Abstract
Changes in the gut microbiota during pathogen infection are often predicted to influence disease outcomes. However, studies exploring whether pathogens induce microbiota shifts have yielded inconsistent results. This suggests that variation in infection, rather than the presence of infection alone, might shape pathogen–microbiota relationships. For example, most hosts are coinfected with multiple pathogens simultaneously, and hosts vary in how long they are infected, which may amplify or diminish microbial shifts expected in response to a focal pathogen. We used a longitudinal anthelmintic treatment study of free-ranging African buffalo (Syncerus caffer) to examine whether (i) coinfection with bovine tuberculosis (Mycobacterium bovis, TB) and gastrointestinal nematodes, and (ii) the duration of TB infection, modified effects of single pathogens on the gut microbiota. By accounting for the interaction between TB and nematodes, we found that coinfection affected changes in microbial abundance associated with single infections. Furthermore, the duration of TB infection predicted more microbiota variation than the presence of TB. Importantly, coinfection and infection duration had nearly as much influence on microbial patterns as demographic and environmental factors commonly examined in microbiota research. These findings demonstrate that acknowledging infection heterogeneities may be crucial to understanding relationships between pathogens and the gut microbiota.
Similar content being viewed by others
Log in or create a free account to read this content
Gain free access to this article, as well as selected content from this journal and more on nature.com
or
Data availability
Sequence data and metadata are available through the Earth Microbiome Project data portal (qiita.microbio.me/emp) under study ID 1736, file 30386.
References
Libertucci J, Young VB. The role of the microbiota in infectious diseases. Nat Microbiol. 2019;4:35–45.
Leung JM, Graham AL, Knowles SCL. Parasite-microbiota interactions with the vertebrate gut: Synthesis through an ecological lens. Front Microbiol. 2018;9:1–20.
Hols P, Ledesma-García L, Gabant P, Mignolet J. Mobilization of microbiota commensals and their bacteriocins for therapeutics. Trends Microbiol. 2019;27:690–702.
Eribo OA, du Plessis N, Ozturk M, Guler R, Walzl G, Chegou NN. The gut microbiome in tuberculosis susceptibility and treatment response: guilty or not guilty? Cell Mol Life Sci. 2020;77:1497–509.
Lv L-X, Jiang H-Y, Yan R, Li L. Interactions between gut microbiota and hosts and their role in infectious diseases. Infect Microbes Dis. 2019;1:3–9.
Cortes A, Peachey LE, Jenkins TP, Scotti R, Cantacessi C. Helminths and microbes within the vertebrate gut—not all studies are created equal. Parasitology. 2019;146:1–8.
Tuddenham SA, Koay WLA, Zhao N, White JR, Ghanem KG, Sears CL. The impact of human immunodeficiency virus infection on gut microbiota α-diversity: an individual-level meta-analysis. Clin Infect Dis. 2020;70:615–27.
Lozupone CA, Li M, Campbell TB, Flores SC, Linderman D, Gebert MJ, et al. Alterations in the gut microbiota associated with HIV-1 infection. Cell Host Microbe. 2013;14:329–39.
McHardy IH, Li X, Tong M, Ruegger P, Jacobs J, Borneman J, et al. HIV Infection is associated with compositional and functional shifts in the rectal mucosal microbiota. Microbiome. 2013;1:1.
Chang JY, Antonopoulos DA, Kalra A, Tonelli A, Khalife WT, Schmidt TM, et al. Decreased diversity of the fecal microbiome in recurrent clostridium difficile-associated diarrhea. J Infect Dis. 2008;197:435–8.
Maji A, Misra R, Dhakan DB, Gupta V, Mahato NK, Saxena R, et al. Gut microbiome contributes to impairment of immunity in pulmonary tuberculosis patients by alteration of butyrate and propionate producers. Environ Microbiol. 2018;20:402–19.
Luo M, Liu Y, Wu P, Luo DX, Sun Q, Zheng H, et al. Alternation of gut microbiota in patients with pulmonary tuberculosis. Front Physiol. 2017;8:822.
Scotti R, Southern S, Boinett C, Jenkins TP, Cortés A, Cantacessi C. MICHELINdb: a web-based tool for mining of helminth-microbiota interaction datasets, and a meta-analysis of current research. Microbiome. 2020;8:10.
Kreisinger J, Bastien G, Hauffe HC, Marchesi J, Perkins SE. Interactions between multiple helminths and the gut microbiota in wild rodents. Philos Trans R Soc B Biol Sci. 2015;370:20140295.
