51
|
Petrick JL, Wilkinson JE, Michaud DS, Cai Q, Gerlovin H, Signorello LB, Wolpin BM, Ruiz-Narváez EA, Long J, Yang Y, Johnson WE, Shu XO, Huttenhower C, Palmer JR. The oral microbiome in relation to pancreatic cancer risk in African Americans. Br J Cancer 2022; 126:287-296. [PMID: 34718358 PMCID: PMC8770575 DOI: 10.1038/s41416-021-01578-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2020] [Revised: 09/14/2021] [Accepted: 10/01/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND African Americans have the highest pancreatic cancer incidence of any racial/ethnic group in the United States. The oral microbiome was associated with pancreatic cancer risk in a recent study, but no such studies have been conducted in African Americans. Poor oral health, which can be a cause or effect of microbial populations, was associated with an increased risk of pancreatic cancer in a single study of African Americans. METHODS We prospectively investigated the oral microbiome in relation to pancreatic cancer risk among 122 African-American pancreatic cancer cases and 354 controls. DNA was extracted from oral wash samples for metagenomic shotgun sequencing. Alpha and beta diversity of the microbial profiles were calculated. Multivariable conditional logistic regression was used to estimate odds ratios (ORs) and 95% confidence intervals (CIs) for associations between microbes and pancreatic cancer risk. RESULTS No associations were observed with alpha or beta diversity, and no individual microbial taxa were differentially abundant between cases and control, after accounting for multiple comparisons. Among never smokers, there were elevated ORs for known oral pathogens: Porphyromonas gingivalis (OR = 1.69, 95% CI: 0.80-3.56), Prevotella intermedia (OR = 1.40, 95% CI: 0.69-2.85), and Tannerella forsythia (OR = 1.36, 95% CI: 0.66-2.77). CONCLUSIONS Previously reported associations between oral taxa and pancreatic cancer were not present in this African-American population overall.
Collapse
Affiliation(s)
| | - Jeremy E Wilkinson
- Department of Biostatistics, Harvard TH Chan School of Public Health, Boston, MA, USA
| | - Dominique S Michaud
- Department of Public Health and Community Medicine, Tufts University School of Medicine, Boston, MA, USA
| | - Qiuyin Cai
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Hanna Gerlovin
- Slone Epidemiology Center, Boston University, Boston, MA, USA
| | - Lisa B Signorello
- Division of Cancer Prevention, National Cancer Institute, Bethesda, MD, USA
| | - Brian M Wolpin
- Department of Medical Oncology, Dana-Farber Cancer Institute and Harvard Medical School, Boston, MA, USA
| | - Edward A Ruiz-Narváez
- Department of Nutritional Sciences, University of Michigan School of Public Health, Ann Arbor, MI, USA
| | - Jirong Long
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Yaohua Yang
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - W Evan Johnson
- Department of Medicine, Division of Computational Biomedicine, Boston University, Boston, MA, USA
| | - Xiao-Ou Shu
- Division of Epidemiology, Department of Medicine, Vanderbilt Epidemiology Center, Vanderbilt-Ingram Cancer Center, Vanderbilt University School of Medicine, Nashville, TN, USA
| | - Curtis Huttenhower
- Department of Biostatistics, Harvard TH Chan School of Public Health, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Julie R Palmer
- Slone Epidemiology Center, Boston University, Boston, MA, USA.
| |
Collapse
|
52
|
Nearing JT, Douglas GM, Hayes MG, MacDonald J, Desai DK, Allward N, Jones CMA, Wright RJ, Dhanani AS, Comeau AM, Langille MGI. Microbiome differential abundance methods produce different results across 38 datasets. Nat Commun 2022; 13:342. [PMID: 35039521 PMCID: PMC8763921 DOI: 10.1038/s41467-022-28034-z] [Citation(s) in RCA: 305] [Impact Index Per Article: 101.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Accepted: 01/04/2022] [Indexed: 12/12/2022] Open
Abstract
Identifying differentially abundant microbes is a common goal of microbiome studies. Multiple methods are used interchangeably for this purpose in the literature. Yet, there are few large-scale studies systematically exploring the appropriateness of using these tools interchangeably, and the scale and significance of the differences between them. Here, we compare the performance of 14 differential abundance testing methods on 38 16S rRNA gene datasets with two sample groups. We test for differences in amplicon sequence variants and operational taxonomic units (ASVs) between these groups. Our findings confirm that these tools identified drastically different numbers and sets of significant ASVs, and that results depend on data pre-processing. For many tools the number of features identified correlate with aspects of the data, such as sample size, sequencing depth, and effect size of community differences. ALDEx2 and ANCOM-II produce the most consistent results across studies and agree best with the intersect of results from different approaches. Nevertheless, we recommend that researchers should use a consensus approach based on multiple differential abundance methods to help ensure robust biological interpretations. Many microbiome differential abundance methods are available, but it lacks systematic comparison among them. Here, the authors compare the performance of 14 differential abundance testing methods on 38 16S rRNA gene datasets with two sample groups, and show ALDEx2 and ANCOM-II produce the most consistent results.
