1
|
Olivares C, Ruppé E, Ferreira S, Corbel T, Andremont A, de Gunzburg J, Guedj J, Burdet C. A modelling framework to characterize the impact of antibiotics on the gut microbiota diversity. Gut Microbes 2025; 17:2442523. [PMID: 39711113 DOI: 10.1080/19490976.2024.2442523] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/08/2024] [Accepted: 12/09/2024] [Indexed: 12/24/2024] Open
Abstract
Metagenomic sequencing deepened our knowledge about the role of the intestinal microbiota in human health, and several studies with various methodologies explored its dynamics during antibiotic treatments. We compared the impact of four widely used antibiotics on the gut bacterial diversity. We used plasma and fecal samples collected during and after treatment from healthy volunteers assigned to a 5-day treatment either by ceftriaxone (1 g every 24 h through IV route), ceftazidime/avibactam (2 g/500 mg every 8 h through IV route), piperacillin/tazobactam (1 g/500 mg every 8 h through IV route) or moxifloxacin (400 mg every 24 h through oral route). Antibiotic concentrations were measured in plasma and feces, and bacterial diversity was assessed by the Shannon index from 16S rRNA gene profiling. The relationship between the evolutions of antibiotic fecal exposure and bacterial diversity was modeled using non-linear mixed effects models. We compared the impact of antibiotics on gut microbiota diversity by simulation, using various reconstructed pharmacodynamic indices. Piperacillin/tazobactam was characterized by the highest impact in terms of intensity of perturbation (maximal [IQR] loss of diversity of 27.3% [1.9; 40.0]), while moxifloxacin had the longest duration of perturbation, with a time to return to 95% of baseline value after the last administration of 13.2 d [8.3; 19.1]. Overall, moxifloxacin exhibited the highest global impact, followed by piperacillin/tazobactam, ceftazidime/avibactam and ceftriaxone. Their AUC between day 0 and day 42 of the change of diversity indices from day 0 were, respectively, -13.2 Shannon unit.day [-20.4; -7.9], -10.9 Shannon unit.day [-20.4; -0.6] and -10.1 Shannon unit.day [-18.3; -4.6]. We conclude that antibiotics alter the intestinal diversity to varying degrees, both within and between antibiotics families. Such studies are needed to help antibiotic stewardship in using the antibiotics with the lowest impact on the intestinal microbiota.
Collapse
Affiliation(s)
| | - Etienne Ruppé
- Université Paris Cité, IAME, INSERM, Paris, France
- APHP, Laboratoire de Bactériologie, Hôpital Bichat, Paris, France
| | | | | | | | | | | | | |
Collapse
|
2
|
Wang R, Ma F, Yin D, Wang H, Wei X. Intestinal Microbes, Metabolites, and Hormones in Alcohol-Associated Liver Disease. Semin Liver Dis 2025. [PMID: 40334703 DOI: 10.1055/a-2601-9480] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/09/2025]
Abstract
Alcohol-associated liver disease (ALD)-encompassing conditions including steatosis, fibrosis, cirrhosis, and hepatocellular carcinoma-refers to hepatic damage arising from excessive or hazardous alcohol consumption, and is now recognized as a significant global health burden. Although the mechanisms underlying ALD remain incompletely understood, several pathways have been substantiated over the last five decades, notably the involvement of intestinal microorganisms and the involvement of the gut-liver axis in alcohol metabolism and ALD pathogenesis. Ethanol intake disrupts the intestinal microbial balance and compromises the gut barrier, resulting in increased permeability to microbial products. The subsequent translocation of microbial metabolites and other antigenic substances to the liver activates hepatic immune responses, thereby contributing to liver injury. In addition, gastrointestinal hormones are also implicated in ALD progression through various mechanisms. Although no therapies for ALD have been approved by the Food and Drug Administration, various therapeutic strategies targeting the intestinal microbiota and gut barrier have been identified. In conclusion, this review discusses the role of the gut-liver axis in alcohol metabolism and ALD pathogenesis and explores the emerging therapeutic strategies.
Collapse
Affiliation(s)
- Ruimeng Wang
- Second Clinical Medical College, Anhui Medical University, Hefei, China
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Fang Ma
- Center for Scientific Research of Anhui Medical University, Anhui Medical University, Hefei, China
| | - Dou Yin
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Hua Wang
- Department of Oncology, the First Affiliated Hospital of Anhui Medical University, Hefei, China
- Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Medical University, Hefei, China
| | - Xiaohui Wei
- Department of Pathophysiology, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| |
Collapse
|
3
|
Ha YJ, Hwang SJ, Hong S, Kim J, Kim M, Lee GS, Lee SH, Kim H, Lee BS, In An H, Kim KH, Lee W, Lee HJ, Kim CS. Erythromycin Stress Upregulates Antiangiogenic Metabolites in the Gut Bacterium Aneurinibacillus aneurinilyticus. J Am Chem Soc 2025; 147:16459-16470. [PMID: 40305844 DOI: 10.1021/jacs.5c03174] [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: 05/02/2025]
Abstract
The interplay among antibiotics, gut microbiota, and disease pathogenesis remains poorly understood, particularly in the context of rare gut bacteria. This study identifies a novel correlation between erythromycin-induced stress and the production of antiangiogenic metabolites in Aneurinibacillus aneurinilyticus, a human gut bacterium. We report the isolation and structural characterization of aneuristatin (1), a metabolite featuring a unique pyrrolo[1,2-a]pyrazine scaffold, along with seven structurally related metabolites (2-8) from A. aneurinilyticus ATCC 12856T. These metabolites were upregulated via the erythromycin-induced activation of the arnA biosynthetic gene. Aneuristatin (1) enhanced prolyl hydroxylase activity, promoting hypoxia-inducible factor-1α (HIF-1α) degradation and reducing downstream targets, including VEGF and EPO. It also exhibited antioxidant effects by reducing ROS levels under hypoxia. Additionally, it inhibited angiogenesis in HUVECs and zebrafish and effectively reduced inflammation, fibrosis, and angiogenesis in a mouse corneal injury model. Our study establishes a molecular basis for the potential of erythromycin-induced aneuristatin (1) to prevent or treat angiogenesis-related diseases such as cancer.
Collapse
Affiliation(s)
- Young Jun Ha
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Su Jung Hwang
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Subin Hong
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Jonghwan Kim
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Minji Kim
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Gyu Sung Lee
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Seung Hwan Lee
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hyemin Kim
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Bum Soo Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hye In An
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Ki Hyun Kim
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Wonsik Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Hyo-Jong Lee
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| | - Chung Sub Kim
- Department of Biopharmaceutical Convergence, Sungkyunkwan University, Suwon 16419, Republic of Korea
- School of Pharmacy, Sungkyunkwan University, Suwon 16419, Republic of Korea
| |
Collapse
|
4
|
Kim SJ, Chung HC, Park SY, Lee JM, Han JH. Beneficial effects of probiotics on dysbiosis of gut microbiota induced by antibiotic treatment in healthy dogs. Res Vet Sci 2025; 191:105674. [PMID: 40347600 DOI: 10.1016/j.rvsc.2025.105674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Revised: 04/16/2025] [Accepted: 05/01/2025] [Indexed: 05/14/2025]
Abstract
The gut microbiota plays a crucial role in maintaining host health. While numerous studies have explored the impact of antibiotics on the gut microbiota in humans, limited research has examined how antibiotics affect the gut microbiome in dogs. This study investigated the effects of antibiotic treatment on the gut microbiota of dogs and assessed whether probiotic supplementation could prevent antibiotic-induced dysbiosis. Fourteen healthy young dogs undergoing castration were included in the study. All dogs received a single injection of cefovecin immediately after surgery. The probiotics group (7 dogs) was given a probiotic complex daily starting on the day of surgery and continuing for two weeks, while the non-probiotics group (7 dogs) received no supplementation. Fecal samples were collected on the day of surgery and two weeks later during the follow-up visit for suture removal for microbiome analysis. In microbial diversity analysis, α-diversity was significantly higher in the probiotic-supplemented group compared to the non-probiotics group (p < 0.05). β-diversity analysis revealed significant differences in microbial community composition in the non-probiotics group after antibiotic treatment (p < 0.05), while no significant differences were observed in the probiotics group. Relative abundance analysis indicated that Clostridioides, a marker of antibiotic-induced dysbiosis, significantly increased in dogs without probiotics after antibiotic treatment (p < 0.05). In contrast, Butyricicoccus, a butyrate-producing bacterium with gut health benefits, was significantly enriched in the probiotics group (p < 0.05). These findings suggest that probiotic supplementation supports healthier gut microbiome recovery following antibiotic treatment and highlights its potential to enhance gut microbiota restoration and mitigate gut dysbiosis caused by antibiotics.
Collapse
Affiliation(s)
- Sung-Jae Kim
- Department of Companion Animal Health, Kyungbok University, Namyangju 12051, South Korea
| | - Hee-Chun Chung
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, South Korea
| | - Soo-Yeon Park
- Department of Companion Animal Health, Seojeong University, Yangju 11429, South Korea
| | - Jae-Myun Lee
- Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul 03722, South Korea; Department of Microbiology and Immunology, Institute for Immunology and Immunological Diseases, Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, South Korea.
| | - Jeong-Hee Han
- College of Veterinary Medicine and Institute of Veterinary Science, Kangwon National University, Chuncheon 24341, South Korea.
| |
Collapse
|
5
|
Fletcher SJ, Ching C, Suprenant MP, Desai D, Zaman MH. Agent-based model of the human colon to investigate mechanisms of pathogen colonization resistance. Biophys J 2025; 124:1446-1461. [PMID: 40156186 DOI: 10.1016/j.bpj.2025.03.022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2024] [Revised: 01/31/2025] [Accepted: 03/24/2025] [Indexed: 04/01/2025] Open
Abstract
Recent global burden of disease studies have shown that bacterial infections are responsible for over 13 million deaths worldwide, or 1 in every 8 deaths, each year. Enteric diarrheal infections, in particular, pose a significant challenge and strain on healthcare systems as many are difficult to address pharmaceutically, and thus rely primarily on the patient's own immune system and gut microbiome to fight the infection. Nonetheless, the specific mechanisms behind gut microbiome colonization resistance of enteric pathogens are not well defined and microbiome diversity is difficult to represent and study experimentally. To address this gap, we have constructed an agent-based computational model of the colonic epithelium cross section to investigate the colonic invasion of enteric pathogens. The model focuses on three main regions: epithelial layer, mucosal bilayer, and adjacent lumen, and utilizes four main cell types as agents: anaerobic bacteria, facultative anaerobic bacteria, human goblet cells, and pathogens. Utilizing this model, we are able to describe the healthy microbiome cell localization and dynamics from our mucosal bilayer. In addition, we are also able to investigate the impact of host dietary fiber consumption and simulate pathogen invasion. The model exemplifies the possibility and potential to explore key gut microbiome colonization resistance mechanisms and environmental impacts on the gut microbiome using computational methods.
Collapse
Affiliation(s)
| | - Carly Ching
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | | | - Darash Desai
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts
| | - Muhammad Hamid Zaman
- Department of Biomedical Engineering, Boston University, Boston, Massachusetts; Howard Hughes Medical Institute, Boston University, Boston, Massachusetts; Center on Forced Displacement, Boston University, Boston, Massachusetts.
| |
Collapse
|
6
|
Fang Y, Wu B, Zhang R, Chen X, Jiang F, Jin Q, Jin T, Huang S, Tao C, Qiang M, Piao Y, Hua Y, Feng X, Cao C. Effects of Antibiotics on First-line Immunotherapy in Patients With Recurrent or Metastatic Nasopharyngeal Carcinoma. J Immunother 2025:00002371-990000000-00137. [PMID: 40223355 DOI: 10.1097/cji.0000000000000556] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2025] [Accepted: 03/06/2025] [Indexed: 04/15/2025]
Abstract
Immunotherapy combined with chemotherapy has become the first-line treatment for recurrent or metastatic nasopharyngeal carcinoma (RM-NPC). However, the impact of antibiotic (ATB) use on the efficacy of immunotherapy in RM-NPC remains unclear. A total of 200 patients with RM-NPC who started first-line immunotherapy between October 2021 and September 2023 were included. Forty-six patients received ATB within 60 days before and 42 days after the first infusion of immunotherapy (group ATB+), and the remaining 154 patients were in group ATB-. The median progression-free survival (PFS) times of the ATB+ and ATB- groups were 11.20 and 19.87 months, respectively (P = 0.061). The 2-year overall survival (OS) rates of the ATB+ and ATB- groups were 52.6% and 76.7%, respectively (P = 0.001). In multivariate analysis, ATB use was significantly associated with worse OS (hazard ratio = 2.549, P = 0.002). No significant differences were observed between the 2 groups in terms of grade 3+ treatment-related adverse events. ATB use in RM-NPC may reduce the effectiveness of first-line immunotherapy.
Collapse
Affiliation(s)
- Yuting Fang
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Postgraduate Training Base Alliance of Wenzhou Medical University, Zhejiang Cancer Hospital, Hangzhou, China
| | - Binhao Wu
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Postgraduate Training Base Alliance of Wenzhou Medical University, Zhejiang Cancer Hospital, Hangzhou, China
| | - Rong Zhang
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Postgraduate Training Base Alliance of Wenzhou Medical University, Zhejiang Cancer Hospital, Hangzhou, China
| | - Xiaozhong Chen
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Postgraduate Training Base Alliance of Wenzhou Medical University, Zhejiang Cancer Hospital, Hangzhou, China
| | - Feng Jiang
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Postgraduate Training Base Alliance of Wenzhou Medical University, Zhejiang Cancer Hospital, Hangzhou, China
| | - Qifeng Jin
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Postgraduate Training Base Alliance of Wenzhou Medical University, Zhejiang Cancer Hospital, Hangzhou, China
| | - Ting Jin
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Postgraduate Training Base Alliance of Wenzhou Medical University, Zhejiang Cancer Hospital, Hangzhou, China
| | - Shuang Huang
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Postgraduate Training Base Alliance of Wenzhou Medical University, Zhejiang Cancer Hospital, Hangzhou, China
| | - Changjuan Tao
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Postgraduate Training Base Alliance of Wenzhou Medical University, Zhejiang Cancer Hospital, Hangzhou, China
| | - Mengyun Qiang
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Postgraduate Training Base Alliance of Wenzhou Medical University, Zhejiang Cancer Hospital, Hangzhou, China
| | - Yongfeng Piao
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Postgraduate Training Base Alliance of Wenzhou Medical University, Zhejiang Cancer Hospital, Hangzhou, China
| | - Yonghong Hua
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Postgraduate Training Base Alliance of Wenzhou Medical University, Zhejiang Cancer Hospital, Hangzhou, China
| | - Xinglai Feng
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Postgraduate Training Base Alliance of Wenzhou Medical University, Zhejiang Cancer Hospital, Hangzhou, China
| | - Caineng Cao
- Department of Radiation Oncology, Zhejiang Cancer Hospital, Hangzhou Institute of Medicine (HIM), Chinese Academy of Sciences, Hangzhou, China
- Key Laboratory of Head and Neck Cancer Translational Research of Zhejiang Province, Postgraduate Training Base Alliance of Wenzhou Medical University, Zhejiang Cancer Hospital, Hangzhou, China
| |
Collapse
|
7
|
Zheng Z, Jin W, Guo W, Jin Z, Zuo Y. Oral Fusobacterium nucleatum exacerbates ulcerative colitis via the oral-gut axis: mechanisms and therapeutic implications. Front Cell Infect Microbiol 2025; 15:1564169. [PMID: 40260115 PMCID: PMC12009839 DOI: 10.3389/fcimb.2025.1564169] [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: 01/21/2025] [Accepted: 03/18/2025] [Indexed: 04/23/2025] Open
Abstract
Background Fusobacterium nucleatum (F. nucleatum) is an anaerobic bacterium known for its association with periodontal disease and oral infections. It has been implicated in the development of gastrointestinal diseases such as inflammatory bowel disease and colorectal cancer. Ulcerative colitis (UC), which is characterized by chronic inflammation of the colon, is a condition of unknown etiology with a rising incidence rate, significantly affecting the quality of life for patients. The increased intestinal permeability during UC may facilitate the adherence or invasion of F. nucleatum into the damaged intestinal barrier, leading to exacerbated inflammation. Methods This article introduces the concept of the oral-gut axis, reviewing existing literature to analyze the role of F. nucleatum in the pathogenesis of UC and exploring its potential pathogenic mechanisms. It also summarizes the latest advances in treating patients with UC who have F. nucleatum and looks forward to prospective therapeutic strategies and the translational prospects of F. nucleatum within the oral-gut axis. Results F. nucleatum may be a key player in the pathogenesis of UC, likely due to its invasiveness during periods of increased intestinal permeability. The paper also discusses innovative approaches for the prevention and management of UC exacerbated by F. nucleatum, paving the way for more effective treatment of UC. Conclusion The review offers new insights into the complex relationship between the oral microbiome and intestinal diseases, enhancing our understanding of their dynamic interactions. There is a paucity of literature on therapeutic approaches, indicating a need for further clinical research.
