• No results found

University of Groningen Environment-host-microbe interactions shape human metabolism Chen, Lianmin

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Environment-host-microbe interactions shape human metabolism Chen, Lianmin"

Copied!
15
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

University of Groningen

Environment-host-microbe interactions shape human metabolism

Chen, Lianmin

DOI:

10.33612/diss.171839167

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from

it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date:

2021

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

Chen, L. (2021). Environment-host-microbe interactions shape human metabolism. University of

Groningen. https://doi.org/10.33612/diss.171839167

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

Chapter 1

Introduction

The introduction is partially based on the review paper:

A system biology perspective on environment-host-microbe interactions

Lianmin Chen, Sanzhima Garmaeva, Alexandra Zhernakova, Jingyuan Fu &

Cisca Wijmenga.

(3)

Complex diseases such as cardiovascular disease (CVD), obesity,

inflammatory bowel disease (IBD), kidney disease, type 2 diabetes (T2D)

and cancers have become a major burden to public health and affect more than 20% of the population worldwide. The aetiology of complex diseases is not yet clear, but they are traditionally thought to be caused by genetics and environmental factors (e.g. dietary habits), and by their interactions. Following the completion of human genome sequencing in 2003, numerous genome-wide association studies (GWAS) have generated a wealth of information on the genetic architecture of complex diseases, which has advanced the development of genomics-based medicine. While the success of GWAS has led to the development of “genomic medicine”, it has become increasingly clear that genetics can only explain a limited proportion of an individual’s risk of developing a complex, non-infectious disease. For instance, the heritability for T2D and Crohn’s disease are estimated at

about 6% and 20% 1, respectively, indicating that a large proportion of the

phenotypic variation is explained by environmental or other unidentified factors. In the past few years, the impact of the gut microbiome on the development of complex human diseases has increasingly been recognized. The human intestines are colonized by a vast number of bacteria, archaea, microbial eukaryotes and viruses that are as abundant as our somatic cells and are collectively known as the gut microbiome.

The microbes in our body are relevant for our health and disease

Human bodies harbour a diverse community of microbes that together compose the human microbiome. With the aid of two culture-independent next generation sequencing technologies, 16s rRNA sequencing and shotgun metagenomics sequencing (Box 1), our knowledge of the composition and functional properties of the human microbiome has been increasing rapidly over the past several decades. The Human Microbiome Project, completed in 2013, and other related projects have now characterized the composition of the human microbiome at 18 different body sites, characterized the inter-individual variation in the microbiome, provided reference genomes for nearly 3,000 microbial strains and constructed a catalogue for ~10 million

microbial genes 2. This work has provided a great reference source for human

microbial research.

The majority of human-inhabiting microbes are found in the human gut. This community, known as the gut microbiome, contains up to a 1000 different

species and can weigh as much as 200 grams in adults 3,4. The number of

bacterial cells in the gut microbiome is similar to the number of human cells

in the body, yet it carries 100 times more genes than the human genome 5.

Given its high diversity in composition and function, the gut microbiome

(4)

against xenobiotic toxins 8, while also strengthening our immune response

9. In line with this, a large body of evidence now supports a role for the gut

microbiome in our predisposition for various diseases, including obesity 10,

T2D 11, CVD 12, IBD 7,13, cancers 14,15, depression 16 and Parkinson disease

17. Moreover, the gut microbiome can also determine individual response to

diet 7 and medication 18. Given these roles, the gut microbiome has become

known as “the second human genome”. However, in comparison to the human genome, the gut microbiome can be modulated by various means, and this has led to it being considered an important player in personalized medicine. The human gut microbiome is thus emerging as an attractive therapeutic target for disease prevention and treatment.

Box 1. Terminology in human microbiome research

Microbiome:the collection of all genomes of microbes in an ecosystem.

16S rRNA sequencing: an analytical method for characterizing the microbiome based on sequencing the 16S rRNA gene of bacteria and archaea. Shotgun metagenomics sequencing: an analytical method for characterizing the microbiome based on sequencing of all DNA fragments.

Homeostasis: stability of the microbiome maintained via internal mechanisms of self-regulation that is resilient to external perturbation. Dysbiosis: imbalance or maladaptation of the microbiome.

Diversity: variety and variability of a microbial community. The most common measures used to characterize these features are alpha diversity and beta diversity.

