throbber
Submit a Manuscript: http://www.wjgnet.com/esps/
`Help Desk: http://www.wjgnet.com/esps/helpdesk.aspx
`DOI: 10.3748/wjg.v20.i47.17727
`
`World J Gastroenterol 2014 December 21; 20(47): 17727-17736
` ISSN 1007-9327 (print) ISSN 2219-2840 (online)
`© 2014 Baishideng Publishing Group Inc. All rights reserved.
`
`WJG 20th Anniversary Special Issues (17): Intestinal microbiota
`Impact of the gut microbiota on rodent models of human
`disease
`
`TOPIC HIGHLIGHT
`
`Axel Kornerup Hansen, Camilla Hartmann Friis Hansen, Lukasz Krych, Dennis Sandris Nielsen
`
`Axel Kornerup Hansen, Camilla Hartmann Friis Hansen,
`Section of Experimental Animal Models, Department of Veteri-
`nary Disease Biology, University of Copenhagen, 1871 Freder-
`iksberg, Denmark
`Lukasz Krych, Dennis Sandris Nielsen, Department of Food
`Science, Faculty of Sciences, University of Copenhagen, 1958
`Frederiksberg, Denmark
`Author contributions: All authors contributed to this paper
`equally.
`Correspondence to: Axel Kornerup Hansen, Professor, DVSc,
`DVM, DipECLAM, Section of Experimental Animal Models, De-
`partment of Veterinary Disease Biology, University of Copenha-
`gen, 57 Thorvaldsensvej, 1871 Frederiksberg,
`Denmark. akh@sund.ku.dk
`Telephone: +45-353-32726 Fax: +45-353-32755
`Received: February 27, 2014 Revised: September 30, 2014
`Accepted: November 18, 2014
`Published online: December 21, 2014
`
`Abstract
`Traditionally bacteria have been considered as either
`pathogens, commensals or symbionts. The mammal
`gut harbors 1014 organisms dispersed on approximately
`1000 different species. Today, diagnostics, in contrast
`to previous cultivation techniques, allow the identifica-
`tion of close to 100% of bacterial species. This has
`revealed that a range of animal models within differ-
`ent research areas, such as diabetes, obesity, cancer,
`allergy, behavior and colitis, are affected by their gut
`microbiota. Correlation studies may for some diseases
`show correlation between gut microbiota composition
`and disease parameters higher than 70%. Some dis-
`ease phenotypes may be transferred when recolonizing
`germ free mice. The mechanistic aspects are not clear,
`but some examples on how gut bacteria stimulate re-
`ceptors, metabolism, and immune responses are dis-
`cussed. A more deeper understanding of the impact of
`microbiota has its origin in the overall composition of
`the microbiota and in some newly recognized species,
`
`such as Akkermansia muciniphila, Segmented filamen-
`tous bacteria and Faecalibacterium prausnitzii, which
`seem to have an impact on more or less severe disease
`in specific models. Thus, the impact of the microbiota
`on animal models is of a magnitude that cannot be
`ignored in future research. Therefore, either models
`with specific microbiota must be developed, or the mi-
`crobiota must be characterized in individual studies and
`incorporated into data evaluation.
`
`© 2014 Baishideng Publishing Group Inc. All rights reserved.
`
`Key words: Animal models; Gut microbiota; Diabetes;
`Obesity; Cancer; Allergy; Behavior; Colitis
`
`Core tip: Full characterization of the gut microbiota of
`animal models has revealed that animal models within
`different research areas, such as diabetes, obesity,
`cancer, allergy, behavior and colitis, are highly affected
`by their gut microbiota. The mechanistic aspects are
`not clear; however, the impact of the microbiota on
`animal models is of a magnitude that cannot be ig-
`nored in future research. Therefore, either models with
`specific microbiota must be developed, or the micro-
`biota must be characterized in individual studies and
`incorporated into data evaluation.
`
`Hansen AK, Friis Hansen CH, Krych L, Nielsen DS. Impact of
`the gut microbiota on rodent models of human disease. World J
`Gastroenterol 2014; 20(47): 17727-17736 Available from: URL:
`http://www.wjgnet.com/1007-9327/full/v20/i47/17727.htm DOI:
`http://dx.doi.org/10.3748/wjg.v20.i47.17727
`
`INTRODUCTION
`Host-microbiota relationship
`The gut is an ideal incubation chamber for bacteria
`adapted to the mammal body temperature and the anaer-
`
`WJG|www.wjgnet.com
`
`17727
`
`December 21, 2014|Volume 20|Issue 47|
`
`

`

`Hansen AK et al. Gut microbiota and rodent models
`
`Actinobacteria
`Mollicutes
`Spirochaetes
`Proteobacteria
`Verrucomicrobia
`Bacteroidetes
`Firmicutes
`
`100%
`90%
`80%
`70%
`60%
`50%
`40%
`30%
`20%
`10%
`0%
`
`Ileum Caecum Faeces
`
`Figure 1 The approximate composition of the gut microbiota in the ileum,
`cecum and feces of mice[3,7,8,11,16].
