51
|
Jia B, Baek JH, Lee JK, Sun Y, Kim KH, Jung JY, Jeon CO. Expanding the β-Lactamase Family in the Human Microbiome. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2403563. [PMID: 39447121 PMCID: PMC11633517 DOI: 10.1002/advs.202403563] [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: 04/05/2024] [Revised: 09/23/2024] [Indexed: 10/26/2024]
Abstract
β-lactams, the most common antibiotics globally, have resistance primarily determined by β-lactamases. Human microbiota and β-lactams influence mutually; however, β-lactamase variety and abundance in the human microbiome remain partially understood. This study aimed to elucidate the diversity, abundance, and substrate spectrum of β-lactamases. 1369 characterized β-lactamases and 16 204 putative sequences are collected from protein databases. Upon clustering analysis and biochemical assays, nine proteins exhibiting less than 35% identity to those previously characterized are confirmed as β-lactamases. These newly identified β-lactamases originated from eight distinct clusters comprising 1163 β-lactamases. Quantifying healthy participants (n = 2394) across 19 countries using functionally confirmed clusters revealed that Japan have the highest gut β-lactamase abundance (log2[reads per million (RPM)] = 6.52) and Fiji have the lowest (log2[RPM] = 2.31). The β-lactamase abundance is correlated with β-lactam consumption (R = 0.50, p = 0.029) and income (R = 0.51, p = 0.024). Comparing individuals with ailments with healthy participants, β-lactamase abundance in the gut is increased significantly in patients with colorectal cancer, cardiovascular diseases, breast cancer, and epilepsy. These outcomes provide insights into investigating antibiotic resistance, antibiotic stewardship, and gut microbiome-antibiotic interactions.
Collapse
Affiliation(s)
- Baolei Jia
- Xianghu LaboratoryHangzhou311231China
- Department of Life ScienceChung‐Ang UniversitySeoul06974Republic of Korea
| | - Ju Hye Baek
- Department of Life ScienceChung‐Ang UniversitySeoul06974Republic of Korea
| | - Jae Kyeong Lee
- Department of Life ScienceChung‐Ang UniversitySeoul06974Republic of Korea
| | - Ying Sun
- Department of Veterinary and Animal SciencesUniversity of CopenhagenCopenhagen1870Denmark
| | - Kyung Hyun Kim
- Department of Biological Sciences and BiotechnologyHannam UniversityDaejon34054Republic of Korea
| | - Ji Young Jung
- Microbial Research DepartmentNakdonggang National Institute of Biological ResourcesGyeongsangbuk‐do37242Republic of Korea
| | - Che Ok Jeon
- Department of Life ScienceChung‐Ang UniversitySeoul06974Republic of Korea
| |
Collapse
|
52
|
Nath S, Zilm P, Jamieson L, Ketagoda DHK, Kapellas K, Weyrich L. Characterising healthy Australian oral microbiomes for 'super donor' selection. J Dent 2024; 151:105435. [PMID: 39461582 DOI: 10.1016/j.jdent.2024.105435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 10/21/2024] [Accepted: 10/23/2024] [Indexed: 10/29/2024] Open
Abstract
OBJECTIVES Among healthy people, we understand very little about the sociodemographic, lifestyle, and dental hygiene behaviours that shape their oral microbiota. This study investigates how sociodemographic, lifestyle and dental hygiene behaviours shape oral microbiota diversity and composition in an Australian population to better inform healthy oral microbiota donors for Oral Microbiota Transplantation (OMT). METHODS The study comprised 93 healthy adults who underwent comprehensive oral examinations and questionnaires to assess their health status. Participants were excluded if they had any active systemic or oral disease. All completed a questionnaire containing information on socio-economic, lifestyle, behavioural, and oral health factors. Supragingival plaque was collected, and 16S ribosomal RNA (rRNA) amplicon sequencing was used to analyse microbial composition. Associations between the core microbiome, alpha- (within-sample), beta-diversity (between-sample) and an individual's co-variates were tested for statistical significance. A redundancy analysis (RDA), multivariate adonis, differential abundance and correlation analysis were performed to characterise which factors drive the variation in the healthy oral microbiome. RESULTS Streptococcus and Corynebacterium were the most prevalent and abundant genera among healthy Australians. The alpha and beta diversity were higher among unemployed non-Australian-born students who consumed low carbohydrates, fat, and sugar and had not visited the dentist for over 12 months. Additionally, beta diversity was significantly higher among daily flossers who abstained from fluoride treatment and had high salivary pH, although no single factor explained >4 % of the total variation (R2= 0.042). Alloprevotella, Lachnosporacea, and Parvimonas were significantly abundant among non-Australians who did not visit the dentist within a year. The RDA analysis revealed associations between microbiome composition and factors such as high carbohydrate, sugar, and fat consumption, low fibre intake, and regular dental checks among Australian-born individuals. CONCLUSION These findings indicate that alpha and beta diversity of the oral microbiome varied significantly with sociodemographic, lifestyle, and dietary factors, including non-Australian birthplaces, unemployment, diet, and infrequent dental visits. CLINICAL SIGNIFICANCE These findings underscore the importance of considering diverse sociodemographic, lifestyle, and dietary factors in oral health management. Before microbiome transplantations, clinicians should account for individual characteristics that may be beneficial for shaping and maintaining optimal oral microbiome diversity and health.
Collapse
Affiliation(s)
- Sonia Nath
- Adelaide Dental School, The University of Adelaide, Adelaide, SA, Australia.
| | - Peter Zilm
- Adelaide Dental School, The University of Adelaide, Adelaide, SA, Australia
| | - Lisa Jamieson
- Adelaide Dental School, The University of Adelaide, Adelaide, SA, Australia
| | | | - Kostas Kapellas
- Adelaide Dental School, The University of Adelaide, Adelaide, SA, Australia
| | - Laura Weyrich
- School of Biological Sciences, University of Adelaide, Adelaide, SA, Australia; Department of Anthropology and Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802, USA
| |
Collapse
|
53
|
Manus MB, Lucore J, Kuthyar S, Moy M, Savo Sardaro ML, Amato KR. Technical note: A biological anthropologist's guide for applying microbiome science to studies of human and non-human primates. AMERICAN JOURNAL OF BIOLOGICAL ANTHROPOLOGY 2024; 185:e25020. [PMID: 39222382 DOI: 10.1002/ajpa.25020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/27/2024] [Revised: 05/28/2024] [Accepted: 08/18/2024] [Indexed: 09/04/2024]
Abstract
A central goal of biological anthropology is connecting environmental variation to differences in host physiology, biology, health, and evolution. The microbiome represents a valuable pathway for studying how variation in host environments impacts health outcomes. While there are many resources for learning about methods related to microbiome sample collection, laboratory analyses, and genetic sequencing, there are fewer dedicated to helping researchers navigate the dense portfolio of bioinformatics and statistical approaches for analyzing microbiome data. Those that do exist are rarely related to questions in biological anthropology and instead are often focused on human biomedicine. To address this gap, we expand on existing tutorials and provide a "road map" to aid biological anthropologists in understanding, selecting, and deploying the data analysis and visualization methods that are most appropriate for their specific research questions. Leveraging an existing dataset of fecal samples and survey data collected from wild geladas living in Simien Mountains National Park in Ethiopia (Baniel et al., 2021), this paper guides researchers toward answering three questions related to variation in the gut microbiome across host and environmental factors. By providing explanations, examples, and a reproducible workflow for different analytic methods, we move beyond the theoretical benefits of considering the microbiome within anthropological research and instead present researchers with a guide for applying microbiome science to their work. This paper makes microbiome science more accessible to biological anthropologists and paves the way for continued research into the microbiome's role in the ecology, evolution, and health of human and non-human primates.
Collapse
Affiliation(s)
- Melissa B Manus
- Department of Anthropology, University of Texas at San Antonio, San Antonio, Texas, USA
- Department of Anthropology, Northwestern University, Evanston, Illinois, USA
| | - Jordan Lucore
- Department of Anthropology, University of Michigan, Ann Arbor, Michigan, USA
| | - Sahana Kuthyar
- Division of Biological Sciences, University of California San Diego, La Jolla, California, USA
| | - Madelyn Moy
- Department of Anthropology, Northwestern University, Evanston, Illinois, USA
| | - Maria Luisa Savo Sardaro
- Department of Anthropology, Northwestern University, Evanston, Illinois, USA
- Department of Human Science and Promotion of the Quality of Life, University of San Raffaele, Rome, Italy
| | - Katherine R Amato
- Department of Anthropology, Northwestern University, Evanston, Illinois, USA
| |
Collapse
|
54
|
Sheng Y, Wang J, Gao Y, Peng Y, Li X, Huang W, Zhou H, Liu R, Zhang W. Combined analysis of cross-population healthy adult human microbiome reveals consistent differences in gut microbial characteristics between Western and non-Western countries. Comput Struct Biotechnol J 2024; 23:87-95. [PMID: 38116074 PMCID: PMC10730331 DOI: 10.1016/j.csbj.2023.11.047] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2023] [Revised: 11/23/2023] [Accepted: 11/24/2023] [Indexed: 12/21/2023] Open
Abstract
Despite extensive research on the gut microbiome of healthy individuals from a single country, there are still a limited number of population-level comparative studies. Moreover, the sequencing approach used in most related studies involves 16 S ribosomal RNA (rRNA) sequencing with a limited resolution, which cannot provide detailed functional profiles. In the present study, we applied a combined analysis approach to analyze whole metagenomic shotgun sequencing data from 2035 healthy adult samples from six countries across four continents. Analysis of core species revealed that 13 species were present in more than 90 % of all investigated individuals, the majority of which produced short-chain fatty acids (SCFA)-producing bacteria. Our analysis revealed consistently significant differences in gut microbial species and pathways between Western and non-Western countries, such as Escherichia coli and the relation of MetaCyc pathways to the TCA cycle. Specific changes in microbial species and pathways are potentially related to lifestyle and diet. Furthermore, we identified several noteworthy microbial species and pathways that exhibit distinct characteristics specific to China. Interestingly, we observed that China (CHN) was more similar to the United States (USA) and United Kingdom (GBR) in terms of the taxonomic and functional composition of the gut microbiome than India (IND) and Madagascar (MDG), which were more similar to the China (CHN) diet. The current study identified consistent microbial features associated with population and geography, which will inspire further clinical translations that consider paying attention to differences in microbiota backgrounds and confounding factors.
Collapse
Affiliation(s)
- Yanghao Sheng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Central South University, Changsha, China
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Jue Wang
- Department of Pharmacy, Xiangya Hospital, Central South University, Changsha, China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Yongchao Gao
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Yilei Peng
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Central South University, Changsha, China
| | - Xiong Li
- Center for Clinical Precision Pharmacy, School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
| | - Weihua Huang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Central South University, Changsha, China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Honghao Zhou
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Central South University, Changsha, China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Rong Liu
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Central South University, Changsha, China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Wei Zhang
- Department of Clinical Pharmacology, Xiangya Hospital, Central South University, Changsha, China
- Institute of Clinical Pharmacology, Central South University, Changsha, China
- Institute for Rational and Safe Medication Practices, National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
- Center for Clinical Precision Pharmacy, School of Clinical Pharmacy, Guangdong Pharmaceutical University, Guangzhou, China
- The First Affifiliated Hospital of Shantou University Medical College, Shantou, China
- Key Laboratory of Clinical Precision Pharmacy of Guangdong Higher Education, Institutes, The First Affiliated Hospital, Guangdong Pharmaceutical University, Guangzhou, China
| |
Collapse
|
55
|
Liu R, Wang Y, Cheng D. Micro-DeMix: a mixture beta-multinomial model for investigating the heterogeneity of the stool microbiome compositions. Bioinformatics 2024; 40:btae667. [PMID: 39563467 PMCID: PMC11645251 DOI: 10.1093/bioinformatics/btae667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/15/2023] [Revised: 10/29/2024] [Accepted: 11/15/2024] [Indexed: 11/21/2024] Open
Abstract
MOTIVATION Extensive research has uncovered the critical role of the human gut microbiome in various aspects of health, including metabolism, nutrition, physiology, and immune function. Fecal microbiota is often used as a proxy for understanding the gut microbiome, but it represents an aggregate view, overlooking spatial variations across different gastrointestinal (GI) locations. Emerging studies with spatial microbiome data collected from specific GI regions offer a unique opportunity to better understand the spatial composition of the stool microbiome. RESULTS We introduce Micro-DeMix, a mixture beta-multinomial model that deconvolutes the fecal microbiome at the compositional level by integrating stool samples with spatial microbiome data. Micro-DeMix facilitates the comparison of microbial compositions across different GI regions within the stool microbiome through a hypothesis-testing framework. We demonstrate the effectiveness and efficiency of Micro-DeMix using multiple simulated datasets and the inflammatory bowel disease data from the NIH Integrative Human Microbiome Project. AVAILABILITY AND IMPLEMENTATION The R package is available at https://github.com/liuruoqian/MicroDemix.
Collapse
Affiliation(s)
- Ruoqian Liu
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ 85251, United States
| | - Yue Wang
- Department of Biostatistics and Informatics, Colorado School of Public Health, Aurora, CO 80045, United States
| | - Dan Cheng
- School of Mathematical and Statistical Sciences, Arizona State University, Tempe, AZ 85251, United States
| |
Collapse
|
56
|
Shao L, Zhang B, Song Y, Lyu Z, Zhang W, Yang W, Fu J, Li J, Shi J. Mucosal Hub Bacteria as Potential Targets for Improving High-Fat Diet-Related Intestinal Barrier Injury. THE CANADIAN JOURNAL OF INFECTIOUS DISEASES & MEDICAL MICROBIOLOGY = JOURNAL CANADIEN DES MALADIES INFECTIEUSES ET DE LA MICROBIOLOGIE MEDICALE 2024; 2024:3652740. [PMID: 39634324 PMCID: PMC11617042 DOI: 10.1155/cjid/3652740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Accepted: 11/13/2024] [Indexed: 12/07/2024]
Abstract
Background: Intestinal barrier injury contributes to multiple diseases such as obesity and diabetes, whereas no treatment options are available. Methods: Due to close interactions between mucosal microbiome and intestinal barrier, we evaluated the potential of mucosal bacteria in providing targets for high-fat diet (HFD)-related intestinal barrier injury. Whole-genome metagenomics was used to evaluate mucosal microbiome, while intestinal barrier injury was estimated using serum LPS, FITC-dextran intensity, and ZO-1 protein. Results: We found that HFD induced significant fat accumulation in epididymal tissue at weeks 4 and 12, while ALT, LDL, and TC increased at week 12. Intestinal barrier injury was confirmed by elevated serum LPS at both weeks, upregulated FITC-dextran intensity, and decreased ZO-1 protein at week 12. Fourteen species such as Phocaeicola vulgatus differed in HFD-fed mice. The co-occurrence network of mucosal microbiome shifted from scale-free graph in controls to nearly random graph in HFD-fed mice. Besides, 10 hub bacteria especially Bacteroides ovatus decreased drastically in both mucosal and fecal samples of HFD-fed mice, correlated with intestinal permeability, ALT, and KEGG pathways such as "Mitochondrial biogenesis" and "metabolism". Moreover, Bacteroides ovatus has been confirmed to improve intestinal barrier function in a recent study. Conclusion: Mucosal hub bacteria can provide potential targets for improving HFD-related intestinal barrier function.
Collapse
Affiliation(s)
- Li Shao
- School of Clinical Medicine, Hangzhou Normal University, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 311121, Zhejiang, China
- Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, Hangzhou 310015, Zhejiang, China
- Institute of Translational Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 311121, Zhejiang, China
| | - Binbin Zhang
- School of Clinical Medicine, Hangzhou Normal University, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 311121, Zhejiang, China
- Institute of Translational Medicine, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 311121, Zhejiang, China
| | - Yu Song
- Department of Hepatology, 2nd Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| | - Zhe Lyu
- Department of Stomatology, The First Affiliated Hospital, College of Medicine, Zhejiang University, Hangzhou 310003, Zhejiang, China
| | - Weishi Zhang
- Department of Otolaryngology, Affiliated Hospital 2 of Nantong University, Nantong 226001, Jiangsu, China
| | - Wenjun Yang
- School of Clinical Medicine, Hangzhou Normal University, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 311121, Zhejiang, China
- Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, Hangzhou 310015, Zhejiang, China
| | - Jinlong Fu
- School of Clinical Medicine, Hangzhou Normal University, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 311121, Zhejiang, China
- Department of Gastroenterology and Hepatology, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 310015, Zhejiang, China
| | - Jie Li
- Department of Infectious Diseases, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, Jiangsu, China
| | - Junping Shi
- School of Clinical Medicine, Hangzhou Normal University, The Affiliated Hospital of Hangzhou Normal University, Hangzhou 311121, Zhejiang, China
- Institute of Hepatology and Metabolic Diseases, Hangzhou Normal University, Hangzhou 310015, Zhejiang, China
- Department of Hepatology, 2nd Affiliated Hospital of Zhejiang Chinese Medical University, Hangzhou 310053, Zhejiang, China
| |
Collapse
|
57
|
Sall I, Foxall R, Felth L, Maret S, Rosa Z, Gaur A, Calawa J, Pavlik N, Whistler JL, Whistler CA. Gut dysbiosis was inevitable, but tolerance was not: temporal responses of the murine microbiota that maintain its capacity for butyrate production correlate with sustained antinociception to chronic morphine. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.15.589671. [PMID: 38659831 PMCID: PMC11042308 DOI: 10.1101/2024.04.15.589671] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
The therapeutic benefits of opioids are compromised by the development of analgesic tolerance, which necessitates higher dosing for pain management thereby increasing the liability for drug dependence and addiction. Rodent models indicate opposing roles of the gut microbiota in tolerance: morphine-induced gut dysbiosis exacerbates tolerance, whereas probiotics ameliorate tolerance. Not all individuals develop tolerance which could be influenced by differences in microbiota, and yet no study design has capitalized upon this natural variation. We leveraged natural behavioral variation in a murine model of voluntary oral morphine self-administration to elucidate the mechanisms by which microbiota influences tolerance. Although all mice shared similar morphine-driven microbiota changes that largely masked informative associations with variability in tolerance, our high-resolution temporal analyses revealed a divergence in the progression of dysbiosis that best explained sustained antinociception. Mice that did not develop tolerance maintained a higher capacity for production of the short-chain fatty acid (SCFA) butyrate known to bolster intestinal barriers and promote neuronal homeostasis. Both fecal microbial transplantation (FMT) from donor mice that did not develop tolerance and dietary butyrate supplementation significantly reduced the development of tolerance independently of suppression of systemic inflammation. These findings could inform immediate therapies to extend the analgesic efficacy of opioids.
Collapse
Affiliation(s)
- Izabella Sall
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
- Graduate program in Molecular and Evolutionary Systems Biology, University of New Hampshire, Durham, NH, USA
| | - Randi Foxall
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Lindsey Felth
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
| | - Soren Maret
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Zachary Rosa
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
| | - Anirudh Gaur
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
| | - Jennifer Calawa
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
- Microbiology Graduate Program, University of New Hampshire, Durham, NH, USA
| | - Nadia Pavlik
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| | - Jennifer L. Whistler
- Center for Neuroscience, University of California–Davis, Davis, CA, USA
- Department of Physiology and Membrane Biology, UC Davis School of Medicine, Davis, CA, USA
| | - Cheryl A. Whistler
- Department of Molecular, Cellular, & Biomedical Sciences, University of New Hampshire, Durham, NH, USA
| |
Collapse
|
58
|
Kilonzo-Nthenge A, Rafiqullah I, Netherland M, Nzomo M, Mafiz A, Nahashon S, Hasan NA. Comparative metagenomics of microbial communities and resistome in southern farming systems: implications for antimicrobial stewardship and public health. Front Microbiol 2024; 15:1443292. [PMID: 39659424 PMCID: PMC11628260 DOI: 10.3389/fmicb.2024.1443292] [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: 06/06/2024] [Accepted: 10/14/2024] [Indexed: 12/12/2024] Open
Abstract
Agricultural practices significantly influence microbial diversity and the distribution of virulence and antimicrobial resistance (AMR) genes, with implications for ecosystem health and food safety. This study used metagenomic sequencing to analyze 60 samples (30 per state) including water, soil, and manure (10 each) from Alabama (a mix of cattle and poultry sources) and Tennessee (primarily from cattle). The results highlighted a rich microbial diversity, predominantly comprising Bacteria (67%) and Viruses (33%), with a total of over 1,950 microbial species identified. The dominant bacterial phyla were Proteobacteria, Cyanobacteria, Actinobacteria, Firmicutes, and Bacteroidetes, with the viral communities primarily represented by Phixviricota and Uroviricota. Distinct state-specific microbial profiles were evident, with Alabama demonstrating a higher prevalence of viral populations and unique bacterial phyla compared to Tennessee. The influence of environmental and agricultural practices was reflected in the microbial compositions: soil samples were notably rich in Actinobacteria, water samples were dominated by Proteobacteria and Cyanobacteria, and manure samples from Alabama showed a predominance of Actinobacteria. Further analyses, including diversity assessment and enterotype clustering, revealed complex microbial structures. Tennessee showed higher microbial diversity and phylogenetic complexity across most sample types compared to Alabama, with poultry-related samples displaying distinct diversity trends. Principal Coordinate Analysis (PCoA) highlighted notable state-specific variations, particularly in manure samples. Differential abundance analysis demonstrated elevated levels of Deinococcus and Ligilactobacillus in Alabama, indicating regional effects on microbial distributions. The virulome analysis revealed a significant presence of virulence genes in samples from Alabama. The community resistome was extensive, encompassing 109 AMR genes across 18 antibiotic classes, with manure samples displaying considerable diversity. Ecological analysis of the interactions between AMR gene subtypes and microbial taxa revealed a sophisticated network, often facilitated by bacteriophages. These findings underscore the critical role of agricultural practices in shaping microbial diversity and resistance patterns, highlighting the need for targeted AMR mitigation strategies in agricultural ecosystems to protect both public health and environmental integrity.