Collapse
Affiliation(s)
- Zhaoyu Zheng
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Wenqin Jin
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Weiwei Guo
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Zhao Jin
- Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Yuling Zuo
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, China
| |
Collapse
|
8
|
Shahzad M, Arshad M, Ahmad HA, Iddrissu I, Bailey EH, Dru N, Khan S, Khan H, Andrews SC. Nutritional status reshapes gut microbiota composition in adolescent Afghan refugees in Peshawar, Pakistan. Nutr Res 2025; 138:55-67. [PMID: 40311534 DOI: 10.1016/j.nutres.2025.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Revised: 04/03/2025] [Accepted: 04/03/2025] [Indexed: 05/03/2025]
Abstract
Although the human gut microbiome, and its role in health and disease, have been extensively studied in different populations, a comprehensive assessment of gut microbiome composition has not been performed in vulnerable refugee populations. In this study, we hypothesized that overall nutritional status, as indicated by serum micronutrients concentrations, is an important driver of variations in gut microbiome composition. Therefore, gut-microbiome diversity and associated demographic, health and nutritional factors were assessed in adolescent Afghan refugees (n=206). Blood and faecal samples were collected and analysed for nutrition status markers and 16S rRNA gene amplicon-based community profiling, respectively. Bioinformatics and statistical analysis were performed using SPSS, QIIME and R. Overall, 56 distinct phyla, 117 families and 252 genera were identified in the faecal samples. Bacterial diversity (alpha and beta diversity) and the Firmicutes:Bacteroidetes (F/B) ratio were significantly higher in the 15 to 19 year old age group (cf. the 10-14 age group) but were lower in the underweight and vitamin D deficient groups. Furthermore, LEfSe analysis identified significant differences in the relative abundance of bacterial genera based on age, BMI and micronutrient (vitamins and minerals) status. These results were further scrutinised by correlation analysis which confirmed that age, BMI and micronutrient status show significant correlations with F/B ratio and the relative abundance of specific bacterial taxa. Collectively, our study provides the first indication of how the gut-microbiota profile of adolescent Afghan refugees is associated with a range of nutrition-status factors. These findings can thus provide a basis for translational microbiota research aimed at improving the health of such understudied and vulnerable populations.
Collapse
Affiliation(s)
- Muhammad Shahzad
- Faculty of Dentistry, Zarqa University, Zarqa, Jordan; Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Muhammad Arshad
- Center for Genomics and Systems Biology, New York University, Abu Dhabi, United Arab Emirates
| | - Habab Ali Ahmad
- Department of Biological and Health Sciences, Pak-Austria Fachhochschule Institute of Applied Science and Technology (PAF-IAST), Haripur, Pakistan
| | - Ishawu Iddrissu
- School of Biological Sciences, Health and Life Sciences Building, University of Reading, Reading, United Kingdom
| | - Elizabeth H Bailey
- School of Biosciences, Sutton Bonington Campus, University of Nottingham, Loughborough, Leicestershire, United Kingdom
| | - Nizar Dru
- Center for Genomics and Systems Biology, New York University, Abu Dhabi, United Arab Emirates
| | - Shabir Khan
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Haris Khan
- Institute of Basic Medical Sciences, Khyber Medical University, Peshawar, Pakistan
| | - Simon C Andrews
- School of Biological Sciences, Health and Life Sciences Building, University of Reading, Reading, United Kingdom.
| |
Collapse
|
9
|
Smibert OC, Vogrin S, Sinclair M, Majumdar A, Nasra M, Pandey D, Jahanabadi H, Trubiano JA, Markey KA, Slavin MA, Testro A, Kwong JC. Antibiotic Exposure and Risk of Allograft Rejection and Survival After Liver Transplant: An Observational Cohort Study From a Tertiary Referral Centre. Transpl Infect Dis 2025:e70026. [PMID: 40153422 DOI: 10.1111/tid.70026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2025] [Revised: 03/07/2025] [Accepted: 03/12/2025] [Indexed: 03/30/2025]
Abstract
INTRODUCTION Our goal is to understand whether there is an association between Abx exposure-and the inferred downstream damage to the intestinal microbiome-and the key patient outcomes of overall survival and rejection following liver transplant. METHODS We conducted a retrospective cohort study of 462 liver transplant recipients treated at a multistate liver transplant (LTx) service during a 7-year period. The association between antibiotic exposure and outcome was tested across models that addressed antibiotic spectrum, duration, and timing relative to transplant. Cox proportional hazard regression was used to evaluate the relationship between antibiotics with survival and rejection. RESULTS The observed 1-year survival in this cohort was 95% (95% CI: 93%, 97%), and 20.8% of patients (96/462) experienced rejection at 1 year. In multivariable analyses, exposure to anaerobe-targeting antibiotics for longer than 14 days pretransplant (p = 0.055) or posttransplant (p = 0.040) was significantly associated with reduced 1-year survival. In multivariable analyses, exposure to any anaerobe-targeting Abx posttransplant was significantly associated with an increased risk of rejection (p = 0.001). CONCLUSIONS Exposure to anaerobic spectrum antibiotics either before or after LTx was associated with poor outcomes during the first year posttransplant and provides an impetus to further characterize the relationship between antibiotic use, microbiota disruption, and cellular immunity in liver transplantation.
Collapse
Affiliation(s)
- Olivia C Smibert
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Infectious Diseases and Immunology, Austin Health, Melbourne, Victoria, Australia
- Department of Infectious Diseases, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Sara Vogrin
- Department of Medicine, St Vincent's Hospital, Victoria Parade, University of Melbourne, Fitzroy, Australia
| | - Marie Sinclair
- Liver Transplant Unit, Austin Health, Melbourne, Victoria, Australia
- School of Medicine, Dentistry and Health Sciences, the University of Melbourne, Melbourne, Victoria, Australia
| | - Avik Majumdar
- Liver Transplant Unit, Austin Health, Melbourne, Victoria, Australia
- School of Medicine, Dentistry and Health Sciences, the University of Melbourne, Melbourne, Victoria, Australia
| | - Mohamed Nasra
- Department of Infectious Diseases and Immunology, Austin Health, Melbourne, Victoria, Australia
| | - Dinesh Pandey
- Data Analytics Research and Evaluation (DARE) Centre, The University of Melbourne and Austin Hospital, Melbourne, Victoria, Australia
- Clinical Analytics and Reporting, Performance Reporting and Decision Support, Austin Health, Melbourne, Victoria, Australia
| | - Hossein Jahanabadi
- Data Analytics Research and Evaluation (DARE) Centre, The University of Melbourne and Austin Hospital, Melbourne, Victoria, Australia
- Clinical Analytics and Reporting, Performance Reporting and Decision Support, Austin Health, Melbourne, Victoria, Australia
| | - Jason A Trubiano
- National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Infectious Diseases and Immunology, Austin Health, Melbourne, Victoria, Australia
- Department of Infectious Diseases, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Kate A Markey
- Translational Science and Therapeutics Division, Fred Hutchinson Cancer Center (FHCC), Seattle, Washington, USA
- Department of Medicine, University of Washington, Seattle, Washington, USA
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, New York, USA
- Weill Cornell Medical College, New York, New York, USA
| | - Monica A Slavin
- Sir Peter MacCallum Department of Oncology, University of Melbourne, Parkville, Victoria, Australia
- Department of Infectious Diseases, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- National Centre for Infections in Cancer, Peter MacCallum Cancer Centre, Melbourne, Victoria, Australia
- Department of Infectious Diseases, University of Melbourne, The Peter Doherty Institute for Infection and Immunity, Melbourne, Victoria, Australia
| | - Adam Testro
- Liver Transplant Unit, Austin Health, Melbourne, Victoria, Australia
- School of Medicine, Dentistry and Health Sciences, the University of Melbourne, Melbourne, Victoria, Australia
| | - Jason C Kwong
- Department of Infectious Diseases and Immunology, Austin Health, Melbourne, Victoria, Australia
- Department of Microbiology & Immunology, The Peter Doherty Institute for Infection and Immunity, the University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
10
|
Yang SY, Han SM, Lee JY, Kim KS, Lee JE, Lee DW. Advancing Gut Microbiome Research: The Shift from Metagenomics to Multi-Omics and Future Perspectives. J Microbiol Biotechnol 2025; 35:e2412001. [PMID: 40223273 PMCID: PMC12010094 DOI: 10.4014/jmb.2412.12001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2024] [Revised: 02/14/2025] [Accepted: 02/24/2025] [Indexed: 04/15/2025]
Abstract
The gut microbiome, a dynamic and integral component of human health, has co-evolved with its host, playing essential roles in metabolism, immunity, and disease prevention. Traditional microbiome studies, primarily focused on microbial composition, have provided limited insights into the functional and mechanistic interactions between microbiota and their host. The advent of multi-omics technologies has transformed microbiome research by integrating genomics, transcriptomics, proteomics, and metabolomics, offering a comprehensive, systems-level understanding of microbial ecology and host-microbiome interactions. These advances have propelled innovations in personalized medicine, enabling more precise diagnostics and targeted therapeutic strategies. This review highlights recent breakthroughs in microbiome research, demonstrating how these approaches have elucidated microbial functions and their implications for health and disease. Additionally, it underscores the necessity of standardizing multi-omics methodologies, conducting large-scale cohort studies, and developing novel platforms for mechanistic studies, which are critical steps toward translating microbiome research into clinical applications and advancing precision medicine.
Collapse
Affiliation(s)
- So-Yeon Yang
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Seung Min Han
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Ji-Young Lee
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Kyoung Su Kim
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Jae-Eun Lee
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| | - Dong-Woo Lee
- Department of Biotechnology, Yonsei University, Seoul 03722, Republic of Korea
| |
Collapse
|
11
|
Lewis N, Villani A, Lagopoulos J. Gut dysbiosis as a driver of neuroinflammation in attention-deficit/hyperactivity disorder: A review of current evidence. Neuroscience 2025; 569:298-321. [PMID: 39848564 DOI: 10.1016/j.neuroscience.2025.01.031] [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: 07/11/2024] [Revised: 01/12/2025] [Accepted: 01/13/2025] [Indexed: 01/25/2025]
Abstract
There is mounting evidence for the involvement of the immune system, neuroinflammation and disturbed gut microbiota, or dysbiosis, in attention-deficit/hyperactivity disorder (ADHD). Gut dysbiosis is strongly implicated in many physical, autoimmune, neurological, and neuropsychiatric conditions, however knowledge of its particular pathogenic role in ADHD is sparse. As such, this narrative review examines and synthesizes the available evidence related to inflammation, dysbiosis, and neural processes in ADHD. Minimal differences in microbiota diversity measures between cases and controls were found, however many relative abundance differences were observed at all classification levels (phylum to strain). Compositional differences of taxa important to key gut-brain axis pathways, in particular Bacteroides species and Faecalibacterium, may contribute to inflammation, brain functioning differences, and symptoms, in ADHD. We have identified one possible model of ADHD etiopathogenesis involving systemic inflammation, an impaired blood-brain barrier, and neural disturbances as downstream consequences of gut dysbiosis. Nevertheless, studies conducted to date have varied degrees of methodological rigour and involve diverse participant characteristics and analytical techniques, highlighting a need for additional research.
Collapse
Affiliation(s)
- Naomi Lewis
- School of Health, University of the Sunshine Coast, 90 Sippy Downs Dr, Sippy Downs, QLD 4556, Australia; Thompson Institute, University of the Sunshine Coast, 12 Innovation Pkwy, Birtinya, QLD 4575, Australia.
| | - Anthony Villani
- School of Health, University of the Sunshine Coast, 90 Sippy Downs Dr, Sippy Downs, QLD 4556, Australia.
| | - Jim Lagopoulos
- Thompson Brain and Mind Healthcare, Eccles Blvd, Birtinya, QLD 4575, Australia.
| |
Collapse
|
12
|
Li S, Liu J, Zhang X, Gu Q, Wu Y, Tao X, Tian T, Pan G, Chu M. The Potential Impact of Antibiotic Exposure on the Microbiome and Human Health. Microorganisms 2025; 13:602. [PMID: 40142495 PMCID: PMC11944296 DOI: 10.3390/microorganisms13030602] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 02/21/2025] [Accepted: 02/28/2025] [Indexed: 03/28/2025] Open
Abstract
Antibiotics are a cornerstone of modern medicine, saving countless lives. However, their widespread use presents two major challenges. First, antibiotic-induced changes in the microbiome can disrupt immune function, increasing the susceptibility to diseases associated with these alterations. Second, prolonged antibiotic use fosters the proliferation of antibiotic resistance genes, leading to the emergence of resistant strains and threatening our ability to control infections. These challenges highlight an urgent global health crisis, necessitating in-depth investigation into the multifaceted effects of antibiotic exposure on microbiome dynamics and human health. In this review, we explore the potential effects of antibiotic exposure on the microbiome and its implications for overall health. Additionally, we examine the role of emerging technologies in addressing these challenges and in shaping future antibiotic development. Our goal is to provide insights that will inform more effective public health strategies and interventions aimed at mitigating the adverse consequences of antibiotic use, restoring microbial balance, and improving overall health outcomes.
Collapse
Affiliation(s)
- Siqi Li
- Department of Epidemiology, School of Public Health, Nantong University, 9 Seyuan Road, Nantong 226019, China; (S.L.); (J.L.); (Q.G.); (Y.W.); (X.T.); (T.T.)
| | - Jiahao Liu
- Department of Epidemiology, School of Public Health, Nantong University, 9 Seyuan Road, Nantong 226019, China; (S.L.); (J.L.); (Q.G.); (Y.W.); (X.T.); (T.T.)
| | - Xinyang Zhang
- School of Medical, Nantong University, Nantong 226019, China;
| | - Qihong Gu
- Department of Epidemiology, School of Public Health, Nantong University, 9 Seyuan Road, Nantong 226019, China; (S.L.); (J.L.); (Q.G.); (Y.W.); (X.T.); (T.T.)
| | - Yutong Wu
- Department of Epidemiology, School of Public Health, Nantong University, 9 Seyuan Road, Nantong 226019, China; (S.L.); (J.L.); (Q.G.); (Y.W.); (X.T.); (T.T.)
| | - Xiaobo Tao
- Department of Epidemiology, School of Public Health, Nantong University, 9 Seyuan Road, Nantong 226019, China; (S.L.); (J.L.); (Q.G.); (Y.W.); (X.T.); (T.T.)
| | - Tian Tian
- Department of Epidemiology, School of Public Health, Nantong University, 9 Seyuan Road, Nantong 226019, China; (S.L.); (J.L.); (Q.G.); (Y.W.); (X.T.); (T.T.)
| | - Gongbu Pan
- Department of Epidemiology, School of Public Health, Nantong University, 9 Seyuan Road, Nantong 226019, China; (S.L.); (J.L.); (Q.G.); (Y.W.); (X.T.); (T.T.)
