throbber
Downloaded from
`Downloaded from
`Downloaded from
`
`http://aem.asm.org/
`http://aem.asm.org/
`http://aem.asm.org/
`
` on November 15, 2016 by guest
` on November 15, 2016 by guest
` on November 15, 2016 by guest
`
`APPLIED AND ENVIRONMENTAL MICROBIOLOGY, Feb. 2008, p. 660–666
`0099-2240/08/$08.00⫹0 doi:10.1128/AEM.01261-07
`Copyright © 2008, American Society for Microbiology. All Rights Reserved.
`
`Vol. 74, No. 3
`
`Gnotobiotic Mouse Immune Response Induced by Bifidobacterium sp.
`Strains Isolated from Infants䌤
`Odile Me´nard,1 Marie-Jose´ Butel,1 Vale´rie Gaboriau-Routhiau,2 and Anne-Judith Waligora-Dupriet1*
`EA4065, Faculte´ des Sciences Pharmaceutiques et Biologiques, Universite´ Paris Descartes, 4 Avenue de l’Observatoire,
`75270 Paris Cedex 06, France,1 and Unite´ d’Ecologie et de Physiologie du Syste`me Digestif, Institut National de
`la Recherche Agronomique, Domaine de Vilvert, 78352 Jouy en Josas Cedex, France2
`
`Received 7 June 2007/Accepted 28 November 2007
`
`Bifidobacterium, which is a dominant genus in infants’ fecal flora and can be used as a probiotic, has shown
`beneficial effects in various pathologies, including allergic diseases, but its role in immunity has so far been
`little known. Numerous studies have shown the crucial role of the initial intestinal colonization in the
`development of the intestinal immune system, and bifidobacteria could play a major role in this process. For
`a better understanding of the effect of Bifidobacterium on the immune system, we aimed at determining the
`impact of Bifidobacterium on the T-helper 1 (TH1)/TH2 balance by using gnotobiotic mice. Germfree mice were
`inoculated with Bifidobacterium longum NCC2705, whose genome is sequenced, and with nine Bifidobacterium
`strains isolated from infants’ fecal flora. Five days after inoculation, mice were killed. Transforming growth
`factor ␤1 (TGF-␤1), interleukin-4 (IL-4), IL-10, and gamma interferon (IFN-␥) gene expressions in the ileum
`and IFN-␥, tumor necrosis factor alpha (TNF-␣), IL-10, IL-4, and IL-5 secretions by splenocytes cultivated for
`48 h with concanavalin A were quantified. Two Bifidobacterium species had no effect (B. adolescentis) or little
`effect (B. breve) on the immune system. Bifidobacterium bifidum, Bifidobacterium dentium, and one B. longum
`strain induced TH1 and TH2 cytokines at the systemic and intestinal levels. One B. longum strain induced a TH2
`orientation with high levels of IL-4 and IL-10, both secreted by splenocytes, and of TGF-␤ gene expression in
`the ileum. The other two strains induced TH1 orientations with high levels of IFN-␥ and TNF-␣ splenocyte
`secretions. Bifidobacterium’s capacity to stimulate immunity is species specific, but its influence on the orien-
`tation of the immune system is strain specific.
`
`Bifidobacteria are gram-positive, anaerobic bacteria that
`represent up to 90% of the total gut microflora in breast-fed
`babies (7) and up to 15% in adults (22). The presence of such
`high levels of bifidobacteria in the human intestine is suggested
`to contribute to human health, leading to the use of bifidobac-
`teria as probiotics. Indeed, beneficial effects of bifidobacteria
`are shown for various diseases, e.g., diarrhea associated with
`rotavirus or antibiotics and some inflammatory intestinal dis-
`eases (27). Though some studies suggest a role for bifidobac-
`teria in prevention of allergic diseases, data are scarce and
`controversial (2, 5). Moreover, the role of bifidobacteria in
`immunity has so far been little known.
`For a few years, the incidences of allergic diseases have been
`increasing in developed countries. A disturbance in the bal-
`ance of T-helper 1 (TH1)/TH2 lymphocyte responses to exog-
`enous antigens toward a TH2 phenotype is considered a major
`event in the onset of allergic diseases. Though the mechanisms
`of these disturbances are still controversial, several studies
`suggest a prominent role for microorganisms from the envi-
`ronment and the normal commensal flora of the gastrointes-
`tinal tract in these disregulations or in the prevention of aller-
`gic sensitization (15, 28). In fact, intestinal colonization, in
`
`* Corresponding author. Mailing address: EA4065 Ecosyste`me
`Intestinal, Probiotiques, Antibiotiques, Faculte´ des Sciences Phar-
`maceutiques et Biologiques, Universite´ Paris Descartes, 4 avenue
`de l’Observatoire, 75270 Paris Cedex 06, France. Phone: 33 (1) 53
`73 99 20. Fax: 33 (1) 53 73 99 23. E-mail: anne-judith.waligora
`@univ-paris5.fr.
