Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Short Article

Diet-Microbiota Interactions Mediate Global


Epigenetic Programming in Multiple Host Tissues
Graphical Abstract Authors
Kimberly A. Krautkramer,
Julia H. Kreznar,
Kymberleigh A. Romano, ...,
Alan D. Attie, Federico E. Rey,
John M. Denu

Correspondence
ferey@wisc.edu (F.E.R.),
john.denu@wisc.edu (J.M.D.)

In Brief
The gut microbiota is an important
metabolic organ and associated with a
number of robust metabolic and
immunologic host phenotypes.
Krautkramer et al. report that diet-gut
microbiota interactions mediate host
epigenetic programming in a variety of
host tissues.

Highlights Accession Numbers


d Gut microbiota alter host histone acetylation and methylation GSE81115
in multiple tissues GSE81117

d Western diet suppresses microbiota-driven SCFA production


and chromatin effects

d SCFAs recapitulate microbiota-driven chromatin and


transcriptional effects

Krautkramer et al., 2016, Molecular Cell 64, 982–992


December 1, 2016 ª 2016 Elsevier Inc.
http://dx.doi.org/10.1016/j.molcel.2016.10.025
Molecular Cell

Short Article

Diet-Microbiota Interactions Mediate Global


Epigenetic Programming in Multiple Host Tissues
Kimberly A. Krautkramer,1,2 Julia H. Kreznar,3 Kymberleigh A. Romano,3 Eugenio I. Vivas,3 Gregory A. Barrett-Wilt,4
Mary E. Rabaglia,5 Mark P. Keller,5 Alan D. Attie,5 Federico E. Rey,3,* and John M. Denu1,2,6,7,*
1Wisconsin Institute for Discovery, Madison, WI 53715, USA
2Department of Biomolecular Chemistry, University of Wisconsin School of Medicine and Public Health – Madison, Madison, WI 53706, USA
3Department of Bacteriology, University of Wisconsin – Madison, Madison, WI 53706, USA
4Biotechnology Center, University of Wisconsin – Madison, Madison, WI 53706, USA
5Department of Biochemistry, University of Wisconsin – Madison, Madison, WI 53706, USA
6Morgridge Institute for Research, Madison, WI 53715, USA
7Lead Contact

*Correspondence: ferey@wisc.edu (F.E.R.), john.denu@wisc.edu (J.M.D.)


http://dx.doi.org/10.1016/j.molcel.2016.10.025

SUMMARY is a necessary substrate for histone acetyltransferases (HATs),


and increased availability can increase HAT activity.
Histone-modifying enzymes regulate transcription The gut microbiota produces a variety of metabolites detect-
and are sensitive to availability of endogenous able in host circulation, including small organic acids, bile acids,
small-molecule metabolites, allowing chromatin to vitamins, choline metabolites, and lipids (reviewed in Nicholson
respond to changes in environment. The gut micro- et al., 2012). Dietary poly- and oligosaccharides resistant to
biota produces a myriad of metabolites that affect digestion by the mammalian host pass to the distal gut where
they serve as a source of carbon and energy for gut bacteria.
host physiology and susceptibility to disease; how-
Through fermentative reactions, the gut microbiota can metabo-
ever, the underlying molecular events remain largely
lize complex carbohydrates to produce small organic acids, the
unknown. Here we demonstrate that microbial colo- majority of which are comprised of the short-chain fatty acids
nization regulates global histone acetylation and (SCFAs) acetate, propionate, and butyrate (R95%) (den Besten
methylation in multiple host tissues in a diet-depen- et al., 2013). SCFAs have been implicated in both disease pro-
dent manner: consumption of a ‘‘Western-type’’ moting and therapeutic effects (Canfora et al., 2015; Belcheva
diet prevents many of the microbiota-dependent et al., 2014; Perry et al., 2016; Tan et al., 2014), prompting a
chromatin changes that occur in a polysaccharide- need for increased understanding of the underlying molecular
rich diet. Finally, we demonstrate that supplementa- mechanisms. Robust associations between gut microbiota and
tion of germ-free mice with short-chain fatty acids, host metabolic outcomes include cardiovascular disease
major products of gut bacterial fermentation, is suffi- (Wang et al., 2011; Karlsson et al., 2012), metabolic syndrome
(Suez et al., 2014; Vijay-Kumar et al., 2010), obesity (Ba €ckhed
cient to recapitulate chromatin modification states
et al., 2004; Ley et al., 2005; Ridaura et al., 2013), diabetes mel-
and transcriptional responses associated with colo-
litus (Amar et al., 2011), and inflammatory bowel disorders and
nization. These findings have profound implications malignancy (Chassaing et al., 2015; Donohoe et al., 2012; 2014).
for understanding the complex functional interac- Eukaryotic histone-modifying enzymes have evolved to sense
tions between diet, gut microbiota, and host health. and integrate environmental signals, ultimately programming
gene expression patterns and mediating phenotype. Given the
sensitivity of these enzymes to endogenous metabolites, we hy-
INTRODUCTION pothesized that gut microbial metabolites absorbed and metab-
olized by the host may exert similar control. Here, we explore
The eukaryotic genome is organized into a highly compressed whether the gut microbiota affects host epigenetic programming
nucleoprotein structure known as chromatin. Histone proteins in a variety of tissues and how this relationship is affected by host
(H2A, H2B, H3, and H4) are major components of chromatin diet. We provide evidence of gut microbiota-mediated changes
and act as spools, wrapping DNA into fundamental nucleosome in global histone acetylation and methylation not only in colon,
units that can fold into higher-order structures. Histones undergo which is in direct contact with microbes and their metabolites,
a myriad of covalent post-translational modifications (PTMs) but also in tissues outside the gut. We demonstrate that
and, along with histone variant replacement, comprise what is this regulatory relationship is sensitive to host diet, wherein a
known as the ‘‘histone code,’’ wherein the local PTM state dic- ‘‘Western-type’’ diet limits microbial SCFA production, abolishes
tates whether chromatin is repressive or activating toward tran- the effects of microbiota on host chromatin states, and results
scription (Jenuwein and Allis, 2001). Enzymes that add or remove in functionally relevant alterations in hepatic gene expression.
PTMs are sensitive to the availability of endogenous metabolites Finally, we identify an underlying mechanism that reveals
(Fan et al., 2015). For example, acetyl-coenzyme A (acetyl-CoA) that SCFA supplementation of germ-free mice is sufficient