- Wicking Dementia Research and Education Centre, University of Tasmania, Hobart, TAS 7005, Australia
| | - Minjie Chu
- Department of Epidemiology, School of Public Health, Nantong University, 9 Seyuan Road, Nantong 226019, China; (S.L.); (J.L.); (Q.G.); (Y.W.); (X.T.); (T.T.)
| |
Collapse
|
13
|
Safarchi A, Al-Qadami G, Tran CD, Conlon M. Understanding dysbiosis and resilience in the human gut microbiome: biomarkers, interventions, and challenges. Front Microbiol 2025; 16:1559521. [PMID: 40104586 PMCID: PMC11913848 DOI: 10.3389/fmicb.2025.1559521] [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: 01/13/2025] [Accepted: 02/19/2025] [Indexed: 03/20/2025] Open
Abstract
The healthy gut microbiome is important in maintaining health and preventing various chronic and metabolic diseases through interactions with the host via different gut-organ axes, such as the gut-brain, gut-liver, gut-immune, and gut-lung axes. The human gut microbiome is relatively stable, yet can be influenced by numerous factors, such as diet, infections, chronic diseases, and medications which may disrupt its composition and function. Therefore, microbial resilience is suggested as one of the key characteristics of a healthy gut microbiome in humans. However, our understanding of its definition and indicators remains unclear due to insufficient experimental data. Here, we review the impact of key drivers including intrinsic and extrinsic factors such as diet and antibiotics on the human gut microbiome. Additionally, we discuss the concept of a resilient gut microbiome and highlight potential biomarkers including diversity indices and some bacterial taxa as recovery-associated bacteria, resistance genes, antimicrobial peptides, and functional flexibility. These biomarkers can facilitate the identification and prediction of healthy and resilient microbiomes, particularly in precision medicine, through diagnostic tools or machine learning approaches especially after antimicrobial medications that may cause stable dysbiosis. Furthermore, we review current nutrition intervention strategies to maximize microbial resilience, the challenges in investigating microbiome resilience, and future directions in this field of research.
Collapse
Affiliation(s)
- Azadeh Safarchi
- Microbiome for One Systems Health FSP, CSIRO, Westmead, NSW, Australia
- Health and Biosecurity Research Unit, CSIRO, Adelaide, SA, Australia
| | - Ghanyah Al-Qadami
- Microbiome for One Systems Health FSP, CSIRO, Westmead, NSW, Australia
- Health and Biosecurity Research Unit, CSIRO, Adelaide, SA, Australia
| | - Cuong D Tran
- Health and Biosecurity Research Unit, CSIRO, Adelaide, SA, Australia
| | - Michael Conlon
- Health and Biosecurity Research Unit, CSIRO, Adelaide, SA, Australia
| |
Collapse
|
14
|
Jena PK, Arditi M, Rivas MN. Gut Microbiota Alterations in Patients With Kawasaki Disease. Arterioscler Thromb Vasc Biol 2025; 45:345-358. [PMID: 39846163 PMCID: PMC11998981 DOI: 10.1161/atvbaha.124.321201] [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: 10/14/2024] [Revised: 12/09/2024] [Accepted: 01/09/2025] [Indexed: 01/24/2025]
Abstract
The intestinal microbiota influences many host biological processes, including metabolism, intestinal barrier functions, and immune responses in the gut and distant organs. Alterations in its composition have been associated with the development of inflammatory disorders and cardiovascular diseases, including Kawasaki disease (KD). KD is an acute pediatric vasculitis of unknown etiology and the leading cause of acquired heart disease in children in the United States. The presence of gastrointestinal symptoms in the acute phase of KD has been associated with an increased risk of treatment resistance and the development of coronary artery aneurysms. Studies report alterations in fecal bacterial communities of patients with KD, characterized by the blooming of pathogenic bacteria and decreased relative abundance of short-chain fatty acid-producing bacteria. However, causality and functionality cannot be established from these observational patient cohorts of KD. This highlights the need for more advanced and rigorous studies to establish causality and functionality in both experimental models of KD vasculitis and patient cohorts. Here, we review the evidence linking an altered gut microbiota composition to the development of KD, assess the potential mechanisms involved in this process, and discuss the potential therapeutic value of these observations.
Collapse
Affiliation(s)
- Prasant K. Jena
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children’s, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Moshe Arditi
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children’s, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Smidt Heart Institute, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| | - Magali Noval Rivas
- Department of Pediatrics, Division of Pediatric Infectious Diseases, Guerin Children’s, Cedars-Sinai Medical Center, Los Angeles, CA, USA
- Infectious and Immunologic Diseases Research Center (IIDRC), Department of Biomedical Sciences, Cedars-Sinai Medical Center, Los Angeles, CA, USA
| |
Collapse
|
15
|
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
|
16
|
Amato KR, Lake BR, Ozminkowski S, Jiang H, Moy M, Sardaro MLS, Fultz A, Hopper LM. Exploring the Utility of the Gut Microbiome as a Longitudinal Health Monitoring Tool in Sanctuary Chimpanzees (Pan troglodytes). Am J Primatol 2025; 87:e70004. [PMID: 40089976 PMCID: PMC11910989 DOI: 10.1002/ajp.70004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 11/20/2024] [Accepted: 01/03/2025] [Indexed: 03/18/2025]
Abstract
The primary goal of captive primate management is to ensure optimal health and welfare of the animals in our care. Given that the gut microbiome interacts closely with host metabolism, immunity, and even cognition, it represents a potentially powerful tool for identifying subtle changes in health status across a range of body systems simultaneously. However, thus far, it has not been widely tested or implemented as a monitoring tool. In this study, we used longitudinal microbiome sampling of newly arrived chimpanzees at Chimp Haven to explore the feasibility of using the gut microbiome as a health and welfare biomarker in a sanctuary environment. We also tested the hypothesis that a transition to a new living environment, and integration into new social groupings, would result in temporal changes in chimpanzee gut microbiome composition. The collection of longitudinal microbiome data at Chimp Haven was feasible, and it revealed temporal shifts that were unique to each individual and, in some cases, correlated to other known impacts on health and behavior. We found limited evidence for microbial change over time after arrival at Chimp Haven that was consistent across individuals. In contrast, social group and enclosure, and to a lesser extent, age and sex, were associated with differences in gut microbiome composition. Microbiome composition was also associated with overall health status categories. However, many of the effects we detected were most apparent when using longitudinal data, as opposed to single time point samples. Additionally, we found important effects of technical factors, specifically outdoor temperature and time to collection, on our data. Overall, we demonstrate that the gut microbiome has the potential to be effectively deployed as a tool for health and environmental monitoring in a population of sanctuary chimpanzees, but the design must be carefully considered. We encourage other institutions to apply these approaches and integrate health and physiology data to build on the utility of gut microbiome analysis for ensuring the welfare of captive primates in a range of contexts.
Collapse
Affiliation(s)
| | - Benjamin R. Lake
- Chimp HavenKeithvilleLouisianaUSA
- Ecology & Evolutionary Biology ProgramTexas A&M UniversityCollege StationTexasUSA
| | - Samuel Ozminkowski
- Department of Statistics and Data ScienceNorthwestern UniversityEvanstonIllinoisUSA
| | - Hongmei Jiang
- Department of Statistics and Data ScienceNorthwestern UniversityEvanstonIllinoisUSA
| | - Madelyn Moy
- Department of AnthropologyNorthwestern UniversityEvanstonIllinoisUSA
| | - Maria Luisa Savo Sardaro
- Department of AnthropologyNorthwestern UniversityEvanstonIllinoisUSA
- Department of Human Science and Promotion of the Quality of LifeUniversity of San RaffaeleRomeItaly
| | | | - Lydia M. Hopper
- Lester E. Fisher Center for the Study and Conservation of Apes, Lincoln Park ZooChicagoIllinoisUSA
- Department of Molecular and Comparative PathobiologyJohns Hopkins University School of MedicineBaltimoreMarylandUSA
| |
Collapse
|
17
|
Tsouprou M, Koumpagioti D, Botsa E, Douros K, Moriki D. Utilization of Inhaled Antibiotics in Pediatric Non-Cystic Fibrosis Bronchiectasis: A Comprehensive Review. Antibiotics (Basel) 2025; 14:165. [PMID: 40001409 PMCID: PMC11851904 DOI: 10.3390/antibiotics14020165] [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: 12/30/2024] [Revised: 01/25/2025] [Accepted: 02/05/2025] [Indexed: 02/27/2025] Open
Abstract
The lack of available treatments in pediatric non-cystic fibrosis (non-CF) bronchiectasis is a major concern, especially in the context of the increasing disease burden due to better detection rates with advanced imaging techniques. Recurrent infections in these patients are the main cause of deterioration, leading to impaired lung function and increasing the risk of morbidity and mortality. Since pediatric non-CF bronchiectasis with early recognition and appropriate treatment can be reversible, optimal management is an issue of growing significance. The use of inhaled antibiotics as a treatment option, although a standard of care for CF patients, has been poorly studied in patients with non-CF bronchiectasis, especially in children. In this review, we present the current data on the potential use of inhaled antibiotics in the treatment of non-CF bronchiectasis and assess their safety and efficacy profile, focusing mainly on children. We conclude that inhaled antibiotics as an adjuvant maintenance treatment option could be tried in a subgroup of patients with frequent exacerbations and recent or chronic Pseudomonas aeruginosa infection as they appear to have beneficial effects on exacerbation rate and bacterial load with minimal safety concerns. However, the level of evidence in children is extremely low; therefore, further research is needed on the validity of this recommendation.
Collapse
Affiliation(s)
- Maria Tsouprou
- Pediatric Allergy and Respiratory Unit, 3rd Department of Pediatrics, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (M.T.); (D.M.)
- Department of Pediatrics, 1st Pediatric Clinic, Agia Sofia Hospital, 11527 Athens, Greece;
| | | | - Evanthia Botsa
- Department of Pediatrics, 1st Pediatric Clinic, Agia Sofia Hospital, 11527 Athens, Greece;
| | - Konstantinos Douros
- Pediatric Allergy and Respiratory Unit, 3rd Department of Pediatrics, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (M.T.); (D.M.)
| | - Dafni Moriki
- Pediatric Allergy and Respiratory Unit, 3rd Department of Pediatrics, “Attikon” University Hospital, School of Medicine, National and Kapodistrian University of Athens, 12462 Athens, Greece; (M.T.); (D.M.)
| |
Collapse
|
18
|
Han Y, Teng TM, Han J, Kim HS. Antibiotic-associated changes in Akkermansia muciniphila alter its effects on host metabolic health. MICROBIOME 2025; 13:48. [PMID: 39920776 PMCID: PMC11804010 DOI: 10.1186/s40168-024-02023-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/17/2024] [Accepted: 12/19/2024] [Indexed: 02/09/2025]
Abstract
BACKGROUND Altered gut microbiota has emerged as a major contributing factor to the etiology of chronic conditions in humans. Antibiotic exposure, historically dating back to the mass production of penicillin in the early 1940s, has been proposed as a primary contributor to the cumulative alteration of microbiota over generations. However, the mechanistic link between the antibiotics-altered microbiota and chronic conditions remains unclear. RESULTS In this study, we discovered that variants of the key beneficial gut microbe, Akkermansia muciniphila, were selected upon exposure to penicillin. These variants had mutations in the promoter of a TEM-type β-lactamase gene or pur genes encoding the de novo purine biosynthesis pathway, and they exhibited compromised abilities to mitigate host obesity in a murine model. Notably, variants of A. muciniphila are prevalent in the human microbiome worldwide. CONCLUSIONS These findings highlight a previously unknown mechanism through which antibiotics influence host health by affecting the beneficial capacities of the key gut microbes. Furthermore, the global prevalence of A. muciniphila variants raises the possibility that these variants contribute to global epidemics of chronic conditions, warranting further investigations in human populations. Video Abstract.
Collapse
Affiliation(s)
- Yumin Han
- Division of Biosystems & Biomedical Sciences, College of Health Sciences, 145 Anam-Ro, Seongbuk-Gu, Seoul, 02841, Korea
| | - Teh Min Teng
- Division of Biosystems & Biomedical Sciences, College of Health Sciences, 145 Anam-Ro, Seongbuk-Gu, Seoul, 02841, Korea
| | - Juwon Han
- Division of Biosystems & Biomedical Sciences, College of Health Sciences, 145 Anam-Ro, Seongbuk-Gu, Seoul, 02841, Korea
| | - Heenam Stanley Kim
- Division of Biosystems & Biomedical Sciences, College of Health Sciences, 145 Anam-Ro, Seongbuk-Gu, Seoul, 02841, Korea.
| |
Collapse
|
19
|
Xinhua C, Yang W, Jinyang S, Hongyue X, Wanlu Y, Mingmei Z, Jiazhang Q, Lu Y. The Effects of Baitouweng Decoction on Salmonella Typhimurium Infection and Its Underlying Mechanisms Evaluated by In Vivo and In Vitro Experiments, Network Pharmacology Analysis, and Molecular Docking Technology. Foodborne Pathog Dis 2025; 22:140-158. [PMID: 39298327 DOI: 10.1089/fpd.2023.0169] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/21/2024] Open
Abstract
Salmonella Typhimurium is a foodborne pathogen threatening livestock and human health. It is highly resistant to commonly used clinical antibiotics, and it is urgently needed to explore new anti-Salmonella treatment schemes. In this study, first, our in vivo mouse experiments showed that Baitouweng decoction (BTW), a classical Traditional Chinese Medicine (TCM) prescription, had good efficacy against Salmonella Typhimurium infection: mitigating weight loss of mice; lowering the bacterial load of liver, spleen, and colon; reducing the production of serum inflammatory factors (interleukin-1β and tumor necrosis factor-α); and decreasing histological index scores than that in the Salmonella Typhimurium infection group. Furthermore, we explored the potential active components and molecular mechanism of BTW in the treatment of Salmonella Typhimurium infection. A total of 465 compounds of BTW were retrieved from herb website and 227 bioactive compounds were identified, 911 potential BTW-related targets and 1,602 disease targets of Salmonella Typhimurium infection were acquired by ten public analytical databases, among them, 188 genes were overlay targets of BTW-Salmonella Typhimurium; String, Metascape, and Cytoscape plug-in Molecular Complex Detection and ClueGo analysis pointed that BTW exerted an anti-Salmonella effect through a multicomponent, multitarget, and multipathway manner, including 10 hub targets (TNF, AKT CASP3, ALB, EGFR, JUN, MAPK, STAT3, VEGFA, and TP53) and 94 pathways such as cell apoptosis, inflammation, and metabolism. Finally, AutoDock Vina showed that the hub target AKT1 with menispermine and quercetin had good binding energy, which was confirmed by the in vitro cellular thermal shift assay and drug affinity responsive target stability assay. This study laid the foundation for further study of BTW mechanism and for further development of BTW anti-Salmonella.