`䌤 Published ahead of print on 14 December 2007.
`
`660
`
`which the earliest contact with microbes occurs, has a marked
`impact on the maturation of the immune system (IS), which is
`underdeveloped at birth (9). The natural sequential coloniza-
`tion process of the digestive tract is complex and was shown to
`crucially influence immune response establishment during the
`neonatal period, especially TH1/TH2 balance (8, 34, 35). Sev-
`eral clinical studies showed a relationship between allergic
`diseases and the gut microbiota, pointing out quantitative and
`qualitative differences in bifidobacterial colonization (4, 12, 26,
`37). These studies showed that allergic patients had lower
`counts of Bifidobacterium than healthy control subjects. Fur-
`thermore, differences in species were observed. Bifidobacte-
`rium adolescentis and Bifidobacterium longum were isolated
`from allergic infants as
`the predominant bifidobacteria,
`whereas the predominant ones isolated from age-matched
`healthy infants were Bifidobacterium infantis, Bifidobacterium
`bifidum, and Bifidobacterium breve (26). Although these data
`suggested a link between bifidobacterial species and atopy or
`tolerance, no clear relation of causes and effects has been
`demonstrated yet. An in vivo study of newborn mice showed
`that B. infantis establishment was important for restoration of
`the usual oral tolerance process and especially for immuno-
`globulin E suppression (34). The impact of bifidobacteria on
`immunity has been specified with in vitro studies showing the
`roles of individual bifidobacterial species (11, 21, 29, 39).
`Nonetheless, no in vivo study has confirmed such observations.
`Thus, it is essential, at a time when dramatic increases in the
`prevalences of allergic diseases are a major concern in western
`countries, to understand the connections between allergy and
`intestinal flora and more particularly between allergy and bi-
`
`Genome & Co. v. Univ. of Chicago, PGR2019-00002
`UNIV. CHICAGO EX. 2004 - 1/8
`
`

`

`VOL. 74, 2008
`
`IMPACT OF BIFIDOBACTERIUM ON IMMUNITY
`
`661
`
`Downloaded from
`
`http://aem.asm.org/
`
` on November 15, 2016 by guest
`
`FIG. 1. Levels of pro-TH1 cytokine secretion by splenocytes stimulated for 48 h with ConA (3 ␮g/␮l). (A) IFN-␥; (B) TNF-␣. Data obtained
`by ELISA are presented as means plus SEMs. P was ⬍0.05 (ⴱ) and ⬍0.01 (ⴱⴱ) for comparison with GF mice. Reference groups are represented
`by open bars.
`
`fidobacteria, which may be used as a probiotic for preventing
`these diseases (3, 5).
`In this study, using gnotobiotic mice and quantification of
`pro- and anti-inflammatory-cytokine production and/or gene
`expression, we aimed to assess the roles of bifidobacterial
`strains belonging to different species and isolated from the
`fecal flora of infants on TH1/TH2 balance.
`
`MATERIALS AND METHODS
`
`Bacterial strains. Ten Bifidobacterium strains were studied. Nine strains were
`isolated from the dominant fecal flora of 3- to 24-month-old healthy infants,
`including three B. longum strains (BS64, BS49, and CUETM 89-215), three B.
`breve strains (BS94, BS52, BS56), one B. bifidum strain (BS42), one B. dentium
`strain (BS21), and one B. adolescentis strain (BS27). The tenth strain was B.
`longum NCC2705, whose genome is entirely sequenced (31). Bifidobacterial
`genera were identified according to their morphological characteristics, by the
`presence of fructose-6-phospho-phosphoketolase, and by PCR according to Kok
`et al. (18). Species identification was realized using a multiplex PCR targeting the
`16S-23S rRNA gene intergenic spacer according to Mullie´ et al. (23). All strains
`were grown on Wilkins Chalgren agar base containing D-glucose (10 g/liter),
`L-cysteine (0.5 g/liter), and Tween 80 (0.5%, vol/vol) (6) and in trypticase-
`glucose-yeast extract-hemin broth. They were incubated at 37°C in an anaerobic
`chamber (dw Scientific, AES laboratoires, Bruzz, France).
`Experimental animals. Six- to 7-week-old female germfree (GF) C3H/HeN
`and conventional C3H/HeN mice were purchased from the INRA (Jouy-en-
`
`Josas, France). GF and conventional mice were maintained in sterile isolators
`(JCE Biotechnology, Hauterive, France) and fed ad libitum with a commercial
`rodent diet sterilized by gamma irradiation (40 megarads). The animal experi-
`mentation was conducted in accordance with the rules of our institution and with
`Council of Europe Guidelines, with license for experimental studies on living
`animals and animal facility agreement no. A750602 (Direction of Veterinary
`Services, Prefecture de Police de Paris, France).
