Widespread Transfer of Mobile Antibiotic Resistance Genes Within

Total Page:16

File Type:pdf, Size:1020Kb

Widespread Transfer of Mobile Antibiotic Resistance Genes Within bioRxiv preprint doi: https://doi.org/10.1101/2020.03.19.998526; this version posted March 20, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 1 Widespread transfer of mobile antibiotic resistance genes within individual gut microbiomes 2 revealed through bacterial Hi-C 3 4 Alyssa Kent1*, Albert Vill1*, Qiaojuan Shi1, Michael J. Satlin2, Ilana Lauren Brito1 5 6 1 Meinig School of Biomedical Engineering, Cornell University, Ithaca, NY 7 2 Department of Internal Medicine, Weill Cornell Medicine, New York, NY 8 9 * Denotes equal contribution 10 11 Abstract 12 13 The gut microbiome harbors a ‘silent reservoir’ of antibiotic resistance (AR) genes that is thought to 14 contribute to the emergence of multidrug-resistant pathogens through the process of horizontal gene 15 transfer (HGT). To counteract the spread of AR genes, it is paramount to know which organisms harbor 16 mobile AR genes and with which organisms they engage in HGT. Despite methods to characterize the 17 bulk presence1, abundance2 and function3 of AR genes in the gut, technological limitations of short-read 18 sequencing have precluded linking bacterial taxa to specific mobile genetic elements (MGEs) and their 19 concomitant AR genes. Here, we apply and evaluate a high-throughput, culture-independent method for 20 surveilling the bacterial carriage of MGEs, based on bacterial Hi-C protocols. We compare two healthy 21 individuals with a cohort of seven neutropenic patients undergoing hematopoietic stem cell 22 transplantation, who receive multiple courses of antibiotics throughout their prolonged hospitalizations, 23 and are thus acutely vulnerable to the threat of multidrug-resistant infections4. We find that the networks 24 of HGT are surprisingly distinct between individuals, yet AR and mobile genes are more dispersed across 25 taxa within the neutropenic patients than the healthy subjects. Our data further suggest that HGT is 26 occurring throughout the course of treatment in the microbiomes of neutropenic patients and within the 27 guts of healthy individuals over a similar timeframe. Whereas most efforts to understand the spread of AR 28 genes have focused on pathogenic species, our findings shed light on the role of the human gut 29 microbiome in this process. bioRxiv preprint doi: https://doi.org/10.1101/2020.03.19.998526; this version posted March 20, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 30 The acquisition of antibiotic resistance (AR) genes has rendered important pathogens, such as 31 multidrug-resistant (MDR) Enterobacteriaceae and Pseudomonas aeruginosa, nearly or fully 32 unresponsive to antibiotics. It is widely accepted that these so-called ‘superbugs’ acquire AR genes 33 through the process of horizontal gene transfer (HGT) with members of the human microbiome with 34 whom they come into contact5. The emergence of these MDR bacteria threatens our ability to perform 35 life-saving interventions, such as curative hematopoietic cell transplants for patients with hematologic 36 malignancies6. Furthermore, antibiotic use, required for vital prophylaxis in these patients, has been 37 proposed as a trigger for HGT. Although tools are available to identify AR genes within the gut 38 microbiome, and characterize their function7, abundance8,9 and their host-associations10, no studies have 39 attempted to monitor the bacterial host associations of AR genes and mobile elements during relatively 40 short periods, such as during these patients’ hospitalizations. 41 42 To determine the bacterial hosts of mobile AR genes, we utilized a high-throughput chromatin 43 conformation capture (Hi-C) method aimed at sampling long-range interactions within single bacterial 44 genomes11,12,13. Briefly, while cells are still intact, DNA within individual cells is crosslinked by 45 formaldehyde. Cells are then lysed and the DNA is cut with restriction enzymes, biotinylated, and 46 subjected to dilute ligation to promote intra-molecular linkages between crosslinked DNA. Crosslinking 47 is reversed and then ligated DNA molecules are pulled-down and made into DNA libraries for 48 sequencing. As is, this protocol has been used to improve metagenomic assemblies of bacterial genomes14 49 and has identified a handful of strong plasmid- and phage-bacterial host associations15,16,17,18, suggesting 50 that this technique could be applied to link mobile genes with specific taxa more broadly and to observe 51 the process of HGT over time. 52 53 Here, we develop a modified version of current Hi-C protocols and analytical pipelines (Extended Data 54 Figure 1) in conjunction with metagenomic shotgun sequencing to surveil the bacterial taxa harboring 55 specific mobile AR genes in the gut microbiomes of two healthy individuals and seven patients 56 undergoing hematopoietic