`
`obic environment. Thousands of years of co-existence
`has led to such adaptation, and the mammal gut harbors
`1014 organisms dispersed over approximately 1000 dif-
`ferent species, dependent on how the cut-offs are set for
`similarity. Within the traditional approach to laboratory
`animal bacteriology, bacteria have been considered as
`either pathogens, commensals or symbionts; however,
`there seems to be a need for a broader understanding
`of this. When first inside the gut, the bacteria will be fed
`and will be allowed to propagate, while the host organ-
`ism will benefit from otherwise unavailable products of
`microbial digestion. Generally, pathogenicity is not in
`the interest of the microorganism, because it induces a
`strong and eradicating immune response from the host,
`and even in the case of microbial victory in this battle,
`the end result may be the death of the host and the need
`for the microbe to relocate to a new habitat. The host
`immune system, on the other hand, needs to protect the
`host from invasion without being so aggressive that it
`loses the microbe and thereby all its benefits.
`
`Complexity of microbial impact on the host
`A more advanced understanding of the impact of the
`microbiota takes into consideration both the overall
`composition and the balance between the members of
`the microbiota, as well as some newly recognized species,
`which, by themselves, seem to have an effect on the spe-
`cific models. Some of these have a symbiotic effect, while
`others push disease development in a more detrimental
`direction. However, same species may act in favor of the
`development of one disease, while being more protective
`against another disease, and the mechanistic potential of
`the species may differ between different parts of the gut.
`For most of these bacteria, it is their abundance, rather
`than their qualitative presence or absence, which are re-
`
`sponsible for their effect on the host[1-4]. The microbiota
`is normally not very diverse in the upper part of the gut,
`e.g. in the ileum, where there is a huge accumulation of
`lymphatic tissue available for stimulation[3,5-10]. It gradually
`becomes more diverse as the gut contents pass through
`the large intestine and become feces (Figure 1)[3,5-11]. In
`both man and mouse, a microbiota with a low diversity is
`indicative of an increased risk of developing inflammato-
`ry disease[12,13]. Furthermore, in animals, a microbiota that
`is roughly similar in the upper part of the gut, may differ
`substantially in the lower part of the gut and vice versa[3,14].
`Finally, there might be essential differences between the
`effects of the various species at different ages of the
`animals, which may explain why some species favor the
`development of one disease, while protecting against an-
`other.
`
`Modern microbiological identification techniques
`Over recent decades, new methods based upon molecu-
`lar biology diagnostics have been developed. Such meth-
`ods, which include quantitative real-time polymerase
`chain reaction (qPCR) assays[15], pyrosequencing[16] and
`metagenomic sequencing, have permitted identification
`of close to 100% of the gut’s operational taxonomic
`units (OTU), which include both cultivable and non-
`cultivable bacterial species, and in principle, viral, eukary-
`otes and Archea[17], although they are seldom specifically
`tested for at present. In contrast, previous cultivation
`techniques only allowed cultivation and identification of
`10%-20% of the bacterial species present in the gut[18].
`These molecular biology-based tools have enabled de-
`tailed correlation studies. Such studies have revealed that
`a range of animal models within a range of different
`research areas are affected by their gut microbiota[19].
`
`GENERAL MECHANISMS UNDERLYING
`THE GUT MICROBIOTA EFFECT
`As described below, the impact of the microbiota on an-
`imal models is well documented, while the mechanisms
`underlying this are less clear. Some hypotheses, though,
`make more sense than others. As techniques for the
`full characterization of the microbiota have been devel-
`oped over the last decade, we are only now beginning to
`achieve an understanding of how the microbiota actually
`exerts its effect on the host; however, some examples
`can be given.
`
`Window of opportunity
`In early life, there is a window for the induction of oral
`tolerance in the gut[20]. This seems essential to avoid in-
`flammatory disease later in life[21]. Molecular structures
`in bacteria known as microbial-associated molecular pat-
`terns (MAMP) stimulate pattern-recognition receptors
`(PRR) in the host, thereby inducing innate responses[22].