982 Molecular Cell 64, 982–992, December 1, 2016 ª 2016 Elsevier Inc.
to recapitulate epigenetic phenotypes associated with gut for these modified nucleosomes along the genome (Figure 1B).
colonization. Interestingly, the singly acetylated peptides K9ac + K14un and
K9un + K14ac decrease concomitantly with an increase in the
RESULTS AND DISCUSSION doubly acetylated K9ac + K14ac peptide, and a similar pattern
occurs on the singly acetylated and coeluting K18ac/K23ac pep-
Gut Microbiota Affect Host Tissue Epigenetic States tides (Figure 1B). These results are consistent with a shift away
To investigate whether gut microbes and their metabolites affect from a singly acetylated state toward a maximally acetylated
host chromatin states, we examined histone PTM states as a state.
function of colonization. We focused our analysis on proximal H3 methylation patterns are also altered as a function of gut
colon, liver, and white adipose tissue (WAT). The experimental colonization status. There is a modest, yet statistically significant
workflow is described in Figure 1A. Briefly, mice were either increase in H3 K27me3 + K36un in proximal colon, liver, and
maintained germ-free (GF) throughout the experiment, allowed adipose tissues from ConvD mice versus their GF controls
to acquire a microbiota from birth to adulthood (conventionally (1.4- to 1.5-fold increase; Figure 1B), accounting for 12% of
raised, ConvR), or colonized with a complete (uncultured) micro- total PTM states and suggesting more broad regulatory effects
biota (conventionalized, ConvD) harvested from ConvR donors. in comparison to highly acetylated states of H3 and H4. There
Use of a ConvD mouse model allows for the determination of were increases in peptides containing highly methylated forms
whether the phenotype observed in ConvR animals is transfer- of K27 and K36 (i.e., me2 and me3) on both the canonical H3
rable via the gut microbiota alone. Additionally, since ConvR an- and the variant H3.3; however, these effects were not present
imals experience different environmental exposure early in life consistently across all three tissues, suggesting some tissue
and have developmental differences (Smith et al., 2007) that specificity in the response to colonization (Figure 1B). Notably,
may exhibit phenotypic differences versus their GF controls, H3 K18me1 decreased across all three ConvD tissues (2.1- to
the use of ConvD mice for relatively short time periods allows 8.3-fold across liver, proximal colon, and adipose tissue; Fig-
for dissection of effects more directly related to differences in mi- ure 1B). A similar pattern was present for the combinatorial
crobial metabolism. Histones were extracted from tissues and K27me2 + K36me1 peptide on H3 and variant H3.3 (Figure 1B).
prepared for mass spectrometry analysis using an in-house Together, these results demonstrate that gut microbiota affect
workflow (Krautkramer et al., 2015). host tissue acetylated and methylated chromatin states in a
We surveyed 55 unique and combinatorial acetylated and site-specific and combinatorial fashion, strongly supporting a
methylated histone PTM states in proximal colon, liver, and role for the gut microbiota as a driver of host tissue chromatin
WAT (Table S1). Colonization induced robust increases in H4 regulation. While some histone PTM states appear to be similarly
acetylation in all three tissues (Figure 1B). This peptide includes regulated across all tissues surveyed, other changes are unique
the first 4 lysines (K5, K8, K12, and K16) on the H4 N-terminal within a tissue.
tail. Thus, H4: 0ac indicates peptides where no lysine residues The ability to detect histone PTMs within the context of neigh-
are acetylated, whereas H4: 1ac–4ac indicates peptides where boring modifiable sites allowed us to uncover combinatorial PTM
any 1–4 of the 4 lysines are acetylated. In ConvR animals, there states that account for a very small percentage of the total and
was a significant 2.1-fold increase in both triply and quadruply that are not necessarily resolvable by orthologous techniques
acetylated H4 (H4: 3ac and H4: 4ac, respectively). Similarly, such as western blot analysis. Indeed, we performed several
ConvR animals showed a 3-fold and 1.30-fold increase in the western blots and found no statistically significant differences
highly acetylated H4: 4ac peptide in proximal colon and adipose in H3 K9ac, H3 K27me3, or pan-acetyl(K) detectable via western
tissue, respectively, relative to GF mice (Figure 1B). The effects of blot in histone extracts from GF, ConvR, and ConvD mouse
colonization on H4 acetylation were even more robust in ConvD livers (Figures S1A and S1B). To compare methods, we summed
mice, with a 4.5-, 6.0-, and 12.0-fold increase in H4: 4ac of prox- all possible permutations of peptides containing K9ac and
imal colon, adipose, and liver, respectively (Figure 1B). Triply K27me3 on canonical histone H3 to obtain single western blot-
acetylated H4 peptides also increased 2.1- to 4.2-fold in proximal like estimates of K9ac and K27me3 abundance for GF, ConvR,
colon, adipose, and liver (Figure 1B). It is noteworthy that these and ConvD mouse liver histone PTM states (see Supplemental
two H4 states collectively account for just over 1% of the total Experimental Procedures). Calculating fold changes relative to
H4 states, suggesting that this open chromatin state is confined GF, both K9ac and K27me3, as a total among all combinatorial
to very specific loci along the genome. Consistent with the forms quantified, remained relatively unchanged between colo-
conversion of unacetylated states to higher acetylation, the nized and GF mice (Figure S1C), consistent with quantitative
completely unmodified form decreased significantly (H4: 0ac, western blot results (Figure S1B).
1.33- to 3.3-fold across tissues surveyed) in colonized animals.
Microbes similarly induced acetylation of canonical H3 and the Gut Microbiota-Mediated Changes in Chromatin State
variant H3.3. The doubly acetylated canonical H3 K9ac + K14ac Are Sensitive to Host Diet
and H3 K18ac + K23ac peptides increased significantly in ConvD Given the known effect of host diet on gut microbial community
mouse livers and trended toward a similar magnitude increase composition and metabolism (Shoaie et al., 2015; Daniel et al.,
in proximal colon of ConvR and ConvD mice (Figure 1B). These 2014; David et al., 2013; Turnbaugh et al., 2008, 2009b), we
two doubly acetylated canonical H3 peptides account for next evaluated the effects of host diet on microbiota-mediated
roughly 2% or less of total histone PTM states observed in regulation of host chromatin states (Table S1). ConvR and GF
each peptide family, again supporting a more loci-specific role mice were fed a ‘‘Western-type’’ high-fat, high-sucrose diet