Collapse
Affiliation(s)
- Jacob T Nearing
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada.
| | - Gavin M Douglas
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Molly G Hayes
- Department of Mathematics and Statistics, Dalhousie University, Halifax, NS, Canada
| | - Jocelyn MacDonald
- Department of Computer Science, Dalhousie University, Halifax, NS, Canada
| | - Dhwani K Desai
- Integrated Microbiome Resource, Dalhousie University, Halifax, NS, Canada
| | - Nicole Allward
- Department of Civil and Resource Engineering, Dalhousie University, Halifax, NS, Canada
| | - Casey M A Jones
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Robyn J Wright
- Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| | - Akhilesh S Dhanani
- Integrated Microbiome Resource, Dalhousie University, Halifax, NS, Canada
| | - André M Comeau
- Integrated Microbiome Resource, Dalhousie University, Halifax, NS, Canada
| | - Morgan G I Langille
- Integrated Microbiome Resource, Dalhousie University, Halifax, NS, Canada.,Department of Pharmacology, Dalhousie University, Halifax, NS, Canada
| |
Collapse
|
53
|
Gönenc II, Wolff A, Schmidt J, Zibat A, Müller C, Cyganek L, Argyriou L, Räschle M, Yigit G, Wollnik B. OUP accepted manuscript. Hum Mol Genet 2022; 31:2185-2193. [PMID: 35099000 PMCID: PMC9262399 DOI: 10.1093/hmg/ddab373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2021] [Revised: 12/02/2021] [Accepted: 12/27/2021] [Indexed: 11/12/2022] Open
Abstract
Bloom syndrome (BS) is an autosomal recessive disease clinically characterized by primary microcephaly, growth deficiency, immunodeficiency and predisposition to cancer. It is mainly caused by biallelic loss-of-function mutations in the BLM gene, which encodes the BLM helicase, acting in DNA replication and repair processes. Here, we describe the gene expression profiles of three BS fibroblast cell lines harboring causative, biallelic truncating mutations obtained by single-cell (sc) transcriptome analysis. We compared the scRNA transcription profiles from three BS patient cell lines to two age-matched wild-type controls and observed specific deregulation of gene sets related to the molecular processes characteristically affected in BS, such as mitosis, chromosome segregation, cell cycle regulation and genomic instability. We also found specific upregulation of genes of the Fanconi anemia pathway, in particular FANCM, FANCD2 and FANCI, which encode known interaction partners of BLM. The significant deregulation of genes associated with inherited forms of primary microcephaly observed in our study might explain in part the molecular pathogenesis of microcephaly in BS, one of the main clinical characteristics in patients. Finally, our data provide first evidence of a novel link between BLM dysfunction and transcriptional changes in condensin complex I and II genes. Overall, our study provides novel insights into gene expression profiles in BS on an sc level, linking specific genes and pathways to BLM dysfunction.
Collapse
Affiliation(s)
| | | | - Julia Schmidt
- Institute of Human Genetics, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Arne Zibat
- Institute of Human Genetics, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Christian Müller
- Institute of Human Genetics, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Lukas Cyganek
- Stem Cell Unit, Clinic for Cardiology and Pneumology, University Medical Center Göttingen, 37075 Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Loukas Argyriou
- Institute of Human Genetics, University Medical Center Göttingen, 37073 Göttingen, Germany
| | - Markus Räschle
- Department of Molecular Genetics, Technical University of Kaiserslautern, 67663 Kaiserslautern, Germany
| | - Gökhan Yigit
- Institute of Human Genetics, University Medical Center Göttingen, 37073 Göttingen, Germany
- German Center for Cardiovascular Research (DZHK), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Bernd Wollnik
- To whom correspondence should be addressed at: Institute of Human Genetics, University Medical Center Göttingen, Heinrich-Düker-Weg 12, 37073 Göttingen, Germany. Tel: +49 5513960606; Fax: +49 5513969303;
| |
Collapse
|
54
|
Mordant A, Kleiner M. Evaluation of Sample Preservation and Storage Methods for Metaproteomics Analysis of Intestinal Microbiomes. Microbiol Spectr 2021; 9:e0187721. [PMID: 34908431 PMCID: PMC8672883 DOI: 10.1128/spectrum.01877-21] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2021] [Accepted: 10/31/2021] [Indexed: 12/20/2022] Open
Abstract
A critical step in studies of the intestinal microbiome using meta-omics approaches is the preservation of samples before analysis. Preservation is essential for approaches that measure gene expression, such as metaproteomics, which is used to identify and quantify proteins in microbiomes. Intestinal microbiome samples are typically stored by flash-freezing and storage at -80°C, but some experimental setups do not allow for immediate freezing of samples. In this study, we evaluated methods to preserve fecal microbiome samples for metaproteomics analyses when flash-freezing is not possible. We collected fecal samples from C57BL/6 mice and stored them for 1 and 4 weeks using the following methods: flash-freezing in liquid nitrogen, immersion in RNAlater, immersion in 95% ethanol, immersion in a RNAlater-like buffer, and combinations of these methods. After storage, we extracted protein and prepared peptides for liquid chromatography with tandem mass spectrometry (LC-MS/MS) analysis to identify and quantify peptides and proteins. All samples produced highly similar metaproteomes, except for ethanol-preserved samples that were distinct from all other samples in terms of protein identifications and protein abundance profiles. Flash-freezing and RNAlater (or RNAlater-like treatments) produced metaproteomes that differed only slightly, with less than 0.7% of identified proteins differing in abundance. In contrast, ethanol preservation resulted in an average of 9.5% of the identified proteins differing in abundance between ethanol and the other treatments. Our results suggest that preservation at room temperature in RNAlater or an RNAlater-like solution performs as well as freezing for the preservation of intestinal microbiome samples before metaproteomics analyses. IMPORTANCE Metaproteomics is a powerful tool to study the intestinal microbiome. By identifying and quantifying a large number of microbial, dietary, and host proteins in microbiome samples, metaproteomics provides direct evidence of the activities and functions of microbial community members. A critical step for metaproteomics workflows is preserving samples before analysis because protein profiles are susceptible to fast changes in response to changes in environmental conditions (air exposure, temperature changes, etc.). This study evaluated the effects of different preservation treatments on the metaproteomes of intestinal microbiome samples. In contrast to prior work on preservation of fecal samples for metaproteomics analyses, we ensured that all steps of sample preservation were identical so that all differences could be attributed to the preservation method.
