intricate gut microbiome-host relationship and pave the way for future experimental validation and therapeutic strategies targeting the gut microbiome interplay.
Collapse
Affiliation(s)
- Despoina P. Kiouri
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (D.P.K.); (G.C.B.)
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15772 Athens, Greece;
| | - Georgios C. Batsis
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (D.P.K.); (G.C.B.)
| | - Thomas Mavromoustakos
- Laboratory of Organic Chemistry, Department of Chemistry, National and Kapodistrian University of Athens, 15772 Athens, Greece;
| | - Alessandro Giuliani
- Environment and Health Department, Istituto Superiore di Sanità, 00161 Rome, Italy;
| | - Christos T. Chasapis
- Institute of Chemical Biology, National Hellenic Research Foundation, 11635 Athens, Greece; (D.P.K.); (G.C.B.)
| |
Collapse
|
39
|
Valdés-Mas R, Leshem A, Zheng D, Cohen Y, Kern L, Zmora N, He Y, Katina C, Eliyahu-Miller S, Yosef-Hevroni T, Richman L, Raykhel B, Allswang S, Better R, Shmueli M, Saftien A, Cullin N, Slamovitz F, Ciocan D, Ouyang KS, Mor U, Dori-Bachash M, Molina S, Levin Y, Atarashi K, Jona G, Puschhof J, Harmelin A, Stettner N, Chen M, Suez J, Honda K, Lieb W, Bang C, Kori M, Maharshak N, Merbl Y, Shibolet O, Halpern Z, Shouval DS, Shamir R, Franke A, Abdeen SK, Shapiro H, Savidor A, Elinav E. Metagenome-informed metaproteomics of the human gut microbiome, host, and dietary exposome uncovers signatures of health and inflammatory bowel disease. Cell 2025; 188:1062-1083.e36. [PMID: 39837331 DOI: 10.1016/j.cell.2024.12.016] [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: 07/02/2024] [Revised: 10/08/2024] [Accepted: 12/11/2024] [Indexed: 01/23/2025]
Abstract
Host-microbiome-dietary interactions play crucial roles in regulating human health, yet their direct functional assessment remains challenging. We adopted metagenome-informed metaproteomics (MIM), in mice and humans, to non-invasively explore species-level microbiome-host interactions during commensal and pathogen colonization, nutritional modification, and antibiotic-induced perturbation. Simultaneously, fecal MIM accurately characterized the nutritional exposure landscape in multiple clinical and dietary contexts. Implementation of MIM in murine auto-inflammation and in human inflammatory bowel disease (IBD) characterized a "compositional dysbiosis" and a concomitant species-specific "functional dysbiosis" driven by suppressed commensal responses to inflammatory host signals. Microbiome transfers unraveled early-onset kinetics of these host-commensal cross-responsive patterns, while predictive analyses identified candidate fecal host-microbiome IBD biomarker protein pairs outperforming S100A8/S100A9 (calprotectin). Importantly, a simultaneous fecal nutritional MIM assessment enabled the determination of IBD-related consumption patterns, dietary treatment compliance, and small intestinal digestive aberrations. Collectively, a parallelized dietary-bacterial-host MIM assessment functionally uncovers trans-kingdom interactomes shaping gastrointestinal ecology while offering personalized diagnostic and therapeutic insights into microbiome-associated disease.
Collapse
Affiliation(s)
- Rafael Valdés-Mas
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Avner Leshem
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; Department of Surgery, Tel Aviv Sourasky Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Danping Zheng
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Yotam Cohen
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Lara Kern
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Niv Zmora
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; School of Medicine, Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Research Center for Digestive Tract and Liver Diseases, Tel Aviv Sourasky Medical Center, Tel Aviv University, Tel Aviv, Israel
| | - Yiming He
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Corine Katina
- de Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalized Medicine (G-INCPM), Weizmann Institute of Science, Rehovot, Israel
| | | | - Tal Yosef-Hevroni
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Liron Richman
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Barbara Raykhel
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Shira Allswang
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Reut Better
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Merav Shmueli
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Nyssa Cullin
- Division of Microbiome & Cancer, DKFZ, Heidelberg, Germany
| | - Fernando Slamovitz
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Dragos Ciocan
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | | | - Uria Mor
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Mally Dori-Bachash
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Shahar Molina
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Yishai Levin
- de Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalized Medicine (G-INCPM), Weizmann Institute of Science, Rehovot, Israel
| | - Koji Atarashi
- RIKEN Center for Integrative Medical Sciences (IMS), Tsurumi, Yokohama, Kanagawa, Japan; Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Ghil Jona
