Megasphaera in the Stool Microbiota Is Negatively Associated With Diarrheal Cryptosporidiosis

Maureen A Carey, Gregory L Medlock, Masud Alam, Mamun Kabir, Md Jashim Uddin, Uma Nayak, Jason Papin, A S G Faruque, Rashidul Haque, William A Petri, Carol A Gilchrist, Maureen A Carey, Gregory L Medlock, Masud Alam, Mamun Kabir, Md Jashim Uddin, Uma Nayak, Jason Papin, A S G Faruque, Rashidul Haque, William A Petri, Carol A Gilchrist

Abstract

Background: The protozoan parasites in the Cryptosporidium genus cause both acute diarrheal disease and subclinical (ie, nondiarrheal) disease. It is unclear if the microbiota can influence the manifestation of diarrhea during a Cryptosporidium infection.

Methods: To characterize the role of the gut microbiota in diarrheal cryptosporidiosis, the microbiome composition of both diarrheal and surveillance Cryptosporidium-positive fecal samples from 72 infants was evaluated using 16S ribosomal RNA gene sequencing. Additionally, the microbiome composition prior to infection was examined to test whether a preexisting microbiome profile could influence the Cryptosporidium infection phenotype.

Results: Fecal microbiome composition was associated with diarrheal symptoms at 2 timepoints. Megasphaera was significantly less abundant in diarrheal samples compared with subclinical samples at the time of Cryptosporidium detection (log2 [fold change] = -4.3; P = 10-10) and prior to infection (log2 [fold change] = -2.0; P = 10-4); this assigned sequence variant was detected in 8 children who had diarrhea and 30 children without diarrhea. Random forest classification also identified Megasphaera abundance in the pre- and postexposure microbiota as predictive of a subclinical infection.

Conclusions: Microbiome composition broadly, and specifically low Megasphaera abundance, was associated with diarrheal symptoms prior to and at the time of Cryptosporidium detection. This observation suggests that the gut microenvironment may play a role in determining the severity of a Cryptosporidium infection. Clinical Trials Registration. NCT02764918.

Keywords: Cryptosporidium; Bangladesh; diarrhea; microbiome/microbiota; parasite.

© The Author(s) 2021. Published by Oxford University Press for the Infectious Diseases Society of America.

