throbber
RESEARCH ARTICLE SUMMARY
`
`Gut Microbiota from Twins Discordant
`for Obesity Modulate Metabolism
`in Mice
`
`Vanessa K. Ridaura, Jeremiah J. Faith, Federico E. Rey, Jiye Cheng, Alexis E. Duncan, Andrew
`L. Kau, Nicholas W. Griffi n, Vincent Lombard, Bernard Henrissat, James R. Bain, Michael J.
`Muehlbauer, Olga Ilkayeva, Clay F. Semenkovich, Katsuhiko Funai, David K. Hayashi, Barbara
`J. Lyle, Margaret C. Martini, Luke K. Ursell, Jose C. Clemente, William Van Treuren, William A.
`Walters, Rob Knight, Christopher B. Newgard, Andrew C. Heath, Jeffrey I. Gordon*
`
`Introduction: Establishing whether specifi c structural and functional confi gurations of a human gut
`microbiota are causally related to a given physiologic or disease phenotype is challenging. Twins dis-
`cordant for obesity provide an opportunity to examine interrelations between obesity and its associ-
`ated metabolic disorders, diet, and the gut microbiota. Transplanting the intact uncultured or cultured
`human fecal microbiota from each member of a discordant twin pair into separate groups of recipient
`germ-free mice permits the donors’ communities to be replicated, differences between their properties
`to be identifi ed, the impact of these differences on body composition and metabolic phenotypes to be
`discerned, and the effects of diet-by-microbiota interactions to be analyzed. In addition, cohousing
`coprophagic mice harboring transplanted microbiota from discordant pairs provides an opportunity to
`determine which bacterial taxa invade the gut communities of cage mates, how invasion correlates with
`host phenotypes, and how invasion and microbial niche are affected by human diets.
`
`Methods: Separate groups of germ-free mice were colonized with uncultured fecal microbiota from
`each member of four twin pairs discordant for obesity, or with culture collections from an obese (Ob) or
`lean (Ln) co-twin. Animals were fed a mouse chow low in fat and rich in plant polysaccharides (LF-HPP)
`or one of two diets refl ecting the upper or lower (Hi or Lo) tertiles of consumption of saturated fats (SF)
`and fruits and vegetables (FV) based on the U.S. National Health and Nutrition Examination Survey
`(NHANES). Ln or Ob mice were cohoused 5 days after colonization. Body composition changes were
`defi ned by quantitative magnetic resonance. Microbiota or microbiome structure, gene expression, and
`metabolism were assayed by 16S ribosomal RNA profi ling, whole-community shotgun sequencing, RNA-
`sequencing, and mass spectrometry. Host gene expression and metabolism were also characterized.
`
`Results and Discussion: The intact uncultured and culturable bacterial component of Ob co-twins’
`fecal microbiota conveyed signifi cantly greater increases in body mass and adiposity than those of
`Ln communities. Differences in body composition were correlated with differences in fermentation of
`short-chain fatty acids (increased in Ln), metabolism of branched-chain amino acids (increased in Ob),
`and microbial transformation of bile acid species (increased in Ln and correlated with down-regulation
`of host farnesoid X receptor signaling). Cohousing Ln and Ob mice prevented development of increased
`adiposity and body mass in Ob cage mates and transformed their microbiota’s metabolic profi le to a
`leanlike state. Transformation correlated with invasion of members of Bacteroidales from Ln into Ob
`microbiota. Invasion and phenotypic rescue were diet-dependent and occurred with the diet represent-
`ing the lower tertile of U.S. consumption of saturated fats and upper tertile of fruits and vegetables but
`not with the diet representing the upper tertile of saturated fats and lower tertile of fruit and vegetable
`consumption. These results reveal that transmissible and modifi able interactions between diet and
`microbiota infl uence host biology.
`
`READ THE FULL ARTICLE ONLINE
`http://dx.doi.org/10.1126/science.1241214
`
`Cite this article as V. K. Ridaura et al.,
`Science 341, 1241214 (2013).
`DOI: 10.1126/science.1241214
`
`FIGURES IN THE FULL ARTICLE
`
`Fig. 1. Reliable replication of human donor
`microbiota in gnotobiotic mice.
`
`Fig. 2. Cohousing Obch and Lnch mice
`transforms the adiposity phenotype of cage
`mates harboring the obese co-twin’s culture
`collection to a leanlike state.
`
`Fig. 3. Effect of cohousing on metabolic
`profi les in mice consuming the LF-HPP diet.
`
`Fig. 4. Effects of NHANES-based LoSF-HiFV
`and HiSF-LoFV diets on bacterial invasion,
`body mass, and metabolic phenotypes.