Collapse
Affiliation(s)
- Angie Mordant
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, North Carolina, USA
| | - Manuel Kleiner
- Department of Plant and Microbial Biology, North Carolina State University, Raleigh, North Carolina, USA
| |
Collapse
|
55
|
Wertman JN, Dunn KA, Kulkarni K. The impact of the host intestinal microbiome on carcinogenesis and the response to chemotherapy. Future Oncol 2021; 17:4371-4387. [PMID: 34448411 DOI: 10.2217/fon-2021-0087] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
The microbiome consists of all microbes present on and within the human body. An unbalanced, or 'dysbiotic' intestinal microbiome is associated with inflammatory bowel disease, diabetes and some cancer types. Drug treatment can alter the intestinal microbiome composition. Additionally, some chemotherapeutics interact with microbiome components, leading to changes in drug safety and/or efficacy. The intestinal microbiome is a modifiable target, using strategies such as antibiotic treatment, fecal microbial transplantation or probiotic administration. Understanding the impact of the microbiome on the safety and efficacy of cancer treatment may result in improved treatment outcome. The present review seeks to summarize relevant research and look to the future of cancer treatment, where the intestinal microbiome is recognized as an actionable treatment target.
Collapse
Affiliation(s)
- Jaime N Wertman
- Department of Pediatrics/Division of Pediatric Hematology-Oncology, Dalhousie University/IWK Health Centre, Halifax, Canada
- College of Pharmacy, Dalhousie University, Halifax, Canada
| | - Katherine A Dunn
- Department of Pediatrics/Division of Pediatric Hematology-Oncology, Dalhousie University/IWK Health Centre, Halifax, Canada
- Department of Biology, Dalhousie University, Halifax, Canada
| | - Ketan Kulkarni
- Department of Pediatrics/Division of Pediatric Hematology-Oncology, Dalhousie University/IWK Health Centre, Halifax, Canada
| |
Collapse
|
56
|
Mallick H, Rahnavard A, McIver LJ, Ma S, Zhang Y, Nguyen LH, Tickle TL, Weingart G, Ren B, Schwager EH, Chatterjee S, Thompson KN, Wilkinson JE, Subramanian A, Lu Y, Waldron L, Paulson JN, Franzosa EA, Bravo HC, Huttenhower C. Multivariable association discovery in population-scale meta-omics studies. PLoS Comput Biol 2021; 17:e1009442. [PMID: 34784344 PMCID: PMC8714082 DOI: 10.1371/journal.pcbi.1009442] [Citation(s) in RCA: 854] [Impact Index Per Article: 213.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 12/28/2021] [Accepted: 09/09/2021] [Indexed: 12/13/2022] Open
Abstract
It is challenging to associate features such as human health outcomes, diet, environmental conditions, or other metadata to microbial community measurements, due in part to their quantitative properties. Microbiome multi-omics are typically noisy, sparse (zero-inflated), high-dimensional, extremely non-normal, and often in the form of count or compositional measurements. Here we introduce an optimized combination of novel and established methodology to assess multivariable association of microbial community features with complex metadata in population-scale observational studies. Our approach, MaAsLin 2 (Microbiome Multivariable Associations with Linear Models), uses generalized linear and mixed models to accommodate a wide variety of modern epidemiological studies, including cross-sectional and longitudinal designs, as well as a variety of data types (e.g., counts and relative abundances) with or without covariates and repeated measurements. To construct this method, we conducted a large-scale evaluation of a broad range of scenarios under which straightforward identification of meta-omics associations can be challenging. These simulation studies reveal that MaAsLin 2's linear model preserves statistical power in the presence of repeated measures and multiple covariates, while accounting for the nuances of meta-omics features and controlling false discovery. We also applied MaAsLin 2 to a microbial multi-omics dataset from the Integrative Human Microbiome (HMP2) project which, in addition to reproducing established results, revealed a unique, integrated landscape of inflammatory bowel diseases (IBD) across multiple time points and omics profiles.