`Among the most important PRRs are the toll-like recep-
`tors (TLR), which are present in different types on a
`range of different cell types[22-29] (Figure 2). An impor-
`
`WJG|www.wjgnet.com
`
`17728
`
`December 21, 2014|Volume 20|Issue 47|
`
`

`

`Hansen AK et al. Gut microbiota and rodent models
`
`Mum
`
`AMP-activated
`protein kinase
`
`H
`
`H
`
`H
`
`C
`
`H
`
`H
`
`C
`
`H
`
`H
`
`C
`
`H
`
`H
`
`C
`
`H
`
`C
`
`H
`H
`
`C
`
`H
`
`C
`
`H
`
`O
`
`O
`
`C
`
`OH
`
`OH
`
`Short chain fatty acid
`
`GPR
`
`Lipids
`
`Lipids
`
`Lipids
`
`CH2OH
`O
`
`OH
`OH
`
`OH
`OH
`
`CH2OH
`O
`
`OH
`OH
`
`OH
`OH
`
`CH2OH
`O
`
`OH
`OH
`
`OH
`OH
`
`Acetyl-CoA carboxylase
`fatty acid synthase
`
`Lipopolysaccharide
`
`TLR4
`
`Flagellin
`
`TLR5
`
`Peptidoglycan
`Lipoproteins
`
`Lipoteichoic acid
`Polysaccharide A
`
` Zymosan
`
`TLR10
`
`TLR6
`
`TLR2
`
`TLR1
`
`Figure 2 Examples of some theories on potential pathways for the impact of gut microbiota on animal models of human disease. Bacterial colonization may
`double the density of capillaries in the small intestinal epithelium, thereby promoting intestinal monosaccharide absorption[28]. Undigested food components may be
`fermented into SCFAs and subsequently act as signals for GPRs of importance for the development of obesity[26,29]. Bacteria may express several key enzymes rele-
`vant for hepatic lipogenesis[27,50], and hepatic and muscular fatty acid oxidation[31]. Molecular structures in the cell walls of bacteria may act as MAMPs, which stimulate
`TLRs on the host cells to induce innate immune responses. The complex of TLR1, TLR2, TLR6 and TLR10 is expressed on a range of cell types such as enterocytes,
`macrophages, dendritic cells, natural killer cells, mast cells, T cells, B cells, neutrophilic cells and Schwann cells, and may be stimulated by various MAMPs, e.g. pep-
`tidoglycan, from Gram positive bacteria cell types[21,23-25,31-33]. TLR4, expressed by e.g. macrophages, dendritic cells, mast cells, natural killer cells and enterocytes, is
`stimulated by lipopolysaccharides from Gram negative bacteria[30], while flagellin from various bacteria may stimulate TLR5 expressed by e.g. mucosal dendritic cells
`and macrophages[33]. Mucin-degrading Akkermansia muciniphila may reduce the mucus layer to increase TLR-stimulation[79]. SCFAs: Short chain fatty acids; GPR:
`G-protein receptor; MAMP: Microbial-associated molecular pattern; TLR: Toll-like receptor.
`
`tant example of a MAMP is lipopolysaccharides (LPS),
`which are important parts of the cell wall of Gram
`negative bacteria[30], such as Proteobacteria[31], from
`which it stimulates TLR4. Another important example is
`peptidoglycan, found in the cell walls of Gram positive
`bacteria, which stimulates TLR2[32] and flagellin deriving
`from flagellated bacteria, leading to stimulation of TLR5[33].
`Therefore, as different types of MAMPs stimulate differ-
`ent TLR’s dispersed on a variety of different cell types[23],
`and as MAMPs are also dispersed and shared between
`members of the microbiota[22], there is a vast range of
`innate host responses to bacteria.
`
`Adult life stimulation
`The age of the animal also makes a difference. For
`example, stimulation of TLR1, TLR2 and TLR4 in early
`life leads to higher production of interleukin (IL)-6 than
`stimulation later in life[34]. Germ free animals have more
`T helper cells type 2 (TH2) and less TH1 cells[35], as the
`
`stimulation of the gut lamina propria dendritic cells, e.g.
`by polysaccharide A (PSA) from Bacteroides fragilis, induces
`IL-12 secretion, which favors TH1 at the cost of TH2[36].
`Host-bacterial interactions, probably mediated through
`glucagon-like peptide 2 (GLP-2), seem to control the gut
`barrier function[37]. Metabolic endotoxaemia is responsible
`for the phenomenon whereby excess intake of dietary
`fat increases plasma LPS levels[38,39], which in mice, is
`a sufficient molecular mechanism to trigger metabolic
`diseases, such as obesity and diabetes[40].