`Twelve groups of 6 to 12 mice were studied. GF and conventional mice as well
`as mice which had been mono-associated with one of the 10 Bifidobacterium
`strains were included.
`Colonization of GF mice. Bifidobacterial mono-associated mice were obtained
`by gastric intubation of GF mice with a 48-h culture of one of the bifidobacterial
`strains comprising 106 to 108 CFU/ml. This range of doses did not influence the
`colonization level. Both GF and conventional mice received the same volume
`(300 ␮l) of sterile water. Feces were collected 48 h after force feeding to check
`the sterility in GF mice and the bacterial establishment in monobiotic mice.
`Cecal contents were collected after sacrifice in order to measure the bifidobac-
`terial levels in monobiotic mice, to determine the fecal microbiota compositions
`in conventional mice, and to check the sterility in GF mice. Dilutions of feces and
`cecal contents in a prereduced peptone liquid medium were performed in anaer-
`obiosis and spread on Wilkins Chalgren agar base for monobiotic mice and on
`various media allowing the isolation and the quantification of the main bacterial
`genera for conventional mice, as previously described (6).
`Lymphocyte culture. Six days after their inoculation, mice were sacrificed with
`an intraperitoneal injection of pentobarbital sodium (CEVA sante´ animale,
`Libourne, France). Spleens were gently crushed, filtered through a 70-␮m nylon
`filter (Falcon, VWR, Val de Fontenay, France), and rinsed in RPMI 1640
`
`Genome & Co. v. Univ. of Chicago, PGR2019-00002
`UNIV. CHICAGO EX. 2004 - 2/8
`
`

`

`662
`
`ME´NARD ET AL.
`
`APPL. ENVIRON. MICROBIOL.
`
`Downloaded from
`
`http://aem.asm.org/
`
` on November 15, 2016 by guest
`
`FIG. 2. Levels of pro-TH2 cytokine secretion by splenocytes stimulated for 48 h with ConA (3 ␮g/␮l). (A) IL-10; (B) IL-4. Data obtained by
`ELISA are presented as means plus SEMs. P was ⬍0.05 (ⴱ) and ⬍0.01 (ⴱⴱ) for comparison with GF mice. Reference groups are represented by
`open bars.
`
`(Gibco, Fisher-Bioblock, Illkirch, France). Spleen cells (SC) were then purified
`for 5 minutes in ice with Tris-buffered NH4Cl according to Nicaise et al. (25). SC
`were rinsed and resuspended in 1 ml of RPMI 1640 containing 25 mM of HEPES
`buffer, 1% of L-glutamine, 1% of penicillin, streptomycin, 1% of fungizon, and
`10% of fetal calf serum. The number of viable cells was determined by the trypan
`blue dye (0.25%) exclusion method. SC were adjusted to 2.106 cells/well and
`cultured in 24-well plates with and without 3 ␮g/␮l of concanavalin A (ConA)
`(Sigma, France) at 37°C in a 5% CO2-95% air atmosphere. Supernatants were
`collected after 48 h of culture by centrifugation at 1,800 rpm, 20°C, for 5 minutes
`and conserved at ⫺20°C.
`Cytokine level measurement. Levels of gamma interferon (IFN-␥), tumor
`necrosis factor alpha (TNF-␣), interleukin 10 (IL-10), IL-4, and IL-5 in culture
`supernatants were quantified using enzyme-linked immunosorbent assay
`(ELISA) kits (eBiosciences, Montrouge, France) according to the manufactur-
`er’s instructions. Detection limits for ELISAs were as follows: IL-4 and IL-5, 4
`pg/ml; TNF-␣, 8 pg/ml; and IFN-␥ and IL-10, 15 pg/ml. Duplicate wells were run
`for each sample. Results are presented as mean values plus standard errors of the
`means (SEMs) for each group.
`Cytokine expression from the terminal ileum. A total of 2.5 cm of the entire
`terminal ileum, including the Peyer patches, was crushed with an Ultra-Turrax
`J25 instrument (Fisher-Bioblock) for 40 seconds, and total mRNA was extracted
`using the TRIzol reagent method (Invitrogen, Illkirch, France) according to the
`manufacturer’s instructions. mRNA was treated by DNase I (Invitrogen) and
`converted into cDNA by reverse transcription using the Superscript II and Oligo
`dT12-18 primers (Invitrogen). The cDNA obtained was subjected to real-time
`PCR using Smart Cycler (Cepheid, Sunnyvale, Canada). The Quantitect Probe
`PCR master mix and Quantitect gene expression assay (Qiagen, Courtaboeuf,
`
`France) kits were used to quantify the transforming growth factor ␤1 (TGF-␤1)-
`and IL-4 gene expressions directly on mouse ileum. IFN-␥- and IL-10 gene
`expressions were quantified by using a Smart kit for Sybr green 1 (Eurogentec,
`Angers, France) and 900 nM specific primers (IFN-␥ forward [5⬘-AGCAACAG
`CAAGGCGAAAA-3⬘] and reverse [5⬘-CTGGACCTGTGGGTTGTTGA-3⬘]
`and IL-10 forward [5⬘-TTTGAATTCCCTGGGTGAGAA-3⬘] and reverse [5⬘-A
`CAGGGGAGAAATCGATGACA-3⬘]) (38). Dosages were performed in dupli-
`cate. As the efficacy of amplification for each gene was confirmed to be similar
`to that of the ␤-actin-encoding gene, which was used as a reference (Qiagen),
`data analysis was performed with the 2⫺⌬⌬Ct method as described by Livak and
`Schmittgen (20). For each group and each cytokine, gene expression level is
`determined by comparison with that in the GF mouse group.