Collapse
Affiliation(s)
- Cui Xinhua
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine Jilin University, Center of Infectious Diseases and Pathogen Biology, Department of Infectious Diseases, First Hospital of Jilin University, Changchun, China
| | - Wang Yang
- State Key Laboratory for Diagnosis and Treatment of Severe Zoonotic Infectious Diseases, Key Laboratory for Zoonosis Research, Ministry of Education, Institute of Zoonosis, College of Veterinary Medicine Jilin University, Center of Infectious Diseases and Pathogen Biology, Department of Infectious Diseases, First Hospital of Jilin University, Changchun, China
| | | | | | | | | | | | | |
Collapse
|
20
|
Calvez V, Puca P, Di Vincenzo F, Del Gaudio A, Bartocci B, Murgiano M, Iaccarino J, Parand E, Napolitano D, Pugliese D, Gasbarrini A, Scaldaferri F. Novel Insights into the Pathogenesis of Inflammatory Bowel Diseases. Biomedicines 2025; 13:305. [PMID: 40002718 PMCID: PMC11853239 DOI: 10.3390/biomedicines13020305] [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: 12/07/2024] [Revised: 01/17/2025] [Accepted: 01/21/2025] [Indexed: 02/27/2025] Open
Abstract
Inflammatory bowel diseases (IBDs), encompassing Crohn's disease and ulcerative colitis, are complex chronic disorders characterized by an intricate interplay between genetic predisposition, immune dysregulation, gut microbiota alterations, and environmental exposures. This review aims to synthesize recent advances in IBD pathogenesis, exploring key mechanisms and potential avenues for prevention and personalized therapy. A comprehensive literature search was conducted across major bibliographic databases, selecting the most recent and impactful studies on IBD pathogenesis. The review integrates findings from multi-omics analyses, single-cell transcriptomics, and longitudinal cohort studies, focusing on immune regulation, gut microbiota dynamics, and environmental factors influencing disease onset and progression. Immune dysregulation, including macrophage polarization (M1 vs. M2) and Th17 activation, emerges as a cornerstone of IBD pathogenesis. Dysbiosis, as a result of reduced alpha and beta diversity and overgrowth of harmful taxa, is one of the main contributing factors in causing inflammation in IBD. Environmental factors, including air and water pollutants, maternal smoking, and antibiotic exposure during pregnancy and infancy, significantly modulate IBD risk through epigenetic and microbiota-mediated mechanisms. While recent advances have supported the development of new therapeutic strategies, deeply understanding the complex dynamics of IBD pathogenesis remains challenging. Future efforts should aim to reduce the burden of disease with precise, personalized treatments and lower the incidence of IBD through early-life prevention and targeted interventions addressing modifiable risk factors.
Collapse
Affiliation(s)
- Valentin Calvez
- IBD Unit, UOC CEMAD Medicina Interna e Gastroenterologia, Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche Addominali ed Endocrino Metaboliche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (V.C.); (P.P.); (D.N.); (D.P.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Pierluigi Puca
- IBD Unit, UOC CEMAD Medicina Interna e Gastroenterologia, Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche Addominali ed Endocrino Metaboliche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (V.C.); (P.P.); (D.N.); (D.P.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Federica Di Vincenzo
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Angelo Del Gaudio
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Bianca Bartocci
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Marco Murgiano
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Jacopo Iaccarino
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Erfan Parand
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Daniele Napolitano
- IBD Unit, UOC CEMAD Medicina Interna e Gastroenterologia, Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche Addominali ed Endocrino Metaboliche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (V.C.); (P.P.); (D.N.); (D.P.)
| | - Daniela Pugliese
- IBD Unit, UOC CEMAD Medicina Interna e Gastroenterologia, Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche Addominali ed Endocrino Metaboliche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (V.C.); (P.P.); (D.N.); (D.P.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Antonio Gasbarrini
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| | - Franco Scaldaferri
- IBD Unit, UOC CEMAD Medicina Interna e Gastroenterologia, Centro Malattie dell’Apparato Digerente, Dipartimento di Scienze Mediche e Chirurgiche Addominali ed Endocrino Metaboliche, Fondazione Policlinico Universitario Agostino Gemelli IRCCS, 00168 Rome, Italy; (V.C.); (P.P.); (D.N.); (D.P.)
- Dipartimento di Medicina e Chirurgia Traslazionale, Università Cattolica del Sacro Cuore, 00168 Rome, Italy; (F.D.V.); (A.D.G.); (B.B.); (M.M.); (J.I.); (E.P.); (A.G.)
| |
Collapse
|
21
|
Takamaru H, Tsay C, Shiba S, Yachida S, Saito Y. Microbiome and Colorectal Cancer in Humans: A Review of Recent Studies. J Anus Rectum Colon 2025; 9:20-24. [PMID: 39882224 PMCID: PMC11772794 DOI: 10.23922/jarc.2024-080] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Accepted: 08/04/2024] [Indexed: 01/31/2025] Open
Abstract
The tumor microenvironment has recently been well-studied in various gastrointestinal cancers, including colorectal cancer (CRC). The gut microbiota, a collection of microorganisms in the human gastrointestinal tract, is one of the microenvironments associated with colon carcinogenesis. It has been challenging to elucidate the mechanisms by which gut microbiota contributes to carcinogenesis and cancer progression due to complex interactions with the host, including its metabolites and immune and inflammatory responses. Various studies described the influence of diet on reported changes in the composition and microbiota of gut bacteria and its association with CRC. In recent years, metagenomic techniques such as shotgun sequencing and genome-wide association studies focused on understanding the role of the microbiota and the metabolome on early CRCs and colon carcinogenesis to determine if there are modifiable or intervenable targets for CRC. In this review, we will attempt to provide an overview of gut microbiota related to CRC, with particular attention to the findings of recent studies.
Collapse
Affiliation(s)
| | - Cynthia Tsay
- Division of Gastroenterology and Hepatology, Weill Cornell Medical College, New York, USA
| | - Satoshi Shiba
- Division of Cancer Genomics, National Cancer Center Research Institute, Tokyo, Japan
| | - Shinichi Yachida
- Department of Cancer Genome Informatics, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Yutaka Saito
- Endoscopy Division, National Cancer Center Hospital, Tokyo, Japan
| |
Collapse
|
22
|
Mendes Pedro D, Santos D, Meneses M, Gonçalves F, Domingos GJ, Caneiras C. Risk of Colonization with Multidrug-Resistant Gram-Negative Bacteria Among Travellers and Migrants: A Narrative Review. Trop Med Infect Dis 2025; 10:26. [PMID: 39852677 PMCID: PMC11769174 DOI: 10.3390/tropicalmed10010026] [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: 12/17/2024] [Revised: 01/10/2025] [Accepted: 01/14/2025] [Indexed: 01/26/2025] Open
Abstract
Globalization in the 21st century has posed several challenges. In particular, the spread of multidrug-resistant bacterial strains, especially Gram-negative bacteria, which are prevalent in certain regions of the world, is one of the most critical issues. This raises concerns about the risks associated with the booming tourism industry and migratory flows. In fact, even transient colonization with multidrug-resistant strains can present significant challenges to individual, family, and public health. Understanding the epidemiology and mechanisms of resistance, associated risk factors and prevention policies is therefore essential to ensure that strategies are in place to limit the global spread of high-risk bacterial clones and thereby protect public health.
Collapse
Affiliation(s)
- Diogo Mendes Pedro
- Laboratory of Microbiology Research in Environmental Health (EnviHealthMicro Lab), Institute of Environmental Health (ISAMB), Associate Laboratory TERRA, Faculdade de Medicina, Universidade de Lisboa, 1249-028 Lisboa, Portugal; (D.S.); (M.M.); (C.C.)
- Infectious Diseases Department, ULS Santa Maria, 1649-028 Lisboa, Portugal; (F.G.); (G.J.D.)
- Infectious Diseases University Clinic, Faculdade de Medicina, Universidade de Lisboa, 1249-028 Lisboa, Portugal
- Institute of Pharmacology and Neurosciences, Faculdade de Medicina, Universidade de Lisboa, 1249-028 Lisboa, Portugal
| | - Daniela Santos
- Laboratory of Microbiology Research in Environmental Health (EnviHealthMicro Lab), Institute of Environmental Health (ISAMB), Associate Laboratory TERRA, Faculdade de Medicina, Universidade de Lisboa, 1249-028 Lisboa, Portugal; (D.S.); (M.M.); (C.C.)
| | - Maria Meneses
- Laboratory of Microbiology Research in Environmental Health (EnviHealthMicro Lab), Institute of Environmental Health (ISAMB), Associate Laboratory TERRA, Faculdade de Medicina, Universidade de Lisboa, 1249-028 Lisboa, Portugal; (D.S.); (M.M.); (C.C.)
| | - Fátima Gonçalves
- Infectious Diseases Department, ULS Santa Maria, 1649-028 Lisboa, Portugal; (F.G.); (G.J.D.)
| | | | - Cátia Caneiras
- Laboratory of Microbiology Research in Environmental Health (EnviHealthMicro Lab), Institute of Environmental Health (ISAMB), Associate Laboratory TERRA, Faculdade de Medicina, Universidade de Lisboa, 1249-028 Lisboa, Portugal; (D.S.); (M.M.); (C.C.)
- Egas Moniz Interdisciplinary Research Center, Egas Moniz School of Health & Science, Monte da Caparica, 2829-511 Almada, Portugal
- Institute of Preventive Medicine and Public Health (IMP&SP), Faculdade de Medicina, Universidade de Lisboa, 1249-028 Lisboa, Portugal
| |
Collapse
|
23
|
Mukuda K, Inoue R, Takata M, Takazawa K, Noma H, Morishima S, Oda M, Ma'arif AS, Endo Y, Sunada H, Doi A, Matsuda R, Nishikawa Y, Okada K, Kitaura T, Nakamoto M, Yamasaki A, Chikumi H. Temporal effects of lascufloxacin on human gut and salivary microbiota: Analysis using next-generation sequencing method. J Infect Chemother 2025; 31:102483. [PMID: 39089446 DOI: 10.1016/j.jiac.2024.07.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/26/2024] [Accepted: 07/28/2024] [Indexed: 08/04/2024]
Abstract
INTRODUCTION Antimicrobial treatment disrupts human microbiota. The effects of lascufloxacin (LSFX), a new fluoroquinolone, on human microbiota remains unknown. Therefore, in this study, we aimed to evaluate the effects of LSFX administration on the gut and salivary microbiota of healthy participants and those with pneumonia. METHODS LSFX (75 mg, once a day, orally) was administered to healthy adults (healthy group) and adult patients with pneumonia (pneumonia group), and fecal and saliva samples were collected at five time points (Days 0, 3, 7, 14, and 28). Using the collected samples, α- and β-diversity indices, as well as bacterial composition of the gut microbiota and salivary microbiota were analyzed using next-generation sequencing. RESULTS In the healthy group, α-diversity indices of the gut and salivary microbiota were reduced and the lowest values on Day 3. For the gut microbiota, the Chao1 index (richness) recovered on Day 28, whereas the Shannon index (evenness) did not. In the salivary microbiota, the Chao1 and Shannon indices did not recover within the 28 day period. The β-diversity indices changed after LSFX administration and subsequently recovered on Day 28. After LSFX administration, the abundance of the Lachnospiraceae family decreased in the gut microbiota, and the abundance of Granulicatella, Streptococcus, Prevotella, Absconditabacteriales(SR1), and Saccharimonadales decreased in the salivary microbiota. In the pneumonia group, the α-diversity indices were lowest on Day 14 after LSFX administration. CONCLUSIONS We elucidated that LSFX administration differentially affected the gut and salivary microbiota; however, the richness and beta diversity recovered within 28 days.
Collapse
Affiliation(s)
- Kengo Mukuda
- Division of Infectious Diseases, Faculty of Medicine, Tottori University, Nishi-cho 86, Yonago, Tottori, 683-8503, Japan.
| | - Ryo Inoue
- Laboratory of Animal Science, Department of Applied Biological Sciences, Faculty of Agriculture, Setsunan University, 45-1 Nagaotogecho, Hirakata, Osaka, 573-0101, Japan.
| | - Miyako Takata
- Department of Pathobiological Science and Technology, Graduate School of Medical Science, School of Health Science, Faculty of Medicine, Tottori University, Nishi-cho 86, Yonago, Tottori, 683-8503, Japan.
| | - Kenji Takazawa
- Medical Corporation Shinanokai Shinanozaka Clinic, Yotsuya Medical Bldg. 3F, 20, Samon-cho, Shinjuku-ku, Tokyo, 160-0017, Japan.
| | - Hisashi Noma
- Department of Interdisciplinary Statistical Mathematics, The Institute of Statistical Mathematics, 10-3 Midori-cho, Tachikawa City, Tokyo, 683-8503, Japan.
| | - So Morishima
- Laboratory of Animal Science, Department of Applied Biological Sciences, Faculty of Agriculture, Setsunan University, 45-1 Nagaotogecho, Hirakata, Osaka, 573-0101, Japan.
| | - Machi Oda
- Laboratory of Animal Science, Department of Applied Biological Sciences, Faculty of Agriculture, Setsunan University, 45-1 Nagaotogecho, Hirakata, Osaka, 573-0101, Japan.
| | - Athok Shofiudin Ma'arif
- Division of Infectious Diseases, Faculty of Medicine, Tottori University, Nishi-cho 86, Yonago, Tottori, 683-8503, Japan.
| | - Yusuke Endo
- Organisation for Research Institute and Promotion, Tottori University, Nishi-cho 86, Yonago, Tottori, 683-8503, Japan.
| | - Hiroshi Sunada
- Advanced Medicine, Innovation and Clinical Research Center, Tottori University Hospital, Nishi-cho 36-1, Yonago, Tottori, 683-8504, Japan.
| | - Ayumu Doi
- Division of Infectious Diseases, Faculty of Medicine, Tottori University, Nishi-cho 86, Yonago, Tottori, 683-8503, Japan.
| | - Risa Matsuda
- Division of Infectious Diseases, Faculty of Medicine, Tottori University, Nishi-cho 86, Yonago, Tottori, 683-8503, Japan.
| | - Yukari Nishikawa
- Division of Infectious Diseases, Faculty of Medicine, Tottori University, Nishi-cho 86, Yonago, Tottori, 683-8503, Japan.
| | - Kensaku Okada
- Division of Infectious Diseases, Faculty of Medicine, Tottori University, Nishi-cho 86, Yonago, Tottori, 683-8503, Japan.
| | - Tsuyoshi Kitaura
- Division of Infectious Diseases, Faculty of Medicine, Tottori University, Nishi-cho 86, Yonago, Tottori, 683-8503, Japan.
| | - Masaki Nakamoto
- Division of Infectious Diseases, Faculty of Medicine, Tottori University, Nishi-cho 86, Yonago, Tottori, 683-8503, Japan.
| | - Akira Yamasaki
- Division of Respiratory Medicine and Rheumatology Department of the Multidisciplinary Internal Medicine, Faculty of Medicine, Tottori University, Nishi-cho 86, Yonago, Tottori, 683-8503, Japan.
| | - Hiroki Chikumi
- Division of Infectious Diseases, Faculty of Medicine, Tottori University, Nishi-cho 86, Yonago, Tottori, 683-8503, Japan.
| |
Collapse
|
24
|
Junca H, Steube A, Mrowietz S, Stallhofer J, Vital M, dos Anjos Borges LG, Pieper DH, Stallmach A. Bacterial and viral assemblages in ulcerative colitis patients following fecal microbiota and fecal filtrate transfer. ISME COMMUNICATIONS 2025; 5:ycae167. [PMID: 39830095 PMCID: PMC11740987 DOI: 10.1093/ismeco/ycae167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Revised: 12/11/2024] [Accepted: 12/19/2024] [Indexed: 01/22/2025]
Abstract
Fecal microbiota filtrate transfer is discussed as a safe alternative to fecal microbiota transfer (FMT) to treat ulcerative colitis. We investigated modulation of viral and bacterial composition during fecal microbiota filtrate transfer followed by FMT in six patients with active ulcerative colitis (where clinical activity improved in three patients after filtrate transfer) and combined 16S ribosomal RNA gene amplicon sequencing with a virome analysis pipeline including fast viral particle enrichment and metagenome mapping to detect frequencies of 45,033 reference bacteriophage genomes. We showed that after antibiotic treatment and during filtrate transfer, the bacterial community typically adopted a stable composition distinct to that before antibiotic treatment, with no change toward a donor community. FMT in contrast typically changed the bacterial community to a community with similarity to donor(s). There were no indications of an establishment of predominant donor viruses during filtrate transfer but a remodeling of the virome. In contrast, the establishment of donor viruses during FMT correlated with the predicted hosts established during such transfer. Our approach warrants further investigation in a randomized trial to evaluate larger therapeutic interventions in a comparable and efficient manner.