Alpha diversity:a measure of the diversity of species or other taxa within a sample. Widely used alpha diversity measures include species richness, the number of taxa present in a sample and the entropy-based Shannon and Simpson indices.

Beta diversity: a measure that describes the difference in taxonomic composition between samples, which can be represented as a square distance matrix. Commonly used beta diversity measures are UniFrac distance and Bray-Curtis dissimilarity.

The gut microbiome is a complex trait

To study the potential of microbiome-modulating approaches in personalized medicine, we first need to bear in mind that the gut microbiome is itself a complex trait that can be affected by host genetics and exogenous factors,

(5)

and by their interactions. The colonization of the human gut begins at birth, after which it is shaped to become mature around three years of age, when its composition and diversity are similar to those found in adults. During the course of human life, gut microbiome composition can be affected by many perturbation factors and its status can change from homeostasis to dysbiosis through ageing or the development of disease (Figure 1). In the general population, up to 20% of the inter-individual variation in the gut microbiome

can be explained by various intrinsic and exogenous factors 19-22. It is also

consistently observed in humans and mice that environmental factors

dominate over host genetics in shaping the gut microbiome 19,21,23.

Figure 1. Development and dynamic changes of the gut microbiome over the course of human life. The colonization of the human gut begins at birth. It is then rapidly shaped over the first years of life to reach a mature state at the age of three, when its composition and diversity is close to that of an adult. Over the course of life, gut microbiome composition can be affected by many perturbation factors, including diet, medication use, lifestyle and host physiological status. The gut microbiome can thus show high inter-individual variation. Over a lifetime, the microbiome can preserve its homeostasis, but dysbiosis can also occur with ageing or during the development of disease, when diversity is significantly decreased.

Diet is one dominant environmental factor affecting the gut microbiome

23,24. The gut microbiome of vegans, for example, is rather different from that

of people with an omnivorous diet 20, and a western diet (i.e. one with high

calorie and high fat intake) is associated with a less diverse microbial ecology

than a diet with high fibre intake 7. Higher consumption of fruit, vegetables,

fibre and red wine has been linked to higher abundances of beneficial

Early development Homeostasis Dysbiosis

Div ersit y low high perturbation factors Microbial status Host status

(6)

bacteria including the butyrate-producing Faecalibacterium prausnitzii,

anti-inflammatory Clostridiales and mucin-degrading Akkermansia muciniphila

7,21. The impact of diet on the gut microbiome suggests that dietary invention

could be a means of improving microbial composition to improve human health. One recent dietary intervention study showed that a high-fibre diet can promote short-chain fatty acid (SCFA)-producing bacterial species, and these species sequentially exerted a beneficial effect on their host by lowering haemoglobin A1c level and diminishing metabolically detrimental

compounds, thereby alleviating T2D 25. These SCFA-producers can also be

promoted by metformin, a common drug in the treatment of T2D 11,26. Many

other prescribed medications have also been shown to affect gut microbiome

composition 7,19,21,27,28, including antibiotics 29-31, proton pump inhibitors

6,7, statins and laxatives 11,21,32.

The impact of host genetics on the gut microbiome is also emerging. Twin studies have estimated the heritability of individual bacteria and microbial

pathways in humans 22,33 and identified a proportion of gut bacteria

that are substantially heritable. The highest estimated heritability for a given species is up to 0.4, which is comparable to the heritability of many common diseases. Interestingly, several heritable taxa and pathways are also associated with complex traits. For instance, the most heritable bacteria

taxa (Christensenellaceae, Archaea, Tenericutes and Bifidobacteriaceae)

are associated to traits including body mass index (BMI) and bacterial biosynthesis of branched-chain amino acids, which are both also linked to

insulin resistance 34. These observations strongly suggest that host microbe

interactions have a role in the development of complex traits and diseases. Several GWAS of >1000 samples had identified genetic loci associated to

microbial diversity, species abundance and bacterial pathways 35-37. Although

there was limited overlap in the associated loci due to the heterogeneity

of the statistical methods used and the relatively small samples sizes 38,

the identified loci converged toward several common processes, including innate immunity, metabolism and food processing. In particular, consistent

associations were found for C-type lectin genes in independent human 35,36

and animal 39-41 studies and for the lactase gene (LCT locus) that affects

the abundance of milk fermenting Bifidobacteria 35,37,42, an association

found to be dependent on the consumption of milk products 35. To increase

the statistical power to discover more of these relationships, the MiBioGen consortium has now been established to analyse the genetics of microbiome in >19,000 subjects from 18 participating groups using a harmonised

methodology and analytical pipeline 43,44.