Ezenwa VO. Helminth-microparasite co-infection in wildlife: lessons from ruminants, rodents and rabbits. Parasite Immunol. 2016;38:527–34.
Babu S, Nutman TB. Helminth-tuberculosis co-infection: an immunologic perspective. Trends Immunol. 2016;37:597–607.
Rooks MG, Garrett WS. Gut microbiota, metabolites and host immunity. Nat Rev Immunol. 2016;16:341–52.
Cooper P, Walker AW, Reyes J, Chico M, Salter SJ, Vaca M, et al. Patent human infections with the whipworm, trichuris trichiura, are not associated with alterations in the faecal microbiota. PLoS ONE. 2013;8:e76573.
Welsh MD, Cunningham RT, Corbett DM, Girvin RM, McNair J, Skuce RA, et al. Influence of pathological progression on the balance between cellular and humoral immune responses in bovine tuberculosis. Immunology. 2005;114:101–11.
Hoffmann M, Pantazis N, Martin GE, Hickling S, Hurst J, Meyerowitz J, et al. Exhaustion of activated CD8 T cells predicts disease progression in primary HIV-1 infection. PLoS Pathog. 2016;12:1–19.
Murphy L, Nalpas N, Stear M, Cattadori IM. Explaining patterns of infection in free-living populations using laboratory immune experiments. Parasite Immunol. 2011;33:287–302.
Rothschild D, Weissbrod O, Barkan E, Kurilshikov A, Korem T, Zeevi D, et al. Environment dominates over host genetics in shaping human gut microbiota. Nature. 2018;555:210–5.
Yatsunenko T, Rey FE, Manary MJ, Trehan I, Dominguez-Bello MG, Contreras M, et al. Human gut microbiome viewed across age and geography. Nature. 2012;486:222–7.
Xu Z, Knight R. Dietary effects on human gut microbiome diversity. Br J Nutr. 2015;113:S1–5.
Graf D, Di Cagno R, Fåk F, Flint HJ, Nyman M, Saarela M, et al. Contribution of diet to the composition of the human gut microbiota. Micro Ecol Heal Dis. 2015;26:1–11.
Rosa BA, Supali T, Gankpala L, Djuardi Y, Sartono E, Zhou Y, et al. Differential human gut microbiome assemblages during soil-transmitted helminth infections in Indonesia and Liberia. Microbiome. 2018;6:1–19.
Wasimuddin Brändel SD, Tschapka M, Page R, Rasche A, Corman VM, et al. Astrovirus infections induce age-dependent dysbiosis in gut microbiomes of bats. ISME J. 2018;12:2883–93.
Arrazuria R, Elguezabal N, Juste RA, Derakhshani H, Khafipour E. Mycobacterium avium subspecies paratuberculosis infection modifies gut microbiota under different dietary conditions in a rabbit model. Front Microbiol. 2016;7:1–14.
Cattadori IM, Sebastian A, Hao H, Katani R, Albert I, Eilertson KE, et al. Impact of helminth infections and nutritional constraints on the small intestine microbiota. PLoS ONE. 2016;11:1–23.
Ezenwa VO, Jolles AE, Beechler BR, Budischak SA, Gorsich EE. The causes and consequences of parasite interactions: African buffalo as a case study. In: Wilson K, Fenton A, Tompkins D (eds). Wildlife Disease Ecology: Linking Theory to Data and Application. 1st edn. (Cambridge University Press, Cambridge, 2019). pp 129–60.
Ezenwa VO, Etienne RS, Luikart G, Beja-Pereira A, Jolles AE. Hidden consequences of living in a wormy world: nematode‐induced immune suppression facilitates tuberculosis invasion in African buffalo. Am Nat. 2010;176:613–24.
Jolles AE, Ezenwa VO, Etienne RS, Turner WC, Olff H. Interactions between macroparasites and microparasites drive infection patterns in free-ranging African buffalo. Ecology. 2008;89:2239–50.
Ezenwa VO, Jolles AE. Opposite effects of anthelmintic treatment on microbial infection at individual versus population scales. Science. 2015;347:175–7.
Ezenwa VO, Budischak SA, Buss P, Seguel M, Luikart G, Jolles AE, et al. Natural resistance to worms exacerbates bovine tuberculosis severity independently of worm coinfection. bioRxiv. 2020.
Budischak SA, Hoberg EP, Abrams ART, Jolles AE, Ezenwa VO. A combined parasitological molecular approach for noninvasive characterization of parasitic nematode communities in wild hosts. Mol Ecol Resour. 2015;15:1112–9.
