lactobacillus gasseri in the upper small intestine impacts

23
Article Lactobacillus gasseri in the Upper Small Intestine Impacts an ACSL3-Dependent Fatty Acid-Sensing Pathway Regulating Whole-Body Glucose Homeostasis Graphical Abstract Highlights d Upper small intestinal fatty acid-ACSL3 sensing impacts glucose homeostasis d HFD decreases Lactobacillus gasseri (LG) and disrupts ACSL3-fatty acid sensing d Regular chow microbiota transplant restores LG and ACSL3- lipid sensing d Lactobacillus gasseri administration restores ACSL3-lipid sensing in HFD rodents Authors Paige V. Bauer, Frank A. Duca, T.M. Zaved Waise, ..., Mozhgan Rasti, Catherine A. O’Brien, Tony K.T. Lam Correspondence [email protected] In Brief Bauer et al. report that a glucoregulatory pre-absorptive ACSL3-dependent fatty acid-sensing pathway in the upper small intestine is compromised by a high-fat diet. Fatty acid sensing and glucose homeostasis are restored by probiotic Lactobacillus gasseri administration or by transplantation of microbiota from chow- fed animals. Bauer et al., 2018, Cell Metabolism 27, 572–587 March 6, 2018 ª 2018 Elsevier Inc. https://doi.org/10.1016/j.cmet.2018.01.013

Upload: others

Post on 28-Jul-2022

1 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: Lactobacillus gasseri in the Upper Small Intestine Impacts

Article

Lactobacillus gasseri in the

Upper Small IntestineImpacts an ACSL3-Dependent Fatty Acid-SensingPathway RegulatingWhole-Body Glucose Homeostasis

Graphical Abstract

Highlights

d Upper small intestinal fatty acid-ACSL3 sensing impacts

glucose homeostasis

d HFD decreases Lactobacillus gasseri (LG) and disrupts

ACSL3-fatty acid sensing

d Regular chow microbiota transplant restores LG and ACSL3-

lipid sensing

d Lactobacillus gasseri administration restores ACSL3-lipid

sensing in HFD rodents

Bauer et al., 2018, Cell Metabolism 27, 572–587March 6, 2018 ª 2018 Elsevier Inc.https://doi.org/10.1016/j.cmet.2018.01.013

Authors

Paige V. Bauer, Frank A. Duca,

T.M. Zaved Waise, ..., Mozhgan Rasti,

Catherine A. O’Brien, Tony K.T. Lam

[email protected]

In Brief

Bauer et al. report that a glucoregulatory

pre-absorptive ACSL3-dependent fatty

acid-sensing pathway in the upper small

intestine is compromised by a high-fat

diet. Fatty acid sensing and glucose

homeostasis are restored by probiotic

Lactobacillus gasseri administration or by

transplantation of microbiota from chow-

fed animals.

Page 2: Lactobacillus gasseri in the Upper Small Intestine Impacts

Cell Metabolism

Article

Lactobacillus gasseri in the Upper Small IntestineImpacts an ACSL3-Dependent Fatty Acid-SensingPathwayRegulatingWhole-BodyGlucoseHomeostasisPaige V. Bauer,1,2 Frank A. Duca,1 T.M. Zaved Waise,1 Helen J. Dranse,1 Brittany A. Rasmussen,1,2 Akshita Puri,4

Mozhgan Rasti,1 Catherine A. O’Brien,2,4,5 and Tony K.T. Lam1,2,3,6,7,*1Toronto General Hospital Research Institute, UHN, MaRS Centre, Toronto Medical Discovery Tower, Room 10-705, 101 College Street,

Toronto, ON M5G 1L7, Canada2Department of Physiology, University of Toronto, Toronto, ON M5S 1A8, Canada3Department of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada4Princess Margaret Cancer Centre, UHN, Toronto, ON M5G 2M9, Canada5Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, ON M5S 1A8, Canada6Banting and Best Diabetes Centre, University of Toronto, Toronto, ON M5G 2C4, Canada7Lead Contact

*Correspondence: [email protected]

https://doi.org/10.1016/j.cmet.2018.01.013

SUMMARY

Long-chain acyl-CoA synthetase (ACSL)-dependentupper small intestinal lipid metabolism activatespre-absorptive pathways to regulate metabolic ho-meostasis, but whether changes in the upper smallintestinal microbiota alter specific fatty acid-depen-dent pathways to impact glucose homeostasis re-mains unknown. We here first find that upper smallintestinal infusion of Intralipid, oleic acid, or linoleicacid pre-absorptively increases glucose toleranceand lowers glucose production in rodents. High-fatfeeding impairs pre-absorptive fatty acid sensingand reduces upper small intestinal Lactobacillusgasseri levels and ACSL3 expression. Transplanta-tion of healthy upper small intestinal microbiota tohigh-fat-fed rodents restores L. gasseri levels andfatty acid sensing via increased ACSL3 expression,while L. gasseri probiotic administration to non-transplanted high-fat-fed rodents is sufficient torestore upper small intestinal ACSL3 expressionand fatty acid sensing. In summary, we unveil a glu-coregulatory role of upper small intestinal L. gasserithat impacts an ACSL3-dependent glucoregulatoryfatty acid-sensing pathway.

INTRODUCTION

Glucose intolerance is a risk factor for type 2 diabetes (Roglic

and World Health Organization, 2016). An elevation of glucose

production (GP) by the liver contributes to glucose intolerance

and is the result of impaired insulin secretion and action, as

well as defects in glucose effectiveness and nutrient-sensing

pathways in the gut and brain (Defronzo, 2009; Lam, 2010; Ros-

setti et al., 1990). Metformin and bariatric surgery are effective

therapies that improve glucose tolerance by lowering GP in dia-

572 Cell Metabolism 27, 572–587, March 6, 2018 ª 2018 Elsevier Inc

betes (Batterham andCummings, 2016; Hundal et al., 2000), and

the mechanisms are linked to a small intestinal nutrient sensing-

dependent neuronal network (Breen et al., 2012; Duca et al.,

2015b; Jiao et al., 2013). Metformin and bariatric surgery also

changes gut microbiota (Forslund et al., 2015; Tremaroli et al.,

2015;Wu et al., 2017), suggesting themicrobiota interact with in-

testinal nutrient sensing to impact glucose homeostasis.

The gut microbiota rapidly changes in response to a high-fat

diet (HFD) (David et al., 2014; Turnbaugh et al., 2009), and themi-

crobiota regulates body weight and adiposity (Backhed et al.,

2004; Ridaura et al., 2013; Turnbaugh et al., 2006). Despite prog-

ress in identifying functional and compositional alterations in the

fecal metagenome of type 2 diabetic individuals (Karlsson et al.,

2013; Qin et al., 2012), most studies have overlooked the role of

the microbiota residing in the upper small intestine, an important

site for nutrient sensing and metabolic regulation (Duca et al.,

2015a). For example, the transfer of feces from healthy individ-

uals to individuals with metabolic syndrome via a duodenal cath-

eter alters the fecal microbiota of recipients in association with

improvements in insulin sensitivity (Vrieze et al., 2012). This mi-

crobiota transfer could alter the microbial community in the up-

per small intestine, contributing to this insulin-sensitizing effect.

Similarly, pre- and probiotics improve glucose regulation and

robustly alter distal intestinal and fecal microbiota of rodents

and humans (Balakumar et al., 2016; Cani et al., 2006a; Ejtahed

et al., 2012; Everard andCani, 2013); however, these substances

must first pass the upper small intestine where they could elicit

their glucoregulatory effects. This is in line with the fact that

most probiotics are derived from Lactobacillus (Asemi et al.,

2013; Balakumar et al., 2016; Ejtahed et al., 2012; Yadav et al.,

2007), which is a prominent inhabitor of the small intestine (Gu

et al., 2013; Wirth et al., 2014).

The upper small intestine contacts ingested nutrients that

initiate negative feedback pathways to regulate glucose homeo-

stasis (Duca et al., 2015a). For example, lipid sensing (achieved

via Intralipid infusion) in the upper small intestine activates a gut-

brain axis to lower GP and plasma glucose levels (Wang et al.,

2008). This effect is dependent upon formation of long-chain

fatty acyl-coenzyme A (CoA) via long-chain acyl-CoA synthetase

.

Page 3: Lactobacillus gasseri in the Upper Small Intestine Impacts

Figure 1. Upper Small Intestinal Infusion of Intralipid, Oleic Acid, or Linoleic Acid, Increases Glucose Tolerance in RC but Not HFD Rodents

(A) Experimental procedure and intravenous glucose tolerance test (i.v.GTT) protocol.

(B–G) Percent change in plasma glucose levels (inset: integrated area under the curve [AUC]) (B, D, and F) and absolute plasma insulin levels (C, E, and G) during

the i.v.GTT in regular chow rats that received an upper small intestinal infusion of saline (n = 6), Intralipid (n = 6), PBS with bile salts (n = 6), oleic acid (n = 8), or

linoleic acid (n = 6).

(H–J) Percent change in plasma glucose levels (inset: integrated AUC) during the i.v.GTT in 3-day high-fat diet rats that received an upper small intestinal infusion

of saline (n = 7), Intralipid (n = 6) (H), PBS + bile salts (n = 6), oleic acid (n = 6) (I), or linoleic acid (n = 6) (J).

*p < 0.05, **p < 0.01, ***p < 0.001 determined by t test. Data are shown as the mean ± SEM. See also Table S1 and Figure S1. i.v.GTT, intravenous glucose

tolerance test; AUC, area under the curve; HFD, high-fat diet.

(ACSL), as general inhibition of ACSL isoforms in the upper small

intestine negates lipid sensing (Wang et al., 2008). This glucore-

gulatory lipid-sensing pathway is abolished in HFD rodents

(Cheung et al., 2009), but whether HFD feeding affects upper

small intestinal ACSL expression via changes in microbiota,

and whether this postulated change in ACSL expression affects

lipid sensing, is unknown. Interestingly, inoculation of germ-free

mice with the microbiota of healthy conventionally raised mice

alters the expression of hundreds of genes involved in glucose

and lipid metabolism in the upper and distal gut (Derrien et al.,

2011; El Aidy et al., 2013), including ACSL expression (El Aidy

et al., 2013), suggesting that the microbiota could alter intestinal

ACSL expression. Based on these findings, we tested the hy-

pothesis that upper small intestinal microbiota alter upper small

intestinal ACSL-dependent glucoregulatory fatty acid-sensing

pathways in rodents.

RESULTS

Upper Small Intestinal Fatty Acid-Sensing MechanismsIncrease Glucose Tolerance and Lower GP in Healthybut Not High-Fat-Fed RodentsTo begin addressing the potential interaction between the upper

small intestinal microbiota and glucoregulatory lipid-sensing

pathways, we first investigated whether gut lipid sensing regu-

lates whole-body glucose tolerance under physiological condi-

tions. Intralipid (lipid emulsion) was infused for a total of 50 min

into the upper small intestine of conscious, unrestrained, regular

chow (RC)-fed rats to activate pre-absorptive lipid-sensing path-

ways as described previously (Wang et al., 2008), and glucose

tolerance was assessed with an intravenous glucose toler-

ance test (i.v.GTT) (Figure 1A). Intralipid versus saline infusion

increased glucose tolerance (Figures 1B and S1A) independently

of a rise in plasma insulin levels (Figure 1C). To examine the effect

of individual fatty acids within Intralipid, oleic acid or linoleic acid

(prevalent fatty acids found in human circulation) was dissolved

in PBS with bile salts (containing 0.1% [wt/vol] lecithin and 1%

[wt/vol] sodium taurocholate) and infused for a total of 50 min

into the upper small intestine during the i.v.GTT. Both oleic

acid and linoleic acid improved glucose tolerance (Figures 1D,

1F, S1B, and S1C) independently of a rise in plasma insulin levels

(Figures 1E and 1G) when compared with an infusion of PBSwith

bile salts alone. Next, surgically recovered rats were given a

saturated fat-enriched HFD (Table S1) for 3 days, which has pre-

viously been shown to induce hyperphagia and hepatic insulin

resistance in rodents (Cote et al., 2015), before being subjected

to the i.v.GTTwith a gut infusion of Intralipid, oleic acid, or linoleic

acid. Rats exhibited hyperphagia in response to the HFD diet

(Figure S1D) but had comparable body weight (Figure S1E),

which is consistent with reports documenting that 3-day

Cell Metabolism 27, 572–587, March 6, 2018 573

Page 4: Lactobacillus gasseri in the Upper Small Intestine Impacts

Figure 2. Upper Small Intestinal Oleic Acid or Linoleic Acid Infusion Lowers GP through a Gut Peptide-Dependent Neuronal Network in RC

but Not HFD Rodents

(A) Experimental procedure for pancreatic (basal insulin)-euglycemic clamp protocol.

(B–D) The glucose infusion rate (B), glucose production (C), and glucose uptake (D) during the pancreatic clamps of healthy rats that received an upper small

intestinal infusion of saline (n = 12), PBS with bile salts (n = 7), oleic acid (n = 11), or linoleic acid (n = 7). **p < 0.01 versus saline and PBS with bile salts as

determined by ANOVA with Tukey’s post hoc test.

(E) Glucose production during clamps of healthy rats that received an upper small intestinal infusion of MK-329 (n = 7), Exendin-9 (n = 5), MK-329 + oleic acid

(n = 5), Exendin-9 + oleic acid (n = 6), MK-329 + linoleic acid (n = 5), or Exendin-9 + linoleic acid (n = 5). **p < 0.01 versus all other groups as determined by ANOVA

with Tukey’s post hoc test.

(F) Glucose production during clamps of healthy rats that received an upper small intestinal infusion of tetracaine (n = 5), tetracaine + oleic acid (n = 5), and

tetracaine + linoleic acid (n = 5).

(G) Glucose production during clamps in 3-day high-fat diet-fed rats that received an upper small intestinal infusion of saline (n = 5), oleic acid (n = 5), and linoleic

acid (n = 5). Values are shown asmean + SEM. See also Tables S1, S2, and Figure S2. SRIF, somatostatin; basal, the average GP over the period from 60 to 90min

during the pancreatic (basal insulin)-euglycemic clamp; clamp, the average GP from 180 to 200 min; Ex-9, exendin-9.

HFD-induced hyperphagia induces insulin and leptin resistance

but does not increase body weight in rodents (Morgan et al.,

2004; Scherer et al., 2012; Thaler et al., 2012). We found that up-

per small intestinal infusion of Intralipid, oleic acid, and linoleic

acid failed to improve glucose tolerance (Figures 1H, 1I, 1J,

S1F, S1G, and S1H) in HFD rats at comparable plasma insulin

levels (Figures S1I, S1J, and S1K). Thus, upper small intestinal

lipid (specifically oleic acid and linoleic acid) sensing, improves

glucose tolerance in healthy but not HFD rodents.

To determine whether upper small intestinal fatty acid-sensing

regulates glucose tolerance via gut-mediated changes in GP or

glucose uptake independent of changes in plasma insulin levels

(Table S2), oleic acid or linoleic acid were infused for 50 min

into the upper small intestine and changes in glucose kinetics

were evaluated via tracer dilution methodology under pancreatic

basal insulin-euglycemic clamp conditions (Figure 2A; Table S2).