In addition to microbial interactions with diet and host genetics, the gut microbiome is an ecosystem in which microbes can exchange or compete for nutrients, signalling molecules, or immune-evasion mechanisms through

(7)

Enthusiasm has thus been rising to decipher these microbial interactions in

order to detect key microbes in health and disease 48,49. One way of doing

this is to create co-abundance networks based on correlations, a method that has the potential to study interactions between microbes and thereby to

generate hypotheses for experimental validation 48,49. Microbial taxonomic

interactions have recently been inferred in healthy individuals and in individuals with extreme longevity, gestational diabetes, Crohn’s disease

and colorectal cancer 50-55. These studies have identified microbial genera

that are potentially key in health and disease, e.g. Porphyromonas and

Bacteroides in gestational diabetes 53.

Environment-genetics-microbiome interactions in complex diseases

Increasing evidence of the impact of genetic and environmental factors on the gut microbiome has led to a paradigm shift in our perspective on the complexity of the development of complex diseases. The typical view of complex diseases was that these disorders resulted from multiple genetic factors and their interaction with environmental factors. Now, the gut microbiome has been included as a third factor that can affect susceptibility to complex diseases via its interaction with genetics and environment (Figure 2A). Understanding the causal role of the microbiome in this complex interaction is essential for the development of microbiome-targeting therapy for the prevention and treatment of complex diseases. Such complex interactions can fall into three global scenarios: an additive model, a mediator model, or an interaction model.

Figure 2. Complex environment-genetics-microbiome interactions in complex diseases. A. Genetics, environment, gut microbiome and their interactions can contribute to individual susceptibility to complex diseases. B. Additive model that assumes genetics, environment and the gut microbiome exert independent and additive effects on the susceptibility to complex diseases. C. Mediator model in which the gut microbiome mediates the effects of genetics and environment on complex disease susceptibility. D. Interaction model in which the impact of genetics and environmental factors on a complex disease may not occur via their direct effects on the gut microbiota but may depend on the gut microbiome and their interactions. In an additive model, the gut microbiome exerts an additive effect on the susceptibility to complex diseases, i.e. in addition to the known genetic and environmental factors (Figure 2B). The gut microbiome can explain extra inter-individual variation of a trait, suggesting that microbiome-targeting approaches may have a better control on a complex trait on top of other approaches of modulating genetic and environmental effects. For example,

(8)

one population-based microbiome study showed that gut microbiota explains an extra 4.5-6% of the variance in BMI and blood lipid levels and an additive model with age, sex and genetic risk factors can explain up to 25.9% of the

variation in HDL cholesterol level 56. Similar findings have been observed in

other studies in mouse and humans. In mice, the combined and integrated effects of diet, host background and gut microbiome drive dynamic changes

in faecal and plasma metabolites 24. In Israeli and Dutch human cohorts, the

gut microbiome and host genetics have been shown to be largely independent and a combination of both factors had a higher power to predict host

phenotypes 19.

In a mediator model, the gut microbiome mediates the effects of genetic and environmental factors on complex diseases (Figure 2C). Here, adding the gut microbiome to the prediction model may not explain extra inter-individual variation, but microbiome-targeting approaches can alleviate the impact of genetics and environmental factors on the susceptibility of complex diseases. This kind of causal model can be tested by transferring genetic- or diet-modulated faecal microbiomes from donors to receivers and assessing whether the corresponding phenotype was transmitted or not. Using this approach, the mediating role of the gut microbiome has been reported for the

increased susceptibility to NLRP3 inflammasome deficiency in non-alcoholic

steatohepatitis 57, the therapeutic effect of metformin in the treatment of

T2D and the protective effects of dietary capsaicin against obesity-associated

chronic low-grade inflammation 58.

