• No results found

Willow_Liu_HonorsThesis.pdf

N/A
N/A
Protected

Academic year: 2020

Share "Willow_Liu_HonorsThesis.pdf"

Copied!
37
0
0

Loading.... (view fulltext now)

Full text

(1)

The Effect of Dietary Fibers on Gastrointestinal Anatomy and

Intestinal Microbial Communities in Mice

Jintong Liu

Honors Thesis Department of Nutrition

Gillings School of Global Public Health University of North Carolina at Chapel Hill

April 2020

Approved by

(2)

Abstract

Objectives: Grain-based diets (GBDs) are widely used in rodent studies, but the lack of control of ingredients in GBDs and potential batch to batch variation limit their utility. Purified diets (PDs)—diets with known sources and quantities of all nutrients—offer solutions and permit investigators to control for diet, a major environmental factor, in rodent studies. However, mice fed a standard PD display abnormal gastrointestinal (GI) anatomy and greater adiposity

compared to mice fed a GBD—Purina 5001. Interestingly, the addition of a soluble fiber (inulin) to PDs (which usually only contain the insoluble fiber cellulose) ameliorates these adverse effects. Yet, the impact of PDs modified with soluble fibers from mixed sources on GI anatomy and the intestinal microbiota has not been investigated. We therefore sought to identify a

combination of fibers in PDs that best recapitulate the GI health and intestinal microbiota of mice fed a GBD, while also including an additional reference GBD (Teklad 2020SX).

Methods: 7-week-old C57BL/6J male mice were individually housed and randomly assigned to a diet (two GBDs and four PDs with varying fiber composition) for 28 days. To assess changes in GI anatomy, small intestinal length, colon length, cecal weight, and colon weight were recorded at tissue harvest. Cecal contents, colon contents, and fecal pellets were collected for 16S rRNA gene sequencing to compare microbial profiles across different GI niches and between diets using the Quantitative Insights Into Microbial Ecology 2 pipeline (QIIME 2) pipeline.

(3)

consuming either GBD in all GI niches (p<0.05, ANOSIM). Microbiotas were distinct between mice fed either Purina 5001 or Teklad 2020SX GBDs (p<0.01, ANOSIM). Mice fed low soluble fiber PDs (≤ 25% soluble fiber) also produced significantly different microbial communities than mice fed high soluble PDs (≥ 75% soluble fiber) (ANOSIM, p<0.001). Unexpectedly, the

microbial richness of mice decreased with increasing soluble fiber content in PDs. The relative abundance of Lactococcus spp. and the [Eubacterium] xylanophilum group were different between mice fed GBDs and PDs, while Romboutsia spp. had a higher relative abundance in mice fed low soluble fiber PDs compared to other diet groups.

Conclusions: Mice fed PDs with high soluble fiber content (≥ 75% soluble fiber) best

(4)

Introduction

Gut Microbiota

The gut microbiota is a complex and dynamic community of microorganisms—including bacteria, archaea, fungi, protists, and viruses—that reside within the gastrointestinal (GI) tract of animals1. In contrast to pathogenic microorganisms that cause disease, the gut microbial

community largely contains symbiotic and commensal microbes, which provide various benefits to their host. With 3.8×1013 cells and over 1,900 species residing in the human intestine, bacteria

comprise the most diverse group of these microorganisms2–4. Therefore, while all members

contribute to the makeup and function of this community, bacteria are the most extensively studied.

Initially, investigators were restricted to studying the gut microbiota using culture-based methods. However, this approach provides limited information because estimates report that only 10-50% of intestinal bacteria are culturable5. In addition, the costly and time-consuming nature

of this approach limits the number of samples investigators can analyze. With the reduced cost of high throughput sequencing and the advances of -omics technologies (e.g., metabolomics,

metagenomics, and transcriptomics), researchers are now able to investigate the gut microbiota more comprehensively6. These technologies allow researchers to identify the taxonomic

composition of the majority of microbes within a community, including non-culturable microbes, by sequencing microbial genes. Present in all bacteria but not in eukaryotes, the 16S rRNA gene is often used in characterizing complex microbial communities and reconstructing

phylogenies7,8. With conserved regions that are universal across bacteria and variable regions

(5)

advances have led to seminal discoveries regarding the intricate relationships between the intestinal microbiota and human health6.

The gut microbiota serves numerous functions in the host. For example, the intestinal microbiota helps break down nutrients in the intestine, maintains the integrity of the intestinal epithelium, and ensures the proper development of the immune system9. The gut microbiota also

influences the host’s behavior by exerting its effect via the gut-brain axis10. While a healthy gut

microbiota promotes the health of the host, a disturbed (or dysbiotic) gut microbiota contributes to GI dysfunction and factors related to metabolic syndrome (i.e., obesity), inflammatory bowel diseases (IBD), and cardiovascular diseases11–14.

Modulation of enteric microbial communities to restore normal GI functions, with probiotics, prebiotics, and fecal microbiota transplantation (FMT), has also been an active field of research14. Probiotics are living microorganisms that are beneficial to the host when

consumed, while prebiotics are non-digestible carbohydrates that selectively promote the growth and metabolism of certain beneficial bacteria15. In theory, both probiotics and prebiotics promote

the restoration of a healthy microbial community; however, more research is required to uncover the mechanism and efficacy of treating diseases with probiotics and prebiotics. FMT is a

procedure where the fecal microbiota from a healthy donor is transferred to a recipient believed to harbor an altered gut microbiota16. FMT has shown to be most effective with treating

Clostridium difficile infection (CDI), with the resolution rate usually higher than 85%16,17.