Figures

Figure 1.
Figure 1.
Study design. A, Overall cohort design and sample collection. For more information, see [1, 14]. Samples from Mirzapur were only used in post hoc analysis in Figure 4F. B, Paired samples were selected to assess Cryptosporidium-positive samples (time of detection) and the preceding surveillance sample (predetection). Cryptosporidium-positive samples were identified from both monthly surveillance and diarrheal stool samples, generating our subclinical and diarrheal sample groups.
Figure 2.
Figure 2.
Diarrheal infection and antibiotic treatment were common and heterogenous in infants from Mirpur. A, Prevalence of diarrhea. Frequency of diarrheal episodes per child. Full Mirpur cohort is shown in in gray; red subset indicates the children whose samples were used in the microbiome study (and all subsequent figures). B, All-cause diarrhea was heterogenous among children with divergent Cryptosporidium outcomes. Number of diarrheal events per child based on cumulative Cryptosporidium status, both over the first 2 years of life. C, Antibiotic usage was heterogenous among children with divergent Cryptosporidium outcomes. Number of antibiotic events per child based on cumulative Cryptosporidium status, both over the first 2 years of life. Combination therapies were treated as separate doses. For B and C, the full cohort was used and statistics are shown if significant. For B and C, each box represents the median (inner line), 25th percentile, and 75th percentile. Upper whiskers extend from the top of the box to the largest value within 1.5 times the interquartile range (distance between 25th and 75th percentile), and the lower whisker extends to the smallest value within 1.5 times the interquartile range. P values were generated from a t test without multiple testing correction.
Figure 3.
Figure 3.
Microbiome samples were highly variable. A, Most abundant amplicon sequence variants (ASVs) in the study. Only the top 10 most abundant ASVs are shown; the abundance of these common ASVs per sample is also represented in Supplementary Figure 3. Nearly 25% of all reads were assigned to an ASV in the Bifidobacterium genus. B, Richness of each sample, or the number of ASVs present in a sample, was not significantly different across sample groups. B and C, Each box represents the median (inner line), 25th percentile, and 75th percentile. Upper whiskers extend from the top of the box to the largest value within 1.5 times the interquartile range (distance between 25th and 75th percentile), and the lower whisker extends to the smallest value within 1.5 times the interquartile range. C, Evenness was also minimally different across sample groups. Evenness is a diversity metric calculated to represent how many different species are present and how well distributed those species are across samples; it is calculated using the inverse Simpson index. No significant differences in evenness was observed among any comparisons of clinical type (2-way analysis of variance with multiple testing correction via Tukey honest significant difference). D, Fraction of all samples containing a particular ASV, ordered by from highest to lowest. Very few ASVs were detected in many samples; however, almost all samples contain the most common Bifidobacterium ASVs.
Figure 4.
Figure 4.
Identifying associations between diarrheal symptoms and the microbiota. A, Predetection (PD) and time-of-detection (TOD) sample microbiota were indistinguishable via principal coordinate analysis using a permutational multivariate analysis of variance using distance matrices and a significance cutoff of P < .05, as were subclinical and diarrheal Cryptosporidium-positive samples. Principal coordinate analysis of amplicon sequence variants (ASV) quantification across all samples using Euclidean distance. B, Univariate statistics identifies ASVs associated with symptoms in the PD samples and TOD samples. Statistically significant differential expressed ASVs are colored, whereas gray points represent ASVs that were not different or not significantly different, using DESeq2. Large points indicate ASVs that were also identified as important using random forest classification, whereas small points were not among the top 15 most important variables. Random forest classifiers were built to predict the presence of diarrhea upon Cryptosporidium infection. Importantly, purple points represent statistically significant ASVs that were also among the most important variables for classifiers made at both timepoints. C, Random forest classifiers were built from the TOD microbiota (blue) or predetection microbiota (red). Area under the curve (AUC), a metric of classifier accuracy, is listed for each classifier. D, Most important variables, as ranked by mean decrease in node impurity (or Gini importance), from the PD and TOD classifiers. Important variables were similarly important, within and across models. Of note, age was not an important variable in the TOD classifier. E, One ASV assigned to the Megasphaera genus was significantly less abundant in diarrheal cases via univariate analyses (at both timepoints) and was among the top 15 most important variables for the classifiers for both timepoints. Relative abundance of each ASV is plotted for each sample, with each box representing the median (inner line), 25th percentile, and 75th percentile. Upper whiskers extend from the top of the box to the largest value within 1.5 times the interquartile range (distance between 25th and 75th percentile), and the lower whisker extends to the smallest value within 1.5 times the interquartile range. F, The Megasphaera ASV was also more likely to be high-abundance (above dashed line) in samples at the second study site, Mirzapur, where diarrheal cryptosporidiosis was less common when compared to Mirpur; however, environmental factors, including the causal Cryptosporidium species, were also different in Mirzapur [1]. Increased Megasphaera abundance in Mirzapur may partially explain reduced diarrhea associated with cryptosporidiosis in that community.