- Department of Life Sciences Core Facilities, Weizmann Institute of Science, Rehovot, Israel
| | - Jens Puschhof
- Division of Microbiome & Cancer, DKFZ, Heidelberg, Germany
| | - Alon Harmelin
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Noa Stettner
- Department of Veterinary Resources, Weizmann Institute of Science, Rehovot, Israel
| | - Minhu Chen
- Department of Gastroenterology, The First Affiliated Hospital, Sun Yat-Sen University, Guangzhou, China
| | - Jotham Suez
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Kenya Honda
- RIKEN Center for Integrative Medical Sciences (IMS), Tsurumi, Yokohama, Kanagawa, Japan; Department of Microbiology and Immunology, Keio University School of Medicine, Tokyo, Japan
| | - Wolfgang Lieb
- Institute of Epidemiology and Biobank Popgen, University Hospital of Schleswig-Holstein (UKSH), Kiel, Germany
| | - Corinna Bang
- Institute of Clinical Molecular Biology, Christian-Albrechts-Universität Zu Kiel, Kiel, Germany; University Hospital of Schleswig-Holstein (UKSH), Kiel, Germany
| | - Michal Kori
- Pediatric Gastroenterology Unit, Kaplan Medical Center, Rehovot, Israel; Faculty of Medicine, Hebrew University of Jerusalem, Jerusalem, Israel
| | - Nitsan Maharshak
- School of Medicine, Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Gastroenterology and Hepatology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Yifat Merbl
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Oren Shibolet
- School of Medicine, Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Gastroenterology and Hepatology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Zamir Halpern
- School of Medicine, Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Department of Gastroenterology and Hepatology, Tel Aviv Medical Center, Tel Aviv, Israel
| | - Dror S Shouval
- School of Medicine, Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Institute of Gastroenterology, Nutrition, and Liver Diseases, Schneider Children's Medical Centre, Petach-Tikva, Israel
| | - Raanan Shamir
- School of Medicine, Faculty of Medicine and Health Sciences, Tel Aviv University, Tel Aviv, Israel; Institute of Gastroenterology, Nutrition, and Liver Diseases, Schneider Children's Medical Centre, Petach-Tikva, Israel
| | - Andre Franke
- Institute of Clinical Molecular Biology, Christian-Albrechts-Universität Zu Kiel, Kiel, Germany; University Hospital of Schleswig-Holstein (UKSH), Kiel, Germany
| | - Suhaib K Abdeen
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Hagit Shapiro
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel
| | - Alon Savidor
- de Botton Institute for Protein Profiling, The Nancy and Stephen Grand Israel National Center for Personalized Medicine (G-INCPM), Weizmann Institute of Science, Rehovot, Israel
| | - Eran Elinav
- Department of Systems Immunology, Weizmann Institute of Science, Rehovot, Israel; Division of Microbiome & Cancer, DKFZ, Heidelberg, Germany.
| |
Collapse
|
40
|
Blok L, Hanssen N, Nieuwdorp M, Rampanelli E. From Microbes to Metabolites: Advances in Gut Microbiome Research in Type 1 Diabetes. Metabolites 2025; 15:138. [PMID: 39997763 PMCID: PMC11857261 DOI: 10.3390/metabo15020138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Revised: 02/12/2025] [Accepted: 02/13/2025] [Indexed: 02/26/2025] Open
Abstract
Background: Type 1 diabetes (T1D) is a severe chronic T-cell mediated autoimmune disease that attacks the insulin-producing beta cells of the pancreas. The multifactorial nature of T1D involves both genetic and environmental components, with recent research focusing on the gut microbiome as a crucial environmental factor in T1D pathogenesis. The gut microbiome and its metabolites play an important role in modulating immunity and autoimmunity. In recent years, studies have revealed significant alterations in the taxonomic and functional composition of the gut microbiome associated with the development of islet autoimmunity and T1D. These changes include reduced production of short-chain fatty acids, altered bile acid and tryptophan metabolism, and increased intestinal permeability with consequent perturbations of host (auto)immune responses. Methods/Results: In this review, we summarize and discuss recent observational, mechanistic and etiological studies investigating the gut microbiome in T1D and elucidating the intricate role of gut microbes in T1D pathogenesis. Moreover, we highlight the recent advances in intervention studies targeting the microbiota for the prevention or treatment of human T1D. Conclusions: A deeper understanding of the evolution of the gut microbiome before and after T1D onset and of the microbial signals conditioning host immunity may provide us with essential insights for exploiting the microbiome as a prognostic and therapeutic tool.
Collapse
Affiliation(s)
- Lente Blok
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, 1105 AZ Amsterdam, The Netherlands; (N.H.); (M.N.)
| | - Nordin Hanssen
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, 1105 AZ Amsterdam, The Netherlands; (N.H.); (M.N.)
| | - Max Nieuwdorp
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, 1105 AZ Amsterdam, The Netherlands; (N.H.); (M.N.)
| | - Elena Rampanelli
- Department of Internal and Vascular Medicine, Amsterdam University Medical Center, Location AMC, 1105 AZ Amsterdam, The Netherlands; (N.H.); (M.N.)