(6)

The composition of the intestinal microbiota is influenced by multiple host factors, including diet, age, geographic location, sex, and antibiotic use14,21. Host diet has a significant

effect on the resulting composition of the microbial community. For example, it was reported that short-term consumption of diets composed entirely of animal or plant products rapidly altered microbial community structure22. Rodent studies also show that mice fed a high-fat diet

harbor a gut microbiota distinct from mice fed a low-fat diet23. Further, recent studies have

demonstrated that supplementing dietary fibers alone significantly alter the microbial community, in both human subjects and rodents24,25.

Dietary Fibers and Short Chain Fatty Acids (SCFAs)

Dietary fibers are carbohydrate polymers that cannot be broken down by human digestive enzymes26,27. Fibers therefore escape digestion in the small intestine and pass into the cecum and

colon, where they become available to the microbial community for fermentation26. There are

two main types of dietary fibers: water insoluble and water soluble. Insoluble fibers, such as cellulose and hemicellulose, are poorly digested by the gut microbiota28. Insoluble fibers increase

fecal bulking and stool water content in the intestine, resulting in increased GI transit time by stimulating mucus secretion and peristalsis29. On the other hand, the viscosity of soluble fibers

induces gastric distention and slows gastric emptying rate to promote satiety and reduce calorie intake29. Recent studies also reveal that the lack of soluble fiber causes microbes to extract

energy from the colonic mucus, degrading the colonic mucus layer that protects against

pathogens30,31. Therefore, dietary fibers are essential for maintaining the integrity of the GI tract.

(7)

propionate, and butyrate, are bacterial metabolites produced from the fermentation of soluble fibers and are beneficial to the host. Butyrate is the primary energy source for colonocytes, while propionate is a substrate for gluconeogenesis in liver26,30. Colonic delivery of propionate has

been shown to increase postprandial peptide YY and glucagon-like peptide-1 secretion in humans, resulting in reduced food intake32. SCFAs also affect the nervous system. For example,

acetate crosses the blood-brain barrier and activates hypothalamic neurons to drive satiety33. In

addition, SCFAs regulate sympathetic nervous system activity via G protein-coupled receptor 41 (GPR41), allowing control of body energy expenditure by promoting the maintenance of

metabolic homeostasis34.

Laboratory diets in rodent models

The two commonly used diet types for rodent models in laboratory studies are grain-based diets (GBDs) and purified diets (PDs). GBDs, also called chow diets, are produced from agricultural and animal by-products, such as ground corn, ground oats, alfalfa meal, soybean meal, and ground wheat35. Though they are relatively inexpensive to produce and provide

adequate nutrition, there are several drawbacks with GBDs in rodent studies. Chow formulas are generally “closed” formulas, meaning commercial manufacturers do not disclose the exact amount and source of each ingredient35. Even when some diets are open formulas, the

composition of the components varies with the environmental conditions under which they were grown and processed36. The variability of GBDs makes it hard, if not impossible, for researchers

(8)

reproducibility37. These GBD drawbacks also have significant implications regarding gut

microbiota research, as the complex microbial community is significantly influenced by diet. Fortunately, purified diets (PDs), also called purified ingredient diets, are a potential solution to these issues. PDs are composed of highly refined ingredients, producing well-defined diets with precisely measured amounts of each ingredient and minimal batch-to-batch

variability36,38. PDs not only increase experimental reproducibility by controlling for a known

environmental variable, but they also allow researchers to modify any specific ingredient to investigate the effects of diets with varying macro- and micronutrient compositions35.

Unfortunately, most of the currently used PDs, including AIN-76A and AIN-93, contain little or no soluble fiber39,40. A recent study by Chassaing et al. showed that the lack of soluble fibers in

PDs resulted in increased adiposity and abnormal GI anatomy in mice, which were ameliorated when inulin, a soluble fiber, was added into the diet41. With the current knowledge on the

importance of soluble fibers and the recognition of PDs being a powerful tool, PDs containing soluble fibers are highly desirable.

(9)

Methods

Diets

Six different diets, with similar macro-nutrient contents and essential micronutrients, were used in this study. The two GB diets used, Teklad 2020SX from (Envigo; Madison, WI) and Purina 5001 (Nestlé Purina PetCare; St. Louis, MO), consisted of unknown dietary fiber amounts and compositions. Four purified diets with varying composition of fibers were

(10)
(11)

Mice

Six-week-old male wild-type (WT) C57BL/6J mice were purchased from Jackson Laboratory (Bar Harbor, ME). All animals (n=30 mice) were housed in individually ventilated cages with ad libitum access to autoclaved water and food. All autoclaved cages contained bedding, a hut, and a nestlet. During the seven-day acclimation period, all mice were fed Teklad 2020X. Mice were then randomly assigned to one of the six diet groups for 28 days (n=5

mice/group). Food consumption and body weight were measured on day 0 (when mice were placed in individual cages with Teklad 2020SX), on day 7 (when mice were randomly assigned to receive one of the six diets), on day 21, and on day 35 (when the study terminated). Food was replaced and cages were changed on day 7 and day 21. All animal care and experimental

protocols were approved by Institutional Animal Care and Use Committee (IACUC) of UNC-Chapel Hill on protocol 17-136.

Fecal and Tissue Sample Collection

(12)

DNA Extraction

Bacterial DNA was isolated from fecal pellets, cecal contents, and colon pellets, using a phenol-chloroform extraction method followed by a DNA clean-up42. One fecal pellet, two colon

pellets, or 100-mg of cecal contents, were suspended in 750 µl of lysis buffer (200 mM NaCl, 100 mM Tris (pH 8.0), 20 mM EDTA, and 20 mg/ml lysozyme) with 300 mg of 0.1 mm glass beads (BioSpec, Bartlesville, OK). The mixture was vortexed briefly and incubated at 37 °C for 30 minutes. For each sample, 85 µL of 10% SDS and 20 µL of Proteinase K (15 mg/mL) from the Qiagen DNA Stool Mini Kit (Qiagen, Valencia, CA) was added, and the mixture was incubated for another 30 minutes at 60 °C.