`
`Fig. 5. Invasion analysis of species-level taxa
`in Obch or Lnch mice fed the NHANES-based
`LoSF-HiFV diet.
`
`Fig. 6. Acylcarnitine profi le in the skeletal
`muscle of mice colonized with the Ob or Ln
`culture collections from dizygotic twin pair 1
`and fed the LoSF-HiFV diet.
`
`SUPPLEMENTARY MATERIALS
`
`Materials and Methods
`Supplementary Text
`Figs. S1 to S17
`Tables S1 to S17
`References and Notes
`
`RELATED ITEMS IN SCIENCE
`
`A. W. Walker, J. Parkhill, Fighting obesity
`with bacteria. Science 341, 1069–1070 (2013).
`DOI: 10.1126/science.1243787
`
`Cohousing Ln and Ob
`mice prevents adiposity
`phenotype in Ob cage
`mates (Obch). (A) The adi-
`posity change after 10
`days of cohousing. *P <
`0.05 versus Ob controls
`(Student’s t test). (B) Bac-
`teroidales from Lnch micro-
`biota invade the Obch
`microbiota. Columns show
`individual mice.
`
`The list of author affi liations is available in the full article online.
`*Corresponding author. E-mail: jgordon@wustl.edu
`
`www.sciencemag.org SCIENCE VOL 341 6 SEPTEMBER 2013
`Published by AAAS
`
`1079
`
`Genome Ex. 1045
`Page 1 of 12
`
`

`

`RESEARCH ARTICLE
`
`Gut Microbiota from Twins
`Discordant for Obesity Modulate
`Metabolism in Mice
`
`Vanessa K. Ridaura,1 Jeremiah J. Faith,1 Federico E. Rey,1 Jiye Cheng,1 Alexis E. Duncan,2,3
`Andrew L. Kau,1 Nicholas W. Griffin,1 Vincent Lombard,4 Bernard Henrissat,4,5
`James R. Bain,6,7,8 Michael J. Muehlbauer,6 Olga Ilkayeva,6 Clay F. Semenkovich,9
`Katsuhiko Funai,9 David K. Hayashi,10 Barbara J. Lyle,11 Margaret C. Martini,11
`Luke K. Ursell,12 Jose C. Clemente,12 William Van Treuren,12 William A. Walters,13
`Rob Knight,12,14,15 Christopher B. Newgard,6,7,8 Andrew C. Heath,2 Jeffrey I. Gordon1*
`
`The role of specific gut microbes in shaping body composition remains unclear. We transplanted
`fecal microbiota from adult female twin pairs discordant for obesity into germ-free mice fed
`low-fat mouse chow, as well as diets representing different levels of saturated fat and fruit and
`vegetable consumption typical of the U.S. diet. Increased total body and fat mass, as well as
`obesity-associated metabolic phenotypes, were transmissible with uncultured fecal communities
`and with their corresponding fecal bacterial culture collections. Cohousing mice harboring an
`obese twin’s microbiota (Ob) with mice containing the lean co-twin’s microbiota (Ln) prevented the
`development of increased body mass and obesity-associated metabolic phenotypes in Ob cage
`mates. Rescue correlated with invasion of specific members of Bacteroidetes from the Ln
`microbiota into Ob microbiota and was diet-dependent. These findings reveal transmissible,
`rapid, and modifiable effects of diet-by-microbiota interactions.
`
`Microbial community configurations
`
`vary substantially between unrelated in-
`dividuals (1–9), which creates a chal-
`lenge in designing surveys of sufficient power to
`determine whether observed differences between
`disease-associated and healthy communities dif-
`fer significantly from normal interpersonal var-
`iation. This challenge is especially great if, for
`a given disease state, there are many associated
`states of the microbial species (microbiota) or mi-
`crobial gene repertoire (microbiome), each shared
`by relatively few individuals. Microbiota config-
`
`1Center for Genome Sciences and Systems Biology, Washing-
`ton University School of Medicine, St. Louis, MO 63108, USA.
`2Department of Psychiatry, Washington University School of
`Medicine, St. Louis, MO 63110, USA. 3George Warren Brown
`School of Social Work, Washington University, St. Louis, MO
`63130, USA. 4Architecture et Fonction des Macromolécules
`Biologiques, CNRS and Aix Marseille Université, CNRS UMR
`7257, 13288 Marseille, France. 5Department of Cellular and
`Molecular Medicine, Faculty of Health and Medical Sciences,
`University of Copenhagen, DK-2200, Copenhagen, Denmark.