`Statistical analysis. The Kruskal-Wallis test was used to determine the signif-
`icance of the differences between the groups, and the Mann-Whitney U test was
`performed for pairwise comparison. P values of less than 0.05 were considered to
`be statistically significant. Data were analyzed using SPSS (version 12.0).
`
`RESULTS
`
`Bacterial colonization level. GF mice were sterile before
`inoculation. After inoculation, bifidobacteria were established
`at a high level, with a mean of 9.5 ⫾ 0.8 log10 CFU/g of cecal
`content, ranging from 8.0 ⫾ 0.8 log10 CFU/g of cecal content
`for B. adolescentis to 10.3 ⫾ 0.3 log10 CFU/g of cecal content
`for B. breve BS56. No exogenous bifidobacteria were found.
`
`Genome & Co. v. Univ. of Chicago, PGR2019-00002
`UNIV. CHICAGO EX. 2004 - 3/8
`
`

`

`Downloaded from
`
`http://aem.asm.org/
`
` on November 15, 2016 by guest
`
`VOL. 74, 2008
`
`IMPACT OF BIFIDOBACTERIUM ON IMMUNITY
`
`663
`
`TABLE 1. Orientation of the IS in GF mice mono-associated with different Bifidobacterium strains
`
`Bifidobacterium strain
`
`Bacterial
`colonization
`levela
`
`Splenocyte secretion (peripheral IS)b
`
`Gene expression in final
`ileum (intestinal IS)b
`
`IL-10
`
`IL-4
`
`IL-5
`
`TNF-␣
`
`IFN-␥
`
`IFN-␥
`
`TGF-␤
`
`IL-4
`
`Orientation of TH1/TH2 balance
`according to our in vivo study
`
`B. adolescentis BS27
`B. breve BS94
`
`8.0 ⫾ 0.8
`10.3 ⫾ 0.3
`
`B. breve BS52
`
`B. breve BS56
`B. longum BS64
`
`9.9 ⫾ 0.9
`
`9.1 ⫾ 0.1
`9.6 ⫾ 0.2
`
`B. longum NCC2705
`
`9.8 ⫾ 0.1
`
`B. longum BS49
`
`9.9 ⫾ 0.1
`
`0c
`/4d
`
`0
`
`0
`0
`
`0
`
`0
`⫻11.5d
`
`⫻7d
`⫻11e
`
`0
`
`0
`
`0
`
`0
`
`0
`/2
`
`0
`
`0
`0
`
`0
`
`0
`
`B. longum CUETM
`89–215
`B. bifidum BS42
`B. dentium BS21
`
`10.0 ⫾ 0.1
`
`⫻4.5e
`
`⫻6.5e ⫻1.5
`
`9.1 ⫾ 0.2
`8.7 ⫾ 0.3
`
`0
`0
`
`⫻2e
`⫻3.5e
`
`0
`0
`
`0
`/2
`
`0
`
`0
`0
`
`⫻2e
`
`⫻2d
`
`⫻2e
`
`⫻1.5
`0
`
`0
`0
`
`0
`
`0
`/4d
`
`⫻3e
`
`⫻2d
`
`⫻7.5e
`
`⫻3.5e
`⫻13e
`
`0
`0
`
`0
`
`0
`0
`
`0
`
`/7d
`
`/5
`
`0
`0
`
`0
`⫻2d
`
`⫻5.5d
`
`⫻2.5d
`⫻3.5e
`
`⫻4e
`
`0
`
`⫻3e
`
`⫻2
`⫻3e
`
`0
`/5d
`
`0
`0
`
`0
`
`Noneffector
`Suppressor (peripheral IS)/TH1/
`TH2 (intestinal IS)
`/12d Non inducer (peripheral IS)/
`TH1/TH2 (intestinal IS)
`Low TH2 inducer
`TH2 (peripheral IS)/suppressor
`(intestinal IS)
`TH1 (peripheral IS)/suppressor
`(intestinal IS)
`TH1 (peripheral IS)/TH2
`(intestinal IS)
`TH1/TH2
`
`0
`
`0
`
`/8d
`0
`
`TH1/TH2
`TH1/TH2
`
`a Expressed as log10 numbers of UFC/g cecal content.