- Amsterdam Institute for Infection and Immunity (AII), Amsterdam, The Netherlands
| |
Collapse
|
41
|
Ke H, Yao H, Wei P. Advances in research on gut microbiota and allergic diseases in children. CURRENT RESEARCH IN MICROBIAL SCIENCES 2025; 8:100362. [PMID: 40123594 PMCID: PMC11930230 DOI: 10.1016/j.crmicr.2025.100362] [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] [Indexed: 03/25/2025] Open
Abstract
Epidemiological studies indicate a rising prevalence of allergic diseases, now recognized as a major global public health concern. In children, the progression of these diseases often follows the "atopic march," beginning with eczema, followed by food allergies, allergic rhinitis, and asthma. Recent research has linked gut microbiota dysbiosis to the development of allergic diseases in children. The gut microbiota, a crucial component of human health, plays a vital role in maintaining overall well-being, highlighting its potential in preventing and modifying the course of allergic diseases. This review examines the relationship between childhood allergic diseases and gut microbiota, drawing on the latest evidence. We first elaborated the concepts of allergic diseases and gut microbiota, followed by a discussion of the developmental trajectory of the gut microbiota in healthy children. This review further explored the richness, diversity, and composition of the gut microbiota, as well as specific microbial taxa associated with allergic disease. Lastly, we discussed the current status and future potential of probiotic interventions in managing pediatric allergic diseases.
Collapse
Affiliation(s)
- Heng Ke
- Department of Otolaryngology, The Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Hongbing Yao
- Department of Otolaryngology, The Children's Hospital of Chongqing Medical University, Chongqing, PR China
| | - Ping Wei
- Department of Otolaryngology, The Children's Hospital of Chongqing Medical University, Chongqing, PR China
| |
Collapse
|
42
|
Kerezoudi EN, Saxami G, Zervakis GI, Pletsa V, Brummer RJ, Kyriacou A, Rangel I. Effects of In Vitro Fermented Pleurotus eryngii on Intestinal Barrier Integrity and Immunomodulation in a Lipopolysaccharide-Induced Colonic Model. Biomedicines 2025; 13:430. [PMID: 40002843 PMCID: PMC11853518 DOI: 10.3390/biomedicines13020430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2025] [Revised: 02/05/2025] [Accepted: 02/08/2025] [Indexed: 02/27/2025] Open
Abstract
Background: This study investigates the impact of fermentation supernatants (FSs) from Pleurotus eryngii whole mushrooms (PEWS), as well as its subcomponents, digested (PEWSD) and extracted (PEWSE) forms, on intestinal barrier function and immune modulation in lipopolysaccharide (LPS) -stimulated Caco-2 cells. Methods: Gene expression of tight junction (TJs) genes, cytokines, and key immune/metabolic receptors was assessed via qRT-PCR, while cytokine protein levels were measured using ELISA to explore post-transcriptional regulation. Results: LPS challenge significantly downregulated TJs zonula occludens-1 (ZO-1,) occludin, and claudin-1, compromising epithelial integrity. Treatment with FS-PEWS notably restored ZO-1 and occludin expression, outperforming FS-PEWSD and FS-PEWSE, which only partially mitigated the LPS-induced damage. FS-PEWS further demonstrated potent immunomodulatory effects, upregulating anti-inflammatory IL-10 and pro-inflammatory cytokines such as IL-8 and TNF-α. The activation of key receptors like TLR-2 and mTOR suggests that FS-PEWS modulates critical immune and metabolic pathways, such as NF-kB signaling, to maintain immune homeostasis. Although mRNA expression of pro-inflammatory cytokines was altered, no corresponding protein release was detected, suggesting potential post-transcriptional regulation. Conclusions: FS-PEWS preserves intestinal barrier integrity and modulates immune responses, particularly in low-grade inflammation, highlighting the whole food matrix's role in enhancing its bioactivity and functional food potential.
Collapse
Affiliation(s)
- Evangelia N. Kerezoudi
- Nutrition-Gut-Brain Interactions Research Centre, School of Medical Sciences, Örebro University, 70182 Örebro, Sweden; (R.J.B.); (I.R.)
- Department of Nutrition and Dietetics, Harokopio University, 17676 Athens, Greece; (G.S.); (A.K.)
| | - Georgia Saxami
- Department of Nutrition and Dietetics, Harokopio University, 17676 Athens, Greece; (G.S.); (A.K.)
- Institute of Technology of Agricultural Products, Hellenic Agricultural Organization—DIMITRA (ELGO-DIMITRA), Sof. Venizelou 1, 14123 Athens, Greece
| | - Georgios I. Zervakis
- Laboratory of General and Agricultural Microbiology, Department of Crop Science, Agricultural University of Athens, 11855 Athens, Greece;
| | - Vasiliki Pletsa
- National Hellenic Research Foundation, Institute of Chemical Biology, 11635 Athens, Greece;
| | - Robert J. Brummer
- Nutrition-Gut-Brain Interactions Research Centre, School of Medical Sciences, Örebro University, 70182 Örebro, Sweden; (R.J.B.); (I.R.)
| | - Adamantini Kyriacou
- Department of Nutrition and Dietetics, Harokopio University, 17676 Athens, Greece; (G.S.); (A.K.)
| | - Ignacio Rangel
- Nutrition-Gut-Brain Interactions Research Centre, School of Medical Sciences, Örebro University, 70182 Örebro, Sweden; (R.J.B.); (I.R.)