Williams JC. Efficacy of albendazole, levamisole and fenbendazole against gastrointestinal nematodes of cattle, with emphasis on inhibited early fourth stage Ostertagia ostertagi larvae. Vet Parasitol. 1991;40:59–71.
Jolles AE, Cooper DV, Levin SA. Hidden effects of chronic tuberculosis in African buffalo. Ecology. 2005;86:2358–64.
Ezenwa VO, Jolles AE, O’Brien MP. A reliable body condition scoring technique for estimating condition in African buffalo. Afr J Ecol. 2009;47:476–81.
Ryan SJ, Cross PC, Winnie J, Hay C, Bowers J, Getz WM. The utility of normalized difference vegetation index for predicting African buffalo forage quality. J Wildl Manag. 2012;76:1499–508.
Budischak SA, O’Neal D, Jolles AE, Ezenwa VO. Differential host responses to parasitism shape divergent fitness costs of infection. Funct Ecol. 2018;32:324–33.
Coetsee C, February EC, Bond WJ. Nitrogen availability is not affected by frequent fire in a South African savanna. J Trop Ecol. 2008;24:647–54.
Thompson LR, Sanders JG, McDonald D, Amir A, Ladau J, Locey KJ, et al. A communal catalogue reveals Earth’s multiscale microbial diversity. Nature. 2017;551:457–63.
Caporaso JG, Lauber CL, Walters WA, Berg-Lyons D, Lozupone CA, Turnbaugh PJ, et al. Global patterns of 16S rRNA diversity at a depth of millions of sequences per sample. Proc Natl Acad Sci. 2011;108:4516–22.
Caporaso JG, Lauber CL, Walters WA, Berg-Lyons D, Huntley J, Fierer N, et al. Ultra-high-throughput microbial community analysis on the Illumina HiSeq and MiSeq platforms. ISME J. 2012;6:1621–4.
Gonzalez A, Navas-Molina JA, Kosciolek T, McDonald D, Vázquez-Baeza Y, Ackermann G, et al. Qiita: rapid, web-enabled microbiome meta-analysis. Nat Methods. 2018;15:796–8.
Bokulich NA, Subramanian S, Faith JJ, Gevers D, Gordon JI, Knight R, et al. Quality-filtering vastly improves diversity estimates from Illumina amplicon sequencing. Nat Methods. 2013;10:57–9.
Amir A, McDonald D, Navas-Molina JA, Kopylova E, Morton JT, Zech Xu Z, et al. Deblur rapidly resolves single-nucleotide community sequence patterns. mSystems. 2017;2:e00191–16.
Bolyen E, Rideout JR, Dillon MR, Bokulich NA, Abnet CC, Al-Ghalith GA, et al. Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2. Nat Biotechnol. 2019;37:852–7.
Janssen S, McDonald D, Gonzalez A, Navas-Molina JA, Jiang L, Xu ZZ, et al. Phylogenetic placement of exact amplicon sequences improves associations with clinical information. mSystems. 2018;3:e00021–18.
Weiss S, Xu ZZ, Peddada S, Amir A, Bittinger K, Gonzalez A, et al. Normalization and microbial differential abundance strategies depend upon data characteristics. Microbiome. 2017;5:27.
McDonald D, Price MN, Goodrich J, Nawrocki EP, DeSantis TZ, Probst A, et al. An improved Greengenes taxonomy with explicit ranks for ecological and evolutionary analyses of bacteria and archaea. ISME J. 2012;6:610–8.
Sonnenburg JL, Bäckhed F. Diet-microbiota interactions as moderators of human metabolism. Nature. 2016;535:56–64.
Viney M. The gut microbiota of wild rodents: challenges and opportunities. Lab Anim. 2019;53:252–8.
Koren O, Goodrich JK, Cullender TC, Spor A, Laitinen K, Kling Bäckhed H, et al. Host remodeling of the gut microbiome and metabolic changes during pregnancy. Cell. 2012;150:470–80.
Beechler BR, Jolles AE, Budischak SA, Corstjens PLAM, Ezenwa VO, Smith M, et al. Host immunity, nutrition and coinfection alter longitudinal infection patterns of schistosomes in a free ranging African buffalo population. PLoS Negl Trop Dis. 2017;11:1–24.
Simpson GL. permute: Functions for generating restricted permutations of data. R package version 0.9-4. https://cran.r-project.org/package=permute.
Oksanen JF, Blanchet G, Friendly M, Kindt R, Legendre P, McGlinn D, et al. vegan: Community ecology package. R package version 2.5-2. https://cran.r-project.org/package=vegan.