Collapse
Affiliation(s)
- Agnes Kilonzo-Nthenge
- Department of Food and Animal Sciences, Tennessee State University, Nashville, TN, United States
| | | | | | - Maureen Nzomo
- Department of Food and Animal Sciences, Tennessee State University, Nashville, TN, United States
| | - Abdullah Mafiz
- Department of Food and Animal Sciences, Tennessee State University, Nashville, TN, United States
| | - Samuel Nahashon
- Department of Food and Animal Sciences, Tennessee State University, Nashville, TN, United States
| | - Nur A. Hasan
- EzBiome Inc., Gaithersburg, MD, United States
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, MD, United States
| |
Collapse
|
59
|
Wozniak H, Gaïa N, Lazarevic V, Le Terrier C, Beckmann TS, Balzani E, Urner M, Pugin J, Schrenzel J, Heidegger CP. Early reduction in gut microbiota diversity in critically ill patients is associated with mortality. Ann Intensive Care 2024; 14:174. [PMID: 39589673 PMCID: PMC11599525 DOI: 10.1186/s13613-024-01407-x] [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: 07/01/2024] [Accepted: 11/04/2024] [Indexed: 11/27/2024] Open
Abstract
BACKGROUND Critical illness is associated with an altered gut microbiota, yet its association with poor outcomes remains unclear. This study evaluates the early gut microbiota diversity changes in intensive care unit patients and its association with mortality. Additionally, it explores fecal pH as a potential biomarker for these changes. METHODS In this prospective observational cohort study, fecal samples were collected at two time points: S1, the first stool passed upon intensive care unit admission, and S2, the first stool passed at least 24 h after S1. Full-length 16S rRNA gene sequencing was performed for gut microbiota analysis, with α-diversity measured using the Shannon index. Bayesian joint models were used to estimate the association between time-varying changes in gut microbiota diversity and 60-day mortality, as well as the association between daily changes in stool pH and in diversity. RESULTS Twenty-four of 96 patients overall died during follow-up. Daily Shannon index decreased on average by -0.1 points [95% Credible Intervals (CrI) -0.20 to -0.10]. Every point decrease in Shannon index was associated with a 1.99-fold increase in the hazard of death (95% CrI, 1.04 to 4.51). Time-varying fecal pH levels were not associated with changes in Shannon index. CONCLUSIONS Gut microbiota diversity decreased over time, associated with increased mortality. Fecal pH is an unreliable marker of gut microbiota change. Future studies on gut microbiota and related biomarkers should focus on the initial days in the intensive care unit to detect and mitigate a decline in gut microbiota diversity.
Collapse
Affiliation(s)
- Hannah Wozniak
- Intensive Care Unit, Department of Acute Medicine, Geneva University Hospitals, Geneva, Switzerland.
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Canada.
| | - Nadia Gaïa
- Genomic Research Laboratory, Department of Medicine, Geneva University, Geneva, Switzerland
| | - Vladimir Lazarevic
- Genomic Research Laboratory, Department of Medicine, Geneva University, Geneva, Switzerland
| | - Christophe Le Terrier
- Intensive Care Unit, Department of Acute Medicine, Geneva University Hospitals, Geneva, Switzerland
- Department of Medicine, Emerging Antibiotic Resistance Unit, Medical and Molecular Microbiology, University of Fribourg, Fribourg, Switzerland
| | - Tal Sarah Beckmann
- Intensive Care Unit, Department of Acute Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Eleonora Balzani
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Canada
| | - Martin Urner
- Interdepartmental Division of Critical Care Medicine, University of Toronto, Toronto, Canada
- Department of Anesthesiology & Pain Medicine, University of Toronto, Toronto, Canada
| | - Jérôme Pugin
- Intensive Care Unit, Department of Acute Medicine, Geneva University Hospitals, Geneva, Switzerland
| | - Jacques Schrenzel
- Genomic Research Laboratory, Department of Medicine, Geneva University, Geneva, Switzerland
| | - Claudia-Paula Heidegger
- Intensive Care Unit, Department of Acute Medicine, Geneva University Hospitals, Geneva, Switzerland
| |
Collapse
|
60
|
Steinberg R, Mostacci N, Kieninger E, Frauchiger B, Casaulta C, Usemann J, Moeller A, Trachsel D, Rochat I, Blanchon S, Mueller-Suter D, Kern B, Zanolari M, Frey U, Ramsey KA, Hilty M, Latzin P, Korten I. Early nasal microbiota and subsequent respiratory tract infections in infants with cystic fibrosis. COMMUNICATIONS MEDICINE 2024; 4:246. [PMID: 39580540 PMCID: PMC11585651 DOI: 10.1038/s43856-024-00616-6] [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: 03/08/2024] [Accepted: 09/18/2024] [Indexed: 11/25/2024] Open
Abstract
BACKGROUND Respiratory tract infections (RTIs) drive lung function decline in children with cystic fibrosis (CF). While the respiratory microbiota is clearly associated with RTI pathogenesis in infants without CF, data on infants with CF is scarce. We compared nasal microbiota development between infants with CF and controls and assessed associations between early-life nasal microbiota, RTIs, and antibiotic treatment in infants with CF. METHODS We included 50 infants with CF and 30 controls from two prospective birth cohorts followed throughout the first year of life. We collected 1511 biweekly nasal swabs and analyzed the microbiota after amplifying the V3-V4 region of the 16S rRNA gene. We conducted structured weekly interviews to assess respiratory symptoms and antibiotic treatment. We calculated generalized additive mixed models and permutational analysis of variance. RESULTS Here, we show that the nasal microbiota is already altered before the first RTI or antibiotic treatment in infants with CF. Microbiota diversity differs between infants with CF and controls following RTIs and/or antibiotic treatment. CF infants with lower α-diversity have a higher number of subsequent RTIs. CONCLUSIONS Early nasal microbiota alterations may reflect predisposition or predispose to RTIs in infants with CF, and further change after RTIs and antibiotic treatment. This highlights the potential of targeting the nasal microbiota in CF-related RTI management, while also questioning current practices in the era of novel modulator therapies.
Collapse
Affiliation(s)
- Ruth Steinberg
- Division of Paediatric Respiratory Medicine and Allergology, Departement of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Graduate School for Cellular and Biomedical Sciences, University of Bern, Bern, Switzerland
- Paediatric Intensive Care and Pulmonology, University Children's Hospital Basel (UKBB), Basel, Switzerland
| | - Nadja Mostacci
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Elisabeth Kieninger
- Division of Paediatric Respiratory Medicine and Allergology, Departement of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Bettina Frauchiger
- Division of Paediatric Respiratory Medicine and Allergology, Departement of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Carmen Casaulta
- Division of Paediatric Respiratory Medicine and Allergology, Departement of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Jakob Usemann
- Paediatric Intensive Care and Pulmonology, University Children's Hospital Basel (UKBB), Basel, Switzerland
- Department of Respiratory Medicine and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Alexander Moeller
- Department of Respiratory Medicine and Children's Research Center, University Children's Hospital Zurich, Zurich, Switzerland
| | - Daniel Trachsel
- Paediatric Intensive Care and Pulmonology, University Children's Hospital Basel (UKBB), Basel, Switzerland
| | - Isabelle Rochat
- Pediatric Pulmonology and Cystic Fibrosis Unit, Division of Pediatrics, Department of Woman-Mother-Child, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | - Sylvain Blanchon
- Pediatric Pulmonology and Cystic Fibrosis Unit, Division of Pediatrics, Department of Woman-Mother-Child, Lausanne University Hospital and University of Lausanne, Lausanne, Switzerland
| | | | - Barbara Kern
- Division of Pediatric Pneumology, Kantonsspital Aarau, Aarau, Switzerland
| | - Maura Zanolari
- Division of Pediatrics, Hospital Bellinzona, Bellinzona, Switzerland
| | - Urs Frey
- Paediatric Intensive Care and Pulmonology, University Children's Hospital Basel (UKBB), Basel, Switzerland
| | - Kathryn A Ramsey
- Division of Paediatric Respiratory Medicine and Allergology, Departement of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
- Wal-yan Respiratory Research Centre, Telethon Kids Institute, University of Western Australia, Perth, WA, Australia
| | - Markus Hilty
- Institute for Infectious Diseases, University of Bern, Bern, Switzerland
| | - Philipp Latzin
- Division of Paediatric Respiratory Medicine and Allergology, Departement of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland
| | - Insa Korten
- Division of Paediatric Respiratory Medicine and Allergology, Departement of Paediatrics, Inselspital, Bern University Hospital, University of Bern, Bern, Switzerland.
| |
Collapse
|
61
|
Luo Q, Zhang S, Butt H, Chen Y, Jiang H, An L. PhyImpute and UniFracImpute: two imputation approaches incorporating phylogeny information for microbial count data. Brief Bioinform 2024; 26:bbae653. [PMID: 39708838 DOI: 10.1093/bib/bbae653] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 11/16/2024] [Accepted: 12/05/2024] [Indexed: 12/23/2024] Open
Abstract
Sequencing-based microbial count data analysis is a challenging task due to the presence of numerous non-biological zeros, which can impede downstream analysis. To tackle this issue, we introduce two novel approaches, PhyImpute and UniFracImpute, which leverage similar microbial samples to identify and impute non-biological zeros in microbial count data. Our proposed methods utilize the probability of non-biological zeros and phylogenetic trees to estimate sample-to-sample similarity, thus addressing this challenge. To evaluate the performance of our proposed methods, we conduct experiments using both simulated and real microbial data. The results demonstrate that PhyImpute and UniFracImpute outperform existing methods in recovering the zeros and empowering downstream analyses such as differential abundance analysis, and disease status classification.
Collapse
Affiliation(s)
- Qianwen Luo
- Department of Biosystems Engineering, University of Arizona, Tucson, AZ 85721, United States
| | - Shanshan Zhang
- Interdisciplinary Program in Statistics and Data Science, University of Arizona, Tucson, AZ 85721, United States
| | - Hamza Butt
- Department of Epidemiology and Biostatistics, University of Arizona, Tucson, AZ 85721, United States
| | - Yin Chen
- Department of Pharmacology and Toxicology, School of Pharmacy, University of Arizona, Tucson, AZ 85721, United States
| | - Hongmei Jiang
- Department of Statistics and Data Science, Northwestern University, Evanston, IL 60208, United States
| | - Lingling An
- Department of Biosystems Engineering, University of Arizona, Tucson, AZ 85721, United States
- Interdisciplinary Program in Statistics and Data Science, University of Arizona, Tucson, AZ 85721, United States
- Department of Epidemiology and Biostatistics, University of Arizona, Tucson, AZ 85721, United States
| |
Collapse
|
62
|
Pereira H, Chakarov N, Caspers BA, Gilles M, Jones W, Mijoro T, Zefania S, Székely T, Krüger O, Hoffman JI. The gut microbiota of three avian species living in sympatry. BMC Ecol Evol 2024; 24:144. [PMID: 39574002 PMCID: PMC11580620 DOI: 10.1186/s12862-024-02329-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2024] [Accepted: 11/06/2024] [Indexed: 11/24/2024] Open
Abstract
BACKGROUND Evolutionary divergence and genetic variation are often linked to differences in microbial community structure and diversity. While environmental factors and diet heavily influence gut microbial communities, host species contributions are harder to quantify. Closely related species living in sympatry provide a unique opportunity to investigate species differences without the confounding effects of habitat and dietary variation. We therefore compared and contrasted the gut microbiota of three sympatric plover species: the widespread Kittlitz's and white-fronted plovers (Anarhynchus pecuarius and A. marginatus) and the endemic and vulnerable Madagascar plover (A. thoracicus). RESULTS We found no significant differences in the beta diversity (composition) of the gut microbiota of the three species. However, A. thoracicus exhibited higher intraspecific compositional similarity (i.e. lower pairwise distances) than the other two species; this pattern was especially pronounced among juveniles. By contrast, microbial alpha diversity varied significantly among the species, being highest in A. pecuarius, intermediate in A. marginatus and lowest in A. thoracicus. This pattern was again stronger among juveniles. Geographical distance did not significantly affect the composition of the gut microbiota, but genetic relatedness did. CONCLUSION While patterns of microbial diversity varied across species, the lack of compositional differences suggests that habitat and diet likely exert a strong influence on the gut microbiota of plovers. This may be enhanced by their precocial, ground-dwelling nature, which could facilitate the horizontal transmission of microbes from the environment. We hypothesise that gut microbiota diversity in plovers primarily reflects the ecological pool of microbiota, which is subsequently modified by host-specific factors including genetics. The reduced microbial and genetic diversity of the endemic A. thoracicus may hinder its ability to adapt to environmental changes, highlighting the need for increased conservation efforts for this vulnerable species.
Collapse
Grants
- 316099922 Deutsche Forschungsgemeinschaft
- 316099922 Deutsche Forschungsgemeinschaft
- 316099922 Deutsche Forschungsgemeinschaft
- 316099922 Deutsche Forschungsgemeinschaft
- 316099922 Deutsche Forschungsgemeinschaft
- 316099922 Deutsche Forschungsgemeinschaft
- 316099922 Deutsche Forschungsgemeinschaft
- 316099922 Deutsche Forschungsgemeinschaft
- 316099922 Deutsche Forschungsgemeinschaft
- 316099922 Deutsche Forschungsgemeinschaft
- 396780709 Deutsche Forschungsgemeinschaft,
- 396780709 Deutsche Forschungsgemeinschaft,
- 396780709 Deutsche Forschungsgemeinschaft,
- 396780709 Deutsche Forschungsgemeinschaft,
- 396780709 Deutsche Forschungsgemeinschaft,
- 396780709 Deutsche Forschungsgemeinschaft,
- 396780709 Deutsche Forschungsgemeinschaft,
- 396780709 Deutsche Forschungsgemeinschaft,
- 396780709 Deutsche Forschungsgemeinschaft,
- 396780709 Deutsche Forschungsgemeinschaft,
- Universität Bielefeld (3146)
Collapse
Affiliation(s)
- Hugo Pereira
- Department of Animal Behaviour, Bielefeld University, Konsequenz 45, Bielefeld, 33615, Germany.
| | - Nayden Chakarov
- Department of Animal Behaviour, Bielefeld University, Konsequenz 45, Bielefeld, 33615, Germany
- Joint Institute for Individualisation in a Changing Environment (JICE), Bielefeld University and University of Münster, Konsequenz 45, Bielefeld, 33615, Germany
| | - Barbara A Caspers
- Department of Behavioural Ecology, Bielefeld University, Konsequenz 45, Bielefeld, 33615, Germany
- Joint Institute for Individualisation in a Changing Environment (JICE), Bielefeld University and University of Münster, Konsequenz 45, Bielefeld, 33615, Germany
| | - Marc Gilles
- Department of Behavioural Ecology, Bielefeld University, Konsequenz 45, Bielefeld, 33615, Germany
| | - William Jones
- Institut Supérieur de Technologie de Menabe, Université of Toliara & Morondava, Toliara, 601, Madagascar
| | - Tafitasoa Mijoro
- HUN-REN-Debrecen University Reproductive Strategies Research Group, University of Debrecen, Egyetem tér 1, Debrecen, H-4032, Hungary
| | - Sama Zefania
- HUN-REN-Debrecen University Reproductive Strategies Research Group, University of Debrecen, Egyetem tér 1, Debrecen, H-4032, Hungary
| | - Tamás Székely
- Institut Supérieur de Technologie de Menabe, Université of Toliara & Morondava, Toliara, 601, Madagascar
- Milner Centre for Evolution, Department of Life Sciences, University of Bath, Claverton Down, Bath, BA2 7AY, UK
| | - Oliver Krüger
- Department of Animal Behaviour, Bielefeld University, Konsequenz 45, Bielefeld, 33615, Germany
- Joint Institute for Individualisation in a Changing Environment (JICE), Bielefeld University and University of Münster, Konsequenz 45, Bielefeld, 33615, Germany
| | - Joseph I Hoffman
- Department of Animal Behaviour, Bielefeld University, Konsequenz 45, Bielefeld, 33615, Germany
- Department of Evolutionary Population Genetics, Bielefeld University, Konsequenz 45, Bielefeld, 33615, Germany
- Joint Institute for Individualisation in a Changing Environment (JICE), Bielefeld University and University of Münster, Konsequenz 45, Bielefeld, 33615, Germany
- British Antarctic Survey, High Cross, Madingley Road, Cambridge, CB3 OET, UK
- Center for Biotechnology (CeBiTec), Faculty of Biology, Bielefeld University, Universitätsstraße 25, Bielefeld, 33615, Germany
| |
Collapse
|
63
|
Hu W, Naimi S, Trainel N, Liévin-Le Moal V, Perlemuter G, Chassaing B, Ciocan D, Cassard AM. Minibioreactor arrays to model microbiome response to alcohol and tryptophan in the context of alcohol-associated liver disease. NPJ Biofilms Microbiomes 2024; 10:132. [PMID: 39567509 PMCID: PMC11579296 DOI: 10.1038/s41522-024-00602-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 11/05/2024] [Indexed: 11/22/2024] Open
Abstract
The intestinal microbiota (IM) plays a role in the severity of alcohol-associated liver disease. Modifying severe alcohol-associated hepatitis (AH) dysbiosis improves liver injury through tryptophan (Trp) metabolites and the aryl hydrocarbon receptor (AhR). However, Trp's effect on the IM in alcohol use disorder (AUD) patients remains unclear. Here, we used an in vitro microbiota modeling system named Minibioreactor arrays (MBRAs). Feces from AUD patients with or without AH were treated with low, normal, or high Trp concentrations, with subsequent treatment with alcohol. Microbiota composition and AhR activity were studied. We showed that microbial communities from donors were maintained in MBRAs. High and low Trp increased the abundance of pathogen Escherichia Shigella. In the absence of alcohol, Trp changed more bacteria in AUD IM compared to AH IM. Normal Trp increased the AhR activity. Overall, maintaining normal Trp levels may prevent dysbiosis in AUD or AH, pending in vivo confirmation.
Collapse
Affiliation(s)
- Wanchao Hu
- Université Paris-Saclay, Inserm U996, Inflammation, Microbiome and Immunosurveillance, Orsay, France
| | - Sabrine Naimi
- INSERM U1016, Team "Mucosal Microbiota in Chronic Inflammatory Diseases", CNRS UMR 8104, Université Paris Cité, Paris, France
| | - Nicolas Trainel
- Université Paris-Saclay, Inserm U996, Inflammation, Microbiome and Immunosurveillance, Orsay, France
| | - Vanessa Liévin-Le Moal
- Université Paris-Saclay, Inserm U996, Inflammation, Microbiome and Immunosurveillance, Orsay, France
| | - Gabriel Perlemuter
- Université Paris-Saclay, Inserm U996, Inflammation, Microbiome and Immunosurveillance, Orsay, France
- AP-HP, Hepato-Gastroenterology and Nutrition, Hôpital Antoine-Béclère, Clamart, France
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France
| | - Benoit Chassaing
- INSERM U1016, Team "Mucosal Microbiota in Chronic Inflammatory Diseases", CNRS UMR 8104, Université Paris Cité, Paris, France.