| |
Collapse
|
43
|
Liu X, Han Z, Ma W, Cui W, Zhen D, Jiang S, Zhang J. Effects of Lactiplantibacillus plantarum HNU082 intervention on fungi and bacteriophages in different intestinal segments of mice. BMC Microbiol 2025; 25:69. [PMID: 39922998 PMCID: PMC11806771 DOI: 10.1186/s12866-025-03784-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 01/24/2025] [Indexed: 02/10/2025] Open
Abstract
BACKGROUND Gut fungi and bacteriophages, as members of the gut microbiota, can affect the interactions between gut bacteria and the host, participate in host metabolism, and are associated with various diseases. Probiotics substantially influence gut fungi and bacteriophages, modulating their composition through both direct and indirect mechanisms, thereby influencing host health. Current research primarily focuses on the effects of probiotics on the intestinal bacterial community. However, the alterations in the compositions of gut fungi and bacteriophages following probiotic intervention are not yet fully understood. Therefore, this study used Lactiplantibacillus plantarum HNU082 (Lp082) as the research subject and aimed to investigate the changes of the gut fungi and bacteriophages in the small intestine and the large intestine after the gavage of Lp082. RESULTS After probiotics entered the gut, the changes of the gut fungi and bacteriophages caused by the probiotics were more pronounced in the small intestine compared to the large intestine. The relative abundance of pathogenic fungi, such as Candida albicans, decreased in the small intestine. Furthermore, a strong positive correlation between the relative abundance of bacteriophages and their host bacteria in the gut was observed. The relative abundance of both Clostridia class bacteria and their bacteriophages increased. CONCLUSIONS In summary, the effects of probiotics on gut fungi and bacteriophages differed between the small intestine and the large intestine. This study contributed to a better understanding of the impact of probiotics on gut fungi and bacteriophages and provided data support for the association and dynamic changes between gut bacteria and their bacteriophages.
Collapse
Affiliation(s)
- Xinlei Liu
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou, 570228, Hainan, China
| | - Zhe Han
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou, 570228, Hainan, China
| | - Wenyao Ma
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou, 570228, Hainan, China
| | - Weipeng Cui
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou, 570228, Hainan, China
| | - Dongyu Zhen
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou, 570228, Hainan, China
| | - Shuaiming Jiang
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou, 570228, Hainan, China.
| | - Jiachao Zhang
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou, 570228, Hainan, China.
- Collaborative Innovation Center of One Health, Hainan University, Haikou, 570228, Hainan, China.
| |
Collapse
|
44
|
Nai S, Song J, Su W, Liu X. Bidirectional Interplay Among Non-Coding RNAs, the Microbiome, and the Host During Development and Diseases. Genes (Basel) 2025; 16:208. [PMID: 40004537 PMCID: PMC11855195 DOI: 10.3390/genes16020208] [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: 12/30/2024] [Revised: 01/24/2025] [Accepted: 01/29/2025] [Indexed: 02/27/2025] Open
Abstract
It is widely known that the dysregulation of non-coding RNAs (ncRNAs) and dysbiosis of the gut microbiome play significant roles in host development and the progression of various diseases. Emerging evidence has highlighted the bidirectional interplay between ncRNAs and the gut microbiome. This article aims to review the current understanding of the molecular mechanisms underlying the crosstalk between ncRNAs, especially microRNA (miRNA), and the gut microbiome in the context of development and diseases, such as colorectal cancer, inflammatory bowel diseases, neurological disorders, obesity, and cardiovascular disease. Ultimately, this review seeks to provide a foundation for exploring the potential roles of ncRNAs and gut microbiome interactions as biomarkers and therapeutic targets for clinical diagnosis and treatment, such as ncRNA mimics, antisense oligonucleotides, and small-molecule compounds, as well as probiotics, prebiotics, and diets.
Collapse
Affiliation(s)
| | | | | | - Xiaoqian Liu
- State Key Laboratory of Microbial Technology, Shandong University, Qingdao 266237, China; (S.N.); (J.S.); (W.S.)