Molecular Cell 64, 982–992, December 1, 2016 983


Figure 1. Gut Microbiota Affect Host Tissue Epigenetic States
(A) Experimental design: (1) tissues harvested from germ-free (GF), conventionally raised (ConvR), and conventionalized (ConvD) mice. (2 and 3) Histones extracted,
chemically derivatized, and trypsinized. (4 and 5) Histone peptides injected onto mass spectrometer and data acquired on >50 unique histone PTM states.
(B) Relative abundance of histone PTMs on H3, H3.3, and H4. Values are reported as a fold change relative to GF controls (log2). Mean percentage of peptide
family total across all samples (rightmost column).
*p < 0.05, **p < 0.01, ***p < 0.001; n = 4 mice per condition.

984 Molecular Cell 64, 982–992, December 1, 2016


A
GF GF

ion intensity
chow HF/HS
or 1 2-5

ConvR ConvR 51.0 52.0 53.0 54.0


chow HF/HS Retention Time

*
B C 6 *** D 40000
45 *
***
***

(μg / mg total protein)


30000
40
Total Cholesterol
(μg / mg tissue)
4

Triglycerides
Weight (g)

* 20000
35
*
2
30 10000 *

25 0 0
GF ConvR ConvD GF ConvR GF ConvR ConvD GF ConvR
GF ConvR
chow chow chow HF/HS HF/HS chow chow chow HF/HS HF/HS
HF/HS HF/HS

E F G 5 Adipose
5
Liver
30 ** ConvD chow
ConvR chow ConvR, chow 4
*** ConvR, chow
4 ConvD, chow ConvD, chow
GF chow
ConvR, HF/HS ConvR, HF/HS *
mM/g cecal contents

3
fold change vs. GF (log2)

ConvR HF/HS
fold change vs. GF (log2)
20 GF HF/HS 3
** *** 2
** 2 *** **
*
10 ** ** * 1
**
***
1 **
** ** 0 *** **
* *
0
0 -1
acetate propionate butyrate
-1
0ac 1ac 2ac 3ac 4ac -2
0ac 1ac 2ac 3ac 4ac

H I J
Proximal Colon 3 Liver
5
4 Proximal Colon
ConvR, chow **
ConvD, chow
fold change vs. GF (log2)

4 2 ConvR, chow 3
ConvR, HF/HS
fold change vs. GF (log2)

ConvD, chow ConvR, chow


fold change vs. GF (log2)

ConvR, HF/HS ConvD, chow


3 2 **
p = 0.05

* 1 ConvR, HF/HS

2 *** *** 1 * * *
0 ** *
** ** 0 *
1 * *
*** ** n
-1 c 3u e1
* ** ** -1
3a ac m 3u
n
23 K2 23
n

e1

0 K2 K2
3u

1 K
n

K
c

/
3m

e
3u
3a

3a

c c n n
K2

8a 8a 8m 8u 8u
K2
K2

K2

K2

K1 K1 K1 K1 K1
e1

-2
c/

n
c

n
8m
8a

8a

8u
8u

-1
K1

K1

K1
K1

K1

0ac 1ac 2ac 3ac 4ac

Figure 2. Gut Microbiota-Mediated Epigenetic Changes Are Sensitive to Diet


(A) Experimental design: (1) GF and ConvR mice on either chow or a HF/HS (‘‘Western’’) diet. (2–5) Tissues were prepared and analyzed as described in Figure 1.
(B) Weights of GF and ConvR mice fed a HF/HS diet at the time of sacrifice.

(legend continued on next page)