Bacterial cells were physically disrupted by adding 500 µL of 25:24:1 solution of phenol:chloroform:isoamyl alcohol and homogenizing in a bead beater (TeSeE Precess 48 Homogenizer, Bio-Rad, Hercules, CA) for 90 seconds at 5300 rpm. Samples were then centrifuged at 13000 rpm for 5 minutes at 20 °C. The resulting supernatant was transferred to another 500 µL volume of 25:24:1 phenol:chloroform:isoamyl alcohol, vortexed, and

centrifuged. The supernatant was then transferred to 500 µL of 100% chloroform, again

vortexed, and centrifuged. Lastly, the supernatant was transferred to 1 mL100% ethanol with 50

µL of 3 M sodium acetate. Samples were then stored at -80 °C for 60 minutes to promote DNA precipitation.

Samples were centrifuged at 13000 rpm for 5 minutes at 20 °C. After carefully discarding the supernatant, the pelleted DNA was air dried for 5 minutes. The precipitated DNA was

suspended in 200 µL molecular-grade, nuclease-free water and then cleaned with 100% ethanol and buffers (AL, AW1, AW2) from the Qiagen QIAamp DNA Stool Mini Kit per the

(13)

quantify the amount and assess purity of final DNA preparations. All samples were diluted to 40 ng/ml as the working concentration and were stored at -20 °C for subsequent amplification via PCR.

Sequencing of 16S rRNA Genes

Following DNA extraction, two consecutive polymerase chain reactions (PCR) were performed to amplify the V4 variable region of 16S rRNA gene. The first PCR contained 120 ng of template DNA, six forward primers, six reverse primers (10 µM each primer) and Buffer A, Enhancer, dNTPs, and Robust DNA polymerase from the KAPA2G Robust PCR Kit (Kapa Biosystems, Wilmington, MA). The mixture was amplified using the following conditions: 95°C/3min; [95°C/30s; 50°C/30s; 72°C/30s] x 10 cycles; 72°C/5 min; 4°C/hold. The second PCR further amplified the V4 variable region and added the Illumina MiSeq adapter primers and a single 12-nucleotide Golay error-correcting barcode for multiplexing43. The KAPA HiFi

HotStart ReadyMix reagent (Kapa Biosystems, Wilmington, MA) was combined with 5 µL PCR product from the first reaction, the forward MiSeq adapter primer, and reverse Golay barcode primer (10 µM) for the second PCR, using the following parameters: 95°C/3min; [95°C/30s; 50°C/30s; 72°C/30s] x 22 cycles; 72°C/5 min; 4°C/hold.

Products from each PCR were purified with the HighPrep PCR clean-up kit (MagBio, Lausanne, Switzerland) and a DynaMag-96 side magnet (Life Technologies, Carlsbad, CA) per the manufacturer’s instructions. Final purified products for each sample were quantified with a NanoDrop (Thermo Fisher Scientific, Madison, WI) and were pooled for sequencing at

(14)

Sequencing Facility at UNC School of Medicine for 250bp paired-end sequencing on an Illumina MiSeq benchtop sequencer (Illumina, San Diego, CA).

Analysis of 16S rRNA Gene Sequences

16S rRNA gene forward sequence read classification was performed on Quantitative Insights Into Microbial Ecology 2 pipeline (QIIME 2, version 2019.4)44. Single-end reads were

demultiplexed based on the associated barcode sequence with each sample. Primers and adapters were then removed. DADA2 was used to correct for Illumina-sequenced amplicon errors and produce sequence variants (SVs) at a 100% identity threshold45. Sequences were truncated at 215

base pair (bp) for cecal contents, 203 bp for colon contents, 213 bp for fecal samples at day 35, and 215 bp for all fecal samples.

To eliminate rare SVs, SVs with lower than 0.01% of total reads were filtered out for each run. Number of SVs decreased from 524 to 321 with a total number of reads of 1,781,133 after filtering cecal contents. For colon contents, number of SVs decreased from 493 to 318, and the number of reads were reduced to 1,826,413. Fecal samples at day 35 had 272 SVs filtered, resulting in 358 SVs and 1,787,123 reads. 1086 SVs were filtered out for the run with all fecal samples, leaving 379 SVs and 4,885,632 reads. Unfiltered cecal contents, colon contents, and fecal samples at day 35 were merged and filtered. The number of SVs decreased from 1647 to 685, with 5,291,216 reads after filtering.

(15)

a-diversity, an indication of microbial richness within each group, was characterized with

Shannon diversity and the absolute number of SVs. b-diversity, representing the differences between groups, was characterized with Bray-Curtis dissimilarity. The SILVA classifier (132, 99% OTUs) was used for taxonomy assignment46. A differential abundance analysis (Analysis of

Composition of Microbiomes; ANCOM) was used to identify specific genera of bacteria that drove differences in the microbial communities between groups across all three sties47.

Statistical Analysis

(16)

Results

Differential effect between GBDs and PDs on food consumption and body weight

In general, mice fed GBDs (Purina and Teklad) consumed significantly more food than mice fed PDs (Fig. 1b-c). Daily calorie intake was calculated (daily food intake × energy

density) to account for the different calorie density between different diets. Interestingly, calorie intake differed between the two GBD groups, Teklad and Purina (Fig. 1d). Mice fed the 75C/25I diet also had a higher daily calorie intake than mice fed the 100C and 25C/I/G/P diets (Fig. 1d). Consistent with the Chassaing et al. study, we found that a PD without soluble fiber (100C) was

Fig. 1. Food consumption and body weight changed by diet groups during the study. (a) Timeline of the study. Mice were fed Teklad 2020X for 7 days to acclimate, and were randomly assigned to one of the diets for 28 days.

