1
|
Tram NDT, Xu J, Chan KH, Rajamani L, Ee PLR. Bacterial clustering biomaterials as anti-infective therapies. Biomaterials 2025; 316:123017. [PMID: 39708775 DOI: 10.1016/j.biomaterials.2024.123017] [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: 08/13/2024] [Revised: 11/23/2024] [Accepted: 12/13/2024] [Indexed: 12/23/2024]
Abstract
In Nature, bacterial clustering by host-released peptides or nucleic acids is an evolutionarily conserved immune defense strategy employed to prevent adhesion of pathogenic microbes, which is prerequisite for most infections. Synthetic anti-adhesion strategies present as non-lethal means of targeting bacteria and may potentially be used to avoid resistance against antimicrobial therapies. From bacteria-agglutinating biomolecules discovered in nature to synthetic designs involving peptides, cationic polymers and nanoparticles, the modes of actions appear broad and unconsolidated. Herein, we present a critical review and update of the state-of-the-art in synthetic bacteria-clustering designs with proposition of a more streamlined nomenclature and classification. Overall, this review aims to consolidate the conceptual framework in the field of bacterial clustering and highlight its potentials as an avenue for discovering novel antibacterial biomaterials.
Collapse
Affiliation(s)
- Nhan Dai Thien Tram
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, 18 Science Drive 4, Singapore, 117559, Singapore
| | - Jian Xu
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, 18 Science Drive 4, Singapore, 117559, Singapore
| | - Kiat Hwa Chan
- Division of Science, Yale-NUS College, 16 College Avenue West, Singapore, 138527, Singapore; NUS College, National University of Singapore, 18 College Avenue East, Singapore, 138593, Singapore
| | - Lakshminarayanan Rajamani
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, 18 Science Drive 4, Singapore, 117559, Singapore; Ocular Infections and Anti-Microbials Research Group, Singapore Eye Research Institute, Singapore, 169856, Singapore; Ophthalmology and Visual Sciences Academic Clinical Program, Duke-NUS Graduate Medical School, Singapore, 169857, Singapore
| | - Pui Lai Rachel Ee
- Department of Pharmacy and Pharmaceutical Sciences, National University of Singapore, 18 Science Drive 4, Singapore, 117559, Singapore.
| |
Collapse
|
2
|
Schille TB, Sprague JL, Naglik JR, Brunke S, Hube B. Commensalism and pathogenesis of Candida albicans at the mucosal interface. Nat Rev Microbiol 2025:10.1038/s41579-025-01174-x. [PMID: 40247134 DOI: 10.1038/s41579-025-01174-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 03/18/2025] [Indexed: 04/19/2025]
Abstract
Fungi are important and often underestimated human pathogens. Infections with fungi mostly originate from the environment, from soil or airborne spores. By contrast, Candida albicans, one of the most common and clinically important fungal pathogens, permanently exists in the vast majority of healthy individuals as a member of the human mucosal microbiota. Only under certain circumstances will these commensals cause infections. However, although the pathogenic behaviour and disease manifestation of C. albicans have been at the centre of research for many years, its asymptomatic colonization of mucosal surfaces remains surprisingly understudied. In this Review, we discuss the interplay of the fungus, the host and the microbiome on the dualism of commensal and pathogenic life of C. albicans, and how commensal growth is controlled and permitted. We explore hypotheses that could explain how the mucosal environment shapes C. albicans adaptations to its commensal lifestyle, while still maintaining or even increasing its pathogenic potential.
Collapse
Affiliation(s)
- Tim B Schille
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany
| | - Jakob L Sprague
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany
| | - Julian R Naglik
- Centre for Host-Microbiome Interactions, Faculty of Dentistry, Oral and Craniofacial Sciences, King's College London, London, UK
| | - Sascha Brunke
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany.
| | - Bernhard Hube
- Department of Microbial Pathogenicity Mechanisms, Leibniz Institute for Natural Product Research and Infection Biology-Hans Knöll Institute (HKI), Jena, Germany.
- Cluster of Excellence Balance of the Microverse, Friedrich Schiller University Jena, Jena, Germany.
- Institute of Microbiology, Friedrich Schiller University Jena, Jena, Germany.
| |
Collapse
|
3
|
Kowalski CH, Nguyen UT, Lawhorn S, Smith TJ, Corrigan RM, Suh WS, Kalan L, Barber MF. Skin mycobiota-mediated antagonism against Staphylococcus aureus through a modified fatty acid. Curr Biol 2025:S0960-9822(25)00371-9. [PMID: 40233753 DOI: 10.1016/j.cub.2025.03.055] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 02/17/2025] [Accepted: 03/21/2025] [Indexed: 04/17/2025]
Abstract
Microbiota promote host health by inhibiting pathogen colonization, yet how host-resident fungi or mycobiota contribute to this process remains unclear. The human skin mycobiota is uniquely stable compared with other body sites and dominated by skin-adapted yeasts of the genus Malassezia. We observe that colonization of human skin by Malassezia sympodialis significantly reduces subsequent colonization by the prominent bacterial pathogen Staphylococcus aureus. In vitro, M. sympodialis generates a hydroxyl palmitic acid isomer from environmental sources that has potent bactericidal activity against S. aureus in the context of skin-relevant stressors and is sufficient to impair S. aureus skin colonization. Leveraging experimental evolution to pinpoint mechanisms of S. aureus adaptation in response to antagonism by Malassezia, we identified multiple mutations in the stringent response regulator Rel that promote survival against M. sympodialis and provide a competitive advantage on human skin when M. sympodialis is present. Similar Rel alleles have been reported in S. aureus clinical isolates, and natural Rel variants are sufficient for tolerance to M. sympodialis antagonism. Partial stringent response activation underlies tolerance to clinical antibiotics, with both laboratory-evolved and natural Rel variants conferring multidrug tolerance in a manner that is dependent on the alternative sigma factor SigB. These findings demonstrate the ability of the mycobiota to mediate pathogen colonization resistance through generation of a hydroxy palmitic acid isomer, identify new mechanisms of bacterial adaptation in response to microbiota antagonism, and reveal the potential for microbiota-driven evolution to shape pathogen antibiotic susceptibility.
Collapse
Affiliation(s)
- Caitlin H Kowalski
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR 97403, USA.
| | - Uyen Thy Nguyen
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada; Microbiology Doctoral Training Program, University of Wisconsin-Madison, Madison, WI 53705, USA
| | - Susannah Lawhorn
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA
| | - T Jarrod Smith
- Institute of Molecular Biology, University of Oregon, Eugene, OR 97403, USA
| | - Rebecca M Corrigan
- Florey Institute, School of Biosciences, University of Sheffield, Sheffield S10 2TN, UK; The School of Medicine, University College Dublin, Belfield, Dublin 4, Dublin D04 V1W8, Ireland
| | - Won Se Suh
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada; M.G. DeGroote Institute for Infectious Disease Research, David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Lindsay Kalan
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, ON L8N 3Z5, Canada; Department of Medical Microbiology and Immunology, School of Medicine and Public Health, University of Wisconsin-Madison, Madison, WI 53706, USA; M.G. DeGroote Institute for Infectious Disease Research, David Braley Centre for Antibiotic Discovery, McMaster University, Hamilton, ON L8N 3Z5, Canada
| | - Matthew F Barber
- Institute of Ecology and Evolution, University of Oregon, Eugene, OR 97403, USA; Department of Biology, University of Oregon, Eugene, OR 97403, USA.
| |
Collapse
|
4
|
Zhang J, Yang J, Luo J, Wu W, Luo H, Wei W, Lyu H, Wang Y, Yi H, Zhang Y, Fan Z, Lyu H, Kanakaveti VP, Qin B, Yuan P, Yang R, Zhang H, Zuo T, Felsher DW, Lee MH, Li K. Lactobacillus acidophilus potentiates oncolytic virotherapy through modulating gut microbiota homeostasis in hepatocellular carcinoma. Nat Commun 2025; 16:3315. [PMID: 40195307 PMCID: PMC11976979 DOI: 10.1038/s41467-025-58407-z] [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: 03/20/2025] [Indexed: 04/09/2025] Open
Abstract
Oncolytic viruses (OVs) hold promise for cancer treatment. However, the antitumor efficacy is limited. Microbiota plays a pivotal role in cancer treatment and its impact on oncolytic virotherapy is unknown. Here, we show that VSVΔ51 has higher antitumor efficacy for hepatocellular carcinoma in the absence of microbiota in female mouse models. VSVΔ51 infection causes microbiota dysbiosis, increasing most of the gut bacteria abundance, while decreasing the commensal Lactobacillus. VSVΔ51 reduced intestinal expression of SLC20A1 that binds to Lactobacillus acidophilus (L. acidophilus) CdpA cell wall protein through IL6-JAK-STAT3 signaling, thereby attenuating attachment and colonization of L. acidophilus. L. acidophilus supplementation confers sensitivity to VSVΔ51 through restoring gut barrier integrity and microbiota homeostasis destroyed by VSVΔ51. In this work, we show that targeting microbiota homostasis holds substantial potential in improving therapeutic outcomes of oncolytic virotherapy.
Collapse
Affiliation(s)
- Jiayu Zhang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jinneng Yang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jinyan Luo
- The First Affiliated Hospital of Nanjing Medical University, 300 Guangzhou Road, Nanjing, 210029, China
| | - Weili Wu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Department of Clinical Laboratory, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Haidan Luo
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Wenxia Wei
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Haimei Lyu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yuzhi Wang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Hairong Yi
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Yijing Zhang
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Zongmin Fan
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Haiwen Lyu
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | | | - Baifu Qin
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Ping Yuan
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Runxiang Yang
- Department of the Second Medical Oncology, The Third Affiliated Hospital of Kunming Medical University, 519, Kunzhou Road, Kunming, 650118, China
| | - Haipeng Zhang
- Department of Pharmacology, School of Medicine, Jinan University, Guangzhou, Guangdong, 510632, China
| | - Tao Zuo
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Dean W Felsher
- Division of Oncology, Department of Medicine, Stanford University, Stanford, CA, 94305, USA
| | - Mong-Hong Lee
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China.
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China.
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| | - Kai Li
- Guangdong Provincial Key Laboratory of Colorectal and Pelvic Floor Disease, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China.
- Guangdong Research Institute of Gastroenterology, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, 510655, China.
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China.
- Biomedical Innovation Center, The Sixth Affiliated Hospital, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
5
|
Heidrich V, Valles-Colomer M, Segata N. Human microbiome acquisition and transmission. Nat Rev Microbiol 2025:10.1038/s41579-025-01166-x. [PMID: 40119155 DOI: 10.1038/s41579-025-01166-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2025] [Indexed: 03/24/2025]
Abstract
As humans, we host personal microbiomes intricately connected to our biology and health. Far from being isolated entities, our microbiomes are dynamically shaped by microbial exchange with the surroundings, in lifelong microbiome acquisition and transmission processes. In this Review, we explore recent studies on how our microbiomes are transmitted, beginning at birth and during interactions with other humans and the environment. We also describe the key methodological aspects of transmission inference, based on the uniqueness of the building blocks of the microbiome - single microbial strains. A better understanding of human microbiome transmission will have implications for studies of microbial host regulation, of microbiome-associated diseases, and for effective microbiome-targeting strategies. Besides exchanging strains with other humans, there is also preliminary evidence we acquire microorganisms from animals and food, and thus a complete understanding of microbiome acquisition and transmission can only be attained by adopting a One Health perspective.
Collapse
Affiliation(s)
| | | | - Nicola Segata
- Department CIBIO, University of Trento, Trento, Italy.
- Department of Experimental Oncology, IEO European Institute of Oncology IRCCS, Milan, Italy.
- Department of Twins Research and Genetic Epidemiology, King's College London, London, UK.
| |
Collapse
|
6
|
Huang Y, Liao L, Jiang Y, Tao S, Tang D. Role of gut microbiota in predicting chemotherapy-induced neutropenia duration in leukemia patients. Front Microbiol 2025; 16:1507336. [PMID: 40177485 PMCID: PMC11961871 DOI: 10.3389/fmicb.2025.1507336] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Accepted: 02/18/2025] [Indexed: 04/05/2025] Open
Abstract
Background Acute leukemia is an aggressive malignancy with high morbidity and mortality, and chemotherapy is the primary treatment modality. However, chemotherapy often induces neutropenia (chemotherapy-induced neutropenia, CIN), increasing the risk of infectious complications and mortality. Current research suggests that gut microbiota may play a significant role in chemotherapy's efficacy and side effects. Objective This study aimed to investigate whether gut microbiota can predict the duration of chemotherapy-induced neutropenia in leukemia patients. Methods We included 56 leukemia patients from the Hematology Department of the Second Affiliated Hospital of Nanchang University, collecting fecal samples 1 day before and 1 day after chemotherapy. The diversity and community structure of gut microbiota were analyzed using 16S rRNA gene sequencing. Patients were divided into two groups based on the duration of neutropenia post-chemotherapy: Neutropenia ≤7 Days Group (NLE7 Group) and Neutropenia > 7 Days Group (NGT7 Group). Comparative analysis identified characteristic microbiota. Results After chemotherapy, gut microbiota diversity significantly decreased (p < 0.05). In the NGT7 Group, the relative abundance of Enterococcus before chemotherapy was significantly higher than in the NLE7 Group (p < 0.05). ROC curve analysis showed that the relative abundance of Enterococcus had high predictive accuracy for the duration of neutropenia (AUC = 0.800, 95% CI: 0.651-0.949). Conclusion The abundance of Enterococcus before chemotherapy can predict the duration of chemotherapy-induced neutropenia. These findings provide new evidence for gut microbiota as a predictive biomarker for chemotherapy side effects and may guide personalized treatment for leukemia patients.
Collapse
Affiliation(s)
- Yezi Huang
- Department of Hematology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Hematological Diseases, Nanchang, China
| | - Lihong Liao
- Department of Hematology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Hematological Diseases, Nanchang, China
| | - Yanjun Jiang
- Department of Hematology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Hematological Diseases, Nanchang, China
| | - Si Tao
- Department of Oncology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
| | - Duozhuang Tang
- Department of Hematology, The Second Affiliated Hospital of Nanchang University, Nanchang, China
- Jiangxi Provincial Key Laboratory of Hematological Diseases, Nanchang, China
| |
Collapse
|
7
|
Ding W, Cheng Y, Liu X, Zhu Z, Wu L, Gao J, Lei W, Li Y, Zhou X, Wu J, Gao Y, Ling Z, Jiang R. Harnessing the human gut microbiota: an emerging frontier in combatting multidrug-resistant bacteria. Front Immunol 2025; 16:1563450. [PMID: 40165964 PMCID: PMC11955657 DOI: 10.3389/fimmu.2025.1563450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2025] [Accepted: 02/25/2025] [Indexed: 04/02/2025] Open
Abstract
Antimicrobial resistance (AMR) has become a major and escalating global health threat, undermining the effectiveness of current antibiotic and antimicrobial therapies. The rise of multidrug-resistant bacteria has led to increasingly difficult-to-treat infections, resulting in higher morbidity, mortality, and healthcare costs. Tackling this crisis requires the development of novel antimicrobial agents, optimization of current therapeutic strategies, and global initiatives in infection surveillance and control. Recent studies highlight the crucial role of the human gut microbiota in defending against AMR pathogens. A balanced microbiota protects the body through mechanisms such as colonization resistance, positioning it as a key ally in the fight against AMR. In contrast, gut dysbiosis disrupts this defense, thereby facilitating the persistence, colonization, and dissemination of resistant pathogens. This review will explore how gut microbiota influence drug-resistant bacterial infections, its involvement in various types of AMR-related infections, and the potential for novel microbiota-targeted therapies, such as fecal microbiota transplantation, prebiotics, probiotics, phage therapy. Elucidating the interactions between gut microbiota and AMR pathogens will provide critical insights for developing novel therapeutic strategies to prevent and treat AMR infections. While previous reviews have focused on the general impact of the microbiota on human health, this review will specifically look at the latest research on the interactions between the gut microbiota and the evolution and spread of AMR, highlighting potential therapeutic strategies.