`6Sarah W. Stedman Nutrition and Metabolism Center, Duke
`University Medical Center, Durham, NC 27710, USA. 7Depart-
`ment of Medicine, Duke University Medical Center, Durham,
`NC 27710, USA. 8Department of Pharmacology and Cancer
`Biology, Duke University Medical Center, Durham, NC 27710,
`USA. 9Department of Medicine, Washington University School
`of Medicine, St. Louis, MO 63110, USA. 10Mondelez Interna-
`tional, Glenview, IL60025, USA. 11Kraft Foods Group, Glenview,
`IL 60025, USA. 12Department of Chemistry and Biochemistry,
`University of Colorado, Boulder, CO 80309, USA. 13Molecular,
`Cellular and Developmental Biology, University of Colorado,
`Boulder, CO 80309, USA. 14Biofrontiers Institute, University of
`Colorado, Boulder, CO 80309, USA. 15Howard Hughes Medical
`Institute, University of Colorado, Boulder, CO 80309, USA.
`*To whom correspondence should be sent. E-mail: jgordon@
`wustl.edu
`
`urations are influenced by early environmental
`exposures and are generally more similar among
`family members (2, 7, 10, 11).
`There have been conflicting reports about
`the relation between interpersonal differences
`in the structure of the gut microbiota and host
`body mass index (BMI). Taxonomic profiles
`for obese and lean individuals may have dis-
`tinct patterns between human populations, but
`technical issues related to how gut samples are
`processed and community members are identified
`by 16S ribosomal RNA (rRNA) gene sequencing
`may also play a role in observed differences.
`The relative contributions of the microbiota and
`dietary components to obesity and obesity-related
`metabolic phenotypes are unclear and likely multi-
`faceted (2, 12–17). Transplants of fecal microbiota
`from healthy donors to recipients with metabolic
`syndrome have provided evidence that the micro-
`biota can ameliorate insulin-resistance, although
`the underlying mechanisms remain unclear (18).
`Monozygotic (MZ) or dizygotic (DZ) twins
`discordant for obesity (19, 20) provide an attract-
`ive model for studying the interrelations between
`obesity, its associated dietary and lifestyle risk fac-
`tors, and the gut microbiota/microbiome. In the
`case of same-sex twins discordant for a disease
`phenotype, the healthy co-twin provides a valuable
`reference control to contrast with the co-twin’s disease-
`associated gut community. However, this comparison
`is fundamentally descriptive and cannot establish
`causality. Transplanting a fecal sample obtained
`from each twin in a discordant pair into separate
`groups of recipient germ-free mice provides an
`opportunity to (i) identify structural and functional
`
`differences between their gut communities; (ii)
`generate and test hypotheses about the impact of
`these differences on host biology, including body
`composition and metabolism; and (iii) determine
`the effects of diet-by-microbiota interactions
`through manipulation of the diets fed to these
`“humanized” animals and/or the representation of
`microbial taxa in their gut communities.
`
`Reproducibility of Microbiota Transplants
`from Discordant Twins
`We surveyed data collected from 21- to 32-year-old
`female twin pairs (n = 1539) enrolled in the Mis-
`souri Adolescent Female Twin Study [MOAFTS;
`(21, 22); for further details, see ref. (23)]. We
`recruited four twin pairs, discordant for obesity
`(obese twin BMI > 30 kg/m2) with a sustained
`multiyear BMI difference of ≥5.5 kg/m2 (n =
`1 MZ and 3 DZ pairs) (Fig. 1A). Fecal samples
`were collected from each twin, frozen immedi-
`ately after they were produced, and stored at
`–80°C. Each fecal sample was introduced, via a
`single oral gavage, into a group of 8- to 9-week-
`old adult male germ-free C57BL/6J mice (one
`gnotobiotic isolator per microbiota sample; each
`recipient mouse was individually caged within
`the isolator; n = 3 to 4 mice per donor microbiota
`sample per experiment; n = 1 to 5 independent
`experiments per microbiota). All recipient mice
`were fed, ad libitum, a commercial, sterilized mouse
`chow that was low in fat (4% by weight) and high
`in plant polysaccharides (LF-HPP) (23). Fecal
`pellets were obtained from each mouse 1, 3, 7,
`10, and 15 days post colonization (dpc) and, for
`more prolonged experiments, on days 17, 22, 24,
`29, and 35.