Molecular Cell 64, 982–992, December 1, 2016 985


(HF/HS), which contains low levels of fermentable substrate for basal histone H4 acetylation in liver, proximal colon, and adipose
the gut microbiota, for 16 weeks prior to sacrifice at 19 weeks tissue. In comparison to H4 acetylation in chow-fed ConvR and
of age. Tissues were harvested, and physiological parameters GF animals, there were significant increases in nearly all forms of
and histone PTM states were measured (Figure 2A). As antici- 1ac–4ac H4 peptides in HF/HS-fed tissues ranging from 1.2- to
pated, ConvR mice fed a HF/HS diet weighed more than diet- 7.2-fold (Figure 1B).
matched GF controls (Figure 2B). HF/HS-fed ConvR mice dis- Although the direction of change in both acetylated and
played higher hepatic total cholesterol and triglycerides versus methylated histone PTM states was similar in ConvR and
diet-matched GF controls and chow-fed mice (Figures 2C and ConvD tissues, differences in magnitude (Figures 1B and 2F–
2D). Thus, HF/HS feeding impacted host metabolic state in a 2J) trended with cecal SCFA levels (Figure 2E). To investigate
microbiota-dependent manner. To determine whether HF/HS whether these differences reflected alterations to microbial
feeding altered SCFA production, we measured acetate, propio- community composition, we performed 16S rRNA sequencing.
nate, and butyrate levels in cecal contents (i.e., principal site of Principal Coordinates Analysis (PCoA) of weighted UniFrac dis-
fermentation) of ConvR, ConvD, and GF mice on both chow tances revealed that microbial community compositions of
and HF/HS diets. Gut microbial colonization increased these ConvR and ConvD mice on a chow diet were more similar to
metabolites in the ceca of mice, and this increase was more pro- each other than they were to the microbial community from
nounced in mice fed a chow diet compared to a HF/HS diet low in mice fed HF/HS diet (Figure S2A). ConvR mice fed a HF/HS
fermentable complex polysaccharides (Figure 2E). In HF/HS-fed diet had significantly fewer Bacteroidetes and a greater abun-
ConvR mice, cecal acetate, propionate, and butyrate levels were dance of Firmicutes (Figures S2B and S2C) than chow-fed
lower relative to chow-fed ConvR mice (1.9- to 6.0-fold; Figures ConvR and ConvD mice, consistent with previous observations
2E and S4A). Interestingly, chow-fed ConvD mice had greater that diet and obesity alter this ratio in the gut (Ley et al., 2005).
cecal SCFAs than ConvR mice (Figure 2E). This pattern in cecal Relative abundance of these two major phyla in chow-fed
SCFA levels is consistent with that of histone PTM changes in ConvD mice was intermediate between chow- and HF/HS-fed
ConvD and ConvR mice on chow, wherein PTM states trend ConvR mice (Figures S2B–S2D). Together, these data suggest
in the same direction, but the magnitude of change is larger in that gut microbial community composition and metabolite pro-
ConvD mice versus ConvR (Figure 1B). Thus, SCFA availability duction are important factors that connect gut microbiota and
influences histone PTM states. In peripheral venous blood, host chromatin states.
levels of these SCFAs were not significantly different (Fig-
ure S4B), consistent with these organic acids having undergone Functional Impact: Co-regulation of Hepatic Genes
significant metabolism in the liver prior to reaching peripheral Associates with Altered Chromatin States
venous blood. To assess whether microbiota-dependent changes in host chro-
As predicted by cecal SCFA data, the gut microbiota-host epi- matin state affected tissue gene expression, we performed RNA-
genome relationship was altered in response to HF/HS feeding. seq analyses on livers of colonized (both ConvR and ConvD) and
While there was a microbiota-dependent increase in H4 acetyla- GF mice on the two diets described above. Each group of colo-
tion in ConvR and ConvD tissues of chow-fed mice (Figure 1B), nized mice was compared to its diet-matched GF control: i.e.,
HF/HS feeding abolished the effects of gut colonization in liver chow-fed ConvR versus GF, chow-fed ConvD versus GF, and
and WAT (Figures 2F and 2G). Interestingly, the microbiota- HF/HS-fed ConvR versus GF. A total of 623 genes were differen-
dependent effects on H4 acetylation were attenuated but still tially expressed (DE) among these three groups, as determined
significantly increased relative to GF controls in HF/HS-fed by an FDR cut-off of 0.05 (Table S2). K-means clustering of he-
mouse proximal colon (Figure 2H). This pattern of diet depen- patic DE genes revealed 6 optimal clusters, each enriched for
dence was also present in other histone PTM states. The unique biological pathways (Figure 3A; Table S2). When
response to gut microbiota on H3 K18 and K23 also trended comparing ConvR mice on either chow or HF/HS diets to their
as a function of diet: K18me1 and K23me1 peptides both respective GF controls, cluster 2 contained genes that are co-
decreased significantly in livers of both ConvR and ConvD regulated as a function of both diet and microbiota (Figures 3A
chow-fed mice but remained unchanged in HF/HS-fed mice (Fig- and 3B). This group of genes was enriched for processes
ure 2I). The coeluting peptides K18ac and K23ac (i.e., K18ac/ involved in insulin, SREBP, and PPAR signaling, and adaptive
K23ac) were unchanged in response to gut microbiota in livers immunity. Additionally, this cluster contains a number of genes
of chow-fed mice yet decreased in HF/HS-fed mouse livers (Fig- that may regulate histone PTM states via modulation of small
ure 2I). However, in proximal colon this diet dependency was molecule metabolite availability. Clusters 4 and 6 contain genes
again absent, and histone PTM states trended in similar direc- whose expression patterns differ in ConvR animals as a function
tions regardless of dietary conditions (Figure 2J). There was of diet (Figures 3A, 3C, and 3D). Cluster 4 genes are enriched
also a diet-dependent, microbiota-independent increase in for pathways involved in cholesterol, retinol, and amino acid

(C and D) Hepatic total cholesterol (C) and hepatic triglycerides (D) in GF, ConvD, and ConvR mice on chow and HF/HS diets.
(E) SCFA measurement in cecal contents from GF, ConvR, and ConvD animals on chow and HF/HS diet. *p < 0.05, **p < 0.01; error bars represent SD; n = 4 mice
per condition.
(F–H) H4 (K5, K8, K12, and K16) acetylation in colonized liver (F), adipose tissue (G), and proximal colon (H) relative to GF controls (fold change, log2).
(I and J) H3 K18 and K23 methylation and acetylation in colonized liver (I) and proximal colon (J) relative to GF controls (fold change, log2).
*p < 0.05, **p < 0.01, ***p < 0.001; error bars represent SEM; n = 4 mice per condition.

986 Molecular Cell 64, 982–992, December 1, 2016


Figure 3. Hepatic Genes Are Co-regulated in Colonized Mice as a Function of Diet or Colonization Status
(A) K-means clustering of differentially expressed hepatic genes (left, fold change versus GF, log2) and KEGG pathway enrichment terms (right).
(B–D) Interaction network for cluster 2 (B), cluster 4 (C), and cluster 6 (D). Only genes with at least one reported interaction are graphed. Edges indicate interaction.
Node size indicates relative expression in HF/HS-fed mouse livers. Node color indicates relative expression in chow-fed mouse livers.