Collapse
Affiliation(s)
- Wenwen Ding
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
- Medical School of Nantong University, Nantong, Jiangsu, China
| | - Yiwen Cheng
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xia Liu
- Department of Intensive Care Unit, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Zhangcheng Zhu
- Department of Preventive Medicine, School of Public Health and Management, Wenzhou Medical University, Wenzhou, Zhejiang, China
| | - Lingbin Wu
- Department of Intensive Care Unit, Lishui Second People’s Hospital, Lishui, Zhejiang, China
| | - Jie Gao
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Wenhui Lei
- Jinan Microecological Biomedicine Shandong Laboratory, Jinan, Shandong, China
| | - Yating Li
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Xin Zhou
- Department of Genetics, Stanford University School of Medicine, Stanford, CA, United States
- Stanford Center for Genomics and Personalized Medicine, Stanford, CA, United States
- Stanford Diabetes Research Center, Stanford, CA, United States
- The Jackson Laboratory for Genomic Medicine, Farmington, CT, United States
| | - Jian Wu
- Department of Clinical Laboratory, Suzhou Municipal Hospital, Suzhou, Jiangsu, China
| | - Yongtao Gao
- Department of Anesthesiology, Affiliated Hospital of Nantong University, Nantong, Jiangsu, China
- Medical School of Nantong University, Nantong, Jiangsu, China
| | - Zongxin Ling
- Collaborative Innovation Center for Diagnosis and Treatment of Infectious Diseases, State Key Laboratory for Diagnosis and Treatment of Infectious Diseases, National Clinical Research Center for Infectious Diseases, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, China
| | - Ruilai Jiang
- Department of Intensive Care Unit, Lishui Second People’s Hospital, Lishui, Zhejiang, China
| |
Collapse
|
8
|
Cherrak Y, Younes AA, Perez-Molphe-Montoya E, Maurer L, Yilmaz K, Enz U, Zeder C, Kiefer P, Christen P, Gül E, Vorholt JA, von Mering C, Hardt WD. Neutrophil recruitment during intestinal inflammation primes Salmonella elimination by commensal E. coli in a context-dependent manner. Cell Host Microbe 2025; 33:358-372.e4. [PMID: 40023150 DOI: 10.1016/j.chom.2025.02.004] [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: 08/16/2024] [Revised: 12/19/2024] [Accepted: 02/05/2025] [Indexed: 03/04/2025]
Abstract
Foodborne bacterial diarrhea involves complex pathogen-microbiota-host interactions. Pathogen-displacing probiotics are increasingly popular, but heterogeneous patient outcomes highlighted the need to understand individualized host-probiotic activity. Using the mouse gut commensal Escherichia coli 8178 and the human probiotic E. coli Nissle 1917, we found that the degree of protection against the enteric pathogen Salmonella enterica serovar Typhimurium (S. Tm) varies across mice with distinct gut microbiotas. Pathogen clearance is linked to enteropathy severity and subsequent recruitment of intraluminal neutrophils, which differs in a microbiota-dependent manner. By combining mouse knockout and antibody-mediated depletion models with bacterial genetics, we show that neutrophils and host-derived reactive oxygen species directly influence E. coli-mediated S. Tm displacement by potentiating siderophore-bound toxin killing. Our work demonstrates how host immune factors shape pathogen-displacing probiotic efficiency while also revealing an unconventional antagonistic interaction where a gut commensal and the host synergize to displace an enteric pathogen.
Collapse
Affiliation(s)
- Yassine Cherrak
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland.
| | - Andrew Abi Younes
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Eugenio Perez-Molphe-Montoya
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, 8057 Zurich, Switzerland
| | - Luca Maurer
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Koray Yilmaz
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Ursina Enz
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Christophe Zeder
- Laboratory of Nutrition and Metabolic Epigenetics, Department of Health Science and Technology, 8092 Zurich, Switzerland
| | - Patrick Kiefer
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Philipp Christen
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Ersin Gül
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Julia A Vorholt
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland
| | - Christian von Mering
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, 8057 Zurich, Switzerland
| | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, ETH Zurich, 8093 Zurich, Switzerland.
| |
Collapse
|
9
|
Saad MJA, Santos A. The Microbiota and Evolution of Obesity. Endocr Rev 2025; 46:300-316. [PMID: 39673174 PMCID: PMC11894537 DOI: 10.1210/endrev/bnae033] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/18/2024] [Revised: 09/03/2024] [Accepted: 12/12/2024] [Indexed: 12/16/2024]
Abstract
Obesity is a major global concern and is generally attributed to a combination of genetic and environmental factors. Several hypotheses have been proposed to explain the evolutionary origins of obesity epidemic, including thrifty and drifty genotypes, and changes in thermogenesis. Here, we put forward the hypothesis of metaflammation, which proposes that due to intense selection pressures exerted by environmental pathogens, specific genes that help develop a robust defense mechanism against infectious diseases have had evolutionary advantages and that this may contribute to obesity in modern times due to connections between the immune and energy storage systems. Indeed, incorporating the genetic variations of gut microbiota into the complex genetic framework of obesity makes it more polygenic than previously believed. Thus, uncovering the evolutionary origins of obesity requires a multifaceted approach that considers the complexity of human history, the unique genetic makeup of different populations, and the influence of gut microbiome on host genetics.
Collapse
Affiliation(s)
- Mario J A Saad
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, CEP 13083-887 Campinas, SP, Brazil
| | - Andrey Santos
- Department of Internal Medicine, School of Medical Sciences, University of Campinas, CEP 13083-887 Campinas, SP, Brazil
| |
Collapse
|
10
|
Ku JY, Lee MJ, Jung Y, Choi HJ, Park J. Changes in the gut microbiome due to diarrhea in neonatal Korean indigenous calves. Front Microbiol 2025; 16:1511430. [PMID: 40109976 PMCID: PMC11921620 DOI: 10.3389/fmicb.2025.1511430] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2024] [Accepted: 02/17/2025] [Indexed: 03/22/2025] Open
Abstract
Studies on gut microbiome changes in neonatal Korean indigenous calves with diarrhea are rare. In this study, 14 normal calves and 11 calves with diarrhea were selected from Korean indigenous calves up to 30 days of age and classified into three groups at 10-day intervals (1-10, 11-20, and 21-30 days). Feces from 25 calves were collected, and the diversity, similarity, structure, and correlation of the gut microbiome were analyzed. Firmicutes, Bacteroidetes, and Proteobacteria were predominant in the taxonomic composition of the gut microbiome of the calves regardless of the presence of diarrhea. However, Proteobacteria increased and Bacteroidetes and Actinobacteria decreased in calves with diarrhea. In addition, calves with diarrhea showed a significant decrease in the diversity of the gut microbiome, especially for anaerobic microorganisms Faecalibacterium prausnitzii, Gemmiger formicilis, and Collinsella aerofaciens. The microbial communities in calves with diarrhea and normal calves were distinct. By analyzing the microorganisms that showed correlation with diarrhea and age using linear discriminant analysis effect size, at the genus level, Prevotella and Lachnospiraceae_uc were significantly related in the normal (11-20 days) group whereas Enterobacterales, Gammaproteobacteria, Enterobacteriaceae, Escherichia, and Proteobacteria were significantly associated with diarrhea in the 11-20 days group. Futhermore, the normal (21-30 days) group showed significant correlation with Blautia, Provotellaceae, Muribaculaceae, Christensenellaceae, and Catenella, whereas the diarrhea (21-30 days) group showed significant correlation with Dorea. The microorganisms associated with diarrhea in calves were mainly known as harmful microorganisms, we confirmed that there is a relationship between the increase in harmful bacteria and diarrhea. These results show that diarrhea significantly affects the gut microbiome of Korean indigenous calves. The changes in the gut microbiome of Korean indigenous calves observed in this study could be helpful in predicting and managing diarrhea calves, and furthermore, in establishing preventive measures for calf diarrhea through management of gut microbiome.
Collapse
Affiliation(s)
- Ji-Yeong Ku
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Jeonbuk University, Iksan, Republic of Korea
| | - Mi-Jin Lee
- Department of Veterinary Nursing, College of Health Science, Wonkwang University, Iksan, Republic of Korea
| | - Youngwoo Jung
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Jeonbuk University, Iksan, Republic of Korea
| | - Hak-Jong Choi
- Technology Innovation Research Division, World Institute of Kimchi, Gwangju, Republic of Korea
| | - Jinho Park
- Department of Veterinary Internal Medicine, College of Veterinary Medicine, Jeonbuk University, Iksan, Republic of Korea
| |
Collapse
|
11
|
Meng JX, Li MH, Wang XY, Li S, Zhang Y, Ni HB, Ma H, Liu R, Yan JC, Li XM, Sun YZ, Yang X, Zhang XX. Temporal variability in the diversity, function and resistome landscapes in the gut microbiome of broilers. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2025; 292:117976. [PMID: 40037072 DOI: 10.1016/j.ecoenv.2025.117976] [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: 10/03/2024] [Revised: 02/15/2025] [Accepted: 02/24/2025] [Indexed: 03/06/2025]
Abstract
Understanding the dynamic and stability of gut microbiota over the course of production cycle of broiler chicken can help identify microbial features that associate with better health and productivity. In the present study, we profile the changes in the composition and stability of gut microbiota of commercially raised broilers at nine distinct time points using shotgun metagenomics and culturomics approaches. We demonstrate, within the first week post-hatching, a rapid decline in relative abundance of 122 pioneer microbial species including Bacteroides fragilis, Lachnospira eligens and Ruminococcus gnavus, accompanied by a substantial decrease in both microbial richness and diversity. This was followed by a gradual increase and stabilization in the microbial diversity and population structure that persisted until the broilers reached the marketing age. Throughout the production cycle, key bacterial families such as Lachnospiraceae, Bacteroidaceae, and Ruminococcaceae were identified. However, significant shifts at the lower taxonomic levels occurred at different production stages, influencing the functional capacities and resistance profiles of the microbiota. During the rapid growth phase, enzymes crucial to vitamin and amino acid metabolism dominated, whereas enzymes associated with carbohydrate and energy metabolism were notably more abundant during the fattening stage. Many predicted antibiotic resistance genes were detected in association with typical commensal bacterial species in the gut microbiota, indicating a sustained resistance of the gut microbiota to antibiotic classes such as aminoglycosides and tetracyclines, which persist even in the absence of antibiotic selection pressure. Our research carries important implications for the management and health surveillance of broiler production.
Collapse
Affiliation(s)
- Jin-Xin Meng
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province 266109, PR China
| | - Ming-Han Li
- College of Animal Medicine, Jilin Agricultural University Changchun, Jilin Province 130118, PR China
| | - Xiang-Yu Wang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province 266109, PR China
| | - Shenghui Li
- Puensum Genetech Institute, Wuhan, Hubei Province 430223, PR China
| | - Yue Zhang
- Puensum Genetech Institute, Wuhan, Hubei Province 430223, PR China
| | - Hong-Bo Ni
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province 266109, PR China
| | - He Ma
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province 266109, PR China
| | - Rui Liu
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province 266109, PR China
| | - Jin-Chu Yan
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province 266109, PR China
| | - Xiao-Man Li
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province 266109, PR China
| | - Yu-Zhe Sun
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province 266109, PR China
| | - Xing Yang
- Department of Medical Microbiology and Immunology, School of Basic Medicine, Dali University, Dali, Yunnan Province 671000, PR China
| | - Xiao-Xuan Zhang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province 266109, PR China.
| |
Collapse
|
12
|
Younge N, Patel RM. Probiotics and the Risk of Infection. Clin Perinatol 2025; 52:87-100. [PMID: 39892956 PMCID: PMC11789005 DOI: 10.1016/j.clp.2024.10.006] [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: 02/04/2025]
Abstract
Probiotic use has increased in preterm infants and may reduce the risk of necrotizing enterocolitis. Probiotic-associated infection is a concern for infants receiving probiotic supplementation in the neonatal intensive care unit, as highlighted by a recent case and subsequent action by the United States Food and Drug Administration. Based on reports to date, invasive infection is an infrequent but known risk of probiotic supplementation. In this article, we discuss the epidemiology and pathophysiology of invasive infection in preterm infants, review the benefits and risks of probiotic as regulations and available products continue to evolve.
Collapse
Affiliation(s)
- Noelle Younge
- Department of Pediatrics, Duke University, 2400 Pratt Street, DUMC Box 2739, Durham, NC 27705, USA
| | - Ravi M Patel
- Department of Pediatrics, Emory University and Children's Healthcare of Atlanta, Arthur M. Blank Hospital, 2220 North Druid Hills Road NE, CL.06323, Atlanta, GA 30329, USA.
| |
Collapse
|
13
|
Szajewska H, Scott KP, de Meij T, Forslund-Startceva SK, Knight R, Koren O, Little P, Johnston BC, Łukasik J, Suez J, Tancredi DJ, Sanders ME. Antibiotic-perturbed microbiota and the role of probiotics. Nat Rev Gastroenterol Hepatol 2025; 22:155-172. [PMID: 39663462 DOI: 10.1038/s41575-024-01023-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 11/08/2024] [Indexed: 12/13/2024]
Abstract
The disruptive effect of antibiotics on the composition and function of the human microbiota is well established. However, the hypothesis that probiotics can help restore the antibiotic-disrupted microbiota has been advanced, with little consideration of the strength of evidence supporting it. Some clinical data suggest that probiotics can reduce antibiotic-related side effects, including Clostridioides difficile-associated diarrhoea, but there are no data that causally link these clinical effects to microbiota protection or recovery. Substantial challenges hinder attempts to address this hypothesis, including the absence of consensus on the composition of a 'normal' microbiota, non-standardized and evolving microbiome measurement methods, and substantial inter-individual microbiota variation. In this Review, we explore these complexities. First, we review the known benefits and risks of antibiotics, the effect of antibiotics on the human microbiota, the resilience and adaptability of the microbiota, and how microbiota restoration might be defined and measured. Subsequently, we explore the evidence for the efficacy of probiotics in preventing disruption or aiding microbiota recovery post-antibiotic treatment. Finally, we offer insights into the current state of research and suggest directions for future research.
Collapse
Affiliation(s)
- Hania Szajewska
- Department of Paediatrics, The Medical University of Warsaw, Warsaw, Poland
| | - Karen P Scott
- Rowett Institute, University of Aberdeen, Aberdeen, UK
| | - Tim de Meij
- Department of Paediatric Gastroenterology, Emma Children's Hospital, Amsterdam UMC, Academic Medical Centre, Amsterdam, The Netherlands
| | - Sofia K Forslund-Startceva
- Experimental and Clinical Research Center, a joint cooperation of Max Delbruck Center for Molecular Medicine and Charité - Universitätsmedizin Berlin, Berlin, Germany
- Charité - Universitätsmedizin Berlin, corporate member of Freie Universität Berlin and Humboldt-Universität zu Berlin, Berlin, Germany
- Max Delbruck Center for Molecular Medicine in the Helmholtz Association (MDC), Berlin, Germany
- DZHK (German Centre for Cardiovascular Research), Berlin, Germany
| | - Rob Knight
- Department of Pediatrics, University of California San Diego, San Diego, CA, USA
- Department of Computer Science & Engineering, University of California San Diego, San Diego, CA, USA
- Shu Chien - Gene Lay Department of Bioengineering, University of California San Diego, San Diego, CA, USA
- Halıcıoğlu Data Science Institute, University of California San Diego, San Diego, CA, USA
- Center for Microbiome Innovation, University of California San Diego, San Diego, CA, USA
| | - Omry Koren
- Azrieli Faculty of Medicine, Bar Ilan University, Safed, Israel
| | - Paul Little
- Primary Care Research Centre, University of Southampton, Southampton, UK
| | - Bradley C Johnston
- Department of Nutrition, College of Agriculture and Life Sciences, Texas A&M University, College Station, TX, USA
- Department of Epidemiology and Biostatistics, School of Public Health, Texas A&M University, College Station, TX, USA
| | - Jan Łukasik
- Department of Paediatrics, The Medical University of Warsaw, Warsaw, Poland
| | - Jotham Suez
- W. Harry Feinstone Department of Molecular Microbiology and Immunology, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, USA
| | - Daniel J Tancredi
- Department of Pediatrics, School of Medicine, University of California Davis, Sacramento, CA, USA
| | - Mary Ellen Sanders
- International Scientific Association for Probiotics and Prebiotics, Consulting Scientific Advisor, Centennial, CO, USA.
| |
Collapse
|
14
|
Tadese DA, Mwangi J, Luo L, Zhang H, Huang X, Michira BB, Zhou S, Kamau PM, Lu Q, Lai R. The microbiome's influence on obesity: mechanisms and therapeutic potential. SCIENCE CHINA. LIFE SCIENCES 2025; 68:657-672. [PMID: 39617855 DOI: 10.1007/s11427-024-2759-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/21/2024] [Accepted: 10/16/2024] [Indexed: 01/03/2025]
Abstract
In 2023, the World Obesity Atlas Federation concluded that more than 50% of the world's population would be overweight or obese within the next 12 years. At the heart of this epidemic lies the gut microbiota, a complex ecosystem that profoundly influences obesity-related metabolic health. Its multifaced role encompasses energy harvesting, inflammation, satiety signaling, gut barrier function, gut-brain communication, and adipose tissue homeostasis. Recognizing the complexities of the cross-talk between host physiology and gut microbiota is crucial for developing cutting-edge, microbiome-targeted therapies to address the global obesity crisis and its alarming health and economic repercussions. This narrative review analyzed the current state of knowledge, illuminating emerging research areas and their implications for leveraging gut microbial manipulations as therapeutic strategies to prevent and treat obesity and related disorders in humans. By elucidating the complex relationship between gut microflora and obesity, we aim to contribute to the growing body of knowledge underpinning this critical field, potentially paving the way for novel interventions to combat the worldwide obesity epidemic.