`
`EXAMPLES OF SOME ANIMAL
`MODELS UNDER IMPACT OF THE GUT
`MICROBIOTA
`Impact of germ free status
`The clearest documentation of a general microbial impact
`on rodent models is observed when comparing a conven-
`
`WJG|www.wjgnet.com
`
`17729
`
`December 21, 2014|Volume 20|Issue 47|
`
`

`

`Hansen AK et al. Gut microbiota and rodent models
`
`Table 1 Examples of rodent models in which germ free status
`has a documented impact
`
`Model
`Models with increased disease incidence or severity
` β-lactoglobulin induced mouse[51]
` NOD mouse[42]
` MyD88 KO NOD mouse[42]
` Restrained mouse[43]
`Models with decreased disease incidence or severity
` Ovalbumin-specific TCR TG mouse[44]
` Swiss-Webster mouse[45]
` Collagen induced rat[52]
` HLA-B27 TG rat[53]
` IL-2 KO mouse[54,55]
` IL-10 KO mouse[56]
` TCRα KO mouse[57]
` Dextran sulfate sodium induced mouse[46]
` SAMP1/Yit mouse[47]
` Adoptive T-cell transfer in the mouse[48]
` Carrageenan, LPS, or formalin induced mouse[49]
`
` C57BL/6 mouse[65]
` C57BL/6 mouse[65]
`
`Disease
`
`Allergy
`Type 1 diabetes
`Type 1 diabetes
`Stress
`
`Allergy
`Anxiety
`Arthritis
`IBD
`IBD
`IBD
`IBD
`IBD
`IBD
`IBD
`Inflammatory
`pain
`Obesity
`Type 2 diabetes
`
`NOD: Non-obese diabetic; MyD88: Myeloid differentiation primary
`response gene 88; KO: Knockout; TCR: T cell receptor; TG: Transgenic;
`HLA-B27: Human leucocyte antigen subtype B27; IL-2: Interleukin 2;
`SAMP1/Yit: Senescence accelerated mice prone line 1 Yakult; LPS: Lipo-
`polysaccharide. IBD: Inflammatory bowel disease.
`
`tional model with a microbiota with a germ free version.
`In several studies, this has revealed essential differences
`in disease expression (Table 1)[22,41-57]. Although germ free
`mice eat more, they are leaner, and they have less body
`fat compared with conventional mice because they are
`less efficient in extracting energy from their diet[50]. Germ
`free mice have increased expression of obesity-related
`peptides, such as glucagon-like peptide 1 (GLP-1) in the
`brain[58], which is relevant, because central GLP-1 reduces
`food intake in rats[59]. Germ free mice also behave differ-
`ently from microbiota-harboring mice and this behavior
`may be normalized by colonization[43]. However, for this
`phenotype there also seems to be an important time win-
`dow in early life[60]. Germ free mice with a mutation caus-
`ing a defect in the skin barrier suffer from a more severe
`B-lymphoproliferative disorder, because they express sig-
`nificantly higher levels of the proinflammatory cytokine
`thymic stromal lymphopoietin[61]. Inflammatory bowel dis-
`ease (IBD) occurs either because of a TH1/TH17 response
`(Crohn’s disease) or a TH2 response (ulcerative colitis) to
`gut commensals[62]. Therefore, IBD under germ free con-
`ditions does not develop at all in, e.g. Human Leucocyte
`Antigen subtypes B27 (HLA-B27) transgenic rats[53] and
`IL-10 knockout mice[56]. For the IL-10 knockout mice[63]
`it does not occur even under barrier protected conditions
`(Table 1). IL-2 knockout mice may, under germ free con-
`ditions, show mild focal intestinal inflammation[64] (Table
`1).