Molecular Cell 64, 982–992, December 1, 2016 987


metabolism as well as host immunity, whereas cluster 6 contains is not unexpected that colonized mice have altered expression
a number of genes involved in lipid and amino acid metabolism of immunomodulatory genes relative to GF mice. Gut microbiota
as well as a group of genes involved in regulation of folate, which are important for the development and function of host immunity
ultimately affects the availability of the one-carbon donor SAM to (Belkaid and Hand, 2014), and SCFAs play an important role in
histone methyltransferases. Notably, 4% of DE genes are known both adaptive and innate immunity (Belkaid and Hand, 2014;
hepatic targets of SREBP and PPAR. Smith et al., 2013). More detailed information about specific
To assess the effects of diet alone, we compared GF and gene cluster membership, expression, and pathway enrichment
ConvR mice across dietary conditions (i.e., HF/HS versus among clusters is described in Tables S2 and S3, Figure S3, and
chow). Of the 868 differentially expressed genes, 413 increased the Supplemental Information.
and 455 decreased in expression in HF/HS-fed mice relative to
chow-fed controls (Figures S3A and S3B). Although a fraction SCFA-Supplementation Partially Phenocopies the
of DE up (Figure S3A) and DE down (Figure S3B) genes are regu- Effects of Colonization on Host Epigenetic
lated in both ConvR and GF mice, the fact that there are 1.8-fold Programming
more total DE genes in response to diet in ConvR mice versus GF We hypothesized that SCFAs acetate, propionate, and butyrate
mice suggests that gut microbiota drive a significant portion of were key mediators of systemic microbiota-induced changes in
the response to HF/HS feeding in liver. KEGG pathway analysis host chromatin states. To explore this idea, we supplemented
of unique and overlapping DE genes in GF and ConvR mice re- germ-free mice with acetate, propionate, and butyrate (GF +
vealed several oppositely regulated pathways as a function of SCFA) and harvested liver and proximal colon for histone PTM
diet (Figures S3C and S3D). For example, while starch and su- and gene expression analyses (Figure 4A). These GF + SCFA
crose metabolism is enriched in DE up genes of ConvR mice, mice were then compared to GF and ConvD mice, as negative
this same pathway is enriched in DE down genes of GF mice. and positive controls, respectively. Hierarchical clustering re-
The same pattern is present for pathways involved in arachidonic vealed that histone PTM states of GF + SCFA and ConvD mice
acid metabolism, cytokine-cytokine receptor interaction, and were strikingly similar across both acetylated and methylated
endocytosis. Other key differences include DE up gene enrich- peptides (Figure 4B). Further, GF + SCFA and ConvD histone
ment for processes involved in the TCA cycle and propionate PTM signatures were highly correlated, with a Pearson’s correla-
metabolism in GF mice only, and butyrate metabolism in ConvR tion coefficient of 0.74–0.75 for proximal colon and liver samples
mice only (Figure S3C). Importantly, genes involved in PPAR (p = 1.2 3 10 10 and 5.7 3 10 11, respectively; Figures 4C and
signaling, insulin signaling, and diabetes mellitus were enriched 4D). These data reveal that the global chromatin states induced
in DE up genes shared by both GF and ConvR mice (Figure S3C). by SCFAs mimic gut colonization.
Pathways involved in de novo cholesterol synthesis are signifi- While levels of acetate, propionate, and butyrate were signifi-
cantly enriched in DE down genes shared by both GF and ConvR cantly increased in the cecal contents of ConvD mice, there were
mice, suggesting that HF/HS feeding decreases host de novo no significant differences between GF + SCFA mice and GF con-
cholesterol synthesis irrespective of gut colonization status. trols (Figure S4C). Similar to previous observations (Figure S4B),
Gut microbiota altered expression of genes linked to metabo- peripheral blood levels of SCFAs were unchanged as a function
lites that directly regulate histone PTM addition or removal. For of either gut colonization or SCFA supplementation (Figure S4D).
example, expression of ATP citrate lyase (Acly) is decreased in Thus, while conventionalization results in increased local con-
ConvR versus GF mice, under both chow and HF/HS feeding. centration of cecal SCFAs, oral supplementation of SCFAs did
Acly is essential for glucose-driven, but not acetate-driven, his- not, likely due to absorption along alimentary tract. Despite these
tone acetylation in mammalian cells (Wellen et al., 2009). Acly potential differences, the physiological impacts are likely to be
expression decreases most in HF/HS-fed ConvR mouse livers, similar, given that venous blood from the small intestine also
suggesting that the presence of other sources of carbon and en- drains to the liver via the hepatic portal vein where it mixes
ergy, such as bacterial SCFAs or highly energetic lipids from HF/ with blood traveling from the colon.
HS feeding, may suppress glucose-driven histone modification. To determine whether these highly similar global chromatin
Availability of NAD+, a necessary co-substrate for class III HDACs, states elicited similar biological effects in ConvD and GF +
may also be modulated by gut microbiota. NAMPT, which cata- SCFA mice, we used RNA-seq analyses to examine hepatic
lyzes the rate-limiting step in NAD+ biosynthesis, was differentially gene expression in ConvD, GF + SCFA, and GF mice. Consis-
expressed in colonized versus GF livers. Lastly, there were four tent with histone PTM observations, GF + SCFA mice had
genes in cluster 6 linked to regulation of folate, which can affect transcriptional profiles highly similar to those of ConvD mice
the availability of the methyl donor SAM for histone methyltrans- (Figures 4E and 4G). K-means clustering of 537 DE genes re-
ferases: Sardh, Dmgdh, Amt, and Gldc (Figure 3D). Loss of vealed 6 clusters of co-regulated genes that were enriched for
Gldc has been associated with neural tube defects, growth retar- a number of metabolic and immunological processes (Fig-
dation, and decreased levels of one-carbon-carrying folates (Pai ures 4E and 4F; Table S4). In particular, clusters b and c were
et al., 2015). Thus, decreased expression of these enzymes enriched for GO terms involved in immunity (cluster b) and
may affect tissue SAM availability, via modulation of folate levels. regulation, storage, and metabolism of lipids and cholesterol
Finally, a number of genes associated with host immunity (cluster c; Table S4). Finally, there was striking overlap of DE
and inflammation were differentially expressed in colonized genes between GF + SCFA and ConvD mice, wherein >50%
and GF + SCFA mice relative to their GF controls (Figures 3A– of DE genes in GF + SCFA livers overlapped with DE genes in
3D, 4F, and 4G). Given that GF mice are naive to microbiota, it ConvD mice (Figure 4G). Together, these data suggest that