References

    1. Steiner KL, Ahmed S, Gilchrist CA, et al. . Species of cryptosporidia causing subclinical infection associated with growth faltering in rural and urban Bangladesh—a birth cohort study. Clin Infect Dis 2018; 67:1347–55.
    1. Checkley W, Epstein LD, Gilman RH, Black RE, Cabrera L, Sterling CR. Effects of Cryptosporidium parvum infection in Peruvian children: growth faltering and subsequent catch-up growth. Am J Epidemiol 1998; 148:497–506.
    1. Korpe PS, Haque R, Gilchrist C, et al. . Natural history of cryptosporidiosis in a longitudinal study of slum-dwelling Bangladeshi children: association with severe malnutrition. PLoS Negl Trop Dis 2016; 10:e0004564.
    1. Lima AA, Fang G, Schorling JB, et al. . Persistent diarrhea in northeast Brazil: etiologies and interactions with malnutrition. Acta Paediatr Suppl 1992; 381:39–44.
    1. Guerrant DI, Moore SR, Lima AA, Patrick PD, Schorling JB, Guerrant RL. Association of early childhood diarrhea and cryptosporidiosis with impaired physical fitness and cognitive function four-seven years later in a poor urban community in northeast Brazil. Am J Trop Med Hyg 1999; 61:707–13.
    1. Sallon S, Deckelbaum RJ, Schmid II, Harlap S, Baras M, Spira DT. Cryptosporidium, malnutrition, and chronic diarrhea in children. Am J Dis Child 1988; 142:312–5.
    1. Platts-Mills JA, Babji S, Bodhidatta L, et al. ; MAL-ED Network Investigators . Pathogen-specific burdens of community diarrhoea in developing countries: a multisite birth cohort study (MAL-ED). Lancet Glob Health 2015; 3:e564–75.
    1. Khalil IA, Troeger C, Rao PC, et al. . Morbidity, mortality, and long-term consequences associated with diarrhoea from Cryptosporidium infection in children younger than 5 years: a meta-analyses study. Lancet Glob Health 2018; 6:e758–68.
    1. GBD 2016 Diarrhoeal Disease Collaborators. Estimates of the global, regional, and national morbidity, mortality, and aetiologies of diarrhoea in 195 countries: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Infect Dis 2018; 18:1211–28.
    1. Manjunatha UH, Vinayak S, Zambriski JA, et al. . A Cryptosporidium PI(4)K inhibitor is a drug candidate for cryptosporidiosis. Nature 2017; 546:376–80.
    1. Wojcik GL, Korpe P, Marie C, et al. . Genome-wide association study of cryptosporidiosis in infants implicates PRKCA. mBio 2020; 11:e..
    1. Kirkpatrick BD, Haque R, Duggal P, et al. . Association between Cryptosporidium infection and human leukocyte antigen class I and class II alleles. J Infect Dis 2008; 197:474–8.
    1. Nader JL, Mathers TC, Ward BJ, et al. . Evolutionary genomics of anthroponosis in Cryptosporidium. Nat Microbiol 2019; 4:826–36.
    1. Gilchrist CA, Cotton JA, Burkey C, et al. . Genetic diversity of Cryptosporidium hominis in a Bangladeshi community as revealed by whole-genome sequencing. J Infect Dis 2018; 218:259–64.
    1. Heiges M, Wang H, Robinson E, et al. . CryptoDB: a Cryptosporidium bioinformatics resource update. Nucleic Acids Res 2006; 34:D419–22.
    1. Sateriale A, Šlapeta J, Baptista R, et al. . A genetically tractable, natural mouse model of cryptosporidiosis offers insights into host protective immunity. Cell Host Microbe 2019; 26:135–46.e5.
    1. Chappell CL, Darkoh C, Shimmin L, et al. . Fecal indole as a biomarker of susceptibility to Cryptosporidium infection. Infect Immun 2016; 84:2299–306.
    1. Harp JA, Wannemuehler MW, Woodmansee DB, Moon HW. Susceptibility of germfree or antibiotic-treated adult mice to Cryptosporidium parvum. Infect Immun 1988; 56:2006–10.
    1. Harp JA, Chen W, Harmsen AG. Resistance of severe combined immunodeficient mice to infection with Cryptosporidium parvum: the importance of intestinal microflora. Infect Immun 1992; 60:3509–12.
    1. Faith JJ, Guruge JL, Charbonneau M, et al. . The long-term stability of the human gut microbiota. Science 2013; 341:1237439.