In an interaction model, the impact of genetics and environmental factors

Genetics Environment Microbiome Complex diseases Genetics Environment Microbiome Complex diseases Genetics Environment Microbiome Complex diseases Genetics Environment Microbiome Complex diseases A B C D

(9)

on a complex disease will depend on the gut microbiome and on the interactions between all three (Figure 2D). Adding an interaction term with the gut microbiome to a prediction model could explain more inter-individual variation and improve power for phenotype prediction. Moreover, modulating the gut microbiome, or its products, could alleviate or enhance the genetic or environmental effects. For instance, individual response to PD-1/PD-L1 blockers or anti-CTLA4 immunotherapy have been found to

be dependent on the gut microbiome 14,15, very likely through its anabolic

functions, which can enhance systemic and anti-tumour immune response by increasing antigen presentation and improving effector T cell function. Moreover, the genetic susceptibility for IBD at the autophagy-related 16–like 1 (ATG16L1) and nucleotide-binding oligomerization domain–containing

protein 2 (NOD2) genes requires microbial triggers, e.g. the microbial

secreted outer membrane vesicles of the human commensal Bacteroides

fragilis 59, illustrating the importance of genetic-microbiome interaction in

the pathogenesis of IBD.

Metabolites are at the centre of environment-genetics microbiome interactions

Environment-genetics-microbiome interaction effects on complex disease are largely mediated through metabolites. There are thousands of metabolites within the human body, some of which are endogenously produced, e.g. bile acids and lipids, and some that are taken up from the environment, e.g. vitamins and SCFAs. Many of these molecules can affect our health by modulating processes like metabolism and immune responses that provide useful indicators of our health status. However, our knowledge about the origin of most of these molecules and why they vary in concentration between individuals is still very limited. Potential determinants of these molecules include, but are not limited to, the environment (e.g. diet), genetic variants and the gut microbiome. GWAS have identified over 150 loci related

to abnormal lipid levels, explaining around 40% of the total variation 60. Part

of the unexplained variance has been attributed to environmental factors

including diet, as foods act as a direct source of lipids 61,62. Other factors

that influence lipid metabolism include the gut microbiome 56, which plays

an important role in the digestion of different dietary components including fats and polysaccharides. Furthermore, genetics, environment and the gut microbiome can also interact with each other in metabolite regulation, as is the case for trimetlylamine oxide (TMAO). This molecule, which promotes the artery-narrowing disease atherosclerosis, is generated as a result of the metabolism - by both microbes and their host - of certain dietary compounds

that are abundant in red meat 63. However, the key determinants and

(10)

molecules that directly affect human health, investigating their determinants and how environment-genetics-microbiome interactions regulate them may help us to understand the aetiology of complex diseases and yield new clinical strategies for complex disease prevention and treatment.

Research aims and outline of this thesis

The overall aim of the studies presented in this thesis is to obtain a mechanistic understanding of the potential roles of the gut microbiome, in interaction with host genetics, environmental factors and with itself, to human metabolic health. For this purpose, I have made use of various layers of “omics” datasets (metagenomics, metabolomics and genetics) in combination with the extensive phenotypic information that has been generated for a unique series of population-based prospective cohorts and patient cohorts.

In Chapter 1, I introduce the state of knowledge regarding the roles of the gut microbiome and its interactions with host genetics and environmental factors in human health and disease at the onset of the project. We then realized that description of differential microbial taxonomies between healthy subjects and patients failed to capture the functional potential of the gut microbiome. In addition, the gut microbiome itself is a complex trait that can interact with host genetics, various exogenous factors (including diet components and the use of certain drugs) and interactions between microbes. Thus, we pinpointed the importance of integrating multiple layers of functional omics information generated for several prospective human cohorts in order to understand how the gut microbiome interacts with the host, e.g. which signalling molecules are involved and how environmental factors modulate this interaction in human health and disease. With this purpose in mind, the five research chapters were designed to investigate the functional roles of the gut microbiome in humans, particularly in relation to CVD, obesity and its co-morbidities, and IBD.

Chapter 2 assesses microbial contributions to plasma lipid concentrations and composition in relation to CVD. Altered gut microbial composition has been linked to CVD, but its functional links to lipid metabolism of the host in relation to CVD development have remained unclear. To systematically assess functional links between the microbiome and plasma lipid profiles, we determined metagenomics-based microbial associations between more than 200 plasma lipidomic traits and microbial species and pathways in the population-based Lifelines-DEEP cohort and a clinical obesity cohort (300OB). This study represents the largest metagenome-based association study on plasma lipids and microbiome relevant to CVD risk and cardiometabolic phenotypes in humans to date. This work identified

(11)

novel bacterial species and pathways that associate to specific lipoprotein subclasses and revealed functional links between the gut microbiome and cardiovascular health of the host.