Collapse
Affiliation(s)
- Howard Junca
- Microbial Interactions and Processes Research Group, Helmholtz Centre for Infection Research, D-38124 Braunschweig, Germany
| | - Arndt Steube
- Department of Internal Medicine IV (Gastroenterology, Hepatology and Infectious Diseases), University Hospital Jena, D-07747 Jena, Germany
| | - Simon Mrowietz
- Department of Internal Medicine IV (Gastroenterology, Hepatology and Infectious Diseases), University Hospital Jena, D-07747 Jena, Germany
| | - Johannes Stallhofer
- Department of Internal Medicine IV (Gastroenterology, Hepatology and Infectious Diseases), University Hospital Jena, D-07747 Jena, Germany
| | - Marius Vital
- Microbial Interactions and Processes Research Group, Helmholtz Centre for Infection Research, D-38124 Braunschweig, Germany
| | - Luiz Gustavo dos Anjos Borges
- Microbial Interactions and Processes Research Group, Helmholtz Centre for Infection Research, D-38124 Braunschweig, Germany
| | - Dietmar H Pieper
- Microbial Interactions and Processes Research Group, Helmholtz Centre for Infection Research, D-38124 Braunschweig, Germany
| | - Andreas Stallmach
- Department of Internal Medicine IV (Gastroenterology, Hepatology and Infectious Diseases), University Hospital Jena, D-07747 Jena, Germany
| |
Collapse
|
25
|
Charitos IA, Inchingolo AM, Ferrante L, Inchingolo F, Inchingolo AD, Castellaneta F, Cotoia A, Palermo A, Scacco S, Dipalma G. The Gut Microbiota's Role in Neurological, Psychiatric, and Neurodevelopmental Disorders. Nutrients 2024; 16:4404. [PMID: 39771025 PMCID: PMC11677138 DOI: 10.3390/nu16244404] [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: 11/28/2024] [Revised: 12/17/2024] [Accepted: 12/20/2024] [Indexed: 01/11/2025] Open
Abstract
AIM This article aims to explore the role of the human gut microbiota (GM) in the pathogenesis of neurological, psychiatric, and neurodevelopmental disorders, highlighting its influence on health and disease, and investigating potential therapeutic strategies targeting GM modulation. MATERIALS AND METHODS A comprehensive analysis of the gut microbiota's composition and its interaction with the human body, particularly, its role in neurological and psychiatric conditions, is provided. The review discusses factors influencing GM composition, including birth mode, breastfeeding, diet, medications, and geography. Additionally, it examines the GM's functions, such as nutrient absorption, immune regulation, and pathogen defense, alongside its interactions with the nervous system through the gut-brain axis, neurotransmitters, and short-chain fatty acids (SCFAs). RESULTS Alterations in the GM are linked to various disorders, including Parkinson's disease, multiple sclerosis, depression, schizophrenia, ADHD, and autism. The GM influences cognitive functions, stress responses, and mood regulation. Antibiotic use disrupts GM diversity, increasing the risk of metabolic disorders, obesity, and allergic diseases. Emerging therapies such as probiotics, prebiotics, and microbiota transplantation show promise in modulating the GM and alleviating symptoms of neurological and psychiatric conditions. CONCLUSIONS The modulation of the GM represents a promising approach for personalized treatment strategies. Further research is needed to better understand the underlying mechanisms and to develop targeted therapies aimed at restoring GM balance for improved clinical outcomes.
Collapse
Affiliation(s)
- Ioannis Alexandros Charitos
- Istituti Clinici Scientifici Maugeri IRCCS, Pneumology and Respiratory Rehabilitation Unit, “Institute” of Bari, 70124 Bari, Italy;
| | - Angelo Michele Inchingolo
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (L.F.); (A.D.I.); (G.D.)
| | - Laura Ferrante
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (L.F.); (A.D.I.); (G.D.)
| | - Francesco Inchingolo
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (L.F.); (A.D.I.); (G.D.)
| | - Alessio Danilo Inchingolo
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (L.F.); (A.D.I.); (G.D.)
| | - Francesca Castellaneta
- U.O.C. Immunohematology and Transfusion Medicine—S.I.M.T. Di Venere Hospital, 70131 Bari, Italy;
| | - Antonella Cotoia
- Department of Intensive Care, University Hospital of Foggia, 71121 Foggia, Italy;
| | - Andrea Palermo
- Department of Experimental Medicine, University of Salento, 73100 Lecce, Italy;
| | - Salvatore Scacco
- Department of Translational Biomedicine and Neuroscience (DiBraiN), Aldo Moro University, 70121 Bari, Italy;
| | - Gianna Dipalma
- Interdisciplinary Department of Medicine, University of Bari “Aldo Moro”, 70124 Bari, Italy; (A.M.I.); (L.F.); (A.D.I.); (G.D.)
| |
Collapse
|
26
|
Shahid A, Chambers S, Scott-Thomas A, Bhatia M. Gut Microbiota and Liver Dysfunction in Sepsis: The Role of Inflammatory Mediators and Therapeutic Approaches. Int J Mol Sci 2024; 25:13415. [PMID: 39769181 PMCID: PMC11678143 DOI: 10.3390/ijms252413415] [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: 11/28/2024] [Revised: 12/12/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
Sepsis is a life-threatening complication caused by an uncontrolled immune response to infection that can lead to multi-organ dysfunction, including liver injury. Recent research has shown the critical role of gut microbiota in sepsis pathogenesis, with the gut-liver axis playing a crucial role in disease progression. Mechanisms such as the disruption of the gut barrier and liver injury pathways mediated by cytokines, chemokines, adhesion molecules, hydrogen sulfide (H2S). and substance P (SP) have been the focus of recent studies. Some potential biomarkers and gut microbiota-targeted therapies have shown promise as emerging tools for predicting and managing sepsis. This review describes the role of the gut-liver axis in sepsis and the potential of microbiota-targeted therapies and biomarker-driven interventions to improve sepsis outcomes.
Collapse
Affiliation(s)
| | | | | | - Madhav Bhatia
- Department of Pathology and Biomedical Science, University of Otago, Christchurch 8140, New Zealand; (A.S.); (S.C.); (A.S.-T.)
| |
Collapse
|
27
|
Cabezas-Cruz A, Bermúdez-Humarán LG. Exploring the relationship between Faecalibacterium duncaniae and Escherichia coli in inflammatory bowel disease (IBD): Insights and implications. Comput Struct Biotechnol J 2024; 23:1-9. [PMID: 38094217 PMCID: PMC10716368 DOI: 10.1016/j.csbj.2023.11.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 11/14/2023] [Accepted: 11/14/2023] [Indexed: 03/04/2025] Open
Abstract
Inflammatory bowel disease (IBD) is a group of disorders characterized by an inflammation of the gastrointestinal tract (GIT) and represents a major social and economic burden. Despite ongoing research into the etiology and pathophysiology of this multifactorial disease, treatment options remain limited. From this perspective, the gut microbiota has emerged as a potential player in the pathogenesis of IBD, and animal and human studies support this hypothesis. Indeed, the human gut is one of the most complex ecological communities (composed of 1013-1014 microorganisms) that plays a critical role in human health by influencing normal physiology and disease susceptibility through its collective metabolic activities and host interactions. In addition, live probiotic bacteria present in some food products (which transit through the GIT) have been shown to interact with the host immune system and confer several health benefits. The aim of this review is to provide an overview of the link between Faecalibacterium duncaniae and Escherichia coli and IBD, highlighting the main areas of research in this field. An ecological perspective on the gut microbiota may offer new insights for the development of clinical therapies targeting this bacterial community to improve human health.
Collapse
Affiliation(s)
- Alejandro Cabezas-Cruz
- Anses, INRAE, Ecole Nationale Vétérinaire d’Alfort, UMR BIPAR, Laboratoire de Santé Animale, Maisons-Alfort F-94700, France
| | | |
Collapse
|
28
|
Wasti QZ, Sabar MF, Farooq A, Khan MU. Stepping towards pollen DNA metabarcoding: A breakthrough in forensic sciences. Forensic Sci Med Pathol 2024; 20:1464-1474. [PMID: 38147285 DOI: 10.1007/s12024-023-00770-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 12/20/2023] [Indexed: 12/27/2023]
Abstract
This review is engaged in determining the capability of plant pollen as a significant source of evidence for the linkage between suspects and crime location in forensic sciences. Research and review articles were collected from Google Scholar, the Web of Science, and PubMed. Articles were searched using specific keywords such as "Forensic Palynology," "Pollen metabarcoding," "Plant forensics," and "Pollen" AND "criminal investigation." Boolean logic was also utilized to narrow the articles to be included in this review article. Through the literature and exploratory research, it has been observed in the current study that with advancements in technology, forensic palynology has found its application in creating an association between the crime scene and suspected individuals to have a link to it, as pollen DNA is a long-lasting investigative tool that can effectively help forensic investigations. Moreover, the literature shows that the DNA of pollen and spores has helped forensic scientists link suspects to crime scenes, and the introduction of pollen DNA metabarcoding tools has eased the efforts of palynologists to analyze pollen DNA. The introduction of DNA metabarcoding techniques to analyze pollen from plants has helped identify the geological locations of the plants and ultimately identify the culprit.
Collapse
Affiliation(s)
- Qandeel Zaineb Wasti
- Centre for Applied Molecular Biology, University of the Punjab, Lahore, Pakistan
| | | | - Abeera Farooq
- Punjab University College of Pharmacy, University of the Punjab, Lahore, Pakistan
| | - Muhammad Umer Khan
- Institute of Molecular Biology and Biotechnology, The University of Lahore, Lahore, Pakistan.
| |
Collapse
|
29
|
Herbin SR, Crum H, Gens K. Breaking the Cycle of Recurrent Clostridioides difficile Infections: A Narrative Review Exploring Current and Novel Therapeutic Strategies. J Pharm Pract 2024; 37:1361-1373. [PMID: 38739837 DOI: 10.1177/08971900241248883] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Clostridioides difficile is a toxin-producing bacteria that is a main cause of antibiotic-associated diarrhea. Clostridioides difficile infections (CDI) are associated with disruptions within the gastrointestinal (GI) microbiota which can be further exacerbated by CDI-targeted antibiotic treatment thereby causing recurrent CDI (rCDI) and compounding the burden placed on patients and the healthcare system. Treatment of rCDI consists of antibiotics which can be paired with preventative therapeutics, such as bezlotoxumab or fecal microbiota transplants (FMTs), if sustained clinical response is not obtained. Newer preventative strategies have been recently approved to assist in restoring balance within the GI system with the goal of preventing recurrent infections.
Collapse
Affiliation(s)
- Shelbye R Herbin
- Antimicrobial Stewardship and Medication Safety, John D. Dingell VA Medical Center, Detroit, MI, USA
| | - Hannah Crum
- Mercy Hospital Southeast, Cape Girardeau, MO, USA
| | - Krista Gens
- Allina Health, Minneapolis, MN, USA
- Abbott Northwestern Hospital, Minneapolis, MN, USA
| |
Collapse
|
30
|
Rosenberg E. Diversity of bacteria within the human gut and its contribution to the functional unity of holobionts. NPJ Biofilms Microbiomes 2024; 10:134. [PMID: 39580487 PMCID: PMC11585559 DOI: 10.1038/s41522-024-00580-y] [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: 03/13/2024] [Accepted: 10/07/2024] [Indexed: 11/25/2024] Open
Abstract
The composition of bacteria in the human colon has been a subject of interest since the beginning of microbiology. With the development of methods for culturing strict anaerobic bacteria under multiple culture conditions, it was shown the gut contained more than 400 bacterial species and different people harbor different abundant species. The term "gut microbiome" in this review refers to bacteria studied in stool samples. Molecular methods for determining the bacterial composition of human gut has revealed more than 3000 species and less than 130 genera, indicating that the diversity of human colonic bacteria is concentrated at the species and strain levels. This review concludes with a discussion of how diversity can lead to unity of individual holobionts, between holobionts, and between populations. One of the reasons for the unity is that different bacterial species can have similar functional genes.
Collapse
Affiliation(s)
- Eugene Rosenberg
- Department of Microbiology, Tel Aviv University, Tel Aviv, Israel.
| |
Collapse
|
31
|
Backman T, Burbano HA, Karasov TL. Tradeoffs and constraints on the evolution of tailocins. Trends Microbiol 2024; 32:1084-1095. [PMID: 39504934 DOI: 10.1016/j.tim.2024.04.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Revised: 03/25/2024] [Accepted: 04/02/2024] [Indexed: 11/08/2024]
Abstract
Phage tail-like bacteriocins (tailocins) are protein complexes produced by bacteria with the potential to kill their neighbors. Widespread throughout Gram-negative bacteria, tailocins exhibit extreme specificity in their targets, largely killing closely related strains. Despite their presence in diverse bacteria, the impact of these competitive weapons on the surrounding microbiota is largely unknown. Recent studies revealed the rapid evolution and genetic diversity of tailocins in microbial communities and suggest that there are constraints on the evolution of specificity and resistance. Given the precision of their targeted killing and the ease of engineering new specificities, understanding the evolution and ecological impact of tailocins may enable the design of promising candidates for novel targeted antibiotics.
Collapse
Affiliation(s)
- Talia Backman
- School of Biological Sciences, University of Utah 257S 1400E, Salt Lake City, UT 84112, USA.
| | - Hernán A Burbano
- Centre for Life's Origins and Evolution, Department of Genetics, Evolution, and Environment, University College London, London, UK.
| | - Talia L Karasov
- School of Biological Sciences, University of Utah 257S 1400E, Salt Lake City, UT 84112, USA.
| |
Collapse
|
32
|
Dutta R, Stothers L, Ackerman AL. Manipulating the Gut Microbiome in Urinary Tract Infection-Prone Patients. Urol Clin North Am 2024; 51:525-536. [PMID: 39349020 DOI: 10.1016/j.ucl.2024.07.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/02/2024]
Abstract
Although antibiotics remain the mainstay of urinary tract infection treatment, many affected women can be caught in a vicious cycle in which antibiotics given to eradicate one infection predispose them to develop another. This effect is primarily mediated by disturbances in the gut microbiome that both directly enrich for uropathogenic overgrowth and induce systemic alterations in inflammation, tissue permeability, and metabolism that also decrease host resistance to infection recurrences. Here, we discuss nonantibiotic approaches to manipulating the gut microbiome to reverse the systemic consequences of antibiotics, including cranberry supplementation and other dietary approaches, probiotic administration, and fecal microbiota transplantation.
Collapse
Affiliation(s)
- Rahul Dutta
- Division of Urogynecology and Reconstructive Pelvic Surgery, David Geffen School of Medicine at UCLA, Box 951738, Los Angeles, CA 90095-1738, USA
| | - Lynn Stothers
- Division of Urogynecology and Reconstructive Pelvic Surgery, David Geffen School of Medicine at UCLA, Box 951738, Los Angeles, CA 90095-1738, USA
| | - A Lenore Ackerman
- Division of Urogynecology and Reconstructive Pelvic Surgery, David Geffen School of Medicine at UCLA, Box 951738, Los Angeles, CA 90095-1738, USA.
| |
Collapse
|
33
|
dos Santos Pereira E, de Oliveira Raphaelli C, Massaut KB, Ribeiro JA, Soares Vitola HR, Pieniz S, Fiorentini ÂM. Probiotics: Therapeutic Strategy on the Prevention and Treatment of
Inflammatory Diseases: Obesity, Type 2 Diabetes Mellitus and Celiac
Disease. CURRENT NUTRITION & FOOD SCIENCE 2024; 20:1112-1125. [DOI: 10.2174/0115734013252358231016181809] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 07/29/2023] [Accepted: 08/24/2023] [Indexed: 01/03/2025]
Abstract
Background:
Recent evidence demonstrates the fundamental role of the gut microbiota
in inflammatory diseases, and several mechanisms of action of probiotics in improvement of inflammatory
parameters.