stem cell transplantation. These patients have prolonged hospitalizations 57 during their transplant (21 ± 4 days) and often receive multiple courses of antibiotic therapy, increasing 58 the likelihood of an MDR infection. As a result of their condition and treatment, these patients face 59 mortality rates of 40-70% when bacteremic with carbapenem-resistant Enterobacteriaceae (CRE) or 60 carbapenem-resistant Pseudomonas aeruginosa19, and therefore represent a salient population for 61 surveillance and one in which MDR pathogens may emerge and/or amplify under antibiotic selection. Gut 62 microbiome samples for patients and healthy subjects were collected over a 2-3-week period, which, for 63 the neutropenic patients started upon admission for transplant and continued during their hospitalization 64 until neutrophil engraftment (Figure 1A, Extended Data Table 1,2). 65 66 We introduced a number of improvements to current bacterial Hi-C protocols. We changed sample 67 storage and optimized restriction enzymes to improve the congruence between the composition of 68 metagenomic and Hi-C sequencing libraries (Extended Data Figure 2). We also integrated Nextera XT 69 sequencing library preparation directly into the Hi-C experimental protocol, streamlining operations and 70 decreasing sample preparation time. Importantly, within diverse bacterial communities such as the gut 71 microbiome, MGEs may be highly promiscuous and recombinogenic, complicating both assembly20 and 72 linkage analyses21. Therefore, we implemented a computational workflow to assemble genomes, 73 separating large integrated MGEs onto their own contigs, and allowing them to associate with genomes 74 via binning or Hi-C connections. In a mock community of three organisms, each harboring an identifiable 75 plasmid, we are able to confidently link each plasmid to its nascent genome (Extended Data Figure 3). 76 77 Our Hi-C experimental and computational approach results in robust linkages between contigs in human 78 microbiome samples. Hi-C read pairs linking non-mobile contigs with contradictory taxonomic 79 annotations are rarely observed (3.4% at the genus-level) and likely represent homologous sequence 80 matches, highlighting the purity of our Hi-C libraries (Figure 1B). Hi-C read pairs linking two contigs are bioRxiv preprint doi: https://doi.org/10.1101/2020.03.19.998526; this version posted March 20, 2020. The copyright holder for this preprint (which was not certified by peer review) is the author/funder, who has granted bioRxiv a license to display the preprint in perpetuity. It is made available under aCC-BY-NC-ND 4.0 International license. 81 preferentially recruited to contigs that are longer and more abundant, but to a lesser degree than expected, 82 reducing potential bias in our dataset toward highly abundant organisms (Extended Data Figure 4). We 83 binned contigs using several tools (Maxbin22, MetaBat and Concoct), and applied a binning aggregation 84 strategy, DAS Tool23, to obtain a set of draft genomic assemblies. As misassembly can resemble HGT, we 85 removed assemblies with greater than 10% contamination, as determined by CheckM, resulting in 86 taxonomically coherent assemblies (Extended Data Figure 5), albeit a greater number of unbinned contigs 87 (24.6% of the total) (Extended Data Table 3). We then apply conservative criteria to link mobile and 88 mobile AR-containing contigs with the genomic draft assemblies, considering an MGE part of a genome 89 assembly only if it is directly linked to it by at least two uniquely-mapped Hi-C read-pairs. As MGEs are 90 known to recombine, this mitigates the potential for falsely linking contigs that merely share common 91 mobile genes. However, this also potentially reduces our ability for overall detection, especially for larger 92 MGEs, since mobile contigs are often fragmented in metagenomic assemblies24. Nevertheless, we 93 restricted our analysis to those AR-organism and MGE-organism linkages derived from high-confidence 94 read mappings. 95 96 Hi-C significantly improved our ability to detect mobile gene-bacterial host linkages beyond standard 97 metagenomic assembly alone. Hi-C confirms many of the AR gene-taxa and mobile gene-taxa 98 associations observed in the metagenomic assemblies (30.49% ± 11.49% of the AR genes; 30.1% ± 99 9.52% of the mobile genes), but importantly adds on average 31.81% ± 16.28% AR gene associations and 100 36.64% ± 11.56% mobile gene associations to those observed by metagenome assembly alone (Figure 101 1C). Furthermore, whereas metagenomic assembly methods can generally link a single mobile gene 102 cluster to one or two organisms, our Hi-C method was able to identify up to 15 bacterial hosts harboring 103 the same AR or mobile gene, requiring two or more Hi-C linkages within a single individual (mean = 104 3.53 ± 5.69 bacterial hosts per AR gene, Figure 1D; mean = 6.85 ± 10.88 bacterial hosts per gene, 105 Extended Data Figure 6).