    1. Caporaso JG, Lauber CL, Walters WA, et al. . Ultra-high-throughput microbial community analysis on the Illumina HiSeq and MiSeq platforms. ISME J 2012; 6:1621–4.
    1. Bushnell B;Joint Genome Institute. BBTools: a suite of fast, multithreaded bioinformatics tools designed for analysis of DNA and RNA sequence data.2018. Available at: . Accessed 24 July 2020.
    1. Martin M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet. J 2011; 17:10–2.
    1. Callahan BJ, McMurdie PJ, Rosen MJ, Han AW, Johnson AJ, Holmes SP. DADA2: high-resolution sample inference from Illumina amplicon data. Nat Methods 2016; 13:581–3.
    1. Weiss S, Xu ZZ, Peddada S, et al. . Normalization and microbial differential abundance strategies depend upon data characteristics. Microbiome 2017; 5:27.
    1. R Computing Team. A language and environment for statistical computing. Vienna, Austria: R Foundation for Statistical Computing, 2015.
    1. R Computing Team. RStudio: integrated development for R. Boston, MA: RStudio, Inc,2015.
    1. Morgan M, Anders S, Lawrence M, Aboyoun P, Pagès H, Gentleman R. ShortRead: a bioconductor package for input, quality assessment and exploration of high-throughput sequence data. Bioinformatics 2009; 25:2607–8.
    1. McMurdie PJ, Holmes S. phyloseq: an R package for reproducible interactive analysis and graphics of microbiome census data. PLoS One 2013; 8:e61217.
    1. Breiman L. Random forests. Mach Learn 2001; 45:5–32.
    1. Wickham H. reshape2: Flexibly reshape data: a reboot of the reshape package.R package version 2012:1. Available at: . Accessed 1 October 2018.
    1. Wickham H, Chang W, Others. ggplot2: an implementation of the grammar of graphics. R package version 0 7.2008. Available at: . Accessed 1 October 2018.
    1. Attali D, Baker C. ggExtra: add marginal histograms to “ggplot2,” and more “ggplot2”enhancements. R package version 0 3. 2016:4.
    1. Kassambara A. ggpubr":ggplot2” based publication ready plots. R package version 0 1.2017:6.
    1. Allaire JJ, Xie Y, McPherson J, et al. . rmarkdown: dynamic documents for R. 2018. Available at: . Accessed 1 October 2018.
    1. Xie Y. knitr: a comprehensive tool for reproducible research in R. Implement Reprod Res 2014. Available at: . Accessed 1 October 2018.
    1. Liaw A, Wiener M, Others. Classification and regression by randomForest. R News 2002; 2:18–22.
    1. Paluszynska A, Biecek P. randomForestExplainer: explaining and visualizing random forests in terms of variable importance.2017. Available at: . Accessed 1 October 2018.
    1. Urrea V, Calle M. AUCRF: variable selection with random forest and the area under the curve. R package version 1 1. 2012.
    1. Robin X, Turck N, Hainard A, et al. . pROC: an open-source package for R and S+ to analyze and compare ROC curves. BMC Bioinformatics 2011; 12:77.
    1. Wickham H, Francois R, Henry L, Müller K. dplyr: a grammar of data manipulation. R package version 0 4. 2015:3.
    1. Wagner H. Vegan: community ecology package. R package. 2015.
    1. Wickham H, Henry L, Others. tidyr: Easily Tidy Data with “spread ()”and “gather ()”Functions. R package version 0 8. 2018:2.
    1. Calle ML. Statistical analysis of metagenomics data. Genomics Inform 2019; 17:e6.
    1. Love MI, Huber W, Anders S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol 2014; 15:550.
    1. Duvallet C, Gibbons SM, Gurry T, Irizarry RA, Alm EJ. Meta-analysis of gut microbiome studies identifies disease-specific and shared responses. Nat Commun 2017; 8:1784.
    1. Topçuoğlu BD, Lesniak NA, Ruffin MT 4th, Wiens J, Schloss PD. A framework for effective application of machine learning to microbiome-based classification problems. mBio 2020; 11:.
    1. Bokulich NA, Chung J, Battaglia T, et al. . Antibiotics, birth mode, and diet shape microbiome maturation during early life. Sci Transl Med 2016; 8:343ra82.
