The Integrative Human Microbiome Project

Integrative HMP (iHMP) Research Network Consortium, Lita M Proctor, Heather H Creasy, Jennifer M Fettweis, Jason Lloyd-Price, Anup Mahurkar, Wenyu Zhou, Gregory A Buck, Michael P Snyder, Jerome F Strauss 3rd, George M Weinstock, Owen White, Curtis Huttenhower, Integrative HMP (iHMP) Research Network Consortium, Lita M Proctor, Heather H Creasy, Jennifer M Fettweis, Jason Lloyd-Price, Anup Mahurkar, Wenyu Zhou, Gregory A Buck, Michael P Snyder, Jerome F Strauss 3rd, George M Weinstock, Owen White, Curtis Huttenhower

Abstract

The NIH Human Microbiome Project (HMP) has been carried out over ten years and two phases to provide resources, methods, and discoveries that link interactions between humans and their microbiomes to health-related outcomes. The recently completed second phase, the Integrative Human Microbiome Project, comprised studies of dynamic changes in the microbiome and host under three conditions: pregnancy and preterm birth; inflammatory bowel diseases; and stressors that affect individuals with prediabetes. The associated research begins to elucidate mechanisms of host-microbiome interactions under these conditions, provides unique data resources (at the HMP Data Coordination Center), and represents a paradigm for future multi-omic studies of the human microbiome.

Conflict of interest statement

M.S. is a cofounder of Personalis, Qbio, Sensomics, January, Filtricine and Akna and advisor for Genapsys. The other authors declare no competing interests.

Figures

Fig. 1. The first and second phases…
Fig. 1. The first and second phases of the NIH Human Microbiome Project.
The ten-year NIH Human Microbiome Project (HMP) program, organized into two phases (HMP1 and HMP2), developed reference sequences, multi-omic data sets, computational and statistical tools, and analytical and clinical protocols as resources for the broader research community. The HMP1 focused on the characterization of microbial communities from numerous body sites (oral, nasal, vaginal, gut, and skin) in a baseline study of healthy adult subjects, and included a set of demonstration projects that focused on specific diseases or disorders. The HMP2 expanded the repertoire of biological properties analysed for both host and microbiome in three longitudinal cohort studies of representative microbiome-associated conditions: pregnancy and preterm birth (vaginal microbiomes of pregnant women), inflammatory bowel diseases (gut microbiome) and prediabetes (gut and nasal microbiomes). These studies followed the dynamics of these conditions through multi-omic analyses of multiple measurement types over time, including changes in microbial community composition, viromics, metabolomic profiles, gene expression and protein profiles from both host and microbiome, and host-specific properties such as genetic, epigenomic, antibody, and cytokine profiles, along with other study-specific features. All sequences and multi-omic data, clinical information, and tools from both HMP1 and HMP2 are housed in the HMP Data Coordination Center (DCC) or referenced public or controlled-access repositories to serve as a central resource for the research community.
Fig. 2. The vaginal microbiome and its…
Fig. 2. The vaginal microbiome and its relationships with host factors in pregnancy and preterm birth.
The MOMS-PI project followed 1,527 pregnancies longitudinally and involved the collection of 206,437 biospecimens for analysis of host and microbial factors (16S amplicon, metagenomic, and metatranscriptomic sequencing; cytokine profiling; metabolomics; proteomics; genomics; and microbial isolate culture). Around 600 pregnancies were analysed in depth to assess features that lead to preterm birth; this analysis identified both host (for example, cytokine) and microbial (for example, ecological and specific strain) factors. As pregnancy progresses, with predictable changes in systemic oestradiol levels, the uterine and vaginal environments undergo various changes. The uterus switches from an early pro-inflammatory condition to an anti-inflammatory condition in the second trimester, and then back to a pro-inflammatory condition before the onset of labour. Meanwhile, specific changes in the microbiome of the vaginal lumen can be associated with preterm birth, possibly through mechanisms involving microorganisms travelling from the vagina to the uterus. The figure depicts an overview of longitudinal changes in the vaginal mucosal ecosystem and uterus during pregnancy.
Fig. 3. Host–microbiome dynamics in IBD.
Fig. 3. Host–microbiome dynamics in IBD.
The IBDMDB followed more than 100 participants with IBD (Crohn’s disease or ulcerative colitis), as well control individuals without IBD, for one year each to assess host and microbial molecular activity during changes in disease activity and gastrointestinal inflammation. Nearly all measured host–microbiome properties showed changes in either activity or stability during disease, including those shown here—not only microbial taxa and microbial transcription, but also host- and microbiota-derived small molecules in the gut, epithelial transcriptional responses at multiple points along the colon, and circulating antibody levels in peripheral blood serology.
Fig. 4. Differential host and microbial responses…
Fig. 4. Differential host and microbial responses to dietary perturbations and infectious disease in individuals with prediabetes.
For integrated personal ’omics profiling of the microbiome during prediabetes, 106 participants were followed for up to four years, with samples (primarily blood and stool) collected quarterly and additional samples collected during periods of RVI and other stresses. The genomes of the participants were sequenced and measurements of transcriptomes, proteomes, metabolomes, and microbiomes taken at each visit, in addition to clinical details. Insulin-resistant individuals showed differences from insulin-sensitive participants in various measurements, both at baseline and in response to the stresses such as weight loss and RVI.
Fig. 5. Resources from HMP1 and HMP2…
Fig. 5. Resources from HMP1 and HMP2 available at the DCC.
The HMP DCC (http://ihmpdcc.org) hosts raw and processed data from both phases of the Human Microbiome Project, comprising in total more than 42 Tb of multi-omic data. From the HMP1, these include 16S rRNA gene amplicons and metagenomes from the healthy human subjects (HHS) baseline cohort, as well as resources from demonstration projects (https://www.hmpdacc.org/health/projectdemos.php) and genomes of associated microbial isolates. From the HMP2, these include data that IRBs gave permission to be made publicly available from the pregnancy and preterm birth (MOMS-PI), inflammatory bowel disease (IBDMDB), and prediabetes (IPOP) projects, with links to raw data in other repositories. Additional data deposited elsewhere including microbial reference genomes, HMP1 human genomes, and controlled access data for all HMP2 projects is also linked from the DCC. Categories of data are colour coded, and the number of items in each dataset is indicated by the size of the circles.