(17)

associated with increased adiposity, when compared to Purina41 (Fig. 1e,f). Despite consuming

more food and calorie, mice fed a Purina diet gained the least weight over the 28-day experiment period (Fig. 1e). Consistent with the same study, mice fed 100C had an increased gonadal fat weight than mice fed Purina41 (Fig. 1f).

Addition of soluble fibers in PDs differentially impact GI anatomy

Increased soluble fiber content in PDs was associated with increased small intestinal length and cecum weight. The small intestine of mice fed 100C were shorter than those fed 75C/25I, 25C/75I, and 25C/I/G/P, while mice fed 25C/I/G/P had longer small intestines than those fed with all other diets, except for 25C/75I (Fig. 2a, e-j). Mice fed 100C had a lower cecum weight when compared to mice fed GBDs, 25C/75I, and 25C/I/G/P (Fig. 2b).

(18)
(19)

No difference in fecal microbiota composition observed between diet groups before diet

randomization

We first characterized the fecal microbiotas at day 0 (baseline) and day 7 (study onset) (Fig. 3a,d). As expected, no clustering based on diet groups was seen by Bray-Curtis Principal Coordinates Analysis (PCoA) plots for day 0 and day 7 (ANOSIM, p>0.1), suggesting that the diet group assignments were adequately randomized. Similarly, the two major measures of a -diversity—Shannon index and number of SVs—showed no significant differences in fecal samples at day 0 and day 7 (Fig. 3b,c,e,f).

Fig. 3. Characterization of the microbial communities in fecal sample at baseline (day 0) and study onset (day 7). (a)

PCoA at baseline. (b) Shannon index at baseline. (c) Number of SVs at baseline. (d) PCoA when study began. (e)

Shannon index when study began. (f) Number of SVs when study began. N= 5 mice/group.

(20)

in the cecum were significantly different between diet groups (ANOSIM, p<0.05 between all groups, except for 25C/75I vs. 25C/I/G/P, 25C/75I vs. 75C/25I) (Fig. 4a). Characterization of microbial communities in the colon and feces produced similar results (Fig. 4b,c). Interestingly, the microbial communities in mice fed 100C clustered distinctly (as Fig. 4a black circle

indicates), whereas the microbial communities in mice fed GBDs separated from those fed PDs (as Fig. 4a blue circle indicates) (ANOSIM, p<0.001 between GBDs and PDs). In addition, mice fed low soluble fiber PDs (100C and 75C/25I) produced significantly different microbial

communities than mice fed high soluble PDs (25C/25I and 25C/I/G/P) (ANOSIM, p<0.001). These results indicated that within each GI niche, microbiotas clustered separately by diet.

Fig. 4. PCoA showing the microbial composition by diet groups in cecum, colon and feces at the end of the study (day 35). (a) Microbial communities in cecal content by diet group. (b) Microbial communities in colon content by diet group. (c) Microbial communities in fecal sample by diet group. N= 5 mice/group. ANOSIM was used to test for significance.

Impact of diet on microbial richness (a-diversity) was consistent with b-diversity measures

Microbial richness in GBDs was significantly higher than that in PDs across all three GI sites (Fig. 5a-f). In addition, increased soluble fiber was associated with decreased a-diversity in both number of SVs and Shannon index. Mice fed high soluble fiber PDs (25C/25I and

(21)
(22)

Microbial communities did not cluster by GI niche

PCoA revealed clustering of microbial communities between cecum and colon, cecum and feces (ANOSIM, p<0.005), but not between colon and feces (Fig. 6a). Two measures of a -diversity—Shannon index and number of SVs—also indicated no significant differences in microbial richness between niches (Fig. 6b,c). Consistent with previous results, microbial communities clustered based on diets after merging samples from all three GI sites (Fig. 6d). These data illustrated that diet exerted a strong influence on shaping the microbial communities.

Fig. 6. Characterization of microbial communities in the cecum, colon, and feces at the end of study (day 35). (a)

PCoA plot (Bray-Curtis) of microbial communities from the cecum, colon, and feces. p<0.005 between cecum and colon, and between cecum and feces. No significant difference between feces and colon. (b) Shannon index across three GI sites. (c) Number of SVs across three GI sites. (d) microbial communities clustered according to diet. ANOSIM was used to test for significances for b-diversity. One-way ANOVA was used to test for significance for

a-diversity. N=29 for cecum, n=29 for colon, and n=30 for feces.

Lactococcus species drove differences in microbiotas between PDs and GBDs

(23)

across all three sties (Fig. 7a). The relative abundance of Lactococcus spp. was differentially higher in the cecum, colon, and feces of mice fed PDs than in mice fed GBDs (Fig. 7b).

[Eubacterium] ventriosum group was mostly found in the cecum and feces of mice fed the Teklad diet (Fig. 7c). Mice that consumed a Purina diet had a higher relative abundance of

Tyzzerella 3 spp. in the cecum and feces, when compared to all other groups (Fig. 7d). The relative abundance of Romboutsia spp. were also higher in the cecum and colon of mice fed 75C/25I and 100C than other groups (Fig. 7e). Akkermansia spp. were more abundant in the cecum of mice fed PDs, while [Eubacterium] xylanophilum group were more abundant in the cecum of mice fed GBDs (Fig. 7f-g).