    1. Chen RY, Kung VL, Das S, et al. . Duodenal microbiota in stunted undernourished children with enteropathy. N Engl J Med 2020; 383:321–33.
    1. Zozaya-Hinchliffe M, Martin DH, Ferris MJ. Prevalence and abundance of uncultivated Megasphaera-like bacteria in the human vaginal environment. Appl Environ Microbiol 2008; 74:1656–9.
    1. Guo C, Li Y, Wang P, et al. . Alterations of gut microbiota in cholestatic infants and their correlation with hepatic function. Front Microbiol 2018; 9:2682.
    1. Chappell CL, Okhuysen PC, Langer-Curry RC, Akiyoshi DE, Widmer G, Tzipori S. Cryptosporidium meleagridis: infectivity in healthy adult volunteers. Am J Trop Med Hyg 2011; 85:238–42.
    1. Cama VA, Bern C, Roberts J, et al. . Cryptosporidium species and subtypes and clinical manifestations in children, Peru. Emerg Infect Dis 2008; 14:1567–74.
    1. Odamaki T, Kato K, Sugahara H, et al. . Age-related changes in gut microbiota composition from newborn to centenarian: a cross-sectional study. BMC Microbiol 2016; 16:90.
    1. Yatsunenko T, Rey FE, Manary MJ, et al. . Human gut microbiome viewed across age and geography. Nature 2012; 486:222–7.
    1. Costello EK, Carlisle EM, Bik EM, Morowitz MJ, Relman DA. Microbiome assembly across multiple body sites in low-birthweight infants. mBio 2013; 4:e00782–13.
    1. Avershina E, Storrø O, Øien T, Johnsen R, Pope P, Rudi K. Major faecal microbiota shifts in composition and diversity with age in a geographically restricted cohort of mothers and their children. FEMS Microbiol Ecol 2014; 87:280–90.
    1. Kurokawa K, Itoh T, Kuwahara T, et al. . Comparative metagenomics revealed commonly enriched gene sets in human gut microbiomes. DNA Res 2007; 14:169–81.
    1. Koenig JE, Spor A, Scalfone N, et al. . Succession of microbial consortia in the developing infant gut microbiome. Proc Natl Acad Sci U S A 2011; 108:4578–85.
    1. McKenney EA, Greene LK, Drea CM, Yoder AD. Down for the count: Cryptosporidium infection depletes the gut microbiome in Coquerel’s sifakas. Microb Ecol Health Dis 2017; 28:1335165.
    1. Ichikawa-Seki M, Motooka D, Kinami A, et al. . Specific increase of Fusobacterium in the faecal microbiota of neonatal calves infected with Cryptosporidium parvum. Sci Rep 2019; 9:12517.
    1. Ras R, Huynh K, Desoky E, Badawy A, Widmer G. Perturbation of the intestinal microbiota of mice infected with Cryptosporidium parvum. Int J Parasitol 2015; 45:567–73.
    1. Oliveira BCM, Widmer G. Probiotic product enhances susceptibility of mice to cryptosporidiosis. Appl Environ Microbiol 2018; 84:.
    1. Charania R, Wade BE, McNair NN, Mead JR. Changes in the microbiome of Cryptosporidium-infected mice correlate to differences in susceptibility and infection levels. Microorganisms 2020; 8:879.
    1. Oliveira BCM, Bresciani KDS, Widmer G. Deprivation of dietary fiber enhances susceptibility of mice to cryptosporidiosis. PLoS Negl Trop Dis 2019; 13:e0007411.
    1. Shetty SA, Marathe NP, Lanjekar V, Ranade D, Shouche YS. Comparative genome analysis of Megasphaera sp. reveals niche specialization and its potential role in the human gut. PLoS One 2013; 8:e79353.
    1. Parada Venegas D, De la Fuente MK, Landskron G, et al. . Short chain fatty acids (SCFAs)-mediated gut epithelial and immune regulation and its relevance for inflammatory bowel diseases. Front Immunol 2019; 10:277.
    1. Bachem A, Makhlouf C, Binger KJ, et al. . Microbiota-derived short-chain fatty acids promote the memory potential of antigen-activated CD8+ T cells. Immunity 2019; 51:285–97.e5.
    1. Binder HJ. Role of colonic short-chain fatty acid transport in diarrhea. Annu Rev Physiol 2010; 72:297–313.
    1. Sedighi R, Alipour D. Assessment of probiotic effects of isolated Megasphaera elsdenii strains in Mehraban sheep and Holstein lactating cows. Anim Feed Sci Technol 2019; 248:126–31.

Source: PubMed

3
구독하다