`
`Impact of fluctuations in the gut microbiota composition
`Within animal models of the metabolic syndrome, there
`
`seems to be an association between the gut microbiota
`and at least some of the metabolic parameters. For ex-
`ample, in leptin-deficient obese mice, there is a strong
`correlation between glycated hemoglobin levels and the
`composition of the gut microbiota[1]. Further, these mice
`have significantly more Firmicutes and fewer Bacteriode-
`tes members compared with their wild-type and heterozy-
`gous litter mates[10]. Their obese phenotype may be trans-
`ferred with the microbiota by recolonizing germ free lean
`wild-type mice[65]. In C57 Black substrain 6 (C57BL/6)
`mice on both high and low calorie diet, continuous oral
`ampicillin improves glucose tolerance[66,67]. However, this
`effect is mainly caused by an early life impact on glucose
`tolerance, and the effect ceases immediately after ter-
`mination of treatment; thereafter, the glucose tolerance
`may even decrease[68,69]. Several studies describe cross-
`talk between the brain and the gut through both the vagal
`system and the hypothalamus-pituitary-adrenal (HPA)
`axis[70]. Stressing animal models changes their micro-
`biota[71], and the composition of the gut microbiota has
`an impact on responses in rodent stress tests[72,73]. Innate
`immune system cytokines, such as IL-1, IL-6 and tumor
`necrosis factor α (TNFα), which may originate from a
`gut microbiota provocation, induce “sickness behav-
`ior”, changing the priorities of the organism to enhance
`recovery and survival[74]. However, metabolites formed
`by microbial decomposition in the gut may also have a
`direct impact on the brain[75]. In mouse models of atopic
`dermatitis, more than 70% of the variation observed in
`the local tissue cytokine response may be shared with the
`variation in gut microbiota[76]. Changes in the structure
`of the microbial community seem to reduce the number,
`as well as the size, of tumors in azoxymethane/dextran
`sodium sulfate (AOM/DSS) colon cancer-induced mice,
`and tumor induction may be achieved by colonizing germ
`free mice with microbiota from induced mice[77].
`
`EXAMPLES OF THE IMPACTS OF
`SPECIFIC BACTERIAL SPECIES
`Verrucomicrobioa
`Akkermansia muciniphila (A. muciniphila) is a Gram negative
`bacterium, which in mice is the only species belonging to
`the phylum Verrucomicrobia[78]. It interacts via its mucin
`degrading capabilities with enteroendocrine cells to mod-
`ulate gut barrier function, and it is capable of producing
`certain short chain fatty acids (SCFA’s) with a direct ac-
`tion on the receptor G-protein receptor 43 (GPR43)[79].
`Abundance of A. muciniphila is reduced in mice with
`obesity and type 2 diabetes[80], and it gradually disappears
`as aging leptin deficient obese mice develop insulin resis-
`tance[1]. In non-obese diabetic (NOD) mice it becomes
`more abundant when mice are fed a gluten-free diet,
`which decreases the incidence of type 1 diabetes[81]. Early
`life treatment with vancomycin in NOD mice allows A.
`muciniphila to become a dominant gut microbiota mem-
`ber, which reduces the incidence of type 1 diabetes[3],
`but enhances susceptibility to allergic asthma[82], which
`
`WJG|www.wjgnet.com
`
`17730
`
`December 21, 2014|Volume 20|Issue 47|
`
`

`

`is in accordance with other studies showing allergy and
`diabetes to counteract one another in NOD mice[83,84].
`Induction of IBD in mice with dextran sodium sulfate
`(DSS) reduces the number of extracellular vesicles de-
`rived from A. muciniphila, and feeding DSS induced mice
`such vesicles reduces the severity of IBD[85], which fits
`well with observations in humans[4]. However, it not only
`reduces the severity of diseases: its presence is correlated
`with higher severity when infecting mice with Salmonella
`typhimurium[86], and AOM/DSS colon cancer-induced
`mice have an increased abundance of A. muciniphila[77],
`which may be explained by its ability to downregulate the
`natural killer cell receptor, NKG2D, which is part of the
`anti-carcinogenic defense[87].
`
`Firmicutes
`Segmented filamentous bacteria (SFB’s) are clostridia-re-
`lated Gram-positive bacteria[88]. The term has been applied
`for decades to describe intestinal bacteria of a uniform
`morphology[89]. However, today the term refers to one
`single species, also known as Candidatus Savagella[90]. SFBs
`induce secretion of the pro-inflammatory cytokine IL-17
`from TH17 cells[91], which in the adult mouse is correlated
`with a low number of regulatory T cells[92]. The presence
`of SFB’s differs between mice from different vendors[92],
`and SFB positive NOD mice have a significantly lower
`incidence of type 1 diabetes compared with SFB nega-
`tive ones[93]. In the adoptive transfer severe combined
`immune deficiency (SCID) mouse model of IBD, SFBs
`are essential for the induction of severe inflammation[48].
`Furthermore, SFBs and the induced TH17 are important
`in the defense against intestinal pathogens. For example,
`mice infected with Citrobacter rodentium, a potent murine
`colon pathogen, exhibit severe symptoms if they lack
`SFBs[91].