Recommended publications
  • WO 2018/064165 A2 (.Pdf)
    (12) INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PCT) (19) World Intellectual Property Organization International Bureau (10) International Publication Number (43) International Publication Date WO 2018/064165 A2 05 April 2018 (05.04.2018) W !P O PCT (51) International Patent Classification: Published: A61K 35/74 (20 15.0 1) C12N 1/21 (2006 .01) — without international search report and to be republished (21) International Application Number: upon receipt of that report (Rule 48.2(g)) PCT/US2017/053717 — with sequence listing part of description (Rule 5.2(a)) (22) International Filing Date: 27 September 2017 (27.09.2017) (25) Filing Language: English (26) Publication Langi English (30) Priority Data: 62/400,372 27 September 2016 (27.09.2016) US 62/508,885 19 May 2017 (19.05.2017) US 62/557,566 12 September 2017 (12.09.2017) US (71) Applicant: BOARD OF REGENTS, THE UNIVERSI¬ TY OF TEXAS SYSTEM [US/US]; 210 West 7th St., Austin, TX 78701 (US). (72) Inventors: WARGO, Jennifer; 1814 Bissonnet St., Hous ton, TX 77005 (US). GOPALAKRISHNAN, Vanch- eswaran; 7900 Cambridge, Apt. 10-lb, Houston, TX 77054 (US). (74) Agent: BYRD, Marshall, P.; Parker Highlander PLLC, 1120 S. Capital Of Texas Highway, Bldg. One, Suite 200, Austin, TX 78746 (US). (81) Designated States (unless otherwise indicated, for every kind of national protection available): AE, AG, AL, AM, AO, AT, AU, AZ, BA, BB, BG, BH, BN, BR, BW, BY, BZ, CA, CH, CL, CN, CO, CR, CU, CZ, DE, DJ, DK, DM, DO, DZ, EC, EE, EG, ES, FI, GB, GD, GE, GH, GM, GT, HN, HR, HU, ID, IL, IN, IR, IS, JO, JP, KE, KG, KH, KN, KP, KR, KW, KZ, LA, LC, LK, LR, LS, LU, LY, MA, MD, ME, MG, MK, MN, MW, MX, MY, MZ, NA, NG, NI, NO, NZ, OM, PA, PE, PG, PH, PL, PT, QA, RO, RS, RU, RW, SA, SC, SD, SE, SG, SK, SL, SM, ST, SV, SY, TH, TJ, TM, TN, TR, TT, TZ, UA, UG, US, UZ, VC, VN, ZA, ZM, ZW.