- Institut Pasteur, Université Paris Cité, INSERM U1306, Microbiome-Host Interactions, Paris, France.
| | - Dragos Ciocan
- Université Paris-Saclay, Inserm U996, Inflammation, Microbiome and Immunosurveillance, Orsay, France.
- AP-HP, Hepato-Gastroenterology and Nutrition, Hôpital Antoine-Béclère, Clamart, France.
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France.
| | - Anne-Marie Cassard
- Université Paris-Saclay, Inserm U996, Inflammation, Microbiome and Immunosurveillance, Orsay, France.
- Paris Center for Microbiome Medicine (PaCeMM) FHU, Paris, France.
| |
Collapse
|
64
|
Soueidan A, Idiri K, Becchina C, Esparbès P, Legrand A, Le Bastard Q, Montassier E. Pooled analysis of oral microbiome profiles defines robust signatures associated with periodontitis. mSystems 2024; 9:e0093024. [PMID: 39445812 PMCID: PMC11575188 DOI: 10.1128/msystems.00930-24] [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: 07/12/2024] [Accepted: 07/23/2024] [Indexed: 10/25/2024] Open
Abstract
Oral microbial dysbiosis has been associated with periodontitis in studies using 16S rRNA gene sequencing analysis. However, this technology is not sufficient to consistently separate the bacterial species to species level, and reproducible oral microbiome signatures are scarce. Obtaining these signatures would significantly enhance our understanding of the underlying pathophysiological processes of this condition and foster the development of improved therapeutic strategies, potentially personalized to individual patients. Here, we sequenced newly collected samples from 24 patients with periodontitis, and we collected available oral microbiome data from 24 samples in patients with periodontitis and from 214 samples in healthy individuals (n = 262). Data were harmonized, and we performed a pooled analysis of individual patient data. By metagenomic sequencing of the plaque microbiome, we found microbial signatures for periodontitis and defined a periodontitis-related complex, composed by the most discriminative bacteria. A simple two-factor decision tree, based on Tannerella forsythia and Fretibacterium fastidiosum, was associated with periodontitis with high accuracy (area under the curve: 0.94). Altogether, we defined robust oral microbiome signatures relevant to the pathophysiology of periodontitis that can help define promising targets for microbiome therapeutic modulation when caring for patients with periodontitis. IMPORTANCE Oral microbial dysbiosis has been associated with periodontitis in studies using 16S rRNA gene sequencing analysis. However, this technology is not sufficient to consistently separate the bacterial species to species level, and reproducible oral microbiome signatures are scarce. Here, using ultra-deep metagenomic sequencing and machine learning tools, we defined a simple two-factor decision tree, based on Tannerella forsythia and Fretibacterium fastidiosum, that was highly associated with periodontitis. Altogether, we defined robust oral microbiome signatures relevant to the pathophysiology of periodontitis that can help define promising targets for microbiome therapeutic modulation when caring for patients with periodontitis.
Collapse
Affiliation(s)
- Assem Soueidan
- Nantes Université, CHU Nantes, INSERM, Regenerative Medicine and Skeleton, Nantes, France
- Department of Periodontology, Faculty of Dental Surgery, Nantes, France
| | - Katia Idiri
- Department of Periodontology, Faculty of Dental Surgery, Nantes, France
| | - Camille Becchina
- Department of Periodontology, Faculty of Dental Surgery, Nantes, France
| | - Pauline Esparbès
- Department of Periodontology, Faculty of Dental Surgery, Nantes, France
| | - Arnaud Legrand
- CHU Nantes, Direction de la Recherche Clinique, Nantes, France
| | - Quentin Le Bastard
- Cibles et médicaments des infections et de l'immunité, IICiMed, Nantes Université, Nantes, France
- CHU Nantes, Service des urgences, Nantes, France
| | - Emmanuel Montassier
- CHU Nantes, Service des urgences, Nantes, France
- Nantes Université, Inserm, CHU Nantes, Center for Research in Transplantation and Translational Immunology, Nantes, France
| |
Collapse
|
65
|
O'Hare MA, Swart PC, Malan-Müller S, van den Heuvel LL, Bröcker E, Seedat S, Hemmings SMJ. The saNeuroGut Initiative: Investigating the Gut Microbiome and Symptoms of Anxiety, Depression, and Posttraumatic Stress. Neuroimmunomodulation 2024; 32:1-15. [PMID: 39561720 DOI: 10.1159/000542696] [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: 09/02/2024] [Accepted: 11/15/2024] [Indexed: 11/21/2024] Open
Abstract
INTRODUCTION Common mental disorders, such as anxiety disorders, depression, and posttraumatic stress disorder (PTSD), present a substantial health and economic burden. The gut microbiome has been associated with these psychiatric disorders via the microbiome-gut-brain axis. However, previous studies have focused on the associations between the gut microbiome and common mental disorders in European, North American, and Asian populations. As part of the saNeuroGut Initiative, we assessed associations between gut microbial composition and self-reported symptoms of anxiety, depression, and posttraumatic stress (PTS) among South African adults. METHODS Participants completed validated, online self-report questionnaires to evaluate symptoms of state anxiety, trait anxiety, depression, and PTSD. Eighty-six stool-derived microbial DNA samples underwent sequencing of the V4 region of the 16S rRNA gene to characterise gut bacterial taxa in the sample. RESULTS No significant associations were observed between symptom severity scores and alpha (Shannon and Simpson indices) and beta (Aitchison distances) diversity metrics. Linear regression models revealed that the abundances of Catenibacterium, Collinsella, and Holdemanella were significantly positively associated with the severity of PTS symptoms. CONCLUSION Catenibacterium, Collinsella, and Holdemanella have each previously been associated with various psychiatric disorders, with Catenibacterium having been positively associated with symptoms of PTSD in another South African cohort. This study sheds light on the relationship between the human gut microbiome and symptoms of anxiety, depression, and PTS in a South African adult sample.
Collapse
Affiliation(s)
- Michaela A O'Hare
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council / Stellenbosch University Genomics of Brain Disorders Research Unit, Cape Town, South Africa
- Department of Biomedical Sciences, Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Patricia C Swart
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council / Stellenbosch University Genomics of Brain Disorders Research Unit, Cape Town, South Africa
| | - Stefanie Malan-Müller
- Department of Pharmacology and Toxicology, Faculty of Medicine, University Complutense of Madrid (UCM), Research Institute of Hospital 12 de Octubre (Imas12), Instituto Universitario de Investigación Neuroquímica (IUIN-UCM), Madrid, Spain
- Biomedical Research Network Centre in Mental Health, Institute of Health Carlos III (CIBERSAM, ISCIII), Madrid, Spain
| | - Leigh L van den Heuvel
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council / Stellenbosch University Genomics of Brain Disorders Research Unit, Cape Town, South Africa
| | - Erine Bröcker
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council / Stellenbosch University Genomics of Brain Disorders Research Unit, Cape Town, South Africa
| | - Soraya Seedat
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council / Stellenbosch University Genomics of Brain Disorders Research Unit, Cape Town, South Africa
| | - Sian M J Hemmings
- Department of Psychiatry, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
- South African Medical Research Council / Stellenbosch University Genomics of Brain Disorders Research Unit, Cape Town, South Africa
| |
Collapse
|
66
|
Alex AM, Levendosky AA, Bogat GA, Muzik M, Nuttall AK, Knickmeyer RC, Lonstein JS. Stress and mental health symptoms in early pregnancy are associated with the oral microbiome. BMJ MENTAL HEALTH 2024; 27:e301100. [PMID: 39562142 PMCID: PMC11580324 DOI: 10.1136/bmjment-2024-301100] [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: 03/29/2024] [Accepted: 07/18/2024] [Indexed: 11/21/2024]
Abstract
BACKGROUND Research has revealed associations between microbes of the gastrointestinal tract and stress, anxiety and depression in pregnant or postpartum women. While these studies suggest a gut-brain-behaviour axis, no studies have examined microbes of the oral cavity in relation to maternal mental health. OBJECTIVE To explore a potential oral-brain-behaviour axis related to maternal mental health. METHODS Microbes were measured in saliva obtained from 224 second-trimester (mean±SD = 17±2 weeks) women oversampled for stress. Oralome data were associated with women's recent or cumulative pregnancy stress, trait and state anxiety, depression symptoms and post-traumatic stress disorder (PTSD) symptoms. Covariates explored included age, income, alcohol and tobacco use, dental issues and physical health problems. RESULTS Pregnant women in the high trait anxiety or depression symptom groups had higher oral alpha diversity, indicating higher richness of species within samples. Groups with high and low PTSD symptoms differed in beta diversity, reflecting differences in community composition. Linear discriminant analysis showed differently abundant microbes in women with high stress versus low life stress, anxiety, depression, and PTSD, with the affected microbes mostly differing by symptom. Notably, members of phylum Proteobacteria were more abundant in women with high recent life stress and Spirochaetes was more abundant in women with high depression symptoms. Members of phylum Firmicutes were more abundant in the high trait anxiety and high depression groups. Genus Dialister (previously found to be lower in the gut of depressed non-pregnant people) was higher in women experiencing either high trait or state anxiety, or experiencing high depression symptoms, while genus Eikenella was elevated with high trait anxiety, depression or PTSD. CONCLUSIONS The oral microbiome is associated with stress and mental health in pregnant women, in ways different from the gut microbiome or what has been found in non-pregnant people. CLINICAL IMPLICATIONS Understanding oral microbiome-mental health relations may reveal future microbial targets to improve maternal psychological well-being.
Collapse
Affiliation(s)
- Ann M Alex
- Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, Michigan, USA
| | - Alytia A Levendosky
- Department of Psychology, Michigan State University, East Lansing, Michigan, USA
| | - G Anne Bogat
- Department of Psychology, Michigan State University, East Lansing, Michigan, USA
| | - Maria Muzik
- Department of Psychiatry and Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, Michigan, USA
| | - Amy K Nuttall
- Department of Human Development and Family Studies, Michigan State University, East Lansing, Michigan, USA
| | - Rebecca C Knickmeyer
- Institute for Quantitative Health Science & Engineering, Michigan State University, East Lansing, Michigan, USA
- Department of Pediatrics and Human Development, Michigan State University, East Lansing, Michigan, USA
| | - Joseph S Lonstein
- Department of Psychology, Michigan State University, East Lansing, Michigan, USA
| |
Collapse
|
67
|
Yu J, Duan Y, Zhang M, Li Q, Cao M, Song W, Zhao F, Kwok LY, Zhang H, Li R, Sun Z. Effect of combined probiotics and doxycycline therapy on the gut-skin axis in rosacea. mSystems 2024; 9:e0120124. [PMID: 39475254 PMCID: PMC11575305 DOI: 10.1128/msystems.01201-24] [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: 09/05/2024] [Accepted: 09/29/2024] [Indexed: 11/05/2024] Open
Abstract
Rosacea is a chronic inflammatory skin condition marked by facial erythema, telangiectasia, and acne-like eruptions, affecting millions worldwide. While antibiotics remain a common treatment, prolonged use has significant adverse effects and can lead to antibiotic resistance. This study evaluated the impact of combined probiotics and doxycycline treatment on rosacea, emphasizing the gut-skin axis. Sixty rosacea patients were randomly assigned to the probiotic, placebo, or control groups. After a 2-week doxycycline treatment, participants underwent a 3-month intervention with either a placebo, probiotic, or no further treatment. Clinical outcomes were assessed at baseline and after the 14-week intervention. Our results showed that probiotic administration improved facial skin conditions, alleviated inflammation, and reduced facial skin microbiota diversity while enhancing gut microbiota heterogeneity. Multivariate analysis identified microbial markers distinguishing the probiotic group from the control and placebo groups, and some markers were associated with skin health parameters. After the probiotic intervention, some facial skin-associated taxa, such as Aquabacterium sp., UBA4096 sp. 1, UBA4096 sp. 2, and Yimella indica, decreased in abundance. Additionally, the fecal microbiota of the probiotic group was enriched in specific gut microbes, including Streptococcus parasanguinis, Erysipelatoclostridium ramosum, and Coprobacillus cateniformis, while showing a reduced abundance of Bacteroides vulgatus. These changes were associated with reduced facial sebum levels and a lower physician's global assessment score. Finally, fewer antibiotic resistance genes, particularly tetracycline resistance genes, were detected in the probiotic group compared with the control and placebo groups. Our study supports the existence of a gut-skin axis and the application of probiotics in managing rosacea. IMPORTANCE This research elucidates rosacea management with novel insights into probiotic use alongside doxycycline, showing dual benefits in symptom relief and inflammation reduction in patients. The study maps probiotic-induced shifts in gut and skin microbiota, underscoring microbial shifts correlating with skin health improvements. Crucially, it deciphers the gut-skin axis modulation by probiotics, proposing a method to curb antibiotic resistance in rosacea therapies. This study furnishes robust evidence for probiotics in rosacea, advancing our grasp of the gut-skin relationship.
Collapse
Affiliation(s)
- Jie Yu
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Yan Duan
- Department of Dermatology, Inner Mongolia People’s Hospital, Hohhot, China
| | - Meng Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Qi Li
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Miao Cao
- Department of Dermatology, Inner Mongolia People’s Hospital, Hohhot, China
| | - Weixin Song
- Department of Dermatology, Inner Mongolia People’s Hospital, Hohhot, China
| | - Feiyan Zhao
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Lai-Yu Kwok
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Heping Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| | - Ruiya Li
- Department of Dermatology, Inner Mongolia People’s Hospital, Hohhot, China
| | - Zhihong Sun
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education, Inner Mongolia Agricultural University, Hohhot, China
- Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs, Inner Mongolia Agricultural University, Hohhot, China
- Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, China
| |
Collapse
|
68
|
Fei T, Funnell T, Waters NR, Raj SS, Baichoo M, Sadeghi K, Dai A, Miltiadous O, Shouval R, Lv M, Peled JU, Ponce DM, Perales MA, Gönen M, van den Brink MRM. Scalable log-ratio lasso regression for enhanced microbial feature selection with FLORAL. CELL REPORTS METHODS 2024; 4:100899. [PMID: 39515336 DOI: 10.1016/j.crmeth.2024.100899] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 09/03/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024]
Abstract
Identifying predictive biomarkers of patient outcomes from high-throughput microbiome data is of high interest, while existing computational methods do not satisfactorily account for complex survival endpoints, longitudinal samples, and taxa-specific sequencing biases. We present FLORAL, an open-source tool to perform scalable log-ratio lasso regression and microbial feature selection for continuous, binary, time-to-event, and competing risk outcomes, with compatibility for longitudinal microbiome data as time-dependent covariates. The proposed method adapts the augmented Lagrangian algorithm for a zero-sum constraint optimization problem while enabling a two-stage screening process for enhanced false-positive control. In extensive simulation and real-data analyses, FLORAL achieved consistently better false-positive control compared to other lasso-based approaches and better sensitivity over popular differential abundance testing methods for datasets with smaller sample sizes. In a survival analysis of allogeneic hematopoietic cell transplant recipients, FLORAL demonstrated considerable improvement in microbial feature selection by utilizing longitudinal microbiome data over solely using baseline microbiome data.
Collapse
Affiliation(s)
- Teng Fei
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA.
| | - Tyler Funnell
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Nicholas R Waters
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Sandeep S Raj
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Mirae Baichoo
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Keimya Sadeghi
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, USA
| | - Anqi Dai
- Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Oriana Miltiadous
- Department of Pediatrics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Roni Shouval
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Meng Lv
- Institute of Hematology, Peking University People's Hospital, Beijing, China
| | - Jonathan U Peled
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Doris M Ponce
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Miguel-Angel Perales
- Adult Bone Marrow Transplantation Service, Department of Medicine, Memorial Sloan Kettering Cancer Center, New York, NY, USA; Department of Medicine, Weill Cornell Medical College, New York, NY, USA
| | - Mithat Gönen
- Department of Epidemiology and Biostatistics, Memorial Sloan Kettering Cancer Center, New York, NY, USA
| | - Marcel R M van den Brink
- Department of Hematology and Hematopoietic Cell Transplantation, City of Hope National Medical Center, Los Angeles, CA, USA; Hematologic Malignancies Research Institute, City of Hope National Medical Center, Los Angeles, CA, USA; Comprehensive Cancer Center, City of Hope National Medical Center, Los Angeles, CA, USA.
| |
Collapse
|
69
|
Bennett AE, Kelsey S, Saup C, Wilkins M, Malacrinò A. Selenium alters the gene content but not the taxonomic composition of the soil microbiome. ENVIRONMENTAL MICROBIOME 2024; 19:92. [PMID: 39558431 PMCID: PMC11575018 DOI: 10.1186/s40793-024-00641-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 11/11/2024] [Indexed: 11/20/2024]
Abstract
BACKGROUND Microbiomes, essential to ecosystem processes, face strong selective forces that can drive rapid evolutionary adaptation. However, our understanding of evolutionary processes within natural systems remains limited. We investigated evolution in response to naturally occurring selenium in soils of different geological parental materials on the Western Slope of Colorado. Our study focused on examining changes in gene frequencies within microbial communities in response to selenium exposure. RESULTS Despite expectations of taxonomic composition shifts and increased gene content changes at high-selenium sites, we found no significant alterations in microbial diversity or community composition. Surprisingly, we observed a significant increase in differentially abundant genes within high-selenium sites. CONCLUSIONS These findings are suggestive that selection within microbiomes primarily drives the accumulation of genes among existing microbial taxa, rather than microbial species turnover, in response to strong stressors like selenium. Our study highlights an unusual system that allows us to examine evolution in response to the same stressor annually in a non-model system, contributing to understanding microbiome evolution beyond model systems.
Collapse
Affiliation(s)
- Alison E Bennett
- Department of Evolution, Ecology, and Organismal Biology, The Ohio State University, Columbus, OH, USA
| | - Scott Kelsey
- Department of Evolution, Ecology, and Organismal Biology, The Ohio State University, Columbus, OH, USA
| | - Casey Saup
- School of Earth Sciences, The Ohio State University, Columbus, OH, USA
| | - Mike Wilkins
- Department of Soil and Crop Sciences, Colorado State University, Fort Collins, CO, USA
| | - Antonino Malacrinò
- Department of Agriculture, Università degli Studi Mediterranea di Reggio Calabria, Reggio Calabria, Italy.
- Department of Biological Sciences, Clemson University, Clemson, SC, USA.
| |
Collapse
|
70
|
Rebeck ON, Wallace MJ, Prusa J, Ning J, Evbuomwan EM, Rengarajan S, Habimana-Griffin L, Kwak S, Zahrah D, Tung J, Liao J, Mahmud B, Fishbein SRS, Ramirez Tovar ES, Mehta R, Wang B, Gorelik MG, Helmink BA, Dantas G. A yeast-based oral therapeutic delivers immune checkpoint inhibitors to reduce intestinal tumor burden. Cell Chem Biol 2024:S2451-9456(24)00452-5. [PMID: 39571582 DOI: 10.1016/j.chembiol.2024.10.013] [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: 03/15/2024] [Revised: 08/09/2024] [Accepted: 10/28/2024] [Indexed: 12/13/2024]
Abstract
Engineered probiotics are an emerging platform for in situ delivery of therapeutics to the gut. Herein, we developed an orally administered, yeast-based therapeutic delivery system to deliver next-generation immune checkpoint inhibitor (ICI) proteins directly to gastrointestinal tumors. We engineered Saccharomyces cerevisiae var. boulardii (Sb), a probiotic yeast with high genetic tractability and innate anticancer activity, to secrete "miniature" antibody variants that target programmed death ligand 1 (Sb_haPD-1). When tested in an ICI-refractory colorectal cancer (CRC) mouse model, Sb_haPD-1 significantly reduced intestinal tumor burden and resulted in significant shifts to the immune cell profile and microbiome composition. This oral therapeutic platform is modular and highly customizable, opening new avenues of targeted drug delivery that can be applied to treat a myriad of gastrointestinal malignancies.