During the oleic acid and linoleic acid gut infusion, the exoge-

574 Cell Metabolism 27, 572–587, March 6, 2018

nous glucose infusion rate increased by �5-fold relative to the

vehicle infusion to prevent a drop in plasma glucose levels and

to maintain euglycemia (Figure 2B; Table S2). This glucose-

lowering effect was due to a reduction in GP (Figure 2C) rather

than an increase in glucose uptake (Figure 2D). Of note, there

was no change in plasma or portal free fatty acid levels at the

end of the 50 min gut infusion relative to the pre-infused basal

condition, as well as between treatment groups (Figure S2A),

ensuring the effects seen throughout the clamp and i.v.GTT

(Figures 1 and S1) are due to pre-absorptive fatty acid sensing

in healthy rats. In addition, the ability of oleic acid and linoleic

acid to lower GP is conserved in healthy mice during the basal

insulin pancreatic clamp (Figures S2B–S2E). Together with the

fact that upper small intestinal Intralipid infusion lowers GP

(Wang et al., 2008), our data collectively indicate that upper small

intestinal fatty acid sensing increases glucose tolerance at least

in part by lowering GP in rats and mice in vivo.

Page 5: Lactobacillus gasseri in the Upper Small Intestine Impacts

Increased consumption of oleic acid and linoleic acid has

beneficial glucoregulatory effects in rodents and humans (Ris-

erus et al., 2009); however, the mechanisms involved have

beendebated. Although Intralipid infusion into the upper small in-

testine activates a CCK-1 receptor-dependent pathway to lower

GP (Cheung et al., 2009), oleic acid stimulates the release of both

CCK (Chang et al., 2000) and GLP-1 (Iakoubov et al., 2007)

in vitro. While CCK appears to mediate the satiety response to

a duodenal infusion of oleic acid (Woltman and Reidelberger,

1995), the glucoregulatory effects of diets high in oleic acid

havebeenattributed toGLP-1action (Rocca et al., 2001). Linoleic

acid also stimulates the release of CCK (Shah et al., 2012) and

GLP-1 (Richards et al., 2016) in vitro; however, intestinal linoleic

acid infusion lowers food intake through a GLP-1-dependent

mechanism (Dailey et al., 2011). Thus, to evaluate whether a

gut CCK- and/or GLP-1-dependent pathway is necessary for up-

per small intestinal fatty acid sensing, each fatty acid was co-

infused with the CCK-1 receptor inhibitor MK-329 or the GLP-1

receptor (GLP-1R) inhibitor Exendin-9. Co-infusion of oleic acid

with either MK-329 or Exendin-9 abolished the ability of an oleic

acid 50 min infusion to increase the glucose infusion rate (Fig-

ure S2F) and decrease GP (Figure 2E) with no change in glucose

uptake (Figure S2G) during the pancreatic clamp, suggesting

(although warrants future investigation) that oleic acid-induced

CCK-1 receptor and GLP-1R effect is additive in the current

experimental context. In contrast, co-infusion of MK-329 with li-

noleic acid did not influence the ability of linoleic acid to decrease

GP (Figures 2E, S2F, and S2G) compared with MK-329 alone

(Figures 2E, S2F, and S2G). However, co-infusion of Exendin-9

with linoleic acid negated the ability of linoleic acid to regulate

glucose metabolism compared with Exendin-9 alone (Figures

2E, S2F, and S2G). Therefore, oleic acid activates CCK-1- and

GLP-1R-dependent pathways, while linoleic acid stimulates a

CCK-1-independent and GLP-1R-dependent pathway in the up-

per small intestine to lower GP in healthy rats. The underlying

mechanisms responsible for the oleic acid- versus linoleic acid-

induced differential effect on the gut peptide-dependent glucor-

egulatory pathways warrant future investigation.

To determine whether a downstream neuronal pathway is

necessary for oleic acid and linoleic acid sensing, each fatty

acid was co-infused into the upper small intestine with the local

anesthetic, tetracaine. Infusion of tetracaine alone had no meta-

bolic consequences but negated the ability of both fatty acids to

increase the glucose infusion rate (Figure S2H) and lower GP

(Figure 2F) with no effect on glucose uptake (Figure S2I), indi-

cating that a gut-brain neuronal network is required for the

suppressive effects of these fatty acids on GP. Taken together,

oleic and linoleic acids trigger a common GLP-1R-dependent

neuronal network to lower GP in healthy rats.

To address the pathological relevance of these GP-lowering

pathways, oleic acid or linoleic acid were infused into the upper

small intestine of HFD rats. These HFD rats were hyperphagic

relative to their RC counterparts (Figure S2J), but were not obese

(Figure S2K). During the pancreatic clamps, oleic acid and

linoleic acid failed to increase the glucose infusion rate (Fig-

ure S2L) and lower GP (Figure 2G) compared with RC-fed rats,

while glucose uptake remained unaltered (Figure S2M). Taken

together with the i.v.GTT data, we have validated, via three

different fatty acid treatments using two complementary tests,

that an HFD disrupts specific upper small intestinal fatty acid-

sensing pathways, leading to a dysregulation of GP and glucose

tolerance.

Upper Small Intestinal Microbiota Transplantation fromRC-Fed Donors Normalizes the Upper Small IntestinalMicrobiota in HFD Recipient RatsA potential link between HFD and impaired upper small intestinal

fatty acid sensing could be diet-induced changes in the gut mi-

crobiota. Indeed, the gut microbiota rapidly changes in response

to an HFD (David et al., 2014; Turnbaugh et al., 2009), and

increasing evidence suggests the gut microbiota plays an impor-

tant role in glucose homeostasis (Kovatcheva-Datchary et al.,

2015; Plovier et al., 2017; Shin et al., 2014; Vrieze et al., 2012;

Wu et al., 2017), as a recent study indicates metformin alters mi-

crobiota composition in the upper small intestine to impact a

glucose-sensing mechanism that regulates GP as well (Bauer

et al., 2018). More importantly, conventionalization of germ-

free mice alters the expression of proteins involved in upper

small intestinal lipid sensing, such as CD36, FABP4, and ACSL

(Derrien et al., 2011; El Aidy et al., 2013). Thus, we investigated

whether an upper small intestinal microbiota transplant from

RC-fed donors could normalize the upper small intestinal micro-

biota in HFD-recipient rats, and whether the change in upper

small intestinal microbiota of transplanted HFD rats could rescue

fatty acid-sensing mechanisms.

We first characterized themicrobiota composition in the upper

small intestine of RC and HFD rats, as well as HFD rats receiving

an upper small intestinal RCmicrobiota transplant. To ensure the

microbiota transplants were targeted to the upper small intes-

tine, upper small intestinal luminal contents of the donor rats

were transplanted into recipient rats by slowly administering

the transplanted material over the course of 30 s via the upper

small intestinal cannula. RC, HFD, and HFD rats receiving an

upper small intestinal RC microbiota transplant received a gut

saline infusion and were sacrificed following the pancreatic

clamp studies. The luminal contents were collected from the up-

per small intestine (between 6 and 15 cm distal to the pyloric

sphincter), and the variable region 3 (V3) of the bacterial 16S

rRNA gene was amplified by PCR and sequenced using an Illu-

mina MiSeq platform (Table S3). Of note, the luminal contents

used for bacterial transplantation and sequencing were from

the same region of the small intestine.

Principal coordinate analysis of weighted UniFrac distance

between the upper small intestinal samples from each group

showed a clear separation between RC and HFD communities

(Figure 3A), while samples from HFD rats that received an upper

small intestinal RCmicrobiota transplant clustered with RC sam-

ples and were well separated from HFD samples (Figure 3A).

Analysis at the phylum level indicated that the upper small intes-

tinal microbiota in all three groups was dominated by two phyla:

Firmicutes and Proteobacteria (Figure 3B). The high abundance

of Firmicutes in these groups (especially in the RC or the HFD

rats transplanted with RC microbiota) is consistent with studies

characterizing the upper small intestinal microbiota in RC mice

(Gu et al., 2013) and RC Wistar rats (Wirth et al., 2014), as well

as in the cecum and feces of RC Sprague-Dawley rats (Di Luccia

et al., 2015; Liu et al., 2015). HFD increased the relative abun-

dance of Proteobacteria and reduced relative abundance of

Cell Metabolism 27, 572–587, March 6, 2018 575

Page 6: Lactobacillus gasseri in the Upper Small Intestine Impacts

Figure 3. HFD-Induced Changes in the Upper Small Intestinal Microbiota Can Be Reversed with a Microbiota Transplant from RC Rats

(A) Principal coordinate analysis of weighted UniFrac displays separation between samples from RC rats, HFD rats, and HFD rats that received an upper small

intestinal RC microbiota transplant (HFDwRCM). The percentage of variation explained by the plotted principal coordinates is indicated in the axis labels. Each

dot represents an upper small intestinal community from one rat.

(B) Relative abundance at the phylum level in the upper small intestinal community of RC, HFD, and HFDwRCM rats. Each column corresponds to one sample.

(C) Relative abundance of families that are significantly altered by HFD feeding and restored with the upper small intestinal RC microbiota transplant (expressed

as a percent of the total upper small intestinal community). **p < 0.01, ***p < 0.001HFD versus all other groups, as assessed by ANOVAwith Tukey’s post hoc test.

Values are shown as the mean + SEM.

(D) Relative abundance at the genus level in the upper small intestinal community of RC, HFD, or HFDwRCM rats. Each column corresponds to one sample.

(E) Heatmap of the relative abundance of species from the Lactobacillus genus. Each row corresponds to one sample. **p < 0.01 HFD versus all other groups, as

assessed by ANOVAwith Tukey’s post hoc test. See also Table S3 and Figure S3. RC, regular chow (n = 6); HFD, high-fat diet (n = 6); HFDwRCM, high-fat diet with

RC microbiota transplant (n = 5).

Firmicutes compared with RC rats (Figure 3B and Table S3, as

identified by LefSE and assessed by ANOVA). At the family level,

HFD versus RC reduced Lactobacillaceae and modestly yet

significantly increased Clostridiaceae (both Lactobacillaceace

and Clostridiaceae are from the Firmicutes Phylum) (Figure 3C;

Table S3). At the genus level, and consistent with previous liter-

ature (Almiron et al., 2013), the upper small intestinal microbiota

of RC rats was dominated by Lactobacillus (Figure 3D; Table S3).

However, the abundance of Lactobacillus was reduced in

response to HFD (Figure 3D; Table S3). Importantly, analysis

(as identified by LefSE and assessed by ANOVA) of the upper

small intestinal luminal contents of HFD rats transplanted with

576 Cell Metabolism 27, 572–587, March 6, 2018

RC microbiota revealed that HFD-induced changes in the upper

small intestinal microbiota at the phylum (Firmicutes and Proteo-

bacteria) (Figure 3B; Table S3), family (Lactobacillaceae and

Clostridiaceae) (Figure 3C; Table S3), and genus (Lactobacillus)

(Figure 3D; Table S3) levels were all reversed to RC conditions

with transplantation of upper small intestinal RC microbiota.

We next performed a detailed characterization of the species

within the Lactobacillus genus and found that HFD versus

RC reduced the abundance of Lactobacillus (L.) gasseri and

increased L. animalis, while the effect of HFD on L. gasseri and

L. animalis were abolished in HFD rats transplanted with upper

small intestinal RC microbiota (Figure 3E; Table S3). Of note,

Page 7: Lactobacillus gasseri in the Upper Small Intestine Impacts

Figure 4. Transplantation of Upper Small Intestinal RCMicrobiota Restores Fatty Acid-Sensing Pathways that IncreaseGlucose Tolerance inHFD Rodents

(A) Experimental procedure of the microbiota transplant protocol.

(B andC) Percent change in plasma glucose levels (inset: integrated AUC) (B) and absolute plasma insulin levels (C) during the i.v.GTT in HFD rats that received an

RC upper small intestinal microbiota transplant and an upper small intestinal saline (n = 6) or Intralipid (n = 6) infusion and HFD rats that received an HFD upper

small intestinal microbiota transplant and an upper small intestinal saline (n = 6) or Intralipid (n = 6) infusion.

(D) Percent change in plasma glucose levels (inset: integrated AUC) during the i.v.GTT in HFD rats that received heat-shock (n = 6) or filtered (n = 5) RC upper small

intestinal microbiota and an upper small intestinal Intralipid infusion.

(E) Percent change in plasma glucose levels (inset: integrated AUC) during the i.v.GTT in HFD rats that received a colonic RCmicrobiota transplant and an upper

small intestinal saline (n = 5) or Intralipid (n = 5) infusion.

*p < 0.05, **p < 0.01 HFD + RCM + Intralipid versus all other groups, as assessed by ANOVA with Tukey’s post hoc test. Values are shown as mean ± SEM. See

also Figure S4. IVGTT, intravenous glucose tolerance test; RC, regular chow; HFD, high-fat diet.

all changes in the microbiota among the three groups occurred

in the context of comparable bacterial diversity (Figure S3).

Thus, HFD-induced changes in upper small intestinal microbiota

composition can be reversed in HFD rats with a direct upper

small intestinal RC microbiota transplant.

Upper Small Intestinal Microbiota Transplantation fromRC-Fed Donors Restores Upper Small Intestinal FattyAcid-Sensing Mechanisms in HFD-Recipient RatsGiven that an HFD disrupts selective upper small intestinal fatty

acid-sensing pathways and glucose homeostasis (Figures 1

and 2) in parallel to changes in upper small intestinal microbiota

composition (Figure 3), and that an upper small intestinal RC mi-

crobiota transplant is sufficient to normalize upper small intesti-

nal microbiota in HFD rats (Figure 3), we next assessed the glu-

coregulatory relationship between the gut microbiota and fatty

acid-sensing mechanisms in the transplanted rats (Figure 4A).

We found that upper small intestinal infusion of Intralipid versus

saline improved glucose tolerance in HFD rats that received an

upper small intestinal RCmicrobiota transplant (Figure 4B), inde-

pendent of changes in plasma insulin levels (Figure 4C), and this

improvement in glucose tolerance recapitulated the gut lipid-

sensing effect seen in RC rats without a transplant (Figures 1B

and S1A). To ensure the transplant procedure per se did not

improve glucose tolerance, the microbiota of HFD rats was

transferred to HFD recipients and we found that gut Intralipid

versus saline failed to improve glucose tolerance in these rats

(Figure 4B). These data demonstrate that RCmicrobiota restores

lipid sensing to regulate glucose homeostasis in HFD rats. To

ensure the detected changes in gut lipid sensing were due to

the transfer of microbiota rather than metabolites found within

the luminal contents, the upper small intestinal microbiota was

either heat killed (Giannakis et al., 2009; Kovatcheva-Datchary

et al., 2015) or removed via filtering (Gareau et al., 2011) before

the transplant. Transfer of heat-killed or filtered luminal contents

of RC rats failed to restore gut lipid sensing in HFD-recipient

rats (Figure 4D), illustrating that restoration of upper small intes-

tinal lipid sensing in HFD rats transplanted with RC upper small

intestinal luminal contents is dependent upon the transfer of

live bacteria.

Changes in the distal intestinal microbiota correlate with

improved glucose regulation following microbiota manipulation

with pre- and probiotics (Balakumar et al., 2016; Cani et al.,

2006b; Ejtahed et al., 2012). Therefore, to assess whether

changes in the distal gut microbiota are responsible for the

improvements in upper small intestinal glucoregulatory lipid-

sensing pathways following upper small intestinal RCmicrobiota

transplantation, we transplanted the upper small intestinal mi-

crobiota of healthy RC rats directly into the colon of HFD rats

that will receive upper small intestinal Intralipid infusion during

the i.v.GTT the following day. Intralipid versus saline infusion

failed to improve glucose tolerance in HFD rats receiving a colon

RCmicrobiota transplant (Figure 4E), indicating the restoration of

upper small intestinal lipid sensing in HFD rats transplanted with

upper small intestinal RC microbiota is reliant on the normaliza-

tion of the upper small intestinal microbiota, and not the micro-

biota in the lower intestinal tract.