`IBD in IL-10 knockout mice is enhanced by Enterococ-
`cus fecalis[94,95], which is probably linked to its production of
`gelatinase[96].
`Faecalibacterium prausnitzii (F. prausnitzii) is a clostridia-
`related bacterium[97] linked to a protective effect against
`human Crohn’s disease[98]. Oral feeding of F. prausnitzii
`reduced the severity of 2,4,6-trinitrobenzenesulfonic
`acid (TNBS)-induced colitis in mice, and some studies
`indicated that this may also be the case in both multidrug
`resistance gene deficient (mdr1a knockout)[99] and in the
`DSS-induced mouse models of colitis[100].
`High abundances of Lactobacillus spp. and bifidobac-
`teria are correlated strongly with low levels of inflamma-
`tion in mice[101] and leptin in rats[102], which also fits well
`with these bacteria acting protectively against IBD in
`IL-10 knockout mice[103], allergic sensitization in mice[104],
`and myocardial infarction in rats[102]. Lachnospiraceae seems
`quantitatively correlated to improved glucose tolerance
`in leptin-deficient obese mice[1].
`In stressed mice, there is correlation between their Fir-
`micutes levels and their responses in the stress tests[73].
`Ingestion of Lactobacillus rhamnosus in mice regulates their
`emotional behavior and central γ-aminobutyric acid (GABA)
`
`Hansen AK et al. Gut microbiota and rodent models
`
`receptor expression via the vagus nerve[72].
`
`Bacteroidetes
`A high abundance of the Gram negative family Prevotel-
`laceae, perhaps restricted to one unclassified genus, in
`the gut of leptin-deficient obese mice correlated with
`impaired glucose tolerance[1].By contrast, in AOM/DSS
`induced colon cancer mice, a high abundance of Pre-
`votellaceae correlated with a low tumor burden[77]. P. copri,
`which has been correlated with the development of ar-
`thritis in humans, seems to increase the severity of DSS
`induced colitis in mice[5]. Caspase-3 knockout mice exhibit
`a lower inflammatory response to DSS induction of
`colitis compared with wild-type mice; however, this pro-
`tective effect of the mutation is decreased by cohousing
`knockout mice with wild-type mice, which significantly
`increases the abundance of Prevotella spp. in the knockout
`mice[105].
`Bacteroides vulgatus seems to enhance IBD in HLA-B27
`transgenic rats[106] and IL-10 knockout mice[95], and in
`the Bio Breeding (BB) rat, a spontaneous type 1 diabetes
`model. The fecal microbiota differ and contain an in-
`creased number of Bacteroides spp. before onset of diabe-
`tes[107]. As in all other mammals, Bacteroides spp. form an
`important part of the Bacteroidetes fraction of the rodent
`gut[16]. These Gram negative bacteria are important for the
`processing of complex molecules to simpler ones in the
`gut[108]: complex glycans are their key source of energy[109].
`B. fragilis toxins cause symptoms of diarrhea and IBD
`in germ-free mice[110], and they induce colonic tumors
`strongly in multiple intestinal neoplasia (MIN) mice[111].
`On the other hand, B. fragilis PSA, which is important for
`the inflammatory gut response to pathogens[36], also pro-
`tects against Helicobacter hepaticus-induced colitis in mice;
`probably via the prevention of IL-17 secretion[112]. Feeding
`the maternal immune activation (MIA) mouse model with
`B. fragilis reduces symptoms of autism, which is probably
`linked to the normalization of the levels of a specific gut
`metabolite[113].
`The abundance of Alistipes spp., a bacterium of the
`Rikenellaceae family, seems to increase when mice are
`stressed by grid floor housing[73].
`
`Proteobacteria
`Escherichia coli (E. coli) enhances IBD in HLA-B27 over-
`expressing rats[106], although E. coli Nissle stabilizes the
`enteric barrier in mice[114]. When reducing type 1 diabetes
`by pre-weaning treatment of NOD mice with vancomy-
`cin, a vast increase in the abundance of Proteobacteria
`in the pups was observed[3].
`
`Actinobacteria
`Bifidobacterium spp. in rodents have a positive impact on
`the regulatory and innate immunity[101,115]. Perinatal sup-
`plementation of B. longum reduced TH1 and TH2 responses
`in allergen sensitized mice[104]. On the other hand, their
`numbers are also increased in gluten-fed NOD mice with
`a high incidence of type 1 diabetes compared with NOD
`
`WJG|www.wjgnet.com
`
`17731
`
`December 21, 2014|Volume 20|Issue 47|
`
`

`

`Hansen AK et al. Gut microbiota and rodent models
`
`mice on a gluten-free diet[81].