Collapse
Affiliation(s)
- Dawit Adisu Tadese
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - James Mwangi
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lei Luo
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Hao Zhang
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
| | - Xiaoshan Huang
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Brenda B Michira
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Shengwen Zhou
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Peter Muiruri Kamau
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Qiumin Lu
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China
| | - Ren Lai
- Engineering Laboratory of Peptides of Chinese Academy of Sciences, Key Laboratory of Bioactive Peptides of Yunnan Province, KIZ-CUHK Joint Laboratory of Bioresources and Molecular Research in Common Diseases, National Resource Center for Non-Human Primates, National Research Facility for Phenotypic & Genetic Analysis of Model Animals (Primate Facility), Key Laboratory of Genetic Evolution & Animal Models, Sino-African Joint Research Center, and New Cornerstone Science Laboratory, Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, 650201, China.
- Kunming College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
15
|
Schubert C, Nguyen BD, Sichert A, Näpflin N, Sintsova A, Feer L, Näf J, Daniel BBJ, Steiger Y, von Mering C, Sauer U, Hardt WD. Monosaccharides drive Salmonella gut colonization in a context-dependent or -independent manner. Nat Commun 2025; 16:1735. [PMID: 39966379 PMCID: PMC11836396 DOI: 10.1038/s41467-025-56890-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 02/04/2025] [Indexed: 02/20/2025] Open
Abstract
The carbohydrates that fuel gut colonization by S. Typhimurium are not fully known. To investigate this, we designed a quality-controlled mutant pool to probe the metabolic capabilities of this enteric pathogen. Using neutral genetic barcodes, we tested 35 metabolic mutants across five different mouse models with varying microbiome complexities, allowing us to differentiate between context-dependent and context-independent nutrient sources. Results showed that S. Typhimurium uses D-mannose, D-fructose and likely D-glucose as context-independent carbohydrates across all five mouse models. The utilization of D-galactose, N-acetylglucosamine and hexuronates, on the other hand, was context-dependent. Furthermore, we showed that D-fructose is important in strain-to-strain competition between Salmonella serovars. Complementary experiments confirmed that D-glucose, D-fructose, and D-galactose are excellent niches for S. Typhimurium to exploit during colonization. Quantitative measurements revealed sufficient amounts of carbohydrates, such as D-glucose or D-galactose, in the murine cecum to drive S. Typhimurium colonization. Understanding these key substrates and their context-dependent or -independent use by enteric pathogens will inform the future design of probiotics and therapeutics to prevent diarrheal infections such as non-typhoidal salmonellosis.
Collapse
Affiliation(s)
- Christopher Schubert
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland.
| | - Bidong D Nguyen
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Andreas Sichert
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Nicolas Näpflin
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Anna Sintsova
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Lilith Feer
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Jana Näf
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Benjamin B J Daniel
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Yves Steiger
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Christian von Mering
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Uwe Sauer
- Institute of Molecular Systems Biology, ETH Zurich, Zurich, Switzerland
| | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
16
|
Casas Deza D, Alcedo J, Lafuente M, López FJ, Perez-Aisa Á, Pavoni M, Tepes B, Jonaitis L, Castro-Fernandez M, Pabón-Carrasco M, Keco-Huerga A, Voynovan I, Bujanda L, Lucendo AJ, Brglez Jurecic N, Denkovski M, Vologzanina L, Rodrigo L, Martínez-Domínguez SJ, Fadieienko G, Huguet JM, Abdulkhakov R, Abdulkhakov SR, Alcaide N, Velayos B, Hernández L, Bordin DS, Gasbarrini A, Kupcinskas J, Babayeva G, Gridnyev O, Leja M, Rokkas T, Marcos-Pinto R, Lerang F, Boltin D, Mestrovic A, Smith SM, Venerito M, Boyanova L, Milivojevic V, Doulberis M, Kunovsky L, Parra P, Cano-Català A, Moreira L, Nyssen OP, Megraud F, Morain CO, Gisbert JP. Probiotics Prescribed With Helicobacter pylori Eradication Therapy in Europe: Usage Pattern, Effectiveness, and Safety. Results From the European Registry on Helicobacter pylori Management (Hp-EuReg). Am J Gastroenterol 2025:00000434-990000000-01573. [PMID: 39902822 DOI: 10.14309/ajg.0000000000003351] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Accepted: 12/12/2024] [Indexed: 02/06/2025]
Abstract
INTRODUCTION To evaluate the prescription patterns, effectiveness, and safety of adding probiotics to Helicobacter pylori eradication therapy, in Europe. METHODS International, prospective, noninterventional registry of the clinical practice of the European gastroenterologists. Data were collected and quality reviewed until March 2021 at AEG-REDCap. The effectiveness was evaluated by modified intention-to-treat analysis, differentiating by geographic areas. Adverse events (AEs) were categorized as mild, moderate, and severe. RESULTS Overall, 36,699 treatments were recorded, where 8,233 (22%) were prescribed with probiotics. Probiotics use was associated with higher effectiveness in the overall analysis (odds ratio [OR] 1.631, 95% confidence interval [CI] 1.456-1.828), as well as in triple (OR 1.702, 95% CI 1.403-2.065), quadruple (OR 1.383, 95% CI 0.996-1.920), bismuth quadruple (OR 1.248, 95% CI 1.003-1.554), and sequential therapies (OR 3.690, 95% CI 2.686-5.069). Lactobacillus genus was associated with a higher therapy effectiveness in Eastern Europe when triple (OR 2.625, 95% CI 1.911-3.606) and bismuth quadruple (OR 1.587, 95% CI 1.117-2.254) first-line therapies were prescribed. In Central Europe, the use of probiotics was associated with a decrease in both the overall incidence of AEs (OR 0.656, 95% CI 0.516-0.888) and severe AEs (OR 0.312, 95% CI 0.217-0.449). Bifidobacterium genus was associated with lower overall (OR 0.725, 95% CI 0.592-0.888) and severe (OR 0.254, 95% CI 0.185-0.347) AEs, and Saccharomyces was associated with reduced overall (OR 0.54, 95% CI 0.32-0.91) and severe (OR 0.257, 95% CI 0.123-0.536) AEs under quadruple-bismuth regimen. DISCUSSION In Europe, the use of probiotics was associated with higher effectiveness and safety of H. pylori eradication therapy. Lactobacillus improved treatment effectiveness, whereas Bifidobacterium and Saccharomyces were associated with a better safety profile.
Collapse
Affiliation(s)
- Diego Casas Deza
- Department of Gastroenterology, Hospital Universitario Miguel Servet, Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Zaragoza, Spain
| | - Javier Alcedo
- Department of Gastroenterology, Hospital Universitario Miguel Servet, Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Zaragoza, Spain
| | - Miguel Lafuente
- Department of Statistical Methods, Faculty of Science, Universidad de Zaragoza, Zaragoza, Spain
- Instituto de Biocomputación y Física de Sistemas Complejos (BIFI), Universidad de Zaragoza, Zaragoza, Spain
| | - F Javier López
- Department of Statistical Methods, Faculty of Science, Universidad de Zaragoza, Zaragoza, Spain
- Instituto de Biocomputación y Física de Sistemas Complejos (BIFI), Universidad de Zaragoza, Zaragoza, Spain
| | - Ángeles Perez-Aisa
- Digestive Unit, Agencia Sanitaria Costa del Sol, Marbella, Spain
- Redes de Investigación Cooperativa Orientada a Resultados en Salud (RICORS), Marbella, Spain
| | - Matteo Pavoni
- Department of Medical and Surgical Sciences, Sant'Orsola-Malpighi University Hospital, Bologna, Italy
| | - Bojan Tepes
- Department of Gastroenterology, DC Rogaska, Slatina, Slovenia
| | - Laimas Jonaitis
- Research and Department of Gastroenterology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | | | | | - Alma Keco-Huerga
- Department of Gastroenterology, Hospital Universitario de Valme, Seville, Spain
| | - Irina Voynovan
- Department of Gastroenterology, A.S. Loginov Moscow Clinical Scientific Center, Moscow, Russia
| | - Luis Bujanda
- Department of Gastroenterology, Biodonostia Health Research Institute, San Sebastián, Spain
- CIBERehd, Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
- Department of Medicine, Universidad del País Vasco (UPV/EHU), San Sebastián, Spain
| | - Alfredo J Lucendo
- Department of Gastroenterology, Hospital General de Tomelloso, Tomelloso, Spain
- CIBERehd, Centro de Investigación en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
- Gastroenterology, Instituto de Investigación Sanitaria Princesa, Madrid, Spain
| | | | - Maja Denkovski
- Department of Gastroenterology, Interni Oddelek, Diagnostic Centre, Bled, Slovenia
| | | | - Luis Rodrigo
- Gastroenterology, University of Oviedo, Oviedo, Spain
| | - Samuel J Martínez-Domínguez
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), Zaragoza, Spain
- Servicio de Aparato Digestivo, Hospital Clínico Universitario "Lozano Blesa," Zaragoza, Spain
- CIBERehd, Zaragoza, Spain
| | - Galyna Fadieienko
- Departments the Division for the Study of the Digestive Diseases and Its Comorbidity With Noncommunicable Diseases, Government Institution L.T. Malaya Therapy National Institute of NAMS of Ukraine, Kharkiv, Ukraine
| | - Jose M Huguet
- Department of Gastroenterology, Hospital General Universitario de Valencia, Valencia, Spain
| | - Rustam Abdulkhakov
- Department of Hospital Medicine, Kazan State Medical University, Kazan, Russia
| | - Sayar R Abdulkhakov
- Department of Hospital Medicine, Kazan State Medical University, Kazan, Russia
| | - Noelia Alcaide
- Department of Gastroenterology, Hospital Clínico de Valladolid, Valladolid, Spain
| | - Benito Velayos
- Department of Gastroenterology, Hospital Clínico de Valladolid, Valladolid, Spain
| | - Luis Hernández
- Gastroenterology Unit, Hospital Santos Reyes, Aranda de Duero, Spain
| | - Dmitry S Bordin
- Department of Pancreatic, Biliary and Upper Digestive Tract Disorders, A.S. Loginov Moscow Clinical Scientific Center, Moscow, Russia
- Department of Propaedeutic of Internal Diseases and Gastroenterology, Russian University of Medicine, Moscow, Russia
- Department of Outpatient Therapy and Family Medicine, Tver State Medical University, Tver, Russia
| | - Antonio Gasbarrini
- Medicina interna e Gastroenterologia, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, Rome, Italy
| | - Juozas Kupcinskas
- Research and Department of Gastroenterology, Lithuanian University of Health Sciences, Kaunas, Lithuania
| | - Gülüstan Babayeva
- Department of Internal Medicine and Gastroenterology, Azerbaijan State Advanced Training Institute for Doctors named by A. Aliyev, Baku, Azerbaijan
| | - Oleksiy Gridnyev
- Departments the Division for the Study of the Digestive Diseases and Its Comorbidity With Noncommunicable Diseases, Government Institution L.T. Malaya Therapy National Institute of NAMS of Ukraine, Kharkiv, Ukraine
| | - Mārcis Leja
- Department of Gastroenterology, Digestive Diseases Centre, Riga, Latvia
- Institute of Clinical and Preventive Medicine, University of Latvia, Riga, Latvia
| | - Theodore Rokkas
- Gastroenterology Clinic, Henry Dunant Hospital, Athens, Greece
| | - Ricardo Marcos-Pinto
- Gastroenterology Department, Centro Hospitalar do Porto, Porto, Portugal
- Instituto De Ciências Biomédicas de Abel Salazar, Universidade do Porto, Porto, Portugal
- Center for Research in Health Technologies and Information Systems (CINTESIS), Porto, Portugal
| | - Frode Lerang
- Department of Gastroenterology, Østfold Hospital Trust, Grålum, Norway
| | - Doron Boltin
- Division of Gastroenterology, Rabin Medical Center, TelAviv, Israel
- Tel Aviv University, TelAviv, Israel
| | - Antonio Mestrovic
- Department of Gastroenterology, University Hospital of Split, Split, Croatia
| | - Sinead M Smith
- School of Medicine, Faculty of Health Sciences, Trinity College Dublin, Dublin, Ireland
| | - Marino Venerito
- Department of Gastroenterology, Hepatology and Infectious Diseases, University Hospital of Magdeburg, Magdeburg, Germany
| | - Lyudmila Boyanova
- Department of Medical Microbiology, Medical University of Sofia, Sofia, Bulgaria
| | - Vladimir Milivojevic
- Clinic of Gastroenterology and Hepatology, Clinical Center of Serbia, Belgrade, Serbia
- Faculty of Medicine, University of Belgrade, Belgrade, Serbia
| | - Michael Doulberis
- Gastroenterology Department, Kantonsspital Aarau, Aarau, Switzerland
| | - Lumir Kunovsky
- 2nd Department of Internal Medicine-Gastroenterology and Geriatrics, University Hospital Olomouc, Faculty of Medicine and Dentistry, Palacky University Olomouc, Olomouc, Czech Republic
- Department of Surgery, University Hospital Brno, Faculty of Medicine, Masaryk University, Brno, Czech Republic
- Department of Gastroenterology and Digestive Endoscopy, Masaryk Memorial Cancer Institute, Brno, Czech Republic
| | - Pablo Parra
- Servicio de Aparato Digestivo, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | - Anna Cano-Català
- Gastrointestinal Oncology, Endoscopy and Surgery (GOES) Research Group, Althaia Xarxa Assistencial Universitària de Manresa FP, Manresa, Spain
- Institut de Recerca i Innovació en Ciències de la Vida i de la Salut de la Catalunya Central (IRIS-CC), Vic, Spain
| | - Leticia Moreira
- Hospital Clínic de Barcelona, Centro de Investigación Biomédica en Red en Enfermedades Hepáticas y Digestivas (CIBERehd), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS), University of Barcelona, Barcelona, Spain
| | - Olga P Nyssen
- Servicio de Aparato Digestivo, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| | | | - Colm O Morain
- School of Medicine, Faculty of Health Sciences, Trinity College Dublin, Dublin, Ireland
| | - Javier P Gisbert
- Servicio de Aparato Digestivo, Hospital Universitario de La Princesa, Instituto de Investigación Sanitaria Princesa (IIS-Princesa), Universidad Autónoma de Madrid (UAM), Centro de Investigación Biomédica en Red de Enfermedades Hepáticas y Digestivas (CIBERehd), Madrid, Spain
| |
Collapse
|
17
|
Colmenarejo C, Rodríguez-Jiménez C, Navarro FJ, Mateo AB, Pellejero EM, Belda-Moreno RM, Ureña-Méndez R, Pérez-Serrano R, Illescas S, Ramón Muñoz-Rodríguez J, del Campo R. One-year monitorization of the gut colonization by multidrug resistant bacteria in elderly of a single long-term care facility. JAC Antimicrob Resist 2025; 7:dlaf008. [PMID: 39927311 PMCID: PMC11806259 DOI: 10.1093/jacamr/dlaf008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2024] [Accepted: 01/02/2025] [Indexed: 02/11/2025] Open
Abstract
Objective To monitor the gut colonization by multidrug resistant (MDR) bacteria in residents of a single long-term care facility (LTCF) in relation to their clinical evolution, antibiotic consumption and mortality risk. Methods In a total of 187 voluntarily enrolled residents, five rectal swabs samples were recovered over 1 year. Selective media were used to isolate MDR bacteria. Clinical data related to infections, antibiotic consumption and mortality were recovered. Mortality risk among residents who were MDR colonized and non-colonized was compared by Kaplan-Meier curves. Results Globally, 25% of residents have gut colonization by ESBL-producing Escherichia coli with a lack of other pathogens such as Acinetobacter baumannii or Clostridioides difficile. Monitoring of ESBL-producing E. coli colonization for 1 year allowed to us to establish three categories among residents: 48.6% never colonized, 15.5% had a persistent colonization, and the remaining 35.8% presented intermittent colonization. The rates of mortality, infections and antibiotic exposure were comparable among ESBL-producing E. coli colonized and non-colonized residents, except for the intermittent colonization group in which a higher and statistically significant mortality rate was observed. As expected, urinary and respiratory tract infections were the most prevalent infectious pathologies in the LTCF, with amoxicillin/clavulanate and fluoroquinolones being the most prescribed antibiotics. A high percentage of ESBL-producing E. coli (28%), and fluoroquinolone resistance were detected in clinical samples. Conclusions The monitoring of gut colonization by MDR microorganisms in a single LTCF for 1 year demonstrated the predominance of ESBL-producing E. coli. Almost half of the residents were resistant to its colonization, whereas in 15.5% of them gut colonization was stable. Incidence of infectious episodes and antibiotic exposure were comparable between colonized and non-colonized subjects, but the group with the highest risk of mortality was that with intermittent colonization by ESBL-producing E. coli.