988 Molecular Cell 64, 982–992, December 1, 2016


(legend on next page)

Molecular Cell 64, 982–992, December 1, 2016 989


SCFAs are partially causative metabolites in the complex regu- 14.8% protein). Diets were sterilized by irradiation and autoclaving. Sterility
latory relationship between diet, gut microbiota, and host tissue of germ-free animals was assessed by incubating freshly collected fecal sam-
ples under aerobic and anaerobic conditions using standard microbiology
epigenetic programming.
methods. Final dissection and data collection were performed at 19 weeks
SCFAs play a dual role both as substrates for metabolism and of age.
as signaling molecules (den Besten et al., 2013). These SCFAs ConvD mice were generated by colonizing GF C57BL/6J mice with fresh
can be either directly converted (acetate) or oxidized (propionate cecal contents collected from 15-week-old conventionally raised C57BL/6J
and butyrate) to acetyl-CoA, the substrate for HAT enzymes. As mice maintained on a control breeder chow diet (n = 2 mice per donor cecal
much as 70% of acetate and roughly 30% of propionate is taken microbiota sample per experiment; 5021, Lab Diet). Immediately after sacri-
fice, fresh cecal contents from donor mice were re-suspended in Mega
up by the liver, where both serve as sources of energy. Further,
Medium (1:100 w/v) in an anaerobic chamber (Romano et al., 2015). Suspen-
butyrate is a known HDAC inhibitor. Both scenarios could in-
sions were transferred into anaerobic sealed tubes and used to colonize mice
crease histone acetylation. Acetate can also serve as a substrate in a sterile biological safety cabinet. Germ-free 15-week-old C57BL/6J male
for cholesterol, long-chain fatty acid, glutamine, and glutamate mice were inoculated via a single oral gavage with 0.2 mL of cecal inocula
synthesis in the liver. The remainder is metabolized by other (Turnbaugh et al., 2009b) and kept in sealed filter-top gnotobiotic cages for
tissues, including white adipose. As ligands of the G protein- 4 weeks.
coupled receptors FFAR2 (GPR43) and FFAR3 (GPR41), SCFAs
play a role in lipid and glucose metabolism (Samuel et al., 2008; SCFA Supplementation
GF C57BL/6J male mice were maintained in sterile HEPA filter cages and fed
Xiong et al., 2004). Thus, these metabolites play complex roles in
water and autoclaved chow ad libitum. At 12 weeks of age a subset of GF mice
regulation of host metabolic phenotype. How combinations of or were supplemented with a mixture of SCFAs (acetate 67.5 mM, butyrate
specific SCFAs contribute to beneficial or pathogenic effects in 40 mM, propionate 25.9 mM; Smith et al., 2013) via drinking water supplied
the host remains unclear. ad libitum or ConvD with a C57BL/6J intestinal microbial community by
Though the link between histone acetylation and SCFAs may gavage. Inoculum for colonization was prepared by suspending freshly
be direct, the link between SCFAs and alterations in histone collected fecal pellets in Mega Medium. SCFA supplemented water was
freshly prepared and changed every 5 days and again 24 hr before sacrifice.
methylation is less clear. Moreover, the effects of SCFA supple-
Sterility of GF animals was assessed by incubating freshly collected fecal sam-
mentation broadly recapitulated both chromatin states and gene ples under aerobic and anaerobic conditions using standard microbiology
expression patterns of ConvD mice; however, it is notable that methods. Prior to sacrifice and tissue collection at 14 weeks of age, all mice
the magnitude of the effects was generally less in SCFA + GF (GF, GF + SCFA, ConvD) were anesthetized with 1%–5% inhalant isoflurane
tissues relative to ConvD (Figures 4B and 4E). Collectively, these supplied in oxygen.
results suggest that while SCFAs alone can induce aspects The methods used to measure histone PTMs, 16S rRNA sequencing,
gene expression, hepatic total cholesterol and triglycerides, and cecal and
of a ‘‘colonized’’-like state in host chromatin, there are likely
peripheral blood SCFAs are described in the Supplemental Experimental
other bacterial metabolites affecting host epigenome. Particu-
Procedures.
larly interesting are those directing linked to methyl-donor ca- The Histone Spectral Library (Data S1) is a curated in-house library prepared
pacity for epigenetic methylation. We postulate that eukaryotic from Mascot search results from numerous data-dependent acquisition (DDA)
histone-modifying enzymes have evolved to ‘‘sense’’ not only injections of histone peptides that were aggregated using the redundant library
endogenous metabolites, but those produced by commensal feature of Skyline.
microbiota.
ACCESSION NUMBERS
EXPERIMENTAL PROCEDURES
The accession numbers for data reported in this paper are Gene Expression
Omnibus (GEO): GSE81115, GSE81117.
Mouse Husbandry
Animal care and study protocols were approved by the UW-Madison Animal
Care and Use Committee. Mice were housed in the Microbial Sciences SUPPLEMENTAL INFORMATION
Building vivarium. ConvR and GF C57BL/6J mice were bred at UW-Madi-
son. GF mice were housed in separate plastic flexible vinyl gnotobiotic Supplemental Information includes Supplemental Experimental Procedures,
isolators. four figures, four tables, and the Histone Spectral Library and can be found
Mice were group housed by colonization status and diet (3–5 mice/cage) with this article online at http://dx.doi.org/10.1016/j.molcel.2016.10.025.
under standard conditions (12 hr light:dark, temperature- and humidity-con-
trolled conditions) and received ad libitum access to water and food. After AUTHOR CONTRIBUTIONS
3 weeks of age, mice were maintained on either a control breeder chow
(5021, Lab Diet, 23.7% kcal fat, 53.2% carbohydrate, 23.1% protein) or a K.A.K. conceived of the project, performed experiments, interpreted
HF/HS diet (TD.08811, Envigo Teklad, 44.6% kcal fat, 40.6% carbohydrate, results, prepared figures, and wrote the manuscript. J.H.K., K.A.R., and