| |
Collapse
|
45
|
Ma X, Li M, Zhang Y, Xu T, Zhou X, Qian M, Yang Z, Han X. Akkermansia muciniphila identified as key strain to alleviate gut barrier injury through Wnt signaling pathway. eLife 2025; 12:RP92906. [PMID: 39912727 PMCID: PMC11801796 DOI: 10.7554/elife.92906] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2025] Open
Abstract
As the largest mucosal surface, the gut has built a physical, chemical, microbial, and immune barrier to protect the body against pathogen invasion. The disturbance of gut microbiota aggravates pathogenic bacteria invasion and gut barrier injury. Fecal microbiota transplantation (FMT) is a promising treatment for microbiome-related disorders, where beneficial strain engraftment is a significant factor influencing FMT outcomes. The aim of this research was to explore the effect of FMT on antibiotic-induced microbiome-disordered (AIMD) models infected with enterotoxigenic Escherichia coli (ETEC). We used piglet, mouse, and intestinal organoid models to explore the protective effects and mechanisms of FMT on ETEC infection. The results showed that FMT regulated gut microbiota and enhanced the protection of AIMD piglets against ETEC K88 challenge, as demonstrated by reduced intestinal pathogen colonization and alleviated gut barrier injury. Akkermansia muciniphila (A. muciniphila) and Bacteroides fragilis (B. fragilis) were identified as two strains that may play key roles in FMT. We further investigated the alleviatory effects of these two strains on ETEC infection in the AIMD mice model, which revealed that A. muciniphila and B. fragilis relieved ETEC-induced intestinal inflammation by maintaining the proportion of Treg/Th17 cells and epithelial damage by moderately activating the Wnt/β-catenin signaling pathway, while the effect of A. muciniphila was better than B. fragilis. We, therefore, identified whether A. muciniphila protected against ETEC infection using basal-out and apical-out intestinal organoid models. A. muciniphila did protect the intestinal stem cells and stimulate the proliferation and differentiation of intestinal epithelium, and the protective effects of A. muciniphila were reversed by Wnt inhibitor. FMT alleviated ETEC-induced gut barrier injury and intestinal inflammation in the AIMD model. A. muciniphila was identified as a key strain in FMT to promote the proliferation and differentiation of intestinal stem cells by mediating the Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Xin Ma
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang UniversityHangzhouChina
- Hainan Institute of Zhejiang University, Yongyou Industry Park, Yazhou Bay Sci-Tech CitySanyaChina
| | - Meng Li
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang UniversityHangzhouChina
- Hainan Institute of Zhejiang University, Yongyou Industry Park, Yazhou Bay Sci-Tech CitySanyaChina
| | - Yuanyuan Zhang
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang UniversityHangzhouChina
| | - Tingting Xu
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang UniversityHangzhouChina
| | - Xinchen Zhou
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang UniversityHangzhouChina
- Hainan Institute of Zhejiang University, Yongyou Industry Park, Yazhou Bay Sci-Tech CitySanyaChina
| | - Mengqi Qian
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang UniversityHangzhouChina
| | - Zhiren Yang
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang UniversityHangzhouChina
- Hainan Institute of Zhejiang University, Yongyou Industry Park, Yazhou Bay Sci-Tech CitySanyaChina
| | - Xinyan Han
- Key Laboratory of Animal Nutrition and Feed Science in East China, Ministry of Agriculture, College of Animal Sciences, Zhejiang UniversityHangzhouChina
- Hainan Institute of Zhejiang University, Yongyou Industry Park, Yazhou Bay Sci-Tech CitySanyaChina
| |
Collapse
|
46
|
Ma ZF, Lee YY. The Role of the Gut Microbiota in Health, Diet, and Disease with a Focus on Obesity. Foods 2025; 14:492. [PMID: 39942085 PMCID: PMC11817362 DOI: 10.3390/foods14030492] [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: 12/31/2024] [Revised: 01/22/2025] [Accepted: 01/27/2025] [Indexed: 02/16/2025] Open
Abstract
The gut microbiota has been increasingly recognised as a critical determinant of human health, influencing a wide range of physiological processes. A healthy gut microbiota is essential for maintaining metabolic, immune, and gastrointestinal homeostasis, contributing to overall well-being. Alterations in its composition and functionality, often referred to as microbial dysbiosis, are strongly associated with the development of gut-related and systemic diseases. The gut microbiota synthesises several components and interacts with epithelial cell receptors, influencing processes that extend beyond nutritional status to the pathogenesis of diseases such as obesity, which extend beyond their known contribution to nutritional status. Therefore, this state-of-the-art review synthesises findings from recent studies on the composition, functions, and influencing factors of the gut microbiota, with a focus on its role in obesity. A systematic search of peer-reviewed literature was conducted to ensure comprehensive coverage, while expert insights are incorporated to discuss emerging research directions and future perspectives in the field.
Collapse
Affiliation(s)
- Zheng Feei Ma
- Centre for Public Health, School of Health and Social Wellbeing, College of Health, Science and Society, University of the West of England, Bristol BS16 1QY, UK
| | - Yeong Yeh Lee
- School of Medical Sciences, University Sains Malaysia, Kota Bharu 15200, Malaysia
| |
Collapse
|
47
|
Richardson M, Zhao S, Lin L, Sheth RU, Qu Y, Lee J, Moody T, Ricaurte D, Huang Y, Velez-Cortes F, Urtecho G, Wang HH. SAMPL-seq reveals micron-scale spatial hubs in the human gut microbiome. Nat Microbiol 2025; 10:527-540. [PMID: 39901058 DOI: 10.1038/s41564-024-01914-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 12/12/2024] [Indexed: 02/05/2025]
Abstract
The local arrangement of microbes can profoundly impact community assembly, function and stability. However, our understanding of the spatial organization of the human gut microbiome at the micron scale is limited. Here we describe a high-throughput and streamlined method called Split-And-pool Metagenomic Plot-sampling sequencing (SAMPL-seq) to capture spatial co-localization in a complex microbial consortium. The method obtains microbial composition of micron-scale subcommunities through split-and-pool barcoding. SAMPL-seq analysis of the healthy human gut microbiome identified bacterial taxa pairs that consistently co-occurred both over time and across multiple individuals. These co-localized microbes organize into spatially distinct groups or 'spatial hubs' dominated by Bacteroidaceae, Ruminococcaceae and Lachnospiraceae families. Using inulin as a dietary perturbation, we observed reversible spatial rearrangement of the gut microbiome where specific taxa form new local partnerships. Spatial metagenomics using SAMPL-seq can unlock insights into microbiomes at the micron scale.