Collapse
Affiliation(s)
- Cristina Colmenarejo
- Servicio de Microbiología, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
| | - Concepción Rodríguez-Jiménez
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal, and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | | | - Ana Belén Mateo
- Long-term care facility Gregorio Marañón, Ciudad Real, Spain
| | | | | | | | - Raúl Pérez-Serrano
- Servicio de Farmacia, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
| | - Soledad Illescas
- Servicio de Microbiología, Hospital General Universitario de Ciudad Real, Ciudad Real, Spain
- Departmento de Microbiología, Facultad de Medicina, Universidad de Castilla-La Mancha, Ciudad Real, Spain
| | | | - Rosa del Campo
- Servicio de Microbiología, Hospital Universitario Ramón y Cajal, and Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
- CIBER de Enfermedades Infecciosas, Instituto de Salud Carlos III, Madrid, Spain
- Facultad de Ciencias de la Salud, Universidad Alfonso X El Sabio, Villanueva de la Cañada, Madrid, Spain
| |
Collapse
|
18
|
Andrei C, Zanfirescu A, Ormeneanu VP, Negreș S. Evaluating the Efficacy of Secondary Metabolites in Antibiotic-Induced Dysbiosis: A Narrative Review of Preclinical Studies. Antibiotics (Basel) 2025; 14:138. [PMID: 40001382 PMCID: PMC11852119 DOI: 10.3390/antibiotics14020138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2024] [Revised: 01/20/2025] [Accepted: 01/23/2025] [Indexed: 02/27/2025] Open
Abstract
BACKGROUND/OBJECTIVES Drug-induced dysbiosis, particularly from antibiotics, has emerged as a significant contributor to chronic diseases by disrupting gut microbiota composition and function. Plant-derived secondary metabolites, such as polysaccharides, polyphenols, alkaloids, and saponins, show potential in mitigating antibiotic-induced dysbiosis. This review aims to consolidate evidence from preclinical studies on the therapeutic effects of secondary metabolites in restoring gut microbial balance, emphasizing their mechanisms and efficacy. METHODS A narrative review was conducted using PubMed, Scopus, and Web of Science. Studies were selected based on specific inclusion criteria, focusing on animal models treated with secondary metabolites for antibiotic-induced dysbiosis. The search terms included "gut microbiota", "antibiotics", and "secondary metabolites". Data extraction focused on microbial alterations, metabolite-specific effects, and mechanisms of action. Relevant findings were systematically analyzed and summarized. RESULTS Secondary metabolites demonstrated diverse effects in mitigating the impact of dysbiosis by modulating gut microbial composition, reducing inflammation, and supporting host biological markers. Polysaccharides and polyphenols restored the Firmicutes/Bacteroidetes ratio, increased beneficial taxa such as Lactobacillus and Bifidobacterium, and suppressed pathogenic bacteria like Escherichia-Shigella. Metabolites such as triterpenoid saponins enhanced gut barrier integrity by upregulating tight junction proteins, while alkaloids reduced inflammation by modulating proinflammatory cytokines (e.g., TNF-α, IL-1β). These metabolites also improved short-chain fatty acid production, which is crucial for gut and systemic health. While antibiotic-induced dysbiosis was the primary focus, other drug classes (e.g., PPIs, metformin) require further investigation. CONCLUSIONS Plant-derived secondary metabolites show promise in managing antibiotic-induced dysbiosis by restoring microbial balance, reducing inflammation, and improving gut barrier function. Future research should explore their applicability to other types of drug-induced dysbiosis and validate findings in human studies to enhance clinical relevance.
Collapse
Affiliation(s)
| | - Anca Zanfirescu
- Faculty of Pharmacy, “Carol Davila” University of Medicine and Pharmacy, Traian Vuia 6, 020956 Bucharest, Romania; (C.A.); (V.-P.O.); (S.N.)
| | | | | |
Collapse
|
19
|
Geng J, Dong Y, Huang H, Wen X, Xu T, Zhao Y, Liu Y. Role of nasal microbiota in regulating host anti-influenza immunity in dogs. MICROBIOME 2025; 13:27. [PMID: 39871363 PMCID: PMC11773957 DOI: 10.1186/s40168-025-02031-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 01/07/2025] [Indexed: 01/29/2025]
Abstract
BACKGROUND Numerous studies have confirmed a close relationship between the pathogenicity of influenza and respiratory microbiota, but the mechanistic basis for this is poorly defined. Also, the majority of these studies have been conducted on murine models, and it remains unclear how far these findings can be extrapolated from murine models to other animals. Considering that influenza A virus is increasingly recognized as an important canine respiratory pathogen, this study investigated the cross-talk between nasal and lung tissues mediated by microbes and its association with influenza susceptibility in a beagle dog model. RESULTS Using 16S rRNA gene sequencing, combined with comparative transcriptomic, anatomical, and histological examinations, we investigated viral presence, gene expression profiles, and microbiota in the nasal cavity and lung after influenza infection in the beagles with antibiotic-induced nasal dysbiosis. Our data showed that dysbiosis of the nasal microbiome exacerbates influenza-induced respiratory disease and the epithelial barrier disruption, and impairs host antiviral responses in the nasal cavity and lung. Moreover, dysregulation of nasal microbiota exacerbates the influenza-induced disturbance in lung microbiota. Further, we also identified a strain of Lactobacillus plantarum isolated from canine nasal cavity with a significant antiviral effect in vitro, and found that its antiviral activity might be associated with the activation of the interferon (IFN) pathway and modulation of the impaired autophagy flux induced by influenza infection. CONCLUSIONS Our investigation reveals that nasal microbiota dysbiosis exerts a prominent impact on host antiviral responses, inflammation thresholds, and mucosal barrier integrity during influenza infection. Lactobacilli, as part of the nasal microbiota, may contribute to host antiviral defenses by modulating the IFN and autophagy pathways. Collectively, this study underscores the importance of nasal microbiota homeostasis in maintaining respiratory health. Video Abstract.
Collapse
Affiliation(s)
- Jinzhu Geng
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yuhao Dong
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Hao Huang
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Xia Wen
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Ting Xu
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yanbing Zhao
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China
| | - Yongjie Liu
- Joint International Research Laboratory of Animal Health and Food Safety, College of Veterinary Medicine, Nanjing Agricultural University, Nanjing, China.
| |
Collapse
|
20
|
Cheng Q, Chen S. Using In Vitro Models to Study the Interactions Between Environmental Exposures and Human Microbiota. Microorganisms 2025; 13:247. [PMID: 40005610 PMCID: PMC11857843 DOI: 10.3390/microorganisms13020247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2024] [Revised: 01/03/2025] [Accepted: 01/09/2025] [Indexed: 02/27/2025] Open
Abstract
Research has demonstrated a close correlation between human microbiota and overall health, highlighting their intimate connection. Exposure to environmental factors, such as chemical contaminants and biological agents, has the potential to alter the composition and function of microbiota, thereby influencing health outcomes. Meanwhile, microbiota may contribute to host protection by degrading, or rendering harmless, exposures. Environmental exposures demonstrate significant diversity and dynamism; however, conventional methods for exposure-microbiota research, such as animal and epidemiological studies, are often both time-consuming and costly. Additionally, they may raise ethical concerns. This review aimed to examine the existing understanding of employing in vitro models to investigate the interactions between environmental exposures and human microbiota, particularly those located outside the large intestine. A comprehensive search was conducted across the Web of Science, PubMed, and Scopus databases, employing a range of keywords related to microbiota, exposures, and in vitro models. A total of 58 studies fulfilled the search criteria, revealing instances of microbial modulation of exposures and vice versa. It was observed that, although considerable research has been conducted on these interactions in vitro, there remains a pressing need for enhanced model designs and application contexts.
Collapse
Affiliation(s)
- Qiwen Cheng
- Biodesign Center for Bioenergetics, Arizona State University, Tempe, AZ 85281, USA
- Center for Big Data, Qilu Hospital of Shandong University, Jinan 250012, China
| | - Shengxi Chen
- Biodesign Center for Bioenergetics, Arizona State University, Tempe, AZ 85281, USA
| |
Collapse
|
21
|
Beattie GA, Edlund A, Esiobu N, Gilbert J, Nicolaisen MH, Jansson JK, Jensen P, Keiluweit M, Lennon JT, Martiny J, Minnis VR, Newman D, Peixoto R, Schadt C, van der Meer JR. Soil microbiome interventions for carbon sequestration and climate mitigation. mSystems 2025; 10:e0112924. [PMID: 39692482 PMCID: PMC11748500 DOI: 10.1128/msystems.01129-24] [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: 12/19/2024] Open
Abstract
Mitigating climate change in soil ecosystems involves complex plant and microbial processes regulating carbon pools and flows. Here, we advocate for the use of soil microbiome interventions to help increase soil carbon stocks and curb greenhouse gas emissions from managed soils. Direct interventions include the introduction of microbial strains, consortia, phage, and soil transplants, whereas indirect interventions include managing soil conditions or additives to modulate community composition or its activities. Approaches to increase soil carbon stocks using microbially catalyzed processes include increasing carbon inputs from plants, promoting soil organic matter (SOM) formation, and reducing SOM turnover and production of diverse greenhouse gases. Marginal or degraded soils may provide the greatest opportunities for enhancing global soil carbon stocks. Among the many knowledge gaps in this field, crucial gaps include the processes influencing the transformation of plant-derived soil carbon inputs into SOM and the identity of the microbes and microbial activities impacting this transformation. As a critical step forward, we encourage broadening the current widespread screening of potentially beneficial soil microorganisms to encompass functions relevant to stimulating soil carbon stocks. Moreover, in developing these interventions, we must consider the potential ecological ramifications and uncertainties, such as incurred by the widespread introduction of homogenous inoculants and consortia, and the need for site-specificity given the extreme variation among soil habitats. Incentivization and implementation at large spatial scales could effectively harness increases in soil carbon stocks, helping to mitigate the impacts of climate change.
Collapse
Affiliation(s)
- Gwyn A. Beattie
- Department of Plant Pathology, Entomology and Microbiology, Iowa State University, Ames, Iowa, USA
| | | | - Nwadiuto Esiobu
- Department of Biological Sciences, Microbiome Innovation Cluster, Florida Atlantic University, Boca Raton, Florida, USA
| | - Jack Gilbert
- Department of Pediatrics and Scripps Institution of Oceanography, UC San Diego School of Medicine, La Jolla, California, USA
| | | | - Janet K. Jansson
- Biological Sciences Division, Pacific Northwest National Laboratory, Richland, Washington, USA
| | - Paul Jensen
- Scripps Institution of Oceanography, University of California San Diego, La Jolla, California, USA
| | - Marco Keiluweit
- Soil Biogeochemistry Group, Faculty of Geosciences and the Environment, University of Lausanne, Lausanne, Switzerland
| | - Jay T. Lennon
- Department of Biology, Indiana University, Bloomington, Indiana, USA
| | - Jennifer Martiny
- School of Biological Sciences, University of California, Irvine, Irvine, California, USA
| | - Vanessa R. Minnis
- Department of Pediatrics and Scripps Institution of Oceanography, UC San Diego School of Medicine, La Jolla, California, USA
| | - Dianne Newman
- Division of Biology & Biological Engineering and Division of Geological and Planetary Sciences, California Institute of Technology, Pasadena, California, USA
| | - Raquel Peixoto
- Biological and Environmental Science and Engineering Division, King Abdullah University of Science and Technology, Thuwal, Saudi Arabia
| | - Christopher Schadt
- Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, Tennessee, USA
| | | |
Collapse
|
22
|
Zheng ZL, Zheng QF, Wang LQ, Liu Y. Bowel preparation before colonoscopy: Consequences, mechanisms, and treatment of intestinal dysbiosis. World J Gastroenterol 2025; 31:100589. [PMID: 39811511 PMCID: PMC11684204 DOI: 10.3748/wjg.v31.i2.100589] [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: 09/25/2024] [Revised: 10/22/2024] [Accepted: 11/12/2024] [Indexed: 12/18/2024] Open
Abstract
The term "gut microbiota" primarily refers to the ecological community of various microorganisms in the gut, which constitutes the largest microbial community in the human body. Although adequate bowel preparation can improve the results of colonoscopy, it may interfere with the gut microbiota. Bowel preparation for colonoscopy can lead to transient changes in the gut microbiota, potentially affecting an individual's health, especially in vulnerable populations, such as patients with inflammatory bowel disease. However, measures such as oral probiotics may ameliorate these adverse effects. We focused on the bowel preparation-induced changes in the gut microbiota and host health status, hypothesized the factors influencing these changes, and attempted to identify measures that may reduce dysbiosis, thereby providing more information for individualized bowel preparation for colonoscopy in the future.
Collapse
Affiliation(s)
- Ze-Long Zheng
- Department of Gastroenterology (Endoscopy Center), China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Qing-Fan Zheng
- Department of Gastroenterology (Endoscopy Center), China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Li-Qiang Wang
- Department of Gastroenterology (Endoscopy Center), China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| | - Yi Liu
- Department of Gastroenterology (Endoscopy Center), China-Japan Union Hospital of Jilin University, Changchun 130033, Jilin Province, China
| |
Collapse
|
23
|
Chen Z, Yang Y, Chen X, Bei C, Gao Q, Chao Y, Wang C. An RNase III-processed sRNA coordinates sialic acid metabolism of Salmonella enterica during gut colonization. Proc Natl Acad Sci U S A 2025; 122:e2414563122. [PMID: 39792291 PMCID: PMC11745405 DOI: 10.1073/pnas.2414563122] [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: 07/21/2024] [Accepted: 12/06/2024] [Indexed: 01/12/2025] Open
Abstract
Sialic acids derived from colonic mucin glycans are crucial nutrients for enteric bacterial pathogens like Salmonella. The uptake and utilization of sialic acid in Salmonella depend on coordinated regulons, each activated by specific metabolites at the transcriptional level. However, the mechanisms enabling crosstalk among these regulatory circuits to synchronize gene expression remain poorly understood. Here, we identify ManS, a small noncoding RNA derived from the 3' UTR of STM1128 mRNA transcribed from a Salmonella enterica-specific genetic locus, as an important posttranscriptional regulator coordinating sialic acid metabolism regulons. ManS is primarily processed by RNase III and, along with its parental transcripts, is specifically activated by N-acetylmannosamine (ManNAc), the initial degradation product of sialic acid. We found that the imperfect stem-loop structure at the 5' end of ManS allows RNase III to cleave in a noncanonical manner, generating two functional types of ManS with the assistance of RNase E and other RNases: short isoforms with a single seed region that regulate the uptake of N-acetylglucosamine, an essential intermediate in sialic acid metabolism; and long isoforms with an additional seed region that regulate multiple genes involved in central and secondary metabolism. This sophisticated regulation by ManS significantly impacts ManNAc metabolism and S. enterica's competitive behavior during infection. Our findings highlight the role of sRNA in coordinating transcriptional circuits and advance our understanding of RNase III-mediated processing of 3' UTR-derived sRNAs, underscoring the important role of ManNAc in Salmonella adaptation within host environments.