To further address whether HFDmicrobiota per se is sufficient

to disrupt glucoregulatory lipid-sensing pathways, upper small

intestinal microbiota obtained from 3-day HFD rats was trans-

planted into RC rats receiving gut saline or Intralipid infusion dur-

ing the i.v.GTT (Figure S4A). Intralipid failed to improve glucose

tolerance relative to a saline infusion in these rats (Figure S4B),

independent of changes in plasma insulin levels (Figure S4C),

while upper small intestinal Intralipid versus saline infusion for

50 min improved glucose tolerance in RC rats transplanted

Cell Metabolism 27, 572–587, March 6, 2018 577

Page 8: Lactobacillus gasseri in the Upper Small Intestine Impacts

Figure 5. Upper Small Intestinal Microbiota Alters Fatty Acid-Sensing Pathways that Impact GP Regulation

(A–C) The glucose infusion rate (A), glucose production (B), and glucose uptake (C) during the clamps with an oleic acid upper small intestinal infusion in HFD rats

that received an HFD microbiota transplant (n = 6), HFD rats that received an RC microbiota transplant (n = 6), RC rats that received an RC microbiota transplant

(n = 6), and RC rats that received an HFD microbiota transplant (n = 6).

(D–F) The glucose infusion rate (D), glucose production (E), and glucose uptake (F) during the clamp with a linoleic acid upper small intestinal infusion in HFD rats

that received an HFD microbiota transplant (n = 6), HFD rats that received an RC microbiota transplant (n = 6), RC rats that received an RC microbiota transplant

(n = 6), and RC rats that received an HFD microbiota transplant (n = 6).

**p < 0.01 versus RC + HFDM and HFD + HFDM rats, as determined by ANOVA with Tukey’s post hoc test. Values are shown as mean + SEM. RC, regular chow;

HFD, high-fat diet; basal, the average glucose production over the period of 60–90 min during the pancreatic (basal insulin)-euglycemic clamp; clamp, the

average glucose production from 180 to 200 min.

with RCmicrobiota (Figure S4B), and this elevation was identical

to what was seen in RC rats without a transplant receiving

an Intralipid infusion (Figures 1B and S1A). Heat-killed or filtered

luminal contents of HFD rats failed to impair gut lipid sensing in

RC-recipient rats (Figure S4D). Taken together, changes in the

upper small intestinal microbiota alter lipid-sensing-dependent

pathways to impact glucose homeostasis.

To next examine whether bidirectional changes in the upper

small intestinal microbiota impact a selective glucoregulatory

fatty acid-sensing-dependent pathway, microbiota transplant

studies were repeated in rats that underwent the gut fatty acid

infusion-pancreatic clamp studies the day after transplantation

(Figure 4A). We discovered that RC rats receiving an HFDmicro-

578 Cell Metabolism 27, 572–587, March 6, 2018

biota transplant exhibited impaired fatty acid sensing, as oleic

acid and linoleic acid failed to increase the glucose infusion

rate (Figures 5A and 5D) and lower GP (Figures 5B and 5E) rela-

tive to RC rats receiving an RC microbiota transplant. HFD rats

receiving an RC microbiota transplant responded to oleic acid

and linoleic acid infusion, leading to an increased glucose infu-

sion rate (Figures 5A and 5D) and a suppression of GP (Figures

5B and 5E) compared with the disruption in fatty acid sensing

seen in HFD rats receiving an HFD microbiota transplant.

Glucose uptake was comparable among groups (Figures 5C

and 5F).

Thus, an RCmicrobiota transplant restores themicrobiota to a

healthy condition in HFD rats, leading to the restoration of an

Page 9: Lactobacillus gasseri in the Upper Small Intestine Impacts

upper small intestinal glucoregulatory fatty acid-sensing-depen-

dent pathway.

Upper Small Intestinal Microbiota Alters ACSL3-Dependent Glucoregulatory Fatty Acid-SensingPathwayThe gut microbiota alters the expression of genes involved in

lipid metabolism in the upper and lower gut (Derrien et al.,

2011; El Aidy et al., 2013). For example, inoculation of germ-

free mice alters jejunal expression of ACSL (El Aidy et al.,

2013). Interestingly, HFD feeding lowers hepatic protein expres-

sion of the ACSL isoforms ACSL3 and ACSL4 (Bowman et al.,

2016); however, whether this effect occurs in the upper small

intestine and whether it is dependent upon diet-induced

changes in the upper small intestinal microbiota has not been

investigated. Further, whether these postulated gut microbiota-

induced changes in ACSL expression impact lipid sensing and

subsequent glucose regulation requires consideration. There

are five members of the ACSL gene family, numbered 1, 3,

4, 5, and 6 (Soupene and Kuypers, 2008). Studies exploring

mRNA expression of the genes encoding the five ACSLs in

various rat tissues found that all ACSL isoforms are expressed

in the small intestine (Bowman et al., 2016; Mashek et al.,

2006), with ACSL3 and ACSL5 being the most highly expressed

isoforms (Bowman et al., 2016). However, the protein expression

for the five ACSL isoforms has not yet been examined in the

upper small intestinal mucosa. Therefore, to begin evaluating

whether HFD-induced changes in the upper small intestinal mi-

crobiota alter upper small intestinal fatty acid-sensing pathways

via changes in ACSL expression, we first sought to determine

which isoforms are expressed in the upper small intestinal mu-

cosa and whether protein expression is altered in response to

an HFD with or without an RC microbiota transplant.

The upper small intestinal mucosa of RC, HFD, and HFD rats

with RCmicrobiota transplant was collected following the clamp

procedure. First, ACSL1, 4, 5, and 6 were detected in the upper

small intestine using brain tissue as a positive control (Figures

S5A–S5D). Expression of ACSL1, 4, 5, and 6was not significantly

altered in HFD versus RC rats (Figures S5A–S5D). In addition, the

expression of these ACSL isoforms in HFD rats that received an

upper small intestinal RC microbiota transplant was comparable

with both RC and HFD rats (Figures S5A–S5D). ACSL3 was also

detected in the upper small intestinal mucosa using brain tissue

as a positive control and heart as a negative control (Figure 6A).

However, HFD versus RC feeding consistently and significantly

reduced ACSL3 protein expression in the upper small intestinal

mucosa (Figure 6A), and this reduction was restored with an

upper small intestinal RC microbiota transplant in HFD rats

(Figure 6A).

To next investigate whether the upper small intestinal RC

microbiota transplant restores lipid sensing via upregulation of

ACSL3, we prevented ACSL3 upregulation via injection of lenti-

virus expressing either ACSL3 small hairpin RNA (shRNA) (LV-

ACSL3 shRNA) or mismatch (LV-MM). LV-ACSL3 shRNA or

LV-MM was injected into the upper small intestine 3 days prior

to the upper small intestinal RCmicrobiota transplant procedure.

The recipient rats underwent the pancreatic clamp studies the

following day. A comparable lentiviral shRNA injection protocol

has been demonstrated to knock down protein specifically in

the upper small intestine (Cote et al., 2015). We found that upper

small intestinal lipid versus saline infusion in LV-MM-injected

HFD rats with an RC upper small intestinal microbiota transplant

increased the glucose infusion rate (Figure S5E) and lowered GP

(Figure 6B), as reported previously for oleic acid and linoleic acid

sensing (Figures 5A, 5B, 5D, and 5E). Importantly, LV-ACSL3

shRNA-injected HFD rats that received an upper small intestinal

RC microbiota transplant did not respond to upper small intesti-

nal lipid infusion, as the glucose infusion rate (Figure S5E) andGP

(Figure 6B) was unaltered when compared with saline-infused

LV-ACSL3 shRNA-injected transplanted rats, while glucose up-

take was comparable among groups (Figure S5F). This inability

of upper small intestinal lipid infusion to lower GP was associ-

ated with a significant 50% reduction in protein levels of upper

small intestinal ACSL3 in LV-ACSL3 shRNA-injected HFD

transplanted rats versus LV-MM-injected HFD transplanted

rats (Figure 6C).

Since oleic acid- and linoleic acid-sensing pathways in the

upper small intestine are dependent upon GLP-1R signaling

in RC rats (Figure 2E), and that upper small intestinal infusion

of Intralipid versus saline also failed to increase glucose infu-

sion rate (Figure S5G) and lower GP (Figure S5H) in LV-

ACSL3 shRNA-injected RC rats, compared with the effects

seen in LV-MM-injected RC rats, with no changes in glucose

uptake (Figure S5I), we next investigated whether the restora-

tion of upper small intestinal ACSL3-dependent Intralipid

sensing is also GLP-1R dependent in HFD rats that received

an upper small intestinal RC microbiota transplant. We found

that, while infusion of Exendin-9 alone had no metabolic conse-

quences in these transplanted rats (Figures 6D, S5J, and S5K),

co-infusion of Intralipid with Exendin-9 completely abolished

the ability of Intralipid to increase the glucose infusion rate

(Figure S5J) and lower GP (Figure 6D) in HFD rats that received

an upper small intestinal RC microbiota transplant, with no

changes in glucose uptake (Figure S5K). Thus, transplantation

of upper small intestinal RC microbiota is sufficient to restore

an upper small intestinal ACSL3- and GLP-1R-dependent

glucoregulatory lipid-sensing pathway in HFD rats. Of interest,

given that inhibition of gut CCK-1 receptor is sufficient to

abolish the GP-lowering effect of upper small intestinal Intrali-

pid infusion as well in non-transplanted healthy rats (Cheung

et al., 2009), we put forward a working hypothesis (although

this clearly warrants future investigation) that Intralipid-induced

CCK-1 receptor and GLP-1R effect is additive in the current

experimental context.

Given that lipid sensing in HFD rats transplanted with RC

microbiota is GLP-1R dependent, and that nutrients stimulate

GLP-1 release via cell depolarization and the opening of

voltage-gated Ca2+ channels (Diakogiannaki et al., 2013; Gribble

et al., 2003; Kuhre et al., 2015; Reimann et al., 2008), we postu-

late that a 50% knock down of ACSL3 (Figure 6C) is sufficient to

prevent the threshold for the opening of Ca2+ channels to be

reached, resulting in an absence of GLP-1 release and an

inability of lipid sensing to exert glucose control in these rats.

This hypothesis is consistent with a study demonstrating that a

�40% knock down of PKCz in the rat ileum inhibits oleic acid-

induced GLP-1 release (Iakoubov et al., 2011), although the

working hypothesis warrants future investigation. Interestingly,

upper small intestinal glucose infusion increases portal active

Cell Metabolism 27, 572–587, March 6, 2018 579

Page 10: Lactobacillus gasseri in the Upper Small Intestine Impacts

Figure 6. Upper Small Intestinal Microbiota Alters an ACSL3-Dependent Fatty Acid-Sensing Pathway to Impact GP Regulation

(A) Protein expression of ACSL3 in the upper small intestinal mucosa of RC rats (n = 5), HFD rats (n = 5), and HFD rats transplanted with RC upper small intestinal

microbiota (n = 5). Brain tissue was used as a positive control; heart was used as a negative control. b-Actin and GAPDHwere used as loading controls as b-actin

is normally not detected in heart tissue. **p < 0.01 versus all other groups, as assessed by ANOVA with Tukey’s post hoc test.

(B)Glucoseproductionduring theclampofHFDrats injectedwithmismatchorACSL3 lentiviral shRNAthat receivedanuppersmall intestinalRCmicrobiota transplant

and an upper small intestinal saline (n = 5, 5), or Intralipid (n = 5, 5) infusion. **p < 0.01 versus all other groups, as assessed by ANOVA with Tukey’s post hoc test.

(C) Protein expression of ACSL3 in the upper small intestinal mucosa of mismatch (n = 6) or ACSL3 lentiviral shRNA (n = 5)-injected HFD rats that received an

upper small intestinal RCmicrobiota transplant. Brain tissue was used as a positive control; heart was used as a negative control. b-Actin and GAPDHwere used

as loading controls as b-actin is normally not detected in heart tissue. *p < 0.05 versus mismatch as determined by t test.

(D) Glucose production during the clamp of HFD rats that received an upper small intestinal RC microbiota transplant and a saline (n = 6), Intralipid (n = 6),

Exendin-9 (n = 5), or Exendin-9 + Intralipid (n = 6) upper small intestinal infusion. **p < 0.01 versus all other groups, as assessed by ANOVA with Tukey’s post hoc

test. Values are shown as mean + SEM. See also Figure S5. RC, regular chow; HFD, high-fat diet; HFD + RCM, HFD with RC microbiota transplant; ACSL3, acyl

coA synthetase 3; LV-ACSL3 shRNA, lentiviral ACSL3 shRNA; basal, the average glucose production over the period of 60–90 min during the pancreatic (basal

insulin)-euglycemic clamp; clamp, the average glucose production from 180 to 200 min.

GLP-1 levels and lowers GP in RC, but not HFD, rats during the

pancreatic clamp conditions, while co-infusion of glucose with

Exendin-9 abolishes the glucoregulatory ability of the upper

small intestinal glucose infusion in RC rats (Bauer et al., 2018).

Given that upper small intestinal fatty acid infusion similarly

lowers GP through a GLP-1R-dependent pathway in RC rats,

but failed to exert a GP-lowering effect in HFD rats (Figures 2E

and 2G), it is likely that upper small intestinal fatty acid infusion,

as conducted in the current study, also stimulates the release of

GLP-1 in RC but not HFD rats.

580 Cell Metabolism 27, 572–587, March 6, 2018

Our findings demonstrate that the restoration of upper small

intestinal ACSL3 by transplantation of RC upper small intestinal

microbiota in HFD rats is necessary to restore a glucoregulatory

lipid-dependent pathway.

Upper Small Intestinal L. gasseri AdministrationRestores Upper Small Intestinal ACSL3-DependentLipid Sensing in HFD RatsUpper small intestinal L. gasseri (from the Lactobacillus genus)

levels were reduced in response to HFD and increased to normal

Page 11: Lactobacillus gasseri in the Upper Small Intestine Impacts

Figure 7. Upper Small Intestinal Administration of L. gasseri Alters an ACSL3-Dependent Fatty Acid-Sensing Pathway via Inhibition of FXR to

Impact GP Regulation

(A) Schematic representation of the working hypothesis.

(B) Glucose production during the clamp of HFD rats pre-administered with PBS that received an upper small intestinal saline or Intralipid infusion (n = 5, 6

respectively) and HFD rats pre-administeredwith L. gasseri that received a saline or Intralipid (n = 6, 6 respectively) upper small intestinal infusion. *p < 0.05 versus

all other groups, as assessed by ANOVA with Tukey’s post hoc test.

(C) Protein expression of ACSL3 in the upper small intestinal mucosa of RC rats (n = 5), HFD rats administered with PBS (vehicle) (n = 5), HFD rats administered

with L. gasseri (n = 5), and HFD rats administered with L. gasseri followed by GW4064 (n = 5). Brain tissue was used as a positive control; heart was used as a

negative control. b-Actin and GAPDH were used as loading controls as b-actin is normally not detected in heart tissue. *p < 0.05, **p < 0.01 versus RC and HFD +

LG, as assessed by ANOVA with Tukey’s post hoc test.