`Unweighted UniFrac-based comparisons of
`bacterial 16S rRNA data sets generated from the
`input human donor microbiota, from fecal sam-
`ples collected from gnotobiotic mice and from
`different locations along the length of the mouse
`gut at the time they were killed (table S1A), plus
`comparisons of the representation of genes with
`assignable enzyme commission numbers (ECs)
`in human fecal and mouse cecal microbiomes
`(defined by shotgun sequencing), disclosed that
`transplant recipients efficiently and reproducibly
`captured the taxonomic features of their human
`donor’s microbiota and the functions encoded by
`the donor’s microbiome (see Fig. 1B; fig. S1, A
`to E; fig. S2; table S1B; and table S2, A to D)
`(23). The 16S rRNA data sets allowed us to iden-
`tify bacterial taxa that differentiate gnotobiotic
`mice harboring gut communities transplanted
`from all lean versus all obese co-twins [analysis of
`variance (ANOVA) using Benjamini-Hochberg
`correction for multiple hypotheses] [table S3; see
`(23) for details].
`
`Reproducible Transmission of Donor Body
`Composition Phenotypes
`Quantitative magnetic resonance (QMR) anal-
`ysis was used to assess the body composition
`of transplant recipients 1 day, 15 days, and, in
`
`www.sciencemag.org SCIENCE VOL 341
`
`6 SEPTEMBER 2013
`
`1241214-1
`
`Genome Ex. 1045
`Page 2 of 12
`
`

`

`RESEARCH ARTICLE
`
`the case of longer experiments, 8, 22, 29, and
`35 days after transplantation. The increased adi-
`posity phenotype of each obese twin in a discor-
`dant twin pair was transmissible: The change in
`adipose mass of mice that received an obese co-
`twin’s fecal microbiota was significantly greater
`than the change in animals receiving her lean
`twin’s gut community within a given experiment
`and was reproducible across experiments (P ≤
`0.001, one-tailed unpaired Student’s t test; n = 103
`mice phenotyped) (Fig. 1, C to E). Epididymal
`fat pad weights (normalized to total body weight)
`were also significantly higher in mice colonized
`with gut communities from obese twins (P ≤ 0.05,
`one-tailed unpaired Student’s t test). These dif-
`ferences in adiposity were not associated with
`statistically significant differences in daily chow
`consumption (measured on days 1, 8, and 15 after
`gavage and weekly thereafter for longer exper-
`iments) or with appreciably greater inflamma-
`tory responses in recipients of obese compared
`with lean co-twin fecal microbiota as judged by
`fluorescence-activated cell sorting (FACS) anal-
`ysis of the CD4+ and CD8+ T cell compartments
`in spleen, mesenteric lymph nodes, small intes-
`tine, or colon [see (23) for details].
`
`Functional Differences Between Transplanted
`Microbial Communities
`Fecal samples collected from gnotobiotic mice
`were used to prepare RNA for microbial RNA se-
`quencing (RNA-Seq) of the transplanted mi-
`crobial communities’ meta-transcriptomes (table
`S1C). Transcripts were mapped to a database of
`sequenced human gut bacterial genomes and as-
`signed to Kyoto Encyclopedia of Genes and Ge-
`nomes (KEGG) Enzyme Commission numbers
`(EC numbers) [see ref. (23)]. Significant differences
`and distinguishing characteristics were defined
`using ShotgunFunctionalizeR, which is based on a
`Poisson model (24) (see table S4 and table S5 for
`ECs and KEGG level 2 pathways, respectively).
`Transcripts encoding 305 KEGG ECs were dif-
`ferentially expressed between mice harboring mi-
`crobiomes transplanted from lean or obese donors
`[ShotgunFunctionalizeR, Akaike's information
`−30].
`criterion (AIC) < 5000; P ≤ 10
`Mice harboring the transplanted microbiomes
`from the obese twins exhibited higher expression
`of microbial genes involved in detoxification and
`stress responses; in biosynthesis of cobalamin;
`metabolism of essential amino acids (phenyl-
`alanine, lysine, valine, leucine, and isoleucine)
`
`and nonessential amino acids (arginine, cysteine,
`and tyrosine); and in the pentose phosphate path-
`way (fig. S3, A and B; table S4, B to G; and
`table S5). Follow-up targeted tandem mass spec-
`trometry (MS/MS)–based analysis of amino acids
`in sera obtained at the time mice were killed dem-
`onstrated significant increases in branched-chain
`amino acids (BCAA: Val and Leu/Ile), as well as
`other amino acids (Met, Ser, and Gly), plus trends
`to increase (Phe, Tyr, and Ala), in recipients of
`microbiota from obese compared with lean co-
`twins in discordant twin pairs DZ1 and MZ4
`(tables S1D and S6A). These specific amino
`acids, as well as the magnitude of their differ-
`ences, are remarkably similar to elevations in
`BCAA and related amino acids reported in obese
`and insulin-resistant versus lean and insulin-
`sensitive humans (25). This finding suggested
`that the gut microbiota from obese subjects
`could influence metabolites that characterize
`the obese state.