Collapse
Affiliation(s)
- Ziying Chen
- Key Laboratory of Medical Molecular Virology (Ministry of Education / National Health Commission / Chinese Academy of Medical Sciences), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200033, China
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai200031, China
- Department of Colorectal Surgery, Fudan University Shanghai Cancer Center, Shanghai200032, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai200032, China
| | - Yaomei Yang
- Key Laboratory of Medical Molecular Virology (Ministry of Education / National Health Commission / Chinese Academy of Medical Sciences), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200033, China
| | - Xiaomin Chen
- Key Laboratory of Medical Molecular Virology (Ministry of Education / National Health Commission / Chinese Academy of Medical Sciences), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200033, China
| | - Cheng Bei
- Key Laboratory of Medical Molecular Virology (Ministry of Education / National Health Commission / Chinese Academy of Medical Sciences), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200033, China
| | - Qian Gao
- Key Laboratory of Medical Molecular Virology (Ministry of Education / National Health Commission / Chinese Academy of Medical Sciences), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200033, China
| | - Yanjie Chao
- Key Laboratory of RNA Innovation, Science and Engineering, Shanghai Institute of Immunity and Infection, Chinese Academy of Sciences, Shanghai200031, China
| | - Chuan Wang
- Key Laboratory of Medical Molecular Virology (Ministry of Education / National Health Commission / Chinese Academy of Medical Sciences), Shanghai Frontiers Science Center of Pathogenic Microorganisms and Infection, School of Basic Medical Sciences, Shanghai Medical College, Fudan University, Shanghai200033, China
| |
Collapse
|
24
|
Almási ÉDH, Eisenhard L, Osbelt L, Lesker TR, Vetter AC, Knischewski N, Bielecka AA, Gronow A, Muthukumarasamy U, Wende M, Tawk C, Neumann-Schaal M, Brönstrup M, Strowig T. Klebsiella oxytoca facilitates microbiome recovery via antibiotic degradation and restores colonization resistance in a diet-dependent manner. Nat Commun 2025; 16:551. [PMID: 39789003 PMCID: PMC11717976 DOI: 10.1038/s41467-024-55800-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025] Open
Abstract
Competition among bacteria for carbohydrates is pivotal for colonization resistance (CR). However, the impact of Western-style diets on CR remains unclear. Here we show how the competition between Klebsiella oxytoca and Klebsiella pneumoniae is modulated by consuming one of three Western-style diets characterized by high-starch, high-sucrose, or high-fat/high-sucrose content. In vivo competition experiments in ampicillin-treated mice reveal that K. oxytoca promotes K. pneumoniae decolonization on all dietary backgrounds. However, mice on the high-fat/high-sucrose diet show reduced pathogen clearance. Microbiome analysis reveals that the combination of Western-style diets and ampicillin treatment synergize in microbiome impairment, particularly noticeable in the presence of high dietary fat content. The diet-independent degradation of ampicillin in the gut lumen by K. oxytoca beta-lactamases facilitates rapid commensal outgrowth, which is required for subsequent pathogen clearance. Our findings provide insights into how diet modulates functional microbiome recovery and K. oxytoca-mediated pathogen elimination from the gut.
Collapse
Affiliation(s)
- Éva D H Almási
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Lea Eisenhard
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Lisa Osbelt
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Till Robin Lesker
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Anna C Vetter
- Department of Chemical Biology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Nele Knischewski
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Agata Anna Bielecka
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Achim Gronow
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Uthayakumar Muthukumarasamy
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Marie Wende
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Caroline Tawk
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Meina Neumann-Schaal
- Bacterial Metabolomics, Leibniz Institute DSMZ-German Collection of Microorganisms and Cell Cultures, Braunschweig, Germany
| | - Mark Brönstrup
- Department of Chemical Biology, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany
| | - Till Strowig
- Department of Microbial Immune Regulation, Helmholtz Centre for Infection Research (HZI), Braunschweig, Germany.
- Center for Individualized Infection Medicine, Hannover, Germany.
- German Center for Infection Research (DZIF), partner site Hannover-Braunschweig, Braunschweig, Germany.
| |
Collapse
|
25
|
de Barros Santos HS, Pagnussatti MEL, Arthur RA. Symbiosis Between the Oral Microbiome and the Human Host: Microbial Homeostasis and Stability of the Host. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1472:31-51. [PMID: 40111684 DOI: 10.1007/978-3-031-79146-8_3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
The oral cavity presents a highly diverse microbial composition. All the three domains of life, Bacteria, Eukarya, and Archaea, as well as viruses constitute the oral microbiome. Bacteria are among the most abundant microorganisms in the oral cavity, followed by viruses, fungi, and Archaea. These microorganisms tend to live in harmony with each other and with the host by preventing the colonization of oral sites by exogenous microorganisms. Interactions between the host and its microbiota are crucial for keeping ecological stability in the oral cavity and a condition compatible with oral health. This chapter focuses on describing the oral microbiota in healthy individuals based on both targeted and nontargeted genome sequencing methods and the functional activity played by those microorganisms based on metagenomic, metatranscriptomic, metaproteomic, and metabolomic analyses. Additionally, this chapter explores mutualistic and antagonistic microbe-microbe relationships. These interactions are mediated by complex mechanisms like cross-feeding networks, production of bacteriocins and secondary metabolites, synthesis of pH-buffering compounds, and the use of universal signaling molecules. At last, the role played by host-microbe interactions on colonization resistance and immune tolerance will help provide a better understanding about the harmonious and peaceful coexistence among host and microbial cells under oral health-related conditions.
Collapse
Affiliation(s)
- Heitor Sales de Barros Santos
- Preventive and Community Dentistry Department, Dental School, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Maria Eduarda Lisbôa Pagnussatti
- Preventive and Community Dentistry Department, Dental School, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| | - Rodrigo Alex Arthur
- Preventive and Community Dentistry Department, Dental School, Federal University of Rio Grande do Sul (UFRGS), Porto Alegre, Brazil
| |
Collapse
|
26
|
Colombo APV, Lourenço TGB, de Oliveira AM, da Costa ALA. Link Between Oral and Gut Microbiomes: The Oral-Gut Axis. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2025; 1472:71-87. [PMID: 40111686 DOI: 10.1007/978-3-031-79146-8_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/22/2025]
Abstract
In the last decades, groundbreaking research on the human microbiome has changed our reductionist conception of the etiology and pathogenesis of several chronic diseases. As a result, we have come to appreciate the significance of a balanced microbiome in maintaining human health. In this context, the upper and lower gastrointestinal tracts (GITs) comprise the most abundant and diverse microbiotas of the human body. In addition to its diversity, functional redundancy, and temporal stability, a healthy GIT microbiome is characterized by its body site specificity. In fact, current evidence has indicated that the translocation of oral species to the gut environment through the oral-gut axis is increased in an array of illnesses, including chronic inflammatory and metabolic diseases, neurological disorders, and cancer. Oral pathogens have also been shown to promote gut dysbiosis and systemic inflammation in animal models. Yet, some level of overlapping between oral and gut microbiomes may occur without disruption of these microbial communities and loss of site specificity. The uniqueness of each host-microbiome entity may hinder our ability to define a "universal" normal GIT microbiome. Despite that, this chapter summarizes the predominant health-related taxa along the human GIT, as well as their role in the physiology and immunity of the digestive system. Some mechanisms that may lead to disturbances and relevant shifts in the oral and gut microbiomes of major inflammatory chronic diseases are also pointed out. Lastly, oral-fecal microbial signatures are presented as potential biomarkers for several oral and systemic disorders. The recognition of such symbiotic/dysbiotic microbial profiles may provide insights into the development of more accurate early diagnosis and therapeutic ecological approaches to restore the balance of the GIT microbiome.
Collapse
Affiliation(s)
- Ana Paula Vieira Colombo
- Oral Microbiology Laboratory, Institute of Microbiology Paulo de Góes, UFRJ, Rio de Janeiro, Brazil.
- School of Dentistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil.
| | | | - Adriana Miranda de Oliveira
- Oral Microbiology Laboratory, Institute of Microbiology Paulo de Góes, UFRJ, Rio de Janeiro, Brazil
- School of Dentistry, Federal University of Rio de Janeiro, Rio de Janeiro, Brazil
| | | |
Collapse
|
27
|
Sirko J, Bor B, He X. Microbial dark matter and the future of dentistry. J Am Dent Assoc 2025; 156:81-84. [PMID: 39779069 DOI: 10.1016/j.adaj.2024.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2025]
|
28
|
Dhanawat M, Malik G, Wilson K, Gupta S, Gupta N, Sardana S. The Gut Microbiota-Brain Axis: A New Frontier in Alzheimer's Disease Pathology. CNS & NEUROLOGICAL DISORDERS DRUG TARGETS 2025; 24:7-20. [PMID: 38967078 DOI: 10.2174/0118715273302508240613114103] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Revised: 05/12/2024] [Accepted: 05/17/2024] [Indexed: 07/06/2024]
Abstract
Dr. Aloysius Alzheimer, a German neuropathologist and psychiatrist, recognized the primary instance of Alzheimer's disease (AD) for a millennium, and this ailment, along with its related dementias, remains a severe overall community issue related to health. Nearly fifty million individuals worldwide suffer from dementia, with Alzheimer's illness contributing to between 60 and 70% of the instances, estimated through the World Health Organization. In addition, 82 million individuals are anticipated to be affected by the global dementia epidemic by 2030 and 152 million by 2050. Furthermore, age, environmental circumstances, and inherited variables all increase the likelihood of acquiring neurodegenerative illnesses. Most recent pharmacological treatments are found in original hypotheses of disease, which include cholinergic (drugs that show affective cholinergic system availability) as well as amyloid-accumulation (a single drug is an antagonist receptor of Nmethyl D-aspartate). In 2020, the FDA provided approval on anti-amyloid drugs. According to mounting scientific data, this gut microbiota affects healthy physiological homeostasis and has a role in the etiology of conditions that range between obesity and neurodegenerative disorders like Alzheimer's. The microbiota-gut-brain axis might facilitate interconnection among gut microbes as well as the central nervous system (CNS). Interaction among the microbiota-gut system as well as the brain occurs through the "two-way" microbiota-gut-brain axis. Along this axis, the stomach as well as the brain develop physiologically and take on their final forms. This contact is constant and is mediated by numerous microbiota-derived products. The gut microbiota, for instance, can act as non-genetic markers to set a threshold for maintaining homeostasis or getting ill. The scientific community has conducted research and found that bowel dysbiosis and gastrointestinal tract dysregulation frequently occur in Alzheimer's disease (AD) patients. In this review, the effects of the microbiota- gut-brain axis on AD pathogenesis will be discussed.
Collapse
Affiliation(s)
- Meenakshi Dhanawat
- Amity Institute of Pharmacy, Amity University Haryana, Gurugram 122413, India
| | - Garima Malik
- Department of Pharmaceutics, M.M College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India
| | - Kashish Wilson
- Department of Pharmaceutics, M.M College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India
| | - Sumeet Gupta
- Department of Pharmaceutics, M.M College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India
| | - Nidhi Gupta
- Department of Pharmaceutics, M.M College of Pharmacy, Maharishi Markandeshwar (Deemed to be University), Mullana-Ambala, Haryana 133207, India
| | - Satish Sardana
- Amity Institute of Pharmacy, Amity University Haryana, Gurugram 122413, India
| |
Collapse
|
29
|
Wang Y, Bai M, Peng Q, Li L, Tian F, Guo Y, Jing C. Angiogenesis, a key point in the association of gut microbiota and its metabolites with disease. Eur J Med Res 2024; 29:614. [PMID: 39710789 DOI: 10.1186/s40001-024-02224-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2024] [Accepted: 12/15/2024] [Indexed: 12/24/2024] Open
Abstract
The gut microbiota is a complex and dynamic ecosystem that plays a crucial role in human health and disease, including obesity, diabetes, cardiovascular diseases, neurodegenerative diseases, inflammatory bowel disease, and cancer. Chronic inflammation is a common feature of these diseases and is closely related to angiogenesis (the process of forming new blood vessels), which is often dysregulated in pathological conditions. Inflammation potentially acts as a central mediator. This abstract aims to elucidate the connection between the gut microbiota and angiogenesis in various diseases. The gut microbiota influences angiogenesis through various mechanisms, including the production of metabolites that directly or indirectly affect vascularization. For example, short-chain fatty acids (SCFAs) such as butyrate, propionate, and acetate are known to regulate immune responses and inflammation, thereby affecting angiogenesis. In the context of cardiovascular diseases, the gut microbiota promotes atherosclerosis and vascular dysfunction by producing trimethylamine N-oxide (TMAO) and other metabolites that promote inflammation and endothelial dysfunction. Similarly, in neurodegenerative diseases, the gut microbiota may influence neuroinflammation and the integrity of the blood-brain barrier, thereby affecting angiogenesis. In cases of fractures and wound healing, the gut microbiota promotes angiogenesis by activating inflammatory responses and immune effects, facilitating the healing of tissue damage. In cancer, the gut microbiota can either inhibit or promote tumor growth and angiogenesis, depending on the specific bacterial composition and their metabolites. For instance, some bacteria can activate inflammasomes, leading to the production of inflammatory factors that alter the tumor immune microenvironment and activate angiogenesis-related signaling pathways, affecting tumor angiogenesis and metastasis. Some bacteria can directly interact with tumor cells, activating angiogenesis-related signaling pathways. Diet, as a modifiable factor, significantly influences angiogenesis through diet-derived microbial metabolites. Diet can rapidly alter the composition of the microbiota and its metabolic activity, thereby changing the concentration of microbial-derived metabolites and profoundly affecting the host's immune response and angiogenesis. For example, a high animal protein diet promotes the production of pro-atherogenic metabolites like TMAO, activating inflammatory pathways and interfering with platelet function, which is associated with the severity of coronary artery plaques, peripheral artery disease, and cardiovascular diseases. A diet rich in dietary fiber promotes the production of SCFAs, which act as ligands for cell surface or intracellular receptors, regulating various biological processes, including inflammation, tissue homeostasis, and immune responses, thereby influencing angiogenesis. In summary, the role of the gut microbiota in angiogenesis is multifaceted, playing an important role in disease progression by affecting various biological processes such as inflammation, immune responses, and multiple signaling pathways. Diet-derived microbial metabolites play a crucial role in linking the gut microbiota and angiogenesis. Understanding the complex interactions between diet, the gut microbiota, and angiogenesis has the potential to uncover novel therapeutic targets for managing these conditions. Therefore, interventions targeting the gut microbiota and its metabolites, such as through fecal microbiota transplantation (FMT) and the application of probiotics to alter the composition of the gut microbiota and enhance the production of beneficial metabolites, present a promising therapeutic strategy.
Collapse
Affiliation(s)
- Yan Wang
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Mingshuai Bai
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Qifan Peng
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
| | - Leping Li
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China
| | - Feng Tian
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China.
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| | - Ying Guo
- Department of Breast and Thyroid Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| | - Changqing Jing
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital, Shandong University, Jinan, 250021, Shandong, China.
- Department of Gastrointestinal Surgery, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, 250021, Shandong, China.
| |
Collapse
|
30
|
Avain A, Azad MAK, García Y, García Y, Martínez Y. Effects of Ganoderma lucidum Powder on the Growth Performance, Immune Organ Weights, Cecal Microbiology, Serum Immunoglobulins, and Tibia Minerals of Broiler Chickens. Vet Sci 2024; 11:675. [PMID: 39729015 DOI: 10.3390/vetsci11120675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 12/28/2024] Open
Abstract
A total of 640 one-day-old Cobb 500 MV × Cobb 500 FF mixed broilers were randomly assigned to one of four experimental treatments with four replicates per treatment and 40 birds per replicate for 32 days. The treatments consisted of a basal diet (control group), basal diet + 0.02% zinc bacitracin (AGP group), basal diet + 0.2% G. lucidum powder (GLP; 0.2% GLP group), and basal diet + 0.3% GLP (0.3% GLP group). The results showed that dietary 0.2% GLP supplementation increased body weight compared to the control and 0.3% GLP groups, and decreased feed conversion ratio (FCR) compared to the control group, during 19-32 days (p < 0.05). The feed intake was lower (p < 0.05) in both dietary GLP supplementation groups and the AGP group during 1-8 and 1-32 days compared to the control group. Additionally, the FCR was lower in the dietary GLP supplementation group (0.2%) and the AGP group (p < 0.05) compared to the control group. Moreover, the caeca of broiler chickens in the AGP and 0.2% GLP groups had a higher abundance of lactic acid bacteria (LAB). Supplementation of feed additives (AGP and GLP) increased the relative weight of the thymus, with no effect on the bursa of Fabricius and spleen. However, AGP supplementation decreased the serum IgM concentration, while supplementing a higher dose of GLP (0.3%) increased the ash content in the tibia. The findings indicate that 0.2% GLP is the recommended supplementation dose as a natural growth promoter to replace AGP in apparently normal chickens.