Fig. 7. Specific bacterial genera identified with a differential abundance analysis (ANCOM). (a) Significant genera identified across the three GI sites. (b) Log10 relative abundance of Lactococcus spp. in cecum. (c) Log10 relative

abundance of [Eubacterium] ventriosum group in cecum. (d) Log10 relative abundance of Tyzzerella 3 spp. in

cecum. (e) Log10 relative abundance of Romboutsia spp. in cecum. (f) Log10 relative abundance of Akkermansia spp.

(24)

Discussion

Consistent with the Chassaing et al. study, our data found that the PD lacking soluble fibers (100C) resulted in significantly increased gonadal fat weight (a standard measure for body adiposity) and decreased colon weight, when compared to a specific GBD, Purina 500141.

However, as this previous study only used Purina 5001 as the GBD, we also evaluated the effect of a second GBD (Teklad 2020SX) on GI anatomy. Unexpectedly, unlike Purina 5001, mice that consumed the Teklad 2020SX diet did not exhibit significantly different gonadal fat or colon weights, when compared to the PDs. In addition, GBDs and PDs produced significantly different microbial communities in mice. Even within GBDs, the microbial communities clustered

distinctly between Teklad and Purina.

These results suggest that using a GBD as a control for rodent studies is not reliable and decreases the generalizability of results. Such variability highlights the importance of using PDs for rodent studies, yet, many studies investigating the effect of high-fat diets (HFD) on the gut microbiota continue to use GBDs, instead of low-fat PDs, as controls48,49,50. A recent study

demonstrated that the lack of soluble fiber in PDs, instead of the addition of fat, drove

alternations in the gut microbiota of mice48. Moreover, another study reported that the microbial

communities between mice fed GBDs and PDs were more distinct than those between mice fed a high-fat PD and a low-fat PD51. Consistent with these findings, our study also illustrated that

GBDs are unreliable controls, as mice fed GBDs produce distinct microbial communities when compared to those fed PDs.

(25)

addition of soluble fiber to PDs promoted these GI anatomical measurements, which are usually associated with healthier GI functions41. Unexpectedly, the addition of soluble fibers in PDs led

to decreased a-diversity—a measurement of microbial richness—of the gut microbiota.

Decreased a-diversity has typically been associated with a low-fiber high-sugar diet and linked to metabolic syndromes and IBD in human and mouse studies52. Additionally, a study showed

that piglets fed a diet with only soluble fiber had a significant increase in a-diversity, when compared to piglets fed a baseline diet, a diet with only insoluble fiber, or a diet with a mixture of soluble and insoluble fiber53. Yet, it was also recently reported that soluble fiber interventions

in human studies did not result in a difference in a-diversity, when compared to the control group with no additional soluble fiber27. It was therefore unexpected that the addition of soluble

fibers to rodent diets was beneficial to the GI tract (based on gross anatomy) yet accompanied with a decreased a-diversity. One hypothesis to explain this finding is that more bacterial groups are needed to break down the insoluble fiber to extract energy, thus increasing the number of different microbes in the gut. Future studies are needed to validate this hypothesis and to uncover a potential mechanism.

Though the validity of using fecal samples—as opposed to cecal or colon contents—to represent the gut microbiota has been questioned, our study showed similar clustering by diet groups across three GI sites on PCoA plots54. While we did find separate clustering between

fecal and cecal microbiotas, the microbiotas clustered more distinctly by diet groups irrespective of the GI niche, suggesting that diet plays a more significant role in driving the microbial

(26)

studies also reported that luminal microbiotas were not distinct between sites, but the mucosa-associated microbiota was distinct from luminal ones and exhibited greater associations with certain diseases, such as Crohn’s disease56. The mucosa-associated microbiotas were not

characterized in our study, but represents an important future direction.

Lactococcus spp. had a higher relative abundance in mice fed PDs than in mice fed GBD. It has been reported that the presence of Lactococcus spp. in rodent gut microbiota resulted from contamination in PDs51,57,58. Specifically, Lactococcus spp. are involved in industrial production

of casein, which is the main protein source in PDs but not in GBDs. As high throughput

sequencing of the 16S rRNA gene is unable to distinguish between live and dead microbes, it is difficult to determine the source of Lactococcus spp. in our study. In an independent study, investigators concluded that the Lactococcus spp. detected in their fecal samples originated from dead bacteria DNA leftover from casein production as they were unable to culture viable

bacteria, yet detected Lactococcus spp. in the diet57. Since we did not perform these experiments

in our study, we cannot conclude that the Lactococcus spp. found in PDs were leftover DNA from casein production, but it is a potential explanation for this increased abundance in the PDs compared to GBDs.

Limited research has been carried out to investigate the role of [Eubacterium]ventriosum

group and Tyzzerella 3 spp. in the gut microbiota.More studies are needed to help explain why the [Eubacterium]ventriosum group was more abundant in mice fed Teklad and why Tyzzerella 3 spp. was more abundant in mice fed Purina, when compared to other diet groups in our study.

Romboutsia spp.have incredible metabolic capabilities, including the ability to ferment carbohydrates, glycerol, and amino acids59. This interesting feature might help explain the high

(27)

with the hypothesis that increased types of bacteria are needed to utilize low soluble fiber in PDs, which results in a higher a-diversity.

Akkermansia spp. are mucin-degrading bacteria that reside in the mucus layer in the intestine. Mucin constitutes carbon and energy sources that can be released and used by the gut microbiota when broken down by mucin-degrading bacteria60. Increased relative abundance of Akkermansia spp. is associated with reduced mucosa inflammation and an increased metabolic profile61. As Akkermansia spp. had a higher relative abundance in PDs than in GBDs, our study

suggests that the PDs may promote an anti-inflammatory environment in the gut.

[Eubacterium] xylanophilum was found to be able to break down xylan, which is a hemicellulose62. A recent study also found that the relative abundance of [Eubacterium] xylanophilum increased when cultured inoculants supplemented with wheat barn63.