    [Show full text]
  • Table S4. Phylogenetic Distribution of Bacterial and Archaea Genomes in Groups A, B, C, D, and X
    Table S4. Phylogenetic distribution of bacterial and archaea genomes in groups A, B, C, D, and X. Group A a: Total number of genomes in the taxon b: Number of group A genomes in the taxon c: Percentage of group A genomes in the taxon a b c cellular organisms 5007 2974 59.4 |__ Bacteria 4769 2935 61.5 | |__ Proteobacteria 1854 1570 84.7 | | |__ Gammaproteobacteria 711 631 88.7 | | | |__ Enterobacterales 112 97 86.6 | | | | |__ Enterobacteriaceae 41 32 78.0 | | | | | |__ unclassified Enterobacteriaceae 13 7 53.8 | | | | |__ Erwiniaceae 30 28 93.3 | | | | | |__ Erwinia 10 10 100.0 | | | | | |__ Buchnera 8 8 100.0 | | | | | | |__ Buchnera aphidicola 8 8 100.0 | | | | | |__ Pantoea 8 8 100.0 | | | | |__ Yersiniaceae 14 14 100.0 | | | | | |__ Serratia 8 8 100.0 | | | | |__ Morganellaceae 13 10 76.9 | | | | |__ Pectobacteriaceae 8 8 100.0 | | | |__ Alteromonadales 94 94 100.0 | | | | |__ Alteromonadaceae 34 34 100.0 | | | | | |__ Marinobacter 12 12 100.0 | | | | |__ Shewanellaceae 17 17 100.0 | | | | | |__ Shewanella 17 17 100.0 | | | | |__ Pseudoalteromonadaceae 16 16 100.0 | | | | | |__ Pseudoalteromonas 15 15 100.0 | | | | |__ Idiomarinaceae 9 9 100.0 | | | | | |__ Idiomarina 9 9 100.0 | | | | |__ Colwelliaceae 6 6 100.0 | | | |__ Pseudomonadales 81 81 100.0 | | | | |__ Moraxellaceae 41 41 100.0 | | | | | |__ Acinetobacter 25 25 100.0 | | | | | |__ Psychrobacter 8 8 100.0 | | | | | |__ Moraxella 6 6 100.0 | | | | |__ Pseudomonadaceae 40 40 100.0 | | | | | |__ Pseudomonas 38 38 100.0 | | | |__ Oceanospirillales 73 72 98.6 | | | | |__ Oceanospirillaceae
    [Show full text]
  • Extensive Microbial Diversity Within the Chicken Gut Microbiome Revealed by Metagenomics and Culture
    Extensive microbial diversity within the chicken gut microbiome revealed by metagenomics and culture Rachel Gilroy1, Anuradha Ravi1, Maria Getino2, Isabella Pursley2, Daniel L. Horton2, Nabil-Fareed Alikhan1, Dave Baker1, Karim Gharbi3, Neil Hall3,4, Mick Watson5, Evelien M. Adriaenssens1, Ebenezer Foster-Nyarko1, Sheikh Jarju6, Arss Secka7, Martin Antonio6, Aharon Oren8, Roy R. Chaudhuri9, Roberto La Ragione2, Falk Hildebrand1,3 and Mark J. Pallen1,2,4 1 Quadram Institute Bioscience, Norwich, UK 2 School of Veterinary Medicine, University of Surrey, Guildford, UK 3 Earlham Institute, Norwich Research Park, Norwich, UK 4 University of East Anglia, Norwich, UK 5 Roslin Institute, University of Edinburgh, Edinburgh, UK 6 Medical Research Council Unit The Gambia at the London School of Hygiene and Tropical Medicine, Atlantic Boulevard, Banjul, The Gambia 7 West Africa Livestock Innovation Centre, Banjul, The Gambia 8 Department of Plant and Environmental Sciences, The Alexander Silberman Institute of Life Sciences, Edmond J. Safra Campus, Hebrew University of Jerusalem, Jerusalem, Israel 9 Department of Molecular Biology and Biotechnology, University of Sheffield, Sheffield, UK ABSTRACT Background: The chicken is the most abundant food animal in the world. However, despite its importance, the chicken gut microbiome remains largely undefined. Here, we exploit culture-independent and culture-dependent approaches to reveal extensive taxonomic diversity within this complex microbial community. Results: We performed metagenomic sequencing of fifty chicken faecal samples from Submitted 4 December 2020 two breeds and analysed these, alongside all (n = 582) relevant publicly available Accepted 22 January 2021 chicken metagenomes, to cluster over 20 million non-redundant genes and to Published 6 April 2021 construct over 5,500 metagenome-assembled bacterial genomes.