`In contrast, the transplanted microbiomes from
`lean co-twins exhibited higher expression of genes
`involved in (i) digestion of plant-derived poly-
`saccharides [e.g., a-glucuronidase (EC 3.2.1.139),
`a-L-arabinofuranosidase (EC 3.2.1.55)]; (ii) fermen-
`
`***
`
`15
`
`10
`
`05
`
`-5
`
`D
`
`% Change in fat mass
`
`Fat mass
`Lean body mass
`
`All lean
`
`co-twin donors
`
`All obese
`co-twin donors
`
`All lean
`co-twin donors
`
`All obese
`co-twin donors
`
`Twin pair 1
`Obese co-twin
`Lean co-twin
`
`0246
`
`-2
`
`C
`
`% Change in body composition
`
`E
`
`30
`
`20
`
`10
`
`0
`
`% Change in fat mass
`
`Twin Pair
`
`1 (DZ)
`
`2 (DZ)
`
`3 (DZ)
`
`4 (MZ)
`
`BMI (kg/m2)
`
`23
`
`32
`
`25.5
`
`31
`
`19.5 30.7
`
`24
`
`33
`
`Twin pair 1
`
`Lean donor
`
`Obese donor
`
`A
`
`B
`
`PC1 (23%)
`
`dpc
`
`1
`
`3
`
`7
`
`10
`
`15
`
`8dpc
`
`15dpc
`
`22dpc
`
`29dpc
`
`35dpc
`
`Input samples
`Experiment 1
`Experiment 2
`Experiment 3
`Fig. 1. Reliable replication of human donor microbiota in gnotobiotic
`mice. (A) Features of the four discordant twin pairs. (B) Assembly of bacterial
`communities in mice that had received intact and uncultured fecal microbiota
`transplants from the obese and lean co-twins in DZ pair 1. Principal coordinates
`analysis plot of principal coordinate 1 (PC1) based on an unweighted UniFrac
`distance matrix and 97%ID OTUs present in sampled fecal communities. Circles
`correspond to a single fecal sample obtained at a given time point from a given
`mouse and are colored according to the experiment (n = 3 independent ex-
`periments). Note that assembly is reproducible within members of a group of
`mice that have received a given microbiota, as well as between experiments. (C)
`Body composition, defined by QMR, was performed 1 day and 15 dpc of each
`mouse in each recipient group. Mean values (T SEM) are plotted for the percent
`increase in fat mass and lean body mass at 15 dpc for all recipient mice of each
`of the four obese co-twins’ or lean co-twins’ fecal microbiota, normalized to the
`
`-10
`initial body mass of each recipient mouse. A two-way ANOVA indicated that
`there was a significant donor effect (P ≤ 0.05), driven by a significant difference
`in adiposity and total body mass between mice colonized with a lean or obese
`co-twin donor’s fecal microbiota (adjusted P ≤ 0.05; Siˇdák’s multiple comparison
`test). (D) Mean values (T SEM) are plotted for the percent change in fat mass at
`15 dpc for all recipient mice of each of the four obese co-twins’ or lean co-twins’
`fecal microbiota. Data are normalized to initial fat mass (n = 3 to 12 animals
`per donor microbiota; 51 to 52 mice per BMI bin; total of 103 mice). ***P ≤
`0.001, as judged by a one-tailed unpaired Student’s t test. (E) More prolonged
`time course study for recipients of fecal microbiota from co-twins in discordant
`DZ pair 1 (mean values T SEM plotted; n = 4 mice per donor microbiota).
`The difference between the gain in adiposity calculated relative to initial fat
`mass (1 dpc) between the two recipient groups of mice is statistically signif-
`icant (P ≤ 0.001, two-way ANOVA).
`
`1241214-2
`
`6 SEPTEMBER 2013 VOL 341 SCIENCE www.sciencemag.org
`
`Genome Ex. 1045
`Page 3 of 12
`
`

`

`RESEARCH ARTICLE
`
`biota of Obch mice were reconfigured so that they
`came to resemble the microbiota of Lnch cage
`mates. In contrast, the microbiota of the Lnch cage
`mates remained stable (fig. S9, A to C). We per-
`formed a follow-up analysis to identify species-
`level taxa that had infiltrated into and/or had been
`displaced from the guts of mice harboring the Ln
`andOb culture collections. We did so by character-
`izingthedirectionandsuccessofinvasion.Microbial
`SourceTracker estimates, for every species-level
`taxon or 97%ID OTU, the Bayesian probability
`(P) of its being derived from each of a set of source
`communities (30). The fecal microbiota of Ln or
`Ob controls sampled 5 days after colonization
`were used as source communities to determine the
`direction of invasion. The fecal communities
`belonging to each Lnch and Obch mouse were then
`traced to these sources. We defined the direction of
`invasion for these bacterial taxa, by calculating the
`log odds ratio of the probability of a Ln origin
`(PLn) or an Ob origin (POb) for each species-level
`taxon or 97%ID OTU, i, as follows:
`
`
`log2 PLni=PObi
`
`
`
`
`
`.