Objective:
The objective of this review was to relate the consumption of probiotic bacteria and its
effects on inflammatory diseases, including obesity, type II diabetes and celiac disease.
Methods:
A search was carried out in English, between the years 2011 and 2022, for research articles
and clinical trials with humans and in vivo studies. Research showed improvement in cardiovascular
risk markers, and improvement in insulin sensitivity, lipid profile and plasma atherogenic
index, in obesity with the use of probiotics. In type II diabetes, decreased levels of fasting glucose,
glycated hemoglobin, insulin and glycemic index, and increased levels of peptide 1, superoxide
dismutase and glutathione peroxidase were observed.
Results:
In addition to cellular protection of the islets of Langerhans and positive alteration of TNF-
α and IL-1β markers. Improvement in the condition of patients with celiac disease was observed,
since the neutralization of the imbalance in serotonin levels was observed, reducing the expression
of genes of interest and also, a decrease in cytokines.
Conclusion:
Therefore, the use of probiotics should be encouraged.
Collapse
Affiliation(s)
| | | | - Khadija Bezerra Massaut
- Department of Food Science and Technology, Universidade Federal de Pelotas, Pelotas, Rs, Brazil
| | - Jardel Araújo Ribeiro
- Department of Food Science and Technology, Universidade Federal de Pelotas, Pelotas, Rs, Brazil
| | | | - Simone Pieniz
- Department of Food Science and Technology, Universidade Federal de Pelotas, Pelotas, Rs, Brazil
| | - Ângela Maria Fiorentini
- Department of Food Science and Technology, Universidade Federal de Pelotas, Pelotas, Rs, Brazil
| |
Collapse
|
34
|
Ansai H, Yamada M, Masuda H, Imadome KI, Yashiro M, Noval Rivas M, Arditi M, Nakamura Y, Abe J. Association of recent antibiotic exposure and coronary artery lesions in Kawasaki disease: nationwide study. Front Pediatr 2024; 12:1467288. [PMID: 39554306 PMCID: PMC11563978 DOI: 10.3389/fped.2024.1467288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/19/2024] [Accepted: 10/21/2024] [Indexed: 11/19/2024] Open
Abstract
Objectives To investigate the relationship between recent antibiotic exposure and the development of coronary artery lesions (CALs) during the clinical course of Kawasaki disease (KD). Design Data were obtained from the 25th nationwide epidemiological survey of KD conducted in Japan from 2017 to 2018. Baseline characteristics and clinical course were compared between Antibiotics (+) and Antibiotics (-) groups. Setting Nationwide survey of KD in Japan. Participants KD patients were enrolled by response to a questionnaire sent to physicians working in pediatrics at hospitals with >100 beds. Exposure Antibiotic exposure within one week before the first hospital visit as KD patients. Main outcome measures The relationship between recent antibiotic exposure and the development of coronary artery lesions (CALs). Results Out of 28,265 KD patients, 12,918 (45.7%) received antibiotics. In KD patients who received antibiotics in the week before KD diagnosis, the frequency of coronary artery lesions (CALs) at each phase were significantly higher compared to those who did not receive antibiotics. In further analysis using propensity score matching, recent antibiotic exposure and the initial IVIG resistance were associated with CALs at the acute and the sequelae phase. After adjusting for the status of initial IVIG resistance, recent antibiotic exposure remained associated with CALs during the acute phase (adjusted OR 1.29, 95%CI 1.16, 1.43) and the sequelae phase (1.26, 95%CI 1.04, 1.52). Conclusions These observations suggest that recent antibiotic exposure might be associated with higher frequency of CAL development in KD patients, possibly by altering the gut microbiota and diminishing beneficial bacteria.
Collapse
Affiliation(s)
- Hideto Ansai
- Department of General Pediatrics and Interdisciplinary Medicine, National Center for Child Health and Development, Tokyo, Japan
- Department of Advanced Medicine for Viral Infections, National Center for Child Health and Development, Tokyo, Japan
| | - Masaki Yamada
- Department of Advanced Medicine for Viral Infections, National Center for Child Health and Development, Tokyo, Japan
| | - Hiroshi Masuda
- Department of General Pediatrics and Interdisciplinary Medicine, National Center for Child Health and Development, Tokyo, Japan
| | - Ken-Ichi Imadome
- Department of Advanced Medicine for Viral Infections, National Center for Child Health and Development, Tokyo, Japan
| | - Mayumi Yashiro
- Department of Public Health, Jichi Medical University, Tochigi, Japan
| | - Magali Noval Rivas
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Biomedical Sciences, Infectious and Immunologic Diseases Research Center (IIDRC), Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Moshe Arditi
- Department of Pediatrics, Division of Infectious Diseases and Immunology, Guerin Children’s at Cedars-Sinai Medical Center, Los Angeles, CA, United States
- Department of Biomedical Sciences, Infectious and Immunologic Diseases Research Center (IIDRC), Cedars-Sinai Medical Center, Los Angeles, CA, United States
| | - Yosikazu Nakamura
- Department of Public Health, Jichi Medical University, Tochigi, Japan
| | - Jun Abe
- Department of Advanced Medicine for Viral Infections, National Center for Child Health and Development, Tokyo, Japan
| |
Collapse
|
35
|
Bejcek A, Ancha A, Lewis M, Beaver R, Tecson K, Bomar J, Johnson C. Antibiotic use and risk of Clostridioides difficile infection in patients with inflammatory bowel disease. J Gastroenterol Hepatol 2024; 39:2417-2423. [PMID: 39148287 DOI: 10.1111/jgh.16720] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2023] [Revised: 06/23/2024] [Accepted: 07/30/2024] [Indexed: 08/17/2024]
Abstract
BACKGROUND AND AIM Patients with inflammatory bowel disease (IBD) have an increased risk of Clostridioides difficile infection (CDI) compared with those without IBD, which is worsened with antibiotic usage. While prior studies have shown a correlation between CDI development and certain classes of antibiotics, the IBD population has not been well represented. This study evaluates the rates of CDI with outpatient antibiotic use in patients with IBD. METHODS We conducted a retrospective cohort study composed of patients with IBD and compared the incidence of CDI in patients who received an outpatient prescription for antibiotics (6694 patients) against those without prescriptions (6025 patients) from 2014 to 2020 at our institution. We compared CDI rates based on nine antibiotic classes: penicillins, cephalosporins, sulfonamides, tetracyclines, macrolides, quinolones, clindamycin, metronidazole, and nitrofurantoin. RESULTS The risk of CDI was low (0.7%) but significantly higher for those with antibiotic exposure (0.9% vs 0.5%, P = 0.005) and had a positive correlation with a smoking history. The increased risk of CDI in the IBD population was attributable to the clindamycin and metronidazole classes (odds ratio = 4.7, 95% confidence interval: 1.9-11.9, P = 0.001; odds ratio = 3.6, 95% confidence interval: 2.1-6.2, P < 0.0001, respectively). CONCLUSIONS The use of clindamycin or metronidazole prescribed in an outpatient setting was associated with a statistically significant increased risk of CDI in patients with IBD. Although the association between clindamycin and CDI is a well-established and common finding, the association between metronidazole and CDI is unique in this study.
Collapse
Affiliation(s)
- Alexis Bejcek
- Division of Gastroenterology, Department of Medicine, Baylor Scott & White Medical Center, Temple, Texas, USA
| | - Anupama Ancha
- Division of Internal Medicine, Department of Medicine, Baylor Scott & White Medical Center, Temple, Texas, USA
| | - Megan Lewis
- Division of Gastroenterology, Department of Medicine, Baylor Scott & White Medical Center, Temple, Texas, USA
| | - Ryan Beaver
- Division of Infectious Diseases, Department of Medicine, Baylor Scott & White Medical Center, Temple, Texas, USA
| | - Kristen Tecson
- Baylor Scott & White Research Institute, Baylor Scott & White Health, Dallas, Texas, USA
| | - Jaccallene Bomar
- Baylor Scott & White Research Institute, Baylor Scott & White Health, Dallas, Texas, USA
| | - Christopher Johnson
- Division of Gastroenterology, Department of Medicine, Baylor Scott & White Medical Center, Temple, Texas, USA
- Department of Medicine, Baylor College of Medicine, Temple, Texas, USA
| |
Collapse
|
36
|
Heidari H, Lawrence DA. An integrative exploration of environmental stressors on the microbiome-gut-brain axis and immune mechanisms promoting neurological disorders. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2024; 27:233-263. [PMID: 38994870 DOI: 10.1080/10937404.2024.2378406] [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: 07/13/2024]
Abstract
The microbiome-gut-brain axis is altered by environmental stressors such as heat, diet, and pollutants as well as microbes in the air, water, and soil. These stressors might alter the host's microbiome and symbiotic relationship by modifying the microbial composition or location. Compartmentalized mutualistic microbes promote the beneficial interactions in the host leading to circulating metabolites and hormones such as insulin and leptin that affect inter-organ functions. Inflammation and oxidative stress induced by environmental stressors may alter the composition, distribution, and activities of the microbes in the microbiomes such that the resultant metabolite and hormone changes are no longer beneficial. The microbiome-gut-brain axis and immune adverse changes that may accompany environmental stressors are reviewed for effects on innate and adaptive immune cells, which may make host immunity less responsive to pathogens and more reactive to self-antigens. Cardiovascular and fluid exchanges to organs might adversely alter organ functionality. Organs, especially the brain, need a consistent supply of nutrients and clearance of debris; disruption of these exchanges by stressors, and involvement of gut microbiome are discussed regarding neural dysfunctions with Alzheimer's disease, autistic spectrum disorders, viral infections, and autoimmune diseases. The focus of this review includes the manner in which environmental stressors may disrupt gut microbiota leading to adverse immune and hormonal influences on development of neuropathology related to hyperhomocysteinemia, inflammation, and oxidative stress, and how certain therapeutics may be beneficial. Strategies are explored to lessen detrimental effects of environmental stressors on central and peripheral health navigated toward (1) understanding neurological disorders and (2) promoting environmental and public health and well-being.
Collapse
Affiliation(s)
- Hajar Heidari
- Department of Biomedical Sciences, University at Albany School of Public Health, Rensselaer, NY, USA
| | - David A Lawrence
- Department of Biomedical Sciences, University at Albany School of Public Health, Rensselaer, NY, USA
- Department of Environmental Health Sciences, University at Albany School of Public Health, Rensselaer, NY, USA
- New York State Department of Health, Wadsworth Center, Albany, NY, USA
| |
Collapse
|
37
|
Aruwa CE, Sabiu S. Interplay of poultry-microbiome interactions - influencing factors and microbes in poultry infections and metabolic disorders. Br Poult Sci 2024; 65:523-537. [PMID: 38920059 DOI: 10.1080/00071668.2024.2356666] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2024] [Accepted: 03/06/2024] [Indexed: 06/27/2024]
Abstract
1. The poultry microbiome and its stability at every point in time, either free range or reared under different farming systems, is affected by several environmental and innate factors. The interaction of the poultry birds with their microbiome, as well as several inherent and extraneous factors contribute to the microbiome dynamics. A poor understanding of this could worsen poultry heath and result in disease/metabolic disorders.2. Many diseased states associated with poultry have been linked to dysbiosis state, where the microbiome experiences some perturbation. Dysbiosis itself is too often downplayed; however, it is considered a disease which could lead to more serious conditions in poultry. The management of interconnected factors by conventional and emerging technologies (sequencing, nanotechnology, robotics, 3D mini-guts) could prove to be indispensable in ensuring poultry health and welfare.3. Findings showed that high-throughput technological advancements enhanced scientific insights into emerging trends surrounding the poultry gut microbiome and ecosystem, the dysbiotic condition, and the dynamic roles of intrinsic and exogenous factors in determining poultry health. Yet, a combination of conventional, -omics based and other techniques further enhance characterisation of key poultry microbiome actors, their mechanisms of action, and roles in maintaining gut homoeostasis and health, in a bid to avert metabolic disorders and infections.4. In conclusion, there is an important interplay of innate, environmental, abiotic and biotic factors impacting on poultry gut microbiome homoeostasis, dysbiosis, and overall health. Associated infections and metabolic disorders can result from the interconnected nature of these factors. Emerging concepts (interkingdom or network signalling and neurotransmitter), and future technologies (mini-gut models, cobots) need to include these interactions to ensure accurate control and outcomes.
Collapse
Affiliation(s)
- C E Aruwa
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, Durban, South Africa
| | - S Sabiu
- Department of Biotechnology and Food Science, Faculty of Applied Sciences, Durban University of Technology, Durban, South Africa
| |
Collapse
|
38
|
Liu A, Tian B, Qiu C, Su KJ, Jiang L, Zhao C, Song M, Liu Y, Qu G, Zhou Z, Zhang X, Gnanesh SSM, Thumbigere-Math V, Luo Z, Tian Q, Zhang LS, Wu C, Ding Z, Shen H, Deng HW. Multi-View Integrative Approach For Imputing Short-Chain Fatty Acids and Identifying Key factors predicting Blood SCFA. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.614767. [PMID: 39386638 PMCID: PMC11463355 DOI: 10.1101/2024.09.25.614767] [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: 10/12/2024]
Abstract
Short-chain fatty acids (SCFAs) are the main metabolites produced by bacterial fermentation of dietary fiber within gastrointestinal tract. SCFAs produced by gut microbiotas (GMs) are absorbed by host, reach bloodstream, and are distributed to different organs, thus influencing host physiology. However, due to the limited budget or the poor sensitivity of instruments, most studies on GMs have incomplete blood SCFA data, limiting our understanding of the metabolic processes within the host. To address this gap, we developed an innovative multi-task multi-view integrative approach (M2AE, Multi-task Multi-View Attentive Encoders), to impute blood SCFA levels using gut metagenomic sequencing (MGS) data, while taking into account the intricate interplay among the gut microbiome, dietary features, and host characteristics, as well as the nuanced nature of SCFA dynamics within the body. Here, each view represents a distinct type of data input (i.e., gut microbiome compositions, dietary features, or host characteristics). Our method jointly explores both view-specific representations and cross-view correlations for effective predictions of SCFAs. We applied M2AE to two in-house datasets, which both include MGS and blood SCFAs profiles, host characteristics, and dietary features from 964 subjects and 171 subjects, respectively. Results from both of two datasets demonstrated that M2AE outperforms traditional regression-based and neural-network based approaches in imputing blood SCFAs. Furthermore, a series of gut bacterial species (e.g., Bacteroides thetaiotaomicron and Clostridium asparagiforme), host characteristics (e.g., race, gender), as well as dietary features (e.g., intake of fruits, pickles) were shown to contribute greatly to imputation of blood SCFAs. These findings demonstrated that GMs, dietary features and host characteristics might contribute to the complex biological processes involved in blood SCFA productions. These might pave the way for a deeper and more nuanced comprehension of how these factors impact human health.