    [Show full text]
  • Characterization of an Adapted Microbial Population to the Bioconversion of Carbon Monoxide Into Butanol Using Next-Generation Sequencing Technology
    Characterization of an adapted microbial population to the bioconversion of carbon monoxide into butanol using next-generation sequencing technology Guillaume Bruant Research officer, Bioengineering group Energy, Mining, Environment - National Research Council Canada Pacific Rim Summit on Industrial Biotechnology and Bioenergy December 8 -11, 2013 Butanol from residue (dry): syngas route biomass → gasification → syngas → catalysis → synfuels (CO, H2, CO2, CH4) (alcohols…) Biocatalysis vs Chemical catalysis potential for higher product specificity may be less problematic when impurities present less energy intensive (low pressure and temperature) Anaerobic undefined mixed culture vs bacterial pure culture mesophilic anaerobic sludge treating agricultural wastes (Lassonde Inc, Rougemont, QC, Canada) PRS 2013 - 2 Experimental design CO Alcohols Serum bottles incubated at Next Generation RDP Pyrosequencing mesophilic temperature Sequencing (NGS) pipeline 35°C for 2 months Ion PGMTM sequencer http://pyro.cme.msu.edu/ sequences filtered CO continuously supplied Monitoring of bacterial and to the gas phase archaeal populations RDP classifier atmosphere of 100% CO, http://rdp.cme.msu.edu/ 1 atm 16S rRNA genes Ion 314TM chip classifier VFAs & alcohol production bootstrap confidence cutoff low level of butanol of 50 % Samples taken after 1 and 2 months total genomic DNA extracted, purified, concentrated PRS 2013 - 3 NGS: bacterial results Bacterial population - Phylum level 100% 80% Other Chloroflexi 60% Synergistetes %
    [Show full text]
  • Table S5. the Information of the Bacteria Annotated in the Soil Community at Species Level
    Table S5. The information of the bacteria annotated in the soil community at species level No. Phylum Class Order Family Genus Species The number of contigs Abundance(%) 1 Firmicutes Bacilli Bacillales Bacillaceae Bacillus Bacillus cereus 1749 5.145782459 2 Bacteroidetes Cytophagia Cytophagales Hymenobacteraceae Hymenobacter Hymenobacter sedentarius 1538 4.52499338 3 Gemmatimonadetes Gemmatimonadetes Gemmatimonadales Gemmatimonadaceae Gemmatirosa Gemmatirosa kalamazoonesis 1020 3.000970902 4 Proteobacteria Alphaproteobacteria Sphingomonadales Sphingomonadaceae Sphingomonas Sphingomonas indica 797 2.344876284 5 Firmicutes Bacilli Lactobacillales Streptococcaceae Lactococcus Lactococcus piscium 542 1.594633558 6 Actinobacteria Thermoleophilia Solirubrobacterales Conexibacteraceae Conexibacter Conexibacter woesei 471 1.385742446 7 Proteobacteria Alphaproteobacteria Sphingomonadales Sphingomonadaceae Sphingomonas Sphingomonas taxi 430 1.265115184 8 Proteobacteria Alphaproteobacteria Sphingomonadales Sphingomonadaceae Sphingomonas Sphingomonas wittichii 388 1.141545794 9 Proteobacteria Alphaproteobacteria Sphingomonadales Sphingomonadaceae Sphingomonas Sphingomonas sp. FARSPH 298 0.876754244 10 Proteobacteria Alphaproteobacteria Sphingomonadales Sphingomonadaceae Sphingomonas Sorangium cellulosum 260 0.764953367 11 Proteobacteria Deltaproteobacteria Myxococcales Polyangiaceae Sorangium Sphingomonas sp. Cra20 260 0.764953367 12 Proteobacteria Alphaproteobacteria Sphingomonadales Sphingomonadaceae Sphingomonas Sphingomonas panacis 252 0.741416341
    [Show full text]
  • Phylogenomic Analysis of 589 Metagenome-Assembled Genomes Encompassing All Major Prokaryotic Lineages from the Gut of Higher Termites
    Phylogenomic analysis of 589 metagenome-assembled genomes encompassing all major prokaryotic lineages from the gut of higher termites Vincent Hervé1, Pengfei Liu1, Carsten Dietrich1, David Sillam-Dussès2, Petr Stiblik3, Jan Šobotník3 and Andreas Brune1 1 Research Group Insect Gut Microbiology and Symbiosis, Max Planck Institute for Terrestrial Microbiology, Marburg, Germany 2 Laboratory of Experimental and Comparative Ethology EA 4443, Université Paris 13, Villetaneuse, France 3 Faculty of Forestry and Wood Sciences, Czech University of Life Sciences, Prague, Czech Republic ABSTRACT “Higher” termites have been able to colonize all tropical and subtropical regions because of their ability to digest lignocellulose with the aid of their prokaryotic gut microbiota. Over the last decade, numerous studies based on 16S rRNA gene amplicon libraries have largely described both the taxonomy and structure of the prokaryotic communities associated with termite guts. Host diet and microenvironmental conditions have emerged as the main factors structuring the microbial assemblages in the different gut compartments. Additionally, these molecular inventories have revealed the existence of termite-specific clusters that indicate coevolutionary processes in numerous prokaryotic lineages. However, for lack of representative isolates, the functional role of most lineages remains unclear. We reconstructed 589 metagenome-assembled genomes (MAGs) from the different Submitted 29 August 2019 gut compartments of eight higher termite species that encompass 17 prokaryotic