(D) Glucose production during the clamp of HFD rats administeredwith PBS vehicle with or without GW4064 and infusedwith Intralipid (n = 6, 6 respectively), HFD

rats administered with L. gasseri that received 1% CMC and a saline or Intralipid infusion (n = 5, 6 respectively), or HFD rats administered with L. gasseri that

received GW4064 and a saline or Intralipid infusion (n = 6, 5, respectively). *p < 0.05 versus all other groups, as assessed by ANOVA with Tukey’s post hoc test.

Values are shown as mean + SEM. See also Figure S6. FXR, farnesoid X receptor; ACSL3, acyl CoA synthetase 3; LCFA-CoA, long-chain fatty acyl CoA; RC,

regular chow; HFD, high-fat diet; LG, L. gasseri; basal, the average glucose production over the period of 60–90 min during the pancreatic (basal insulin)-

euglycemic clamp; clamp, the average glucose production from 180 to 200 min.

levels in HFD rats that received an upper small intestinal RC

microbiota transplant (Figure 3E; Table S3). This suggests that

the increase in upper small intestinal L. gasseri could restore glu-

coregulatory fatty acid-sensing pathways and improve glucose

control in these HFD transplanted rats. L. gasseri exhibits bile

salt hydrolase activity (Rani et al., 2017), and administration of

L. gasseri probiotics alters the bile acid pool and promotes bile

acid excretion in rats (Usman and Hosono, 2001). In parallel,

the bile acid sequestrant colesevelam improves glycemic control

in diabetic humans (Beysen et al., 2012; Marina et al., 2012) and

rodents (Prawitt et al., 2011; Trabelsi et al., 2015) and inhibits in-

testinal FXR in rodents (Prawitt et al., 2011; Trabelsi et al., 2015).

Inhibition of intestinal FXR per se improves metabolic homeosta-

sis (Jiang et al., 2015; Li et al., 2013; Prawitt et al., 2011; Trabelsi

et al., 2015) in the presence of increased ACSL in the intestinal

mucosa (Li et al., 2013), while FXR activation downregulates

nutrient-stimulated GLP-1 secretion (Trabelsi et al., 2015). These

collective and correlative findings raise the possibility that

L. gasseri enhances lipid sensing via FXR inhibition and subse-

quent upregulation of ACSL3 expression in the upper small intes-

tine (Figure 7A). Of note, L. animalis was also changed in our

experimental context (Figure 3E; Table S3). However, given

that the change in L. animalis was in the opposite direction as

L. gasseri and that, to the best of our knowledge, studies to

date have not documented a role for L. animalis in bile acid ho-

meostasis, we here first tested whether L. gasseri is sufficient

to restore upper small intestinal ACSL3-dependent glucoregula-

tory lipid-sensing pathways via FXR in HFD rats.

HFD rats were administered with L. gasseri probiotics (109 col-

ony-forming units) or PBS vehicle by slowly infusing the probi-

otic over the course of 30 s via the upper small intestinal cannula

1 day prior to the pancreatic clamp studies in the same way the

upper small intestinal RC microbiota transplant was performed

(Figure 4A). During the clamps, although upper small intestinal

Intralipid infusion for 50 min had no metabolic impact relative

to a saline infusion in non-probiotic-infused (or PBS-infused)

Cell Metabolism 27, 572–587, March 6, 2018 581

Page 12: Lactobacillus gasseri in the Upper Small Intestine Impacts

HFD rats, HFD rats administered with L. gasseri probiotics ex-

hibited a restoration of upper small intestinal lipid sensing, as

upper small intestinal Intralipid versus saline infusion increased

the glucose infusion rate (Figure S6A) and decreased GP (Fig-

ure 7B) in these rats, independent of changes in glucose uptake

(Figure S6B). To investigate whether this improvement in lipid

sensing was associated with a rise in ACSL3 expression, the

upper small intestinal mucosa of HFD rats administered with

PBS vehicle or L. gasseri was collected immediately following

the saline-infused clamp procedure and ACSL3 expression

was compared with untreated RC upper small intestinal mucosal

samples. HFD rats administered with PBS vehicle (non-probi-

otic-infused) exhibited significantly reduced ACSL3 protein

expression in the upper small intestinal mucosa relative to RC

samples (Figure 7C), which is consistent with ACSL3 protein

expression in untreated HFD rats (Figure 6A). Importantly, this

reduction in ACSL3 expression was prevented with L. gasseri

probiotic administration in HFD rats (Figure 7C).

Finally, to investigate whether L. gasseri restores upper small

intestinal ACSL3-dependent lipid-sensing pathways via FXR in-

hibition, we repeated the probiotic L. gasseri experiments in the

presence or absence of FXR activation. Probiotic- (or non-probi-

otic PBS)-infused HFD rats received GW4064 (FXR agonist) or

vehicle 1% carboxymethyl cellulose (CMC) 30min after probiotic

administration. GW4060 or 1% CMC was slowly administered

over the course of 30 s via the upper small intestinal cannula,

and was administered again the following morning before

commencing the pancreatic clamp studies. First, GW4064 did

not worsen upper small intestinal lipid-sensing mechanisms in

non-probiotic-infused (i.e., PBS-infused) HFD rats compared

with non-probiotic-infused and non-GW4064 HFD rats (Figures

7D and S6C). Second, an Intralipid versus saline 50 min infusion

instead was able to increase the glucose infusion rate (Fig-

ure S6C) and lower GP (Figure 7D) in probiotic-infused HFD

rats but, importantly, this glucoregulatory effect was lost in

probiotic-infused HFD rats administered with GW4064 (Figures

7D and S6C), while glucose uptake remained unchanged

(Figure S6D). Of note, we found that GW4064 inhibited the stim-

ulatory effect of L. gasseri on upper small intestinal ACSL3

expression in these HFD rats as well (Figure 7C).

Taken together, L. gasseri enhances an upper small intestinal

ACSL3-dependent glucoregulatory lipid-sensing pathway via

FXR inhibition in HFD rodents.

DISCUSSION

We here first report that upper small intestinal oleic and linoleic

acid sensing in healthy rodents pre-absorptively increase

whole-body glucose tolerance independent of a rise in plasma in-

sulin levels and lower GP when plasma insulin levels were main-

tained at basal with a pancreatic-euglycemic clamp. Both upper

small intestinal fatty acid-sensing pathways fail to regulate

glucose tolerance and GP in lard oil-enriched HFD-fed rodents,

suggesting that HFD feeding disrupts upper small intestinal fatty

acid sensing to lower GP, leading to a disruption of whole-body

glucose homeostasis. Given that defective fatty acid sensing

arises in response to a saturated fat lard oil-enriched diet, satu-

rated palmitic and stearic acid sensing in the upper small intestine

may not share the samemetabolic effect as themonounsaturated

582 Cell Metabolism 27, 572–587, March 6, 2018

oleic and polyunsaturated linoleic acids, as currently described,

although such aworking hypothesiswarrants future investigation.

HFD reduces upper small intestinal L. gasseri levels and ACSL3

expression, while both reductions were reversed in HFD rats that

received an upper small intestinal RC microbiota transplant,

together with a restoration of the glucoregulatory ACSL3-depen-

dent lipid-sensing pathway. We further discovered that directly

administering non-transplanted HFD rats with L. gasseri probiot-

ics is sufficient to recapitulate the effect of the RC microbiota

transplant, as upper small intestinal ACSL3 expression and the

subsequent glucoregulatory function of upper small intestinal lipid

sensing are also restored in these rats. These studies together

highlight thatL.gasseridirectly impactsanACSL3-dependentglu-

coregulatory lipid-sensing pathway in the upper small intestine.

Given that probiotic supplementation with various species from

the Lactobacillus genus improve glucose parameters in rodents

and humans with diabetes (Asemi et al., 2013; Ejtahed et al.,

2012; Yadav et al., 2007), future studies are warranted to deter-

mine whether L. gasseri-induced restoration of upper small intes-

tinal sensing mechanisms could rescue glucose homeostasis in

long-term HFD-induced obese and/or diabetic rodents.

How does L. gasseri increase ACSL3 expression in the upper

small intestine? Although the mechanistic link remains largely

unknown, our preliminary data suggest that inhibition of the up-

per small intestinal bile acid receptor, FXR, could represent a

possible link. This is consistent with the fact that Lactobacillus

probiotics inhibit intestinal FXR (Degirolamo et al., 2014), and

that FXR deficiency in rodents increases intestinal ACSL expres-

sion (Li et al., 2013). Future gain- and loss-of-function genetic

experiments targeting the upper small intestinal FXR nuclear

receptor are necessary to assess the integrative role of FXR

in microbiota-induced regulation of glucoregulatory intestinal

lipid-sensing mechanisms.

Another urgent question is which bile acid(s) are involved

in L. gasseri-induced FXR inhibition? L. gasseri preferentially de-

conjugates glyco-conjugated bile acids rather than tauro-conju-

gated bile acids (Rani et al., 2017), which would lead to a drop in

glyco-conjugated bile acid with a relatively stable level of tauro-

conjugated bile acids. On one hand, deconjugation of glyco-

conjugated bile acids would result in increased excretion of

these bile acids, as deconjugation increases the hydrophobicity

of bile acids leading to increased excretion (Choi et al., 2015;

Schiff et al., 1972). In fact, similarly to other Lactobacillus species

(Degirolamo et al., 2014; Jeun et al., 2010; Kumar et al., 2011;

Lye et al., 2017), administration of L. gasseri probiotics increases

bile acid excretion in rodents (Usman and Hosono, 2001).

Consequently, Lactobacillus-induced reductions in intestinal

bile acid uptake can reduce activity of the bile acid receptor

FXR in the intestine (Degirolamo et al., 2014). On the other

hand, tauro-conjugated bile acids, such as tauro-b-muricholic

acid, inhibit FXR and improve glucose homeostasis in obese

rats (Li et al., 2013; Sayin et al., 2013). Thus the potential relative

increase in tauro-conjugated bile acids could also contribute to a

L. gasseri-induced suppression of FXR. Nonetheless, assessing

bile acids as links between L. gasseri and ACSL3-dependent

lipid-sensing pathways as well as how microbiota transplanta-

tion affects bile acid pool warrant future investigations.

Microbiota changes in the large intestine and feces correlate

with changes in host metabolism (Forslund et al., 2015; Karlsson

Page 13: Lactobacillus gasseri in the Upper Small Intestine Impacts

et al., 2013; Le Chatelier et al., 2013; Qin et al., 2012; Ridaura

et al., 2013) and transplantation of large intestinal or fecal micro-

biota from HFD obese rodents impairs glucose homeostasis in

recipient rodents (Kovatcheva-Datchary et al., 2015; Parseus

et al., 2017; Perry et al., 2016; Ridaura et al., 2013). In parallel,

administration of probiotics improves glucose homeostasis in

diabetic rodents in association with changes in large intestinal

and fecal microbiota composition (Balakumar et al., 2016; Cani

et al., 2006b; Kovatcheva-Datchary et al., 2015; Neyrinck

et al., 2012; Zhou et al., 2008), together suggesting that changes

in large intestinal microbiota affect glucose homeostasis. These

studies clearly did not (and could not) rule out that upper small

intestinal microbiota does (or does not) impact metabolic ho-

meostasis. Instead, we here report that the upper small intestinal

RC microbiota transplant rescues upper small intestinal lipid-

sensing pathways in HFD-recipient rats via specific changes in

the upper small intestinal microbiota, rather than changes in

the colonic tract. However, our studies are in no way implying

that direct HFD-induced changes in colonic microbiota do not

impact glucose homeostasis as well. Taken together, we pro-

pose a potential parallel role of the distal and upper small intes-

tinal microbiota in glucose homeostasis.

In fact, metformin has been documented to alter both distal in-

testinal (de la Cuesta-Zuluaga et al., 2017; Forslund et al., 2015;

Shin et al., 2014;Wu et al., 2017) and upper small intestinalmicro-

biota (Bauer et al., 2018) in parallel to improvements in glucose

homeostasis. While HFD feeding lowers upper small intestinal

L. gasseri levels (Figure 3) (Bauer et al., 2018), transplantation

of metformin-treated microbiota into the upper small intestine

of HFD-recipient rats does not alter L. gasseri levels (in contrast

to what is currently described for the RC microbiota transplant

in HFD rats (Figure 3)). Transplantation of metformin-treated mi-

crobiota instead increases Lactobacillus salivarius in parallel to

a restoration of an upper small intestinal SGLT1-dependent glu-

coregulatory glucose-sensing pathway (Bauer et al., 2018). Given

that L. gasseri activates an ACSL3-dependent glucoregulatory

lipid-sensing pathway via FXR inhibition in the upper small intes-

tine (Figure 7), and that L. salivarius, like L. gasseri, exhibits bile

salt hydrolase activity (Wang et al., 2012; Xu et al., 2016), which

could impact the bile acid pool and subsequent FXR activity,

we propose (which clearly warrants future investigation) that

upper small intestinal FXR could be a common pathway that links

members of the Lactobacillus genus, such as L. gasseri and

L. salivarius, with nutrient-sensing mechanisms to exert glucore-

gulatory effects in obesity and diabetes.

In summary, L. gasseri probiotic administration in HFD rats, or

transplantation of RC upper small intestinal microbiota that in-

creases upper small intestinal L. gasseri levels in HFD-recipient

rats, enhances an upper small intestinal ACSL3-dependent glu-

coregulatory lipid-sensing pathway in rodents. These findings

strengthen the claim on the causative glucoregulatory role of

the gut microbiota and highlight L. gasseri as an important regu-

lator of nutrient sensing mechanisms in the upper small intestine

to exert metabolic benefits.

Limitations of StudyThe first key limitation of our study is that the current working hy-

pothesis has yet to be tested in chronic obese and/or diabetic

animalmodels aswell as in healthy, obese, and diabetic humans.

A second key limitation is that the glucoregulatory role of ACSL

isoforms other than ACSL3, as well as the definitive glucoregula-

tory role of the bile acids, has not been investigated in the current

as well as chronic obese and/or diabetic models. Lastly, the

glucoregulatory role of the members of the Lactobacillus genus

other than L. gasseri has yet to be assessed.

STAR+METHODS

Detailed methods are provided in the online version of this paper

and include the following:

d KEY RESOURCES TABLE

d CONTACT FOR REAGENT AND RESOURCE SHARING

d EXPERIMENTAL MODEL AND SUBJECT DETAILS

B Animals

d METHOD DETAILS

B Surgical Procedures

B Virus Injection

B High-fat Diet Model

B Intravenous Glucose Tolerance Test

B Rat Pancreatic (Basal Insulin) Clamp Procedure

B Mouse Pancreatic (Basal Insulin) Clamp Procedure

B Fatty Acid Preparation for Upper Small Intestinal In-

fusions

B Upper Small Intestinal Infusions

B Microbiota Transplant

B Microbiota Heat-Shock

B Microbiota Filter

B Lactobacillus Gasseri Administration

B GW4064 Administration

B Genomic DNA Extraction and 16S rRNA Gene

Sequencing

B Sequence Processing and Data Analysis

B Tissue Collection and Western Blotting

B Biochemical Analysis

d QUANTIFICATION AND STATISTICAL ANALYSIS

SUPPLEMENTAL INFORMATION

Supplemental Information includes six figures and three tables and can be

found with this article online at https://doi.org/10.1016/j.cmet.2018.01.013.