Collapse
Affiliation(s)
- Arazay Avain
- Sustainable Tropical Agriculture Master Program, Zamorano University, Valle de Yeguare, San Antonio de Oriente P.O. Box 93, Francisco Morazán, Honduras
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Processes, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
- Department of Monogastric Animals, Institute of Animal Science, Central Highway km 47 ½, San José de las Lajas C.P. 32700, Mayabeque, Cuba
| | - Md Abul Kalam Azad
- CAS Key Laboratory of Agro-Ecological Processes in Subtropical Region, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Processes, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha 410125, China
| | - Yaneisy García
- Department of Monogastric Animals, Institute of Animal Science, Central Highway km 47 ½, San José de las Lajas C.P. 32700, Mayabeque, Cuba
| | - Yanelys García
- Department of Monogastric Animals, Institute of Animal Science, Central Highway km 47 ½, San José de las Lajas C.P. 32700, Mayabeque, Cuba
| | - Yordan Martínez
- Department of Monogastric Animals, Institute of Animal Science, Central Highway km 47 ½, San José de las Lajas C.P. 32700, Mayabeque, Cuba
- Faculty of Veterinary Medicine, University of Fondwa, Léogâne 6320, Haiti
| |
Collapse
|
31
|
Birabaharan M, Kaelber DC, Nizet V, Zarrinpar A. Gut-derived PPAR-γ signaling and risk of bacterial enteric infection: insight from thiazolidinedione users in a US population-based study. MEDRXIV : THE PREPRINT SERVER FOR HEALTH SCIENCES 2024:2024.09.24.24313682. [PMID: 39398997 PMCID: PMC11469470 DOI: 10.1101/2024.09.24.24313682] [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: 10/15/2024]
Abstract
Background and Aims The ongoing antimicrobial resistant crisis heralds the need for new therapeutics against enteric infection. In mouse models, colon epithelial peroxisome proliferator-activated receptor-γ (PPAR-γ) signaling limits oxygen and nitrate luminal bioavailability, thereby preventing bacterial pathogen colonization. However, whether this mechanism operates similarly in humans remains uncertain. Methods To investigate, we used the cloud-based TriNetX Analytics Platform which aggregates health records from 117 million patients across 66 US healthcare organizations, to assess the risk of bacterial enteric infection among diabetic patients prescribed thiazolidinediones, a class of PPAR-γ agonists, to other anti-diabetes medications. Results Among 85,117 thiazolidinedione users, we observed a 22-49% lower risk of bacterial enteric infections compared to users of other anti-diabetes medications. This reduction in risk was consistent across high-risk individuals, regardless of sex or age. Similar results were replicated in high-risk patients when thiazolidinedione users were directly compared to those on DPP-4 inhibitors. Conclusion These findings support the potential protective role of PPAR-γ signaling against bacterial enteric infection and call for further clinical investigation.
Collapse
Affiliation(s)
- Morgan Birabaharan
- Division of Infectious Diseases and Global Public Health, University of California, San Diego, La Jolla, California, USA
| | - David C Kaelber
- Departments of Population and Quantitative Health Sciences, Case Western Reserve University School of Medicine, Cleveland, Ohio, USA
| | - Victor Nizet
- Division of Host-Microbe Systems and Therapeutics, Department of Pediatrics, UC San Diego School of Medicine
- Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California, San Diego, La Jolla, California
| | - Amir Zarrinpar
- Division of Gastroenterology, University of California, San Diego, La Jolla, CA, USA
- Center for Microbiome Innovation, University of California, San Diego, La Jolla, CA, USA
- Division of Gastroenterology, Jennifer Moreno Department of Veterans Affairs Medical Center
- Institute of Diabetes and Metabolic Health, University of California, San Diego, La Jolla, CA, USA
- Shu Chien-Gene Lay Department of Bioengineering, University of California San Diego, La Jolla, CA, USA
| |
Collapse
|
32
|
García G, Soto J, Netherland M, Hasan NA, Buchaca E, Martínez D, Carlin M, de Jesus Cano R. Evaluating the Effects of Sugar Shift ® Symbiotic on Microbiome Composition and LPS Regulation: A Double-Blind, Placebo-Controlled Study. Microorganisms 2024; 12:2525. [PMID: 39770729 PMCID: PMC11678924 DOI: 10.3390/microorganisms12122525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2024] [Revised: 12/04/2024] [Accepted: 12/05/2024] [Indexed: 01/11/2025] Open
Abstract
(1) Background: This study evaluated the effects of BiotiQuest® Sugar Shift®, a novel probiotic formulation, for its impact on gut microbiome composition and metabolic health in type 2 diabetes mellitus (T2D). T2D is characterized by chronic inflammation and gut microbiome imbalances, yet the therapeutic potential of targeted probiotics remains underexplored. (2) Methods: In a 12-week randomized, double-blind, placebo-controlled trial, 64 adults with T2D received either Sugar Shift or placebo capsules twice daily. Each dose provided 18 billion CFU of eight GRAS-certified bacterial strains and prebiotics. Clinical samples were analyzed for metabolic markers, and microbiome changes were assessed using 16S rRNA sequencing and metagenomics. (3) Results: Sugar Shift significantly reduced serum lipopolysaccharide (LPS) levels, improved insulin sensitivity (lower HOMA-IR scores), and increased short-chain fatty acid (SCFA)-producing genera, including Bifidobacterium, Faecalibacterium, Fusicatenibacter, and Roseburia. Pro-inflammatory taxa like Enterobacteriaceae decreased, with reduced LPS biosynthesis genes and increased SCFA production genes. The Lachnospiraceae:Enterobactericeae ratio emerged as a biomarker of reduced inflammation. (4) Conclusions: These findings demonstrate the potential of Sugar Shift to restore gut homeostasis, reduce inflammation, and improve metabolic health in T2D. Further studies are warranted to explore its long-term efficacy and broader application in metabolic disease management.
Collapse
Affiliation(s)
- Gissel García
- Pathology Department, Clinical Hospital “Hermanos Ameijeiras” (HHA), Calle San Lázaro No 701, Esq.a Belascoaín, Centro Habana, La Habana 10400, Cuba;
| | - Josanne Soto
- Clinical Laboratory Department, Clinical Hospital “Hermanos Ameijeiras” (HHA), Calle San Lázaro No 701, Esq.a Belascoaín, Centro Habana, La Habana 10400, Cuba;
| | | | - Nur A. Hasan
- EzBiome, 704 Quince Orchard Rd, Gaithersburg, MD 20878, USA (N.A.H.)
| | - Emilio Buchaca
- Internal Medicine Department, Clinical Hospital “Hermanos Ameijeiras” (HHA), Calle San Lázaro No 701, Esq.a Belascoaín, Centro Habana, La Habana 10400, Cuba;
| | - Duniesky Martínez
- Research and Development Department, Center for Genetic Engineering and Biotechnology of Sancti Spíritus (CIGBSS), Circunvalante Norte S/N, Olivos 3, Apartado Postal 83, Sancti Spíritus 60200, Cuba;
| | - Martha Carlin
- The BioCollective, LLC, 4800 Dahlia Street, G8, Denver, CO 80216, USA;
| | - Raúl de Jesus Cano
- Biological Sciences Department, California Polytechnic State University, San Luis Obispo, CA 93407, USA
| |
Collapse
|
33
|
Xu Z, Wu XM, Luo YB, Li H, Zhou YQ, Liu ZQ, Li ZY. Exploring the therapeutic potential of yeast β-glucan: Prebiotic, anti-infective, and anticancer properties - A review. Int J Biol Macromol 2024; 283:137436. [PMID: 39522898 DOI: 10.1016/j.ijbiomac.2024.137436] [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: 08/24/2024] [Revised: 10/31/2024] [Accepted: 11/07/2024] [Indexed: 11/16/2024]
Abstract
Yeast β-glucan (YBG), an indigestible polysaccharide from yeast cell walls, is multifunctional. It plays a pivotal role in regulating gut microbiota (GM) and boosting the immune system, which is central to research on inflammation, cancer, and metabolic diseases. By modulating the GM, YBG exhibits various prebiotic effects, including hypoglycemic, hypolipidemic, and immune-regulating properties. Additionally, acting as a bioreactor modulator, it activates immune responses, demonstrating potential in anti-infection and anticancer applications. This article synthesizes the latest data from in vitro, in vivo, and clinical studies. It comprehensively evaluates the therapeutic potential of YBG, starting from its structure-function relationship. It particularly focuses on the application prospects of yeast β-glucan in probiotic-like effects, anti-infectious properties, and anti-cancer activity, and explores the underlying mechanisms of these actions. The aim of this article is to elucidate the positive impact of YBG on health by modulating the gut microbiota and enhancing immune responses. Simultaneously, it identifies critical areas for future research to provide theoretical support for its development in biomedical applications.
Collapse
Affiliation(s)
- Zhen Xu
- The Second Clinical Medical College, China Three Gorges University, Yichang, Hubei 443002, China
| | - Xiao Meng Wu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
| | - Yan Bin Luo
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
| | - Hui Li
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443002, China
| | - Yong Qin Zhou
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443002, China.
| | - Zhao Qi Liu
- Hubei Key Laboratory of Tumor Microenvironment and Immunotherapy, College of Basic Medical Science, China Three Gorges University, Yichang 443002, China.
| | - Zhi Ying Li
- The Second Clinical Medical College, China Three Gorges University, Yichang, Hubei 443002, China.
| |
Collapse
|
34
|
Yang X, Hang HC. Chemical genetic approaches to dissect microbiota mechanisms in health and disease. Science 2024; 386:eado8548. [PMID: 39541443 DOI: 10.1126/science.ado8548] [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: 05/08/2024] [Accepted: 10/04/2024] [Indexed: 11/16/2024]
Abstract
Advances in genomics, proteomics, and metabolomics have revealed associations between specific microbiota species in health and disease. However, the precise mechanism(s) of action for many microbiota species and molecules have not been fully elucidated, limiting the development of microbiota-based diagnostics and therapeutics. In this Review, we highlight innovative chemical and genetic approaches that are enabling the dissection of microbiota mechanisms and providing causation in health and disease. Although specific microbiota molecules and mechanisms have begun to emerge, new approaches are still needed to go beyond phenotypic associations and translate microbiota discoveries into actionable targets and therapeutic leads to prevent and treat diseases.
Collapse
Affiliation(s)
- Xinglin Yang
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA, USA
| | - Howard C Hang
- Department of Immunology and Microbiology, Scripps Research Institute, La Jolla, CA, USA
- Department of Chemistry, Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
35
|
Chen Z, Lu Y, Xu Z, Wu L, Wei X, Cai Y. Evaluation of a Burkholderia ambifaria strain from plants as a novel promising probiotic in dental caries management. J Oral Microbiol 2024; 16:2420612. [PMID: 39502190 PMCID: PMC11536693 DOI: 10.1080/20002297.2024.2420612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 10/04/2024] [Accepted: 10/16/2024] [Indexed: 11/08/2024] Open
Abstract
Background Probiotics serve as a novel preventive or therapeutic approach for dental caries owing to their ability to reverse dysbiosis and restore a healthy microbiota. Here, we identified Burkholderia ambifaria AFS098024 as a probiotic candidate isolated from plants. Methods The safety of B. ambifaria was evaluated by hemolytic activity, D-lactic acid production and antibiotic susceptibility. In vitro biofilm model derived from the saliva of caries-free and caries-active donors and in vivo rat caries model were used to assess the efficacy of B. ambifaria in caries prevention and treatment. Results B. ambifaria was safe as a probiotic candidate and it could integrate with in vitro biofilm model. It significantly reduced the biomass and lactate production of biofilms from caries-active donors and disrupted biofilm structures. B. ambifaria effectively reduced the severity of carious lesions in rat molars, regardless of the inoculation sequence. Molars pretreated or treated with B. ambifaria demonstrated notably higher enamel volumes. Additionally, colonization of rat molars by B. ambifaria persisted for 6 weeks. Conclusion The B. ambifaria strain used in this study holds promise as a probiotic for inhibiting dental caries, both in vitro and in vivo.
Collapse
Affiliation(s)
- Zirang Chen
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yangyu Lu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Zhezhen Xu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Lijing Wu
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Xi Wei
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| | - Yanling Cai
- Hospital of Stomatology, Guanghua School of Stomatology, Sun Yat-sen University, Guangzhou, China
- Guangdong Provincial Key Laboratory of Stomatology, Sun Yat-sen University, Guangzhou, China
| |
Collapse
|
36
|
Lamminpää I, Boem F, Amedei A. Health-promoting worms? Prospects and pitfalls of helminth therapy. Bioessays 2024; 46:e2400080. [PMID: 39263744 DOI: 10.1002/bies.202400080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 08/28/2024] [Accepted: 09/02/2024] [Indexed: 09/13/2024]
Abstract
In this manuscript, we explore the potential therapeutic use of helminths. After analyzing helminths' role in connection with human health from the perspective of their symbiotic and evolutionary relationship, we critically examine some studies on their therapeutic applications. In doing so, we focus on some prominent mechanisms of action and potential benefits, but also on the exaggerations and theoretical and methodological difficulties of such proposals. We conclude that further studies are needed to fully explore the potential benefits of this perspective, and we encourage the scientific community in doing so.
Collapse
Affiliation(s)
- Ingrid Lamminpää
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
| | - Federico Boem
- Institut für Philosophie I, Ruhr-Universität Bochum, Bochum, Germany
| | - Amedeo Amedei
- Department of Experimental and Clinical Medicine, University of Florence, Florence, Italy
- Interdisciplinary Internal Medicine Unit, Careggi University Hospital, Florence, Italy
| |
Collapse
|
37
|
Xiang X, Zhu Y, Wang T, Ding P, Cheng K, Ming Y. Association between salivary microbiota and tacrolimus pharmacokinetic variability in kidney transplant. Genomics 2024; 116:110952. [PMID: 39426572 DOI: 10.1016/j.ygeno.2024.110952] [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: 03/15/2024] [Revised: 09/02/2024] [Accepted: 10/14/2024] [Indexed: 10/21/2024]
Abstract
Kidney transplantation (KT) serves as a highly effective treatment for end-stage renal disease (ESRD). Nonetheless, the administration of tacrolimus, a commonly used immunosuppressant in KT, faces challenges due to the lack of dependable biomarkers for its efficacy and the considerable variability in tacrolimus pharmacokinetics (TacIPV). In this study, 183 saliva samples from 48 KT recipients under tacrolimus therapy, alongside 9 healthy control samples, were subjected to 16S rRNA sequencing. The analysis revealed significant differences in the composition of salivary microbiota among KT recipients, patients with ESRD, and healthy controls. Moreover, trough blood concentrations (C0) of tacrolimus were associated with alterations in microbiota composition. Notably, Capnocytophage consistently exhibited a negative correlation in both group-level and individual trends. Furthermore, distinct taxa were identified that effectively distinguished recipients with varying TacIPV, as demonstrated by a cross-validation random forest model (mean AUC = 0.7560), with Anaerolinea emerging as a prominent contributor to the classifier. These findings suggest that salivary microbiota is closely linked to tacrolimus C0 levels and could aid clinicians in differentiating KT recipients based on TacIPV.
Collapse
Affiliation(s)
- Xuyu Xiang
- The Transplantation Center of the Third Xiangya Hospital, Central South University, Changsha 410013, China; Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Yi Zhu
- The Transplantation Center of the Third Xiangya Hospital, Central South University, Changsha 410013, China; Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Tianyin Wang
- The Transplantation Center of the Third Xiangya Hospital, Central South University, Changsha 410013, China; Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Peng Ding
- The Transplantation Center of the Third Xiangya Hospital, Central South University, Changsha 410013, China; Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Ke Cheng
- The Transplantation Center of the Third Xiangya Hospital, Central South University, Changsha 410013, China; Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China
| | - Yingzi Ming
- The Transplantation Center of the Third Xiangya Hospital, Central South University, Changsha 410013, China; Engineering and Technology Research Center for Transplantation Medicine of National Health Commission, Changsha, China.
| |
Collapse
|
38
|
Mehlferber EC, Arnault G, Joshi B, Partida-Martinez LP, Patras KA, Simonin M, Koskella B. A cross-systems primer for synthetic microbial communities. Nat Microbiol 2024; 9:2765-2773. [PMID: 39478083 PMCID: PMC11660114 DOI: 10.1038/s41564-024-01827-2] [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: 02/02/2024] [Accepted: 09/11/2024] [Indexed: 11/02/2024]
Abstract
The design and use of synthetic communities, or SynComs, is one of the most promising strategies for disentangling the complex interactions within microbial communities, and between these communities and their hosts. Compared to natural communities, these simplified consortia provide the opportunity to study ecological interactions at tractable scales, as well as facilitating reproducibility and fostering interdisciplinary science. However, the effective implementation of the SynCom approach requires several important considerations regarding the development and application of these model systems. There are also emerging ethical considerations when both designing and deploying SynComs in clinical, agricultural or environmental settings. Here we outline current best practices in developing, implementing and evaluating SynComs across different systems, including a focus on important ethical considerations for SynCom research.