Chapter 3 goes beyond microbial composition-lipid associations by linking bile acids (BAs) to host lipid metabolism. BAs are amphipathic steroids that are exclusively synthesized in the liver (primary BAs) that can be structurally modified by the gut microbes (secondary BAs) and have been recognized as signalling molecules that act in the aetiologies of several components of metabolic syndrome, including dyslipidaemia. We profiled plasma and faecal BAs in the 300OB cohort and linked BAs with liver fat and plasma lipid parameters. We also characterized potential genetic and microbial determinants of BAs. Our study reveals novel genetic and microbial determinants of BAs in obesity and their relationship to disease-relevant lipid parameters that are important for the design of personalized therapies targeting BA signalling pathways.

Chapter 4 extends insights in microbial impact on plasma metabolite levels to various categories of (biologically-active) compounds generated by untargeted metabolomics platforms to allow for a broader understanding of the functional roles of the microbiome in host metabolic health. We generated profiles of 1,183 plasma metabolites from 1,368 Lifelines-DEEP participants for whom there is extensive information about dietary habits, gut microbiome and genetic background. We assessed metabolite associations to diet, genetics and the gut microbiome and evaluated the relative importance of diet, genetics and the gut microbiome to inter-individual metabolite variations. Additionally, we used Mendelian randomization and mediation

analysis to infer in silico causal relationships in diet-genetics-microbiome

interactions and putative mechanisms underlying complex diseases. Moreover, we assessed the potential of using diet, genetics and microbiome to predict an individual’s future metabolic health using a machine learning algorithm. Our results provide a systematic underpinning of the importance of diet-microbiome-genetics interactions in human metabolism that may act as a basis for developing strategies for the prevention or treatment of metabolic disorders.

Chapter 5 moves forward from cross-sectional to longitudinal analyses to assess whether alterations of the gut microbiome are related to changes in host health status. We present a long-term follow-up analysis of the gut microbiome in 338 participants of the Lifelines-DEEP cohort in which we compare microbiome composition in faecal samples taken 4 years apart. We characterized microbial stability and variations in gut microbial composition and genetic makeup. For temporally stable and individual-specific microbial features, we developed a method to use these microbial features as a “fingerprint” to distinguish samples taken from the same individual.

(12)

For microbial features showing large temporal variations, we linked their temporal variation to changes in the host’s clinical phenotypes and lifestyle. To gain further biological insights, we profiled plasma levels of 1,183

metabolites at both time points and used mediation analysis as an in silico

method to infer whether specific metabolites mediate the microbial impact on host health. Finally, we assessed the changes in antibiotic resistance and virulence factors in the human gut microbiome. This study highlights the importance of longitudinal sampling and of integrating complementary multi-omics data to identify functional mechanisms that can serve as therapeutic targets for human metabolic disease.

Chapter 6 delves into microbial interactions and compares differential microbial co-abundances between healthy subjects and those with specific diseases. Although the gut microbiome is an ecosystem that involves complex interactions, our current knowledge of the potential roles of the gut microbiome in health and disease relies mainly on description of differential microbial abundances. However, little is known about the role of microbial interactions in the context of human disease. Here, we constructed and compared microbial co-abundance networks using 2,379 metagenomes from four human cohorts: an inflammatory bowel disease (1000IBD) cohort, an obese cohort (300OB) and two population-based cohorts (Lifelines-DEEP and 500FG). We compared the microbial taxonomic and functional networks under different host health conditions and, by doing so, identified potential key species and pathways that shape host-associated microbial networks. Our study provides evidence that altered microbial abundances in a specific disease can reflect their co-abundance relationship, which expands our current knowledge regarding microbial dysbiosis in disease.

Finally, Chapter 7 summarizes the main findings of this thesis and

provides my personal perspectives on how the booming field of “microbiome functional omics” should move forward.

References

1 Manolio, T. A. et al. Finding the missing heritability of complex diseases. Nature 461, 747-753, doi:10.1038/nature08494 (2009).

2 Sonnenburg, E. D. et al. Diet-induced extinctions in the gut microbiota compound over generations. Nature 529, 212-215, doi:10.1038/nature16504 (2016).

3 Holmes, E., Li, J. V., Athanasiou, T., Ashrafian, H. & Nicholson, J. K. Understanding the role of gut microbiome-host metabolic signal disruption in health and disease. Trends Microbiol 19, 349-359, doi:10.1016/j.tim.2011.05.006 (2011).