`
`
`
`Aij
`
`tation to butyrate [acetyl-CoA C-acetyltransferase
`(EC 2.3.1.9), 3-hydroxybutyryl-CoA dehydrogenase
`(EC 1.1.1.157), 3-hydroxybutyryl-CoA dehydratase
`(EC 4.2.1.55), butyryl-CoA dehydrogenase (EC
`1.3.8.1)] (fig. S3, C and D); and (iii) fermentation to
`propionate [succinate dehydrogenase (EC 1.3.99.1),
`phosphoenolpyruvate carboxykinase (EC 4.1.1.32),
`methylmalonyl-CoA mutase (EC 5.4.99.2)] (table
`S4A). Follow-up gas chromatography–mass spec-
`trometry (GC-MS) of cecal contents confirmed that
`levels of butyrate and propionate were significantly
`increased and that levels of several mono- and
`disaccharides significantly decreased in animals
`colonized with lean compared with obese co-twin
`gut communities (P ≤ 0.05, unpaired Student’s t
`test) (fig. S4, A and B, and table S6B). Procrustes
`analysis, using a Hellinger distance matrix (26), re-
`vealed significant correlations between taxonomic
`structure [97% identity (97%ID) threshold for de-
`fining distinct operational taxonomic units (OTU)
`in fecal samples], transcriptional profiles (enzyme
`representation in fecal mRNA populations), and
`metabolic profiles (GC-MS of cecal samples), with
`separation of groups based on donor microbiota
`and BMI (Mantel test, P ≤ 0.001) (fig. S5).
`These results suggest that, in this diet context,
`transplanted microbiota from lean co-twins had a
`greater capacity to breakdown and ferment poly-
`saccharides than the microbiota of their obese co-
`twins. Previous reports have shown that increased
`microbial fermentation of nondigestible starches
`is associated with decreased body weight and de-
`creased adiposity in conventionally raised mice that
`harbor a mouse microbiota [e.g., refs. (27–29)].
`
`Phenotypes Produced by Bacterial
`Culture Collections
`We followed up these studies of transplanted, in-
`tact, and uncultured donor communities with a set of
`experiments involving culture collections produced
`from the fecal microbiota of one of the discordant
`twin pairs. Our goal was to determine whether cul-
`tured bacterial members of the co-twins’ microbiota
`could transmit the discordant adiposity phenotypes
`and distinctive microbiota-associated metabolic pro-
`files when transplanted into gnotobiotic mouse re-
`cipients that received the LF-HPP chow diet.
`Collections of cultured anaerobic bacteria were
`generated from each co-twin in DZ pair 1 and
`subsequently introduced into separate groups of
`8-week-old germ-free male C57BL/6J mice (n =
`5 independent experiments; n = 4 to 6 recipient
`mice per culture collection per experiment). The
`culture collections stabilized in the guts of re-
`cipient mice within 3 days after their introduc-
`tion [see (23); fig. S6, A to E; and table S7 for
`documentation of the efficient and reproducible
`capture of cultured taxa and their encoded gene
`functions between groups of recipient mice].
`As in the case of uncultured communities, we
`observed a significantly greater increase in adi-
`posity in recipients of the obese twin’s culture col-
`lection compared with the lean co-twin’s culture
`collection (P ≤ 0.02, one-tailed unpaired Student’s t
`test) (Fig. 2, A and B). Nontargeted GC-MS showed
`
`that the metabolic profiles generated by the trans-
`planted culture collections clustered with the
`profiles produced by the corresponding intact
`uncultured communities (fig. S6E). In addition,
`the fecal biomass of recipients of the culture col-
`lection from the lean twin was significantly greater
`than the fecal biomass of mice receiving the cul-
`ture collection from her obese sibling; these differ-
`ences were manifest within 7 days (P ≤ 0.0001,
`two-way ANOVA) (fig. S7A).
`
`Cohousing Ob and Ln Animals Prevents
`an Increased Adiposity Phenotype
`Because mice are coprophagic, the potential for
`transfer of gut microbiota through the fecal-oral
`route is high. Therefore, we used cohousing to
`determine whether exposure of a mouse harboring
`a culture collection from the lean twin could
`prevent development of the increased adiposity
`phenotype and microbiome-associated metabolic
`profile of a cage mate colonized with the culture
`collection from her obese co-twin or vice versa.