Figure 4. SCFA-Supplementation Mimics Colonization-Induced Epigenetic Programming


(A) Experimental design: (1) GF mice were supplemented with SCFAs (GF + SCFA) or colonized (ConvD), and tissues were harvested. (2–5) Histone extracts were
prepared as described in Figure 1.
(B) Hierarchical clustering of histone PTMs in ConvD and GF + SCFA mouse tissues (fold change versus GF, log2).
(C and D) Pearson’s correlation of ConvD and GF + SCFA mouse tissue histone PTM states in liver (C) and proximal colon (D).
(E) K-means clustering of differentially expressed hepatic genes in ConvD and GF + SCFA mice. FDR cut-off for differential expression = 0.05; n = 3 mice per
condition.
(F) GO term enrichment in clusters b and c.
(G) Overlap of differentially expressed genes between ConvD and GF + SCFA mice.

990 Molecular Cell 64, 982–992, December 1, 2016


G.A.B.-W. performed experiments, interpreted results, and contributed to Fan, J., Krautkramer, K.A., Feldman, J.L., and Denu, J.M. (2015). Metabolic
writing the manuscript. E.I.V. and M.E.R. performed experiments. A.D.A. regulation of histone post-translational modifications. ACS Chem. Biol. 10,
and M.P.K. interpreted results and contributed to writing the manuscript. 95–108.
J.M.D. and F.E.R. conceived of the project, interpreted results, and wrote Jenuwein, T., and Allis, C.D. (2001). Translating the histone code. Science 293,
the manuscript. 1074–1080.
Karlsson, F.H., Fåk, F., Nookaew, I., Tremaroli, V., Fagerberg, B., Petranovic,
ACKNOWLEDGMENTS €ckhed, F., and Nielsen, J. (2012). Symptomatic atherosclerosis is asso-
D., Ba
ciated with an altered gut metagenome. Nat. Commun. 3, 1245.
K.A.K. is supported by NIH F30 DK108494 and J.M.D. by NIH GM059789-15/ Krautkramer, K.A., Reiter, L., Denu, J.M., and Dowell, J.A. (2015).
P250VA. Additional support: Clinical and Translational Science Award pro- Quantification of SAHA-Dependent Changes in Histone Modifications Using
gram through the NIH National Center for Advancing Translational Sciences Data-Independent Acquisition Mass Spectrometry. J. Proteome Res. 14,
grants UL1TR000427 and KL2TR000428 (F.E.R.) and NIH DK108259 (F.E.R.) 3252–3262.
and DK101573 (A.D.A.). Extended personal thanks are provided in the Supple- €ckhed, F., Turnbaugh, P., Lozupone, C.A., Knight, R.D., and
Ley, R.E., Ba
mental Information.
Gordon, J.I. (2005). Obesity alters gut microbial ecology. Proc. Natl. Acad.
Sci. USA 102, 11070–11075.
Received: May 7, 2016
Nicholson, J.K., Holmes, E., Kinross, J., Burcelin, R., Gibson, G., Jia, W., and
Revised: September 22, 2016
Pettersson, S. (2012). Host-gut microbiota metabolic interactions. Science
Accepted: October 18, 2016
336, 1262–1267.
Published: November 23, 2016
Pai, Y.J., Leung, K.-Y., Savery, D., Hutchin, T., Prunty, H., Heales, S., Brosnan,
M.E., Brosnan, J.T., Copp, A.J., and Greene, N.D.E. (2015). Glycine decarbox-
REFERENCES
ylase deficiency causes neural tube defects and features of non-ketotic hyper-
glycinemia in mice. Nat. Commun. 6, 6388.
Amar, J., Serino, M., Lange, C., Chabo, C., Iacovoni, J., Mondot, S., Lepage,
Perry, R.J., Peng, L., Barry, N.A., Cline, G.W., Zhang, D., Cardone, R.L.,
P., Klopp, C., Mariette, J., Bouchez, O., et al.; D.E.S.I.R. Study Group (2011).
Petersen, K.F., Kibbey, R.G., Goodman, A.L., and Shulman, G.I. (2016).
Involvement of tissue bacteria in the onset of diabetes in humans: evidence for
Acetate mediates a microbiome-brain-b-cell axis to promote metabolic syn-
a concept. Diabetologia 54, 3055–3061.
drome. Nature 534, 213–217.
€ckhed, F., Ding, H., Wang, T., Hooper, L.V., Koh, G.Y., Nagy, A.,
Ba
Ridaura, V.K., Faith, J.J., Rey, F.E., Cheng, J., Duncan, A.E., Kau, A.L., Griffin,
Semenkovich, C.F., and Gordon, J.I. (2004). The gut microbiota as an environ-
N.W., Lombard, V., Henrissat, B., and Bain, J.R. (2013). Gut microbiota from
mental factor that regulates fat storage. Proc. Natl. Acad. Sci. USA 101,
twins discordant for obesity modulate metabolism in mice. Science 341,
15718–15723.
1241214.
Belcheva, A., Irrazabal, T., Robertson, S.J., Streutker, C., Maughan, H.,
Romano, K.A., Vivas, E.I., Amador-Noguez, D., and Rey, F.E. (2015). Intestinal
Rubino, S., Moriyama, E.H., Copeland, J.K., Kumar, S., Green, B., et al.
microbiota composition modulates choline bioavailability from diet and accu-
(2014). Gut microbial metabolism drives transformation of MSH2-deficient co-
mulation of the proatherogenic metabolite trimethylamine-N-oxide. MBio 6,
lon epithelial cells. Cell 158, 288–299.
e02481.
Belkaid, Y., and Hand, T.W. (2014). Role of the microbiota in immunity and €ckhed, F., Manchester, J.K.,
Samuel, B.S., Shaito, A., Motoike, T., Rey, F.E., Ba
inflammation. Cell 157, 121–141.
Hammer, R.E., Williams, S.C., Crowley, J., Yanagisawa, M., et al. (2008).
Canfora, E.E., Jocken, J.W., and Blaak, E.E. (2015). Short-chain fatty acids in Effects of the gut microbiota on host adiposity are modulated by the short-
control of body weight and insulin sensitivity. Nature 11, 577–591. chain fatty-acid binding G protein-coupled receptor, Gpr41. PNAS 105,
Chassaing, B., Koren, O., Goodrich, J.K., Poole, A.C., Srinivasan, S., Ley, 16767–16772.
R.E., and Gewirtz, A.T. (2015). Dietary emulsifiers impact the mouse Shoaie, S., Ghaffari, P., Kovatcheva-Datchary, P., Mardinoglu, A., Sen, P.,
gut microbiota promoting colitis and metabolic syndrome. Nature 519, Pujos-Guillot, E., de Wouters, T., Juste, C., Rizkalla, S., Chilloux, J., et al.
92–96. (2015). Quantifying Diet-Induced Metabolic Changes of the Human Gut
Daniel, H., Moghaddas Gholami, A., Berry, D., Desmarchelier, C., Hahne, Microbiome. Cell Metabolism 22, 320–331.
H., Loh, G., Mondot, S., Lepage, P., Rothballer, M., Walker, A., et al. Smith, K., McCoy, K.D., and Macpherson, A.J. (2007). Use of axenic animals in
(2014). High-fat diet alters gut microbiota physiology in mice. ISME J 8, studying the adaptation of mammals to their commensal intestinal microbiota.
295–308. Semin. Immunol. 19, 59–69.
David, L.A., Maurice, C.F., Carmody, R.N., Gootenberg, D.B., Button, J.E., Smith, P.M., Howitt, M.R., Panikov, N., Michaud, M., Gallini, C.A., Bohlooly-Y,
Wolfe, B.E., Ling, A.V., Devlin, A.S., Varma, Y., Fischbach, M.A., et al. M., Glickman, J.N., and Garrett, W.S. (2013). The microbial metabolites, short-
(2013). Diet rapidly and reproducibly alters the human gut microbiome. chain fatty acids, regulate colonic Treg cell homeostasis. Science 341,
Nature 505, 559–563. 569–573.
den Besten, G., van Eunen, K., Groen, A.K., Venema, K., Reijngoud, D.J., Suez, J., Korem, T., Zeevi, D., Zilberman-Schapira, G., Thaiss, C.A., Maza, O.,
and Bakker, B.M. (2013). The role of short-chain fatty acids in the interplay Israeli, D., Zmora, N., Gilad, S., Weinberger, A., et al. (2014). Artificial sweet-
between diet, gut microbiota, and host energy metabolism. J. Lipid Res. 54, eners induce glucose intolerance by altering the gut microbiota. Nature 514,
2325–2340. 181–186.
Donohoe, D.R., Collins, L.B., Wali, A., Bigler, R., Sun, W., and Bultman, Tan, J., McKenzie, C., Potamitis, M., Thorburn, A.N., Mackay, C.R., and Macia,
S.J. (2012). The Warburg effect dictates the mechanism of butyrate- L. (2014). The role of short-chain fatty acids in health and disease. Adv.
mediated histone acetylation and cell proliferation. Mol. Cell 48, Immunol. 121, 91–119.
612–626. Turnbaugh, P.J., Ba €ckhed, F., Fulton, L., and Gordon, J.I. (2008). Diet-induced
Donohoe, D.R., Holley, D., Collins, L.B., Montgomery, S.A., Whitmore, A.C., obesity is linked to marked but reversible alterations in the mouse distal gut mi-
Hillhouse, A., Curry, K.P., Renner, S.W., Greenwalt, A., Ryan, E.P., et al. crobiome. Cell Host and Microbe 3, 213–223.
(2014). A gnotobiotic mouse model demonstrates that dietary fiber protects Turnbaugh, P.J., Ridaura, V.K., Faith, J.J., Rey, F.E., Knight, R., and Gordon,
against colorectal tumorigenesis in a microbiota- and butyrate-dependent J.I. (2009b). The effect of diet on the human gut microbiome: a metagenomic
manner. Cancer Discov. 4, 1387–1397. analysis in humanized gnotobiotic mice. Sci. Transl. Med. 1, 6ra14.