Collapse
Affiliation(s)
- Olivia N Rebeck
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Miranda J Wallace
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jerome Prusa
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jie Ning
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Esse M Evbuomwan
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA
| | - Sunaina Rengarajan
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Division of Dermatology, John T. Milliken Department of Internal Medicine, Washington University School of Medicine, St. Louis MO 63110, USA
| | - LeMoyne Habimana-Griffin
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Radiation Oncology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Suryang Kwak
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - David Zahrah
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jason Tung
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - James Liao
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Bejan Mahmud
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Skye R S Fishbein
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Erick S Ramirez Tovar
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Rehan Mehta
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Bin Wang
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Mark G Gorelik
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Beth A Helmink
- Department of Surgical Oncology, University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | - Gautam Dantas
- The Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pathology and Immunology, Division of Laboratory and Genomic Medicine, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Biomedical Engineering, Washington University in St. Louis, St. Louis, MO 63130, USA; Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, USA; Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110, USA.
| |
Collapse
|
71
|
Chaudhari DS, Francescutti DM, Winters AD, Koka O, Kracht DJ, Greenberg JM, Theis KR, Angoa-Perez M. Contributions of the gut microbiota to Gulf War Illness susceptibility: Findings from a mouse model. Life Sci 2024; 359:123244. [PMID: 39551360 DOI: 10.1016/j.lfs.2024.123244] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 11/01/2024] [Accepted: 11/11/2024] [Indexed: 11/19/2024]
Abstract
AIMS In light of the evidence supporting a significant role of the gut microbiome in Gulf War Illness (GWI) pathology, we sought to examine its contribution to GWI susceptibility in a mouse model. We also aimed to identify bacterial taxa and microbially-derived metabolites associated with disease susceptibility. MAIN METHODS Male mice receiving pyridostigmine bromide (PB) orally, and controls were evaluated for symptoms of GWI at 8 weeks post-treatment. The Kansas criteria were adapted to assess behaviors associated with the following domains: gastrointestinal alterations, pain, mood, cognitive function, skin and respiratory disturbances. PB-treated subjects were classified into susceptible (GWI-S) or resilient (GWI-R). The status of the gut microbiome was assessed via analyses of the 16S rRNA gene and microbial-derived metabolites were evaluated with metabolomics tools. KEY FINDINGS Our results indicate that application of the Kansas criteria to behavioral outcomes in PB-treated mice resulted in a GWI susceptibility rate of ~35 %, similar to the one reported in humans. The composition and structure of the gut microbiome was different in GWI-S subjects compared to both control and GWI-R mice at 8 weeks but differences in microbial community structure were observed prior to PB treatment between GWI-R and GWI-S mice. GWI-S subjects exhibited a pattern of differentially abundant bacterial taxa and microbial metabolites. SIGNIFICANCE To our knowledge, this is the first preclinical report in which a stratification by susceptibility to GWI and its association with the gut microbiome is described. In light of the research conundrum that vulnerability to GWI represents, the use of tools that could provide further insight into this complex factor should be considered.
Collapse
Affiliation(s)
- Diptaraj S Chaudhari
- Research and Development Service, John D. Dingell VA Medical Center, Detroit, MI, USA; Department of Physiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Dina M Francescutti
- Research and Development Service, John D. Dingell VA Medical Center, Detroit, MI, USA; Department of Physiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Andrew D Winters
- Research and Development Service, John D. Dingell VA Medical Center, Detroit, MI, USA; Department of Physiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Orena Koka
- Research and Development Service, John D. Dingell VA Medical Center, Detroit, MI, USA; Department of Physiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - David J Kracht
- Research and Development Service, John D. Dingell VA Medical Center, Detroit, MI, USA
| | - Jonathan M Greenberg
- Research and Development Service, John D. Dingell VA Medical Center, Detroit, MI, USA; Department of Physiology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Kevin R Theis
- Department of Biochemistry, Microbiology and Immunology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Mariana Angoa-Perez
- Research and Development Service, John D. Dingell VA Medical Center, Detroit, MI, USA; Department of Physiology, Wayne State University School of Medicine, Detroit, MI, USA.
| |
Collapse
|
72
|
Liu H, Birk JW, Provatas AA, Vaziri H, Fan N, Rosenberg DW, Gharaibeh RZ, Jobin C. Correlation between intestinal microbiota and urolithin metabolism in a human walnut dietary intervention. BMC Microbiol 2024; 24:476. [PMID: 39548408 PMCID: PMC11566485 DOI: 10.1186/s12866-024-03626-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2024] [Accepted: 11/04/2024] [Indexed: 11/18/2024] Open
Abstract
This study is to investigate the relationship between the intestinal microbiota and urine levels of the ellagic acid-derived polyphenols, the urolithins, in a cohort of subjects following a three-week walnut dietary intervention. We longitudinally collected fecal and urine samples from 39 subjects before and after walnut consumption (2 oz per day for 21 days). 16S RNA gene sequencing was performed on fecal DNA to study the association between microbiota composition and the levels of nine urolithin metabolites, which were measured using UHPLC/Q-TOF-MS/MS. Fecal microbial composition was found to be significantly different between pre- and post-walnut intervention (beta diversity, FDR-p = 0.018; alpha diversity, p = 0.018). Roseburia, Rothia, Parasutterella, Lachnospiraceae UCG-004, Butyricicoccus, Bilophila, Eubacterium eligens, Lachnospiraceae UCG-001, Gordonibacter, Paraprevotella, Lachnospira, Ruminococcus torques, and Sutterella were identified as the 13 most significantly enriched genera after daily intake of walnuts. We observed 26 genera that were significantly associated with 7 urolithin metabolites, with 22 genera positively correlating after walnut supplementation (FDR-p ≤ 0.05). PICRUSt analysis showed that several inferred KEGG orthologs were associated with 4 urolithin metabolites after walnut intake. In this study, we found that walnut supplementation altered urolithin metabolites, which associates with specific changes in bacterial taxa and inferred functional contents.
Collapse
Affiliation(s)
- Huijia Liu
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL, USA
| | - John W Birk
- Division of Gastroenterology, University of Connecticut, Farmington, CT, USA
| | - Anthony A Provatas
- Center for Environmental Sciences and Engineering, University of Connecticut, Storrs, CT, USA
| | - Haleh Vaziri
- Division of Gastroenterology, University of Connecticut, Farmington, CT, USA
| | - Nuoxi Fan
- School of Medicine, University of Connecticut, Farmington, CT, USA
| | | | - Raad Z Gharaibeh
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL, USA.
- Department of Molecular Genetics and Microbiology, University of Florida, Gainesville, FL, USA.
| | - Christian Jobin
- Department of Medicine, University of Florida College of Medicine, Gainesville, FL, USA.
- Department of Infectious Diseases and Immunology, University of Florida College of Medicine, Gainesville, FL, USA.
- Department of Anatomy and Cell Biology, University of Florida College of Medicine, Gainesville, FL, USA.
| |
Collapse
|
73
|
Islam J, Ohtani N, Shimizu Y, Tanimizu M, Goto Y, Sato M, Makino E, Shimada T, Ueda C, Matsuo A, Suyama Y, Sakai Y, Karrow NA, Yoneyama H, Hirakawa R, Furukawa M, Tanaka H, Nochi T. Freeze-dried fecal microorganisms as an effective biomaterial for the treatment of calves suffering from diarrhea. Sci Rep 2024; 14:28078. [PMID: 39543390 PMCID: PMC11564888 DOI: 10.1038/s41598-024-79267-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024] Open
Abstract
Fecal microbiota transplantation (FMT) is a therapeutic modality for treating neonatal calf diarrhea. Several practical barriers, including donor selection, fecal collection, and a limited timeframe for FMT, are the main constraints to using fresh feces for implementing on-farm FMT. We report the utility of FMT with pretreated ready-to-use frozen (F) or freeze-dried (FD) microorganisms for treating calf diarrhea. In total, 19 FMT (F-FMT, n = 10 and FD-FMT, n = 9) treatments were conducted. Both FMT treatments were 100% clinically effective; however, multi-omics analysis showed that FD-FMT was superior to F-FMT. Machine learning analysis with SourceTracker confirmed that donor microbiota was retained four times better in the recipient calves treated with FD-FMT than F-FMT. A predictive model based on receiver operating characteristic curve analysis and area under the curve showed that FD-FMT was more discriminative than F-FMT of the observed changes in microbiota and metabolites during disease recovery. These results provide new insights into establishing methods for preparing fecal microorganisms to increase the quality of FMT in animals and may contribute to FMT in humans.
Collapse
Affiliation(s)
- Jahidul Islam
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, 468-1 Aoba, Aramaki, Aoba-Ku, Sendai, Miyagi, 980-8572, Japan
- Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
- Laboratory of Animal Mucosal Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | - Natsuki Ohtani
- Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
- Laboratory of Animal Mucosal Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
- Chiba Prefectural Federation of Agricultural Mutual Aid Association, Chiba, 299-0126, Japan
| | - Yu Shimizu
- Chiba Prefectural Federation of Agricultural Mutual Aid Association, Chiba, 299-0126, Japan
| | - Masae Tanimizu
- Chiba Prefectural Federation of Agricultural Mutual Aid Association, Chiba, 299-0126, Japan
| | - Yoshiaki Goto
- Chiba Prefectural Federation of Agricultural Mutual Aid Association, Chiba, 299-0126, Japan
| | - Masumi Sato
- Chiba Prefectural Federation of Agricultural Mutual Aid Association, Chiba, 299-0126, Japan
| | - Eiji Makino
- Chiba Prefectural Federation of Agricultural Mutual Aid Association, Chiba, 299-0126, Japan
| | - Toru Shimada
- Chiba Prefectural Federation of Agricultural Mutual Aid Association, Chiba, 299-0126, Japan
| | - Chise Ueda
- Chiba Prefectural Federation of Agricultural Mutual Aid Association, Chiba, 299-0126, Japan
| | - Ayumi Matsuo
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, 468-1 Aoba, Aramaki, Aoba-Ku, Sendai, Miyagi, 980-8572, Japan
| | - Yoshihisa Suyama
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, 468-1 Aoba, Aramaki, Aoba-Ku, Sendai, Miyagi, 980-8572, Japan
| | - Yoshifumi Sakai
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, 468-1 Aoba, Aramaki, Aoba-Ku, Sendai, Miyagi, 980-8572, Japan
| | - Niel A Karrow
- Ontario Agricultural College, University of Guelph, Ontario, N1G 2W1, Canada
| | - Hiroshi Yoneyama
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, 468-1 Aoba, Aramaki, Aoba-Ku, Sendai, Miyagi, 980-8572, Japan
| | - Ryota Hirakawa
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, 468-1 Aoba, Aramaki, Aoba-Ku, Sendai, Miyagi, 980-8572, Japan
- Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
- Laboratory of Animal Mucosal Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | - Mutsumi Furukawa
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, 468-1 Aoba, Aramaki, Aoba-Ku, Sendai, Miyagi, 980-8572, Japan
- Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
- Laboratory of Animal Mucosal Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan
| | - Hidekazu Tanaka
- Chiba Prefectural Federation of Agricultural Mutual Aid Association, Chiba, 299-0126, Japan.
- West Veterinary Clinical Center, Chiba Prefectural Federation of Agricultural Mutual Aid Association, 2-5-12 Midorigaoka, Yachiyo, Chiba, 276-0049, Japan.
| | - Tomonori Nochi
- International Education and Research Center for Food and Agricultural Immunology, Graduate School of Agricultural Science, Tohoku University, 468-1 Aoba, Aramaki, Aoba-Ku, Sendai, Miyagi, 980-8572, Japan.
- Laboratory of Animal Functional Morphology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan.
- Laboratory of Animal Mucosal Immunology, Graduate School of Agricultural Science, Tohoku University, Miyagi, 980-8572, Japan.
- Ontario Agricultural College, University of Guelph, Ontario, N1G 2W1, Canada.
- Division of Mucosal Vaccines, International Vaccine Design Center, The Institute of Medical Science, The University of Tokyo, Tokyo, 108-8639, Japan.
- Center for Professional Development, Institute for Excellence in Higher Education, Tohoku University, Miyagi, 980-8576, Japan.
| |
Collapse
|
74
|
Matthews AE, Trevelline BK, Wijeratne AJ, Boves TJ. Picky eaters: Selective microbial diet of avian ectosymbionts. J Anim Ecol 2024. [PMID: 39538981 DOI: 10.1111/1365-2656.14215] [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: 07/29/2024] [Accepted: 10/14/2024] [Indexed: 11/16/2024]
Abstract
Individual organisms can function as ecosystems inhabited by symbionts. Symbionts may interact with each other in ways that subsequently influence their hosts positively or negatively, although the details of how these interactions operate collectively are usually not well understood. Vane-dwelling feather mites are common ectosymbionts of birds and are proposed to confer benefits to hosts by consuming feather-degrading microbes. However, it is unknown whether these mites exhibit generalist or selective diets, or how their dietary selection could potentially impact their symbiotic functional nature. In this study, we conducted 16S rDNA and ITS1 amplicon sequencing to examine the microbial diet of feather mites. We characterized and compared the diversity and composition of bacteria and fungi in the bodies of mites living on feathers of the Prothonotary Warbler, Protonotaria citrea, to microbial assemblages present on the same feathers. We found less diverse, more compositionally similar microbial assemblages within mites than on feathers. We also found that mites were resource-selective. Based on the identity and known functions of microbes found within and presumably preferred by mites, our results suggest that these mites selectively consume feather-degrading microbes. Therefore, our results support the proposition that mites confer benefits to their hosts. This study provides insight into symbioses operating at multiple biological levels, highlights the ecological and evolutionary importance of the synergistic interactions between species, and greatly expands our understanding of feather mite biology.
Collapse
Affiliation(s)
- Alix E Matthews
- College of Sciences and Mathematics and Molecular Biosciences Program, Arkansas State University, Jonesboro, Arkansas, USA
- Department of Biological Sciences, Arkansas State University, Jonesboro, Arkansas, USA
- Department of Biology, Rhodes College, Memphis, Tennessee, USA
- Department of Biological Sciences, University at Buffalo (SUNY), Buffalo, New York, USA
| | - Brian K Trevelline
- Cornell Lab of Ornithology, Cornell University, Ithaca, New York, USA
- Department of Ecology and Evolutionary Biology, Cornell University, Ithaca, New York, USA
- Department of Biological Sciences, Kent State University, Kent, Ohio, USA
| | - Asela J Wijeratne
- Department of Biological Sciences, Arkansas State University, Jonesboro, Arkansas, USA
| | - Than J Boves
- Department of Biological Sciences, Arkansas State University, Jonesboro, Arkansas, USA
| |
Collapse
|
75
|
Macandog ADG, Catozzi C, Capone M, Nabinejad A, Nanaware PP, Liu S, Vinjamuri S, Stunnenberg JA, Galiè S, Jodice MG, Montani F, Armanini F, Cassano E, Madonna G, Mallardo D, Mazzi B, Pece S, Tagliamonte M, Vanella V, Barberis M, Ferrucci PF, Blank CU, Bouvier M, Andrews MC, Xu X, Santambrogio L, Segata N, Buonaguro L, Cocorocchio E, Ascierto PA, Manzo T, Nezi L. Longitudinal analysis of the gut microbiota during anti-PD-1 therapy reveals stable microbial features of response in melanoma patients. Cell Host Microbe 2024; 32:2004-2018.e9. [PMID: 39481388 PMCID: PMC11629153 DOI: 10.1016/j.chom.2024.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2023] [Revised: 09/15/2024] [Accepted: 10/07/2024] [Indexed: 11/02/2024]
Abstract
Immune checkpoint inhibitors (ICIs) improve outcomes in advanced melanoma, but many patients are refractory or experience relapse. The gut microbiota modulates antitumor responses. However, inconsistent baseline predictors point to heterogeneity in responses and inadequacy of cross-sectional data. We followed patients with unresectable melanoma from baseline and during anti-PD-1 therapy, collecting fecal and blood samples that were surveyed for changes in the gut microbiota and immune markers. Varying patient responses were linked to different gut microbiota dynamics during ICI treatment. We select complete responders by their stable microbiota functions and validate them using multiple external cohorts and experimentally. We identify major histocompatibility complex class I (MHC class I)-restricted peptides derived from flagellin-related genes of Lachnospiraceae (FLach) as structural homologs of tumor-associated antigens, detect FLach-reactive CD8+ T cells in complete responders before ICI therapy, and demonstrate that FLach peptides improve antitumor immunity. These findings highlight the prognostic value of microbial functions and therapeutic potential of tumor-mimicking microbial peptides.
Collapse
Affiliation(s)
- Angeli D G Macandog
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | - Carlotta Catozzi
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | - Mariaelena Capone
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori-IRCCS Fondazione G. Pascale, Naples 80131, Italy
| | - Amir Nabinejad
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | - Padma P Nanaware
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Shujing Liu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104-4238, USA
| | - Smita Vinjamuri
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612-7342, USA
| | - Johanna A Stunnenberg
- Netherlands Cancer Institute (NKI)-AVL, North Holland, Amsterdam 1066 CX, the Netherlands
| | - Serena Galiè
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | - Maria Giovanna Jodice
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | - Francesca Montani
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | - Federica Armanini
- Department of CIBIO, University of Trento, Trento, Povo 38123, Italy
| | - Ester Cassano
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | - Gabriele Madonna
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori-IRCCS Fondazione G. Pascale, Naples 80131, Italy
| | - Domenico Mallardo
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori-IRCCS Fondazione G. Pascale, Naples 80131, Italy
| | | | - Salvatore Pece
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | - Maria Tagliamonte
- Innovative Immunological Models, Istituto Nazionale Tumori-IRCCS Fondazione G. Pascale, Naples 80131, Italy
| | - Vito Vanella
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori-IRCCS Fondazione G. Pascale, Naples 80131, Italy
| | - Massimo Barberis
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | | | - Christian U Blank
- Netherlands Cancer Institute (NKI)-AVL, North Holland, Amsterdam 1066 CX, the Netherlands
| | - Marlene Bouvier
- Department of Microbiology and Immunology, College of Medicine, University of Illinois at Chicago, Chicago, IL 60612-7342, USA
| | - Miles C Andrews
- Department of Medicine, School of Translational Medicine, Monash University, Melbourne, VIC 3004, Australia
| | - Xiaowei Xu
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104-4238, USA
| | - Laura Santambrogio
- Department of Radiation Oncology, Weill Cornell Medicine, New York, NY 10065, USA
| | - Nicola Segata
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy; Department of CIBIO, University of Trento, Trento, Povo 38123, Italy
| | - Luigi Buonaguro
- Innovative Immunological Models, Istituto Nazionale Tumori-IRCCS Fondazione G. Pascale, Naples 80131, Italy
| | - Emilia Cocorocchio
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy
| | - Paolo A Ascierto
- Melanoma, Cancer Immunotherapy and Development Therapeutics Unit, Istituto Nazionale Tumori-IRCCS Fondazione G. Pascale, Naples 80131, Italy
| | - Teresa Manzo
- Department of Molecular Biotechnology and Health Sciences, University of Torino, Turin 10126, Italy
| | - Luigi Nezi
- Department of Experimental Oncology, Istituto Europeo di Oncologia-IRCCS, Milan 20139, Italy.
| |
Collapse
|
76
|
Bae M, Le C, Mehta RS, Dong X, Pieper LM, Ramirez L, Alexander M, Kiamehr S, Turnbaugh PJ, Huttenhower C, Chan AT, Balskus EP. Metatranscriptomics-guided discovery and characterization of a polyphenol-metabolizing gut microbial enzyme. Cell Host Microbe 2024; 32:1887-1896.e8. [PMID: 39471822 PMCID: PMC11585353 DOI: 10.1016/j.chom.2024.10.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2024] [Revised: 09/18/2024] [Accepted: 10/01/2024] [Indexed: 11/01/2024]
Abstract
Gut microbial catechol dehydroxylases are a largely uncharacterized family of metalloenzymes that potentially impact human health by metabolizing dietary polyphenols. Here, we use metatranscriptomics (MTX) to identify highly transcribed catechol-dehydroxylase-encoding genes in human gut microbiomes. We discover a prevalent, previously uncharacterized catechol dehydroxylase (Gp Hcdh) from Gordonibacter pamelaeae that dehydroxylates hydrocaffeic acid (HCA), an anti-inflammatory gut microbial metabolite derived from plant-based foods. Further analyses suggest that the activity of Gp Hcdh may reduce anti-inflammatory benefits of polyphenol-rich foods. Together, these results show the utility of combining MTX analysis and biochemical characterization for gut microbial enzyme discovery and reveal a potential link between host inflammation and a specific polyphenol-metabolizing gut microbial enzyme.