Collapse
Affiliation(s)
- Himel Mallick
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- The Broad Institute, Cambridge, Massachusetts, United States of America
| | - Ali Rahnavard
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, George Washington University, Washington DC, United States of America
| | - Lauren J. McIver
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- The Broad Institute, Cambridge, Massachusetts, United States of America
| | - Siyuan Ma
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- The Broad Institute, Cambridge, Massachusetts, United States of America
| | - Yancong Zhang
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- The Broad Institute, Cambridge, Massachusetts, United States of America
| | - Long H. Nguyen
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Timothy L. Tickle
- The Broad Institute, Cambridge, Massachusetts, United States of America
| | - George Weingart
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- The Broad Institute, Cambridge, Massachusetts, United States of America
| | - Boyu Ren
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- The Broad Institute, Cambridge, Massachusetts, United States of America
| | - Emma H. Schwager
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- The Broad Institute, Cambridge, Massachusetts, United States of America
| | - Suvo Chatterjee
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Kelsey N. Thompson
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Jeremy E. Wilkinson
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Ayshwarya Subramanian
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- The Broad Institute, Cambridge, Massachusetts, United States of America
| | - Yiren Lu
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Levi Waldron
- Department of Epidemiology and Biostatistics, CUNY School of Public Health, New York City, New York, United States of America
| | - Joseph N. Paulson
- Department of Biostatistics, Product Development, Genentech, Inc., South San Francisco, California, United States of America
| | - Eric A. Franzosa
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- The Broad Institute, Cambridge, Massachusetts, United States of America
| | - Hector Corrada Bravo
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, United States of America
| | - Curtis Huttenhower
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- The Broad Institute, Cambridge, Massachusetts, United States of America
| |
Collapse
|
57
|
Mallick H, Rahnavard A, McIver LJ, Ma S, Zhang Y, Nguyen LH, Tickle TL, Weingart G, Ren B, Schwager EH, Chatterjee S, Thompson KN, Wilkinson JE, Subramanian A, Lu Y, Waldron L, Paulson JN, Franzosa EA, Bravo HC, Huttenhower C. Multivariable association discovery in population-scale meta-omics studies. PLoS Comput Biol 2021. [PMID: 34784344 DOI: 10.1101/2021.01.20.427420v1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2023] Open
Abstract
It is challenging to associate features such as human health outcomes, diet, environmental conditions, or other metadata to microbial community measurements, due in part to their quantitative properties. Microbiome multi-omics are typically noisy, sparse (zero-inflated), high-dimensional, extremely non-normal, and often in the form of count or compositional measurements. Here we introduce an optimized combination of novel and established methodology to assess multivariable association of microbial community features with complex metadata in population-scale observational studies. Our approach, MaAsLin 2 (Microbiome Multivariable Associations with Linear Models), uses generalized linear and mixed models to accommodate a wide variety of modern epidemiological studies, including cross-sectional and longitudinal designs, as well as a variety of data types (e.g., counts and relative abundances) with or without covariates and repeated measurements. To construct this method, we conducted a large-scale evaluation of a broad range of scenarios under which straightforward identification of meta-omics associations can be challenging. These simulation studies reveal that MaAsLin 2's linear model preserves statistical power in the presence of repeated measures and multiple covariates, while accounting for the nuances of meta-omics features and controlling false discovery. We also applied MaAsLin 2 to a microbial multi-omics dataset from the Integrative Human Microbiome (HMP2) project which, in addition to reproducing established results, revealed a unique, integrated landscape of inflammatory bowel diseases (IBD) across multiple time points and omics profiles.
Collapse
Affiliation(s)
- Himel Mallick
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- The Broad Institute, Cambridge, Massachusetts, United States of America
| | - Ali Rahnavard
- Computational Biology Institute, Department of Biostatistics and Bioinformatics, Milken Institute School of Public Health, George Washington University, Washington DC, United States of America
| | - Lauren J McIver
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- The Broad Institute, Cambridge, Massachusetts, United States of America
| | - Siyuan Ma
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- The Broad Institute, Cambridge, Massachusetts, United States of America
| | - Yancong Zhang
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- The Broad Institute, Cambridge, Massachusetts, United States of America
| | - Long H Nguyen
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
- Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts, United States of America
| | - Timothy L Tickle
- The Broad Institute, Cambridge, Massachusetts, United States of America
| | - George Weingart
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- The Broad Institute, Cambridge, Massachusetts, United States of America
| | - Boyu Ren
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- The Broad Institute, Cambridge, Massachusetts, United States of America
| | - Emma H Schwager
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- The Broad Institute, Cambridge, Massachusetts, United States of America
| | - Suvo Chatterjee
- Epidemiology Branch, Division of Intramural Population Health Research, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Kelsey N Thompson
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Jeremy E Wilkinson
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Ayshwarya Subramanian