`b Differences are expressed in comparison with the level for GF mice. ⫻n, upregulation by a factor of n; /n, downregulation by a factor of n.
`c No difference.
`d P ⬍ 0.05.
`e P ⬍ 0.01.
`
`The microbiota of conventional mice consisted of 8.2 log10
`CFU total aerobes/g of cecal content and 9.4 log10 CFU total
`anaerobes/g of cecal content, with numerous lactobacilli (8.7
`log10 CFU/g of cecal content) and no bifidobacteria.
`Cytokine production from splenocytes in vitro. Cytokine
`secretions by SC are shown in Fig. 1 and 2, and data are
`compiled in Table 1. Spontaneous secretions of IFN-␥, TNF-␣,
`IL-4, IL-5, and IL-10 were observed in the unstimulated
`splenocytes’ supernatants, but with rates below the detection/
`quantification threshold. Only the results obtained with SC
`stimulated with 3 ␮g/␮l of ConA are considered.
`TH1 cytokines. According to their capacities to influence
`IFN-␥ secretion, strains can be significantly separated into
`three groups in comparison with GF mice, i.e., inducers (three
`B. longum strains out of four, B. bifidum, and B. dentium),
`suppressors (the fourth B. longum strain), and strains with no
`effect (the three B. breve and B. adolescentis strains) (Fig. 1A).
`Various effects were observed for the B. longum strains: B.
`longum CUETM 89-215 was a higher inducer than B. longum
`NCC2705 and BS49, and B. longum BS64 was shown to be a
`suppressor.
`Three strains of B. longum significantly induced TNF-␣ se-
`cretion, and the other strains, regardless of species, had no
`effect in comparison with the level for GF mice (Fig. 1B).
`TH2 cytokines. According to their capacities to influence
`IL-4 secretion, two groups of strains can be observed: two B.
`longum strains, one B. breve strain, B. bifidum, and B. dentium
`significantly induced this secretion, and the other strains had
`no effect in comparison with the level for GF mice (Fig. 2B).
`No strains had a significant effect on IL-5 secretion.
`Regulatory cytokine: IL-10. Strains can be significantly sep-
`arated into three groups, i.e., inducers (two B. longum strains),
`a suppressor (BS94), and strains with no effect on IL-10 secre-
`tion (Fig. 2A).
`
`Cytokine gene expression in the terminal ileum. Cytokine
`gene expressions in the terminal ileum are shown in Fig. 3, and
`data are compiled in Table 1. IL-10 gene expression was below
`the threshold of detection. By contrast, TGF-␤1, IL-4, and
`IFN-␥ gene expressions were detected in conventional, GF,
`and mono-associated mice.
`TGF-␤1 gene expression. All the strains but three (the B.
`bifidum strain, the B. adolescentis strain, and B. longum BS49)
`induced TGF-␤1 gene expression compared with the level for
`GF mice (Fig. 3A).
`IFN-␥ gene expression. Only the establishment of a conven-
`tional flora induced a significant increase in IFN-␥ gene ex-
`pression compared with the level for GF mice (Fig. 3B). This
`expression was significantly inhibited only in mice mono-asso-
`ciated with B. longum BS49 (P ⬍ 0.05) and CUETM 89-215
`(P ⫽ 0.067) compared with the level for GF mice. By contrast,
`the two other B. longum strains (NCC2705 and BS64) did not
`exert such an effect.
`IL-4 gene expression. Two B. breve strains (BS94 and BS52)
`had suppressor effects on IL-4 gene expression, and the other
`strains, regardless of species, had no effect compared with the
`level for GF mice (Fig. 3C). Except for one mouse, B. bifidum
`significantly inhibited IL-4 gene expression compared with the
`level for GF mice.
`
`DISCUSSION
`
`This is the first time that the impacts of various Bifidobac-
`terium strains belonging to several species on immunity have
`been compared in an in vivo model, i.e., gnotobiotic mice. Our
`study has shown that Bifidobacterium’s capacity to stimulate
`immunity is species specific but its influence on the orientation
`of the IS operates in a strain-specific manner.
`In our study, we chose to include four strains of B. longum
`
`Genome & Co. v. Univ. of Chicago, PGR2019-00002
`UNIV. CHICAGO EX. 2004 - 4/8
`
`

`

`664
`
`ME´NARD ET AL.
`
`APPL. ENVIRON. MICROBIOL.
`
`Downloaded from
`
`http://aem.asm.org/
`
` on November 15, 2016 by guest
`
`FIG. 3. Increases (n-fold) in cytokine gene expression in the terminal ileum. (A) TGF-␤1; (B) IFN-␥; (C) IL-4. Results are expressed as
`individual data points, and bars represent means. The y axis represents the amount of the target gene relative to the level for GF mice, normalized
`with ␤-actin. P values were ⬍0.05 (ⴱ) and ⬍0.01 (ⴱⴱ) for comparison with GF mice.