Collapse
Affiliation(s)
- Minwoo Bae
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Chi Le
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Raaj S Mehta
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA 02114, USA
| | - Xueyang Dong
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Lindsey M Pieper
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Lorenzo Ramirez
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Margaret Alexander
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA
| | - Sina Kiamehr
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA
| | - Peter J Turnbaugh
- Department of Microbiology and Immunology, University of California, San Francisco, San Francisco, CA 94143, USA; Chan Zuckerberg Biohub-San Francisco, San Francisco, CA 94158, USA
| | - Curtis Huttenhower
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Department of Biostatistics, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Harvard Chan Microbiome in Public Health Center, Harvard T.H. Chan School of Public Health, Harvard University, Boston, MA 02115, USA
| | - Andrew T Chan
- Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA 02114, USA; Clinical and Translational Epidemiology Unit, Massachusetts General Hospital, Boston, MA 02114, USA; Department of Immunology and Infectious Diseases, Harvard T.H. Chan School of Public Health, Boston, MA 02115, USA; Channing Division of Network Medicine, Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | - Emily P Balskus
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA 02138, USA; Broad Institute of MIT and Harvard, Cambridge, MA 02142, USA; Howard Hughes Medical Institute, Harvard University, Cambridge, MA 02138, USA.
| |
Collapse
|
77
|
Nickols WA, McIver LJ, Walsh A, Zhang Y, Nearing JT, Asnicar F, Punčochář M, Segata N, Nguyen LH, Hartmann EM, Franzosa EA, Huttenhower C, Thompson KN. Evaluating metagenomic analyses for undercharacterized environments: what's needed to light up the microbial dark matter? BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.08.622677. [PMID: 39574575 PMCID: PMC11580994 DOI: 10.1101/2024.11.08.622677] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/29/2024]
Abstract
Non-human-associated microbial communities play important biological roles, but they remain less understood than human-associated communities. Here, we assess the impact of key environmental sample properties on a variety of state-of-the-art metagenomic analysis methods. In simulated datasets, all methods performed similarly at high taxonomic ranks, but newer marker-based methods incorporating metagenomic assembled genomes outperformed others at lower taxonomic levels. In real environmental data, taxonomic profiles assigned to the same sample by different methods showed little agreement at lower taxonomic levels, but the methods agreed better on community diversity estimates and estimates of the relationships between environmental parameters and microbial profiles.
Collapse
Affiliation(s)
- William A. Nickols
- Department of Biostatistics, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
- Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Lauren J. McIver
- Department of Biostatistics, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
- Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, MA, USA
| | - Aaron Walsh
- Department of Biostatistics, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
- Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Yancong Zhang
- Department of Biostatistics, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
- Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Jacob T. Nearing
- Department of Biostatistics, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
- Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Francesco Asnicar
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Michal Punčochář
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Nicola Segata
- Department of Cellular, Computational and Integrative Biology (CIBIO), University of Trento, Trento, Italy
| | - Long H. Nguyen
- Department of Biostatistics, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
- Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
| | - Erica M. Hartmann
- Department of Civil and Environmental Engineering, McCormick School of Engineering, Northwestern University, Evanston, IL, USA
- Center for Synthetic Biology, Northwestern University, Evanston, IL, USA
- Department of Medicine/Division of Pulmonary Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL, USA
| | - Eric A. Franzosa
- Department of Biostatistics, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
- Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Curtis Huttenhower
- Department of Biostatistics, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
- Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA
- Department of Immunology and Infectious Diseases, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
| | - Kelsey N. Thompson
- Department of Biostatistics, T.H. Chan School of Public Health, Harvard University, Boston, MA, USA
- Harvard Chan Microbiome in Public Health Center, Harvard T. H. Chan School of Public Health, Boston, MA, USA
- Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, MA, USA
| |
Collapse
|
78
|
Chen L, Tan KML, Xu J, Mishra P, Mir SA, Gong M, Narasimhan K, Ng B, Lai JS, Tint MT, Cai S, Sadananthan SA, Michael N, Yaligar J, Velan SS, Leow MKS, Tan KH, Chan J, Meaney MJ, Chan SY, Chong YS, Eriksson JG. Exploring multi-omics and clinical characteristics linked to accelerated biological aging in Asian women of reproductive age: insights from the S-PRESTO study. Genome Med 2024; 16:128. [PMID: 39516835 PMCID: PMC11549770 DOI: 10.1186/s13073-024-01403-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Accepted: 10/29/2024] [Indexed: 11/16/2024] Open
Abstract
BACKGROUND Phenotypic age (PhenoAge), a widely used marker of biological aging, has been shown to be a robust predictor of all-cause mortality and morbidity in different populations. Existing studies on biological aging have primarily focused on individual domains, resulting in a lack of a comprehensive understanding of the multi-systemic dysregulation that occurs in aging. METHODS PhenoAge was evaluated based on a linear combination of chronological age (CA) and 9 clinical biomarkers in 952 multi-ethnic Asian women of reproductive age. Phenotypic age acceleration (PhenoAgeAccel), an aging biomarker, represents PhenoAge after adjusting for CA. This study conducts an in-depth association analysis of PhenoAgeAccel with clinical, nutritional, lipidomic, gut microbiome, and genetic factors. RESULTS Higher adiposity, glycaemia, plasma saturated fatty acids, kynurenine pathway metabolites, GlycA, riboflavin, nicotinamide, and insulin-like growth factor binding proteins were positively associated with PhenoAgeAccel. Conversely, a healthier diet and higher levels of pyridoxal phosphate, all-trans retinol, betaine, tryptophan, glutamine, histidine, apolipoprotein B, and insulin-like growth factors were inversely associated with PhenoAgeAccel. Lipidomic analysis found 132 lipid species linked to PhenoAgeAccel, with PC(O-36:0) showing the strongest positive association and CE(24:5) demonstrating the strongest inverse association. A genome-wide association study identified rs9864994 as the top genetic variant (P = 5.69E-07) from the ZDHHC19 gene. Gut microbiome analysis revealed that Erysipelotrichaceae UCG-003 and Bacteroides vulgatus were inversely associated with PhenoAgeAccel. Integrative network analysis of aging-related factors underscored the intricate links among clinical, nutritional and lipidomic variables, such as positive associations between kynurenine pathway metabolites, amino acids, adiposity, and insulin resistance. Furthermore, potential mediation effects of blood biomarkers related to inflammation, immune response, and nutritional and energy metabolism were observed in the associations of diet, adiposity, genetic variants, and gut microbial species with PhenoAgeAccel. CONCLUSIONS Our findings provide a comprehensive analysis of aging-related factors across multiple platforms, delineating their complex interconnections. This study is the first to report novel signatures in lipidomics, gut microbiome and blood biomarkers specifically associated with PhenoAgeAccel. These insights are invaluable in understanding the molecular and metabolic mechanisms underlying biological aging and shed light on potential interventions to mitigate accelerated biological aging by targeting modifiable factors.
Collapse
Affiliation(s)
- Li Chen
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore.
| | - Karen Mei-Ling Tan
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore.
- Department of Laboratory Medicine, National University Hospital, Singapore, Singapore.
| | - Jia Xu
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Priti Mishra
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Sartaj Ahmad Mir
- Singapore Lipidomics Incubator, Life Sciences Institute, National University of Singapore, Singapore, Singapore
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Min Gong
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Kothandaraman Narasimhan
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Bryan Ng
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jun Shi Lai
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Mya Thway Tint
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Shirong Cai
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Suresh Anand Sadananthan
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Navin Michael
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Jadegoud Yaligar
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
| | - Sambasivam Sendhil Velan
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Medicine and Human Potential Translational Research Programme. Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Melvin Khee Shing Leow
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Endocrinology, Tan Tock Seng Hospital, Singapore, Singapore
- Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore, Singapore
- Duke-NUS Medical School, Singapore, Singapore
- Department of Obstetrics and Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Kok Hian Tan
- Duke-NUS Medical School, Singapore, Singapore
- KK Women's and Children's Hospital, Singapore, Singapore
| | - Jerry Chan
- Duke-NUS Medical School, Singapore, Singapore
- KK Women's and Children's Hospital, Singapore, Singapore
| | - Michael J Meaney
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Sackler Program for Epigenetics & Psychobiology, McGill University, Montréal, Canada
- Ludmer Centre for Neuroinformatics and Mental Health, Douglas Mental Health University Institute, McGill University, Montréal, Canada
| | - Shiao-Yng Chan
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Obstetrics and Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Yap Seng Chong
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Obstetrics and Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Johan G Eriksson
- Institute for Human Development and Potential, Agency for Science, Technology and Research (A*STAR), Singapore, Singapore
- Department of Obstetrics and Gynaecology and Human Potential Translational Research Programme, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- Folkhalsan Research Center, Helsinki, Finland
- Department of General Practice and Primary Health Care, University of Helsinki, Helsinki, Finland
| |
Collapse
|
79
|
Rooney CM, Jeffery IB, Mankia K, Wilcox MH, Emery P. Dynamics of the gut microbiome in individuals at risk of rheumatoid arthritis: a cross-sectional and longitudinal observational study. Ann Rheum Dis 2024:ard-2024-226362. [PMID: 39515835 DOI: 10.1136/ard-2024-226362] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2024] [Accepted: 10/03/2024] [Indexed: 11/16/2024]
Abstract
OBJECTIVES This work aimed to resolve the conflicting reports on Prevotellaceae abundance in the development of rheumatoid arthritis (RA) and to observe structural, functional and temporal changes in the gut microbiome in RA progressors versus non-progressors. METHODS Individuals at risk of RA were defined by the presence of anticyclic citrullinated protein (anti-CCP) antibodies and new musculoskeletal symptoms without clinical synovitis. Baseline sampling included 124 participants (30 progressed to RA), with longitudinal sampling of 19 participants (5 progressed to RA) over 15 months at five timepoints. Gut microbiome taxonomic alterations were investigated using 16S rRNA amplicon sequencing and confirmed with shotgun metagenomic DNA sequencing on 49 samples. RESULTS At baseline, CCP+ at risk progressors showed significant differences in Prevotellaceae abundance compared with non-progressors, contingent on intrinsic RA risk factors and time to progression. Longitudinal sampling revealed gut microbiome instability in progressors 10 months before RA onset, a phenomenon absent in non-progressors. This may indicate a late microbial shift before RA onset, with Prevotellaceae contributing but not dominating these changes. Structural changes in the gut microbiome during arthritis development were associated with increased amino acid metabolism. CONCLUSION These data suggest conflicting reports on Prevotellaceae overabundance are likely due to sampling within a heterogeneous population along a dynamic disease spectrum, with certain Prevotellaceae strains/clades possibly contributing to the establishment and/or progression of RA. Gut microbiome changes in RA may appear at the transition to clinical arthritis as a late manifestation, and it remains unclear whether they represent a primary or secondary phenomenon.
Collapse
Affiliation(s)
| | | | - Kulveer Mankia
- University of Leeds, Leeds Institute of Rheumatic and Musculoskeletal Medicine, Leeds, UK
- NIHR Leeds Musculoskeletal Biomedical Research Centre, Leeds, UK
| | - Mark H Wilcox
- Leeds Institute of Medical Research, University of Leeds, Leeds, UK
| | - Paul Emery
- University of Leeds, Leeds Institute of Rheumatic and Musculoskeletal Medicine, Leeds, UK
- NIHR Leeds Musculoskeletal Biomedical Research Centre, Leeds, UK
| |
Collapse
|
80
|
Heo SH, Choi BY, Kang J, Jung JY, Kim HC, Lee SJ, Sul WJ, Lee SW. Dynamics of the airway microbiome in response to exposure to particulate matter 2.5 in patients with chronic obstructive pulmonary disease. THE SCIENCE OF THE TOTAL ENVIRONMENT 2024; 956:177314. [PMID: 39489443 DOI: 10.1016/j.scitotenv.2024.177314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 10/28/2024] [Accepted: 10/29/2024] [Indexed: 11/05/2024]
Abstract
BACKGROUND Particulate matter (PM) and air pollution have been suggested to be associated with chronic obstructive pulmonary disease (COPD), contributing significantly to global respiratory disease-related mortality. This study aimed to investigate whether seasonal exposure to PM influences dysbiosis in the respiratory microbiota of patients with COPD. METHODS Sputum samples were collected four times over 1 year from 102 patients with COPD, and 16S rRNA sequencing was performed. The dynamics of the airway microbiota were analyzed depending on PM exposure levels and season. RESULTS The PM-low exposure group had higher α-diversity compared to the PM-high exposure group, particularly noted in spring. Some bacterial groups, including seven species such as Treponema socranskii, were more abundant in the low exposure group. Additionally, the bacterial community structure in summer significantly differed from that in other seasons, with significantly increased α-diversity in this season. The difference in the airway microbiome due to PM exposure was prominent in patients with moderate COPD. CONCLUSIONS PM exposure may influence changes in the sputum microbiome depending on exposure levels and seasonal variations. Our results suggest that airway microbiomes could vary with PM exposure according to seasonal trends.
Collapse
Affiliation(s)
- Sun-Hee Heo
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea
| | - Bo-Yun Choi
- Department of Systems Biotechnology, Chung-Ang University, Anseong, Gyeonggi-do, Republic of Korea
| | - Jieun Kang
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Ilsan Paik Hospital, Inje University College of Medicine, Goyang, Republic of Korea
| | - Ji Ye Jung
- Division of Pulmonary and Critical Care Medicine, Department of Internal Medicine, Severance Hospital, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Hwan-Cheol Kim
- Department of Occupational and Environmental Medicine, Inha University Hospital, Inha University College of Medicine, Incheon, Republic of Korea
| | - Seon-Jin Lee
- Environmental Disease Research Center, Korea Research Institute of Bioscience and Biotechnology, Daejeon, Republic of Korea
| | - Woo Jun Sul
- Department of Systems Biotechnology, Chung-Ang University, Anseong, Gyeonggi-do, Republic of Korea.
| | - Sei Won Lee
- Department of Pulmonary and Critical Care Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
81
|
Van Den Ham KM, Bower LK, Li S, Lorenzi H, Doumbo S, Doumtabe D, Kayentao K, Ongoiba A, Traore B, Crompton PD, Schmidt NW. The gut microbiome is associated with susceptibility to febrile malaria in Malian children. Nat Commun 2024; 15:9525. [PMID: 39500866 PMCID: PMC11538534 DOI: 10.1038/s41467-024-52953-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Accepted: 09/23/2024] [Indexed: 11/08/2024] Open
Abstract
Malaria is a major public health problem, but many of the factors underlying the pathogenesis of this disease are not well understood, including protection from the development of febrile symptoms, which is observed in individuals residing in areas with moderate-to-high transmission by early adolescence. Here, we demonstrate that susceptibility to febrile malaria following Plasmodium falciparum infection is associated with the composition of the gut microbiome prior to the malaria season in 10-year-old Malian children, but not in younger children. Gnotobiotic mice colonized with the fecal samples of malaria-susceptible children were shown to have a significantly higher parasite burden following Plasmodium infection compared to gnotobiotic mice colonized with the fecal samples of malaria-resistant children. The fecal microbiome of the susceptible children was determined to be enriched for bacteria associated with inflammation, mucin degradation and gut permeability, and to have increased levels of nitric oxide-derived DNA adducts and lower levels of mucus phospholipids compared to the resistant children. Overall, these results indicate that the composition of the gut microbiome is associated with the prospective risk of febrile malaria in Malian children and suggest that modulation of the gut microbiome could decrease malaria morbidity in endemic areas.
Collapse
Affiliation(s)
- Kristin M Van Den Ham
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Layne K Bower
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Shanping Li
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Hernan Lorenzi
- Infectious Diseases Group, J. Craig Venter Institute, Bethesda, MD, USA
| | - Safiatou Doumbo
- Mali International Center of Excellence in Research; Malaria Research and Training Center, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Didier Doumtabe
- Mali International Center of Excellence in Research; Malaria Research and Training Center, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Kassoum Kayentao
- Mali International Center of Excellence in Research; Malaria Research and Training Center, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Aissata Ongoiba
- Mali International Center of Excellence in Research; Malaria Research and Training Center, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Boubacar Traore
- Mali International Center of Excellence in Research; Malaria Research and Training Center, University of Sciences, Techniques and Technologies of Bamako, Bamako, Mali
| | - Peter D Crompton
- Malaria Infection Biology and Immunity Section, Laboratory of Immunogenetics, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Rockville, MD, USA
| | - Nathan W Schmidt
- Ryan White Center for Pediatric Infectious Diseases and Global Health, Herman B Wells Center for Pediatric Research, Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
82
|
Yu X, Xiong T, Yu L, Liu G, Yang F, Li X, Wei Y, Wang X, Wei S, Jiang Y, Kong X, Ren S, Shi Y. Gut microbiome and metabolome profiling in coal workers' pneumoconiosis: potential links to pulmonary function. Microbiol Spectr 2024; 12:e0004924. [PMID: 39283109 PMCID: PMC11537036 DOI: 10.1128/spectrum.00049-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Accepted: 07/02/2024] [Indexed: 11/07/2024] Open
Abstract
Coal workers' pneumoconiosis (CWP) is a severe occupational disease resulting from prolonged exposure to coal dust. However, its pathogenesis remains elusive, compounded by a lack of early detection markers and effective treatments. Although the impact of gut microbiota on lung diseases is acknowledged, its specific role in CWP is unclear. This study aims to explore changes in the gut microbiome and metabolome in CWP, while also assessing the correlation between gut microbes and alterations in lung function. Fecal specimens from 43 CWP patients and 48 dust-exposed workers (DEW) were examined using 16S rRNA gene sequencing for microbiota and liquid chromatography-mass spectrometry for metabolite profiling. We observed similar gut microbial α-diversity but significant differences in flora composition (β-diversity) between patients with CWP and the DEW group. After adjusting for age using multifactorial linear regression analysis (MaAsLin2), the distinct gut microbiome profile in CWP patients revealed an increased presence of pro-inflammatory microorganisms such as Klebsiella and Haemophilus. Furthermore, in CWP patients, alterations in gut microbiota-particularly reduced α-diversity and changes in microbial composition-were significantly correlated with impaired pulmonary function, a relationship not observed in DEW. This underscores the specific impact of gut microbiota on pulmonary health in individuals with CWP. Metabolomic analysis of fecal samples from CWP patients and DEW identified 218 differential metabolites between the two groups, with a predominant increase in metabolites in CWP patients, suggesting enhanced metabolic activity in CWP. Key altered metabolites included various lipids, amino acids, and organic compounds, with silibinin emerging as a potential biomarker. Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis linked these metabolites to pathways relevant to the development of pulmonary fibrosis. Additionally, studies on the interaction between microbiota and metabolites showed positive correlations between certain bacteria and increased metabolites in CWP, further elucidating the complex interplay in this disease state. Our findings suggest a potential contributory role of gut microbiota in CWP pathogenesis through metabolic regulation, with implications for diagnostic biomarkers and understanding disease mechanisms, warranting further molecular investigation. IMPORTANCE The findings have significant implications for the early diagnosis and treatment of coal workers' pneumoconiosis, highlighting the potential of gut microbiota as diagnostic biomarkers. They pave the way for new research into gut microbiota-based therapeutic strategies, potentially focusing on modifying gut microbiota to mitigate disease progression.
Collapse
Affiliation(s)
- Xiao Yu
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Tao Xiong
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Lu Yu
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Gaisheng Liu
- Quality Control Office, Xishan Occupational Disease Prevention and Control Institute, Taiyuan, China
| | - Fan Yang
- Quality Control Office, Xishan Occupational Disease Prevention and Control Institute, Taiyuan, China
| | - Xueqin Li
- Department of Respiratory Medicine, Jincheng General Hospital, Shanxi, China
| | - Yangyang Wei
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaojing Wang
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Shuting Wei
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yi Jiang
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Xiaomei Kong
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Shouan Ren
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| | - Yiwei Shi
- NHC Key Laboratory of Pneumoconiosis, Shanxi Key Laboratory of Respiratory Diseases, Department of Pulmonary and Critical Care Medicine, The First Hospital of Shanxi Medical University, Taiyuan, China
| |
Collapse
|
83
|
Yergaliyev T, Künzel S, Hanauska A, Rees A, Wild KJ, Pétursdóttir ÁH, Gunnlaugsdóttir H, Reynolds CK, Humphries DJ, Rodehutscord M, Camarinha-Silva A. The effect of Asparagopsis taxiformis, Ascophyllum nodosum, and Fucus vesiculosus on ruminal methanogenesis and metagenomic functional profiles in vitro. Microbiol Spectr 2024; 12:e0394223. [PMID: 39347544 PMCID: PMC11542596 DOI: 10.1128/spectrum.03942-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2023] [Accepted: 07/08/2024] [Indexed: 10/01/2024] Open
Abstract
The ruminant-microorganism symbiosis is unique by providing high-quality food from fibrous materials but also contributes to the production of one of the most potent greenhouse gases-methane. Mitigating methanogenesis in ruminants has been a focus of interest in the past decades. One of the promising strategies to combat methane production is the use of feed supplements, such as seaweeds, that might mitigate methanogenesis via microbiome modulation and direct chemical inhibition. We conducted in vitro investigations of the effect of three seaweeds (Ascophyllum nodosum, Asparagopsis taxiformis, and Fucus vesiculosus) harvested at different locations (Iceland, Scotland, and Portugal) on methane production. We applied metataxonomics (16S rRNA gene amplicons) and metagenomics (shotgun) methods to uncover the interplay between the microbiome's taxonomical and functional states, methanogenesis rates, and seaweed supplementations. Methane concentration was reduced by A. nodosum and F. vesiculosus, both harvested in Scotland and A. taxiformis, with the greatest effect of the latter. A. taxiformis acted through the reduction of archaea-to-bacteria ratios but not eukaryotes-to-bacteria. Moreover, A. taxiformis application was accompanied by shifts in both taxonomic and functional profiles of the microbial communities, decreasing not only archaeal ratios but also abundances of methanogenesis-associated functions. Methanobrevibacter "SGMT" (M. smithii, M. gottschalkii, M. millerae or M. thaueri; high methane yield) to "RO" (M. ruminantium and M. olleyae; low methane yield) clades ratios were also decreased, indicating that A. taxiformis application favored Methanobrevibacter species that produce less methane. Most of the functions directly involved in methanogenesis were less abundant, while the abundances of the small subset of functions that participate in methane assimilation were increased. IMPORTANCE The application of A. taxiformis significantly reduced methane production in vitro. We showed that this reduction was linked to changes in microbial function profiles, the decline in the overall archaeal community counts, and shifts in ratios of Methanobrevibacter "SGMT" and "RO" clades. A. nodosum and F. vesiculosus, obtained from Scotland, also decreased methane concentration in the total gas, while the same seaweed species from Iceland did not.