ACKNOWLEDGMENTS

This work was supported by a Canadian Institutes of Health Research (CIHR)

Foundation Grant to T.K.T.L. (FDN-143204). P.V.B. is supported by an Ontario

Graduate Scholarship and a Banting and Best Diabetes Center Graduate Stu-

dentship. F.A.D. was aBanting Fellow. T.M.Z.W. is supported by aBanting and

Best Diabetes Center Post-Doctoral Fellowship. H.J.D. is supported by a CIHR

and a Diabetes Canada post-doctoral fellowship. B.A.R. was a Vanier Scholar.

T.K.T.L. holds the John KitsonMcIvor (1915–1942) Endowed Chair in Diabetes

Research and the Canada Research Chair in Obesity at the Toronto General

Hospital Research Institute and the University of Toronto.

AUTHOR CONTRIBUTIONS

P.V.B. conducted and designed the experiments, performed the data ana-

lyses, and wrote the manuscript. F.A.D., T.M.Z.W., and H.J.D. assisted with

the experiments and edited the manuscript. B.A.R., A.P., M.R., and C.A.O. as-

sisted with the experiments. T.K.T.L. supervised the project, designed the ex-

periments, and edited the manuscript.

Cell Metabolism 27, 572–587, March 6, 2018 583

Page 14: Lactobacillus gasseri in the Upper Small Intestine Impacts

DECLARATION OF INTERESTS

The authors declare no competing interests.

Received: May 15, 2017

Revised: November 9, 2017

Accepted: January 22, 2018

Published: March 6, 2018

REFERENCES

Almiron, M., Traglia, G., Rubio, A., and Sanjuan, N. (2013). Colonization of the

mouse upper gastrointestinal tract by Lactobacillus murinus: a histological,

immunocytochemical, and ultrastructural study. Curr. Microbiol. 67, 395–398.

Asemi, Z., Zare, Z., Shakeri, H., Sabihi, S.S., and Esmaillzadeh, A. (2013).

Effect of multispecies probiotic supplements on metabolic profiles, hs-CRP,

and oxidative stress in patients with type 2 diabetes. Ann. Nutr. Metab. 63, 1–9.

Backhed, F., Ding, H., Wang, T., Hooper, L.V., Koh, G.Y., Nagy, A.,

Semenkovich, C.F., and Gordon, J.I. (2004). The gut microbiota as an environ-

mental factor that regulates fat storage. Proc. Natl. Acad. Sci. USA 101,

15718–15723.

Balakumar, M., Prabhu, D., Sathishkumar, C., Prabu, P., Rokana, N., Kumar,

R., Raghavan, S., Soundarajan, A., Grover, S., Batish, V.K., et al. (2016).

Improvement in glucose tolerance and insulin sensitivity by probiotic strains

of Indian gut origin in high-fat diet-fed C57BL/6J mice. Eur. J. Nutr. https://

doi.org/10.1007/s00394-016-1317-7.

Bartram, A.K., Lynch,M.D., Stearns, J.C., Moreno-Hagelsieb, G., and Neufeld,

J.D. (2011). Generation of multimillion-sequence 16S rRNA gene libraries from

complex microbial communities by assembling paired-end illumina reads.

Appl. Environ. Microbiol. 77, 3846–3852.

Batterham, R.L., and Cummings, D.E. (2016). Mechanisms of diabetes

improvement following bariatric/metabolic surgery. Diabetes Care 39,

893–901.

Bauer, P.V., Duca, F.A., Waise, T.M.Z., Rasmussen, B.A., Abraham, M.A.,

Dranse, H.J., Puri, A., O’Brien, C.A., and Lam, T.K.T. (2018). Metformin alters

upper small intestinal microbiota that impact a glucose-SGLT1-sensing glu-

coregulatory pathway. Cell Metab. 27, 101–117.e5. Published online

October 19, 2017.

Beysen, C., Murphy, E.J., Deines, K., Chan, M., Tsang, E., Glass, A., Turner,

S.M., Protasio, J., Riiff, T., and Hellerstein, M.K. (2012). Effect of bile acid se-

questrants on glucose metabolism, hepatic de novo lipogenesis, and choles-

terol and bile acid kinetics in type 2 diabetes: a randomised controlled study.

Diabetologia 55, 432–442.

Bowman, T.A., O’Keeffe, K.R., D’Aquila, T., Yan, Q.W., Griffin, J.D., Killion,

E.A., Salter, D.M., Mashek, D.G., Buhman, K.K., and Greenberg, A.S. (2016).

Acyl CoA synthetase 5 (ACSL5) ablation in mice increases energy expenditure

and insulin sensitivity and delays fat absorption. Mol. Metab. 5, 210–220.

Breen, D.M., Rasmussen, B.A., Kokorovic, A., Wang, R., Cheung, G.W., and

Lam, T.K. (2012). Jejunal nutrient sensing is required for duodenal-jejunal

bypass surgery to rapidly lower glucose concentrations in uncontrolled dia-

betes. Nat. Med. 18, 950–955.

Breen, D.M., Yue, J.T., Rasmussen, B.A., Kokorovic, A., Cheung, G.W., and

Lam, T.K. (2011). Duodenal PKC-delta and cholecystokinin signaling axis

regulates glucose production. Diabetes 60, 3148–3153.

Brubaker, P.L. (1991). Regulation of intestinal proglucagon-derived peptide

secretion by intestinal regulatory peptides. Endocrinology 128, 3175–3182.

Cani, P.D., Joly, E., Horsmans, Y., and Delzenne, N.M. (2006a). Oligofructose

promotes satiety in healthy human: a pilot study. Eur. J. Clin. Nutr. 60,

567–572.

Cani, P.D., Knauf, C., Iglesias, M.A., Drucker, D.J., Delzenne, N.M., and

Burcelin, R. (2006b). Improvement of glucose tolerance and hepatic insulin

sensitivity by oligofructose requires a functional glucagon-like peptide 1 re-

ceptor. Diabetes 55, 1484–1490.

Caporaso, J.G., Kuczynski, J., Stombaugh, J., Bittinger, K., Bushman, F.D.,

Costello, E.K., Fierer, N., Pena, A.G., Goodrich, J.K., Gordon, J.I., et al.

584 Cell Metabolism 27, 572–587, March 6, 2018

(2010). QIIME allows analysis of high-throughput community sequencing

data. Nat. Methods 7, 335–336.

Chang, C.H., Chey, W.Y., and Chang, T.M. (2000). Cellular mechanism of

sodium oleate-stimulated secretion of cholecystokinin and secretin. Am. J.

Physiol. Gastrointest. Liver Physiol. 279, G295–G303.

Cheung, G.W., Kokorovic, A., Lam, C.K., Chari, M., and Lam, T.K. (2009).

Intestinal cholecystokinin controls glucose production through a neuronal

network. Cell Metab. 10, 99–109.

Chisholm, C., and Greenberg, G.R. (2002). Somatostatin-28 regulates GLP-1

secretion via somatostatin receptor subtype 5 in rat intestinal cultures. Am.

J. Physiol. Endocrinol. Metab. 283, E311–E317.

Choi, S.B., Lew, L.C., Yeo, S.K., Nair Parvathy, S., and Liong, M.T. (2015).

Probiotics and the BSH-related cholesterol lowering mechanism: a Jekyll

and Hyde scenario. Crit. Rev. Biotechnol. 35, 392–401.

Clarysse, S., Tack, J., Lammert, F., Duchateau, G., Reppas, C., and

Augustijns, P. (2009). Postprandial evolution in composition and characteris-

tics of human duodenal fluids in different nutritional states. J. Pharm. Sci.

98, 1177–1192.

Cote, C.D., Rasmussen, B.A., Duca, F.A., Zadeh-Tahmasebi, M., Baur, J.A.,

Daljeet, M., Breen, D.M., Filippi, B.M., and Lam, T.K. (2015). Resveratrol acti-

vates duodenal Sirt1 to reverse insulin resistance in rats through a neuronal

network. Nat. Med. 21, 498–505.

Dailey, M.J., Moghadam, A.A., and Moran, T.H. (2011). Jejunal linoleic acid in-

fusions require GLP-1 receptor signaling to inhibit food intake: implications for

the effectiveness of Roux-en-Y gastric bypass. Am. J. Physiol. Endocrinol.

Metab. 301, E1184–E1190.

David, L.A., Maurice, C.F., Carmody, R.N., Gootenberg, D.B., Button, J.E.,

Wolfe, B.E., Ling, A.V., Devlin, A.S., Varma, Y., Fischbach, M.A., et al.

(2014). Diet rapidly and reproducibly alters the human gut microbiome.

Nature 505, 559–563.

de la Cuesta-Zuluaga, J., Mueller, N.T., Corrales-Agudelo, V., Velasquez-Mejia,

E.P., Carmona, J.A., Abad, J.M., and Escobar, J.S. (2017). Metformin is

associated with higher relative abundance of mucin-degrading Akkermansia

muciniphila and several short-chain fatty acid-producing microbiota in the gut.

Diabetes Care 40, 54–62.

Defronzo, R.A. (2009). Banting Lecture. From the triumvirate to the ominous

octet: a new paradigm for the treatment of type 2 diabetes mellitus.

Diabetes 58, 773–795.

Degirolamo, C., Rainaldi, S., Bovenga, F., Murzilli, S., and Moschetta, A.

(2014). Microbiota modification with probiotics induces hepatic bile acid syn-

thesis via downregulation of the Fxr-Fgf15 axis in mice. Cell Rep. 7, 12–18.

Derrien, M., Van Baarlen, P., Hooiveld, G., Norin, E., Muller, M., and de Vos,

W.M. (2011). Modulation of mucosal immune response, tolerance, and prolif-

eration in mice colonized by the mucin-degrader Akkermansia muciniphila.

Front. Microbiol. 2, 166.

DeSantis, T.Z., Hugenholtz, P., Larsen, N., Rojas, M., Brodie, E.L., Keller, K.,

Huber, T., Dalevi, D., Hu, P., and Andersen, G.L. (2006). Greengenes, a

chimera-checked 16S rRNA gene database and workbench compatible with

ARB. Appl. Environ. Microbiol. 72, 5069–5072.

Di Luccia, B., Crescenzo, R., Mazzoli, A., Cigliano, L., Venditti, P., Walser, J.C.,

Widmer, A., Baccigalupi, L., Ricca, E., and Iossa, S. (2015). Rescue of fruc-

tose-induced metabolic syndrome by antibiotics or faecal transplantation in

a rat model of obesity. PLoS One 10, e0134893.

Diakogiannaki, E., Pais, R., Tolhurst, G., Parker, H.E., Horscroft, J., Rauscher,

B., Zietek, T., Daniel, H., Gribble, F.M., and Reimann, F. (2013). Oligopeptides

stimulate glucagon-like peptide-1 secretion in mice through proton-coupled

uptake and the calcium-sensing receptor. Diabetologia 56, 2688–2696.

Duca, F.A., Bauer, P.V., Hamr, S.C., and Lam, T.K. (2015a). Glucoregulatory

relevance of small intestinal nutrient sensing in physiology, bariatric surgery,

and pharmacology. Cell Metab. 22, 367–380.

Duca, F.A., Cote, C.D., Rasmussen, B.A., Zadeh-Tahmasebi, M., Rutter, G.A.,

Filippi, B.M., and Lam, T.K. (2015b). Metformin activates a duodenal Ampk-

dependent pathway to lower hepatic glucose production in rats. Nat. Med.

21, 506–511.

Page 15: Lactobacillus gasseri in the Upper Small Intestine Impacts

Ejtahed, H.S., Mohtadi-Nia, J., Homayouni-Rad, A., Niafar, M., Asghari-

Jafarabadi, M., and Mofid, V. (2012). Probiotic yogurt improves antioxidant

status in type 2 diabetic patients. Nutrition 28, 539–543.

El Aidy, S., Merrifield, C.A., Derrien, M., van Baarlen, P., Hooiveld, G., Levenez,

F., Dore, J., Dekker, J., Holmes, E., Claus, S.P., et al. (2013). The gut micro-

biota elicits a profound metabolic reorientation in the mouse jejunal mucosa

during conventionalisation. Gut 62, 1306–1314.

Everard, A., and Cani, P.D. (2013). Diabetes, obesity and gut microbiota. Best

Pract. Res. Clin. Gastroenterol. 27, 73–83.

Forslund, K., Hildebrand, F., Nielsen, T., Falony, G., Le Chatelier, E., Sunagawa,

S., Prifti, E., Vieira-Silva, S., Gudmundsdottir, V., Krogh Pedersen, H., et al.

(2015). Disentangling type 2 diabetes and metformin treatment signatures in

the human gut microbiota. Nature 528, 262–266.

Gareau, M.G., Ho, N.K., Brenner, D., Sousa, A.J., Lebourhis, L., Mak, T.W.,

Girardin, S.E., Philpott, D.J., and Sherman, P.M. (2011). Enterohaemorrhagic,

but not enteropathogenic, Escherichia coli infection of epithelial cells disrupts

signalling responses to tumour necrosis factor-alpha.Microbiology 157, 2963–

2973.

Giannakis, M., Backhed, H.K., Chen, S.L., Faith, J.J., Wu, M., Guruge, J.L.,

Engstrand, L., and Gordon, J.I. (2009). Response of gastric epithelial progen-

itors to Helicobacter pylori isolates obtained from Swedish patients with

chronic atrophic gastritis. J. Biol. Chem. 284, 30383–30394.

Greenberg, D., Kava, R.A., Lewis, D.R., Greenwood, M.R., and Smith, G.P.

(1995). Time course for entry of intestinally infused lipids into blood of rats.

Am. J. Physiol. 269, R432–R436.

Gribble, F.M., Williams, L., Simpson, A.K., and Reimann, F. (2003). A novel

glucose-sensing mechanism contributing to glucagon-like peptide-1 secre-

tion from the GLUTag cell line. Diabetes 52, 1147–1154.

Gu, S., Chen, D., Zhang, J.N., Lv, X.,Wang, K., Duan, L.P., Nie, Y., andWu, X.L.

(2013). Bacterial community mapping of the mouse gastrointestinal tract.

PLoS One 8, e74957.

Hansen, L., Hartmann, B., Bisgaard, T., Mineo, H., Jorgensen, P.N., and Holst,

J.J. (2000). Somatostatin restrains the secretion of glucagon-like peptide-1

and -2 from isolated perfused porcine ileum. Am. J. Physiol. Endocrinol.

Metab. 278, E1010–E1018.

Hundal, R.S., Krssak, M., Dufour, S., Laurent, D., Lebon, V., Chandramouli, V.,

Inzucchi, S.E., Schumann, W.C., Petersen, K.F., Landau, B.R., et al. (2000).

Mechanism by which metformin reduces glucose production in type 2 dia-

betes. Diabetes 49, 2063–2069.

Iakoubov, R., Ahmed, A., Lauffer, L.M., Bazinet, R.P., and Brubaker, P.L.

(2011). Essential role for protein kinase Czeta in oleic acid-induced

glucagon-like peptide-1 secretion in vivo in the rat. Endocrinology 152,

1244–1252.

Iakoubov, R., Izzo, A., Yeung, A., Whiteside, C.I., and Brubaker, P.L. (2007).

Protein kinase Czeta is required for oleic acid-induced secretion of

glucagon-like peptide-1 by intestinal endocrine L cells. Endocrinology 148,

1089–1098.