Molecular Cell 64, 982–992, December 1, 2016 991


Vijay-Kumar, M., Aitken, J.D., Carvalho, F.A., Cullender, T.C., Mwangi, S., Wellen, K.E., Hatzivassiliou, G., Sachdeva, U.M., Bui, T.V., Cross, J.R., and
Srinivasan, S., Sitaraman, S.V., Knight, R., Ley, R.E., and Gewirtz, A.T. Thompson, C.B. (2009). ATP-citrate lyase links cellular metabolism to histone
(2010). Metabolic syndrome and altered gut microbiota in mice lacking Toll- acetylation. Science 324, 1076–1080.
like receptor 5. Science 328, 228–231. Xiong, Y., Miyamoto, N., Shibata, K., Valasek, M.A., Motoike, T., Kedzierski,
Wang, Z., Klipfell, E., Bennett, B.J., Koeth, R., Levison, B.S., DuGar, B., Feldstein, R.M., and Yanagisawa, M. (2004). Short-chain fatty acids stimulate leptin pro-
A.E., Britt, E.B., Fu, X., Chung, Y.-M., et al. (2011). Gut flora metabolism of duction in adipocytes through the G protein-coupled receptor GPR41. Proc
phosphatidylcholine promotes cardiovascular disease. Nature 472, 57–63. Natl Acad Sci USA 101, 1045–1050.

992 Molecular Cell 64, 982–992, December 1, 2016

You might also like