`
`DISCUSSION
`The information gained over the last decade on how
`the entire microbiota, as well as some of its individual
`members, affect animal models of very different types,
`has prompted the scientific community to incorporate
`this in future production and quality assurance of animal
`models. It is not possible to regard these matters from a
`“Specific pathogen-free” concept, as some of the spe-
`cies act in favor of the development of one disease,
`while against the development of another disease, e.g.
`SFB’s both protect against type 1 diabetes and induces a
`TH17 response in favor of the development of Crohn’s
`disease. Furthermore, the balance between the different
`fractions of the microbiota is also likely to make a dif-
`ference. Ultimately, it is often a quantitative rather than
`a qualitative presence that makes the difference. There-
`fore, it is likely that we will see more tailor-made rodent
`models, i.e. commercial breeders and research groups
`have sought to produce animals with a specific microbi-
`ota for the conditions under test. One obvious idea may
`be to breed such animals by selective breeding; however,
`this does not seem to increase the microbiota similarity,
`although the microbiota of offspring show a clear clus-
`tering with the mother’s microbiota[116,117]. It is probably
`rational to inoculate germ free mice with a tailor-made
`microbiota around weaning, as they are conventionalized
`in SPF conditions[118]. The window for induction of oral
`tolerance in animal models may also be turned around,
`such that a low bacterial stimulation in the open phase
`of this window may be essential to develop target diseas-
`es in the model. When stimulated later on, the nature of
`this stimulation is also essential, because commonly used
`disease models in rodents are driven by specific subsets
`of T cells[19]. Another alternative will be to characterize
`the microbiota composition for animals in sensitive stud-
`ies and incorporate this in the data evaluation by chemo-
`metric or multifactorial statistical means. The impact of
`the gut microbiota on animal models is of a magnitude
`that cannot be neglected in the future.
`
`2
`
`REFERENCES
`1
`Ellekilde M, Krych L, Hansen CH, Hufeldt MR, Dahl K,
`Hansen LH, Sørensen SJ, Vogensen FK, Nielsen DS, Hansen
`AK. Characterization of the gut microbiota in leptin deficient
`obese mice - Correlation to inflammatory and diabetic pa-
`rameters. Res Vet Sci 2014; 96: 241-250 [PMID: 24556473 DOI:
`10.1016/j.rvsc.2014.01.007]
`Brown CT, Davis-Richardson AG, Giongo A, Gano KA,
`Crabb DB, Mukherjee N, Casella G, Drew JC, Ilonen J, Knip
`M, Hyöty H, Veijola R, Simell T, Simell O, Neu J, Wasserfall
`CH, Schatz D, Atkinson MA, Triplett EW. Gut microbiome
`metagenomics analysis suggests a functional model for the
`development of autoimmunity for type 1 diabetes. PLoS
`One 2011; 6: e25792 [PMID: 22043294 DOI: 10.1371/journal.
`pone.0025792]
`3 Hansen CH, Krych L, Nielsen DS, Vogensen FK, Hansen LH,
`Sørensen SJ, Buschard K, Hansen AK. Early life treatment with
`
`5
`
`4
`
`vancomycin propagates Akkermansia muciniphila and reduc-
`es diabetes incidence in the NOD mouse. Diabetologia 2012; 55:
`2285-2294 [PMID: 22572803 DOI: 10.1007/s00125-012-2564-7]
`Png CW, Lindén SK, Gilshenan KS, Zoetendal EG, McSwee-
`ney CS, Sly LI, McGuckin MA, Florin TH. Mucolytic bacteria
`with increased prevalence in IBD mucosa augment in vitro
`utilization of mucin by other bacteria. Am J Gastroenterol 2010;
`105: 2420-2428 [PMID: 20648002 DOI: 10.1038/ajg.2010.281]
`Scher JU, Sczesnak A, Longman RS, Segata N, Ubeda C,
`Bielski C, Rostron T, Cerundolo V, Pamer EG, Abramson
`SB, Huttenhower C, Littman DR. Expansion of intestinal
`Prevotella copri correlates with enhanced susceptibility to
`arthritis. Elife 2013; 2: e01202 [PMID: 24192039 DOI: 10.7554/
`eLife.01202]
`6 Ubeda C, Taur Y, Jenq RR, Equinda MJ, Son T, Samstein M,
`Viale A, Socci ND, van den Brink MR, Kamboj M, Pamer EG.