Unsurprisingly, [Eubacterium] xylanophilum was more abundant in GBDs, in which xylan was likely present as an agricultural byproduct, than in PDs, where xylan was not present.

In conclusion, mice fed PDs with high soluble fiber content (≥ 75% soluble fiber) best recapitulated GI health compared to mice fed GBDs, but we did not find any PD that

recapitulated the gut microbiota of mice fed GBDs. Furthermore, GBDs are unreliable choices as controls in diet-based studies because of their variable impact on host anatomy and gut

(28)

Acknowledgments

(29)

References

1. Scarpellini E, Ianiro G, Attili F, Bassanelli C, De Santis A, Gasbarrini A. The human gut microbiota and virome: Potential therapeutic implications. Digestive and Liver Disease. 2015;47(12):1007–1012. doi:10.1016/j.dld.2015.07.008

2. Sender R, Fuchs S, Milo R. Revised estimates for the number of human and bacteria cells in the body. PLOS Biol. 2016;157(28). doi:10.1371/journal.pbio.1002533

3. Almeida A, Mitchell AL, Boland M, Forster SC, Gloor GB, Tarkowska A, Lawley TD, Finn RD. A new genomic blueprint of the human gut microbiota. Nature. 2019;568(7753):499–504. doi:10.1038/s41586-019-0965-1

4. Rajilić-Stojanović M, de Vos WM. The first 1000 cultured species of the human

gastrointestinal microbiota. FEMS Microbiol Rev. 2014;38(5):996–1047. doi:10.1111/1574-6976.12075

5. O’Sullivan Ó, Coakley M, Lakshminarayanan B, Claesson MJ, Stanton C, O’Toole PW, Ross RP. Correlation of rRNA gene amplicon pyrosequencing and bacterial culture for microbial compositional analysis of faecal samples from elderly Irish subjects. J Appl Microbiol. 2011;111(2):467–473. doi:10.1111/j.1365-2672.2011.05067.x

6. Prescott SL. History of medicine : Origin of the term microbiome and why it matters. Human Microbiome J. 2017;4:24–25. doi:10.1016/j.humic.2017.05.004

7. Janda JM, Abbott SL. 16S rRNA gene sequencing for bacterial identification in the diagnostic laboratory: Pluses, perils, and pitfalls. J Clin Microbiol. 2007;45(9):2761–2764.

doi:10.1128/JCM.01228-07

(30)

doi:10.1128/CMR.17.4.840

9. Thursby E, Juge N. Introduction to the human gut microbiota. Biochem J. 2017;0:1823–1836. doi:10.1042/BCJ20160510

10. Carabotti M, Scirocco A, Maselli MA, Severi C. The gut-brain axis: interactions between enteric microbiota, central and enteric nervous systems. Biol Med. 2015;1(4):75–81.

doi:10.1038/ajgsup.2012.3

11. Ussar S, Griffin NW, Bezy O, Fujisaka S, Vienberg S, Softic S, Deng L, Bry L, Gordon JI, Kahn CR. Interactions between gut Microbiota, host genetics and diet modulate the

predisposition to obesity and metabolic syndrome. Cell Metab. 2015;22(3):516–530. doi:10.1016/j.cmet.2015.07.007

12. Nishino K, Nishida A, Inoue R, Kawada Y, Ohno M. Analysis of endoscopic brush samples identified mucosa-associated dysbiosis in inflammatory bowel disease. J Gastroenterol.

2018;53(1):95–106. doi:10.1007/s00535-017-1384-4

13. Trøseid M. Gut microbiota and acute coronary syndromes : ready for use in the emergency room ? Eur Heart J. 2017;(C):825–827. doi:10.1093/eurheartj/ehx005

14. Hasan N, Yang H. Factors affecting the composition of the gut microbiota, and its modulation. PeerJ. 2019:1–31. doi:10.7717/peerj.7502

15. Derikx LAAP, Dieleman LA, Hoentjen F. Probiotics and prebiotics in ulcerative colitis. Best Pract and Res Clin Gastroenterol. 2016;30(1):55–71. doi:10.1016/j.bpg.2016.02.005

16. Vindigni SM, Surawicz CM. Fecal microbiota transplantation. Gastroenterol Clin North Am. 2017;46(1):171–185. doi:10.1016/j.gtc.2016.09.012

(31)

Gastroenterol Hepatol. 2019;3(1):4–9. doi:10.1002/jgh3.12093

18. Fang H, Fu L, Wang J. Protocol for fecal microbiota transplantation in inflammatory bowel disease. Biomed Res Int. 2018;2018. doi:10.1155/2018/8941340

19. Costello SP, Hughes PA, Waters O, Bryant R V., Vincent AD, Blatchford P, Katsikeros R, Makanyanga J, Campaniello MA, Mavrangelos C, et al. Effect of fecal microbiota

transplantation on 8-week remission in patients with ulcerative colitis. J Am Med Assoc. 2019;321(2):156–164. doi:10.1001/jama.2018.20046

20. Peyrin-Biroulet L, Lémann M. Review article: Remission rates achievable by current therapies for inflammatory bowel disease. Aliment Pharmacol Ther. 2011;33(8):870–879. doi:10.1111/j.1365-2036.2011.04599.x

21. Org E, Mehrabian M, Parks BW, Shipkova P, Liu X, Drake TA, Lusis AJ. Sex differences and hormonal effects on gut microbiota composition in mice. Gut Microbes. 2016;7(4):313–322. doi:10.1080/19490976.2016.1203502

22. David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, Ling A V, Devlin AS, Varma Y, Fischbach MA, et al. Diet rapidly alters the human gut microbiota. Nature. 2014;505(7484):559–563. doi:10.1038/nature12820.Diet