Jeun, J., Kim, S., Cho, S.Y., Jun, H.J., Park, H.J., Seo, J.G., Chung, M.J., and

Lee, S.J. (2010). Hypocholesterolemic effects of Lactobacillus plantarum

KCTC3928 by increased bile acid excretion in C57BL/6 mice. Nutrition 26,

321–330.

Jiang, C., Xie, C., Lv, Y., Li, J., Krausz, K.W., Shi, J., Brocker, C.N., Desai, D.,

Amin, S.G., Bisson, W.H., et al. (2015). Intestine-selective farnesoid X receptor

inhibition improves obesity-related metabolic dysfunction. Nat. Commun.

6, 10166.

Jiao, J., Bae, E.J., Bandyopadhyay, G., Oliver, J., Marathe, C., Chen, M., Hsu,

J.Y., Chen, Y., Tian, H., Olefsky, J.M., et al. (2013). Restoration of euglycemia

after duodenal bypass surgery is reliant on central and peripheral inputs in

Zucker fa/fa rats. Diabetes 62, 1074–1083.

Karlsson, F.H., Tremaroli, V., Nookaew, I., Bergstrom, G., Behre, C.J.,

Fagerberg, B., Nielsen, J., and Backhed, F. (2013). Gut metagenome in

European women with normal, impaired and diabetic glucose control.

Nature 498, 99–103.

Kokorovic, A., Cheung, G.W., Breen, D.M., Chari, M., Lam, C.K., and Lam, T.K.

(2011). Duodenal mucosal protein kinase C-delta regulates glucose produc-

tion in rats. Gastroenterology 141, 1720–1727.

Kovatcheva-Datchary, P., Nilsson, A., Akrami, R., Lee, Y.S., De Vadder, F.,

Arora, T., Hallen, A., Martens, E., Bjorck, I., and Backhed, F. (2015). Dietary fi-

ber-induced improvement in glucose metabolism is associated with increased

abundance of Prevotella. Cell Metab. 22, 971–982.

Kuhre, R.E., Frost, C.R., Svendsen, B., and Holst, J.J. (2015). Molecular mech-

anisms of glucose-stimulated GLP-1 secretion from perfused rat small intes-

tine. Diabetes 64, 370–382.

Kumar, R., Grover, S., and Batish, V.K. (2011). Hypocholesterolaemic effect of

dietary inclusion of two putative probiotic bile salt hydrolase-producing

Lactobacillus plantarum strains in Sprague-Dawley rats. Br. J. Nutr. 105,

561–573.

Lam, T.K. (2010). Neuronal regulation of homeostasis by nutrient sensing. Nat.

Med. 16, 392–395.

Le Chatelier, E., Nielsen, T., Qin, J., Prifti, E., Hildebrand, F., Falony, G.,

Almeida, M., Arumugam, M., Batto, J.M., Kennedy, S., et al. (2013).

Richness of human gut microbiome correlates with metabolic markers.

Nature 500, 541–546.

Li, F., Jiang, C., Krausz, K.W., Li, Y., Albert, I., Hao, H., Fabre, K.M., Mitchell,

J.B., Patterson, A.D., andGonzalez, F.J. (2013). Microbiome remodelling leads

to inhibition of intestinal farnesoid X receptor signalling and decreased obesity.

Nat. Commun. 4, 2384.

Liu, J., Kandasamy, S., Zhang, J., Kirby, C.W., Karakach, T., Hafting, J.,

Critchley, A.T., Evans, F., and Prithiviraj, B. (2015). Prebiotic effects of diet

supplemented with the cultivated red seaweed Chondrus crispus or with

fructo-oligo-saccharide on host immunity, colonic microbiota and gut micro-

bial metabolites. BMC Complement. Altern. Med. 15, 279.

Lye, H.S., Kato, T., Low, W.Y., Taylor, T.D., Prakash, T., Lew, L.C., Ohno, H.,

and Liong, M.T. (2017). Lactobacillus fermentum FTDC 8312 combats hyper-

cholesterolemia via alteration of gut microbiota. J. Biotechnol. 262, 75–83.

Mansbach, C.M., 2nd, Cohen, R.S., and Leff, P.B. (1975). Isolation and prop-

erties of the mixed lipid micelles present in intestinal content during fat diges-

tion in man. J. Clin. Invest. 56, 781–791.

Marina, A.L., Utzschneider, K.M., Wright, L.A., Montgomery, B.K., Marcovina,

S.M., and Kahn, S.E. (2012). Colesevelam improves oral but not intravenous

glucose tolerance by a mechanism independent of insulin sensitivity and

beta-cell function. Diabetes Care 35, 1119–1125.

Masella, A.P., Bartram, A.K., Truszkowski, J.M., Brown, D.G., and Neufeld,

J.D. (2012). PANDAseq: paired-end assembler for illumina sequences. BMC

Bioinformatics 13, 31.

Mashek, D.G., Li, L.O., and Coleman, R.A. (2006). Rat long-chain acyl-CoA

synthetase mRNA, protein, and activity vary in tissue distribution and in

response to diet. J. Lipid Res. 47, 2004–2010.

Morgan, K., Obici, S., andRossetti, L. (2004). Hypothalamic responses to long-

chain fatty acids are nutritionally regulated. J. Biol. Chem. 279, 31139–31148.

Neyrinck, A.M., Van Hee, V.F., Piront, N., De Backer, F., Toussaint, O., Cani,

P.D., and Delzenne, N.M. (2012). Wheat-derived arabinoxylan oligosaccha-

rides with prebiotic effect increase satietogenic gut peptides and reduce

metabolic endotoxemia in diet-induced obese mice. Nutr. Diabetes 2, e28.

Parseus, A., Sommer, N., Sommer, F., Caesar, R., Molinaro, A., Stahlman, M.,

Greiner, T.U., Perkins, R., and Backhed, F. (2017). Microbiota-induced obesity

requires farnesoid X receptor. Gut 66, 429–437.

Perry, R.J., Peng, L., Barry, N.A., Cline, G.W., Zhang, D., Cardone, R.L.,

Petersen, K.F., Kibbey, R.G., Goodman, A.L., and Shulman, G.I. (2016).

Acetate mediates a microbiome-brain-beta-cell axis to promote metabolic

syndrome. Nature 534, 213–217.

Phifer, C.B., and Berthoud, H.R. (1998). Duodenal nutrient infusions

differentially affect sham feeding and Fos expression in rat brain stem. Am.

J. Physiol. 274, R1725–R1733.

Plovier, H., Everard, A., Druart, C., Depommier, C., Van Hul, M., Geurts, L.,

Chilloux, J., Ottman, N., Duparc, T., Lichtenstein, L., et al. (2017). A purified

Cell Metabolism 27, 572–587, March 6, 2018 585

Page 16: Lactobacillus gasseri in the Upper Small Intestine Impacts

membrane protein from Akkermansia muciniphila or the pasteurized bacterium

improves metabolism in obese and diabetic mice. Nat. Med. 23, 107–113.

Prawitt, J., Abdelkarim, M., Stroeve, J.H., Popescu, I., Duez, H., Velagapudi,

V.R., Dumont, J., Bouchaert, E., van Dijk, T.H., Lucas, A., et al. (2011).

Farnesoid X receptor deficiency improves glucose homeostasis in mouse

models of obesity. Diabetes 60, 1861–1871.

Qin, J., Li, Y., Cai, Z., Li, S., Zhu, J., Zhang, F., Liang, S., Zhang, W., Guan, Y.,

Shen, D., et al. (2012). A metagenome-wide association study of gut micro-

biota in type 2 diabetes. Nature 490, 55–60.

Rani, R.P., Anandharaj, M., andRavindran, A.D. (2017). Characterization of bile

salt hydrolase from Lactobacillus gasseri FR4 and demonstration of its sub-

strate specificity and inhibitory mechanism using molecular docking analysis.

Front. Microbiol. 8, 1004.

Rasmussen, B.A., Breen, D.M., Luo, P., Cheung, G.W., Yang, C.S., Sun, B.,

Kokorovic, A., Rong, W., and Lam, T.K. (2012). Duodenal activation of

cAMP-dependent protein kinase induces vagal afferent firing and lowers

glucose production in rats. Gastroenterology 142, 834–843.e3.

Reimann, F., Habib, A.M., Tolhurst, G., Parker, H.E., Rogers, G.J., and Gribble,

F.M. (2008). Glucose sensing in L cells: a primary cell study. Cell Metab. 8,

532–539.

Richards, P., Pais, R., Habib, A.M., Brighton, C.A., Yeo, G.S., Reimann, F., and

Gribble, F.M. (2016). High fat diet impairs the function of glucagon-like pep-

tide-1 producing L-cells. Peptides 77, 21–27.

Ridaura, V.K., Faith, J.J., Rey, F.E., Cheng, J., Duncan, A.E., Kau, A.L., Griffin,

N.W., Lombard, V., Henrissat, B., Bain, J.R., et al. (2013). Gut microbiota from

twins discordant for obesity modulate metabolism in mice. Science 341,

1241214.

Riserus, U., Willett, W.C., and Hu, F.B. (2009). Dietary fats and prevention of

type 2 diabetes. Prog. Lipid Res. 48, 44–51.

Rocca, A.S., LaGreca, J., Kalitsky, J., and Brubaker, P.L. (2001).

Monounsaturated fatty acid diets improve glycemic tolerance through

increased secretion of glucagon-like peptide-1. Endocrinology 142, 1148–

1155.

Roglic, G., and World Health Organization. (2016). Global Report on Diabetes

(World Health Organization).

Rossetti, L., Giaccari, A., and DeFronzo, R.A. (1990). Glucose toxicity.

Diabetes Care 13, 610–630.

Sayin, S.I., Wahlstrom, A., Felin, J., Jantti, S., Marschall, H.U., Bamberg, K.,

Angelin, B., Hyotylainen, T., Oresic, M., and Backhed, F. (2013). Gut micro-

biota regulates bile acid metabolism by reducing the levels of tauro-beta-mur-

icholic acid, a naturally occurring FXR antagonist. Cell Metab. 17, 225–235.

Scherer, T., Lindtner, C., Zielinski, E., O’Hare, J., Filatova, N., and Buettner, C.

(2012). Short term voluntary overfeeding disrupts brain insulin control of adi-

pose tissue lipolysis. J. Biol. Chem. 287, 33061–33069.

Schiff, E.R., Small, N.C., and Dietschy, J.M. (1972). Characterization of the ki-

netics of the passive and active transport mechanisms for bile acid absorption

in the small intestine and colon of the rat. J. Clin. Invest. 51, 1351–1362.

Segata, N., Izard, J.,Waldron, L., Gevers, D., Miropolsky, L., Garrett, W.S., and

Huttenhower, C. (2011). Metagenomic biomarker discovery and explanation.

Genome Biol. 12, R60.

Shah, B.P., Liu, P., Yu, T., Hansen, D.R., and Gilbertson, T.A. (2012). TRPM5 is

critical for linoleic acid-induced CCK secretion from the enteroendocrine cell

line, STC-1. Am. J. Physiol. Cell Physiol. 302, C210–C219.

Shin, N.R., Lee, J.C., Lee, H.Y., Kim, M.S., Whon, T.W., Lee, M.S., and Bae,

J.W. (2014). An increase in the Akkermansia spp. population induced by met-

formin treatment improves glucose homeostasis in diet-induced obese mice.

Gut 63, 727–735.

Soupene, E., and Kuypers, F.A. (2008). Mammalian long-chain acyl-CoA syn-

thetases. Exp. Biol. Med. (Maywood) 233, 507–521.

Thaler, J.P., Yi, C.X., Schur, E.A., Guyenet, S.J., Hwang, B.H., Dietrich, M.O.,

Zhao, X., Sarruf, D.A., Izgur, V., Maravilla, K.R., et al. (2012). Obesity is asso-

ciated with hypothalamic injury in rodents and humans. J. Clin. Invest. 122,

153–162.

586 Cell Metabolism 27, 572–587, March 6, 2018

Trabelsi, M.S., Daoudi, M., Prawitt, J., Ducastel, S., Touche, V., Sayin, S.I.,

Perino, A., Brighton, C.A., Sebti, Y., Kluza, J., et al. (2015). Farnesoid X recep-

tor inhibits glucagon-like peptide-1 production by enteroendocrine L cells.

Nat. Commun. 6, 7629.

Tremaroli, V., Karlsson, F., Werling, M., Stahlman, M., Kovatcheva-Datchary,

P., Olbers, T., Fandriks, L., le Roux, C.W., Nielsen, J., and Backhed, F.

(2015). Roux-en-Y gastric bypass and vertical banded gastroplasty induce

long-term changes on the human gut microbiome contributing to fat mass

regulation. Cell Metab. 22, 228–238.

Turnbaugh, P.J., Ley, R.E., Mahowald, M.A., Magrini, V., Mardis, E.R., and

Gordon, J.I. (2006). An obesity-associated gut microbiome with increased ca-

pacity for energy harvest. Nature 444, 1027–1031.

Turnbaugh, P.J., Ridaura, V.K., Faith, J.J., Rey, F.E., Knight, R., and Gordon,

J.I. (2009). The effect of diet on the human gut microbiome: a metagenomic

analysis in humanized gnotobiotic mice. Sci. Transl. Med. 1, 6ra14.

Usman, and Hosono, A. (2001). Hypocholesterolemic effect of Lactobacillus

gasseri SBT0270 in rats fed a cholesterol-enriched diet. J. Dairy Res. 68,

617–624.

Vazquez-Baeza, Y., Pirrung, M., Gonzalez, A., and Knight, R. (2013). EMPeror:

a tool for visualizing high-throughput microbial community data. Gigascience

2, 16.

Vrieze, A., Van Nood, E., Holleman, F., Salojarvi, J., Kootte, R.S., Bartelsman,

J.F., Dallinga-Thie, G.M., Ackermans, M.T., Serlie, M.J., Oozeer, R., et al.

(2012). Transfer of intestinal microbiota from lean donors increases insulin

sensitivity in individuals with metabolic syndrome. Gastroenterology 143,

913–916.e7.

Wang, P.Y., Caspi, L., Lam, C.K., Chari, M., Li, X., Light, P.E., Gutierrez-Juarez,

R., Ang,M., Schwartz, G.J., and Lam, T.K. (2008). Upper intestinal lipids trigger

a gut-brain-liver axis to regulate glucose production. Nature 452, 1012–1016.

Wang, Q., Garrity, G.M., Tiedje, J.M., and Cole, J.R. (2007). Naive Bayesian

classifier for rapid assignment of rRNA sequences into the new bacterial tax-

onomy. Appl. Environ. Microbiol. 73, 5261–5267.

Wang, Z., Zeng, X., Mo, Y., Smith, K., Guo, Y., and Lin, J. (2012). Identification

and characterization of a bile salt hydrolase from Lactobacillus salivarius

for development of novel alternatives to antibiotic growth promoters. Appl.

Environ. Microbiol. 78, 8795–8802.

Whelan, F.J., Verschoor, C.P., Stearns, J.C., Rossi, L., Luinstra, K., Loeb, M.,

Smieja, M., Johnstone, J., Surette, M.G., and Bowdish, D.M. (2014). The loss

of topography in themicrobial communities of the upper respiratory tract in the

elderly. Ann. Am. Thorac. Soc. 11, 513–521.