Collapse
Affiliation(s)
- Anqi Liu
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Bo Tian
- Center for System Biology, Data Sciences, and Reproductive Health, School of Basic Medical Science, Central South University, Yuelu, Changsha, P.R. China
| | - Chuan Qiu
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Kuan-Jui Su
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Lindong Jiang
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Chen Zhao
- College of Computing and Software Engineering, Kennesaw State University, GA, USA
| | - Meng Song
- College of Science, Xi'an Shiyou University, Xi'an, P.R. China
| | - Yong Liu
- Center for System Biology, Data Sciences, and Reproductive Health, School of Basic Medical Science, Central South University, Yuelu, Changsha, P.R. China
| | - Gang Qu
- Department of Biomedical Engineering, Tulane University, New Orleans, Louisiana, USA
| | - Ziyu Zhou
- School of Science and Engineering, Tulane University, New Orleans, LA, USA
| | - Xiao Zhang
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Shashank Sajjan Mungasavalli Gnanesh
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Vivek Thumbigere-Math
- Division of Periodontics, University of Maryland Baltimore School of Dentistry, Baltimore, USA
| | - Zhe Luo
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Qing Tian
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Li-Shu Zhang
- School of Physical Science and Engineering, College of Life Sciences and Bioengineering, Beijing Jiaotong University, Beijing, China
| | - Chong Wu
- Department of Biostatistics, The University of Texas MD Anderson Cancer Center, USA
| | - Zhengming Ding
- School of Science and Engineering, Tulane University, New Orleans, LA, USA
| | - Hui Shen
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, USA
| | - Hong-Wen Deng
- Tulane Center for Biomedical Informatics and Genomics, Deming Department of Medicine, School of Medicine, Tulane University, New Orleans, LA, USA
| |
Collapse
|
39
|
Tu V, Ren Y, Tanes C, Mukhopadhyay S, Daniel SG, Li H, Bittinger K. A quantitative approach to measure and predict microbiome response to antibiotics. mSphere 2024; 9:e0048824. [PMID: 39230261 PMCID: PMC11423569 DOI: 10.1128/msphere.00488-24] [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: 07/01/2024] [Accepted: 07/31/2024] [Indexed: 09/05/2024] Open
Abstract
Although antibiotics induce sizable perturbations in the human microbiome, we lack a systematic and quantitative method to measure and predict the microbiome's response to specific antibiotics. Here, we introduce such a method, which takes the form of a microbiome response index (MiRIx) for each antibiotic. Antibiotic-specific MiRIx values quantify the overall susceptibility of the microbiota to an antibiotic, based on databases of bacterial phenotypes and published data on intrinsic antibiotic susceptibility. We applied our approach to five published microbiome studies that carried out antibiotic interventions with vancomycin, metronidazole, ciprofloxacin, amoxicillin, and doxycycline. We show how MiRIx can be used in conjunction with existing microbiome analytical approaches to gain a deeper understanding of the microbiome response to antibiotics. Finally, we generate antibiotic response predictions for the oral, skin, and gut microbiome in healthy humans. Our approach is implemented as open-source software and is readily applied to microbiome data sets generated by 16S rRNA marker gene sequencing or shotgun metagenomics. IMPORTANCE Antibiotics are potent influencers of the human microbiome and can be a source for enduring dysbiosis and antibiotic resistance in healthcare. Existing microbiome data analysis methods can quantify perturbations of bacterial communities but cannot evaluate whether the differences are aligned with the expected activity of a specific antibiotic. Here, we present a novel method to quantify and predict antibiotic-specific microbiome changes, implemented in a ready-to-use software package. This has the potential to be a critical tool to broaden our understanding of the relationship between the microbiome and antibiotics.
Collapse
Affiliation(s)
- Vincent Tu
- Division of Gastroenterology, Hepatology, and Nutrition, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Yue Ren
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Ceylan Tanes
- Division of Gastroenterology, Hepatology, and Nutrition, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Sagori Mukhopadhyay
- Division of Neonatology, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
- Center for Pediatric Clinical Effectiveness, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Scott G Daniel
- Division of Gastroenterology, Hepatology, and Nutrition, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
| | - Hongzhe Li
- Center for Clinical Epidemiology and Biostatistics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Kyle Bittinger
- Division of Gastroenterology, Hepatology, and Nutrition, Children's Hospital of Philadelphia, Philadelphia, Pennsylvania, USA
- Department of Pediatrics, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| |
Collapse
|
40
|
Farnetano M, Carucci L, Coppola S, Oglio F, Masino A, Cozzolino M, Nocerino R, Berni Canani R. Gut microbiome features in pediatric food allergy: a scoping review. FRONTIERS IN ALLERGY 2024; 5:1438252. [PMID: 39386092 PMCID: PMC11461474 DOI: 10.3389/falgy.2024.1438252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2024] [Accepted: 08/13/2024] [Indexed: 10/12/2024] Open
Abstract
Increasing evidence suggests that alterations in the gut microbiome (GM) play a pivotal role in the pathogenesis of pediatric food allergy (FA). This scoping review analyzes the current evidence on GM features associated with pediatric FAs and highlights the importance of the GM as a potential target of intervention for preventing and treating this common condition in the pediatric age. Following the Preferred Reporting Items for Systematic Reviews and Meta-Analysis guidelines, we searched PubMed and Embase using the keywords (gut microbiome OR dysbiosis OR gut microbiota OR microbiome signatures) AND (food allergy OR IgE-mediated food allergy OR food protein-induced allergic proctocolitis OR food protein-induced enterocolitis OR non-IgE food allergy OR cow milk allergy OR hen egg allergy OR peanut allergy OR fish allergy OR shellfish allergy OR tree nut allergy OR soy allergy OR wheat allergy OR rice allergy OR food sensitization). We included 34 studies reporting alterations in the GM in children affected by FA compared with healthy controls. The GM in pediatric FAs is characterized by a higher abundance of harmful microorganisms (e.g., Enterobacteriaceae, Clostridium sensu stricto, Ruminococcus gnavus, and Blautia spp.) and lower abundance of beneficial bacteria (e.g., Bifidobacteriaceae, Lactobacillaceae, some Bacteroides species). Moreover, we provide an overview of the mechanisms of action elicited by these bacterial species in regulating immune tolerance and of the main environmental factors that can modulate the composition and function of the GM in early life. Altogether, these data improve our knowledge of the pathogenesis of FA and can open the way to innovative diagnostic, preventive, and therapeutic strategies for managing these conditions.
Collapse
Affiliation(s)
- Margherita Farnetano
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
| | - Laura Carucci
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
- ImmunoNutritionLab at the CEINGE Advanced Biotechnologies Research Center, University of Naples Federico II, Naples, Italy
| | - Serena Coppola
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
- ImmunoNutritionLab at the CEINGE Advanced Biotechnologies Research Center, University of Naples Federico II, Naples, Italy
| | - Franca Oglio
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
- ImmunoNutritionLab at the CEINGE Advanced Biotechnologies Research Center, University of Naples Federico II, Naples, Italy
| | - Antonio Masino
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
- ImmunoNutritionLab at the CEINGE Advanced Biotechnologies Research Center, University of Naples Federico II, Naples, Italy
| | - Marica Cozzolino
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
- ImmunoNutritionLab at the CEINGE Advanced Biotechnologies Research Center, University of Naples Federico II, Naples, Italy
| | - Rita Nocerino
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
- ImmunoNutritionLab at the CEINGE Advanced Biotechnologies Research Center, University of Naples Federico II, Naples, Italy
- Department of Biomedicine and Prevention, University of Rome “Tor Vergata”, Rome, Italy
| | - Roberto Berni Canani
- Department of Translational Medical Science, University of Naples Federico II, Naples, Italy
- ImmunoNutritionLab at the CEINGE Advanced Biotechnologies Research Center, University of Naples Federico II, Naples, Italy
- Task Force on Microbiome Studies, University of Naples Federico II, Naples, Italy
- European Laboratory for the Investigation of Food-Induced Diseases, University of Naples Federico II, Naples, Italy
| |
Collapse
|
41
|
Dong H, Li R, Zhao N, Dadhania DM, Suthanthiran M, Lee JR, Ling W. Antibiotic subclasses differentially perturb the gut microbiota in kidney transplant recipients. FRONTIERS IN TRANSPLANTATION 2024; 3:1400067. [PMID: 39371270 PMCID: PMC11451434 DOI: 10.3389/frtra.2024.1400067] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 03/13/2024] [Accepted: 08/27/2024] [Indexed: 10/08/2024]
Abstract
Introduction The impact of antibiotics on the gut microbiota in kidney transplant recipients is not well characterized. In this study, we determine the impact of different subclasses of antibiotics on the gut microbiota in a cohort of 168 kidney transplant recipients. Methods Gut microbiome profiling was performed on 510 fecal specimens using 16S rRNA gene sequencing of the V4-V5 hypervariable region. We classified fecal specimens by antibiotic exposure into 5 categories: Beta-lactam, Fluoroquinolone (FQ), Beta-lactam & FQ Group, Other Antibiotics, and No Antibiotic (No Abx). Mixed-effects regression models were utilized to identify changes in microbial diversity and in the centered log-ratio (CLR) transformed abundance of genera while adjusting for important covariates. Results Antibiotic administration was associated with a significant decrease in the Shannon alpha diversity index, a decreased abundance of 11 taxa including Eubacterium and Ruminococcus, and an increased abundance of 16 taxa including Enterococcus and Staphylococcus. Exposure to Beta-lactam antibiotics was associated with an increased abundance of 10 taxa including Enterococcus and a decreased abundance of 5 taxa including Eubacterium while exposure to FQ antibiotics was associated with an increased abundance of 3 taxa and a decreased abundance of 4 taxa including Ruminococcus. Conclusions Beta-lactam antibiotics and FQ antibiotics have a profound impact on the gut microbiota in kidney transplant recipients. Given the link of the gut microbiota to infectious complications, antibiotic associated changes in the microbiota may lead to an increased risk for further infections.
Collapse
Affiliation(s)
- Hanbo Dong
- Division of Biostatistics, Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, United States
| | - Runzhe Li
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Ni Zhao
- Department of Biostatistics, Johns Hopkins Bloomberg School of Public Health, Baltimore, MD, United States
| | - Darshana M. Dadhania
- Division of Nephrology and Hypertension, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
- Department of Transplantation Medicine, New York Presbyterian Hospital–Weill Cornell Medical Center, New York, NY, United States
| | - Manikkam Suthanthiran
- Division of Nephrology and Hypertension, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
- Department of Transplantation Medicine, New York Presbyterian Hospital–Weill Cornell Medical Center, New York, NY, United States
| | - John R. Lee
- Division of Nephrology and Hypertension, Department of Medicine, Weill Cornell Medicine, New York, NY, United States
- Department of Transplantation Medicine, New York Presbyterian Hospital–Weill Cornell Medical Center, New York, NY, United States
| | - Wodan Ling
- Division of Biostatistics, Department of Population Health Sciences, Weill Cornell Medicine, New York, NY, United States
| |
Collapse
|
42
|
Demirturk M, Cinar MS, Avci FY. The immune interactions of gut glycans and microbiota in health and disease. Mol Microbiol 2024; 122:313-330. [PMID: 38703041 DOI: 10.1111/mmi.15267] [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: 10/13/2023] [Revised: 04/11/2024] [Accepted: 04/12/2024] [Indexed: 05/06/2024]
Abstract
The human digestive system harbors a vast diversity of commensal bacteria and maintains a symbiotic relationship with them. However, imbalances in the gut microbiota accompany various diseases, such as inflammatory bowel diseases (IBDs) and colorectal cancers (CRCs), which significantly impact the well-being of populations globally. Glycosylation of the mucus layer is a crucial factor that plays a critical role in maintaining the homeostatic environment in the gut. This review delves into how the gut microbiota, immune cells, and gut mucus layer work together to establish a balanced gut environment. Specifically, the role of glycosylation in regulating immune cell responses and mucus metabolism in this process is examined.
Collapse
Affiliation(s)
- Mahmut Demirturk
- Department of Biochemistry, Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Mukaddes Sena Cinar
- Department of Biochemistry, Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, USA
| | - Fikri Y Avci
- Department of Biochemistry, Emory Vaccine Center, Emory University School of Medicine, Atlanta, Georgia, USA
| |
Collapse
|
43
|
Ullah H, Ali M, Ma R, Alioui Y, Ali S, Ilyas M, Rahman MU, Ahmed Farooqui N, Siddiqui NZ, Xin Y, Wang L. Polysaccharides derived from Deglet Noor dates modulate amoxicillin-induced dysbiosis and enhance intestinal barrier function. J Funct Foods 2024; 120:106350. [DOI: 10.1016/j.jff.2024.106350] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2025] Open
|
44
|
Light SH, Nagler CR. Regulation of immune responses to food by commensal microbes. Immunol Rev 2024; 326:203-218. [PMID: 39285525 PMCID: PMC11472335 DOI: 10.1111/imr.13396] [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: 09/28/2024]
Abstract
The increasing prevalence of immune-mediated non-communicable chronic diseases, such as food allergies, has prompted a deeper investigation into the role of the gut microbiome in modulating immune responses. Here, we explore the complex interactions between commensal microbes and the host immune system, highlighting the critical role of gut bacteria in maintaining immune homeostasis. We examine how modern lifestyle practices and environmental factors have disrupted co-evolved host-microbe interactions and discuss how changes in microbiome composition impact epithelial barrier function, responses to food allergens, and susceptibility to allergic diseases. Finally, we examine the potential of bioengineered microbiome-based therapies, and live biotherapeutic products, for reestablishing immune homeostasis to prevent or treat food allergies.
Collapse
Affiliation(s)
- Samuel H. Light
- Department of Microbiology, University of Chicago, Chicago IL, 60637
| | - Cathryn R. Nagler
- Department of Pathology, University of Chicago, Chicago IL, 60637
- Department of Biological Sciences Division, Pritzker School of Molecular Engineering, University of Chicago, Chicago IL, 60637
| |
Collapse
|
45
|
Lee JW, Park SJ, Park YJ, Jeong S, Song J, Kim HJ, Chang J, Kim KH, Kim JS, Oh YH, Cho Y, Park SM. Association between antibiotics use and osteoporotic fracture risk: a nationally representative retrospective cohort study. Arch Osteoporos 2024; 19:81. [PMID: 39212806 PMCID: PMC11364706 DOI: 10.1007/s11657-024-01438-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Accepted: 08/12/2024] [Indexed: 09/04/2024]
Abstract
This population-based retrospective cohort study aimed to estimate the association between antibiotic exposure and osteoporotic fracture risk. Long-term antibiotic use was associated with the risk of osteoporotic fracture. An increase in the number of antibiotic classes prescribed may also be associated with an increased osteoporotic fracture risk. PURPOSE This study aims to examine the association between antibiotic usage and osteoporotic fractures in a large cohort of Korean adults, with a specific focus on the duration of antibiotic exposure and the number of antibiotic classes used. METHODS This retrospective cohort study from the National Health Insurance Service-National Health Screening Cohort (NHIS-HEALS) database from January 1, 2002, to December 31, 2019, included 167,370 Korean adults aged 50 years or older (mean [SD] age, 59.3 [7.82] years; 65,425 [39.09%] women). The cumulative antibiotic prescription days and the classes of antibiotics prescribed between 2004 and 2008 were exposure variables, respectively. The main outcome was a newly diagnosed osteoporotic fracture during follow-up. Cox proportional hazard regression was used to determine the adjusted hazard ratios (aHRs) and 95% confidence intervals (CIs) for the incident osteoporotic fractures associated with antibiotic exposure. RESULTS The antibiotic user group with 91 days had a higher risk of osteoporotic fracture in comparison to the antibiotic non-user group (aHR, 1.12; 95% CI, 1.03-1.21). Additionally, those who used more than four different antibiotic classes had an elevated risk of osteoporotic fracture compared to the non-user group (aHR, 1.10; 95% CI, 1.02-1.18). CONCLUSION This extensive population-based cohort study conducted on a large population has identified an association between the utilization of antibiotics and an elevated risk of osteoporotic fractures. The cumulative days exposed to antibiotics and osteoporotic fractures may be positively associated.