References

    1. Eckburg PB, et al. Diversity of the human intestinal microbial flora. Science. 2005;308:1635–1638.
    1. Gill SR, et al. Metagenomic analysis of the human distal gut microbiome. Science. 2006;312:1355–1359.
    1. Costello EK, et al. Bacterial community variation in human body habitats across space and time. Science. 2009;326:1694–1697.
    1. Hsiao EY, et al. Microbiota modulate behavioral and physiological abnormalities associated with neurodevelopmental disorders. Cell. 2013;155:1451–1463.
    1. Gopalakrishnan V, et al. Gut microbiome modulates response to anti-PD-1 immunotherapy in melanoma patients. Science. 2018;359:97–103.
    1. Matson V, et al. The commensal microbiome is associated with anti-PD-1 efficacy in metastatic melanoma patients. Science. 2018;359:104–108.
    1. Routy B, et al. Gut microbiome influences efficacy of PD-1-based immunotherapy against epithelial tumors. Science. 2018;359:91–97.
    1. Haiser HJ, et al. Predicting and manipulating cardiac drug inactivation by the human gut bacterium Eggerthella lenta. Science. 2013;341:295–298.
    1. Taur Y, et al. The effects of intestinal tract bacterial diversity on mortality following allogeneic hematopoietic stem cell transplantation. Blood. 2014;124:1174–1182.
    1. Schloissnig S, et al. Genomic variation landscape of the human gut microbiome. Nature. 2013;493:45–50.
    1. Franzosa EA, et al. Identifying personal microbiomes using metagenomic codes. Proc. Natl Acad. Sci. USA. 2015;112:E2930–E2938.
    1. Dominguez-Bello MG, et al. Delivery mode shapes the acquisition and structure of the initial microbiota across multiple body habitats in newborns. Proc. Natl Acad. Sci. USA. 2010;107:11971–11975.
    1. Koenig JE, et al. Succession of microbial consortia in the developing infant gut microbiome. Proc. Natl Acad. Sci. USA. 2010;108:4578–4585.
    1. Vatanen T, et al. The human gut microbiome in early-onset type 1 diabetes from the TEDDY study. Nature. 2018;562:589–594.
    1. Yatsunenko T, et al. Human gut microbiome viewed across age and geography. Nature. 2012;486:222–227.
    1. Pasolli E, et al. Extensive unexplored human microbiome diversity revealed by over 150,000 genomes from metagenomes spanning age, geography, and lifestyle. Cell. 2019;176:649–662.e620.
    1. Faith JJ, et al. The long-term stability of the human gut microbiota. Science. 2013;341:1237439.
    1. Gajer P, et al. Temporal dynamics of the human vaginal microbiota. Sci. Transl. Med. 2012;4:132ra152.
    1. Gevers D, et al. The Human Microbiome Project: a community resource for the healthy human microbiome. PLoS Biol. 2012;10:e1001377.
    1. Turnbaugh PJ, et al. The Human Microbiome Project. Nature. 2007;449:804–810.
    1. Human Microbiome Project Consortium Structure, function and diversity of the healthy human microbiome. Nature. 2012;486:207–214.
    1. Human Microbiome Project Consortium A framework for human microbiome research. Nature. 2012;486:215–221.
    1. Lloyd-Price J, et al. Strains, functions and dynamics in the expanded Human Microbiome Project. Nature. 2017;550:61–66. doi: 10.1038/nature23889.
    1. Ravel J, et al. Vaginal microbiome of reproductive-age women. Proc. Natl Acad. Sci. USA. 2011;108:4680–4687.
    1. Fettweis JM, et al. Differences in vaginal microbiome in African American women versus women of European ancestry. Microbiology. 2014;160:2272–2282.
    1. Kong HH, et al. Temporal shifts in the skin microbiome associated with disease flares and treatment in children with atopic dermatitis. Genome Res. 2012;22:850–859.
    1. Alekseyenko AV, et al. Community differentiation of the cutaneous microbiota in psoriasis. Microbiome. 2013;1:31.