Collapse
Affiliation(s)
- Elijah C Mehlferber
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA.
- Center for Microbial Dynamics and Infection, Georgia Institute of Technology, Atlanta, GA, USA.
| | - Gontran Arnault
- Univ Angers, Institut Agro, INRAE, IRHS, SFR QUASAV, Angers, France
| | - Bishnu Joshi
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
| | - Laila P Partida-Martinez
- Departamento de Ingeniería Genética, Centro de Investigación y de Estudios Avanzados del Instituto Politécnico Nacional (Cinvestav), Irapuato, México
| | - Kathryn A Patras
- Department of Molecular Virology and Microbiology, Baylor College of Medicine, Houston, TX, USA
- Alkek Center for Metagenomics and Microbiome Research, Baylor College of Medicine, Houston, TX, USA
| | - Marie Simonin
- Univ Angers, Institut Agro, INRAE, IRHS, SFR QUASAV, Angers, France
| | - Britt Koskella
- Department of Integrative Biology, University of California, Berkeley, CA, USA.
- San Francisco Chan Zuckerberg Biohub, San Francisco, CA, USA.
| |
Collapse
|
39
|
Shang KM, Elsheikha HM, Ma H, Wei YJ, Zhao JX, Qin Y, Li JM, Zhao ZY, Zhang XX. Metagenomic profiling of cecal microbiota and antibiotic resistome in rodents. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2024; 286:117186. [PMID: 39426111 DOI: 10.1016/j.ecoenv.2024.117186] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/18/2024] [Accepted: 10/10/2024] [Indexed: 10/21/2024]
Abstract
The rodent gut microbiota is a known reservoir of antimicrobial resistance, yet the distribution of antibiotic resistance genes (ARGs) within rodent cecal microbial communities and the specific bacterial species harboring these ARGs remain largely underexplored. This study employed high-throughput sequencing of 122 samples from five distinct rodent species to comprehensively profile the diversity and distribution of ARGs and to identify the bacterial hosts of these genes. A gene catalog of the rodent cecal microbiome was constructed, comprising 22,757,369 non-redundant genes. Analysis of the microbial composition and diversity revealed that Bacillota and Bacteroidota were the dominant bacterial phyla across different rodent species, with significant variations in species composition among the rodents. In total, 3703 putative antimicrobial resistance protein-coding genes were identified, corresponding to 392 unique ARG types classified into 32 resistance classes. The most enriched ARGs in the rodent cecal microbiome were associated with multidrug resistance, followed by glycopeptide and elfamycin antibiotics. Procrustes analysis demonstrated a correlation between the structure of the microbial community and the resistome. Metagenomic assembly-based host tracking indicated that most ARG-carrying contigs originated from the bacterial family Oscillospiraceae. Additionally, 130 ARGs showed significant correlations with mobile genetic elements. These findings provide new insights into the cecal microbiota and the prevalence of ARGs across five rodent species. Future research on a wider range of wild rodent species carrying ARGs will further elucidate the mechanisms underlying the transmission of antimicrobial resistance.
Collapse
Affiliation(s)
- Kai-Meng Shang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, PR China
| | - Hany M Elsheikha
- Faculty of Medicine and Health Sciences, School of Veterinary Medicine and Science, University of Nottingham, Sutton Bonington Campus, Loughborough, United Kingdom
| | - He Ma
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, PR China
| | - Yong-Jie Wei
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, PR China
| | - Ji-Xin Zhao
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, PR China
| | - Ya Qin
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, PR China; College of Veterinary Medicine, Jilin Agricultural University, Changchun, Jilin Province, PR China
| | - Jian-Ming Li
- College of Chinese Medicinal Materials, Jilin Agricultural University, Changchun, Jilin Province, PR China; Jilin Provincial Engineering Research Center for Efficient Breeding and Product Development of Sika Deer, Changchun, Jilin Province, PR China; Key Laboratory of Animal Production and Product Quality and Security, Ministry of Education, Ministry of National Education, Changchun, Jilin Province, PR China
| | - Zi-Yu Zhao
- College of Veterinary Medicine, Jilin Agricultural University, Changchun, Jilin Province, PR China
| | - Xiao-Xuan Zhang
- College of Veterinary Medicine, Qingdao Agricultural University, Qingdao, Shandong Province, PR China.
| |
Collapse
|
40
|
Liu Y, He L, Hu Y, Liao X, Wang H, Yang L. Synthetic bacterial consortia transplantation attenuates vaginal inflammation and modulates the immune response in a mouse model of Gardnerella vaginalis-induced bacterial vaginosis. Heliyon 2024; 10:e38218. [PMID: 39498013 PMCID: PMC11533556 DOI: 10.1016/j.heliyon.2024.e38218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 11/07/2024] Open
Abstract
This study aimed to evaluate the efficacy of synthetic bacterial consortia transplantation (SBCT) and compare it with VMT (vaginal microbiota transplantation) in a mouse model of Gardnerella vaginalis-induced Bacterial vaginosis (BV). A murine model of G. vaginalis-induced BV was established, and mice were treated with SBCT, VMT, or saline. Histopathological changes, inflammatory cytokine levels, pro-inflammatory biomarker expression, helper T cell transcription factor expression, and vaginal microbiota composition were assessed. SBCT and VMT effectively suppressed G. vaginalis growth, reduced inflammation, and restored vaginal microbiota diversity. Both treatments attenuated epithelial damage, downregulated pro-inflammatory cytokines (IL-1β and IL-8), and upregulated the anti-inflammatory cytokine IL-10. SBCT and VMT also inhibited NF-κB activation, suppressed IL-17 expression, and enhanced Foxp3 expression in vaginal tissues. SBCT is a promising therapeutic approach for treating BV, as it effectively modulates the immune response and restores vaginal microbiota diversity in a mouse model of G. vaginalis-induced BV.
Collapse
Affiliation(s)
- Ying Liu
- Department of Gynecology, Yunnan Cancer Hospital/The Third Affiliated Hospital of Kunming Medical University/Yunnan Cancer Center, Kunming, Yunnan Province, 650000,China
| | - Liang He
- Department of Laboratory, Yunnan Cancer Hospital/The Third Affiliated Hospital of Kunming Medical University/Yunnan Cancer Center, Kunming, Yunnan Province, 650000,China
| | - Yan Hu
- Department of Gynecology, Yunnan Cancer Hospital/The Third Affiliated Hospital of Kunming Medical University/Yunnan Cancer Center, Kunming, Yunnan Province, 650000,China
| | - Xingya Liao
- Department of Gynecology, Yunnan Cancer Hospital/The Third Affiliated Hospital of Kunming Medical University/Yunnan Cancer Center, Kunming, Yunnan Province, 650000,China
| | - Hongyan Wang
- Department of Gynecology, Yunnan Cancer Hospital/The Third Affiliated Hospital of Kunming Medical University/Yunnan Cancer Center, Kunming, Yunnan Province, 650000,China
| | - Linlin Yang
- Department of Gynecology, Yunnan Cancer Hospital/The Third Affiliated Hospital of Kunming Medical University/Yunnan Cancer Center, Kunming, Yunnan Province, 650000,China
| |
Collapse
|
41
|
Valiei A, Dickson AM, Aminian-Dehkordi J, Mofrad MRK. Bacterial community dynamics as a result of growth-yield trade-off and multispecies metabolic interactions toward understanding the gut biofilm niche. BMC Microbiol 2024; 24:441. [PMID: 39472801 PMCID: PMC11523853 DOI: 10.1186/s12866-024-03566-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 10/04/2024] [Indexed: 11/02/2024] Open
Abstract
Bacterial communities are ubiquitous, found in natural ecosystems, such as soil, and within living organisms, like the human microbiome. The dynamics of these communities in diverse environments depend on factors such as spatial features of the microbial niche, biochemical kinetics, and interactions among bacteria. Moreover, in many systems, bacterial communities are influenced by multiple physical mechanisms, such as mass transport and detachment forces. One example is gut mucosal communities, where dense, closely packed communities develop under the concurrent influence of nutrient transport from the lumen and fluid-mediated detachment of bacteria. In this study, we model a mucosal niche through a coupled agent-based and finite-volume modeling approach. This methodology enables us to model bacterial interactions affected by nutrient release from various sources while adjusting individual bacterial kinetics. We explored how the dispersion and abundance of bacteria are influenced by biochemical kinetics in different types of metabolic interactions, with a particular focus on the trade-off between growth rate and yield. Our findings demonstrate that in competitive scenarios, higher growth rates result in a larger share of the niche space. In contrast, growth yield plays a critical role in neutralism, commensalism, and mutualism interactions. When bacteria are introduced sequentially, they cause distinct spatiotemporal effects, such as deeper niche colonization in commensalism and mutualism scenarios driven by species intermixing effects, which are enhanced by high growth yields. Moreover, sub-ecosystem interactions dictate the dynamics of three-species communities, sometimes yielding unexpected outcomes. Competitive, fast-growing bacteria demonstrate robust colonization abilities, yet they face challenges in displacing established mutualistic systems. Bacteria that develop a cooperative relationship with existing species typically obtain niche residence, regardless of their growth rates, although higher growth yields significantly enhance their abundance. Our results underscore the importance of bacterial niche dynamics in shaping community properties and succession, highlighting a new approach to manipulating microbial systems.
Collapse
Affiliation(s)
- Amin Valiei
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, CA, 94720, USA
| | - Andrew M Dickson
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, CA, 94720, USA
| | - Javad Aminian-Dehkordi
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, CA, 94720, USA
| | - Mohammad R K Mofrad
- Molecular Cell Biomechanics Laboratory, Departments of Bioengineering and Mechanical Engineering, University of California, Berkeley, CA, 94720, USA.
- Molecular Biophysics and Integrative Bioimaging Division, Lawrence Berkeley National Lab, Berkeley, CA, 94720, USA.
| |
Collapse
|
42
|
Basu A, Adams AND, Degnan PH, Vanderpool CK. Determinants of raffinose family oligosaccharide use in Bacteroides species. J Bacteriol 2024; 206:e0023524. [PMID: 39330254 PMCID: PMC11501099 DOI: 10.1128/jb.00235-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2024] [Accepted: 09/06/2024] [Indexed: 09/28/2024] Open
Abstract
Bacteroides species are successful colonizers of the human colon and can utilize a wide variety of complex polysaccharides and oligosaccharides that are indigestible by the host. To do this, they use enzymes encoded in polysaccharide utilization loci (PULs). While recent work has uncovered the PULs required for the use of some polysaccharides, how Bacteroides utilize smaller oligosaccharides is less well studied. Raffinose family oligosaccharides (RFOs) are abundant in plants, especially legumes, and consist of variable units of galactose linked by α-1,6 bonds to a sucrose (glucose α-1-β-2 fructose) moiety. Previous work showed that an α-galactosidase, BT1871, is required for RFO utilization in Bacteroides thetaiotaomicron. Here, we identify two different types of mutations that increase BT1871 mRNA levels and improve B. thetaiotaomicron growth on RFOs. First, a novel spontaneous duplication of BT1872 and BT1871 places these genes under the control of a ribosomal promoter, driving high BT1871 transcription. Second, nonsense mutations in a gene encoding the PUL24 anti-sigma factor likewise increase BT1871 transcription. We then show that hydrolases from PUL22 work together with BT1871 to break down the sucrose moiety of RFOs and determine that the master regulator of carbohydrate utilization (BT4338) plays a role in RFO utilization in B. thetaiotaomicron. Examining the genomes of other Bacteroides species, we found homologs of BT1871 in a subset and showed that representative strains of species with a BT1871 homolog grew better on melibiose than species that lack a BT1871 homolog. Altogether, our findings shed light on how an important gut commensal utilizes an abundant dietary oligosaccharide. IMPORTANCE The gut microbiome is important in health and disease. The diverse and densely populated environment of the gut makes competition for resources fierce. Hence, it is important to study the strategies employed by microbes for resource usage. Raffinose family oligosaccharides are abundant in plants and are a major source of nutrition for the microbiota in the colon since they remain undigested by the host. Here, we study how the model commensal organism, Bacteroides thetaiotaomicron utilizes raffinose family oligosaccharides. This work highlights how an important member of the microbiota uses an abundant dietary resource.
Collapse
Affiliation(s)
- Anubhav Basu
- Department of Microbiology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| | - Amanda N. D. Adams
- Department of Microbiology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
- Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, Massachusetts, USA
| | - Patrick H. Degnan
- Department of Microbiology and Plant Pathology, University of California Riverside, Riverside, California, USA
| | - Carin K. Vanderpool
- Department of Microbiology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
- Carl R. Woese Institute for Genomic Biology, University of Illinois Urbana-Champaign, Urbana, Illinois, USA
| |
Collapse
|
43
|
Caruso R, Lo BC, Chen GY, Núñez G. Host-pathobiont interactions in Crohn's disease. Nat Rev Gastroenterol Hepatol 2024:10.1038/s41575-024-00997-y. [PMID: 39448837 DOI: 10.1038/s41575-024-00997-y] [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] [Accepted: 09/23/2024] [Indexed: 10/26/2024]
Abstract
The mammalian intestine is colonized by trillions of microorganisms that are collectively referred to as the gut microbiota. The majority of symbionts have co-evolved with their host in a mutualistic relationship that benefits both. Under certain conditions, such as in Crohn's disease, a subtype of inflammatory bowel disease, some symbionts bloom to cause disease in genetically susceptible hosts. Although the identity and function of disease-causing microorganisms or pathobionts in Crohn's disease remain largely unknown, mounting evidence from animal models suggests that pathobionts triggering Crohn's disease-like colitis inhabit certain niches and penetrate the intestinal tissue to trigger inflammation. In this Review, we discuss the distinct niches occupied by intestinal symbionts and the evidence that pathobionts triggering Crohn's disease live in the mucus layer or near the intestinal epithelium. We also discuss how Crohn's disease-associated mutations in the host disrupt intestinal homeostasis by promoting the penetration and accumulation of pathobionts in the intestinal tissue. Finally, we discuss the potential role of microbiome-based interventions in precision therapeutic strategies for the treatment of Crohn's disease.
Collapse
Affiliation(s)
- Roberta Caruso
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA.
| | - Bernard C Lo
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Grace Y Chen
- Department of Internal Medicine and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA
| | - Gabriel Núñez
- Department of Pathology and Rogel Cancer Center, University of Michigan Medical School, Ann Arbor, MI, USA.
| |
Collapse
|
44
|
Jung D, Park S, Kurban D, Dufour S, Ronholm J. The occurrence of Aerococcus urinaeequi and non-aureus staphylococci in raw milk negatively correlates with Escherichia coli clinical mastitis. mSystems 2024; 9:e0036224. [PMID: 39254336 PMCID: PMC11494980 DOI: 10.1128/msystems.00362-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2024] [Accepted: 08/06/2024] [Indexed: 09/11/2024] Open
Abstract
Escherichia coli is a common environmental pathogen associated with clinical mastitis (CM) in dairy cattle. There is an interest in optimizing the udder microbiome to increase the resistance of dairy cattle to E. coli CM; however, the details of which members of the healthy udder microbiome may play a role in antagonizing E. coli are unknown. In this study, we characterized the bacterial community composition in raw milk collected from quarters of lactating Holstein dairy cows that developed E. coli CM during lactation, including milk from both healthy and diseased quarters (n = 1,172). The milk microbiome from infected quarters was compared before, during, and after CM. A combination of 16S rRNA gene amplicon and metagenomic sequencing was used generate data sets with a high level of both depth and breadth. The microbial diversity present in raw milk significantly decreased in quarters experiencing E. coli CM, indicating that E. coli displaces other members of the microbiome. However, the diversity recovered very rapidly after infection. Two genera, Staphylococcus and Aerococcus, and the family Oscillospiraceae were significantly more abundant in healthy quarters with low inflammation. Species of these genera, Staphylococcus auricularis, Staphylococcus haemolyticus, and Aerocussus urinaeequi, were identified by metagenomics. Thus, these species are of interest for optimizing the microbiome to discourage E. coli colonization without triggering inflammation.IMPORTANCEIn this study, we show that E. coli outcompetes and displaces several members of the udder microbiome during CM, but that microbial diversity recovers post-infection. In milk from quarters which remained healthy, the community composition was often highly dominated by S. auricularis, S. haemolyticus, A. urinaeequi, and S. marcescens without increases in somatic cell count (SCC). Community dominance by these organisms, without inflammation, could indicate that these species might have potential as prophylactic probiotics which could contribute to colonization resistance and prevent future instances of E. coli CM.