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- The Broad Institute, Cambridge, Massachusetts, United States of America
| | - Yiren Lu
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
| | - Levi Waldron
- Department of Epidemiology and Biostatistics, CUNY School of Public Health, New York City, New York, United States of America
| | - Joseph N Paulson
- Department of Biostatistics, Product Development, Genentech, Inc., South San Francisco, California, United States of America
| | - Eric A Franzosa
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- The Broad Institute, Cambridge, Massachusetts, United States of America
| | - Hector Corrada Bravo
- Center for Bioinformatics and Computational Biology, University of Maryland, College Park, Maryland, United States of America
| | - Curtis Huttenhower
- Biostatistics Department, Harvard T. H. Chan School of Public Health, Boston, Massachusetts, United States of America
- The Broad Institute, Cambridge, Massachusetts, United States of America
| |
Collapse
|
58
|
Diener C, Qin S, Zhou Y, Patwardhan S, Tang L, Lovejoy JC, Magis AT, Price ND, Hood L, Gibbons SM. Baseline Gut Metagenomic Functional Gene Signature Associated with Variable Weight Loss Responses following a Healthy Lifestyle Intervention in Humans. mSystems 2021; 6:e0096421. [PMID: 34519531 PMCID: PMC8547453 DOI: 10.1128/msystems.00964-21] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2021] [Accepted: 08/23/2021] [Indexed: 12/29/2022] Open
Abstract
Recent human feeding studies have shown how the baseline taxonomic composition of the gut microbiome can determine responses to weight loss interventions. However, the functional determinants underlying this phenomenon remain unclear. We report a weight loss response analysis on a cohort of 105 individuals selected from a larger population enrolled in a commercial wellness program, which included healthy lifestyle coaching. Each individual in the cohort had baseline blood metabolomics, blood proteomics, clinical labs, dietary questionnaires, stool 16S rRNA gene sequencing data, and follow-up data on weight change. We generated additional targeted proteomics data on obesity-associated proteins in blood before and after intervention, along with baseline stool metagenomic data, for a subset of 25 individuals who showed the most extreme weight change phenotypes. We built regression models to identify baseline blood, stool, and dietary features associated with weight loss, independent of age, sex, and baseline body mass index (BMI). Many features were independently associated with baseline BMI, but few were independently associated with weight loss. Baseline diet was not associated with weight loss, and only one blood analyte was associated with changes in weight. However, 31 baseline stool metagenomic functional features, including complex polysaccharide and protein degradation genes, stress-response genes, respiration-related genes, and cell wall synthesis genes, along with gut bacterial replication rates, were associated with weight loss responses after controlling for age, sex, and baseline BMI. Together, these results provide a set of compelling hypotheses for how commensal gut microbiota influence weight loss outcomes in humans. IMPORTANCE Recent human feeding studies have shown how the baseline taxonomic composition of the gut microbiome can determine responses to dietary interventions, but the exact functional determinants underlying this phenomenon remain unclear. In this study, we set out to better understand interactions between baseline BMI, metabolic health, diet, gut microbiome functional profiles, and subsequent weight changes in a human cohort that underwent a healthy lifestyle intervention. Overall, our results suggest that the microbiota may influence host weight loss responses through variable bacterial growth rates, dietary energy harvest efficiency, and immunomodulation.
Collapse
Affiliation(s)
| | - Shizhen Qin
- Institute for Systems Biology, Seattle, Washington, USA
| | - Yong Zhou
- Institute for Systems Biology, Seattle, Washington, USA
| | | | - Li Tang
- Institute for Systems Biology, Seattle, Washington, USA
| | - Jennifer C. Lovejoy
- Institute for Systems Biology, Seattle, Washington, USA
- Lifestyle Medicine Institute, Redlands, California, USA
| | | | - Nathan D. Price
- Institute for Systems Biology, Seattle, Washington, USA
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
- Onegevity (a division of Thorne HealthTech), New York, New York, USA
| | - Leroy Hood
- Institute for Systems Biology, Seattle, Washington, USA
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
| | - Sean M. Gibbons
- Institute for Systems Biology, Seattle, Washington, USA
- Department of Bioengineering, University of Washington, Seattle, Washington, USA
- eScience Institute, University of Washington, Seattle, Washington, USA
| |
Collapse
|
59
|
Zhang Y, Thompson KN, Branck T, Yan Yan, Nguyen LH, Franzosa EA, Huttenhower C. Metatranscriptomics for the Human Microbiome and Microbial Community Functional Profiling. Annu Rev Biomed Data Sci 2021; 4:279-311. [PMID: 34465175 DOI: 10.1146/annurev-biodatasci-031121-103035] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Shotgun metatranscriptomics (MTX) is an increasingly practical way to survey microbial community gene function and regulation at scale. This review begins by summarizing the motivations for community transcriptomics and the history of the field. We then explore the principles, best practices, and challenges of contemporary MTX workflows: beginning with laboratory methods for isolation and sequencing of community RNA, followed by informatics methods for quantifying RNA features, and finally statistical methods for detecting differential expression in a community context. In thesecond half of the review, we survey important biological findings from the MTX literature, drawing examples from the human microbiome, other (nonhuman) host-associated microbiomes, and the environment. Across these examples, MTX methods prove invaluable for probing microbe-microbe and host-microbe interactions, the dynamics of energy harvest and chemical cycling, and responses to environmental stresses. We conclude with a review of open challenges in the MTX field, including making assays and analyses more robust, accessible, and adaptable to new technologies; deciphering roles for millions of uncharacterized microbial transcripts; and solving applied problems such as biomarker discovery and development of microbial therapeutics.