`
`and three strains of B. breve among the 10 Bifidobacterium
`strains tested. Indeed, in a previous prospective study, these
`species were isolated as the predominant bifidobacteria in in-
`fants (personal data). In GF mice, Bifidobacterium coloniza-
`tion occurred at a high level, as high as that in the infants’ fecal
`flora from which the strains were isolated. To investigate the
`influence of bifidobacteria on TH1/TH2 orientation, IL-4 and
`
`IL-5 were studied as pro-TH2 cytokines, IFN-␥ and TNF-␣ as
`pro-TH1 cytokines, and IL-10 and TGF-␤ as regulatory cyto-
`kines (28). The last group consists of cytokines that can inhibit
`T-cell proliferation and differentiation but in some cases can
`be associated with a TH2 profile. Indeed, IL-10 and TGF-␤ can
`be produced by TH2 lymphocytes as well as regulatory T cells
`(16, 28).
`
`Genome & Co. v. Univ. of Chicago, PGR2019-00002
`UNIV. CHICAGO EX. 2004 - 5/8
`
`

`

`Downloaded from
`
`http://aem.asm.org/
`
` on November 15, 2016 by guest
`
`VOL. 74, 2008
`
`IMPACT OF BIFIDOBACTERIUM ON IMMUNITY
`
`665
`
`The Bifidobacterium strains can be separated into three
`groups according to their impacts on cytokine profiles (Table
`1). The first group includes strains which induced TH1 and/or
`TH2 cytokines, which are the B. bifidum and B. dentium strains
`as well as the four B. longum strains. The second group gathers
`the three B. breve strains which had few effects on immunity.
`The third group includes strains without any effect on immu-
`nity compared with the level for GF mice and comprises only
`one strain, namely, that of B. adolescentis.
`In the first group, B. longum strains direct systemic immunity
`toward a TH1 or TH2 profile, depending on the strains. B.
`longum BS64 induced a predominant TH2 profile. By contrast,
`B. longum BS49 and NCC2705 appeared to be high inducers of
`TH1 cytokines. B.
`longum CUETM 89-215 induced a high
`production level of every cytokine tested then, inducing TH1
`and TH2 cytokines at the same time. At the intestinal level, B.
`longum BS49 inhibited only IFN-␥ gene expression and then
`induced a TH2 profile. Three strains induced TGF-␤ gene
`expression; thus, they have a suppressor effect. Among them,
`B. longum CUETM 89-215 also inhibited IFN-␥ gene expres-
`sion. As TGF-␤ produced by TH3 cells could act toward a TH2
`profile (28), this strain was considered to act toward a TH2
`profile. These results, showing that B. longum appeared to
`orientate TH1/TH2 balance depending on the strain, tally with
`in vitro studies using heat-inactivated B. longum strains or
`bacterial extracts (11, 19, 30, 39). Our in vivo model allows us
`to consider interactions between bacteria and intestinal cells
`(whether immune or not, as with epithelial cells) and to take
`the IS’s complexity into account. However, the fact that we are
`working with entire tissues and not with isolated populations of
`immune cells could account for the differences between our
`results and those previously cited (11, 19, 30, 39).
`As with B. longum CUETM 89-215, B. bifidum and B. den-
`tium colonization in GF mice induced TH1 and TH2 cytokines
`at the systemic level at the same time. These data contrast with
`previous in vitro studies suggesting that B. bifidum induces only
`TH2 cytokines (11, 39). In our study, except for the inhibition
`of IL-4 gene expression, B. bifidum had no impact at the gut
`level. This corroborates previous data obtained in vivo and in
`vitro, showing that peritoneal macrophages from B. bifidum
`mono-associated GF mice produced neither TH1 cytokines
`(IL-1 and TNF-␣) nor TH2 ones (IL-6) (24) and showing that
`B. bifidum was unable to induce TNF-␣, IL-6, and IL-1␤ pro-
`duction (29). However, some B. bifidum strains isolated from
`an infant’s intestine could induce IL-8 secretion but not IL-6
`secretion (21). The multiplicity of strains and models used
`(macrophage-like cell line, dendritic cells from cord blood,
`Caco2, etc.) makes it difficult to draw a conclusion and could
`explain discrepancies between data. The last Bifidobacterium
`species of this group, B. dentium, induced TGF-␤ gene expres-
`sion, but that was the only influence of this strain on the
`terminal ileum. This is the first report of the impact on immu-
`nity of B. dentium, which is not among the usual species in the
`dominant gut microbiota.