Wirth, R., Bodi, N., Maroti, G., Bagyanszki, M., Talapka, P., Fekete, E., Bagi, Z.,

and Kovacs, K.L. (2014). Regionally distinct alterations in the composition of

the gut microbiota in rats with streptozotocin-induced diabetes. PLoS One

9, e110440.

Woltman, T., Castellanos, D., and Reidelberger, R. (1995). Role of cholecysto-

kinin in the anorexia produced by duodenal delivery of oleic acid in rats. Am. J.

Physiol. 269, R1420–R1433.

Woltman, T., and Reidelberger, R. (1995). Effects of duodenal and distal ileal

infusions of glucose and oleic acid on meal patterns in rats. Am. J. Physiol.

269, R7–R14.

Wu, H., Esteve, E., Tremaroli, V., Khan, M.T., Caesar, R., Manneras-Holm, L.,

Stahlman, M., Olsson, L.M., Serino, M., Planas-Felix, M., et al. (2017).

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.

Xu, F., Guo, F., Hu, X.J., and Lin, J. (2016). Crystal structure of bile salt hydro-

lase from Lactobacillus salivarius. Acta Crystallogr. F Struct. Biol. Commun.

72, 376–381.

Yadav, H., Jain, S., and Sinha, P.R. (2007). Antidiabetic effect of probiotic dahi

containing Lactobacillus acidophilus and Lactobacillus casei in high fructose

fed rats. Nutrition 23, 62–68.

Ye, Y. (2011). Identification and quantification of abundant species from pyro-

sequences of 16S rRNA by consensus alignment. Proceedings (IEEE Int. Conf.

Bioinformatics Biomed.) 2010, 153–157.

Page 17: Lactobacillus gasseri in the Upper Small Intestine Impacts

Yue, J.T., Abraham, M.A., Bauer, P.V., LaPierre, M.P., Wang, P., Duca, F.A.,

Filippi, B.M., Chan, O., and Lam, T.K. (2016). Inhibition of glycine trans-

porter-1 in the dorsal vagal complex improves metabolic homeostasis in dia-

betes and obesity. Nat. Commun. 7, 13501.

Zadeh-Tahmasebi, M., Duca, F.A., Rasmussen, B.A., Bauer, P.V., Cote, C.D.,

Filippi, B.M., and Lam, T.K. (2016). Activation of short and long chain fatty acid

sensing machinery in the ileum lowers glucose production in vivo. J. Biol.

Chem. 291, 8816–8824.

Zhou, J., Martin, R.J., Tulley, R.T., Raggio, A.M., McCutcheon, K.L., Shen, L.,

Danna,S.C., Tripathy,S.,Hegsted,M., andKeenan,M.J. (2008).Dietary resistant

starch upregulates total GLP-1 andPYY in a sustained day-longmanner through

fermentation in rodents. Am. J. Physiol. Endocrinol. Metab. 295, E1160–E1166.

Cell Metabolism 27, 572–587, March 6, 2018 587

Page 18: Lactobacillus gasseri in the Upper Small Intestine Impacts

STAR+METHODS

KEY RESOURCES TABLE

REAGENT or RESOURCE SOURCE IDENTIFIER

Antibodies

GAPDH antibody Cell Signaling Biotechnology Cat# 2118S

b-actin antibody Sigma Aldrich Cat# A1978

ACSL1 antibody Cell Signaling Technology Cat# 4047

ACSL3 antibody Santa Cruz Biotechnology Cat# sc-166374

ACSL4 antibody Abcam Cat# ab155282

ACSL5 antibody Novus Biologicals Cat# NPB1-59645

ACSL6 antibody Abcam Cat# ab154094

Bacterial and Virus Strains

ACSL3 shRNA (r) Lentiviral Particles Santa Cruz Cat# sc-270649-V

Control shRNA Lentiviral Particles-A Santa Cruz Cat# sc-108080

Lactobacillus gasseri Lauer and Kandler (ATCC 33323) American Type Culture Collection Cat # 33323

Chemicals, Peptides, and Recombinant Proteins

D-(+)-Glucose solution Sigma Aldrich Cat# G8769

[3-3H] Glucose Perkin Elmer Part# NEC042V250UC

Oleic acid Sigma Aldrich Cat# O1008

Linoleic acid Sigma Aldrich Cat# L1376

L-a-phosphatidylcholine Sigma Aldrich Cat# P7443

Sodium salt of taurcholic acid Sigma Aldrich Cat# 86339

Intralipid Sigma Aldrich Cat# I141

MK-329 Tocris Bioscience Cat# 2304

Exendin-9 Tocris Bioscience Cat# 2081

Tetracaine Sigma Aldrich Cat #T7383

Lactobacilli MRS Broth (ATCC Medium 416) Fischer Scientific Cat# DF0882170

GW4064 Sigma Aldrich Cat# G5172

Sodium carboxymethyl cellulose Sigma Aldrich Cat# 419273

Mutaniolysin Sigma Aldrich Cat# M9901

RNase A Qiagen Cat# 19101

Tris-HCl Roche Cat# 10812846001

EGTA Sigma Aldrich Cat# E3889

EDTA Sigma Aldrich Cat# E9884

Nonidet P40 Roche Cat# 11332473001

Sodium orthovanadate Sigma Aldrich Cat# S6508

Sodium fluoride Sigma Aldrich Cat# S7920

Dithiotritol Sigma Aldrich Cat# D9779

Protease inhibitor cocktail Roche Cat# 11836153001

HR series NEFA-HR color reagent A Wako Diagnostics Cat# 999-34691

HR series NEFA-HR solvent A Wako Diagnostics Cat# 995-34791

HR series NEFA-HR color reagent B Wako Diagnostics Cat# 991-34891

HR series NEFA-HR solvent B Wako Diagnostics Cat# 993-35191

NEFA standard solution Wako Diagnostics Cat# 276-76491

Wako NEFA linearity set Wako Diagnostics Cat# 997-76491

Critical Commercial Assays

Rat Insulin Radioimmunoassay EMD Millipore Cat# RI-13K

Pierce 660 nm Protein Assay Thermo Fischer Scientific Cat# 22660

DNA Clean and Concentrator Kit Zymo Research Cat# D4003T

(Continued on next page)

e1 Cell Metabolism 27, 572–587.e1–e6, March 6, 2018

Page 19: Lactobacillus gasseri in the Upper Small Intestine Impacts

Continued

REAGENT or RESOURCE SOURCE IDENTIFIER

Experimental Models: Organisms/Strains

Sprague Dawley Rats Charles River Laboratories Strain# 400

C57BL6/J Mice Jackson Laboratories Strain# 000664

Software and Algorithms

QIIME bioinformatics pipeline Open Source qiime.org

R programming language Open Source RRID: SCR_001905

GraphPad Prism 7 GraphPad Software www.graphpad.com

CONTACT FOR REAGENT AND RESOURCE SHARING

Further information and requests for resources and reagents should be directed to andwill be fulfilled by the Lead Contact, Tony Lam

([email protected]).

EXPERIMENTAL MODEL AND SUBJECT DETAILS

AnimalsEight-week old adult male Sprague-Dawley rats (280-300g) were obtained from Charles River Laboratories (Montreal, Quebec,

Canada). Eighteen-week-old adult male C57/BL/6 mice (25-30g) were obtained from Jackson Laboratories (Bar Harbor, Maine,

USA). Sex-differences were not examined in the current manuscript. Both rats and mice were housed individually and maintained

on a standard light-dark cycle with ad libitum access to chow (Teklad Diet #7002, Harlan Laboratories) (Table S1) and water. All

animal protocols were reviewed and approved by the Toronto General and Western Animal Care Committee at UHN.

METHOD DETAILS

Surgical ProceduresAnimals were given at least 6 days to acclimatize before surgeries were performed. Rat surgeries were performed 4 days prior to

intravenous glucose tolerance tests (ivGTT) and clamp experiments. An upper small intestinal catheter was placed 6 cm distal to

the pyloric sphincter to target the lower duodenum and upper jejunum (between 6 and �12 cm distal to the pyloric sphincter) during

a 50-min upper small intestinal infusion, while carotid artery and jugular vein cannulations were performed for infusion and sampling

during the clamp. Two to three days before the mouse pancreatic clamp studies, a catheter was placed in the upper small intestine

(1.5-2 cm distal to the pyloric sphincter) to target the lower duodenum and upper jejunum, while a catheter was placed in the jugular

vein for infusion purposes and tail sampling was performed during the clamp studies. Following surgery, both rats and mice were

housed individually and maintained on a standard light-dark cycle with ad libitum access to food and water. Food intake and

body weight was monitored to ensure recovery from surgery, and rats that did not recover to 90% of their baseline body weight

were excluded from the study. Rats were randomly designated into treatment groups prior to experiments and no blinding was

done during the experimental procedures described below.

Virus InjectionA subset of rats received an upper small intestinal lentiviral injection prior to the insertion of the intestinal cannula as described (Cote

et al., 2015; Kokorovic et al., 2011). Briefly, the upper small intestine was elevated from 6 to 12 cm distal to the pyloric sphincter and

was ligated with sutures at both ends to restrict outward flow of virus and inward flow of intestinal fluids. The elevated intestinal

portion was first flushed with 0.2-0.5 mL of saline via a 23-gauge needle inserted right below the 6 cm ligation, and then 0.2 mL

of the lentivirus expressing mismatch or ACSL3 shRNA (both at 1.0 x 106 IFU; 20 mL was diluted into a total volume of 0.2 mL for

injection) (Santa Cruz, CA, USA) was administered via the 23-gauge needle. After 20 min, ligations sutures were removed and the

intestine was flushed with saline. A catheter was then inserted in the site of the virus injection and vascular cannulations were per-

formed as described above.

High-fat Diet ModelRats were placed on a lard oil-enriched high fat diet (HFD; TestDiet #571R, PurinaMills, IN, USA) (Table S1) and allowed to overeat for

three days before the ivGTT or pancreatic clamp procedure, which results in hyperphagia (Figures S1D and S2J) and upper small

intestinal lipid sensing defects (Breen et al., 2011; Cheung et al., 2009; Rasmussen et al., 2012) but not obesity (Figures S1E and

S2K). The same 3-day HFD-induced hyperphagic response has been documented by others to be sufficient to induce insulin and

leptin resistance but insufficient to increase body weight (Morgan et al., 2004; Scherer et al., 2012; Thaler et al., 2012), thus repre-

senting an early onset HFD-induced obese model. Under rare circumstances when rats were not hyperphagic they were excluded

from the study.

Cell Metabolism 27, 572–587.e1–e6, March 6, 2018 e2

Page 20: Lactobacillus gasseri in the Upper Small Intestine Impacts

Intravenous Glucose Tolerance TestExperiments were performed in overnight-fasted rats (16-18 hr) 4 days after surgery. Basal blood samples were obtained in

conscious, unrestrained rats immediately before beginning the gut infusion (0.01 mL/min), which began at t = -15 min and was main-

tained until the end of the experiment at t = 35 min. At t = 0 min blood samples were obtained and an intravenous bolus of glucose

(20% glucose, 0.25 g/kg) was injected into the jugular vein and flushed with saline. Blood was collected via the carotid arte. ry cath-

eter tomeasure plasma glucose and insulin levels for 35min following the glucose injection, as previously described (Yue et al., 2016).

Rat Pancreatic (Basal Insulin) Clamp ProcedureThe clamp was performed as described previously (Cheung et al., 2009; Wang et al., 2008; Yue et al., 2016). After a 4-6 hr fast, a

primed continuous infusion of [3-3H]-glucose (40 mCi bolus; 0.4 mCi min�1; Perkin Elmer, Woodbridge, ON, Canada) was given

from t = 0 to t = 200 min (start to end of the experiment) through an intravenous catheter to assess glucose kinetics based on the

tracer dilution methodology. Starting at t = 90min and continuing to the end of the experiment (t = 200), a pancreatic clamp was con-

ducted through infusion of insulin (1.2mU kg�1 min�1) and somatostatin (3 mg kg�1min�1) to inhibit endogenous insulin and glucagon

secretion. Somatostatin-14was used, which has been shown to have a veryminor inhibitory effect on GLP-1 release when compared

with somatostatin-28 (Brubaker, 1991; Chisholm and Greenberg, 2002; Hansen et al., 2000). Furthermore, upper small intestinal

glucose infusion was shown to stimulate GLP-1 release during the pancreatic basal insulin clamp with the same dose of somato-

statin-14 (Bauer et al., 2018), suggesting this dose of somatostatin-14 does not inhibit GLP-1 release under these conditions.

Between t = 120 and t = 200, blood samples were taken every 10 min to determine the rate of an exogenous 25% glucose infusion

needed to maintain basal glucose levels (averaged from t = 60-90). At t = 150 min, the upper small intestinal infusion (0.01 mL/min)

was started and continued until the end of the experiment (t = 200). To determine insulin levels and specific activity of [3-3H]-glucose,

plasma samples were taken every 10 min during the basal period (t = 60-90) and the gut infusion period (t = 150-200). Upon comple-

tion of the experiment rats were anaesthetized and upper small intestinal microbiota luminal samples (between 6 and 15 cm distal to

the pyloric sphincter), as well asmucosal scrapings (between 6 and 10 cmdistal to the pyloric sphincter), were collected, snap-frozen

in liquid nitrogen and stored at -80�C until use.

Mouse Pancreatic (Basal Insulin) Clamp ProcedureMice were fasted for 4-6 hr prior to experimentation. Through an intravenous catheter, a primed-continuous intravenous infusion

of [3-3H]-glucose (1 mCi bolus; 0.1 mCi/min; Perkin Elmer, Woodbridge, ON, Canada) was given from t = 0 to t = 170 min (start to end

of the experiment) to assess glucose kinetics based on the tracer dilution methodology. Starting at t = 60 and continuing to the end of

theexperiment (t=170), a pancreaticclampwasconducted through infusionof insulin (1.4mU/kg/min) andsomatostatin (8.3mg/kg/min).

Blood samples were taken via tail sampling every 10 min to determine if an exogenous 10% glucose infusion was needed to maintain

basal glucose levels (averaged from t = 50-60) between t = 60 to t = 170.Upper small intestinal infusions (2mL/min) of oleic acid or linoleic

acid (4 x 10-3 kcal/min) were performed from t = 120 to t = 170. To determine specific activity of [3-3H]-glucose levels, plasma samples

were taken every 10 minutes during the basal period (t = 50-60) and the upper small intestinal infusion period (t = 120-170).

Fatty Acid Preparation for Upper Small Intestinal InfusionsOleic acid (99% purity; Sigma, St. Louis, MO) and linoleic acid (99% purity; Sigma) were administered at 2 kcal/mL to mimic the con-

centration of the Intralipid infusate, which has been shown to lower glucose production when infused directly into the upper small

intestine at 0.01 mL/min (Wang et al., 2008). In addition, this dose of oleic acid and linoleic acid (total of 1 kcal delivered) has

been shown to inhibit food intake in rats when infused into the upper small intestine (Phifer and Berthoud, 1998; Woltman et al.,

1995). Fatty acid solutions were prepared as previously described (Woltman et al., 1995). Briefly, fats were emulsified in phosphate

buffered saline (PBS; 115 mM NaCl, 16.5 mM NaH2PO4, 6.75 mM Na2HPO4, and 5 mM KCl; pH 6.4) containing 0.1% wt/vol

(1.24 mmol/mL) lecithin (L-a-phosphatidylcholine, 99% pure; Sigma) and 1% wt/vol (17.5 mmol/mL) sodium taurocholate (sodium

salt of taurcholic acid, 98% pure; Sigma). These bile acid concentrations are comparable to levels detected in the upper small intes-

tine of humans in postprandial (90min after a standardizedmeal) conditions [1.0 – 2.5 mmol/mL (lecithin) and 12 – 17 mmol/mL (sodium

taurocholate)] (Clarysse et al., 2009; Mansbach et al., 1975). Solutions were emulsified until homogenous with an Autotune Series

High Intensity Ultrasonic Processor.