    [Show full text]
  • Modulation of the Gut Microbiota Alters the Tumour-Suppressive Efficacy of Tim-3 Pathway Blockade in a Bacterial Species- and Host Factor-Dependent Manner
    microorganisms Article Modulation of the Gut Microbiota Alters the Tumour-Suppressive Efficacy of Tim-3 Pathway Blockade in a Bacterial Species- and Host Factor-Dependent Manner Bokyoung Lee 1,2, Jieun Lee 1,2, Min-Yeong Woo 1,2, Mi Jin Lee 1, Ho-Joon Shin 1,2, Kyongmin Kim 1,2 and Sun Park 1,2,* 1 Department of Microbiology, Ajou University School of Medicine, Youngtongku Wonchondong San 5, Suwon 442-749, Korea; [email protected] (B.L.); [email protected] (J.L.); [email protected] (M.-Y.W.); [email protected] (M.J.L.); [email protected] (H.-J.S.); [email protected] (K.K.) 2 Department of Biomedical Sciences, The Graduate School, Ajou University, Youngtongku Wonchondong San 5, Suwon 442-749, Korea * Correspondence: [email protected]; Tel.: +82-31-219-5070 Received: 22 August 2020; Accepted: 9 September 2020; Published: 11 September 2020 Abstract: T cell immunoglobulin and mucin domain-containing protein-3 (Tim-3) is an immune checkpoint molecule and a target for anti-cancer therapy. In this study, we examined whether gut microbiota manipulation altered the anti-tumour efficacy of Tim-3 blockade. The gut microbiota of mice was manipulated through the administration of antibiotics and oral gavage of bacteria. Alterations in the gut microbiome were analysed by 16S rRNA gene sequencing. Gut dysbiosis triggered by antibiotics attenuated the anti-tumour efficacy of Tim-3 blockade in both C57BL/6 and BALB/c mice. Anti-tumour efficacy was restored following oral gavage of faecal bacteria even as antibiotic administration continued. In the case of oral gavage of Enterococcus hirae or Lactobacillus johnsonii, transferred bacterial species and host mouse strain were critical determinants of the anti-tumour efficacy of Tim-3 blockade.
    [Show full text]
  • Metabolic Network Percolation Quantifies Biosynthetic Capabilities
    RESEARCH ARTICLE Metabolic network percolation quantifies biosynthetic capabilities across the human oral microbiome David B Bernstein1,2, Floyd E Dewhirst3,4, Daniel Segre` 1,2,5,6,7* 1Department of Biomedical Engineering, Boston University, Boston, United States; 2Biological Design Center, Boston University, Boston, United States; 3The Forsyth Institute, Cambridge, United States; 4Harvard School of Dental Medicine, Boston, United States; 5Bioinformatics Program, Boston University, Boston, United States; 6Department of Biology, Boston University, Boston, United States; 7Department of Physics, Boston University, Boston, United States Abstract The biosynthetic capabilities of microbes underlie their growth and interactions, playing a prominent role in microbial community structure. For large, diverse microbial communities, prediction of these capabilities is limited by uncertainty about metabolic functions and environmental conditions. To address this challenge, we propose a probabilistic method, inspired by percolation theory, to computationally quantify how robustly a genome-derived metabolic network produces a given set of metabolites under an ensemble of variable environments. We used this method to compile an atlas of predicted biosynthetic capabilities for 97 metabolites across 456 human oral microbes. This atlas captures taxonomically-related trends in biomass composition, and makes it possible to estimate inter-microbial metabolic distances that correlate with microbial co-occurrences. We also found a distinct cluster of fastidious/uncultivated taxa, including several Saccharibacteria (TM7) species, characterized by their abundant metabolic deficiencies. By embracing uncertainty, our approach can be broadly applied to understanding metabolic interactions in complex microbial ecosystems. *For correspondence: DOI: https://doi.org/10.7554/eLife.39733.001 [email protected] Competing interests: The authors declare that no Introduction competing interests exist.