Bates D, Mächler M, Bolker B, Walker S. Fitting linear mixed-effects models using lme4. J Stat Softw. 2015;67:1–48.
Kuznetsova A, Brockhoff PB, Christensen RHB. lmerTest Package: tests in linear mixed effects models. J Stat Softw. 2017;82:1–26.
Stoffel M, Nakagawa S, Schielzeth H. partR2: Partitioning R2 in GLMMs. R package version 0.9.0. 2020.
Wang Y, Naumann U, Eddelbuettel D, Wilshire J, Warton D. mvabund: Statistical methods for analysing multivariate abundance data. R package version 4.1.3. https://cran.r-project.org/package=mvabund.
Morton JT, Marotz C, Washburne A, Silverman J, Zaramela LS, Edlund A, et al. Establishing microbial composition measurement standards with reference frames. Nat Commun. 2019;10:2719.
Wang Y, Naumann U, Wright ST, Warton DI. mvabund—An R package for model-based analysis of multivariate abundance data. Methods Ecol Evol. 2012;3:471–4.
Leung JM, Budischak SA, Chung The H, Hansen C, Bowcutt R, Neill R, et al. Rapid environmental effects on gut nematode susceptibility in rewilded mice. PLoS Biol. 2018;16:1–28.
Qian LJ, Kang SM, Xie JL, Huang L, Wen Q, Fan YY, et al. Early-life gut microbial colonization shapes Th1/Th2 balance in asthma model in BALB/c mice. BMC Microbiol. 2017;17:1–8.
Bang C, Weidenbach K, Gutsmann T, Heine H, Schmitz RA. The intestinal archaea Methanosphaera stadtmanae and Methanobrevibacter smithii activate human dendritic cells. PLoS ONE. 2014;9:1–9.
Bernatchez E, Gold MJ, Langlois A, Blais-Lecours P, Boucher M, Duchaine C, et al. Methanosphaera stadtmanae induces a type IV hypersensitivity response in a mouse model of airway inflammation. Physiol Rep. 2017;5:1–13.
Peachey LE, Molena RA, Jenkins TP, Di Cesare A, Traversa D, Hodgkinson JE, et al. The relationships between faecal egg counts and gut microbial composition in UK Thoroughbreds infected by cyathostomins. Int J Parasitol. 2018;48:403–12.
Su C, Su L, Li Y, Chang J, Zhang W, Walker WA, et al. Helminth-induced alterations of the gut microbiota exacerbate bacterial colitis. Mucosal Immunol. 2018;11:144–57.
Reynolds LA, Finlay BB, Maizels RM. Cohabitation in the intestine: interactions among helminth parasites, bacterial microbiota, and host immunity. J Immunol. 2015;195:4059–66.
Martin I, Kaisar MMM, Wiria AE, Hamid F, Djuardi Y, Sartono E, et al. The effect of gut microbiome composition on human immune responses: an exploration of interference by helminth infections. Front Genet. 2019;10:1028.
Preston DL, Henderson JS, Johnson PTJ. Community ecology of invasions: direct and indirect effects of multiple invasive species on aquatic communities. Ecology. 2012;93:1254–61.
Sih A, Englund G, Wooster D. Emergent impacts of multiple predators on prey. Trends Ecol Evol. 1998;13:350–5.
Telfer S, Lambin X, Birtles R, Beldomenico P, Burthe S, Paterson S, et al. Species interactions in a parasite community drive infection risk in a wildlife population. Science. 2010;330:243–7.
Johnson PTJ, Hoverman JT. Parasite diversity and coinfection determine pathogen infection success and host fitness. Proc Natl Acad Sci USA. 2012;109:9006–11.
Griffiths EC, Pedersen AB, Fenton A, Petchey OL. Analysis of a summary network of co-infection in humans reveals that parasites interact most via shared resources. Proc R Soc B Biol Sci. 2014;281:20132286.
Kant S, Gupta H, Ahluwalia S. Significance of nutrition in pulmonary tuberculosis. Crit Rev Food Sci Nutr. 2015;55:955–63.
Coop RL, Kyriazakis I. Influence of host nutrition on the development and consequences of nematode parasitism in ruminants. Trends Parasitol. 2001;17:325–30.
Hu Y, Feng Y, Wu J, Liu F, Zhang Z, Hao Y, et al. The gut microbiome signatures discriminate healthy from pulmonary tuberculosis patients. Front Cell Infect Microbiol. 2019;9:1–8.