Collapse
Affiliation(s)
- Ji Won Lee
- Department of Biomedical Sciences, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
- Department of Health Convergence, Ewha Womans University, Seoul, South Korea
| | - Sun Jae Park
- Department of Biomedical Sciences, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Young Jun Park
- Medical Research Center, Genomic Medicine Institute, Seoul National University, Seoul, South Korea
| | - Seogsong Jeong
- Department of Biomedical Informatics, Korea University College of Medicine, Seoul, South Korea
| | - Jihun Song
- Department of Biomedical Sciences, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Hye Jun Kim
- Department of Biomedical Sciences, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Jooyoung Chang
- Department of Biomedical Sciences, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
| | - Kyae Hyung Kim
- Department of Family Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea
- Public Healthcare Center, Seoul National University Hospital, Seoul, South Korea
| | - Ji Soo Kim
- International Healthcare Center, Seoul National University Bundang Hospital, Seongnam, South Korea
| | - Yun Hwan Oh
- Department of Family Medicine, Chung-Ang University Gwangmyeong Hospital, Chung-Ang University College of Medicine, Gwangmyeong-Si, South Korea
| | - Yoosun Cho
- Total Healthcare Center, Kangbuk Samsung Hospital, Sungkyunkwan University School of Medicine, Seoul, South Korea
| | - Sang Min Park
- Department of Biomedical Sciences, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea.
- Department of Family Medicine, Seoul National University Hospital, Seoul National University College of Medicine, Seoul, South Korea.
| |
Collapse
|
46
|
Beekman CN, Penumutchu S, Peterson R, Han G, Belenky M, Hasan MH, Belenky A, Beura LK, Belenky P. Spatial analysis of murine microbiota and bile acid metabolism during amoxicillin treatment. Cell Rep 2024; 43:114572. [PMID: 39116202 DOI: 10.1016/j.celrep.2024.114572] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2024] [Revised: 06/05/2024] [Accepted: 07/17/2024] [Indexed: 08/10/2024] Open
Abstract
Antibiotics cause collateral damage to resident microbes that is associated with various health risks. To date, studies have largely focused on the impacts of antibiotics on large intestinal and fecal microbiota. Here, we employ a gastrointestinal (GI) tract-wide integrated multiomic approach to show that amoxicillin (AMX) treatment reduces bacterial abundance, bile salt hydrolase activity, and unconjugated bile acids in the small intestine (SI). Losses of fatty acids (FAs) and increases in acylcarnitines in the large intestine (LI) correspond with spatially distinct expansions of Proteobacteria. Parasutterella excrementihominis engage in FA biosynthesis in the SI, while multiple Klebsiella species employ FA oxidation during expansion in the LI. We subsequently demonstrate that restoration of unconjugated bile acids can mitigate losses of commensals in the LI while also inhibiting the expansion of Proteobacteria during AMX treatment. These results suggest that the depletion of bile acids and lipids may contribute to AMX-induced dysbiosis in the lower GI tract.
Collapse
Affiliation(s)
- Chapman N Beekman
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA.
| | - Swathi Penumutchu
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Rachel Peterson
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Geongoo Han
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Marina Belenky
- Felicitex Therapeutics Inc., 27 Strathmore Road, Natick, MA 01760, USA
| | - Mohammad H Hasan
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Alexei Belenky
- Felicitex Therapeutics Inc., 27 Strathmore Road, Natick, MA 01760, USA
| | - Lalit K Beura
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA
| | - Peter Belenky
- Department of Molecular Microbiology and Immunology, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
47
|
Liang J, Wang S, Kou S, Chen C, Zhang W, Nie C. Clostridium butyricum Prevents Diarrhea Incidence in Weaned Piglets Induced by Escherichia coli K88 through Rectal Bacteria-Host Metabolic Cross-Talk. Animals (Basel) 2024; 14:2287. [PMID: 39199821 PMCID: PMC11350811 DOI: 10.3390/ani14162287] [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: 06/04/2024] [Revised: 07/15/2024] [Accepted: 07/30/2024] [Indexed: 09/01/2024] Open
Abstract
This study aimed to evaluate the effects of Clostridium butyricum (C. butyricum) on the prevention of the diarrhea rates and growth performances of weaned piglets induced by Escherichia coli K88 (E. coli K88). Twenty-four weaned piglets (6.92 ± 0.11 kg) were randomly assigned to one of three treatment groups for a period of 21 days. Each group consisted of eight pigs, with each pig being housed in an individual pen. Group I received the control diet along with normal saline, Group II received the control diet along with E. coli K88, and Group III received the control diet supplemented with 5 × 108 CFU/kg of C. butyricum and E. coli K88. We examined alterations in rectal microbiota and metabolites, analyzed the incidence of diarrhea, and investigated the interactions between microbiota and metabolites through the application of Illumina MiSeq sequencing and liquid chromatography-mass spectrometry. The results showed that, from days 14 to 21, the diarrhea incidence in Group III decreased significantly by 83.29% compared to Group II (p < 0.05). Over the entire experimental duration, the average daily feed intake of Group III decreased significantly by 11.13% compared to Group I (p < 0.05), while the diarrhea incidence in Group III decreased by 71.46% compared to Group II (p < 0.05). The predominant microbial flora in the rectum consisted of Firmicutes (57.32%), Bacteroidetes (41.03%), and Proteobacteria (0.66%). Administering E. coli K88 orally can elevate the relative abundance of Megasphaera (p < 0.05). Conversely, the supplementation of C. butyricum in the diet reduced the relative abundance of Megasphaera (p < 0.05), while increasing the relative abundance of unclassified_f_Lachnospiraceae (p < 0.05). Rectal metabolomics analysis revealed that supplementing C. butyricum in the feed significantly altered the amino acids and fatty acids of the piglets infected with E. coli K88 (p < 0.05). The correlation analysis showed that the occurrence of diarrhea was inversely related to adipic acid (p < 0.05) and positively associated with (5-hydroxyindol-3-YL) acetic acid and L-aspartic acid (p < 0.05). Prevotella_1 exhibited a negative correlation with octadecanoic acid (p < 0.05). Prevotellaceae_UCG-005 showed a negative correlation with (5-hydroxyindol-3-YL) acetic acid (p < 0.05). The findings from this research study aid in probiotic development and the enhancement of healthy growth in weaned piglets.
Collapse
Affiliation(s)
- Jing Liang
- College of Life Science, Yulin University, Yulin 719000, China; (J.L.); (S.W.)
- College of Animal Science and Technology, Shihezi University, Shihezi 832003, China; (S.K.); (C.C.)
| | - Sihu Wang
- College of Life Science, Yulin University, Yulin 719000, China; (J.L.); (S.W.)
- College of Animal Science and Technology, Northwest A&F University, Xianyang 712100, China
| | - Shasha Kou
- College of Animal Science and Technology, Shihezi University, Shihezi 832003, China; (S.K.); (C.C.)
| | - Cheng Chen
- College of Animal Science and Technology, Shihezi University, Shihezi 832003, China; (S.K.); (C.C.)
| | - Wenju Zhang
- College of Animal Science and Technology, Shihezi University, Shihezi 832003, China; (S.K.); (C.C.)
| | - Cunxi Nie
- College of Animal Science and Technology, Shihezi University, Shihezi 832003, China; (S.K.); (C.C.)
| |
Collapse
|
48
|
Yang J, Gould TJ, Jeon B, Ji Y. Broad-Spectrum Antibacterial Activity of Antioxidant Octyl Gallate and Its Impact on Gut Microbiome. Antibiotics (Basel) 2024; 13:731. [PMID: 39200031 PMCID: PMC11350663 DOI: 10.3390/antibiotics13080731] [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: 07/04/2024] [Revised: 08/01/2024] [Accepted: 08/03/2024] [Indexed: 09/01/2024] Open
Abstract
In this study, we investigated the antibacterial activity of octyl gallate (OG), an antioxidant food additive, against both Gram-positive and Gram-negative bacterial pathogens. OG demonstrated robust bactericidal activity against Gram-positive bacterial pathogens with minimum inhibitory concentrations (MIC) of 4 to 8 µg/mL and minimum bactericidal concentrations (MBC) of 8 to 16 µg/mL in vitro. However, OG exhibited limited antibacterial activity against Gram-negative bacteria, including E. coli, although it could inhibit bacterial growth in vitro. Importantly, OG administration in mice altered the fecal microbiome, significantly reducing microbial diversity, modifying community structure, and increasing the abundance of beneficial bacteria. Additionally, OG displayed low cytotoxicity and hemolytic activity. These findings suggest that OG could be developed as a novel antibacterial agent, particularly against multi-drug-resistant MRSA. Our results provide new insights into the therapeutic potential of OG in modulating the gut microbiome, especially in conditions associated with microbial imbalance, while ensuring food safety.
Collapse
Affiliation(s)
- Junshu Yang
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108, USA
| | - Trevor J. Gould
- Minnesota Supercomputing Institute, University of Minnesota, Minneapolis, MN 55455, USA
| | - Byeonghwa Jeon
- Division of Environmental Health Sciences, School of Public Health, University of Minnesota, Saint Paul, MN 55108, USA
| | - Yinduo Ji
- Department of Veterinary and Biomedical Sciences, College of Veterinary Medicine, University of Minnesota, Saint Paul, MN 55108, USA
| |
Collapse
|
49
|
Fernandes C, Miranda MCC, Roque CR, Paguada ALP, Mota CAR, Florêncio KGD, Pereira AF, Wong DVT, Oriá RB, Lima-Júnior RCP. Is There an Interplay between Environmental Factors, Microbiota Imbalance, and Cancer Chemotherapy-Associated Intestinal Mucositis? Pharmaceuticals (Basel) 2024; 17:1020. [PMID: 39204125 PMCID: PMC11357004 DOI: 10.3390/ph17081020] [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: 05/29/2024] [Revised: 07/24/2024] [Accepted: 08/01/2024] [Indexed: 09/03/2024] Open
Abstract
Interindividual variation in drug efficacy and toxicity is a significant problem, potentially leading to adverse clinical and economic public health outcomes. While pharmacogenetics and pharmacogenomics have long been considered the primary causes of such heterogeneous responses, pharmacomicrobiomics has recently gained attention. The microbiome, a community of microorganisms living in or on the human body, is a critical determinant of drug response and toxicity. Factors such as diet, lifestyle, exposure to xenobiotics, antibiotics use, illness, and genetics can influence the composition of the microbiota. Changes in the intestinal microbiota are particularly influential in drug responsiveness, especially in cancer chemotherapy. The microbiota can modulate an individual's response to a drug, affecting its bioavailability, clinical effect, and toxicity, affecting treatment outcomes and patient quality of life. For instance, the microbiota can convert drugs into active or toxic metabolites, influencing their efficacy and side effects. Alternatively, chemotherapy can also alter the microbiota, creating a bidirectional interplay. Probiotics have shown promise in modulating the microbiome and ameliorating chemotherapy side effects, highlighting the potential for microbiota-targeted interventions in improving cancer treatment outcomes. This opinion paper addresses how environmental factors and chemotherapy-induced dysbiosis impact cancer chemotherapy gastrointestinal toxicity.
Collapse
Affiliation(s)
- Camila Fernandes
- Department of Physiology and Pharmacology, and Drug Research and Development Center (NPDM), Faculty of Medicine, Federal University of Ceara, Rua Cel Nunes de Melo, 1000, Fortaleza 60430-270, Brazil; (C.F.); (A.L.P.P.); (C.A.R.M.); (K.G.D.F.); (A.F.P.); (D.V.T.W.)
| | | | - Cássia Rodrigues Roque
- Laboratory of Tissue Healing, Ontogeny, and Nutrition, Department of Morphology, and Institute of Biomedicine, Faculty of Medicine, Federal University of Ceara, Fortaleza 60430-170, Brazil; (C.R.R.); (R.B.O.)
| | - Ana Lizeth Padilla Paguada
- Department of Physiology and Pharmacology, and Drug Research and Development Center (NPDM), Faculty of Medicine, Federal University of Ceara, Rua Cel Nunes de Melo, 1000, Fortaleza 60430-270, Brazil; (C.F.); (A.L.P.P.); (C.A.R.M.); (K.G.D.F.); (A.F.P.); (D.V.T.W.)
| | - Carlos Adrian Rodrigues Mota
- Department of Physiology and Pharmacology, and Drug Research and Development Center (NPDM), Faculty of Medicine, Federal University of Ceara, Rua Cel Nunes de Melo, 1000, Fortaleza 60430-270, Brazil; (C.F.); (A.L.P.P.); (C.A.R.M.); (K.G.D.F.); (A.F.P.); (D.V.T.W.)
| | - Katharine Gurgel Dias Florêncio
- Department of Physiology and Pharmacology, and Drug Research and Development Center (NPDM), Faculty of Medicine, Federal University of Ceara, Rua Cel Nunes de Melo, 1000, Fortaleza 60430-270, Brazil; (C.F.); (A.L.P.P.); (C.A.R.M.); (K.G.D.F.); (A.F.P.); (D.V.T.W.)
| | - Anamaria Falcão Pereira
- Department of Physiology and Pharmacology, and Drug Research and Development Center (NPDM), Faculty of Medicine, Federal University of Ceara, Rua Cel Nunes de Melo, 1000, Fortaleza 60430-270, Brazil; (C.F.); (A.L.P.P.); (C.A.R.M.); (K.G.D.F.); (A.F.P.); (D.V.T.W.)
| | - Deysi Viviana Tenazoa Wong
- Department of Physiology and Pharmacology, and Drug Research and Development Center (NPDM), Faculty of Medicine, Federal University of Ceara, Rua Cel Nunes de Melo, 1000, Fortaleza 60430-270, Brazil; (C.F.); (A.L.P.P.); (C.A.R.M.); (K.G.D.F.); (A.F.P.); (D.V.T.W.)
| | - Reinaldo Barreto Oriá
- Laboratory of Tissue Healing, Ontogeny, and Nutrition, Department of Morphology, and Institute of Biomedicine, Faculty of Medicine, Federal University of Ceara, Fortaleza 60430-170, Brazil; (C.R.R.); (R.B.O.)
| | - Roberto César Pereira Lima-Júnior
- Department of Physiology and Pharmacology, and Drug Research and Development Center (NPDM), Faculty of Medicine, Federal University of Ceara, Rua Cel Nunes de Melo, 1000, Fortaleza 60430-270, Brazil; (C.F.); (A.L.P.P.); (C.A.R.M.); (K.G.D.F.); (A.F.P.); (D.V.T.W.)
| |
Collapse
|
50
|
Doré J, Sansonetti PJ. [The human microbiome: 340 years of history, 140 years of interrogations, technological innovations and emergence of "microbial medicine"]. Med Sci (Paris) 2024; 40:654-660. [PMID: 39303118 DOI: 10.1051/medsci/2024101] [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: 09/22/2024] Open
Abstract
For 350 years, we have known that the human body hosts microbes, then called "animalcules". For over a century, following the demonstration of the role of some of these microbes in diseases, questions have arisen about the role of the largely predominant ones colonizing human skin and mucous surfaces, particularly the rich microbial ecosystem of the intestine, the gut microbiota. From the invention of germ-free life - axenism - which experimentally validated the human-microbe symbiosis, resulting from a long coevolution, to the development of anaerobic culture methods, then to the invention of molecular diagnosis, deep sequencing opening up metagenomic and omics approaches in general, a remarkable race has taken place between technological innovations and conceptual advances. This race, beyond the exhaustive description of the microbiota in its intra- and inter-human diversity, and the essential symbiotic functions of the microbiome, has paved the way for a new field of medicine: microbial medicine.
Collapse
Affiliation(s)
- Joël Doré
- Université Paris-Saclay, INRAE, MetaGenoPolis, AgroParisTech, MICALIS, Jouy-en-Josas, France
| | | |
Collapse
|