Collapse
Affiliation(s)
- Yancong Zhang
- Harvard Chan Microbiome in Public Health Center and Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, USA; , .,Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| | - Kelsey N Thompson
- Harvard Chan Microbiome in Public Health Center and Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, USA; , .,Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| | - Tobyn Branck
- Harvard Chan Microbiome in Public Health Center and Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, USA; , .,Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA.,Department of Systems, Synthetic, and Quantitative Biology, Harvard Medical School, Boston, Massachusetts 02115, USA
| | - Yan Yan
- Harvard Chan Microbiome in Public Health Center and Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, USA; , .,Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| | - Long H Nguyen
- Harvard Chan Microbiome in Public Health Center and Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, USA; , .,Division of Gastroenterology, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02114, USA.,Clinical and Translational Epidemiology Unit, Massachusetts General Hospital and Harvard Medical School, Boston, Massachusetts 02108, USA
| | - Eric A Franzosa
- Harvard Chan Microbiome in Public Health Center and Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, USA; , .,Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA
| | - Curtis Huttenhower
- Harvard Chan Microbiome in Public Health Center and Department of Biostatistics, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, USA; , .,Infectious Disease and Microbiome Program, Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, USA.,Department of Immunology and Infectious Diseases, Harvard T. H. Chan School of Public Health, Boston, Massachusetts 02115, USA
| |
Collapse
|
60
|
Jiang R, Li WV, Li JJ. mbImpute: an accurate and robust imputation method for microbiome data. Genome Biol 2021; 22:192. [PMID: 34183041 PMCID: PMC8240317 DOI: 10.1186/s13059-021-02400-4] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Accepted: 06/04/2021] [Indexed: 12/22/2022] Open
Abstract
A critical challenge in microbiome data analysis is the existence of many non-biological zeros, which distort taxon abundance distributions, complicate data analysis, and jeopardize the reliability of scientific discoveries. To address this issue, we propose the first imputation method for microbiome data-mbImpute-to identify and recover likely non-biological zeros by borrowing information jointly from similar samples, similar taxa, and optional metadata including sample covariates and taxon phylogeny. We demonstrate that mbImpute improves the power of identifying disease-related taxa from microbiome data of type 2 diabetes and colorectal cancer, and mbImpute preserves non-zero distributions of taxa abundances.
Collapse
Affiliation(s)
- Ruochen Jiang
- Department of Statistics, University of California, Los Angeles, 90095-1554, CA, USA
| | - Wei Vivian Li
- Department of Statistics, University of California, Los Angeles, 90095-1554, CA, USA
- Department of Biostatistics and Epidemiology, Rutgers School of Public Health, Piscataway, 08854, NJ, USA
| | - Jingyi Jessica Li
- Department of Statistics, University of California, Los Angeles, 90095-1554, CA, USA.
- Department of Human Genetics, University of California, Los Angeles, 90095-7088, CA, USA.
- Department of Computational Medicine, University of California, Los Angeles, 90095-1766, CA, USA.
- Department of Biostatistics, University of California, Los Angeles, 90095-1772, CA, USA.
| |
Collapse
|
61
|
Statistical Modeling of High Dimensional Counts. METHODS IN MOLECULAR BIOLOGY (CLIFTON, N.J.) 2021; 2284:97-134. [PMID: 33835440 DOI: 10.1007/978-1-0716-1307-8_7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Statistical modeling of count data from RNA sequencing (RNA-seq) experiments is important for proper interpretation of results. Here I will describe how count data can be modeled using count distributions, or alternatively analyzed using nonparametric methods. I will focus on basic routines for performing data input, scaling/normalization, visualization, and statistical testing to determine sets of features where the counts reflect differences in gene expression across samples. Finally, I discuss limitations and possible extensions to the models presented here.
Collapse
|
62
|
Nichols RG, Davenport ER. The relationship between the gut microbiome and host gene expression: a review. Hum Genet 2021; 140:747-760. [PMID: 33221945 PMCID: PMC7680557 DOI: 10.1007/s00439-020-02237-0] [Citation(s) in RCA: 78] [Impact Index Per Article: 19.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/06/2020] [Indexed: 12/13/2022]
Abstract
Despite the growing knowledge surrounding host-microbiome interactions, we are just beginning to understand how the gut microbiome influences-and is influenced by-host gene expression. Here, we review recent literature that intersects these two fields, summarizing themes across studies. Work in model organisms, human biopsies, and cell culture demonstrate that the gut microbiome is an important regulator of several host pathways relevant for disease, including immune development and energy metabolism, and vice versa. The gut microbiome remodels host chromatin, causes differential splicing, alters the epigenetic landscape, and directly interrupts host signaling cascades. Emerging techniques like single-cell RNA sequencing and organoid generation have the potential to refine our understanding of the relationship between the gut microbiome and host gene expression in the future. By intersecting microbiome and host gene expression, we gain a window into the physiological processes important for fostering the extensive cross-kingdom interactions and ultimately our health.