    [Show full text]
  • Neocallimastix Californiae G1 36,250,970 NA 29,649 95.52 85.2 SRX2598479 (3)
    Supplementary material for: Horizontal gene transfer as an indispensable driver for Neocallimastigomycota evolution into a distinct gut-dwelling fungal lineage 1 1 1 2 Chelsea L. Murphy ¶, Noha H. Youssef ¶, Radwa A. Hanafy , MB Couger , Jason E. Stajich3, Y. Wang3, Kristina Baker1, Sumit S. Dagar4, Gareth W. Griffith5, Ibrahim F. Farag1, TM Callaghan6, and Mostafa S. Elshahed1* Table S1. Validation of HGT-identification pipeline using previously published datasets. The frequency of HGT occurrence in the genomes of a filamentous ascomycete and a microsporidian were determined using our pipeline. The results were compared to previously published results. Organism NCBI Assembly Reference Method used Value Value accession number to original in the original reported obtained study study in this study Colletotrichum GCA_000149035.1 (1) Blast and tree 11 11 graminicola building approaches Encephalitozoon GCA_000277815.3 (2) Blast against 12-22 4 hellem custom database, AI score calculation, and tree building Table S2. Results of transcriptomic sequencing. Accession number Genus Species Strain Number of Assembled Predicted peptides % genome Ref. reads transcriptsa (Longest Orfs)b completenessc coveraged (%) Anaeromyces contortus C3G 33,374,692 50,577 22,187 96.55 GGWR00000000 This study Anaeromyces contortus C3J 54,320,879 57,658 26,052 97.24 GGWO00000000 This study Anaeromyces contortus G3G 43,154,980 52,929 21,681 91.38 GGWP00000000 This study Anaeromyces contortus Na 42,857,287 47,378 19,386 93.45 GGWN00000000 This study Anaeromyces contortus O2 60,442,723 62,300 27,322 96.9 GGWQ00000000 This study Anaeromyces robustus S4 21,955,935 NA 17,127 92.41 88.7 SRX3329608 (3) Caecomyces sp.
    [Show full text]
  • Microbial Communities Associated with the Camel Tick, Hyalomma Dromedarii
    www.nature.com/scientificreports OPEN Microbial communities associated with the camel tick, Hyalomma dromedarii: 16S rRNA gene‑based analysis Nighat Perveen, Sabir Bin Muzafar, Ranjit Vijayan & Mohammad Ali Al‑Deeb* Hyalomma dromedarii is an important blood‑feeding ectoparasite that afects the health of camels. We assessed the profle of bacterial communities associated with H. dromedarii collected from camels in the eastern part of the UAE in 2010 and 2019. A total of 100 partially engorged female ticks were taken from tick samples collected from camels (n = 100; 50/year) and subjected to DNA extraction and sequencing. The 16S rRNA gene was amplifed from genomic DNA and sequenced using Illumina MiSeq platform to elucidate the bacterial communities. Principle Coordinates Analysis (PCoA) was conducted to determine patterns of diversity in bacterial communities. In 2010 and 2019, we obtained 899,574 and 781,452 read counts and these formed 371 and 191 operational taxonomic units (OTUs, clustered at 97% similarity), respectively. In both years, twenty‑fve bacterial families with high relative abundance were detected and the following were the most common: Moraxellaceae, Enterobacteriaceae, Staphylococcaceae, Bacillaceae, Corynebacteriaceae, Flavobacteriaceae, Francisellaceae, Muribaculaceae, Neisseriaceae, and Pseudomonadaceae. Francisellaceae and Enterobacteriaceae coexist in H. dromedarii and we suggest that they thrive under similar conditions and microbial interactions inside the host. Comparisons of diversity indicated that microbial communities difered in terms of richness and evenness between 2010 and 2019, with higher richness but lower evenness in communities in 2010. Principle coordinates analyses showed clear clusters separating microbial communities in 2010 and 2019. The diferences in communities suggested that the repertoire of microbial communities have shifted.