`In the second group, B. breve strains appear to be poor
`inducers of immunity, suppressors, or strains ineffective at the
`systemic level. Only B. breve BS56 had an impact and induced
`a TH2-driven peripheral immune response. This TH2 immune
`orientation confirmed in vitro studies with living B. breve (11)
`and B. breve supernatant (14). B. breve BS94 was a suppressor,
`
`and B. breve BS52 was ineffective; their establishment induced
`an intestinal immune response activating TGF-␤1 gene expres-
`sion but inhibiting IL-4 gene expression. Similarly, a strain-
`specific effect was observed by Morita et al., who tested four B.
`breve strains (21).
`The establishment in GF mice of B. adolescentis, which is the
`constituent of the third group, had no ability to stimulate the
`IS, as observed by numerous authors (17, 21, 32, 39). However,
`He et al. showed that B. adolescentis induced a TH1-driven
`immune response, but they used a murine macrophage-like cell
`line (11), and Hessle et al. showed that one strain of B. ado-
`lescentis induced TNF-␣ secretion by human mononuclear
`cells, but this was with bacteria killed by UV (13). This killing
`process can change the capacity of bacteria to stimulate im-
`mune responses, and these authors used at least 10 times more
`bacteria/ml than the others and we did.
`Therefore, the present study demonstrates that strains more
`than species are linked to IS orientation. This strain-specific
`effect may be linked with specific secretion products (14) and
`structures, like DNA CpG unmethylated patterns (19, 33),
`exopolysaccharides (1), and peptidoglycans (30, 36, 40). The
`last varies in structure between bifidobacterial species but also
`between strains belonging to the same species (10).
`Our study offers a better understanding of the heterogeneity
`of Bifidobacterium species and of their potential role in IS
`stimulation. Interestingly, this study raises numerous questions
`concerning the use of Bifidobacterium as a probiotic in immune
`diseases such as allergies. The matters regarding the choice of
`one or several strains and the interaction between several bac-
`terial genera (synergy or antagonism) for probiotic supplemen-
`tation as a preventive treatment of allergic disease are still to
`be clarified.
`
`ACKNOWLEDGMENTS
`
`The work of O. Me´nard was supported by the Socie´te´ Franc¸aise de
`Nutrition Ente´rale et Parente´rale and Nutricia—Nutrition Clinique.
`Our thanks to M. C. Moreau for her useful advice.
`
`REFERENCES
`1. Amrouche, T., Y. Boutin, G. Prioult, and I. Fliss. 2006. Effects of bifidobac-
`terial cytoplasm, cell wall and exopolysaccharide on mouse lymphocyte pro-
`liferation and cytokine production. Int. Dairy J. 16:70–80.
`2. Bjorksten, B. 2004. Effects of intestinal microflora and the environment on
`the development of asthma and allergy. Springer Semin. Immunopathol.
`25:257–270.
`3. Bjorksten, B. 2005. Evidence of probiotics in prevention of allergy and
`asthma. Curr. Drug Targets Inflamm. Allergy 4:599–604.
`4. Bjorksten, B., P. Naaber, E. Sepp, and M. Mikelsaar. 1999. The intestinal
`microflora in allergic Estonian and Swedish 2-year-old children. Clin. Exp.
`Allergy 29:342–346.
`5. Boyle, R. J., and M. L. Tang. 2006. The role of probiotics in the management
`of allergic disease. Clin. Exp. Allergy 36:568–576.
`6. Butel, M. J., N. Roland, A. Hibert, F. Popot, A. Favre, A. C. Tesse`dre, M.
`Bensaada, A. Rimbault, and O. Szylit. 1998. Clostridial pathogenicity in
`experimental necrotising enterocolitis in gnotobiotic quails and protective
`role of bifidobacteria. J. Med. Microbiol. 47:391–399.
`7. Fanaro, S., R. Chierici, P. Guerrini, and V. Vigi. 2003. Intestinal microflora
`in early infancy: composition and development. Acta Paediatr. Suppl.
`91(441):48–55.
`8. Gaboriau-Routhiau, V., P. Raibaud, C. Dubuquoy, and M. C. Moreau. 2003.
`Colonization of gnotobiotic mice with human gut microflora at birth protects
`against Escherichia coli heat-labile enterotoxin-mediated abrogation of oral
`tolerance. Pediatr. Res. 54:739–746.
`9. Guy-Grand, D., C. Griscelli, and P. Vassalli. 1974. The gut-associated lym-
`phoid system: nature and properties of the large dividing cells. Eur. J. Im-
`munol. 4:435–443.
`10. Habu, Y., M. Nagaoka, T. Yokokura, and I. Azuma. 1987. Structural studies
`of cell wall polysaccharides from Bifidobacterium breve YIT 4010 and re-
`lated Bifidobacterium species. J. Biochem. (Tokyo) 102:1423–1432.
`
`Genome & Co. v. Univ. of Chicago, PGR2019-00002
`UNIV. CHICAGO EX. 2004 - 6/8
`
`

`

`Downloaded from
`
`http://aem.asm.org/
`
` on November 15, 2016 by guest
`
`666
`
`ME´NARD ET AL.