Collapse
Affiliation(s)
- Robert G. Nichols
- Department of Biology, The Pennsylvania State University, University Park, PA 16802 USA
| | - Emily R. Davenport
- Department of Biology, The Pennsylvania State University, University Park, PA 16802 USA
- Huck Institutes of the Life Sciences, The Pennsylvania State University, University Park, PA 16802 USA
| |
Collapse
|
63
|
Zhang J, Cook J, Nearing JT, Zhang J, Raudonis R, Glick BR, Langille MGI, Cheng Z. Harnessing the plant microbiome to promote the growth of agricultural crops. Microbiol Res 2021; 245:126690. [PMID: 33460987 DOI: 10.1016/j.micres.2020.126690] [Citation(s) in RCA: 58] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Revised: 12/11/2020] [Accepted: 12/30/2020] [Indexed: 12/11/2022]
Abstract
The rhizosphere microbiome is composed of diverse microbial organisms, including archaea, viruses, fungi, bacteria as well as eukaryotic microorganisms, which occupy a narrow region of soil directly associated with plant roots. The interactions between these microorganisms and the plant can be commensal, beneficial or pathogenic. These microorganisms can also interact with each other, either competitively or synergistically. Promoting plant growth by harnessing the soil microbiome holds tremendous potential for providing an environmentally friendly solution to the increasing food demands of the world's rapidly growing population, while also helping to alleviate the associated environmental and societal issues of large-scale food production. There recently have been many studies on the disease suppression and plant growth promoting abilities of the rhizosphere microbiome; however, these findings largely have not been translated into the field. Therefore, additional research into the dynamic interactions between crop plants, the rhizosphere microbiome and the environment are necessary to better guide the harnessing of the microbiome to increase crop yield and quality. This review explores the biotic and abiotic interactions that occur within the plant's rhizosphere as well as current agricultural practices, and how these biotic and abiotic factors, as well as human practices, impact the plant microbiome. Additionally, some limitations, safety considerations, and future directions to the study of the plant microbiome are discussed.
Collapse
Affiliation(s)
- Janie Zhang
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Jamie Cook
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Jacob T Nearing
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Junzeng Zhang
- Aquatic and Crop Resource Development Research Centre, National Research Council of Canada, Halifax, NS, Canada
| | - Renee Raudonis
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada
| | - Bernard R Glick
- Department of Biology, University of Waterloo, Waterloo, ON, Canada
| | - Morgan G I Langille
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada; Department of Pharmacology, Dalhousie University, Halifax, NS, Canada; CGEB-Integrated Microbiome Resource (IMR), Dalhousie University, Halifax, NS, Canada
| | - Zhenyu Cheng
- Department of Microbiology and Immunology, Dalhousie University, Halifax, NS, Canada.
| |
Collapse
|
64
|
Guo J, Shao J, Yang Y, Niu X, Liao J, Zhao Q, Wang D, Li S, Hu J. Gut Microbiota in Patients with Polycystic Ovary Syndrome: a Systematic Review. Reprod Sci 2021; 29:69-83. [PMID: 33409871 DOI: 10.1007/s43032-020-00430-0] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Accepted: 12/10/2020] [Indexed: 02/06/2023]
Abstract
Polycystic ovary Syndrome (PCOS) is one of the most popular diseases that cause menstrual dysfunction and infertility in women. Recently, the relationships between the gastrointestinal microbiome and metabolic disorders such as obesity, type 2 diabetes and PCOS have been discovered. However, the association between the gut microbiome and PCOS symptoms has not been well established. We systematically reviewed existing studies comparing gut microbial composition in PCOS and healthy volunteers to explore evidence for this association. A systematic search was carried out in PubMed, Embase, Cochrane Library, and Web of Science from inception to May 26, 2020, for all original cross-sectional, cohort, or case-control studies comparing the fecal microbiomes of patients with PCOS with microbiomes of healthy volunteers (controls). The primary outcomes were differences in specific gut microbes between patients with PCOS and controls. The search identified 256 citations; 10 studies were included. The total population study of these articles consists of 611 participants (including PCOS group and healthy controls group). Among the included 10 studies, nine studies compared α-diversity, and six studies demonstrated that α-diversity has a significant reduction in PCOS patients. Seven of them reported that there was a significant difference of β-diversity composition between healthy controls groups and PCOS patients. The most common bacterial alterations in PCOS patients included Bacteroidaceae, Coprococcus, Bacteroides, Prevotella, Lactobacillus, Parabacteroides, Escherichia/Shigella, and Faecalibacterium prausnitzii. No consensus has emerged from existing human studies of PCOS and gut microbiome concerning which bacterial taxa are most relevant to it. In this systematic review, we identified specific bacteria associated with microbiomes of patients with PCOS vs controls. Higher level of evidence is needed to determine whether these microbes are a product or cause of PCOS.
Collapse
Affiliation(s)
- Jingbo Guo
- School of Nursing, Lanzhou University, Lanzhou, China
| | - Jie Shao
- School of Nursing, Lanzhou University, Lanzhou, China
| | - Yuan Yang
- The Reproductive Medicine Special Hospital of the 1st Hospital of Lanzhou University, Key Laboratory for Reproductive Medicine and Embryo, Lanzhou, China
| | - Xiaodan Niu
- School of Nursing, Lanzhou University, Lanzhou, China
| | - Juan Liao
- School of Nursing, Lanzhou University, Lanzhou, China
| | - Qing Zhao
- School of Nursing, Lanzhou University, Lanzhou, China
| | - Donghui Wang
- School of Nursing, Lanzhou University, Lanzhou, China
| | - Shuaitong Li
- School of Nursing, Lanzhou University, Lanzhou, China
| | - Junping Hu
- School of Nursing, Lanzhou University, Lanzhou, China. .,The Reproductive Medicine Special Hospital of the 1st Hospital of Lanzhou University, Key Laboratory for Reproductive Medicine and Embryo, Lanzhou, China.