Collapse
Affiliation(s)
- Timur Yergaliyev
- Institute of Animal
Science, University of Hohenheim,
Stuttgart, Germany
- HoLMiR - Hohenheim
Center for Livestock Microbiome Research, University of
Hohenheim, Stuttgart,
Germany
| | - Susanne Künzel
- Institute of Animal
Science, University of Hohenheim,
Stuttgart, Germany
- HoLMiR - Hohenheim
Center for Livestock Microbiome Research, University of
Hohenheim, Stuttgart,
Germany
| | - Anna Hanauska
- Institute of Animal
Science, University of Hohenheim,
Stuttgart, Germany
| | - Antonia Rees
- Institute of Animal
Science, University of Hohenheim,
Stuttgart, Germany
| | - Katharina J. Wild
- Institute of Animal
Science, University of Hohenheim,
Stuttgart, Germany
| | | | | | - Christopher K. Reynolds
- School of Agriculture,
Policy and Development, University of
Reading, Reading,
United Kingdom
| | - David J. Humphries
- School of Agriculture,
Policy and Development, University of
Reading, Reading,
United Kingdom
| | - Markus Rodehutscord
- Institute of Animal
Science, University of Hohenheim,
Stuttgart, Germany
- HoLMiR - Hohenheim
Center for Livestock Microbiome Research, University of
Hohenheim, Stuttgart,
Germany
| | - Amélia Camarinha-Silva
- Institute of Animal
Science, University of Hohenheim,
Stuttgart, Germany
- HoLMiR - Hohenheim
Center for Livestock Microbiome Research, University of
Hohenheim, Stuttgart,
Germany
| |
Collapse
|
84
|
Fang Y, Liu X, Ren J, Wang X, Zhou F, Huang S, You L, Zhao Y. Integrated analysis of microbiome and metabolome reveals signatures in PDAC tumorigenesis and prognosis. Microbiol Spectr 2024; 12:e0096224. [PMID: 39387592 PMCID: PMC11540152 DOI: 10.1128/spectrum.00962-24] [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: 04/29/2024] [Accepted: 09/16/2024] [Indexed: 10/15/2024] Open
Abstract
Pancreatic cancer, predominantly pancreatic ductal adenocarcinoma (PDAC), is one of the most malignant tumors of the digestive system. Emerging evidence suggests the involvement of the microbiome and metabolic substances in the development of PDAC, yet the results remain contradictory. This study aims to identify the alterations and relationships in intratumoral microbiome and metabolites in PDAC. We collected matched tumor and normal adjacent tissue (NAT) samples from 105 PDAC patients and performed a 6-year follow-up. 2bRAD-M sequencing, untargeted liquid chromatography-tandem mass spectrometry, and untargeted gas chromatography-mass spectrometry were performed. Compared with NATs, microbial α-diversity decreased in PDAC tumors. The relative abundance of Staphylococcus aureus, Cutibacterium acnes, and Cutibacterium granulosum was higher in PDAC tumor after adjusting for confounding factors body mass index and M stage, and the presence of Ralstonia pickettii_B was found associated with a worse overall survival. Metabolomic analysis revealed distinctive differences in composition between PDAC and NAT, with 553 discriminative metabolites identified. Differential metabolites were revealed to originate from the microbiota and showed significant interactions with shifted bacterial species through KO (KEGG Orthology) genes. These findings suggest that the PDAC microenvironment harbors unique microbial-derived enzymatic reactions, potentially influencing the occurrence and development of PDAC by modulating the levels of glycerol-3-phosphate, succinate, carbonate, and beta-alanine. IMPORTANCE We conducted a large sample-size pancreatic adenocarcinoma microbiome study using a novel microbiome sequencing method and two metabolomic assays. Two significant outcomes of our analysis are: (i) commensal opportunistic pathogens Staphylococcus aureus, Cutibacterium acnes, and Cutibacterium granulosum were enriched in pancreatic ductal adenocarcinoma (PDAC) tumors compared with normal adjacent tissues, and (ii) worse overall survival was found related to the presence of Ralstonia pickettii_B. Microbial species affect the tumorigenesis, metastasis, and prognosis of PDAC via unique microbe-enzyme-metabolite interaction. Thus, our study highlights the need for further investigation of the potential associations between pancreatic microbiota-derived omics signatures, which may drive the clinical transformation of microbiome-derived strategies toward therapy-targeted bacteria.
Collapse
Affiliation(s)
- Yuan Fang
- Department of General
Surgery, Peking Union Medical College Hospital, Peking Union Medical
College, Chinese Academy of Medical
Sciences, Beijing,
China
- Key Laboratory of
Research in Pancreatic Tumor, Chinese Academy of Medical
Sciences, Beijing,
China
- National Science and
Technology Key Infrastructure on Translational Medicine in Peking Union
Medical College Hospital,
Beijing, China
- State Key Laboratory
of Complex Severe and Rare Diseases, Peking Union Medical College
Hospital, Chinese Academy of Medical Sciences and Peking Union Medical
College, Beijing,
China
| | - Xiaohong Liu
- Department of General
Surgery, Peking Union Medical College Hospital, Peking Union Medical
College, Chinese Academy of Medical
Sciences, Beijing,
China
- Key Laboratory of
Research in Pancreatic Tumor, Chinese Academy of Medical
Sciences, Beijing,
China
- National Science and
Technology Key Infrastructure on Translational Medicine in Peking Union
Medical College Hospital,
Beijing, China
- State Key Laboratory
of Complex Severe and Rare Diseases, Peking Union Medical College
Hospital, Chinese Academy of Medical Sciences and Peking Union Medical
College, Beijing,
China
| | - Jie Ren
- Department of General
Surgery, Peking Union Medical College Hospital, Peking Union Medical
College, Chinese Academy of Medical
Sciences, Beijing,
China
- Key Laboratory of
Research in Pancreatic Tumor, Chinese Academy of Medical
Sciences, Beijing,
China
- National Science and
Technology Key Infrastructure on Translational Medicine in Peking Union
Medical College Hospital,
Beijing, China
- State Key Laboratory
of Complex Severe and Rare Diseases, Peking Union Medical College
Hospital, Chinese Academy of Medical Sciences and Peking Union Medical
College, Beijing,
China
| | - Xing Wang
- Department of General
Surgery, Peking Union Medical College Hospital, Peking Union Medical
College, Chinese Academy of Medical
Sciences, Beijing,
China
- Key Laboratory of
Research in Pancreatic Tumor, Chinese Academy of Medical
Sciences, Beijing,
China
- National Science and
Technology Key Infrastructure on Translational Medicine in Peking Union
Medical College Hospital,
Beijing, China
- State Key Laboratory
of Complex Severe and Rare Diseases, Peking Union Medical College
Hospital, Chinese Academy of Medical Sciences and Peking Union Medical
College, Beijing,
China
| | - Feihan Zhou
- Department of General
Surgery, Peking Union Medical College Hospital, Peking Union Medical
College, Chinese Academy of Medical
Sciences, Beijing,
China
- Key Laboratory of
Research in Pancreatic Tumor, Chinese Academy of Medical
Sciences, Beijing,
China
- National Science and
Technology Key Infrastructure on Translational Medicine in Peking Union
Medical College Hospital,
Beijing, China
- State Key Laboratory
of Complex Severe and Rare Diseases, Peking Union Medical College
Hospital, Chinese Academy of Medical Sciences and Peking Union Medical
College, Beijing,
China
| | - Shi Huang
- Faculty of Dentistry,
The University of Hong Kong, Hong
Kong SAR, China
| | - Lei You
- Department of General
Surgery, Peking Union Medical College Hospital, Peking Union Medical
College, Chinese Academy of Medical
Sciences, Beijing,
China
- Key Laboratory of
Research in Pancreatic Tumor, Chinese Academy of Medical
Sciences, Beijing,
China
- National Science and
Technology Key Infrastructure on Translational Medicine in Peking Union
Medical College Hospital,
Beijing, China
- State Key Laboratory
of Complex Severe and Rare Diseases, Peking Union Medical College
Hospital, Chinese Academy of Medical Sciences and Peking Union Medical
College, Beijing,
China
| | - Yupei Zhao
- Department of General
Surgery, Peking Union Medical College Hospital, Peking Union Medical
College, Chinese Academy of Medical
Sciences, Beijing,
China
- Key Laboratory of
Research in Pancreatic Tumor, Chinese Academy of Medical
Sciences, Beijing,
China
- National Science and
Technology Key Infrastructure on Translational Medicine in Peking Union
Medical College Hospital,
Beijing, China
- State Key Laboratory
of Complex Severe and Rare Diseases, Peking Union Medical College
Hospital, Chinese Academy of Medical Sciences and Peking Union Medical
College, Beijing,
China
| |
Collapse
|
85
|
Won S, Cho S, Kim H. rRNA operon improves species-level classification of bacteria and microbial community analysis compared to 16S rRNA. Microbiol Spectr 2024; 12:e0093124. [PMID: 39365049 PMCID: PMC11537084 DOI: 10.1128/spectrum.00931-24] [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: 04/12/2024] [Accepted: 08/09/2024] [Indexed: 10/05/2024] Open
Abstract
Precise identification of species is fundamental in microbial genomics and is crucial for understanding the microbial communities. While the 16S rRNA gene, particularly its V3-V4 regions, has been extensively employed for microbial identification, however has limitations in achieving species-level resolution. Advancements in long-read sequencing technologies have highlighted the rRNA operon as a more accurate marker for microbial classification and analysis than the 16S rRNA gene. This study aims to compare the accuracy of species classification and microbial community analysis using the rRNA operon versus the 16S rRNA gene. We evaluated the species classification accuracy of the rRNA operon,16S rRNA gene, and 16S rRNA V3-V4 regions using a BLAST-based method and a k-mer matching-based method with public data available from NCBI. We further performed simulations to model microbial community analysis. We accessed the performance using each marker in community composition estimation and differential abundance analysis. Our findings demonstrate that the rRNA operon offers an advantage over the 16S rRNA gene and its V3-V4 regions for species-level classification within the genus. When applied to microbial community analysis, the rRNA operon enables a more accurate determination of composition. Using the rRNA operon yielded more reliable results in differential abundance analysis as well. IMPORTANCE We quantitatively demonstrated that the rRNA operon outperformed the 16S rRNA and its V3-V4 regions in accuracy for both individual species identification and species-level microbial community analysis. Our findings can provide guidelines for selecting appropriate markers in the field of microbial research.
Collapse
Affiliation(s)
- Sohyoung Won
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, Republic of Korea
- eGnome, Inc., Seoul, Republic of Korea
| | - Seoae Cho
- eGnome, Inc., Seoul, Republic of Korea
| | - Heebal Kim
- Interdisciplinary Program in Bioinformatics, Seoul National University, Seoul, Republic of Korea
- eGnome, Inc., Seoul, Republic of Korea
- Department of Agricultural Biotechnology, Research Institute of Agriculture and Life Sciences, Seoul National University, Seoul, Republic of Korea
| |
Collapse
|
86
|
Tamburini FB, Tripathi A, Gold MP, Yang JC, Biancalani T, McBride JM, Keir ME, GARDENIA Study Group. Gut Microbial Species and Endotypes Associate with Remission in Ulcerative Colitis Patients Treated with Anti-TNF or Anti-integrin Therapy. J Crohns Colitis 2024; 18:1819-1831. [PMID: 38836628 PMCID: PMC11532613 DOI: 10.1093/ecco-jcc/jjae084] [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: 02/23/2024] [Revised: 04/19/2024] [Accepted: 06/03/2024] [Indexed: 06/06/2024]
Abstract
BACKGROUND AND AIMS The gut microbiota contributes to aberrant inflammation in inflammatory bowel disease, but the bacterial factors causing or exacerbating inflammation are not fully understood. Further, the predictive or prognostic value of gut microbial biomarkers for remission in response to biologic therapy is unclear. METHODS We perform whole metagenomic sequencing of 550 stool samples from 287 ulcerative colitis patients from a large, phase 3, head-to-head study of infliximab and etrolizumab. RESULTS We identify several bacterial species in baseline and/or post-treatment samples that associate with clinical remission. These include previously described associations [Faecalibacterium prausnitzii_F] as well as new associations with remission to biologic therapy [Flavonifractor plautii]. We build multivariate models and find that gut microbial species are better predictors for remission than clinical variables alone. Finally, we describe patient groups that differ in microbiome composition and remission rate after induction therapy, suggesting the potential utility of microbiome-based endotyping. CONCLUSIONS In this large study of ulcerative colitis patients, we show that few individual species associate strongly with clinical remission, but multivariate models including microbiome can predict clinical remission and have better predictive power compared with clinical data alone.
Collapse
Affiliation(s)
- Fiona B Tamburini
- Human Pathobiology & OMNI Reverse Translation, Genentech, South San Francisco, CA, USA
| | | | - Maxwell P Gold
- Biological Research & AI Development, Genentech, South San Francisco, CA, USA
| | - Julianne C Yang
- Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Tommaso Biancalani
- Biological Research & AI Development, Genentech, South San Francisco, CA, USA
| | - Jacqueline M McBride
- Translational Medicine OMNI-Biomarker Development, Genentech, South San Francisco, CA, USA
| | - Mary E Keir
- Human Pathobiology & OMNI Reverse Translation, Genentech, South San Francisco, CA, USA
| | - GARDENIA Study Group
- Human Pathobiology & OMNI Reverse Translation, Genentech, South San Francisco, CA, USA
| |
Collapse
|
87
|
Nishijima S, Stankevic E, Aasmets O, Schmidt TSB, Nagata N, Keller MI, Ferretti P, Juel HB, Fullam A, Robbani SM, Schudoma C, Hansen JK, Holm LA, Israelsen M, Schierwagen R, Torp N, Telzerow A, Hercog R, Kandels S, Hazenbrink DHM, Arumugam M, Bendtsen F, Brøns C, Fonvig CE, Holm JC, Nielsen T, Pedersen JS, Thiele MS, Trebicka J, Org E, Krag A, Hansen T, Kuhn M, Bork P. Fecal microbial load is a major determinant of gut microbiome variation and a confounder for disease associations. Cell 2024:S0092-8674(24)01204-2. [PMID: 39541968 DOI: 10.1016/j.cell.2024.10.022] [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: 03/08/2024] [Revised: 07/12/2024] [Accepted: 10/14/2024] [Indexed: 11/17/2024]
Abstract
The microbiota in individual habitats differ in both relative composition and absolute abundance. While sequencing approaches determine the relative abundances of taxa and genes, they do not provide information on their absolute abundances. Here, we developed a machine-learning approach to predict fecal microbial loads (microbial cells per gram) solely from relative abundance data. Applying our prediction model to a large-scale metagenomic dataset (n = 34,539), we demonstrated that microbial load is the major determinant of gut microbiome variation and is associated with numerous host factors, including age, diet, and medication. We further found that for several diseases, changes in microbial load, rather than the disease condition itself, more strongly explained alterations in patients' gut microbiome. Adjusting for this effect substantially reduced the statistical significance of the majority of disease-associated species. Our analysis reveals that the fecal microbial load is a major confounder in microbiome studies, highlighting its importance for understanding microbiome variation in health and disease.
Collapse
Affiliation(s)
- Suguru Nishijima
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Evelina Stankevic
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Oliver Aasmets
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Thomas S B Schmidt
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Naoyoshi Nagata
- Department of Gastroenterological Endoscopy, Tokyo Medical University, Tokyo, Japan
| | - Marisa Isabell Keller
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Pamela Ferretti
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Helene Bæk Juel
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Anthony Fullam
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | | | - Christian Schudoma
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Johanne Kragh Hansen
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark; Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Louise Aas Holm
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; The Children's Obesity Clinic, Department of Pediatrics, Copenhagen University Hospital Holbæk, Holbæk, Denmark
| | - Mads Israelsen
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark; Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Robert Schierwagen
- Department of Internal Medicine B, University of Münster, Münster, Germany
| | - Nikolaj Torp
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark; Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Anja Telzerow
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Rajna Hercog
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Stefanie Kandels
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Diënty H M Hazenbrink
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Manimozhiyan Arumugam
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Flemming Bendtsen
- Gastrounit, Medical Division, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Charlotte Brøns
- Clinical Research, Steno Diabetes Center Copenhagen, Herlev, Denmark
| | - Cilius Esmann Fonvig
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; The Children's Obesity Clinic, Department of Pediatrics, Copenhagen University Hospital Holbæk, Holbæk, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Jens-Christian Holm
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; The Children's Obesity Clinic, Department of Pediatrics, Copenhagen University Hospital Holbæk, Holbæk, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Trine Nielsen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark; Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark; Medical department, University Hospital Zeeland, Køge, Denmark
| | - Julie Steen Pedersen
- Gastrounit, Medical Division, Copenhagen University Hospital Hvidovre, Hvidovre, Denmark
| | - Maja Sofie Thiele
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark; Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Jonel Trebicka
- Department of Internal Medicine B, University of Münster, Münster, Germany; European Foundation for the Study of Chronic Liver Failure, EFCLIF, Barcelona, Spain
| | - Elin Org
- Institute of Genomics, University of Tartu, Tartu, Estonia
| | - Aleksander Krag
- Faculty of Health Sciences, University of Southern Denmark, Odense, Denmark; Department of Gastroenterology and Hepatology, Odense University Hospital, Odense, Denmark
| | - Torben Hansen
- Novo Nordisk Foundation Center for Basic Metabolic Research, University of Copenhagen, Copenhagen, Denmark
| | - Michael Kuhn
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany.
| | - Peer Bork
- Molecular Systems Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany; Max Delbrück Centre for Molecular Medicine, Berlin, Germany; Department of Bioinformatics, Biocenter, University of Würzburg, Würzburg, Germany.
| |
Collapse
|
88
|
Lin J, Wang H, Chen J, Li Y. Inter-algal associations and nutrients linked by Scenedesmus and Desmodesmus structure eukaryotic algal communities. Mol Ecol 2024; 33:e17528. [PMID: 39283304 DOI: 10.1111/mec.17528] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2023] [Revised: 08/13/2024] [Accepted: 09/03/2024] [Indexed: 10/30/2024]
Abstract
Eutrophication reduces the variability of the community composition of plankton. However, the mechanisms underlying the diversity and restructuring of eukaryotic algal communities remain unknown. This study analysed the diversity and compositional patterns of algal communities in shallow eutrophic lakes. It investigated how these communities were modified by key genera through mediating inter-algal associations under the influence of abiotic factors. Inter-algal associations explained more variance in algal communities than environmental variables, and variation in composition and diversity was primarily derived from Scenedesmus, Desmodesmus and Cryptomonas, rather than nutrients. Scenedesmus and Desmodesmus were positively correlated with the genera of Chlorophyta and formed the hub of the algal association network. When the relative abundance of Scenedesmus and Desmodesmus increased from 0.41% to 13.74%, communities enriched in biomarkers of Bacillariophyta, Chrysophyceae and Cryptophyta transitioned to communities enriched in biomarkers of Chlorophyta. Moreover, negative associations between the Chlorophyta hub genera and other non-Chlorophyta genera increased. High concentrations of total phosphorus altered the composition of algal communities by increasing the abundance of Scenedesmus and Desmodesmus, which in turn had cascading effects through inter-algal associations. Additionally, algal communities with higher abundances of Scenedesmus and Desmodesmus were more susceptible to the effects of total phosphorus. Our study suggested that inter-algal associations, centred on Scenedesmus and Desmodesmus, had a greater influence on algal diversity and community structure than other factors. Nutrient levels were not a direct driver of algal diversity and community structure adjustments, but acted indirectly by enhancing the influence of Scenedesmus and Desmodesmus.