    1. Wylie KM, Mihindukulasuriya KA, Sodergren E, Weinstock GM, Storch GA. Sequence analysis of the human virome in febrile and afebrile children. PLoS One. 2012;7:e27735.
    1. Lewis JD, et al. Inflammation, antibiotics, and diet as environmental stressors of the gut microbiome in pediatric Crohn’s disease. Cell Host Microbe. 2015;18:489–500.
    1. Zupancic ML, et al. Analysis of the gut microbiota in the old order Amish and its relation to the metabolic syndrome. PLoS One. 2012;7:e43052.
    1. Erickson AR, et al. Integrated metagenomics/metaproteomics reveals human host-microbiota signatures of Crohn’s disease. PLoS One. 2012;7:e49138.
    1. Frank DN, et al. Disease phenotype and genotype are associated with shifts in intestinal-associated microbiota in inflammatory bowel diseases. Inflamm. Bowel Dis. 2011;17:179–184.
    1. La Rosa PS, et al. Patterned progression of bacterial populations in the premature infant gut. Proc. Natl Acad. Sci. USA. 2014;111:12522–12527.
    1. Fodor AA, et al. The “most wanted” taxa from the human microbiome for whole genome sequencing. PLoS One. 2012;7:e41294.
    1. Nelson KE, et al. A catalog of reference genomes from the human microbiome. Science. 2010;328:994–999.
    1. Wylie KM, et al. Novel bacterial taxa in the human microbiome. PLoS One. 2012;7:e35294.
    1. Li K, Bihan M, Yooseph S, Methé BA. Analyses of the microbial diversity across the human microbiome. PLoS One. 2012;7:e32118.
    1. Aagaard K, et al. The Human Microbiome Project strategy for comprehensive sampling of the human microbiome and why it matters. FASEB J. 2013;27:1012–1022.
    1. Schloss PD, Gevers D, Westcott SL. Reducing the effects of PCR amplification and sequencing artifacts on 16S rRNA-based studies. PLoS One. 2011;6:e27310.
    1. Jumpstart Consortium Human Microbiome Project Data Generation Working Group Evaluation of 16S rDNA-based community profiling for human microbiome research. PLoS One. 2012;7:e39315.
    1. Gevers D, Pop M, Schloss PD, Huttenhower C. Bioinformatics for the Human Microbiome Project. PLOS Comput. Biol. 2012;8:e1002779.
    1. Huse SM, Ye Y, Zhou Y, Fodor AA. A core human microbiome as viewed through 16S rRNA sequence clusters. PLoS One. 2012;7:e34242.
    1. Markowitz VM, et al. IMG/M-HMP: a metagenome comparative analysis system for the Human Microbiome Project. PLoS One. 2012;7:e40151.
    1. Cantarel BL, Lombard V, Henrissat B. Complex carbohydrate utilization by the healthy human microbiome. PLoS One. 2012;7:e28742.
    1. Rho M, Wu YW, Tang H, Doak TG, Ye Y. Diverse CRISPRs evolving in human microbiomes. PLoS Genet. 2012;8:e1002441.
    1. Faust K, et al. Microbial co-occurrence relationships in the human microbiome. PLOS Comput. Biol. 2012;8:e1002606.
    1. Abubucker S, et al. Metabolic reconstruction for metagenomic data and its application to the human microbiome. PLOS Comput. Biol. 2012;8:e1002358.
    1. Integrative HMP (iHMP) Research Network Consortium The Integrative Human Microbiome Project: dynamic analysis of microbiome-host omics profiles during periods of human health and disease. Cell Host Microbe. 2014;16:276–289.
    1. Lloyd-Price, J. et al. Multi-omics of the gut microbial ecosystem in inflammatory bowel diseases. Nature10.1038/s41586-019-1237-9 (2019). HMP2 Inflammatory Bowel Disease Multi’omics Database (IBDMDB) study profiling 2,965 stool, biopsy, and blood specimens from 132 individuals with Crohn’s disease, ulcerative colitis or neither (control subjects) followed longitudinally for one year each; multi-omic profiling identified taxa, expressed functions, metabolite pools, and host gene expression that were disrupted in the gut during increased disease activity, as well as systemic effects such as serum antibody levels.