Collapse
Affiliation(s)
- Dongyun Jung
- Faculty of Agricultural and Environmental Sciences, Macdonald Campus, McGill University, Quebec, Canada
- Mastitis Network, Saint-Hyacinthe, Québec, Canada
- Regroupement FRQNT Op+Lait, Saint-Hyacinthe, Québec, Canada
| | - Soyoun Park
- Faculty of Agricultural and Environmental Sciences, Macdonald Campus, McGill University, Quebec, Canada
- Mastitis Network, Saint-Hyacinthe, Québec, Canada
- Regroupement FRQNT Op+Lait, Saint-Hyacinthe, Québec, Canada
| | - Daryna Kurban
- Mastitis Network, Saint-Hyacinthe, Québec, Canada
- Regroupement FRQNT Op+Lait, Saint-Hyacinthe, Québec, Canada
- Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec, Canada
| | - Simon Dufour
- Mastitis Network, Saint-Hyacinthe, Québec, Canada
- Regroupement FRQNT Op+Lait, Saint-Hyacinthe, Québec, Canada
- Faculté de Médecine Vétérinaire, Université de Montréal, Saint-Hyacinthe, Québec, Canada
| | - Jennifer Ronholm
- Faculty of Agricultural and Environmental Sciences, Macdonald Campus, McGill University, Quebec, Canada
- Mastitis Network, Saint-Hyacinthe, Québec, Canada
- Regroupement FRQNT Op+Lait, Saint-Hyacinthe, Québec, Canada
| |
Collapse
|
45
|
Ren X, Clark RM, Bansah DA, Varner EN, Tiffany CR, Jaswal K, Geary JH, Todd OA, Winkelman JD, Friedman ES, Zemel BS, Wu GD, Zackular JP, DePas WH, Behnsen J, Palmer LD. Amino acid competition shapes Acinetobacter baumannii gut carriage. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.10.19.619093. [PMID: 39502362 PMCID: PMC11537318 DOI: 10.1101/2024.10.19.619093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/14/2024]
Abstract
Antimicrobial resistance is an urgent threat to human health. Asymptomatic colonization is often critical for persistence of antimicrobial-resistant pathogens. Gut colonization by the antimicrobial-resistant priority pathogen Acinetobacter baumannii is associated with increased risk of clinical infection. Ecological factors shaping A. baumannii gut colonization remain unclear. Here we show that A. baumannii and other pathogenic Acinetobacter evolved to utilize the amino acid ornithine, a non-preferred carbon source. A. baumannii utilizes ornithine to compete with the resident microbiota and persist in the gut in mice. Supplemental dietary ornithine promotes long-term fecal shedding of A. baumannii. By contrast, supplementation of a preferred carbon source-monosodium glutamate (MSG)-abolishes the requirement for A. baumannii ornithine catabolism. Additionally, we report evidence for diet promoting A. baumannii gut carriage in humans. Together, these results highlight that evolution of ornithine catabolism allows A. baumannii to compete with the microbiota in the gut, a reservoir for pathogen spread.
Collapse
Affiliation(s)
- Xiaomei Ren
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, IL, USA
| | - R. Mason Clark
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, IL, USA
| | - Dziedzom A. Bansah
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, IL, USA
| | - Elizabeth N. Varner
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Connor R. Tiffany
- Division of Protective Immunity, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Kanchan Jaswal
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, IL, USA
| | - John H. Geary
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, IL, USA
| | - Olivia A. Todd
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, IL, USA
| | | | - Elliot S. Friedman
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Babette S. Zemel
- Department of Pediatrics, Perelman School of Medicine University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Division of Gastroenterology, Hepatology, and Nutrition, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Gary D. Wu
- Division of Gastroenterology and Hepatology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Joseph P. Zackular
- Division of Protective Immunity, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pathology and Laboratory Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
- Center for Microbial Medicine, Children’s Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - William H. DePas
- Department of Pediatrics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Judith Behnsen
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, IL, USA
| | - Lauren D. Palmer
- Department of Microbiology and Immunology, University of Illinois Chicago, Chicago, IL, USA
| |
Collapse
|
46
|
Li S, Zhang W, Liu S, Zhou Y, Liu W, Yuan W, He M. Effects of Tanreqing injection on the gut microbiota in healthy volunteers. Front Cell Infect Microbiol 2024; 14:1428476. [PMID: 39431053 PMCID: PMC11486765 DOI: 10.3389/fcimb.2024.1428476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2024] [Accepted: 09/13/2024] [Indexed: 10/22/2024] Open
Abstract
Objectives Many studies have confirmed that antibacterial agents can disrupt the human gut microbiota. In China, Tanreqing injection (TRQ) is a drug with antibacterial activity that is widely used in the treatment of respiratory infections. However, its specific influence on gut microbiota remains unclear. This study aimed to investigate the effect of TRQ on the gut microbiota of healthy volunteers. Methods Twelve healthy adults received 20 ml of TRQ intravenously daily for 7 consecutive days. At six timepoints (Pre, on D1, D3, D5, D7 and follow-up visit) fecal samples were collected and analyzed using 16S rRNA gene sequencing. Results Eleven people were included in the analysis finally. TRQ did not significantly alter gut microbiota diversity or richness (Shannon and Simpson and Chao1 index) in healthy people during the intervention. Gut microbial structure was stable (weighted and unweighted Unifrac). Using a machine learning method based on PLS-DA analysis, the separation trend on D7 at the genus level was found, returning to baseline two days after discontinuation. The abundance of major genus fluctuated on D7 compared with that prior to treatment, including an increase of unclassified_f_Enterobacteriaceae (13.0611%), a decrease of Bifidobacterium and Escherichia-Shigella (6.887%, 10.487%). Functional prediction analysis did not reveal any significant difference. Conclusions Our study showed short-term use of TRQ at conventional doses may not cause perturbations to the gut microbiota in healthy adults. This finding provides some useful information for the safe use of TRQ in the treatment of respiratory infections. Clinical trial registration https://www.medicalresearch.org.cn/, identifier MR-31-24-014367.
Collapse
Affiliation(s)
- Shiyu Li
- Clinical Research Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wenxia Zhang
- Department of Clinical Laboratory, Zhoupu Hospital, Shanghai University of Medicine and Health Sciences, Shanghai, China
| | - Sijie Liu
- Department of Pharmacy, The State Administration of Traditional Chinese Medicine (SATCM) Third Grade Laboratory of Traditional Chinese Medicine Preparations, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yichen Zhou
- Clinical Research Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Wei Liu
- Department of Pharmacy, The State Administration of Traditional Chinese Medicine (SATCM) Third Grade Laboratory of Traditional Chinese Medicine Preparations, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Weian Yuan
- Clinical Research Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Min He
- Clinical Research Center, Shuguang Hospital Affiliated to Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
47
|
Wale KR, O’Boyle N, McHugh RE, Serrano E, Mark DR, Douce GR, Connolly JPR, Roe AJ. A master regulator of central carbon metabolism directly activates virulence gene expression in attaching and effacing pathogens. PLoS Pathog 2024; 20:e1012451. [PMID: 39405360 PMCID: PMC11508082 DOI: 10.1371/journal.ppat.1012451] [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: 07/26/2024] [Revised: 10/25/2024] [Accepted: 10/01/2024] [Indexed: 10/26/2024] Open
Abstract
The ability of the attaching and effacing pathogens enterohaemorrhagic Escherichia coli (EHEC) and Citrobacter rodentium to overcome colonisation resistance is reliant on a type 3 secretion system used to intimately attach to the colonic epithelium. This crucial virulence factor is encoded on a pathogenicity island known as the Locus of Enterocyte Effacement (LEE) but its expression is regulated by several core-genome encoded transcription factors. Here, we unveil that the core transcription factor PdhR, traditionally known as a regulator of central metabolism in response to cellular pyruvate levels, is a key activator of the LEE. Through genetic and molecular analyses, we demonstrate that PdhR directly binds to a specific motif within the LEE master regulatory region, thus activating type 3 secretion directly and enhancing host cell adhesion. Deletion of pdhR in EHEC significantly impacted the transcription of hundreds of genes, with pathogenesis and protein secretion emerging as the most affected functional categories. Furthermore, in vivo studies using C. rodentium, a murine model for EHEC infection, revealed that PdhR is essential for effective host colonization and maximal LEE expression within the host. Our findings provide new insights into the complex regulatory networks governing bacterial pathogenesis. This research highlights the intricate relationship between virulence and metabolic processes in attaching and effacing pathogens, demonstrating how core transcriptional regulators can be co-opted to control virulence factor expression in tandem with the cell's essential metabolic circuitry.
Collapse
Affiliation(s)
- Kabo R. Wale
- School of Infection and Immunity, University of Glasgow, Glasgow, United Kingdom
| | - Nicky O’Boyle
- School of Microbiology, University College Cork, Cork, Ireland
- Department of Pathology, School of Medicine, University College Cork, Cork, Ireland
- Department of Microbiology, School of Genetics & Microbiology, Moyne Institute of Preventive Medicine, Trinity College Dublin, Dublin, Ireland
| | - Rebecca E. McHugh
- School of Infection and Immunity, University of Glasgow, Glasgow, United Kingdom
| | - Ester Serrano
- School of Infection and Immunity, University of Glasgow, Glasgow, United Kingdom
| | - David R. Mark
- School of Infection and Immunity, University of Glasgow, Glasgow, United Kingdom
| | - Gillian R. Douce
- School of Infection and Immunity, University of Glasgow, Glasgow, United Kingdom
| | - James P. R. Connolly
- Newcastle University Biosciences Institute, Newcastle University, Newcastle-upon-Tyne, United Kingdom
| | - Andrew J. Roe
- School of Infection and Immunity, University of Glasgow, Glasgow, United Kingdom
| |
Collapse
|
48
|
Cherrak Y, Salazar MA, Näpflin N, Malfertheiner L, Herzog MKM, Schubert C, von Mering C, Hardt WD. Non-canonical start codons confer context-dependent advantages in carbohydrate utilization for commensal E. coli in the murine gut. Nat Microbiol 2024; 9:2696-2709. [PMID: 39160293 PMCID: PMC11445065 DOI: 10.1038/s41564-024-01775-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: 10/02/2023] [Accepted: 07/04/2024] [Indexed: 08/21/2024]
Abstract
Resource competition is a driver of gut microbiota composition. Bacteria can outcompete metabolically similar rivals through the limitation of shared growth-fuelling nutrients. The mechanisms underlying this remain unclear for bacteria with identical sets of metabolic genes. Here we analysed the lactose utilization operon in the murine commensal Escherichia coli 8178. Using in vitro and in vivo approaches, we showed that translation of the lactose utilization repressor gene lacI from its native non-canonical GTG start codon increases the basal expression of the lactose utilization cluster, enhancing adaptation to lactose consumption. Consequently, a strain carrying the wild type lacI GTG start codon outperformed the lacI ATG start codon mutant in the mouse intestine. This advantage was attenuated upon limiting host lactose intake through diet shift or altering the mutant frequency, emphasizing the context-dependent effect of a single nucleotide change on the bacterial fitness of a common member of the gut microbiota. Coupled with a genomic analysis highlighting the selection of non-ATG start codons in sugar utilization regulator genes across the Enterobacteriaceae family, our data exposed an unsuspected function of non-canonical start codons in metabolic competition.
Collapse
Affiliation(s)
- Yassine Cherrak
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland.
| | - Miguel Angel Salazar
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Nicolas Näpflin
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Lukas Malfertheiner
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Mathias K-M Herzog
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Christopher Schubert
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland
| | - Christian von Mering
- Department of Molecular Life Sciences and Swiss Institute of Bioinformatics, University of Zurich, Zurich, Switzerland
| | - Wolf-Dietrich Hardt
- Institute of Microbiology, Department of Biology, ETH Zurich, Zurich, Switzerland.
| |
Collapse
|
49
|
Sansonetti PJ, Doré J. [The human microbiome proofed by the Anthropocene: from correlation to causality and intervention]. Med Sci (Paris) 2024; 40:757-765. [PMID: 39450961 DOI: 10.1051/medsci/2024121] [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: 10/26/2024] Open
Abstract
The deleterious effects of human activities on biodiversity in the vegetal and animal world, and on climate changes are now well-established facts. However, little is yet known on the impact of human activities on microbial diversity on the planet and more specifically on the human microbiota Large implementation of metagenomics allows exaustive microbial cataloguing with broad spatio-temporal resolution of human microbiota. A reduction in bacterial richness and diversity in the human microbiota, particularly in the intestinal tract, is now established and particularly obvious in the most industrialized regions of the planet. Massive, uncontrolled use of antibiotics, drastic changes in traditional food habits and some elements of the "global exposome" that remain to identify are usually considered as stressors accounting for this situation of "missing microbes". As a consequence, a dysbiotic situation develops, a "dysbiosis" being characterized by the erosion of the central core of shared bacterial species across individuals and the development of opportunistic "pathobionts" in response to a weaker barrier capacity of these impoverished microbiota. The current challenge is to establish a causality link between the extension of these dysbiotic situations and the steady emergence of epidemic, non-communicable diseases such as asthma, allergy, obesity, diabetes, autoimmune diseases and some cancers. Experimental animal models combined with controlled, prospective clinical interventions are in demand to consolidate causality links, with the understanding that in the deciphering of the mechanisms of alteration of the human-microbiome symbiosis resides a novel exciting chapter of medicine: "microbial medicine".
Collapse
Affiliation(s)
| | - Joël Doré
- Université Paris-Saclay, INRAE, MetaGenoPolis, AgroParisTech, MICALIS Jouy-en-Josas France
| |
Collapse
|
50
|
Ma Z, Zuo T, Frey N, Rangrez AY. A systematic framework for understanding the microbiome in human health and disease: from basic principles to clinical translation. Signal Transduct Target Ther 2024; 9:237. [PMID: 39307902 PMCID: PMC11418828 DOI: 10.1038/s41392-024-01946-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2024] [Revised: 07/03/2024] [Accepted: 08/01/2024] [Indexed: 09/26/2024] Open
Abstract
The human microbiome is a complex and dynamic system that plays important roles in human health and disease. However, there remain limitations and theoretical gaps in our current understanding of the intricate relationship between microbes and humans. In this narrative review, we integrate the knowledge and insights from various fields, including anatomy, physiology, immunology, histology, genetics, and evolution, to propose a systematic framework. It introduces key concepts such as the 'innate and adaptive genomes', which enhance genetic and evolutionary comprehension of the human genome. The 'germ-free syndrome' challenges the traditional 'microbes as pathogens' view, advocating for the necessity of microbes for health. The 'slave tissue' concept underscores the symbiotic intricacies between human tissues and their microbial counterparts, highlighting the dynamic health implications of microbial interactions. 'Acquired microbial immunity' positions the microbiome as an adjunct to human immune systems, providing a rationale for probiotic therapies and prudent antibiotic use. The 'homeostatic reprogramming hypothesis' integrates the microbiome into the internal environment theory, potentially explaining the change in homeostatic indicators post-industrialization. The 'cell-microbe co-ecology model' elucidates the symbiotic regulation affecting cellular balance, while the 'meta-host model' broadens the host definition to include symbiotic microbes. The 'health-illness conversion model' encapsulates the innate and adaptive genomes' interplay and dysbiosis patterns. The aim here is to provide a more focused and coherent understanding of microbiome and highlight future research avenues that could lead to a more effective and efficient healthcare system.
Collapse
Affiliation(s)
- Ziqi Ma
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| | - Tao Zuo
- Key Laboratory of Human Microbiome and Chronic Diseases (Sun Yat-sen University), Ministry of Education, Guangzhou, China
- Guangdong Institute of Gastroenterology, The Sixth Affiliated Hospital of Sun Yat-sen University, Guangzhou, China
| | - Norbert Frey
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| | - Ashraf Yusuf Rangrez
- Department of Cardiology, Angiology and Pneumology, University Hospital Heidelberg, Heidelberg, Germany.
- DZHK (German Centre for Cardiovascular Research), partner site Heidelberg/Mannheim, Heidelberg, Germany.
| |
Collapse
|