Collapse
Affiliation(s)
- Juan Lin
- College of Pharmacy and Life Science, Jiujiang University, Jiujiang, China
| | - Hongxia Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Jianfeng Chen
- Institute for Ecological Research and Pollution Control of Plateau Lakes, School of Ecology and Environmental Science, Yunnan University, Kunming, China
| | - Yun Li
- State Key Laboratory of Lake Science and Environment, Key Laboratory of Lake and Watershed Science for Water Security, Nanjing Institute of Geography and Limnology, Chinese Academy of Sciences, Nanjing, China
| |
Collapse
|
89
|
Lima Oliveira M, Lima NS, Khara Renaud G, Estrada A, Buitrago D, Hamm A, Nadeem S, Naylor KB, Chen Z, Yanez B, Booms E, Searcy J, Biggers A, Tussing-Humphreys LM. Design of a mindfulness intervention to reduce risk factors for colorectal cancer among at-risk Black women in Chicago. J Appl Physiol (1985) 2024; 137:1484-1493. [PMID: 39417824 PMCID: PMC11573258 DOI: 10.1152/japplphysiol.00608.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/17/2024] [Accepted: 10/05/2024] [Indexed: 10/19/2024] Open
Abstract
Chronic stress can directly and indirectly promote carcinogenesis through immune, metabolic, and microbial pathways. Our overarching hypothesis is that reducing chronic stress will have important implications for colorectal cancer (CRC) risk reduction among vulnerable and high-risk populations. A promising approach for reducing chronic stress is mindfulness. Mindfulness is a meditation-based technique that achieves a state of mind that is used to experience higher awareness or consciousness. Existing small studies suggest mindfulness can positively regulate stress response in a way that translates to anticancer effects, including reduced systemic inflammation. We propose to evaluate an 8-wk mindfulness intervention delivered in a hybrid format (synchronous and asynchronous sessions) among 40 Black women at elevated risk of CRC who reside in vulnerable communities and who report moderate to high perceived stress. At baseline and postintervention, participants will provide blood, hair, and stool; undergo body composition analysis; and complete mood and lifestyle-related surveys. The specific aims are to assess the feasibility and acceptability of the intervention and explore changes on stress, weight, fasting glucose, inflammation markers, and the gut microbiota-risk markers and risk pathways associated with CRC. The data generated through this project will inform if mindfulness is a feasible option for CRC risk reduction among high-risk individuals.NEW & NOTEWORTHY We propose to evaluate an 8-wk mindfulness intervention delivered in a hybrid format (synchronous and asynchronous sessions) among 40 Black women at elevated risk of CRC who reside in vulnerable communities and who report moderate to high perceived stress. The specific aims are to assess the feasibility and acceptability of the intervention and explore changes on stress, weight, fasting glucose, inflammation markers, and the gut microbiota-risk markers and risk pathways associated with CRC.
Collapse
Affiliation(s)
- Manoela Lima Oliveira
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, Illinois, United States
- University of Illinois Cancer Center, Chicago, Illinois, United States
| | - Natalia Salvatierra Lima
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, Illinois, United States
| | | | - Andy Estrada
- Department of Medical Social Sciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States
| | - Diana Buitrago
- Department of Medical Social Sciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States
| | - Alyshia Hamm
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, Illinois, United States
| | - Saba Nadeem
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, Illinois, United States
| | - Keith B Naylor
- University of Illinois Cancer Center, Chicago, Illinois, United States
- College of Medicine, University of Illinois Chicago, Illinois, United States
| | - Zhengjia Chen
- University of Illinois Cancer Center, Chicago, Illinois, United States
| | - Betina Yanez
- Department of Medical Social Sciences, Northwestern University Feinberg School of Medicine, Chicago, Illinois, United States
| | - Emily Booms
- Biology Department, Northeastern Illinois University, Chicago, Illinois, United States
| | - Jasmin Searcy
- Department of Psychology, University of Illinois Chicago, Chicago, Illinois, United States
| | - Alana Biggers
- College of Medicine, University of Illinois Chicago, Illinois, United States
| | - Lisa Marie Tussing-Humphreys
- Department of Kinesiology and Nutrition, University of Illinois Chicago, Chicago, Illinois, United States
- University of Illinois Cancer Center, Chicago, Illinois, United States
| |
Collapse
|
90
|
Nogueira Silva Lima MT, Delayre-Orthez C, Howsam M, Jacolot P, Niquet-Léridon C, Okwieka A, Anton PM, Perot M, Barbezier N, Mathieu H, Ghinet A, Fradin C, Boulanger E, Jaisson S, Gillery P, Tessier FJ. Early- and life-long intake of dietary advanced glycation end-products (dAGEs) leads to transient tissue accumulation, increased gut sensitivity to inflammation, and slight changes in gut microbial diversity, without causing overt disease. Food Res Int 2024; 195:114967. [PMID: 39277266 DOI: 10.1016/j.foodres.2024.114967] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 08/13/2024] [Accepted: 08/20/2024] [Indexed: 09/17/2024]
Abstract
Dietary advanced glycation end-products (dAGEs) accumulate in organs and are thought to initiate chronic low-grade inflammation (CLGI), induce glycoxidative stress, drive immunosenescence, and influence gut microbiota. Part of the toxicological interest in glycation products such as dietary carboxymethyl-lysine (dCML) relies on their interaction with receptor for advanced glycation end-products (RAGE). It remains uncertain whether early or lifelong exposure to dAGEs contributes physiological changes and whether such effects are reversible or permanent. Our objective was to examine the physiological changes in Wild-Type (WT) and RAGE KO mice that were fed either a standard diet (STD - 20.8 ± 5.1 µg dCML/g) or a diet enriched with dCML (255.2 ± 44.5 µg dCML/g) from the perinatal period for up to 70 weeks. Additionally, an early age (6 weeks) diet switch (dCML→STD) was explored to determine whether potential harmful effects of dCML could be reversed. Previous dCML accumulation patterns described by our group were confirmed here, with significant RAGE-independent accumulation of dCML in kidneys, ileum and colon over the 70-week dietary intervention (respectively 3-fold, 17-fold and 20-fold increases compared with controls). Diet switching returned tissue dCML concentrations to their baseline levels. The dCML-enriched diet had no significative effect on endogenous glycation, inflammation, oxidative stress or senescence parameters. The relative expression of TNFα, VCAM1, IL6, and P16 genes were all upregulated (∼2-fold) in an age-dependent manner, most notably in the kidneys of WT animals. RAGE knockout seemed protective in this regard, diminishing age-related renal expression of TNFα. Significant increases in TNFα expression were detectable in the intestinal tract of the Switch group (∼2-fold), suggesting a higher sensitivity to inflammation perhaps related to the timing of the diet change. Minor fluctuations were observed at family level within the caecal microbiota, including Eggerthellaceae, Anaerovoracaceae and Marinifilaceae communities, indicating slight changes in composition. Despite chronic dCML consumption resulting in higher free CML levels in tissues, there were no substantial increases in parameters related to inflammageing. Age was a more important factor in inflammation status, notably in the kidneys, while the early-life dietary switch may have influenced intestinal susceptibility to inflammation. This study affirms the therapeutic potential of RAGE modulation and corroborates evidence for the disruptive effect of dietary changes occurring too early in life. Future research should prioritize the potential influence of dAGEs on disease aetiology and development, notably any exacerbating effects they may have upon existing health conditions.
Collapse
Affiliation(s)
- M T Nogueira Silva Lima
- U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Institut Pasteur de Lille, University Lille, Inserm, CHU Lille, F-59000 Lille, France
| | - C Delayre-Orthez
- Institut Polytechnique UniLaSalle, Université d'Artois, ULR 7519, Equipe PETALES, 60000 Beauvais, France
| | - M Howsam
- U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Institut Pasteur de Lille, University Lille, Inserm, CHU Lille, F-59000 Lille, France
| | - P Jacolot
- Institut Polytechnique UniLaSalle, Université d'Artois, ULR 7519, Equipe PETALES, 60000 Beauvais, France
| | - C Niquet-Léridon
- Institut Polytechnique UniLaSalle, Université d'Artois, ULR 7519, Equipe PETALES, 60000 Beauvais, France
| | - A Okwieka
- University of Reims Champagne-Ardenne, Laboratory of Biochemistry and Molecular Biology, CNRS/URCA UMR 7369 MEDyC, Faculté de Médecine, 51095 Reims, France
| | - P M Anton
- Institut Polytechnique UniLaSalle, Université d'Artois, ULR 7519, Equipe PETALES, 60000 Beauvais, France
| | - M Perot
- Institut Polytechnique UniLaSalle, Université d'Artois, ULR 7519, Equipe PETALES, 60000 Beauvais, France
| | - N Barbezier
- Institut Polytechnique UniLaSalle, Université d'Artois, ULR 7519, Equipe PETALES, 60000 Beauvais, France
| | - H Mathieu
- Institut Polytechnique UniLaSalle, Université d'Artois, ULR 7519, Equipe PETALES, 60000 Beauvais, France
| | - A Ghinet
- U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Institut Pasteur de Lille, University Lille, Inserm, CHU Lille, F-59000 Lille, France; Junia, Health and Environment, Laboratory of Sustainable Chemistry and Health, 59000 Lille, France
| | - C Fradin
- U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Institut Pasteur de Lille, University Lille, Inserm, CHU Lille, F-59000 Lille, France
| | - E Boulanger
- U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Institut Pasteur de Lille, University Lille, Inserm, CHU Lille, F-59000 Lille, France
| | - S Jaisson
- University of Reims Champagne-Ardenne, Laboratory of Biochemistry and Molecular Biology, CNRS/URCA UMR 7369 MEDyC, Faculté de Médecine, 51095 Reims, France; University Hospital of Reims, Laboratory of Biochemistry-Pharmacology-Toxicology, 51092 Reims, France
| | - P Gillery
- University of Reims Champagne-Ardenne, Laboratory of Biochemistry and Molecular Biology, CNRS/URCA UMR 7369 MEDyC, Faculté de Médecine, 51095 Reims, France; University Hospital of Reims, Laboratory of Biochemistry-Pharmacology-Toxicology, 51092 Reims, France
| | - F J Tessier
- U1167-RID-AGE-Facteurs de Risque et Déterminants Moléculaires des Maladies Liées au Vieillissement, Institut Pasteur de Lille, University Lille, Inserm, CHU Lille, F-59000 Lille, France.
| |
Collapse
|
91
|
Grond K, Zur Tulod J, Kurtz CC, Duddleston KN. Effects of the anti-inflammatory drug budesonide on the gut microbiota and cytokine production of 13-lined ground squirrels during prehibernation fattening. Physiol Genomics 2024; 56:711-720. [PMID: 39250427 PMCID: PMC11573255 DOI: 10.1152/physiolgenomics.00034.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2024] [Revised: 09/05/2024] [Accepted: 09/07/2024] [Indexed: 09/11/2024] Open
Abstract
The gut microbiome is essential for maintaining organismal health. Gut microbiota may be disrupted through external factors like dietary change, which can lead to gut inflammation, resulting in obesity. Hibernating mammals develop low-grade gut inflammation when they accumulate fat deposits in preparation for hibernation, making them useful models for studying the relationship between the microbiome, inflammation, and weight gain. Nonsteroidal anti-inflammatory drugs and steroids are commonly used in humans to target gut inflammation, but how these drugs affect the gut microbiome and its stability is unclear. We investigated the effect of the glucocorticoid drug budesonide on the gut microbiome and cytokine levels of an obligate hibernator, the 13-lined ground squirrel, during the fattening season. We used 16S rRNA gene sequencing to characterize bacterial communities in the lumen and mucosa of the cecum and colon and measured proinflammatory [tumor necrosis factor-α (TNF-α)/interleukin 6 (IL-6)] and anti-inflammatory (IL-10) cytokine levels. Budesonide affected the microbiome only in the cecum lumen, where bacterial diversity was higher in the control group, and communities significantly differed between treatments. Across gut sections, Marvinbryantia and Enterococcus were significantly higher in the budesonide group, whereas Sarcina was higher in the control group. TNF-α and IL-6 levels were higher in control squirrels compared with the budesonide group, but there was no difference in IL-10 levels. Overall, budesonide treatment affected the microbial community and diversity of 13-lined ground squirrels in the cecum lumen. Our study presents another step toward developing ground squirrels as a model for studying the interaction between the microbiota and host inflammation.NEW & NOTEWORTHY Disruptions of gut microbiota can lead to inflammation, resulting in weight gain. Inflammation can be treated with budesonide, but how budesonide affects gut microbiota is unclear. Thirteen-lined ground squirrels experience low-grade gut inflammation during prehibernation fattening, which compares with human inflammation-weight gain mechanisms. We showed that budesonide treatment decreased microbiome diversity and lead to a shift in community in the cecum lumen. Our study supports developing ground squirrels as a model for studying microbiome-inflammation interactions.
Collapse
Affiliation(s)
- Kirsten Grond
- Department of Biological Sciences, College of Arts and Sciences, University of Alaska Anchorage, Anchorage, Alaska, United States
| | - Jewel Zur Tulod
- Department of Biology, University of Wisconsin Oshkosh, Oshkosh, Wisconsin, United States
| | - Courtney C Kurtz
- Department of Biology, University of Wisconsin Oshkosh, Oshkosh, Wisconsin, United States
| | - Khrystyne N Duddleston
- Department of Biological Sciences, College of Arts and Sciences, University of Alaska Anchorage, Anchorage, Alaska, United States
| |
Collapse
|
92
|
Chowdhury S, Kullberg RFJ, Haak BW, Duran C, Earny VA, Eshwara VK, Lawley TD, Wiersinga WJ, Mukhopadhyay C. Gut Microbiome in Human Melioidosis: Composition and Resistome Dynamics from Diagnosis to Discovery. Open Forum Infect Dis 2024; 11:ofae654. [PMID: 39553288 PMCID: PMC11568386 DOI: 10.1093/ofid/ofae654] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Accepted: 11/01/2024] [Indexed: 11/19/2024] Open
Abstract
Background Melioidosis, attributable to the soil-dwelling bacterium Burkholderia pseudomallei, stands as a paramount global health challenge, necessitating extended courses of antibiotics. While murine studies identified the gut microbiota as a modulator of bacterial dissemination during melioidosis, the human intestinal microbiota during melioidosis remains uncharacterized. Here, we characterized gut microbiota composition and antimicrobial resistance (AMR) genes at diagnosis, during treatment, and postdischarge for melioidosis. We hypothesized that the gut microbiota of melioidosis patients would be extensively distorted. Methods In this prospective observational cohort, stool samples of patients with culture-confirmed melioidosis admitted to a tertiary care hospital in India were collected at diagnosis, 14 days after diagnosis, or discharge (whichever occurred first) and at 6 months postinfection. Family members or neighbors served as community controls. The gut microbiota and resistome were profiled by shotgun metagenomic sequencing. Results We longitudinally analyzed the gut microbiota of 70 fecal samples from 28 patients and 16 community controls. At diagnosis, the gut microbiota of patients differed from that of controls, characterized by high abundances of potentially pathogenic bacteria, a loss of butyrate-producing bacteria, and higher levels of AMR genes. Microbiota composition and resistome remained different from community controls at 6 months, driven by total antibiotic exposure. During hospitalization, gut microbiota profiles were associated with secondary Klebsiella pneumoniae infections. Conclusions This first study on gut microbiota composition and resistome in human melioidosis showed extensive disruptions during hospitalization, with limited signs of restoration 6 months postinfection. Given the adverse outcomes linked with microbiome perturbations, limiting microbiota disruptions or using microbiota-restorative therapies (eg, butyrate-producing probiotics) may be beneficial.
Collapse
Affiliation(s)
- Soumi Chowdhury
- Department of Microbiology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
- Center of Emerging and Tropical Diseases (CETD), Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Robert F J Kullberg
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Bastiaan W Haak
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Claudio Duran
- Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - Venkat A Earny
- Department of Microbiology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
- Center of Emerging and Tropical Diseases (CETD), Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Vandana K Eshwara
- Department of Microbiology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
- Center of Emerging and Tropical Diseases (CETD), Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
- Center for Antimicrobial Resistance and Education (CARE), Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| | - Trevor D Lawley
- Host-Microbiota Interactions Laboratory, Wellcome Sanger Institute, Wellcome Genome Campus, Hinxton, United Kingdom
| | - W Joost Wiersinga
- Center for Experimental and Molecular Medicine, Amsterdam University Medical Center, University of Amsterdam, Amsterdam, The Netherlands
- Division of Infectious Diseases, Department of Internal Medicine, Amsterdam University Medical Center, Amsterdam, The Netherlands
| | - Chiranjay Mukhopadhyay
- Department of Microbiology, Kasturba Medical College, Manipal, Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
- Center of Emerging and Tropical Diseases (CETD), Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
- Center for Antimicrobial Resistance and Education (CARE), Manipal Academy of Higher Education, Manipal 576104, Karnataka, India
| |
Collapse
|
93
|
Pfau M, Degregori S, Barber PH, Blumstein DT, Philson CS. Differences in Gut Microbes Across Age and Sex Linked to Metabolism and Microbial Stability in a Hibernating Mammal. Ecol Evol 2024; 14:e70519. [PMID: 39524311 PMCID: PMC11550910 DOI: 10.1002/ece3.70519] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2024] [Revised: 10/01/2024] [Accepted: 10/15/2024] [Indexed: 11/16/2024] Open
Abstract
The gut microbiome has a well-documented relationship with host fitness, physiology, and behavior. However, most of what is known comes from captive animals where diets and environments are more homogeneous or controlled. Studies in wild populations that experience dynamic environments and have natural life history variation are less common but are key to understanding the drivers of variation in the gut microbiome. Here we examine a wild population of yellow-bellied marmots (Marmota flaviventer), an obligate winter hibernator, to quantify multivariate associations between host-associated factors (e.g., age, sex, environmental harshness, and social behavior) and gut microbial composition. Across 5 years and 143 individuals, we found that males had a higher relative abundance of microbes associated with mass gain and cellulose digestion, which suggests a metabolic investment in mass gain (such as phylum Firmicutes and family Lachnospiraceae). By contrast, females had higher relative abundances of microbes associated with inflammation and metabolism (from microbial groups such as Tenericutes and Ruminococcus), possibly reflecting the importance of lactation and offspring investment. Post hoc analyses of lactating females showed a negative relationship with the abundance of microbes associated with mass gain but a positive relationship with microbes associated with metabolic energy, suggesting a trade-off between investment in pups and maternal mass gain. Older animals also had reduced Proteobacteria relative abundance, a phylum associated with reduced inflammation. Results demonstrate that sex and age-based traits, not sociality or environmental harshness, are associated with microbe-mediated metabolism and inflammation in a wild, hibernating mammal.
Collapse
Affiliation(s)
- Madison Pfau
- Department of Ecology and Evolutionary BiologyUCLALos AngelesCaliforniaUSA
- Department of Environmental Science, Policy, and ManagementUniversity of California, BerkeleyBerkeleyCaliforniaUSA
| | - Samuel Degregori
- Department of AnthropologyNorthwestern UniversityEvanstonIllinoisUSA
| | - Paul H. Barber
- Department of Ecology and Evolutionary BiologyUCLALos AngelesCaliforniaUSA
| | - Daniel T. Blumstein
- Department of Ecology and Evolutionary BiologyUCLALos AngelesCaliforniaUSA
- Rocky Mountain Biological LaboratoryCrested ButteColoradoUSA
| | - Conner S. Philson
- Department of Ecology and Evolutionary BiologyUCLALos AngelesCaliforniaUSA
- Rocky Mountain Biological LaboratoryCrested ButteColoradoUSA
- Centre for Research in Animal BehaviourUniversity of ExeterExeterUK
| |
Collapse
|
94
|
Blufstein A, Pejcic N, Spettel K, Hausmann B, Seki D, Ertekin T, Hinrichs‐Priller J, Altner S, Nehr M, Bekes K, Makristathis A, Andrukhov O. Salivary microbiome and MRP-8/14 levels in children with gingivitis, healthy children, and their mothers. J Periodontol 2024; 95:1035-1047. [PMID: 38696461 PMCID: PMC11609491 DOI: 10.1002/jper.23-0632] [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: 10/30/2023] [Revised: 03/21/2024] [Accepted: 03/29/2024] [Indexed: 05/04/2024]
Abstract
BACKGROUND Gingivitis is the most common form of periodontal disease among children and adolescents and is associated with disrupted host-microbiome homeostasis. Family is an important factor influencing the prevalence of gingivitis. In the present study, we investigated the salivary microbiome, oral hygiene habits, and the salivary level of myeloid-related protein (MRP)-8/14 in children aged 7-12 years with gingivitis, periodontally healthy children, and their mothers. METHODS This study included 24 children with gingivitis (including four sibling pairs) and 22 periodontally healthy children (including two sibling pairs) and their mothers. The whole saliva was collected, DNA was extracted, the variable V3-V4 region of the eubacterial 16S ribosomal RNA gene was amplified, and sample library preparation was performed according to the Illumina protocol. The salivary levels of MRP-8/14 were analyzed by ELISA. RESULTS Alpha diversity of the salivary microbiome was considerably higher in gingivitis children and mothers of gingivitis children compared to healthy children and their mothers, respectively. Significant differences in beta diversity between healthy and gingivitis children, healthy children and their mothers, and gingivitis children and their mothers were detected. Overall, the number of common core amplicon sequence variants between children and their own mothers was significantly higher than between children and other mothers. The salivary MRP-8/14 levels in children with gingivitis were significantly higher compared to healthy children; a similar tendency was also mentioned for mothers. CONCLUSION Our study underlines the importance of family as an essential factor influencing oral health.