Upper Small Intestinal InfusionsThe following substances were infused into the upper small intestine at a rate of 0.01 mL/min as described: (1) saline, (2) PBS + bile

salts (PBS with lecithin and sodium taurocholate) (3) Intralipid (2 kcal/mL) (4) Oleic acid (2 kcal/mL; in PBS and bile salts) (5) Linoleic

acid (2 kcal/mL; in PBS and bile salts), (5) MK-329 (0.08 mg/mL; Tocris Bioscience), (6) Exendin-9 (15 mg/mL; Tocris Bioscience) and

(6) tetracaine (1 mg/mL; Sigma). The dose of Intralipid (1 kcal delivered over 50 min) was chosen to ensure the effects observed were

in the pre-absorptive state (Greenberg et al., 1995; Wang et al., 2008). The dose used for oleic acid and linoleic acid were chosen to

mimic the Intralipid dose (1 kcal). In addition, this dose of oleic acid and linoleic acid (total of 1 kcal) has been shown to inhibit food

intake in rats (Phifer and Berthoud, 1998; Woltman et al., 1995). The dose of MK-329 was previously shown to inhibit lipid sensing in

the upper small intestine (Cheung et al., 2009). The dose of Exendin-9 has been shown to inhibit the effects of ileal fatty acid sensing

(Zadeh-Tahmasebi et al., 2016). The dose of tetracaine was previously shown to inhibit the glucose production-lowering effects of an

upper small intestinal Intralipid infusion (Wang et al., 2008).

e3 Cell Metabolism 27, 572–587.e1–e6, March 6, 2018

Page 21: Lactobacillus gasseri in the Upper Small Intestine Impacts

Microbiota TransplantDonor rats were anaesthetized after a 4-6 hr fast and the upper small intestinal luminal contents were removed from the upper small

intestine (between �6-15 cm distal to the pyloric sphincter to collect a sufficient amount of luminal contents for transplantation). The

luminal contents were homogenized and diluted 1:4 in PBS and 0.5mL of the luminal contents were slowly infused (over the course of

30 seconds) into 4-6 hr-fasted recipient rats via the upper small intestinal gut line to target the lower duodenum and upper jejunum. To

validate that the transplant effectively altered the upper small intestinal microbiota composition, the upper small intestinal luminal

contents of anaesthetized recipient rats obtained from �6-15 cm distal to the pyloric sphincter was collected immediately after

the infusion experiments, snap frozen in liquid nitrogen and stored at -80�C for subsequent 16S rRNA gene sequencing (see below).

For the colonic microbiota transplant, the upper small intestinal contents were removed and prepared for infusion in the sameway as

the upper small intestinal microbiota transplant. The luminal contents were then transplanted into recipient rats via a colonic cannu-

lation located 2 cmdistal to the cecum. Recipient rats were subjected to the ivGTT or the basal insulin euglycemic clamp the following

day. Recipient rats undergoing the ivGTT were immediately fasted following the transplant procedure (16-18 hr fast). Recipient rats

undergoing the clampswere given their original diet (the diet being consumed prior to transplantation) and fasted for 4-6 hr prior to the

clamp procedure.

Microbiota Heat-ShockBefore transferring the upper small intestinal luminal contents to the recipient rat, bacteria was heat-killed by incubation at 95-100�Cfor two 5 min intervals. The heat-treated samples were cultured on LB agar plates at 37�C for 96 hr to prove that no viable organisms

remained.

Microbiota FilterTo obtain bacteria-free samples, luminal contents were filtered through 0.2 mm (Millex) filters (Gareau et al., 2011) before being trans-

ferred to recipient rats. The filtered samples were cultured on LB agar plates at 37�C for 96 hr to prove that no viable organisms

remained.

Lactobacillus Gasseri AdministrationLactobacillus gasseri Lauer and Kandler (ATCC 33323) was freshly prepared for each administration. Briefly, the probiotic was

grown overnight in Lactobacilli MRS Broth (ATCC Medium 416) under anaerobic conditions at 37�C the evening prior to adminis-

tration. The following day, 109 colony-forming units (cfu) were dissolved in a total volume of 0.5 mL of PBS and slowly infused (over

the course of 30 seconds) into 4-6 hr-fasted recipient rats via the upper small intestinal gut line to target the lower duodenum and

upper jejunum. Control HFD-fed rats received 0.5 mL of vehicle PBS. Basal insulin euglycemic clamps were performed the following

day after a 4-6 hr fast. The Lactobacillus gasseri probiotic dose (109 cfu) was shown to increase bile acid excretion in male Sprague

Dawley rats (Usman and Hosono, 2001). In addition, a comparable dose (5 x 109 cfu) of a probiotic mixture containing a variety of

species from the Lactobacillus genus increased bile acid excretion and decreased small intestinal FXR expression in mice (Degir-

olamo et al., 2014).

GW4064 AdministrationA subset of Lactobacillus gasseri-infused and control PBS-infused HFD rats were administered with the FXR agonist, GW4064

(30mg/kg; Sigma; dissolved in 1% carboxymethyl cellulose; administered via the gut line over the course of 30 seconds and followed

by a saline gut line flush), 30 min following the Lactobacillus gasseri or PBS administration and again the next morning after a 4-6 hr

fast (immediately prior to the basal insulin-euglycemic clamps). Control HFD-fed rats received 1%carboxymethyl cellulose. The dose

of GW4064 (30 mg/kg) was chosen based on previous work reporting an inhibition of glucose-stimulated GLP-1 release in response

to GW4064 (30 mg/kg) administered via gavage for 5 days (Trabelsi et al., 2015). In addition, oral GW4064 was shown to inhibit

probiotic-induced increased bile acid excretion after two treatments at a comparable dose (45 mg/kg) (Degirolamo et al., 2014).

Lactobacillus gasseri-infused HFD rats were initially given a single dose of GW4064 (30 mg/kg) 30 min following the probiotic admin-

istration. However, we found that a single GW4064 treatment only partially blocked the restoration of upper small intestinal lipid

sensing in Lactobacillus gasseri-infused HFD rats during the pancreatic clamps (Gut Lipid-infused, GP was 8.9 mg kg�1 min�1

(n = 1) vs. Gut saline-infused, GP was 11.4 mg kg�1 min�1 (n = 1)). Given that the effect of GW4064 on glucose-stimulated GLP-1

release and bile acid excretion occurred following daily treatment for 5 and 2 days, respectively (Degirolamo et al., 2014; Trabelsi

et al., 2015), we next administered the same dose of GW4064 (30 mg/kg) immediately following the Lactobacillus gasseri-infusion

and again the next morning prior to the clamp. We found that two treatments with GW4064 (30 mg/kg) blocked the restorative effect

of Lactobacillus gasseri administration on upper small intestinal lipid sensing, with no metabolic impact in non-probiotic-infused

(PBS-infused) HFD rats (Figure 7D).

Genomic DNA Extraction and 16S rRNA Gene SequencingUpper small intestinal luminal contents obtained from between �6-15 cm distal to the pyloric sphincter were collected from rats

immediately following upper small intestinal saline-infused basal insulin-euglycemic pancreatic clamps. Genomic DNA extraction

and purification was performed as described previously (Whelan et al., 2014). Intestinal luminal contents were added to a 2mL plastic

screw top tube containing 0.2 g of 0.1 mm glass beads (MO BIO laboratories, Inc.). Subsequently, 800 mL of 200 mM NaPO4 (pH 8)

Cell Metabolism 27, 572–587.e1–e6, March 6, 2018 e4

Page 22: Lactobacillus gasseri in the Upper Small Intestine Impacts

and 100 mL of guanidine thiocyanate-EDTA-N-lauroyl sarcosine were added to the tube and homogenization was performed for 3min

with a bench-top bead-based homogenizer (Minilys personal homogenizer). For the first enzymatic lysis step, 50 mL of mutanolysin

(Sigma-aldrich, 10 U/mL), 10 mL of RNase A (Qiagen, 10 mg/mL) and 100 mL of 5M NaCl were added to each sample. The samples

were vortexed and incubated at 65�C (water bath) for 1 hr. After incubation, the tubes were spun at maximum speed for 5 min in a

bench-top centrifuge and 900 mL of the supernatant was removed and transferred to a 2 mL tube containing 900 mL of phenolchloro-

form-isoamyl alcohol (25:24:1). After vortexing the tubes for 10 seconds (shear DNA), they were again spun at maximum speed in the

bench-top centrifuge for 10 min. After centrifugation, the top layer was carefully transferred to a 1.5 mL microfuge tube. DNA clean

and concentrator-25 kit (Zymo Research) was used for purifying the DNA, where DNAwas eluted from the columnwith 50 mL of water

and the samples were diluted to a final concentration of 20 ng/mL. DNA samples were quantified using Nanodrop spectrophotometer

(NanoDrop 2000c, Thermo Scientific) and isolated DNA was stored at -20�C.PCR amplification of the V3 region of the 16S rRNA gene was performed as previously described (Bartram et al., 2011) with the

following modifications: a 50 mL reaction containing 1.25 mM MgCl2, 2.5 mM of each dNTP, 100 nM of each barcoded primer,

and 1.25 U Taq polymerase. Amplifications were carried out in triplicate, where the amplified product was purified by separation

with agarose gel electrophoresis and gel extraction. PCR conditions consisted of an initial denaturation at 94�C for 2 min followed

by 30 cycles at 94�C for 30 seconds, 50�C for 30 seconds and a final elongation at 72�C for 10 min. Purified PCR products were

sequenced using the Illumina MiSeq platform at the McMaster Genome Facility (Hamilton, ON, Canada).

Sequence Processing and Data AnalysisPCR products were sequenced using Illumina MiSeq with paired-end reads. 16S rRNA gene sequencing was completed as previ-

ously described (Whelan et al., 2014). Custom Perl scripts were developed in-house to process the sequences. First, Cutadapt

was used to trim these sequences to the V3 region, removing any sequences surpassing this region. Next, sequences were aligned

with their pair using PANDAseq (Masella et al., 2012). During this alignment any mismatches or ambiguous bases were culled. Oper-

ational taxonomic units (OTUs) were picked using AbundantOTU and as described previously (Ye, 2011) with a clustering cut-off of

97%. Taxonomy of the resultant OTUs was assigned via comparison of a representative sequence of the unit to the GreenGenes

reference database (4 February 2011 release) (DeSantis et al., 2006) using the Ribosomal Database Project (RDP) classifier

(Wang et al., 2007). Summaries of the relative abundances of taxonomies, as well as beta and alpha diversity measurements

were calculated using Quantitative Insights Into Microbial Ecology (QIIME, v1.9.1) (Caporaso et al., 2010). The rarefaction depth

was 10,000 sequences/sample. Three-dimensional Principal Coordinate Analysis plots were visualized using Emperor (Vazquez-

Baeza et al., 2013). The significance of clustering was determined by passing the UniFrac distance matrices through QIIME using

Adonis and ANOSIM non-parametric statistical methods. LefSe (Linear Discriminant Analysis with Effect Size) analyses were per-

formed on the website https://huttenhower.sph.harvard.edu/galaxy/ (Segata et al., 2011). The differential features were identified

at the Phylum (p), Class (c), Order (o), Family (f), and Genus (g) levels. The alpha value for the factorial Kruskal-Wallis test among

classes was <0.05 and the threshold on the logarithmic LDA score for discriminative features was >2.0. For group comparisons,

the one-way ANOVA test was used with a Tukey’s multiple comparisons test. Statistical analyses were performed using GraphPad

Prism. A p value <0.05 was considered significant.

Tissue Collection and Western BlottingA section of the upper small intestine �6-10 cm distal to the pyloric sphincter, containing both lower duodenum and upper jejunum,

was removed from anaesthetized rats immediately following the infusion experiments. The upper small intestinal mucosa was sepa-

rated from the smooth muscle and quickly placed in liquid nitrogen. The tissues were lysed on ice with a handheld homogenizer in a

lysis buffer containing 50 mM Tris-HCl (pH 7.5), 1 mM EGTA, 1 mM EDTA, 1% (w/v) Nonidet P40, 1 mM sodium orthovanadate,

50 mM sodium fluoride, 5 mM sodium pyrophosphate, 0.27 M sucrose, 1 mM Dithiotritolo (DTT) and protease inhibitor cocktail

(Roche). The protein concentration of homogenized tissues was determined using the Pierce 660 nm protein assay (Thermo Sci-

entific). 50 mg of tissues lysates were subject to electrophoresis on 10% acrylamide gels and transferred to nitrocellulose mem-

branes. The membranes were incubated for 1 hr with blocking buffer (5% bovine serum albumin in TBS-T), followed by incubation

in primary antibody 2 hr at 4�C. b-actin (Sigma, #A1978) and GAPDH (Cell Signaling, #2118S) were used as loading controls. The

antibodies were prepared in 5% bovine serum albumin in TBS-T at the following dilutions: 1:1000 for ACSL1 (#4047, Cell Signaling

Technology), 1:250 for ACSL3 (sc-166374, Santa Cruz Biotechnology), 1:10000 for ACSL4 (ab155282, Abcam), 1:1000 for ACSL5

(NPB1-59645, Novus Biologicals), and 1:10000 for ACSL6 (ab154094, Abcam). The blots were then washed three times in TBS-T

and incubated with a horseradish peroxidase (HRP)-linked secondary antibody (1:4000 dilution in 5% skim milk) for 2 hr. Blots were

washed 5 times in TBS-T and protein expression was detected using an enhanced chemiluminescence reagent (Clarity Western

ECL Blotting Substrate, Bio-Rad). Immunoblots were imaged using a MicroChemi chemiluminescent imaging system (DNR Bio-

Imaging Systems).

Biochemical AnalysisPlasma insulin concentrations were measured using a radioimmunoassay (Linco Research, St Charles, MO) and plasma free fatty

acid concentrations were measured by an enzymatic assay (Wako Pure Chemical Industries, Osaka, Japan).

e5 Cell Metabolism 27, 572–587.e1–e6, March 6, 2018

Page 23: Lactobacillus gasseri in the Upper Small Intestine Impacts

QUANTIFICATION AND STATISTICAL ANALYSIS

The sample size for each group was chosen based on study feasibility and previously published experiments. Data from all groups

showed normal variance. Therefore, the results were analyzed using unpaired Student’s t test when analyzing statistical difference

between two groups.When comparisons weremade acrossmore than two groups, ANOVAwas performed and if significant this was

followed by Tukey’s post hoc test. Measurements that were taken repeatedly over time were compared using repeated measures

two-way ANOVA; if the time and treatment interaction between groups was found to be significant Tukey’s multiple comparisons

test was used to determine the statistical significance at specific time points between groups. p value < 0.05 was considered

statistically significant. For clamp experiments, the time period of 60-90 min was averaged for the basal condition and the period

of 180-200 min was averaged for the clamp condition.

Cell Metabolism 27, 572–587.e1–e6, March 6, 2018 e6