Collapse
Affiliation(s)
- Miles Richardson
- Department of Systems Biology, Columbia University, New York, NY, USA
- Integrated Program in Cellular, Molecular and Biomedical Studies, Columbia University, New York, NY, USA
| | - Shijie Zhao
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Liyuan Lin
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Ravi U Sheth
- Department of Systems Biology, Columbia University, New York, NY, USA
- Kingdom Supercultures, Brooklyn, NY, USA
| | - Yiming Qu
- Department of Systems Biology, Columbia University, New York, NY, USA
- Integrated Program in Cellular, Molecular and Biomedical Studies, Columbia University, New York, NY, USA
| | - Jeongchan Lee
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Thomas Moody
- Department of Systems Biology, Columbia University, New York, NY, USA
- Integrated Program in Cellular, Molecular and Biomedical Studies, Columbia University, New York, NY, USA
| | - Deirdre Ricaurte
- Department of Systems Biology, Columbia University, New York, NY, USA
- Integrated Program in Cellular, Molecular and Biomedical Studies, Columbia University, New York, NY, USA
| | - Yiming Huang
- Department of Systems Biology, Columbia University, New York, NY, USA
- Integrated Program in Cellular, Molecular and Biomedical Studies, Columbia University, New York, NY, USA
| | - Florencia Velez-Cortes
- Department of Systems Biology, Columbia University, New York, NY, USA
- Integrated Program in Cellular, Molecular and Biomedical Studies, Columbia University, New York, NY, USA
| | - Guillaume Urtecho
- Department of Systems Biology, Columbia University, New York, NY, USA
| | - Harris H Wang
- Department of Systems Biology, Columbia University, New York, NY, USA.
- Department of Pathology and Cell Biology, Columbia University, New York, NY, USA.
| |
Collapse
|
48
|
Wyatt NJ, Watson H, Young GR, Doona M, Tilling N, Allerton D, Masi AC, Ahmad T, Doyle JA, Frith K, Hart A, Hildreth V, Irving PM, Jones C, Kennedy NA, Lawrence S, Lees CW, Lees R, Liddle T, Lindsay JO, Marchesi JR, Parkes M, Powell N, Prescott NJ, Raine T, Satsangi J, Whelan K, Wood R, King A, Jostins-Dean L, Speight RA, McGregor N, Stewart CJ, Lamb CA. Evaluation of intestinal biopsy tissue preservation methods to facilitate large-scale mucosal microbiota research. EBioMedicine 2025; 112:105550. [PMID: 39742562 PMCID: PMC11751561 DOI: 10.1016/j.ebiom.2024.105550] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2024] [Revised: 12/13/2024] [Accepted: 12/23/2024] [Indexed: 01/03/2025] Open
Abstract
BACKGROUND Large-scale multicentre studies are needed to understand complex relationships between the gut microbiota, health and disease. Interrogating the mucosal microbiota may identify important biology not captured by stool analysis. Gold standard tissue cryopreservation ('flash freezing') limits large-scale study feasibility. We aimed to compare gut microbiota in gold standard and pragmatic mucosal biopsy storage conditions. METHODS We collected endoscopic recto-sigmoid biopsies from 20 adults with inflammatory bowel disease. Biopsies were preserved using three methods: (i) flash freezing (most proximal and distal biopsy sites); (ii) nucleic acid preservative reagents (QIAGEN Allprotect®, Invitrogen RNAlater™, and Zymo DNA/RNA Shield™); and (iii) formalin fixation with paraffin embedding (FFPE), which is used to preserve tissue for clinical histopathology within healthcare settings. Microbiota were sequenced on the MiSeq platform (V4 region, 16S rRNA gene). FINDINGS Tissue microbiota were consistent between most proximal and distal tissue suggesting any within-patient variation observed reflected storage condition, not biopsy location. There was no significant difference in alpha-diversity or microbial community profiles of reagent-preserved versus gold standard tissue. FFPE community structure was significantly dissimilar to other tissue samples, driven by differential relative abundance of obligate gut anaerobes; Faecalibacterium, Anaerostipes and Lachnospiraceae. Despite these differences, tissue microbiota grouped by participant regardless of preservation and storage conditions. INTERPRETATION Preservative reagents offer a convenient alternative to flash freezing tissue in prospective large-scale mucosal microbiota studies. Whilst less comparable, FFPE provides potential for retrospective microbiota studies using historical samples. FUNDING Medical Research Council (MR/T032162/1) and The Leona M. and Harry B. Helmsley Charitable Trust (G-2002-04255).