4 Sender, R., Fuchs, S. & Milo, R. Revised Estimates for the Number of Human and Bacteria Cells in the Body. PLoS Biol 14, e1002533, doi:10.1371/journal.pbio.1002533 (2016).

(13)

sequencing. Nature 464, 59-65, doi:10.1038/nature08821 (2010).

6 Imhann, F. et al. Proton pump inhibitors affect the gut microbiome. Gut 65, 740-748, doi:10.1136/gutjnl-2015-310376 (2016).

7 Zhernakova, A. et al. Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity. Science 352, 565-569, doi:10.1126/science. aad3369 (2016).

8 Mirzaei, M. K. & Maurice, C. F. Ménage à trois in the human gut: interactions between host, bacteria and phages. Nature Reviews Microbiology 15, 397 (2017).

9 Schirmer, M. et al. Linking the Human Gut Microbiome to Inflammatory Cytokine Production Capacity. Cell 167, 1125-1136 e1128, doi:10.1016/j.cell.2016.10.020 (2016). 10 Li, G. et al. Intermittent Fasting Promotes White Adipose Browning and Decreases Obesity by Shaping the Gut Microbiota. Cell Metab 26, 672-685 e674, doi:10.1016/ j.cmet.2017.08.019 (2017).

11 Wu, H. et al. Metformin alters the gut microbiome of individuals with treatment- naive type 2 diabetes, contributing to the therapeutic effects of the drug. Nat Med 23, 850-858, doi:10.1038/nm.4345 (2017).

12 Atanasovska, B., Kumar, V., Fu, J., Wijmenga, C. & Hofker, M. H. GWAS as a Driver of Gene Discovery in Cardiometabolic Diseases. Trends Endocrinol Metab 26, 722-732, doi:10.1016/j.tem.2015.10.004 (2015).

13 Vazquez-Baeza, Y. et al. Guiding longitudinal sampling in IBD cohorts. Gut, doi:10.1136/gutjnl-2017-315352 (2017).

14 Gopalakrishnan, V. et al. Gut microbiome modulates response to anti–PD-1 immunotherapy in melanoma patients. Science, eaan4236 (2017).

15 Routy, B. et al. Gut microbiome influences efficacy of PD-1–based immunotherapy against epithelial tumors. Science, eaan3706 (2017).

16 Stevens, B. R. et al. Increased human intestinal barrier permeability plasma biomarkers zonulin and FABP2 correlated with plasma LPS and altered gut microbiome in anxiety or depression. Gut, doi:10.1136/gutjnl-2017-314759 (2017).

17 Tremlett, H., Bauer, K. C., Appel-Cresswell, S., Finlay, B. B. & Waubant, E. The gut microbiome in human neurological disease: A review. Ann Neurol 81, 369-382, doi:10.1002/ana.24901 (2017).

18 Bauer, P. V. et al. Metformin Alters Upper Small Intestinal Microbiota that Impact a Glucose-SGLT1-Sensing Glucoregulatory Pathway. Cell Metab, doi:10.1016/ j.cmet.2017.09.019 (2017).

19 Rothschild, D. et al. Environment dominates over host genetics in shaping human gut microbiota. Nature, doi:10.1038/nature25973 (2018).

20 Faith, J. J. et al. The long-term stability of the human gut microbiota. Science 341, 1-8, doi:10.1126/science.1237439 (2013).

21 Falony, G. et al. Population-level analysis of gut microbiome variation. Science 352, 560-564, doi:10.1126/science.aad3503 (2016).

22 Xie, H. et al. Shotgun Metagenomics of 250 Adult Twins Reveals Genetic and Environmental Impacts on the Gut Microbiome. Cell Syst 3, 572-584 e573, doi:10.1016/ j.cels.2016.10.004 (2016).

23 Carmody, R. N. et al. Diet dominates host genotype in shaping the murine gut microbiota. Cell Host Microbe 17, 72-84, doi:10.1016/j.chom.2014.11.010 (2015).

24 Fujisaka, S. et al. Diet, Genetics, and the Gut Microbiome Drive Dynamic Changes in Plasma Metabolites. Cell Rep 22, 3072-3086, doi:10.1016/j.celrep.2018.02.060 (2018).

(14)

25 Zhao, L. et al. Gut bacteria selectively promoted by dietary fibers alleviate type 2 diabetes. Science 359, 1151-1156 (2018).