Li RW, Wu S, Li W, Navarro K, Couch RD, Hill D, et al. Alterations in the porcine colon microbiota induced by the gastrointestinal nematode Trichuris suis. Infect Immun. 2012;80:2150–7.
Hicks AL, Lee KJ, Couto-Rodriguez M, Patel J, Sinha R, Guo C, et al. Gut microbiomes of wild great apes fluctuate seasonally in response to diet. Nat Commun. 2018;9:1786.
Bi Y, Zeng S, Zhang R, Diao Q, Tu Y. Effects of dietary energy levels on rumen bacterial community composition in Holstein heifers under the same forage to concentrate ratio condition. BMC Microbiol. 2018;18:1–11.
Omoniyi LA, Jewell KA, Isah OA, Neumann AP, Onwuka CFI, Onagbesan OM, et al. An analysis of the ruminal bacterial microbiota in West African Dwarf sheep fed grass- and tree-based diets. J Appl Microbiol. 2014;116:1094–105.
Fujishiro MA, Lidbury JA, Pilla R, Steiner JM, Lappin MR, Suchodolski JS. Evaluation of the effects of anthelmintic administration on the fecal microbiome of healthy dogs with and without subclinical Giardia spp. and Cryptosporidium canis infections. PLoS ONE. 2020;15:1–17.
Korte SW, Franklin CL, Dorfmeyer RA, Ericsson AC. Effects of fenbendazole-impregnated feed and topical moxidectin during quarantine on the gut microbiota of C57BL/6 mice. J Am Assoc Lab Anim Sci. 2018;57:229–35.
Martin I, Martin I, Djuardi Y, Supali T, Sartono E, Yazdanbakhsh M, et al. Dynamic changes in human-gut microbiome in relation to a placebo-controlled anthelminthic trial in Indonesia. PLoS Negl Trop Dis. 2018;12:e0006620.
Winglee K, Eloe-Fadrosh E, Gupta S, Guo H, Fraser C, Bishai W. Aerosol mycobacterium tuberculosis infection causes rapid loss of diversity in gut microbiota. PLoS ONE. 2014;9:1–9.
Chackerian AA, Alt JM, Perera TV, Dascher CC, Behar SM. Dissemination of mycobacterium tuberculosis is influenced by host factors and precedes the initiation of T-cell immunity. Infect Immun. 2002;70:4501–9.
Hernández-Pando R, Orozcoe H, Sampieri A, Pavón L, Velasquillo C, Larriva-Sahd J, et al. Correlation between the kinetics of Th1, Th2 cells and pathology in a murine model of experimental pulmonary tuberculosis. Immunology. 1996;89:26–33.
Pedersen AB, Antonovics J. Anthelmintic treatment alters the parasite community in a wild mouse host. Biol Lett. 2013;9:20130205.
Pedersen AB, Fenton A. Emphasizing the ecology in parasite community ecology. Trends Ecol Evol. 2007;22:133–9.
Thakar J, Pathak AK, Murphy L, Albert R, Cattadori IM. Network model of immune responses reveals key effectors to single and co-infection dynamics by a respiratory bacterium and a gastrointestinal helminth. PLoS Comput Biol. 2012;8:e1002345.
Acknowledgements
This work was supported by the National Science Foundation (Grant #s: DEB-1102493, DGE-1545433), the Earth Microbiome Project, and an ARCS Foundation Award. Animal protocols for this study were approved by the University of Georgia (UGA) and Oregon State University (OSU) Institutional Animal Care and Use Committees (UGA AUP A2010 10-190-Y3-A5; OSU AUP 3822 and 4325). We thank South African National Parks (SANParks) for permission to conduct this study in Kruger; M. Hofmeyr, P. Buss, and the entire SANParks Veterinary Wildlife Services Department for assistance with animal captures and project logistics; and R. Spaan, J. Spaan, K. Thompson, B. Beechler, S. Budischak, and K. Lane-deGraaf for work on animal captures and sample processing. We also thank D. Warton, J. Morton, and JP Schmidt for advice on statistical analyses, and members of the Ezenwa Lab for thoughtful comments on drafts of the manuscript.
Author information
Authors and Affiliations
Corresponding author
Ethics declarations
Conflict of interest
The authors declare that they have no conflict of interest.
Additional information
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Supplementary information
Rights and permissions
About this article
Cite this article
Sabey, K.A., Song, S.J., Jolles, A. et al. Coinfection and infection duration shape how pathogens affect the African buffalo gut microbiota. ISME J 15, 1359–1371 (2021). https://doi.org/10.1038/s41396-020-00855-0
Received:
Revised:
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1038/s41396-020-00855-0