23. Kim SJ, Kim S, Kim A, Kang S, Park M, Sung M. Dietary fat intake and age modulate the composition of the gut microbiota and colonic inflammation in C57BL / 6J mice. BMC Microbiol. 2019:1–11. doi:10.1186/s12866-019-1557-9

(32)

25. Patnode ML, Beller ZW, Han ND, Cheng J, Peters SL, Terrapon N, Henrissat B, Le Gall S, Saulnier L, Hayashi DK, et al. Interspecies competition impacts targeted manipulation of human gut bacteria by fiber-derived glycans. Cell. 2019;179(1):59-73.e13.

doi:10.1016/j.cell.2019.08.011

26. Koh A, Vadder F De, Kovatcheva-Datchary P, Ba¨ckhed F. From dietary fiber to host physiology : short-chain fatty acids as key bacterial metabolites. Cell. 2016;165(6):1332–1345. doi:10.1016/j.cell.2016.05.041

27. So D, Whelan K, Rossi M, Morrison M, Holtmann G, Kelly JT, Shanahan ER. Dietary fiber intervention on gut microbiota composition in healthy adults : a systematic review and meta-analysis. Am J Clin Nutr. 2018:965–983. doi:10.1093/ajcn/nqy041

28. Makki K, Deehan EC, Walter J, Bäckhed F. The impact of dietary fiber on gut microbiota in host health and disease. Cell Host Microbe. 2018;23(6):705–715.

doi:10.1016/j.chom.2018.05.012

29. Müller M, Canfora EE, Blaak EE. Gastrointestinal transit time, glucose homeostasis and metabolic Health: modulation by dietary fibers. Nutrients. 2018;Mar; 10(3).

doi:10.3390/nu10030275

30. Desai MS, Seekatz AM, Koropatkin NM, Stappenbeck TS, Martens EC. A dietary fiber-deprived gut microbiota degrades the colonic mucus barrier and enhances pathogen

susceptibility. Cell. 2016:1339–1353. doi:10.1016/j.cell.2016.10.043

31. Schroeder BO, Birchenough GMH, Arike L, Sta M, Johansson ME V, Arike L, Johansson ME V. Bifidobacteria or fiber protects against diet-Induced microbiota-mediated colonic mucus deterioration. Cell Host Microbe. 2018:27–40. doi:10.1016/j.chom.2017.11.004

(33)

Macdougall K, Preston T, Tedford C, Finlayson GS, et al. Effects of targeted delivery of

propionate to the human colon on appetite regulation, body weight maintenance and adiposity in overweight adults. Gut. 2015:1744–1754. doi:10.1136/gutjnl-2014-307913

33. Frost G, Sleeth ML, Sahuri-arisoylu M, Lizarbe B, Cerdan S, Brody L, Anastasovska J, Ghourab S, Hankir M, Zhang S, et al. The short-chain fatty acid acetate reduces appetite via a central homeostatic mechanism. Nat Commun. 2014:1–11. doi:10.1038/ncomms4611

34. Kimura I, Inoue D, Maeda T, Hara T, Ichimura A, Miyauchi S. Short-chain fatty acids and ketones directly regulate sympathetic nervous system via G protein-coupled. Proc Natl Acad Sci U S A. 2011;41. doi:10.1073/pnas.1016088108

35. Ricci MR, Ulman E. Laboratory animal diets: a critical part of your In vivo research web. 36. Nielsen FH. 90th anniversary commentary: The AIN-93 purified diets for laboratory rodents. J Nutr. 2018:1667–1670.

37. Pellizzon M. Arsenic in grain-based laboratory animal diets and effects on the rodent toxicological phenotype.

38. Pellizzon MA, Ricci MR. Effects of rodent diet choice and fiber type on data interpretation of gut microbiome and metabolic disease research. Curr Protoc Toxicol. 2018;77(1):1–14. doi:10.1002/cptx.55

39. Reeves PG, Nielsen FH, George C. Fahey J. AIN-93 Purified Diets for Laboratory Rodents : Final Report of the American Institute of Nutrition Ad Hoc Writing Committee on the

(34)

Vijay-Kumar M, Gewirtz AT. Lack of soluble fiber drives diet-induced adiposity in mice. Am J Physiol Gastrointest Liver Physiol. 2015;309(7):G528–G541. doi:10.1152/ajpgi.00172.2015 42. Bulik-Sullivan EC, Roy S, Elliott RJ, Kassam Z, Lichtman SN, Carroll IM, Gulati AS. Intestinal Microbial and Metabolic Alterations Following Successful Fecal Microbiota Transplant for D-Lactic Acidosis. Journal of Pediatric Gastroenterology and Nutrition. 2018;67(4):483–487. doi:10.1097/mpg.0000000000002043

43. Werner JJ, Zhou D, Caporaso JG, Knight R, Angenent LT. Comparison of Illumina paired-end and single-direction sequencing for microbial 16S rRNA gene amplicon surveys. ISME J. 2011;6(7):1273–1276. doi:10.1038/ismej.2011.186

44. Bolyen E, Rideout JR, Dillon MR, Bokulich NA, Abnet CC, Al-Ghalith GA, Alexander H, Alm EJ, Arumugam M, Asnicar F, et al. Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2. Nat Biotechnol. 2019;37(August). doi:10.1038/s41587-019-0209-9

45. Callahan BJ, Mcmurdie PJ, Rosen MJ, Han AW, A AJ. DADA2: High resolution sample inference from Illumina amplicon data. Nat Methods. 2016;13(7):581–583.