26 Forslund, K. et al. Disentangling type 2 diabetes and metformin treatment signatures in the human gut microbiota. Nature 528, 262-266, doi:10.1038/nature15766 (2015). 27 Luca, F., Kupfer, S. S., Knights, D., Khoruts, A. & Blekhman, R. Functional Genomics of Host-Microbiome Interactions in Humans. Trends Genet 34, 30-40, doi:10.1016/ j.tig.2017.10.001 (2018).

28 Maier, L. et al. Extensive impact of non-antibiotic drugs on human gut bacteria. Nature, doi:10.1038/nature25979 (2018).

29 Zeissig, S. & Blumberg, R. S. Life at the beginning: perturbation of the microbiota by antibiotics in early life and its role in health and disease. Nat Immunol 15, 307-310, doi:10.1038/ni.2847 (2014).

30 Gensollen, T., Iyer, S. S., Kasper, D. L. & Blumberg, R. S. How colonization by microbiota in early life shapes the immune system. Science 352, 539-544 (2016).

31 Knutie, S. A., Wilkinson, C. L., Kohl, K. D. & Rohr, J. R. Early-life disruption of amphibian microbiota decreases later-life resistance to parasites. Nat Commun 8, 86, doi:10.1038/s41467-017-00119-0 (2017).

32 Maruvada, P., Leone, V., Kaplan, L. M. & Chang, E. B. The Human Microbiome and Obesity: Moving beyond Associations. Cell Host Microbe 22, 589-599, doi:10.1016/ j.chom.2017.10.005 (2017).

33 Goodrich, J. K. et al. Human genetics shape the gut microbiome. Cell 159, 789-799, doi:10.1016/j.cell.2014.09.053 (2014).

34 Pedersen, H. K. et al. Human gut microbes impact host serum metabolome and insulin sensitivity. Nature 535, 376-381, doi:10.1038/nature18646 (2016).

35 Bonder, M. J. et al. The effect of host genetics on the gut microbiome. Nat Genet 48, 1407-1412, doi:10.1038/ng.3663 (2016). 36 Wang, J. et al. Genome-wide association analysis identifies variation in vitamin D receptor and other host factors influencing the gut microbiota. Nat Genet 48, 1396-1406, doi:10.1038/ng.3695 (2016). 37 Goodrich, J. K. et al. Genetic Determinants of the Gut Microbiome in UK Twins. Cell Host Microbe 19, 731-743, doi:10.1016/j.chom.2016.04.017 (2016).

38 Wijmenga, C. & Zhernakova, A. The importance of cohort studies in the post-GWAS era. Nat Genet, 322-328, doi:10.1038/s41588-018-0066-3 (2018).

39 Wang, X. W., Xu, J. D., Zhao, X. F., Vasta, G. R. & Wang, J. X. A shrimp C-type lectin inhibits proliferation of the hemolymph microbiota by maintaining the expression of antimicrobial peptides. J Biol Chem 289, 11779-11790, doi:10.1074/jbc.M114.552307 (2014). 40 Pang, X. et al. Mosquito C-type lectins maintain gut microbiome homeostasis. Nat Microbiol 1, 16023, doi:10.1038/nmicrobiol.2016.23 (2016).

41 Iliev, I. D. et al. Interactions between commensal fungi and the C-type lectin receptor Dectin-1 influence colitis. Science 336, 1314-1317 (2012).

42 Blekhman, R. et al. Host genetic variation impacts microbiome composition across human body sites. Genome Biol 16, 191, doi:10.1186/s13059-015-0759-1 (2015).

43 Wang, J. et al. Meta-analysis of human genome-microbiome association studies: the MiBioGen consortium initiative. Microbiome 6, 101, doi:10.1186/s40168-018-0479-3 (2018).

44 Kurilshikov, A. et al. Large-scale association analyses identify host factors influencing human gut microbiome composition. Nat Genet, doi:10.1038/s41588-020-00763-1 (2021).

(15)

45 Baumler, A. J. & Sperandio, V. Interactions between the microbiota and pathogenic bacteria in the gut. Nature 535, 85-93 (2016).

46 Eickhoff, M. J. & Bassler, B. L. SnapShot: Bacterial Quorum Sensing. Cell 174, 1328-+ (2018).

47 Schirmer, M. et al. Linking the Human Gut Microbiome to Inflammatory Cytokine Production Capacity. Cell 167, 1897, doi:10.1016/j.cell.2016.11.046 (2016).