Collapse
Affiliation(s)
- Nicola J Wyatt
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom; Department of Gastroenterology, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Hannah Watson
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Gregory R Young
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Mary Doona
- Department of Gastroenterology, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Ned Tilling
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Dean Allerton
- Newcastle Clinical Trials Unit (NCTU), Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Andrea C Masi
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Tariq Ahmad
- Department of Gastroenterology, Royal Devon University Healthcare NHS Foundation Trust, Exeter, United Kingdom; Exeter Inflammatory Bowel Disease and Pharmacogenetics Research Group, University of Exeter, Exeter, United Kingdom
| | - Jennifer A Doyle
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Katherine Frith
- Newcastle Clinical Trials Unit (NCTU), Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Ailsa Hart
- Department of Gastroenterology, St Marks Hospital and Academic Institute, Gastroenterology, London, United Kingdom; Department of Surgery and Cancer, Imperial College, London, United Kingdom
| | - Victoria Hildreth
- Newcastle Clinical Trials Unit (NCTU), Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Peter M Irving
- Department of Gastroenterology, Guy's & St Thomas' NHS Foundation Trust, London, United Kingdom; School of Immunology & Microbial Sciences, King's College London, London, United Kingdom
| | - Claire Jones
- Department of Histopathology, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Nicholas A Kennedy
- Department of Gastroenterology, Royal Devon University Healthcare NHS Foundation Trust, Exeter, United Kingdom; Exeter Inflammatory Bowel Disease and Pharmacogenetics Research Group, University of Exeter, Exeter, United Kingdom
| | - Sarah Lawrence
- Newcastle Clinical Trials Unit (NCTU), Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Charlie W Lees
- Institute of Genetics & Molecular Medicine, University of Edinburgh, Edinburgh, United Kingdom; Edinburgh IBD Unit, Western General Hospital, NHS Lothian, Edinburgh, United Kingdom
| | - Robert Lees
- Department of Gastroenterology, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Trevor Liddle
- Research Informatics Team, Clinical Research, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - James O Lindsay
- Department of Gastroenterology, Barts Health NHS Trust, The Royal London Hospital, London, United Kingdom; Centre for Immunobiology, Blizard Institute, Barts and the London School of Medicine and Dentistry, Queen Mary University of London, London, United Kingdom
| | - Julian R Marchesi
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, St Mary's Hospital, Imperial College London, London, United Kingdom
| | - Miles Parkes
- Department of Gastroenterology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Nick Powell
- Division of Digestive Diseases, Department of Metabolism, Digestion and Reproduction, St Mary's Hospital, Imperial College London, London, United Kingdom; Department of Gastroenterology, Imperial College Healthcare NHS Trust, London, United Kingdom
| | - Natalie J Prescott
- Department of Medical and Molecular Genetics, King's College London, Guy's Hospital, London, United Kingdom
| | - Tim Raine
- Department of Gastroenterology, Cambridge University Hospitals NHS Foundation Trust, Cambridge, United Kingdom
| | - Jack Satsangi
- Nuffield Department of Medicine, University of Oxford, Old Road Campus, Oxford, United Kingdom
| | - Kevin Whelan
- Department of Nutritional Sciences, King's College London, London, United Kingdom
| | - Ruth Wood
- Newcastle Clinical Trials Unit (NCTU), Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Andrew King
- Department of Gastroenterology, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Luke Jostins-Dean
- Kennedy Institute of Rheumatology, University of Oxford, Oxford, United Kingdom
| | - R Alexander Speight
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom; Department of Gastroenterology, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom
| | - Naomi McGregor
- Newcastle Clinical Trials Unit (NCTU), Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Christopher J Stewart
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Christopher A Lamb
- Translational & Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom; Department of Gastroenterology, The Newcastle upon Tyne Hospitals NHS Foundation Trust, Newcastle upon Tyne, United Kingdom.
| |
Collapse
|
49
|
Marwaha K, Cain R, Asmis K, Czaplinski K, Holland N, Mayer DCG, Chacon J. Exploring the complex relationship between psychosocial stress and the gut microbiome: implications for inflammation and immune modulation. J Appl Physiol (1985) 2025; 138:518-535. [PMID: 39813028 DOI: 10.1152/japplphysiol.00652.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2024] [Revised: 12/16/2024] [Accepted: 01/07/2025] [Indexed: 01/16/2025] Open
Abstract
There is growing interest in understanding the complex relationship between psychosocial stress and the human gastrointestinal microbiome (GIM). This review explores the potential physiological pathways connecting these two and how they contribute to a proinflammatory environment that can lead to the development and progression of the disease. Exposure to psychosocial stress triggers the activation of the sympathetic nervous system (SNS) and hypothalamic-pituitary axis (HPA), leading to various physiological responses essential for survival and coping with the stressor. However, chronic stress in susceptible individuals could cause sustained activation of HPA and SNS, leading to immune dysregulation consisting of redistribution of natural killer (NK) cells in the bloodstream, decreased function of T and B cells, and elevation of proinflammatory cytokines such as interleukin-1, interleukin-6, tumor necrotic factor-α, interferon-gamma. It also leads to disruption of the GIM composition and increased intestinal barrier permeability, contributing to GIM dysbiosis. The GIM dysbiosis and elevated cytokines can lead to reciprocal effects and further stimulate the HPA and SNS, creating a positive feedback loop that results in a proinflammatory state underlying the pathogenesis and progression of stress-associated cardiovascular, gastrointestinal, autoimmune, and psychiatric disorders. Understanding these relationships is critical for developing new strategies for managing stress-related health disorders.