    1. Fettweis, J. M. et al. The vaginal microbiome and preterm birth. Nat. Med. 10.1038/s41591-019-0450-2 (2019). HMP2 preterm birth study of 1,527 pregnancies focusing on 45 spontaneous preterm birth events and 90 case-matched controls, together yielding over 12,000 multi-omically profiled samples; 16S rRNA gene, metagenomic, metatranscriptomic and cytokine profiles identified differential levels ofLactobacillus crispatus, BVAB1, Sneathia amnii, TM7-H1, and additional taxa associated with preterm deliveries.
    1. Zhou, W. et al. Longitudinal multi-omics of host–microbe dynamics in prediabetes. Nature10.1038/s41586-019-1236-x (2019). HMP2 prediabetes study of 106 participants spanning 1,092 visits, profiling host transcriptomes, metabolomes, cytokines, and proteomes, as well as microbial taxonomic and genomic changes during healthy periods, respiratory viral infections, immunizations, and other perturbations; most molecular profiles were personalized, perturbed during infections and immunizations, and associated with changes in insulin sensitivity.
    1. Behrman, R. E. & Butler A. S. (eds.) Preterm Birth: Causes, Consequences, and Prevention (National Academies Press, 2007).
    1. York TP, Eaves LJ, Neale MC, Strauss JF., III The contribution of genetic and environmental factors to the duration of pregnancy. Am. J. Obstet. Gynecol. 2014;210:398–405.
    1. Liu L, et al. Global, regional, and national causes of under-5 mortality in 2000–15: an updated systematic analysis with implications for the Sustainable Development Goals. Lancet. 2016;388:3027–3035.
    1. Mor G, Aldo P, Alvero AB. The unique immunological and microbial aspects of pregnancy. Nat. Rev. Immunol. 2017;17:469–482.
    1. Romero R, Dey SK, Fisher SJ. Preterm labor: one syndrome, many causes. Science. 2014;345:760–765.
    1. Jefferson KK. The bacterial etiology of preterm birth. Adv. Appl. Microbiol. 2012;80:1–22.
    1. Serrano, M. G. et al. Racioethnic diversity in the dynamics of the vaginal microbiome during pregnancy. Nat. Med. 10.1038/s41591-019-0465-8 (2019). HMP2 term birth study of 613 pregnant women, 1,969 non-pregnant women, and an additional 90 pregnant women followed longitudinally profiling vaginal, buccal, blood, urine, and rectal samples; shifts towardLactobacillus-dominated communities tended to occur early during pregnancy, particularly in women of African or Hispanic descent.
    1. DiGiulio DB, et al. Temporal and spatial variation of the human microbiota during pregnancy. Proc. Natl Acad. Sci. USA. 2015;112:11060–11065.
    1. Callahan BJ, et al. Replication and refinement of a vaginal microbial signature of preterm birth in two racially distinct cohorts of US women. Proc. Natl Acad. Sci. USA. 2017;114:9966–9971.
    1. Kindinger LM, et al. The interaction between vaginal microbiota, cervical length, and vaginal progesterone treatment for preterm birth risk. Microbiome. 2017;5:6.
    1. Brown RG, et al. Vaginal dysbiosis increases risk of preterm fetal membrane rupture, neonatal sepsis and is exacerbated by erythromycin. BMC Med. 2018;16:9.
    1. Jefferson, K. K. et al. Relationship between vitamin D status and the vaginal microbiome during pregnancy. J. Perinatol. 10.1038/s41372-019-0343-8 (2019).
    1. Zhou SS, Tao YH, Huang K, Zhu BB, Tao FB. Vitamin D and risk of preterm birth: Up-to-date meta-analysis of randomized controlled trials and observational studies. J. Obstet. Gynaecol. Res. 2017;43:247–256.
    1. Huang H, et al. Fine-mapping inflammatory bowel disease loci to single-variant resolution. Nature. 2017;547:173–178.
    1. Manichanh C, et al. Reduced diversity of faecal microbiota in Crohn's disease revealed by a metagenomic approach. Gut. 2005;55:205–211.