Collapse
Affiliation(s)
- Alice Blufstein
- Competence Center for Periodontal ResearchUniversity Clinic of DentistryMedical University of ViennaViennaAustria
- Division of Conservative Dentistry and PeriodontologyUniversity Clinic of DentistryMedical University of ViennaViennaAustria
| | - Natasa Pejcic
- Competence Center for Periodontal ResearchUniversity Clinic of DentistryMedical University of ViennaViennaAustria
- Department of Preventive and Pediatric DentistryFaculty of Dental MedicineUniversity of BelgradeBelgradeSerbia
| | - Kathrin Spettel
- Division of Clinical MicrobiologyDepartment of Laboratory MedicineMedical University of ViennaViennaAustria
| | - Bela Hausmann
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Division of Clinical Microbiology, Department of Laboratory Medicine, Medical University of ViennaViennaAustria
| | - David Seki
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Division of Clinical Microbiology, Department of Laboratory Medicine, Medical University of ViennaViennaAustria
| | - Tugba Ertekin
- Competence Center for Periodontal ResearchUniversity Clinic of DentistryMedical University of ViennaViennaAustria
| | - Julia Hinrichs‐Priller
- Division of Pediatric DentistryUniversity Clinic of DentistryMedical University of ViennaViennaAustria
| | - Sarra Altner
- Division of Pediatric DentistryUniversity Clinic of DentistryMedical University of ViennaViennaAustria
| | - Marion Nehr
- Division of Clinical MicrobiologyDepartment of Laboratory MedicineMedical University of ViennaViennaAustria
| | - Katrin Bekes
- Division of Pediatric DentistryUniversity Clinic of DentistryMedical University of ViennaViennaAustria
| | - Athanasios Makristathis
- Division of Clinical MicrobiologyDepartment of Laboratory MedicineMedical University of ViennaViennaAustria
- Joint Microbiome Facility of the Medical University of Vienna and the University of Vienna, Division of Clinical Microbiology, Department of Laboratory Medicine, Medical University of ViennaViennaAustria
| | - Oleh Andrukhov
- Competence Center for Periodontal ResearchUniversity Clinic of DentistryMedical University of ViennaViennaAustria
| |
Collapse
|
95
|
Wang M, Fontaine S, Jiang H, Li G. ADAPT: Analysis of Microbiome Differential Abundance by Pooling Tobit Models. BIOINFORMATICS (OXFORD, ENGLAND) 2024; 40:btae661. [PMID: 39509330 DOI: 10.1093/bioinformatics/btae661] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 10/01/2024] [Accepted: 11/05/2024] [Indexed: 11/15/2024]
Abstract
MOTIVATION Microbiome differential abundance analysis (DAA) remains a challenging problem despite multiple methods proposed in the literature. The excessive zeros and compositionality of metagenomics data are two main challenges for DAA. RESULTS We propose a novel method called "Analysis of Microbiome Differential Abundance by Pooling Tobit Models" (ADAPT) to overcome these two challenges. ADAPT interprets zero counts as left-censored observations to avoid unfounded assumptions and complex models. ADAPT also encompasses a theoretically justified way of selecting non-differentially abundant microbiome taxa as a reference to reveal differentially abundant taxa while avoiding false discoveries. We generate synthetic data using independent simulation frameworks to show that ADAPT has more consistent false discovery rate control and higher statistical power than competitors. We use ADAPT to analyze 16S rRNA sequencing of saliva samples and shotgun metagenomics sequencing of plaque samples collected from infants in the COHRA2 study. The results provide novel insights into the association between the oral microbiome and early childhood dental caries. AVAILABILITY AND IMPLEMENTATION The R package ADAPT can be installed from Bioconductor at https://bioconductor.org/packages/release/bioc/html/ADAPT.html or from Github at https://github.com/mkbwang/ADAPT. The source codes for simulation studies and real data analysis are available at https://github.com/mkbwang/ADAPT_example.
Collapse
Affiliation(s)
- Mukai Wang
- Department of Biostatistics, University of Michigan, 1415 Washington Heights, Ann Arbor, Michigan, 48109, United States
| | - Simon Fontaine
- Department of Statistics, University of Michigan, 1085 South University, Ann Arbor, Michigan, 48109, United States
| | - Hui Jiang
- Department of Biostatistics, University of Michigan, 1415 Washington Heights, Ann Arbor, Michigan, 48109, United States
| | - Gen Li
- Department of Biostatistics, University of Michigan, 1415 Washington Heights, Ann Arbor, Michigan, 48109, United States
| |
Collapse
|
96
|
Gancz NN, Querdasi FR, Chu KA, Towner E, Taylor E, Callaghan BL. Characteristics of the oral microbiome in youth exposed to caregiving adversity. Brain Behav Immun Health 2024; 41:100850. [PMID: 39280088 PMCID: PMC11401114 DOI: 10.1016/j.bbih.2024.100850] [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: 08/22/2024] [Accepted: 08/24/2024] [Indexed: 09/18/2024] Open
Abstract
Caregiving adversity (CA) exposure is robustly linked to increased risk for poor oral, physical, and mental health outcomes. Increasingly, the gut microbiome has garnered interest as a contributor to risk for and resilience to such health outcomes in CA-exposed individuals. Though often overlooked, the oral microbiome of CA-exposed individuals may be just as important a contributor to health outcomes as the gut microbiome. Indeed, outside the context of CA, the oral microbiome is well-documented as a regulator of both oral and systemic health, and preliminary data suggest its association with mental health. However, research examining the association between CA and the oral microbiome is extremely sparse, especially in childhood, when the community composition of such organisms is still stabilizing. To address that sparsity, in the current study, we examined composition and differential abundance metrics of the oral microbiome in 152 youth aged 6-16 years, who had either been exposed to significant caregiving adversity (significant separation from or maltreatment by a caregiver; N = 66, CA) or who had always remained with their biological/birth families (N = 86, Comparison). We identified a significant negative association between hair cortisol and oral microbiome richness in the Comparison group that was significantly blunted in the CA group. Additionally, youth in the CA group had altered oral microbiome composition and elevated abundance of potentially pathogenic bacteria relative to youth in the Comparison group. Questionnaire measures of fatigue, somatic complaints, and internalizing symptoms had limited associations with oral microbiome features that were altered in CA. Although we found differences in the oral microbiomes of CA-exposed youth, further research is required to elucidate the implications of those differences for health and well-being.
Collapse
Affiliation(s)
- Naomi N Gancz
- University of California, Los Angeles, 502 Portola Plaza, Los Angeles, CA, 90095, United States
| | - Francesca R Querdasi
- University of California, Los Angeles, 502 Portola Plaza, Los Angeles, CA, 90095, United States
| | - Kristen A Chu
- University of California, Los Angeles, 502 Portola Plaza, Los Angeles, CA, 90095, United States
| | - Emily Towner
- University of California, Los Angeles, 502 Portola Plaza, Los Angeles, CA, 90095, United States
| | - Eason Taylor
- University of California, Los Angeles, 502 Portola Plaza, Los Angeles, CA, 90095, United States
| | - Bridget L Callaghan
- University of California, Los Angeles, 502 Portola Plaza, Los Angeles, CA, 90095, United States
| |
Collapse
|
97
|
Wang L, Lu W, Song Y, Liu S, Fu YV. Using machine learning to identify environmental factors that collectively determine microbial community structure of activated sludge. ENVIRONMENTAL RESEARCH 2024; 260:119635. [PMID: 39025351 DOI: 10.1016/j.envres.2024.119635] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/14/2024] [Revised: 07/12/2024] [Accepted: 07/15/2024] [Indexed: 07/20/2024]
Abstract
Activated sludge (AS) microbial communities are influenced by various environmental variables. However, a comprehensive analysis of how these variables jointly and nonlinearly shape the AS microbial community remains challenging. In this study, we employed advanced machine learning techniques to elucidate the collective effects of environmental variables on the structure and function of AS microbial communities. Applying Dirichlet multinomial mixtures analysis to 311 global AS samples, we identified four distinct microbial community types (AS-types), each characterized by unique microbial compositions and metabolic profiles. We used 14 classical linear and nonlinear machine learning methods to select a baseline model. The extremely randomized trees demonstrated optimal performance in learning the relationship between environmental factors and AS types (with an accuracy of 71.43%). Feature selection identified critical environmental factors and their importance rankings, including latitude (Lat), longitude (Long), precipitation during sampling (Precip), solids retention time (SRT), effluent total nitrogen (Effluent TN), average temperature during sampling month (Avg Temp), mixed liquor temperature (Mixed Temp), influent biochemical oxygen demand (Influent BOD), and annual precipitation (Annual Precip). Significantly, Lat, Long, Precip, Avg Temp, and Annual Precip, influenced metabolic variations among AS types. These findings emphasize the pivotal role of environmental variables in shaping microbial community structures and enhancing metabolic pathways within activated sludge. Our study encourages the application of machine learning techniques to design artificial activated sludge microbial communities for specific environmental purposes.
Collapse
Affiliation(s)
- Lu Wang
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Weilai Lu
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yang Song
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Shuangjiang Liu
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Yu Vincent Fu
- State Key Laboratory of Microbial Resources, Institute of Microbiology, Chinese Academy of Sciences, Beijing, 100101, China; Savaid Medical School, University of Chinese Academy of Sciences, Beijing, 100049, China.
| |
Collapse
|
98
|
Jain V, Dalby MJ, Alexander EC, Burford C, Acford-Palmer H, Serghiou IR, Teng NMY, Kiu R, Gerasimidis K, Zafeiropoulou K, Logan M, Verma A, Davenport M, Hall LJ, Dhawan A. Association of gut microbiota and gut metabolites and adverse outcomes in biliary atresia: A longitudinal prospective study. Hepatol Commun 2024; 8:e0550. [PMID: 39761011 DOI: 10.1097/hc9.0000000000000550] [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] [Received: 07/31/2024] [Accepted: 08/01/2024] [Indexed: 01/07/2025] Open
Abstract
BACKGROUND The Kasai portoenterostomy (KPE) aims to re-establish bile flow in biliary atresia (BA); however, BA remains the commonest indication for liver transplantation in pediatrics. Gut microbiota-host interplay is increasingly associated with outcomes in chronic liver disease. This study characterized fecal microbiota and fatty acid metabolites in BA. METHODS Fecal samples were prospectively collected in newly diagnosed BA infants (n = 55) before and after KPE. Age-matched healthy control (n = 19) and cholestatic control (n = 21) fecal samples were collected. Fecal 16S rRNA gene amplicon sequencing for gut microbiota and gas chromatography for fecal fatty acids was performed. RESULTS Increased abundance of Enterococcus in pre-KPE BA and cholestatic control infants, compared to healthy infants, was demonstrated. At the early post-KPE time points, increased alpha diversity was revealed in BA versus healthy cohorts. A lower relative abundance of Bifidobacterium and increased Enterococcus, Clostridium, Fusobacterium, and Pseudomonas was seen in infants with BA. Fecal acetate was reduced, and fecal butyrate and propionate were elevated in early post-KPE BA infants. Higher post-KPE alpha diversity was associated with nonfavorable clinical outcomes (6-month jaundice and liver transplantation). A higher relative abundance of post-KPE Streptococcus and Fusobacterium and a lower relative abundance of Dorea, Blautia, and Oscillospira were associated with nonfavorable clinical outcomes. Blautia inversely correlated to liver disease severity, and Bifidobacterium inversely correlated to fibrosis biomarkers. Bifidobacterium abundance was significantly lower in infants experiencing cholangitis within 6 months after KPE. CONCLUSIONS Increased diversity, enrichment of pathogenic, and depletion of beneficial microbiota early post-KPE are all factors associated with nonfavorable BA outcomes. Manipulation of gut microbiota in the early postsurgical period could provide therapeutic potential.
Collapse
Affiliation(s)
- Vandana Jain
- Paediatric Liver, GI and Nutrition Centre and Mowatlabs, King's College Hospital, London, UK
| | - Matthew J Dalby
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Emma C Alexander
- Paediatric Liver, GI and Nutrition Centre and Mowatlabs, King's College Hospital, London, UK
| | - Charlotte Burford
- Paediatric Liver, GI and Nutrition Centre and Mowatlabs, King's College Hospital, London, UK
| | | | | | - Nancy M Y Teng
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
| | - Raymond Kiu
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
| | - Konstantinos Gerasimidis
- Human Nutrition, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow Royal Infirmary, Glasgow, UK
| | - Konstantina Zafeiropoulou
- Human Nutrition, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow Royal Infirmary, Glasgow, UK
| | - Michael Logan
- Human Nutrition, School of Medicine, Dentistry and Nursing, College of Medical, Veterinary and Life Sciences, University of Glasgow, Glasgow Royal Infirmary, Glasgow, UK
| | - Anita Verma
- Department of Infection Science, Paediatric Liver, GI and Nutrition Centre and Mowatlabs, King's College Hospital, London, UK
| | - Mark Davenport
- Department of Paediatric Surgery, King's College Hospital, London, UK
| | - Lindsay J Hall
- Quadram Institute Bioscience, Norwich Research Park, Norwich, UK
- Institute of Microbiology and Infection, College of Medical and Dental Sciences, University of Birmingham, Birmingham, UK
- Norwich Medical School, University of East Anglia, Norwich Research Park, Norwich, UK
| | - Anil Dhawan
- Paediatric Liver, GI and Nutrition Centre and Mowatlabs, King's College Hospital, London, UK
| |
Collapse
|
99
|
Rousseau M, Siegenthaler A, Skidmore AK, de Groot GA, Laros I. Further reduction in soil bacterial diversity under severe acidification in European temperate forests. EUROPEAN JOURNAL OF SOIL SCIENCE 2024; 75:e70005. [PMID: 39583947 PMCID: PMC11579971 DOI: 10.1111/ejss.70005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 10/13/2023] [Revised: 10/09/2024] [Accepted: 10/10/2024] [Indexed: 11/26/2024]
Abstract
Despite a decrease in industrial nitrogen and sulfur deposition over recent decades, soil acidification remains a persistent challenge to European forest health, especially in regions of intense agriculture and urbanisation. Using topsoil eDNA metabarcoding and functional annotations from a sample of 49 plots (each 30 × 30 m) located in The Netherlands and Germany, we investigated the effect of severe acidification on bacterial taxonomic diversity under different forest types and explored potential functional implications for nutrient cycling. Furthermore, we assessed which soil parameters known to influence soil bacterial communities affect these acidophilic communities. Here, we are the first to demonstrate under natural conditions that soil bacterial diversity in extremely acidic soils (pH <4.5) continues to decline similarly across forest types as pH further decreases under intensifying human activity. Our results confirmed pH as the key driver of soil bacterial communities, even in extremely acidic soils. Ongoing severe acidification continues to reduce bacterial communities, favouring taxa adapted to extreme acidity and primarily involved in recalcitrant carbon-degradation compounds (e.g. cellulolysis potential = 0.78%-9.99%) while simultaneously diminishing taxa associated with nitrogen cycling (e.g. fixation potential = 6.72%-0.00%). Altogether, our findings indicate a further decline in bacterial diversity in already extremely acidic soils, likely disrupting nutrient cycling through changes in immobilisation and mineralisation processes. Our study highlights the continuous acidification of European temperate forests to extremely low pH levels, further disrupting forest ecosystem functioning. The significant reduction in bacterial diversity under such a severe acidification gradient, as demonstrated here, underscores the necessity to include severely acidified forests in conservation programmes and monitoring to prevent further degradation of European soils beyond repair.
Collapse
Affiliation(s)
- Mélody Rousseau
- Natural Resources Department, Faculty of Geo‐Information Science and Earth ObservationUniversity of TwenteEnschedeThe Netherlands
| | - Andjin Siegenthaler
- Natural Resources Department, Faculty of Geo‐Information Science and Earth ObservationUniversity of TwenteEnschedeThe Netherlands
| | - Andrew K. Skidmore
- Natural Resources Department, Faculty of Geo‐Information Science and Earth ObservationUniversity of TwenteEnschedeThe Netherlands
| | - G. Arjen de Groot
- Wageningen Environmental ResearchWageningen University & ResearchWageningenThe Netherlands
| | - Ivo Laros
- Wageningen Environmental ResearchWageningen University & ResearchWageningenThe Netherlands
| |
Collapse
|
100
|
Zhang M, Yin YS, May KS, Wang S, Purcell H, Zhang XS, Blaser MJ, den Hartigh LJ. The role of intestinal microbiota in physiologic and body compositional changes that accompany CLA-mediated weight loss in obese mice. Mol Metab 2024; 89:102029. [PMID: 39293564 PMCID: PMC11447304 DOI: 10.1016/j.molmet.2024.102029] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/05/2024] [Revised: 09/10/2024] [Accepted: 09/11/2024] [Indexed: 09/20/2024] Open
Abstract
OBJECTIVE Obesity continues to be a major problem, despite known treatment strategies such as lifestyle modifications, pharmaceuticals, and surgical options, necessitating the development of novel weight loss approaches. The naturally occurring fatty acid, 10,12 conjugated linoleic acid (10,12 CLA), promotes weight loss by increasing fat oxidation and browning of white adipose tissue, leading to increased energy expenditure in obese mice. Coincident with weight loss, 10,12 CLA also alters the murine gut microbiota by enriching for microbes that produce short chain fatty acids (SCFAs), with concurrent elevations in fecal butyrate and plasma acetate. METHODS To determine if the observed microbiota changes are required for 10,12 CLA-mediated weight loss, adult male mice with diet-induced obesity were given broad-spectrum antibiotics (ABX) to perturb the microbiota prior to and during 10,12 CLA-mediated weight loss. Conversely, to determine whether gut microbes were sufficient to induce weight loss, conventionally-raised and germ-free mice were transplanted with cecal contents from mice that had undergone weight loss by 10,12 CLA supplementation. RESULTS While body weight was minimally modulated by ABX-mediated perturbation of gut bacterial populations, adult male mice given ABX were more resistant to the increased energy expenditure and fat loss that are induced by 10,12 CLA supplementation. Transplanting cecal contents from donor mice losing weight due to oral 10,12 CLA consumption into conventional or germ-free mice led to improved glucose metabolism with increased butyrate production. CONCLUSIONS These data suggest a critical role for the microbiota in diet-modulated changes in energy balance and glucose metabolism, and distinguish the metabolic effects of orally delivered 10,12 CLA from cecal transplantation of the resulting microbiota.
Collapse
Affiliation(s)
- Meifan Zhang
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, USA
| | - Yue S Yin
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, USA
| | - Karolline S May
- Department of Medicine: Metabolism, Endocrinology, and Nutrition, Seattle, WA, USA; Diabetes Institute, University of Washington, Seattle, WA, USA
| | - Shari Wang
- Department of Medicine: Metabolism, Endocrinology, and Nutrition, Seattle, WA, USA; Diabetes Institute, University of Washington, Seattle, WA, USA
| | - Hayley Purcell
- Mitochondria and Metabolism Center, Department of Anesthesiology and Pain Medicine, University of Washington, Seattle, WA, USA
| | - Xue-Song Zhang
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, USA
| | - Martin J Blaser
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, USA
| | - Laura J den Hartigh
- Department of Medicine: Metabolism, Endocrinology, and Nutrition, Seattle, WA, USA; Diabetes Institute, University of Washington, Seattle, WA, USA.
| |
Collapse
|