`Vancomycin-resistant Enterococcus domination of intestinal
`microbiota is enabled by antibiotic treatment in mice and
`precedes bloodstream invasion in humans. J Clin Invest 2010;
`120: 4332-4341 [PMID: 21099116 DOI: 10.1172/jci43918]
`7 Hildebrandt MA, Hoffmann C, Sherrill-Mix SA, Keilbaugh
`SA, Hamady M, Chen YY, Knight R, Ahima RS, Bushman
`F, Wu GD. High-fat diet determines the composition of the
`murine gut microbiome independently of obesity. Gastro-
`enterology 2009; 137: 1716-1724.e1-e2 [PMID: 19706296 DOI:
`10.1053/j.gastro.2009.08.042]
`8 Antonopoulos DA, Huse SM, Morrison HG, Schmidt TM,
`Sogin ML, Young VB. Reproducible community dynamics of
`the gastrointestinal microbiota following antibiotic perturba-
`tion. Infect Immun 2009; 77: 2367-2375 [PMID: 19307217 DOI:
`10.1128/IAI.01520-08]
`9 Wilson KH, Brown RS, Andersen GL, Tsang J, Sartor B.
`Comparison of fecal biota from specific pathogen free and
`feral mice. Anaerobe 2006; 12: 249-253 [PMID: 17070078 DOI:
`10.1016/j.anaerobe.2006.09.002]
`10 Ley RE, Bäckhed F, Turnbaugh P, Lozupone CA, Knight RD,
`Gordon JI. Obesity alters gut microbial ecology. Proc Natl
`Acad Sci USA 2005; 102: 11070-11075 [PMID: 16033867 DOI:
`10.1073/pnas.0504978102]
`11 Walk ST, Blum AM, Ewing SA, Weinstock JV, Young VB.
`Alteration of the murine gut microbiota during infection with
`the parasitic helminth Heligmosomoides polygyrus. Inflamm
`Bowel Dis 2010; 16: 1841-1849 [PMID: 20848461 DOI: 10.1002/
`ibd.21299]
`12 Hildebrand F, Nguyen TL, Brinkman B, Yunta RG, Cauwe B,
`Vandenabeele P, Liston A, Raes J. Inflammation-associated
`enterotypes, host genotype, cage and inter-individual effects
`drive gut microbiota variation in common laboratory mice.
`Genome Biol 2013; 14: R4 [PMID: 23347395 DOI: 10.1186/gb-
`2013-14-1-r4]
`13 Qin J, Li Y, Cai Z, Li S, Zhu J, Zhang F, Liang S, Zhang W,
`Guan Y, Shen D, Peng Y, Zhang D, Jie Z, Wu W, Qin Y, Xue
`W, Li J, Han L, Lu D, Wu P, Dai Y, Sun X, Li Z, Tang A,
`Zhong S, Li X, Chen W, Xu R, Wang M, Feng Q, Gong M, Yu J,
`Zhang Y, Zhang M, Hansen T, Sanchez G, Raes J, Falony
`G, Okuda S, Almeida M, LeChatelier E, Renault P, Pons N,
`Batto JM, Zhang Z, Chen H, Yang R, Zheng W, Li S, Yang
`H, Wang J, Ehrlich SD, Nielsen R, Pedersen O, Kristiansen
`K, Wang J. A metagenome-wide association study of gut
`microbiota in type 2 diabetes. Nature 2012; 490: 55-60 [PMID:
`23023125 DOI: 10.1038/nature11450]
`14 Pang W, Vogensen FK, Nielsen DS, Hansen AK. Faecal
`and caecal microbiota profiles of mice do not cluster in the
`same way. Lab Anim 2012; 46: 231-236 [PMID: 22723645 DOI:
`10.1258/la.2012.011128]
`15 Bergström A, Licht TR, Wilcks A, Andersen JB, Schmidt
`LR, Grønlund HA, Vigsnaes LK, Michaelsen KF, Bahl MI.
`Introducing GUt low-density array (GULDA): a validated ap-
`proach for qPCR-based intestinal microbial community anal-
`ysis. FEMS Microbiol Lett 2012; 337: 38-47 [PMID: 22967145
`
`WJG|www.wjgnet.com
`
`17732
`
`December 21, 2014|Volume 20|Issue 47|
`
`

`

`DOI: 10.1111/1574-6968.12004]
`16 Krych L, Hansen CH, Hansen AK, van den Berg FW, Nielsen
`DS. Quantitatively different, yet qualitatively alike: a meta-
`analysis of the mouse core gu

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