`Five days after gavage, when each of the inoculated
`microbial consortia had stabilized in the guts of
`recipient animals, a mouse with the lean co-twin’s
`culture collection was cohoused with a mouse with
`the obese co-twin’s culture collection (abbreviated
`Lnch and Obch, respectively). Control groups con-
`sisted of cages of dually housed recipients of the
`lean twin culture collection and dually housed
`recipients of the obese co-twin’s culture collection
`(n = 3 to 5 cages per housing configuration per
`experiment; n = 4 independent experiments; each
`housing configuration in each experiment was
`placed in a separate gnotobiotic isolator) (Fig.
`2A). All mice were 8-week-old C57BL/6J males.
`All were fed the same LF-HPP chow adlibitum that
`was used for transplants involving the correspond-
`ingunculturedcommunities.Beddingwaschanged
`before initiation of cohousing. Fecal samples were
`collectedfrom allrecipients 1,2,3, 5, 6,7,8, 10,and
`15 days after gavage. Body composition was mea-
`sured by QMR 1 and 5 days after gavage, and after
`10 days of cohousing.
`Obch mice exhibited a significantly lower in-
`crease in adiposity compared with control Ob ani-
`mals that had never been exposed to mice harboring
`the lean co-twin’s culture collection (P ≤ 0.05, one-
`tailed unpaired Student’s t test). Moreover, the adi-
`posity of these Obch animals was not significantly
`different from Ln controls (P > 0.05, one-tailed un-
`paired Student’s t test) (Fig. 2B). In addition, expo-
`sure to Obch animals did not produce a significant
`effect on the adiposity of Lnch mice: Their adiposity
`phenotypes and fecal biomass were indistinguish-
`able from dually housed Ln controls (Fig. 2B; and
`fig. S7, B and C). Cohousing caused the cecal meta-
`bolic profile of Obch mice to assume features of
`Lnch and control Ln animals, including higher lev-
`els (compared with dually housed Ob controls) of
`propionate and butyrate and lower levels of cecal
`mono- and disaccharides, as well as BCAA and
`aromatic amino acids (Fig. 2, C and D, and fig. S8).
`Principal coordinates analysis of unweighted
`UniFrac distances revealed that the fecal micro-
`
`A positive log odds ratio indicated that a
`species or 97%ID OTU was derived from a Ln
`source; a negative log odds ratio indicated an Ob
`source. An invasion score was calculated to
`quantify the success of invasion of each species
`or 97%ID OTU, i, into each cohousing group, j, as
`follows:
`Invasion Scoreij ¼ log2
`
`Bij
`where Aij is the average relative abundance of
`taxoniinallfecalsamplescollectedfromgroupjafter
`cohousing, and Bij is its relative abundance in all
`samples taken from that group before cohousing.
`The observed mean of the distribution of
`invasion scores for Obch animals was significantly
`higher than that for dually housed Ob-Ob controls
`(P ≤ 0.0005, Welch’s two-sample t test) (fig.
`S10A). This was not the case for Lnch animals
`when compared with dually housed Ln-Ln
`controls (P > 0.05), which suggested that there
`was significant invasion of components of the
`Lnch microbiota into the microbiota of Obch cage
`mates, but not vice versa. To quantify invasion
`further, we used the mean and standard deviation
`of the null distribution of invasion scores (defined
`as the scores from recipients of the Ln or Ob
`microbiota that had never been cohoused with
`each other) to calculate a z value and a Benjamini-
`Hochberg adjusted P value for the invasion score
`of each species in Lnch and Obch mice. We con-
`servatively defined a taxon as a successful invader
`if it (i) had a Benjamini-Hochberg adjusted P ≤
`0.05, (ii) was represented in ≥75% of Obch or Lnch
`mice when sampled 7 and 10 days after initiation
`of cohousing, and (iii) had a relative abundance of
`≤0.05% before cohousing and ≥0.5% in the fecal
`microbiota at the time mice were killed. We de-
`fined a taxon that was displaced from an animal’s
`microbiota upon cohousing as having a relative
`
`www.sciencemag.org SCIENCE VOL 341
`
`6 SEPTEMBER 2013
`
`1241214-3
`
`Genome Ex. 1045
`Page 4 of 12
`
`

`

`RESEARCH ARTICLE
`
`abundance ≥1% in Lnch or Obch mice before they
`were cohoused and a relative abundance <0.5%
`after cohousing.