| |
Collapse
|
65
|
Sfriso R, Claypool J. Microbial Reference Frames Reveal Distinct Shifts in the Skin Microbiota after Cleansing. Microorganisms 2020; 8:microorganisms8111634. [PMID: 33113896 PMCID: PMC7690701 DOI: 10.3390/microorganisms8111634] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2020] [Revised: 10/05/2020] [Accepted: 10/20/2020] [Indexed: 01/16/2023] Open
Abstract
Skin cleansing represents a process of mechanical and chemical removal of dirt, pollutants as well as microbiota from the skin. While skin cleansing can help maintain good health, protect us from infections, illnesses and ailments, skin cleansing can also strip away lipids and moisture from the skin, leading to irritation, barrier impairment and disturbance of the delicate cutaneous microbiome. This study investigated how skin cleansing impacts skin’s microbial composition. Thirty Caucasian women were enrolled in a placebo controlled clinical study where participants applied on their volar forearms a liquid body wash twice daily for 1 week in order to mimic frequent showering. Skin microbiome samples were collected by swabbing at defined timepoints and 16S rRNA sequencing was performed. Using “reference frames”, we could identify shifts in the microbial composition and several microbiota were identified as being characteristically associated with the presence of saccharide isomerate, a well-known skin moisturizer. The microbial shift was quite immediate, and we could observe it already at 1 h post cleansing. Interestingly, the new microbial composition reached a certain dynamic equilibrium at day 1 which was then maintained until the end of the study. Paracoccus marcusii, a potentially beneficial carotenoid-producer microorganism, was enriched by the active treatment and, at the same time, the abundance of several potential pathogenic taxa, Brevibacterium casei and Rothia mucilaginosa, diminished.
Collapse
Affiliation(s)
- Riccardo Sfriso
- DSM Nutritional Products, Personal Care, Wurmisweg 576, CH-4303 Kaiseraugst, Switzerland
- Correspondence:
| | - Joshua Claypool
- DSM Nutritional Products, Nutrition Innovation Center, Lexington, MA 02421, USA;
| |
Collapse
|
66
|
Diener C, Reyes-Escogido MDL, Jimenez-Ceja LM, Matus M, Gomez-Navarro CM, Chu ND, Zhong V, Tejero ME, Alm E, Resendis-Antonio O, Guardado-Mendoza R. Progressive Shifts in the Gut Microbiome Reflect Prediabetes and Diabetes Development in a Treatment-Naive Mexican Cohort. Front Endocrinol (Lausanne) 2020; 11:602326. [PMID: 33488518 PMCID: PMC7821428 DOI: 10.3389/fendo.2020.602326] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2020] [Accepted: 11/20/2020] [Indexed: 12/26/2022] Open
Abstract
Type 2 diabetes (T2D) is a global epidemic that affects more than 8% of the world's population and is a leading cause of death in Mexico. Diet and lifestyle are known to contribute to the onset of T2D. However, the role of the gut microbiome in T2D progression remains uncertain. Associations between microbiome composition and diabetes are confounded by medication use, diet, and obesity. Here we present data on a treatment-naive cohort of 405 Mexican individuals across varying stages of T2D severity. Associations between gut bacteria and more than 200 clinical variables revealed a defined set of bacterial genera that were consistent biomarkers of T2D prevalence and risk. Specifically, gradual increases in blood glucose levels, beta cell dysfunction, and the accumulation of measured T2D risk factors were correlated with the relative abundances of four bacterial genera. In a cohort of 25 individuals, T2D treatment-predominantly metformin-reliably returned the microbiome to the normoglycemic community state. Deep clinical characterization allowed us to broadly control for confounding variables, indicating that these microbiome patterns were independent of common T2D comorbidities, like obesity or cardiovascular disease. Our work provides the first solid evidence for a direct link between the gut microbiome and T2D in a critically high-risk population. In particular, we show that increased T2D risk is reflected in gradual changes in the gut microbiome. Whether or not these T2D-associated changes in the gut contribute to the etiology of T2D or its comorbidities remains to be seen.
Collapse
Affiliation(s)
- Christian Diener
- Computational Genomics, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Gibbons Lab, Institute for Systems Biology, Seattle, WA, United States
| | | | - Lilia M. Jimenez-Ceja
- Metabolic Research Laboratory, Department of Medicine and Nutrition, University of Guanajuato, León, Mexico
| | - Mariana Matus
- Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Claudia M. Gomez-Navarro
- Metabolic Research Laboratory, Department of Medicine and Nutrition, University of Guanajuato, León, Mexico
| | - Nathaniel D. Chu
- Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Vivian Zhong
- Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - M. Elizabeth Tejero
- Computational Genomics, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
| | - Eric Alm
- Center for Microbiome Informatics and Therapeutics, Massachusetts Institute of Technology, Cambridge, MA, United States
| | - Osbaldo Resendis-Antonio
- Computational Genomics, Instituto Nacional de Medicina Genómica (INMEGEN), Mexico City, Mexico
- Human Systems Biology Laboratory, Coordinación de la Investigación Científica—Red de Apoyo a la Investigación, Universidad Nacional Autónoma de México (UNAM), Mexico City, Mexico
- *Correspondence: Osbaldo Resendis-Antonio, ; Rodolfo Guardado-Mendoza,
| | - Rodolfo Guardado-Mendoza
- Metabolic Research Laboratory, Department of Medicine and Nutrition, University of Guanajuato, León, Mexico
- Research Department, Hospital Regional de Alta Especialidad del Bajío, León, Mexico
- *Correspondence: Osbaldo Resendis-Antonio, ; Rodolfo Guardado-Mendoza,
| |
Collapse
|