doi:10.1038/nmeth.3869.DADA2

46. Quast C, Pruesse E, Yilmaz P, Gerken J, Schweer T, Glo FO, Yarza P. The SILVA

ribosomal RNA gene database project : improved data processing and web-based tools. Nucleic Acids Res. 2013;41(November 2012):590–596. doi:10.1093/nar/gks1219

47. Mandal S, Van Treuren W, White RA, Eggesbø M, Knight R, Peddada SD. Analysis of composition of microbiomes: a novel method for studying microbial composition. Microb Ecol Health Dis. 2015;26(0):1–7. doi:10.3402/mehd.v26.27663

(35)

of dietary challenge on the gut microbiome. Microbiome. 2020;8:15. doi:10.1186/s40168-020-0791-6

49. Pellizzon MA, Ricci MR. The common use of improper control diets in diet-induced

metabolic disease research confounds data interpretation : the fiber factor. Nutr Metabol. 2018:1– 6. doi:10.1186/s12986-018-0243-5

50. Antonioli L, D’Antongiovanni V, Pellegrini C, Fornai M, Benvenuti L, Carlo A, den

Wijngaard R, Caputi V, Cerantola S, Giron MC, et al. Colonic dysmotility associated with high

-fat diet-induced obesity: Role of enteric glia. The FASEB Journal. 2020: 5512–5524. doi:10.1096/fj.201901844r

51. Dalby MJ, Ross AW, Walker AW, Morgan PJ. Dietary uncoupling of gut microbiota and energy harvesting from obesity and glucose tolerance in mice. Cell Rep. 2017;21(6):1521–1533. doi:10.1016/j.celrep.2017.10.056

52. Hills RD, Pontefract BA, Mishcon HR, Black CA, Sutton SC, Theberge CR. Gut microbiome: Profound implications for diet and disease. Nutrients. 2019;11(7):1–40. doi:10.3390/nu11071613

53. Chen T, Chen D, Tian G, Zheng P, Mao X, Yu J, He J, Huang Z, Luo Y, Luo J, et al. Soluble fiber and insoluble fiber regulate colonic microbiota and barrier function in a piglet model. Biomed Res Int. 2019;2019. doi:10.1155/2019/7809171

54. Pang W, Vogensen FK, Nielsen DS, Hansen AK. Faecal and caecal microbiota profiles of mice do not cluster in the same way. Laboratory Animals. 2012;46(3):231–236.

doi:10.1258/la.2012.011128

(36)

ulcerative colitis and control volunteers. Gut. 2015;64(10):1553–1561. doi:10.1136/gutjnl-2014-307873

56. Gevers D, Kugathasan S, Denson LA, Knight R, Xavier RJ. The treatment-naïve microbiome in new-onset Crohn’s disease. Cell Host Microbe. 2015;15(3):382–392.

doi:10.1016/j.chom.2014.02.005

57. Bisanz JE, Upadhyay V, Turnbaugh JA, Ly K, Turnbaugh PJ. Meta-Analysis Reveals Reproducible Gut Microbiome Alterations in Response to a High-Fat Diet. Cell Host and Microbe. 2019;26(2):265-272.e4. doi:10.1016/j.chom.2019.06.013

58. Carmody RN, Gerber GK, Luevano JM, Gatti DM, Somes L, Svenson KL, Turnbaugh PJ. Diet dominates host genotype in shaping the murine gut microbiota. Cell Host Microbe. 2015;17(1):72–84. doi:10.1016/j.chom.2014.11.010

59. Gerritsen J, Hornung B, Ritari J, Paulin L, Rijkers GT, Schaap PJ, Vos WM de, Smidt H. A comparative and functional genomics analysis of the genus Romboutsia provides insight into adaptation to an intestinal lifestyle. bioRxiv. 2019:845511. doi:10.1101/845511

60. Derrien M, Vaughan EE, Plugge CM, de Vos WM. Akkermansia municiphila gen. nov., sp. nov., a human intestinal mucin-degrading bacterium. Int J Syst Evol Microbiol.

2004;54(5):1469–1476. doi:10.1099/ijs.0.02873-0

61. Everard A, Belzer C, Geurts L, Ouwerkerk JP, Druart C, Bindels LB, Guiot Y, Derrien M, Muccioli GG, Delzenne NM, et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. PNAS. 2013;110(22):9066–9071.

doi:10.1073/pnas.1219451110

(37)

J Syst Bacteriol. 1985;35(3):323–326.

Figure

Table 1 Diets used in study.
Fig. 1. Food consumption and body weight changed by diet groups during the study. (a) Timeline of the study
Fig. 2. GI anatomical measurements at the end of study. (a) Small intestine length (#p&lt;0.05 vs
Fig. 3. Characterization of the microbial communities in fecal sample at baseline (day 0) and study onset (day 7)
+5

References

Related documents

Boundary objects for sharing and reporting of video research that are explicitly addressed here include: technological tools supporting video analysis; technological tools

Experiments were designed with different ecological conditions like prey density, volume of water, container shape, presence of vegetation, predator density and time of

Analyses were made of (i) student perceptions of their readiness/confidence to research and prepare a biology essay; (ii) the relative effect of the “getting

This should be the same number of months or years as secured by your financial guarantee to be provided to secure the transaction as collateral... LOAN TERMS /

For this reason, the researcher chose a descriptive study design to describe, observe and document the experiences of medical practitioners regarding the accessing of

Para dar un mayor entendimiento al comportamiento de la descentralización es necesario presentar la dinámica que ha surtido de este proceso, para lo cual nos

All the full duplex MAC protocols mentioned above are proposed on the same assumption that all the nodes, including AP and STAs, have full duplex capability. But this assumption

You can also define global login settings that apply to all login profiles, including enabling Lightweight Directory Access Protocol (LDAP) server authentication and customizing