`
`APPL. ENVIRON. MICROBIOL.
`
`11. He, F., H. Morita, A. C. Ouwehand, M. Hosoda, M. Hiramatsu, J. Kurisaki,
`E. Isolauri, Y. Benno, and S. Salminen. 2002. Stimulation of the secretion of
`pro-inflammatory cytokines by Bifidobacterium strains. Microbiol. Immunol.
`46:781–785.
`12. He, F., A. C. Ouwehand, E. Isolauri, H. Hashimoto, Y. Benno, and S.
`Salminen. 2001. Comparison of mucosal adhesion and species identification
`of bifidobacteria isolated from healthy and allergic infants. FEMS Immunol.
`Med. Microbiol. 30:43–47.
`13. Hessle, C. C., B. Andersson, and A. E. Wold. 2005. Gram-positive and
`Gram-negative bacteria elicit different patterns of pro-inflammatory cyto-
`kines in human monocytes. Cytokine 30:311–318.
`14. Hoarau, C., C. Lagaraine, L. Martin, F. Velge-Roussel, and Y. Lebranchu.
`2006. Supernatant of Bifidobacterium breve induces dendritic cell matura-
`tion, activation, and survival through a Toll-like receptor 2 pathway. J.
`Allergy Clin. Immunol. 117:696–702.
`15. Holt, P. G., P. D. Sly, and B. Bjorksten. 1997. Atopic versus infectious
`diseases in childhood: a question of balance? Pediatr. Allergy Immunol.
`8:53–58.
`16. Janeway, C., P. Travers, M. Walport, and M. Shlomchik. 2007. Immunobi-
`ology: the immune system in health and disease, 8th ed. Garland Science
`Publishing, New York, NY.
`17. Karlsson, H., C. Hessle, and A. Rudin. 2002. Innate immune responses of
`human neonatal cells to bacteria from the normal gastrointestinal flora.
`Infect. Immun. 70:6688–6696.
`18. Kok, R. G., A. de Waal, F. Schut, G. W. Welling, G. Weenk, and K. J.
`Hellingwerf. 1996. Specific detection and analysis of a probiotic Bifidobac-
`terium strain in infant feces. Appl. Environ. Microbiol. 62:3668–3672.
`19. Lammers, K. M., P. Brigidi, B. Vitali, P. Gionchetti, F. Rizzello, E. Cara-
`melli, D. Matteuzzi, and M. Campieri. 2003. Immunomodulatory effects of
`probiotic bacteria DNA: IL-1 and IL-10 response in human peripheral blood
`mononuclear cells. FEMS Immunol. Med. Microbiol. 38:165–172.
`20. Livak, K. J., and T. D. Schmittgen. 2001. Analysis of relative gene expression
`data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method.
`Methods 25:402–408.
`21. Morita, H., F. He, T. Fuse, A. C. Ouwehand, H. Hashimoto, M. Hosoda, K.
`Mizumachi, and J. Kurisaki. 2002. Adhesion of lactic acid bacteria to caco-2
`cells and their effect on cytokine secretion. Microbiol. Immunol. 46:293–297.
`22. Mueller, S., K. Saunier, C. Hanisch, E. Norin, L. Alm, T. Midtvedt, A.
`Cresci, S. Silvi, C. Orpianesi, M. C. Verdenelli, T. Clavel, C. Koebnick, H. J.
`Zunft, J. Dore, and M. Blaut. 2006. Differences in fecal microbiota in dif-
`ferent European study populations in relation to age, gender, and country: a
`cross-sectional study. Appl. Environ. Microbiol. 72:1027–1033.
`23. Mullie´, C., M. F. Odou, E. Singer, M. B. Romond, and D. Izard. 2003.
`Multiplex PCR using 16S rRNA gene-targeted primers for the identification
`of bifidobacteria from human origin. FEMS Microbiol. Lett. 222:129–136.
`24. Nicaise, P., A. Gleizes, F. Forestier, A. M. Quero, and C. Labarre. 1993.
`Influence of intestinal bacterial flora on cytokine (IL-1, IL-6 and TNF-alpha)
`production by mouse peritoneal macrophages. Eur. Cytokine Netw. 4:133–
`138.
`25. Nicaise, P., A. Gleizes, C. Sandre, R. Kergot, H. Lebrec, F. Forestier, and C.
`Labarre. 1999. The intestinal microflora regulates cytokine production pos-
`itively in spleen-derived macrophages but negatively in bone marrow-derived
`macrophages. Eur. Cytokine Netw. 10:365–372.
`26. Ouwehand, A. C., E. Isolauri, F. He, H. Hashimoto, Y. Benno, and S.
`Salminen. 2001. Differences in Bifidobacterium flora composition in allergic
`and healthy infants. J. Alle

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