48 Berry, D. & Widder, S. Deciphering microbial interactions and detecting keystone species with co-occurrence networks. Front Microbiol 5, 219, doi:10.3389/ fmicb.2014.00219 (2014).

49 Banerjee, S., Schlaeppi, K. & van der Heijden, M. G. A. Keystone taxa as drivers of microbiome structure and functioning. Nat Rev Microbiol 16, 567-576, doi:10.1038/s41579-018-0024-1 (2018).

50 Faust, K. et al. Microbial Co-occurrence Relationships in the Human Microbiome. Plos Computational Biology 8 (2012).

51 Biagi, E. et al. Gut Microbiota and Extreme Longevity. Curr Biol 26, 1480-1485 (2016). 52 Flemer, B. et al. Tumour-associated and non-tumour-associated microbiota in colorectal cancer. Gut 66, 633-643 (2017).

53 Wang, J. et al. Dysbiosis of maternal and neonatal microbiota associated with gestational diabetes mellitus. Gut 67, 1614-1625, doi:10.1136/gutjnl-2018-315988 (2018). 54 Gevers, D. et al. The treatment-naive microbiome in new-onset Crohn's disease. Cell Host Microbe 15, 382-392, doi:10.1016/j.chom.2014.02.005 (2014).

55 Yilmaz, B. et al. Microbial network disturbances in relapsing refractory Crohn's disease. Nat Med 25, 323-336, doi:10.1038/s41591-018-0308-z (2019).

56 Fu, J. et al. The Gut Microbiome Contributes to a Substantial Proportion of the Variation in Blood Lipids. Circ Res 117, 817-824, doi:10.1161/CIRCRESAHA.115.306807 (2015).

57 Henao-Mejia, J. et al. Inflammasome-mediated dysbiosis regulates progression of NAFLD and obesity. Nature 482, 179-185, doi:10.1038/nature10809 (2012).

58 Kang, C. et al. Gut Microbiota Mediates the Protective Effects of Dietary Capsaicin against Chronic Low-Grade Inflammation and Associated Obesity Induced by High-Fat Diet. MBio 8, doi:10.1128/mBio.00470-17 (2017).

59 Chu, H. et al. Gene-microbiota interactions contribute to the pathogenesis of inflammatory bowel disease. Science 352, 1116-1120, doi:10.1126/science.aad9948 (2016). 60 Maslowski, K. M. Metabolism at the centre of the host-microbe relationship. Clin Exp Immunol 197, 193-204, doi:10.1111/cei.13329 (2019).

61 Bar, N. et al. A reference map of potential determinants for the human serum metabolome. Nature 588, 135-140, doi:10.1038/s41586-020-2896-2 (2020).

62 Bradley, P. H. & Pollard, K. S. Building a chemical blueprint for human blood. Nature 588, 36-37, doi:10.1038/d41586-020-03122-6 (2020).

63 Tang, W. H. & Hazen, S. L. The Gut Microbiome and Its Role in Cardiovascular Diseases. Circulation 135, 1008-1010, doi:10.1161/CIRCULATIONAHA.116.024251 (2017).

Referenties

GERELATEERDE DOCUMENTEN

AMC Amsterdam Medical Center; EMC Erasmus Medical Center; LUMC Leiden University Medical Center; MUMC Maastricht University Medical Center; UMCN Radboud University Nijmegen

The first data release, detailed in this announcement and released simultaneously with this publication, will contain basic phenotypes for 1,215 participants, genotypes of

Here we present MOLGENIS Research, an open-source web-application to collect, manage, analyse, visualize and share large and complex biomedical data sets, without the need

Reduced diversity of the gut microbiome associated with PPI use In all three cohorts we identified a lower species richness and lower Shannon diversity, although not

However, we did observe similar findings for the iHMP- IBD cohort for 98.2% of species co-abundances and 99.4% of pathway co- abundances between two time points spanning one

Importantly, these cross- sectional associations between microbiome and metabolites could be further confirmed by making use of the longitudinal study design of the Lifelines cohort,

While numerous novel microbial and genetic associations to plasma and fecal bile acids had been identified previously, our in-depth analysis on the functional potential of

Gut microbial changes over time are related to changes in plasma metabolite levels in the host.