Collapse
Affiliation(s)
- Komal Marwaha
- Department of Medical Education, Paul L Foster School of Medicine, Texas Tech University Health Science Center, El Paso, Texas, United States
| | - Ryan Cain
- Department of Medical Education, Paul L Foster School of Medicine, Texas Tech University Health Science Center, El Paso, Texas, United States
| | - Katherine Asmis
- Department of Medical Education, Paul L Foster School of Medicine, Texas Tech University Health Science Center, El Paso, Texas, United States
| | - Katya Czaplinski
- Department of Medical Education, Paul L Foster School of Medicine, Texas Tech University Health Science Center, El Paso, Texas, United States
| | - Nathan Holland
- Department of Medical Education, Paul L Foster School of Medicine, Texas Tech University Health Science Center, El Paso, Texas, United States
| | - Darly C Ghislaine Mayer
- Department of Medical Education, Paul L Foster School of Medicine, Texas Tech University Health Science Center, El Paso, Texas, United States
| | - Jessica Chacon
- Department of Medical Education, Paul L Foster School of Medicine, Texas Tech University Health Science Center, El Paso, Texas, United States
| |
Collapse
|
50
|
Azimzadeh PN, Birchenough GM, Gualbuerto NC, Pinkner JS, Tamadonfar KO, Beatty W, Hannan TJ, Dodson KW, Ibarra EC, Kim S, Schreiber HL, Janetka JW, Kau AL, Earl AM, Miller MJ, Hansson GC, Hultgren SJ. Mechanisms of uropathogenic E. coli mucosal association in the gastrointestinal tract. SCIENCE ADVANCES 2025; 11:eadp7066. [PMID: 39888987 PMCID: PMC11784811 DOI: 10.1126/sciadv.adp7066] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/09/2024] [Accepted: 12/31/2024] [Indexed: 02/02/2025]
Abstract
Urinary tract infections (UTIs) are highly recurrent and frequently caused by Uropathogenic Escherichia coli (UPEC) strains that can be found in patient intestines. Seeding of the urinary tract from this intestinal reservoir likely contributes to UTI recurrence (rUTI) rates. Thus, understanding the factors that promote UPEC intestinal colonization is of critical importance to designing therapeutics to reduce rUTI incidence. Although E. coli is found in high abundance in large intestine mucus, little is known about how it is able to maintain residence in this continuously secreted hydrogel. We discovered that the FimH adhesin of type 1 pili (T1P) bound throughout the secreted mucus layers of the colon and to epithelial cells in mouse and human samples. Disruption of T1P led to reduced association with colon mucus. Notably, this mutant up-regulated flagellar production and infiltrated the protective inner mucus layer of the colon. This could explain how UPEC resists being washed off by the continuously secreted mucus layers of the colon.
Collapse
Affiliation(s)
- Philippe N. Azimzadeh
- Department of Molecular Microbiology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - George M. Birchenough
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden; Wallenberg Centre for Molecular and Translational Medicine, University of Gothenburg, Gothenburg, Sweden
| | - Nathaniel C. Gualbuerto
- Department of Molecular Microbiology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Jerome S. Pinkner
- Department of Molecular Microbiology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Kevin O. Tamadonfar
- Department of Molecular Microbiology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Wandy Beatty
- Department of Molecular Microbiology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Thomas J. Hannan
- Department of Molecular Microbiology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Karen W. Dodson
- Department of Molecular Microbiology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
| | - Enid C. Ibarra
- Department of Internal Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Seonyoung Kim
- Department of Internal Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Henry L. Schreiber
- Department of Molecular Microbiology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
- Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, USA
| | - James W. Janetka
- Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biochemistry and Molecular Biophysics, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Andrew L. Kau
- Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, USA
- Division of Allergy and Immunology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Ashlee M. Earl
- Infectious Disease and Microbiome Program, Broad Institute, Cambridge, MA 02142, USA
| | - Mark J. Miller
- Department of Internal Medicine, Division of Infectious Diseases, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Gunnar C. Hansson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, University of Gothenburg, Gothenburg, Sweden
| | - Scott J. Hultgren
- Department of Molecular Microbiology, Washington University in St. Louis, School of Medicine, St. Louis, MO 63110, USA
- Center for Women’s Infectious Disease Research, Washington University School of Medicine, St. Louis, MO, USA
| |
Collapse
|