    1. Frank DN, et al. Molecular-phylogenetic characterization of microbial community imbalances in human inflammatory bowel diseases. Proc. Natl Acad. Sci. USA. 2007;104:13780–13785.
    1. Kugathasan S, et al. Prediction of complicated disease course for children newly diagnosed with Crohn’s disease: a multicentre inception cohort study. Lancet. 2017;389:1710–1718.
    1. Narula N, et al. Systematic review and meta-analysis: fecal microbiota transplantation for treatment of active ulcerative colitis. Inflamm. Bowel Dis. 2017;23:1702–1709.
    1. Jeon SR, Chai J, Kim C, Lee CH. Current evidence for the management of inflammatory bowel diseases using fecal microbiota transplantation. Curr. Infect. Dis. Rep. 2018;20:21.
    1. Morgan XC, et al. Dysfunction of the intestinal microbiome in inflammatory bowel disease and treatment. Genome Biol. 2012;13:R79.
    1. Hall AB, et al. A novel Ruminococcus gnavus clade enriched in inflammatory bowel disease patients. Genome Med. 2017;9:103.
    1. Schirmer M, et al. Dynamics of metatranscription in the inflammatory bowel disease gut microbiome. Nat. Microbiol. 2018;3:337–346.
    1. Franzosa EA, et al. Gut microbiome structure and metabolic activity in inflammatory bowel disease. Nat. Microbiol. 2019;4:293–305.
    1. Mallick, H. et al. Predictive metabolomic profiling of microbial communities using amplicon or metagenomic sequences. Nat. Commun. (in the press).
    1. Franzosa EA, et al. Species-level functional profiling of metagenomes and metatranscriptomes. Nat. Methods. 2018;15:962–968.
    1. McIver LJ, et al. bioBakery: a meta’omic analysis environment. Bioinformatics. 2018;34:1235–1237.
    1. Willing BP, et al. A pyrosequencing study in twins shows that gastrointestinal microbial profiles vary with inflammatory bowel disease phenotypes. Gastroenterology. 2010;139:1844–1854.e1841.
    1. Gevers D, et al. The treatment-naive microbiome in new-onset Crohn’s disease. Cell Host Microbe. 2014;15:382–392.
    1. Cowie CC, et al. Full accounting of diabetes and pre-diabetes in the U.S. population in 1988–1994 and 2005–2006. Diabetes Care. 2009;32:287–294.
    1. Pickup JC. Inflammation and activated innate immunity in the pathogenesis of type 2 diabetes. Diabetes Care. 2004;27:813–823.
    1. Qin J, et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature. 2012;490:55–60.
    1. Gamble DR, Kinsley ML, FitzGerald MG, Bolton R, Taylor KW. Viral antibodies in diabetes mellitus. BMJ. 1969;3:627–630.
    1. Mehta SH, et al. Prevalence of type 2 diabetes mellitus among persons with hepatitis C virus infection in the United States. Ann. Intern. Med. 2000;133:592–599.
    1. Chen R, et al. Personal omics profiling reveals dynamic molecular and medical phenotypes. Cell. 2012;148:1293–1307.
    1. Piening BD, et al. Integrative personal omics profiles during periods of weight gain and loss. Cell Syst. 2018;6:157–170 e158.
    1. Schüssler-Fiorenza Rose, S. et al. A longitudinal big data approach for precision health. Nat. Med. 25, 792–804 (2019). HMP2 Integrative Personalized ’Omics Profiling of 109 individuals enriched for risk of type 2 diabetes studied genomically, immunologically, transcriptomically, proteomically, metabolomically, and using wearable monitoring; multiple molecular pathways were associated with metabolic, cardiovascular and oncological pathophysiology, and a subset were predictive of insulin resistance.
    1. Wilkinson MD, et al. The FAIR Guiding Principles for scientific data management and stewardship. Sci. Data. 2016;3:160018.
    1. NIH Human Microbiome Portfolio Analysis Team A review of 10 years of human microbiome research activities at the US National Institutes of Health, Fiscal Years 2007–2016. Microbiome. 2019;7:31.

Source: PubMed

3
Prenumerera