    [Show full text]
  • Distinct and Complex Bacterial Profiles in Human Periodontitis and Health Revealed by 16S Pyrosequencing
    The ISME Journal (2012) 6, 1176–1185 & 2012 International Society for Microbial Ecology All rights reserved 1751-7362/12 www.nature.com/ismej ORIGINAL ARTICLE Distinct and complex bacterial profiles in human periodontitis and health revealed by 16S pyrosequencing Ann L Griffen1,6, Clifford J Beall2,6, James H Campbell3,6, Noah D Firestone2, Purnima S Kumar4, Zamin K Yang3, Mircea Podar3,5 and Eugene J Leys2 1Division of Pediatric Dentistry and Community Oral Health, The Ohio State University College of Dentistry, Columbus, OH, USA; 2Division of Oral Biology, The Ohio State University College of Dentistry, Columbus, OH, USA; 3Biosciences Division, Oak Ridge National Laboratory, Oak Ridge, TN, USA; 4Division of Periodontology, The Ohio State University College of Dentistry, Columbus, OH, USA and 5Genome Science and Technology Program, University of Tennessee, Knoxville, TN, USA Periodontitis has a polymicrobial etiology within the framework of a complex microbial ecosystem. With advances in sequencing technologies, comprehensive studies to elucidate bacterial commu- nity differences have recently become possible. We used 454 sequencing of 16S rRNA genes to compare subgingival bacterial communities from 29 periodontally healthy controls and 29 subjects with chronic periodontitis. Amplicons from both the V1-2 and V4 regions of the 16S gene were sequenced, yielding 1 393 579 sequences. They were identified by BLAST against a curated oral 16S database, and mapped to 16 phyla, 106 genera, and 596 species. 81% of sequences could be mapped to cultivated species. Differences between health- and periodontitis-associated bacterial commu- nities were observed at all phylogenetic levels, and UniFrac and principal coordinates analysis showed distinct community profiles in health and disease.
    [Show full text]
  • Impact of Anaerobic Digestion and Centrifugation/Decanting Processes
    View metadata, citation and similar papers at core.ac.uk brought to you by CORE provided by Repositorio Institucional de la Universidad de Oviedo 1 Impact of anaerobic digestion and 2 centrifugation/decanting processes in 3 bacterial communities fractions 4 Short title: “Impact of anaerobic digestion in bacterial communities” 5 Ana Isabel Díaz1, Paula Oulego1, Sergio Collado1, Adriana Laca1, J. Manuel González2 1* 6 and Mario Díaz 7 1Department of Chemical and Environmental Engineering, University of Oviedo, C/ 8 Julián Clavería, s/n, Oviedo, E-33006, Spain 9 2 R&D, COGERSA SAU, C/ La Zoreda, s/n, Gijón, E-33697, Spain 10 *E-mail: [email protected], Phone: +34 985 10 34 39, Fax: +34 985 10 34 34 1 11 ABSTRACT 12 Sewage sludge can be treated by anaerobic processes that frequently are followed by 13 physical separation processes. In this work, a high-throughput sequencing technology, 14 based on variation in the bacterial 16S rRNA gene, has been used to characterise the 15 bacterial populations present in samples taken from different points of an industrial 16 anaerobic digestion process fed with sewage sludge. Relative abundances of phyla and 17 classes throughout the biological process and the subsequent separation steps were 18 determined. Results revealed that the Bacteroidetes, Firmicutes and Proteobacteria phyla 19 were the most representative. However, significant changes in relative abundance were 20 detected along treatments, showing the influence of operational parameters on the 21 distribution of microorganisms throughout the process. After anaerobic digestion, phylum 22 Firmicutes doubled its relative abundance, which seems to indicate that the anaerobic 23 conditions and the nutrients favoured its growth, in contrast to other phyla that almost 24 disappeared.
    [Show full text]