`
`The direction and success of invasion are
`shown in Fig. 2E and table S8A. The most suc-
`cessful Lnch invaders of the Obch microbiota were
`
`members of the Bacteroidetes (rank order of
`their invasion scores: Bacteroides cellulosilyticus,
`B. uniformis, B. vulgatus, B. thetaiotaomicron,
`
`***
`
`***
`
`***
`
`***
`
`***
`
`***
`
`Ob
`Obch
`
`Ln
`Lnch
`
`GFch
`
`D
`
`15
`
`Log2(spectral abundance)
`
`10
`
`5
`
`***
`
`**
`
`*
`
`*
`
`** *** ** ***
`
`C
`
`10
`
`8
`
`6
`
`4
`
`2
`
`µmol/mg wet cecal contents
`
`**
`
`*
`
`*
`
`*
`
`15
`
`10
`
`5
`
`0
`
`-5
`
`B
`
`% Change in fat mass
`
`Controls
`
`A
`
`Isolator
`
`Cage
`
`Ln
`
`Ob
`
`Ob
`
`Ob
`
`Ln
`Ln
`
`Ob
`Ob
`
`Obch
`Lnch
`
`5 dpc
`
`5 dpc
`
`5 dpc
`
`5 dpc
`
`Ln
`
`Ln
`
`Obch
`2xGFch
`Lnch
`
`-10
`
`Ob-Ob Ln-Ln Obch
`
`Lnch GF
`
`ch
`
`0
`
`Propionate
`
`Butyrate
`
`Cellobiose
`
`0
`
`Maltose
`or similar
`disaccharide
`
`GFch
`co-housing
`8
`10
`
`RA
`
`12 15
`
`Lnch
`
`Obch
`
`prior to co-housing
`1
`2
`3
`5
`
`co-housing
`8
`10
`
`7
`
`12
`
`15
`
`RA
`
`prior to co-housing
`1
`2
`3
`
`5
`
`6
`
`co-housing
`10
`7
`8
`
`12 15
`
`RA
`
`6
`
`6
`
`7
`
`E
`
`dpc
`
`*
`
` Clostridium clostridioforme
` Butyricimonas virosa
` Clostridium asparagiforme
` Turicibacter sanguinis
` Ruminococcus sp. 5_1_39BFAA
` Clostridium glycolicum
` Alistipes putredinis
` Clostridium tertium
` Bacteroides finegoldii
` Clostridium sp. MLG480
` Clostridium symbiosum
` Firmicutes Other
` Enterococcus faecium
` Roseburia faecis
` Ruminococcus sp. CCUG 37327 A
` Clostridium disporicum
` Odoribacter splanchnicus
` Alistipes shahii
` Bifidobacterium longum
` Roseburia spp.
` Anaerotruncus colihominis
` Bacteroides thetaiotaomicron
` Bacteroides cellulosilyticus
` Bacteroides vulgatus
` Bacteroides uniformis
` Bacteroides caccae
` Ruminococcus sp. DJF VR70k1
` Parabacteroides merdae
` Akkermansia muciniphila
` Clostridium sp. NML 04A032
` Clostridium ramosum
` Eubacterium limosum
` Oscillibacter spp.
` Enterobacteriaceae Other
` Alistipes indistinctus
` Holdemania filiformis
` Betaproteobacteria Other
` Clostridium spp.
` Bacteroides spp.
` Ruminococcus torques
` Blautia producta
` Bacteroides massiliensis
` Clostridium hathewayi
` Blautia hansenii
` Alistipes finegoldii
`
`**
`
`**
`*
`
`*
`
` -20
`
` 0
`
` 20
`
`Ob source
`
`Ln source
`
`Direction of invasion
`
`Not detected
`Species present in
` both Ob and Ln
`
`0
`
`5
`
`10 15
`
`Relative abundance
`(RA) (%) b (before),
`a (after) co-housing
`
`−6 −2
`
`2
`
`6
`
`Fold change (fc)
`(Log(RAach/RAbch))
`
`Fig. 2. Cohousing Obch and Lnch mice transforms the adiposity pheno-
`type of cage mates harboring the obese co-twin’s culture collection to a
`lean-like state. (A) Design of cohousing experiment: 8-week-old, male, germ-
`free C57BL/6J mice received culture collections from the lean (Ln) twin or the
`obese (Ob) co-twin in DZ twin pair 1. Five days after colonization, mice were
`cohoused in one of three configurations: Control groups consisted of dually
`housed Ob-Ob or Ln-Ln cage mates; the experimental group consisted of dually
`housed Obch-Lnch cage mates (data shown from five cages per experiment; two
`independent experiments) or Obch-Lnch- GFch-GFch cage mates (n = 3 cages per
`experiment). All mice were fed a LF-HPP diet. (B) Effects of cohousing on fat
`mass. Changes from the first day after cohousing to 10 days after cohousing
`were defined using whole-b

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket