throbber
Early Human Development 86 (2010) 51–58
`
`Contents lists available at ScienceDirect
`
`Early Human Development
`
`j o u r n a l h o m e p a g e : w ww. e l s ev i e r. c o m / l o c a t e / e a r l h u md ev
`
`The role of intestinal bifidobacteria on immune system development in young rats
`Ping Dong, Yi Yang, Wei-ping Wang ⁎
`Children's Hospital, Pediatrics Department of Shanghai Medical College, Fudan University, Shanghai, PR China.
`
`a r t i c l e
`
`i n f o
`
`a b s t r a c t
`
`Article history:
`Received 16 September 2009
`Received in revised form 28 December 2009
`Accepted 4 January 2010
`
`Keywords:
`Bifidobacteria
`Immune-system development
`Intestine
`Rats
`
`Aim: The effects of intestinal bifidobacteria on the development of immunity in early life were explored.
`Methods: Neonatal SD rats born and housed under strict barrier systems were fed from birth with sufficient
`antibiotics (bifidobacteria minimisation group) or supplemented daily with 1×1010 colony-forming units of
`live Bifidobacterium longum (bifidobacteria supplementation group). Relevant indices of immune develop-
`ment were determined at one, three and six weeks old.
`Results: Compared to the control group, minimisation of the intestinal bifidobacteria delayed maturation of
`dendritic cells in Peyer's Patches and the development of T cells in the thymus, increased IL-4 secretion in the
`plasma, down-regulated IL-12, IL-10 mRNA and the interferon-γ/IL-4 mRNA ratio in intestinal mucosa,
`decreased interferon-γ mRNA in cultured peripheral blood mononuclear cells (PBMCs), and reduced
`immunoglobulin-M production in cultured PBMCs. Conversely, supplementation with bifidobacteria
`promoted dendritic cell maturation in Peyer's Patches, up-regulated IL-12, IL-10, interferon-γ mRNA and
`the interferon-γ/IL-4 ratio in intestinal mucosa, increased interferon-γ gene expression in cultured PBMCs,
`and raised immunoglobulin-M secretion in cultured PBMCs.
`Conclusions: Intestinal bifidobacteria could promote the maturation of dendritic cells and its expression of IL-
`12 locally in the gut, influence the development of T cells in the thymus, favour the development of T-helper
`cell type 1 response by increasing the local and systemic expression of interferon-γ and ensure the intestinal
`regulatory T cell response by promoting the local expression of IL-10. In addition, they enhance antibody
`synthesis by PBMCs, thereby affecting the development of both the gut and systemic immunity in early life.
`© 2010 Elsevier Ireland Ltd. All rights reserved.
`
`1. Introduction
`
`Comparative studies based on the germ-free animal have provided
`clear evidence that the body's cross-talk with commensal bacteria is
`essential for the development and maturation of the immune system
`in early life [1]. Because the gastrointestinal tract encompasses the
`largest surface of the human body, all kinds of colonising flora and the
`gut-associated lymphoid tissue (GALT) constitute the main elements
`that influence immune system development. Bifidobacteria, known as
`a kind of probiotic bacteria, represent up to 90% of the total intestinal
`flora in breast-fed infants [2]. Premature delivery, decreased breast-
`feeding, abuse of antibiotics and other factors have led to insufficient
`bifidobacteria colonisation of the gut in early infancy, which may be
`related to the increased future incidence of inflammatory, autoim-
`mune and atopic diseases [3]. Thus, the impact of bifidobacteria on the
`
`Abbreviations: DCs, dendritic cells; IFN, interferon; Ig, immunoglobulin; PPs, Peyer's
`Patches; PBMCs, peripheral blood mononuclear cells; Th1, T-helper cell type 1; Th2,
`T-helper cell type 2; Treg, regulatory T cell.
`⁎ Corresponding author. Children's Hospital, Pediatrics Department of Shanghai
`Medical College, Fudan University, 399 Wanyuan Road, Shanghai, 201102, PR China.
`Tel.: +86 21 64931990; fax: +86 21 65644937.
`E-mail address: wpwang@shmu.edu.cn (W. Wang).
`
`0378-3782/$ – see front matter © 2010 Elsevier Ireland Ltd. All rights reserved.
`doi:10.1016/j.earlhumdev.2010.01.002
`
`development of immunity and the precise mechanisms behind it
`require further elucidation.
`One principal character in immune system development in early
`life is the establishment of a balanced T-helper (Th) cell response
`(Th1/Th2). Th1 mainly secretes cytokines, such as interferon (IFN)-γ
`and IL-2, mediates cellular immunity and thus participates in anti-
`virus and intracellular bacterial infection responses. Th2 secretes
`cytokines, such as IL-4 and IL-5, mediates humoral immunity and is
`involved in the immune responses to bacterial infection, as well as
`immediate hypersensitivity reactions. An imbalanced Th1/Th2 may
`contribute, in part, to clinical diseases: e.g. excessive Th2 activity can
`lead to allergic disease, whereas Th1 predominance is found in
`inflammatory bowel disease. Adaptive responses of the newborn
`show a Th2 type and tend toward a Th1/Th2 balance as a result of the
`postnatal stimulation of initially colonising microbes on Th1 response.
`Additionally, the development of regulatory T (Treg) cell response
`is critical in inducing the tolerance to ‘self’ and in controlling in-
`flammatory diseases, including atopic disorders [4]. A retrospective
`study indicated that a preventive colonisation of high-risk infants with
`probiotics after birth decreased the incidence of allergies and repeated
`infections later in life [5], but its mechanism remains unclear.
`Dendritic cells (DCs), the most potent antigen-presenting cells
`(APCs), possess potent T-cell stimulatory capacities and are important
`in initiating the immune response [6]. Immature DCs reside in various
`
`Genome Ex. 1005
`Page 1 of 8
`
`

`

`52
`
`P. Dong et al. / Early Human Development 86 (2010) 51–58
`
`portals of the microorganism entry, such as skin and mucosa, where
`they guard sites of potential pathogen entry in to the host and capture
`invading microorganisms, and then migrate to nearby lymphoid
`organs, with gradual maturation. Mature DCs initiate a primary cell-
`mediated immune response through the expression of major
`histocompatibility complex (MHC) and costimulatory molecules.
`Moreover, DCs are critical in directing the differentiation of naive
`T cells into Th1, Th2 or Treg cells, thus, controlling the type, amplitude
`and intensity of the response [7]. The modulation of probiotics on
`T-cell responses may occur primarily through its action on the
`differentiation, maturation and functional activities of DCs. Recent in
`vitro studies have shown that certain probiotic interactions with DCs
`lead to the polarisation of naive T cells to a Th1 or Treg direction [8,9],
`however, to date, there are no relevant reports in vivo.
`The aim of the present study was to clarify how intestinal
`bifidobacteria affect the development of both the gut and systemic
`immunity in early life, especially its modulation on the differentiation,
`maturation and function of DCs, T-cell development, T-cell responses
`(Th1, Th2 and Treg) and immunoglobulin synthesis. Thus, the SD rats
`used in the study were fed from birth with sufficient antibiotics per
`day (bifidobacteria minimisation group) or administrated a daily dose
`of Bifidobacterium longum (bifidobacteria supplementation group)
`until one, three and six weeks of age, which are equivalent to the
`neonatal, infant and adolescent period of childhood, respectively.
`Various immunological parameters were assessed to study the impact
`of both bifidobacteria minimisation and supplementation on immune
`development.
`It is expected that the research will provide an
`experimental basis for the proper usage of bifidobacteria to improve
`the immunity of children in reality.
`
`2. Materials and methods
`
`2.1. Animals
`
`All experimental procedures involving animals were approved by the
`Ethics Committee of Shanghai Medical Collage of Fudan University and
`the care of animals was in accordance with institution guidelines.
`Newborn specific pathogen-free Sprague–Dawley (SD) rats were used.
`The rat pups were reared in plastic cages in a room kept at 22±3 °C and
`55±5% humidity, on a 12 h light/dark cycle under strict barrier systems
`with the water, diet and bedding material sterilized and high level air
`purity (Animal Centre of Public Health, Clinical Centre Affiliated to Fudan
`University). They were breast fed first and subsequently consume diet
`and water freely throughout the experimental period. The pups were
`divided into three groups, bifidobacteria minimisation (BM) group was
`fed from birth with gentamicin sulphate injection (100 mg/kg) and
`metronidazole injection (80 mg/kg) per day to minimise the amount of
`resident bifidobacteria in intestinal tract, while bifidobacteria supple-
`mentation (BS) group supplemented from birth with live B. longum
`(BL-11) 1×1010 colony-forming units (CFU) per day. The BL-11 (kindly
`provided by George Food Industries, Shanghai, China) were supplied as
`freeze-dried powder in sealed packets containing about 1.5×1011 CFU/g
`and stored at a temperature of −20 °C until dissolved in warm water for
`use. Both antibiotic injections and BL-11 suspension were given to the
`rats through orogastric gavage, which performed in a clean bench placed
`in the room. A third group of non-intervened rats was used as control. Ten
`rats of each group were anaesthetised by intra-peritoneal injection of 7%
`chloral hydrate (5 ml/kg) at one, three and six weeks old respectively.
`Blood was collected from the carotid artery into a heparinised tube, and
`the thymus, spleen and the whole length of the small intestine were
`removed and placed in sterile Petri dishes containing medium.
`
`2.2. Faecal sample preparation and bifidobacteria quantification
`
`Individual rat faeces were collected before rats were sacrificed and
`stored at −70 °C until used. After thawing all of the faecal sample, the
`
`preparation of bacterial DNA were conducted as described by Wang
`RF [10]. An amount of 0.5 g of faeces was added to 9 ml of sterile
`phosphate-buffered saline (PBS, 0.05 M, pH 7.4) and mixed well by
`inverting and surging the tube for 5 to 10 min, then centrifuged at low
`speed (1440 rpm) for 5 min to collect the upper phases; this was
`repeated three times. The upper phases were then centrifuged at high
`speed (9200 rpm) for 3 min to collect the bacterial cells in the pellets.
`After washing four times with PBS and once with distilled water, the
`cells in the pellets were resuspended with 50 µl distilled water and
`50 µl 1% Triton X-100, boiled at 100 °C for 5 min, and immediately
`cooled down in ice water. The bifidobacteria in pure cultures with an
`accurate quantity of 5 ×1010 CFU/ml were directly centrifuged at
`9200 rpm for 3 min and then were washed twice with PBS and once
`with water, suspended, serially diluted, boiled, and cooled in ice water
`used as standard. The primer set was designed from the 16S rRNA
`gene sequence available from GenBank: GGGTGGTAATGCCGGATG
`(P1); CCACCGTTACA CCGGGAA (P2), with the product length 517
`base pair. Two microlitres of each sample was directly added to 48 µl
`of PCR mixture containing 25 µl of Real time PCR Master Mix
`(TOYOBO,
`Japan) and 0.2 µM of each primer. The amplifying
`conditions were: 95 °C for the first 20 min, then 95 °C for 30 s, 60 °C
`for 30 s and 72 °C for 40 s, for 40 cycles. The amount of bifidobacteria
`in the unknown samples was determined by interpolating the
`threshold cycle values to the standard curve, and the results were
`expressed as log10 CFU/g faeces.
`
`2.3. Preparation of single-cell suspensions
`
`The cellular suspensions of thymus and spleen were prepared by
`chopping the tissues into small pieces with a sterile scissor and then
`grinding mechanically with a 100 mesh stainless steel sieve. The
`resulting suspensions were then transferred to a tube and centrifuged
`at 1500 rpm for 5 min to collect the cell pellets. After washing twice
`with Hanks, the cells were resuspended and adjusted to a final
`concentration of 1×106 cells/ml in Hanks. To prepare the cellular
`suspension of Peyer's Patches (PPs), all PPs were carefully excised
`from the whole length of the small intestine using a curved scissor,
`chopped and then ground mechanically with a 200 mesh stainless
`steel sieve. The resulting suspensions were handled in the same way
`as that of the thymus and spleen.
`
`2.4. Fluorescence staining and flow cytometry analysis
`
`By using direct immunofluorescence, the development of T cells
`(immature CD3+CD4+CD8+ and mature CD3+CD4+ or CD3+
`CD8+ T cell) in thymus, the proportion of T-cell subsets (CD3+CD4+
`helper T cell, CD3+CD8+ cytotoxic T cell, and CD4+CD25+
`regulatory T cell, CD161+ natural killer cell), the number and
`maturation of DCs (OX62+ cell [11] and fluorescence intensity of
`CD86) and B cells (CD45RA+ cell [12] and fluorescence intensity of
`immunoglobulin (Ig)M heavy chain (mIgM)) in peripheral blood,
`spleen and PPs were determined. For the staining of thymus, spleen
`and PPs cells, 1 × 105 cells/tube were incubated with antibodies for
`20 min, rinsed with PBS, centrifuged at 1000 rpm for 5 min, and then
`resuspended in PBS. For blood studies, 100 µl whole blood was
`incubated with antibodies for 20 min. The blood was then lysed with
`FACS Lysing Solution (BD Biosciences, USA), rinsed with PBS,
`centrifuged at 1000 rpm for 5 min, and resuspended in PBS. Analysis
`was done with a FACSCalibur flow cytometer (BD Biosciences). The
`following mouse monoclonal antibodies were used: FITC anti-CD3
`(1 µg/106cells), PE anti-CD4 (0.25 µg/106cells), APC anti-CD8 (0.5 µg/
`106cells), FITC anti-CD25 (1 µg/106cells) and PE anti-CD45RA (1 µg/
`106cells) (all from Biolegend, San Diego, CA), PE anti-OX62 (10 µl/
`tube), FITC anti-CD86 (10 µl/tube), PE anti-CD161 (10 µl/tube) and
`FITC anti-mIgM (4 µl/tube) (all from Serotec, UK). Corresponding
`isotype-matched mAbs were used as controls.
`
`Genome Ex. 1005
`Page 2 of 8
`
`

`

`P. Dong et al. / Early Human Development 86 (2010) 51–58
`
`53
`
`2.5. PBMCs cultured in vitro
`
`Peripheral blood mononuclear cells (PBMCs) were isolated from
`heparinised diluted blood by density gradient centrifugation over
`Ficoll–Hypaque gradients (SABC, Shanghai, China). After being
`washed twice with D-Hanks, the cells were resuspended at a
`concentration of 1 × 106 cells/ml
`in RPMI1640 culture medium
`(GIBCO, USA) supplemented with 10% heat-inactivated neonatal calf
`serum (Sijiqing, Hangzhou, China), 100 U/ml penicillin and 100 µg/ml
`streptomycin. The cell suspension was then incubated in a round
`bottom and ninety-six-well culture plate (Falcon, USA) at
`1 × 105 cells/well with the stimulation of phytohaemagglutinin
`(PHA, 5 µg/ml, Sigma, USA) or Staphylococcus aureus cells (SAC,
`0.0056% w/v, Calbiochem, USA). The plates were placed at 37 °C in an
`environment of 5% CO2. After being incubated with PHA for 24 h and
`48 h, the cells were harvested, mixed well with TRIzol (Invitrogen,
`USA) and stored at −70 °C until analysis of cytokine expression by
`RT-PCR. Cell-free supernatants were obtained after the PBMCs were
`incubated with SAC for 5 d and stored at −70 °C until assayed for
`antibody production by ELISA.
`
`2.6. RNA isolation and reverse transcription
`
`To investigate the mRNA levels of IL-12 (mainly secreted by DCs),
`Th1/Th2 (IFN-γ/ IL-4) and regulatory (IL-10, Foxp3) cytokines in
`intestinal mucosa, the mucosa of the small intestine was scraped and
`then put in the mortar and ground quickly with liquid nitrogen. Total
`RNA was isolated using TRIzol (Invitrogen) according to the
`manufacturer's instructions. For the gene expression of these
`cytokines in PHA-stimulated PBMCs, total RNA was isolated from
`the harvested cells, which had been transferred to TRIzol in the same
`way. The RNA concentration was measured by absorbance at 260 nm
`in a spectrophotometer (Shimadzu UV-1601, Japan). Two micrograms
`of total RNA was then reverse transcribed into cDNA with moloney
`murine leukaemia virus reverse transcriptase (Promega, USA)
`according to the manufacturer's instructions. After reverse transcrip-
`tion, the 25 µl reaction was diluted to 125 µl so that 2.5 µl of RT
`product added to a PCR mixture would be equivalent to the reverse
`transcription of 40 ng total RNA.
`
`2.7. Real-time quantitative PCR
`
`SYBR Green dye and Mastercycler ep realplex4 (Eppendorf,
`Germany) were used for Real-Time PCR. The sequence of primers,
`annealing temperature and PCR product size are described in Table 1.
`Quantitative PCR was carried out on a 25 µl final volume: 12.5 µl of
`Real time PCR Master Mix (TOYOBO), 0.2 µM of each primer and 2.5 µl
`of cDNA. The amplifying conditions were: 95 °C for the first 20 min,
`then 95 °C for 30 s, 54–60 °C (Table 1) for 30 s, and 72 °C for 40 s, for
`
`Table 1
`Primers, annealing temperatures and product sizes of PCR for β-actin and cytokine gene
`detection.
`
`Genes
`β-actin
`
`IL-12
`
`IFN-γ
`
`IL-4
`
`IL-10
`
`Foxp3
`
`Primers (5′ →3′)
`
`Ta (°C)
`
`Product (bp)
`
`P1: AAGATCCTGACCGAGCGTGG
`P2: CAGCACTGTGTTGGCATAGAGG
`P1: TCAGGGACATCATCAAACCG
`P2: ACGCACCTTTCTGGTTACACTC
`P1: GAAAGACAACCAGGCCATCAG
`P2: TCATGAATGCATCCTTTTTTGC
`P1: CCACGGAGAACGAGCTCATC
`P2: GAGAACCCCAGACTTGTTCTTCA
`P1: GCCAAGCCTTGTCAGAAATGA
`P2: TCCCAGGGAATTCAAATGCT
`P1: TTCACCTATGCCACCCTC
`P2: CACTGCTCCCTTCTCACTC
`
`58
`
`58
`
`54
`
`60
`
`56
`
`57
`
`320
`
`201
`
`101
`
`101
`
`101
`
`198
`
`P1: sense primer; P2: antisense primer; bp: base pair.
`
`40 cycles. β-actin was used as quality control. Experiments were
`performed in duplicate to ensure the reproducibility of the technique.
`Amplification specificity was checked using a melting curve following
`the manufacturer's instructions. The relative number of molecules of
`each transcript was determined by interpolating the threshold cycle
`values of the unknown samples to each standard curve.
`
`2.8. Enzyme-linked immunosorbent assay
`
`To determine the basal concentration of cytokines (IL-12, IFN-γ,
`IL-4 and IL-10) and immunoglobulin (IgG, IgM, IgA and IgE) in plasma,
`the plasma samples were prepared by having the heparinised blood
`coagulated naturally at room temperature for 15 min and then
`centrifuged at 2000 rpm for 10 min to collect the upper phases. The
`sample quantification was conducted using a double antibody
`sandwich ELISA with the kits (for cytokines, Jingmei, Beijing, China;
`for immunoglobulin,
`ICL, USA) according to the manufacturer's
`instructions. The level of IgG and IgM in cell-free culture supernatants
`was measured in the same way to investigate the ability of
`immunoglobulin secretion by SAC-stimulated PBMCs. The limits of
`detection of these ELISA kits are as follows: IL-12, 4 pg/ml; IFN-γ,
`7 pg/ml; IL-4, 7 pg/ml; IL-10, 6 pg/ml; IgG, IgM and IgA, 5 ng/ml; and
`IgE, 0.2 ng/ml.
`
`2.9. Statistical analyses
`
`Data are expressed as means and standard deviations (SD). One-
`way ANOVA was applied to examine group differences, with further
`multiple comparisons using a Bonferroni test (Stata ver. 7.0, Stata
`Corp., USA). A p-value of less than 0.05 was considered statistically
`significant.
`
`3. Results
`
`3.1. The amount of bifidobacteria in faecal samples
`
`To evaluate the actual results of bifidobacteria minimisation in the
`BM group, as well as the extra colonisation of it in the BS group, the
`amount of bifidobacteria in faecal samples was detected by RT-
`fluorescence quantitative PCR. The results showed that, as compared
`with the control of the same age, the resident bifidobacteria in the BM
`group decreased by three orders of magnitude at the end of three
`weeks and decreased by four orders of magnitude at the end of six
`weeks, whereas that in the BS group increased by four orders of
`magnitude at the end of three weeks and maintained this level to the
`end of six weeks (n=10, p<0.001 for all, Bonferroni test) (Fig. 1). In
`addition, bifidobacteria minimisation or supplementation didn't
`result in any body weight or organ (the thymus, spleen and PPs)
`weight difference at any age (data not shown), as well as the organ
`gross observation, except that at the end of six weeks, the intestine of
`rats in the BM group had excessive gas and mucus (especially the
`cecum).
`
`3.2. The modulation of bifidobacteria on the differentiation, maturation
`and function of DCs
`
`To determine the modulation of intestinal bifidobacteria on DCs,
`the number and maturation of DCs in the peripheral blood, spleen and
`PPs, the gene expression of IL-12 (mainly secreted by DCs) in
`intestinal mucosa and cultured PBMCs, as well as the secretion of it in
`plasma were determined by flow cytometry, RT-PCR and ELISA,
`respectively. For the level of surface marker CD86 (co-stimulatory
`molecule) expression increases as DCs mature gradually and thus
`reflects DC developmental stage [13]. It was found that in PPs and at
`six weeks, as compared with the control (19.17 (2.32)), the Geo Mean
`(SD) fluorescence intensity of CD86 in the BM group decreased
`
`Genome Ex. 1005
`Page 3 of 8
`
`

`

`54
`
`P. Dong et al. / Early Human Development 86 (2010) 51–58
`
`CD8+ cytotoxic T cell, and CD4+CD25+ regulatory T cell, CD161+
`natural killer cell) in the peripheral blood, spleen and PPs was also
`studied by flow cytometry, and none of the T-cell subsets differed
`significantly among the three groups in all areas (Table 2).
`
`3.4. The impact of bifidobacteria on the T-helper cell response
`
`To determine whether intestinal bifidobacteria affect the balance
`of Th1 and Th2, the mRNA levels of Th1/Th2 (IFN-γ/ IL-4) cytokines in
`intestinal mucosa and cultured PBMCs, as well as the secretion of
`them in plasma were measured with RT-PCR and ELISA respectively.
`The results showed that, as compared with the control, the BM group
`had an increased protein level of IL-4 in plasma at six weeks (n=10,
`p=0.006, Bonferroni test), but a decreased IFN-γ/IL-4 mRNA ratio in
`intestinal mucosa both at three weeks and six weeks (n = 10,
`p =0.011, p = 0.015 respectively, Bonferroni test), as well as a
`decreased IFN-γ gene transcript in cultured PBMCs at six weeks
`(n=10, p=0.034, Bonferroni test). The BS group, however, had up-
`regulated IFN-γ mRNA and IFN-γ/IL-4 ratio in intestinal mucosa both
`at three weeks and six weeks (n=10, p<0.001 for all, Bonferroni
`test), as well as increased IFN-γ gene expression in cultured PBMCs at
`six weeks (n=10, p =0.035, Bonferroni test) (Fig. 3).
`
`3.5. The modulation of bifidobacteria on the regulatory T cell response
`
`To further evaluate the modulation of intestinal bifidobacteria on
`the Treg cell response, the number of CD4+CD25+ Treg cells in the
`peripheral blood, spleen and PPs, the expression of regulatory (IL-10,
`Foxp3) cytokines in intestinal mucosa and cultured PBMCs, as well as
`IL-10 secretion in plasma were determined by flow cytometry, RT-PCR
`and ELISA, respectively. We found that at both three weeks and six
`weeks, the BM group had a decreased expression of IL-10 mRNA in the
`intestinal mucosa (n=10, p=0.022, p =0.012 respectively, Bonfer-
`roni test), whereas the BS group had an increased expression (n =10,
`p=0.015, p=0.011 respectively, Bonferroni test), as compared with
`the control (Fig. 4). Expression of IL-10 outside the gut, Foxp3 gene
`expression (data not shown) and the percentage of CD4+CD25+ Treg
`cells (as has been noted) in all areas showed no significant group
`differences.
`
`3.6. The effect of bifidobacteria on immunoglobulin synthesis
`
`To investigate the effect of intestinal bifidobacteria on immuno-
`globulin secretion, the number and maturation of B cells in the
`peripheral blood, spleen and PPs, the basal level of immunoglobulin
`(IgG, IgM, IgA and IgE) in plasma and the production of IgG and IgM by
`SAC-stimulated PBMCs were determined by flow cytometry and
`ELISA, respectively. We found that at six weeks, after PBMCs were
`cultured with SAC for 5d, the BM group had a decreased production of
`IgM (299.91 (100.03) ng/ml), whereas the BS group had an increased
`production of it (749.58 (85.58) ng/ml), as compared with the control
`(507.16 (89.8) ng/ml) (p<0.001, one-way ANOVA; p =0.012 for the
`BM, p=0.009 for the BS, Bonferroni test) (n=10). The number and
`maturation of B cells (the percentage of CD45RA + cells and
`fluorescence intensity of mIgM) in all areas showed no significant
`group differences (Table 2); this was also the case for the level of
`immunoglobulin in plasma (data not shown).
`
`4. Discussion
`
`Early colonising flora of the gut is crucial in the development of the
`body's immune system. Bifidobacteria, as the predominant bacteria in
`the gut during the early period of life, could play a major role in this
`process. In our present study, feeding the neonatal rats daily with
`sufficient antibiotics and keeping them in a strict barrier environment
`effectively minimised the bifidobacteria in the gut; conversely, B.
`
`Fig. 1. Amount of bifidobacteria in faecal samples, measured by RT-fluorescence
`quantitative PCR at the end of one, three and six weeks, showing the effective
`bifidobacteria minimisation in the BM group and bifidobacteria supplementation in the
`BS group. ‡p values were obtained using one-way ANOVA to examine group differences,
`asterisks indicate statistical difference vs. control (**p <0.01) through the further
`multiple comparisons using a Bonferroni test (n=10).
`
`significantly (16.89 (1.77)), whereas that in the BS group increased
`significantly (22.33 (2.09)) (p =0.013, One-way ANOVA; p =0.024
`for the BM, p=0.015 for the BS, Bonferroni test) (n=10). The results
`of RT-PCR showed that the BS group had an increased expression of IL-
`12 mRNA in the intestinal mucosa, both at three weeks and six weeks
`(n=10, p =0.039, p<0.001 respectively, Bonferroni test); whereas
`the BM group had a decreased expression of it at six weeks (n=10,
`p < 0.001, Bonferroni test) (Fig. 2). DC maturation and IL-12
`expression outside the gut showed no significant group differences,
`as well as DC number (the percentage of OX62+ cells) in all areas
`(data not shown).
`
`3.3. The effect of bifidobacteria on the development of T cells in the
`thymus
`
`Using flow cytometry, we studied the effect of intestinal bifido-
`bacteria on the development of T cells in the thymus. We found that in
`the thymus, the immature CD4+CD8+ T cell percentage in the BM
`group was significantly higher than that in the control group, both at
`three weeks and six weeks (n =10, p=0.043, p=0.022 respectively,
`Bonferroni test), but the mature CD4+ or CD8+ T cell percentage was
`markedly lower than that in the control group at three weeks (n =10,
`p=0.036, p =0.031 respectively, Bonferroni test). The BS group was
`not significantly different than the control however. In addition, the
`proportion of each T-cell subset (CD3+CD4+ helper T cell, CD3+
`
`Fig. 2. Effect of intestinal bifidobacteria minimisation (the BM group) or supplemen-
`tation (the BS group) on the expression of IL-12 mRNA in the intestinal mucosa.
`‡p values were obtained using one-way ANOVA to examine group differences, asterisks
`indicate statistical difference vs. control (*p <0.05, **p < 0.01) through the further
`multiple comparisons using a Bonferroni test (n=10).
`
`Genome Ex. 1005
`Page 4 of 8
`
`

`

`Table 2
`Effects of intestinal bifidobacteria on T-cell development in thymus, proportion of each lymphocyte subset and B-cell maturation in peripheral blood, spleen and PPs.
`
`P. Dong et al. / Early Human Development 86 (2010) 51–58
`
`55
`
`dCD3+CD4+CD8+
`1W
`3W
`6W
`dCD3+CD4+
`1W
`3W
`6W
`dCD3+CD8+
`1W
`3W
`6W
`dCD4+CD25+
`1W
`3W
`6W
`dCD161+
`1W
`3W
`6W
`dCD45RA+
`1W
`3W
`6W
`emIgM+
`1W
`3W
`6W
`
`Control
`
`65.13(10.09)
`49.25(6.13)
`30.09(5.06)
`
`8.21(1.12)
`11.46(2.30)
`12.27(2.17)
`
`4.65(0.24)
`9.44(0.76)
`6.76(0.52)
`
`Thymus
`
`aBM
`
`65.31(9.76)
`55.21(7.20)*
`44.31(5.98)*
`
`7.89(0.98)
`8.62(1.51)*
`11.49(1.25)
`
`4.52(0.19)
`6.33(0.51)*
`5.98(0.45)
`
`bBS
`
`64.98(9.55)
`48.81(5.21)
`31.21(4.03)
`
`8.05(1.10)
`11.95(1.76)
`12.12(1.34)
`
`4.31(0.12)
`9.19(0.58)
`6.13(0.60)
`
`–
`–
`–
`
`–
`–
`–
`
`–
`–
`–
`
`–
`–
`–
`
`–
`–
`–
`
`–
`–
`–
`
`–
`–
`–
`
`–
`–
`–
`
`–
`–
`–
`
`–
`–
`–
`
`–
`–
`–
`
`–
`–
`–
`
`cp
`
`0.998
`0.039
`0.012
`
`0.896
`0.043
`0.747
`
`0.841
`0.046
`0.354
`
`–
`–
`–
`
`–
`–
`–
`
`–
`–
`–
`
`–
`–
`–
`
`Control
`
`7.74(1.44)
`2.40(0.71)
`1.70(0.36)
`
`19.84(2.65)
`29.35(5.38)
`44.55(4.23)
`
`7.66(1.44)
`11.73(2.11)
`22.55(3.35)
`
`1.47(0.11)
`1.97(0.21)
`2.30(0.23)
`
`2.08(0.31)
`7.66(1.11)
`5.29(0.48)
`
`17.61(0.93)
`21.82(1.39)
`18.16(2.21)
`
`24.94(3.11)
`17.96(2.61)
`22.47(2.01)
`
`Blood
`
`BM
`
`8.67(2.78)
`2.35(1.13)
`1.98(0.45)
`
`21.13(1.98)
`27.11(6.13)
`42.99(3.29)
`
`6.31(1.05)
`12.47(1.98)
`24.07(4.84)
`
`1.22(0.18)
`2.16(0.15)
`2.65(0.25)
`
`1.79(0.54)
`8.25(0.98)
`5.57(0.55)
`
`15.39(1.12)
`21.16(2.27)
`19.55(3.43)
`
`25.21(2.60)
`17.77(3.58)
`20.35(2.33)
`
`BS
`
`8.09(2.30)
`2.88(0.52)
`1.61(0.28)
`
`20.05(3.14)
`26.45(4.99)
`46.31(3.28)
`
`6.06(2.33)
`14.55(2.31)
`24.10(4.29)
`
`1.28(0.21)
`2.03(0.32)
`2.57(0.18)
`
`1.76(0.37)
`8.31(1.40)
`6.02(0.56)
`
`15.79(2.04)
`20.88(2.41)
`16.64(3.07)
`
`23.39(3.07)
`16.49(3.12)
`20.68(3.14)
`
`p
`
`0.776
`0.584
`0.189
`
`0.581
`0.069
`0.055
`
`0.772
`0.084
`0.163
`
`0.065
`0.282
`0.059
`
`0.069
`0.233
`0.340
`
`0.081
`0.454
`0.052
`
`0.399
`0.652
`0.113
`
`CD3+CD4+CD8+
`1W
`3W
`6W
`CD3+CD4+
`1W
`3W
`6W
`CD3+CD8+
`1W
`3W
`6W
`CD4+CD25+
`1W
`3W
`6W
`CD161+
`1W
`3W
`6W
`CD45RA+
`1W
`3W
`6W
`mIgM+
`1W
`3W
`6W
`
`Control
`
`4.11(0.31)
`3.69(0.37)
`2.02(0.48)
`
`21.94(5.39)
`22.78(4.55)
`20.86(4.08)
`
`13.45(1.57)
`11.31(1.79)
`10.12(2.41)
`
`1.88(0.14)
`1.93(0.17)
`2.39(0.18)
`
`7.42(0.95)
`10.21(0.78)
`8.45(1.24)
`
`37.41(6.28)
`43.51(5.87)
`30.39(5.04)
`
`22.96(3.05)
`31.25(2.67)
`41.03(2.23)
`
`Spleen
`
`BM
`
`4.09(0.45)
`4.06(0.87)
`1.19(1.13)
`
`18.19(4.27)
`24.23(5.17)
`18.06(5.33)
`
`10.53(2.41)
`9.36(1.50)
`7.19(3.68)
`
`1.71(0.21)
`2.11(0.22)
`2.43(0.25)
`
`5.99(2.11)
`10.06(1.03)
`6.39(2.09)
`
`39.15(7.13)
`39.11(6.93)
`30.06(3.98)
`
`19.58(3.66)
`34.29(3.30)
`40.22(3.38)
`
`BS
`
`p
`
`Control
`
`3.75(0.39)
`3.99(0.55)
`1.61(0.85)
`
`19.82(3.66)
`25.01(4.44)
`17.87(5.51)
`
`13.07(3.42)
`10.55(2.30)
`9.48(2.19)
`
`1.77(0.13)
`1.85(0.19)
`2.16(0.20)
`
`5.92(1.72)
`11.05(0.67)
`6.88(1.56)
`
`40.01(6.03)
`42.55(7.02)
`33.19(3.71)
`
`23.11(4.05)
`30.97(4.15)
`41.18(4.06)
`
`0.072
`0.060
`0.225
`
`0.060
`0.082
`0.224
`
`0.115
`0.644
`0.098
`
`0.150
`0.373
`0.551
`
`0.068
`0.197
`0.058
`
`0.062
`0.055
`0.266
`
`0.077
`0.064
`0.549
`
`0.99(0.12)
`1.17(0.14)
`1.58(0.19)
`
`18.13(1.35)
`20.38(4.02)
`21.29(2.87)
`
`5.56(1.94)
`5.37(1.39)
`7.43(1.82)
`
`5.36(0.18)
`6.02(0.19)
`8.47(1.35)
`
`1.99(0.12)
`2.94(0.41)
`2.77(0.37)
`
`53.39(11.45)
`60.01(16.13)
`57.62(14.61)
`
`12.12(3.31)
`19.37(2.60)
`19.19(4.05)
`
`PPs
`
`BM
`
`0.98(0.24)
`1.26(0.05)
`1.39(0.22)
`
`16.76(2.21)
`19.61(2.19)
`19.59(2.95)
`
`4.11(1.19)
`5.29(1.43)
`5.78(1.31)
`
`6.03(1.12)
`5.79(0.34)
`7.11(0.79)
`
`1.64(0.11)
`3.03(0.58)
`3.49(0.71)
`
`49.75(15.42)
`64.12(15.52)
`54.19(10.09)
`
`15.13(4.54)
`22.27(3.79)
`17.81(3.99)
`
`BS
`
`1.23(0.25)
`1.22(0.18)
`1.56(0.27)
`
`15.48(1.39)
`21.11(4.67)
`22.18(2.06)
`
`6.31(1.71)
`6.56(2.42)
`7.76(2.07)
`
`5.93(0.77)
`6.11(0.25)
`9.09(1.16)
`
`1.73(0.19)
`3.43(0.44)
`2.84(0.62)
`
`51.11(13.39)
`59.98(11.31)
`56.05(13.36)
`
`13.93(5.16)
`20.11(4.12)
`20.15(5.63)
`
`p
`
`0.255
`0.774
`0.069
`
`0.079
`0.661
`0.177
`
`0.064
`0.113
`0.472
`
`0.054
`0.344
`0.092
`
`0.221
`0.057
`0.121
`
`0.077
`0.060
`0.112
`
`0.227
`0.545
`0.066
`
`Asterisks indicate statistical difference vs. control (*p <0.05) through the further multiple comparisons using a Bonferroni test, n=10.
`a : the bifidobacteria minimisation group (BM).
`b : the bifidobacteria supplementation group (BS).
`c : p values were obtained using one-way ANOVA to examine group differences.
`d : data were expressed as the percentage of positive cells with respect to total lymphocytes (mean (SD)).
`e : data were expressed as the fluorescence intensity of mIgM in CD45RA+ cells (mean (SD)).
`
`longum (a bifidobacterial species that is widely used in infant formula
`and probiotic functional foods) successfully colonised the gut after the
`daily supplementation of 1× 1010 CFU from birth. Although none of
`the immunological parameters observed changed at the end of one
`
`week (the neonatal period), several aspects of the immunity were
`affected by both bifidobacteria minimisation and supplementation at
`the end of three weeks (the infant period), with the most obvious
`change at six weeks of age (the adolescent period). These suggest that
`
`Genome Ex. 1005
`Page 5 of 8
`
`

`

`56
`
`P. Dong et al. / Early Human Development 86 (2010) 51–58
`
`Fig. 3. Effect of intestinal bifidobacteria minimisation (the BM group) or supplementation (the BS group) on Th1/Th2 (IFN-γ/ IL-4) cytokines profile. (A) Protein level of IL-4 in the
`plasma; (B) expression of IFN-γ mRNA in the intestinal mucosa; (C) IFN-γ/IL-4 mRNA ratio in the intestinal mucosa; (D) expression of IFN-γ mRNA in phytohaemagglutinin (PHA)-
`stimulated PBMCs. ‡p values were obtained using one-way ANOVA to examine group differences, asterisks indicate statistical difference vs. control (*p < 0.05, **p< 0.01) through the
`further multiple comparisons using a Bonferroni test (n=10).
`
`intestinal bifidobacteria minimisation or extra
`the influence of
`colonisation on immune development may become apparent only if
`this kind of change in bifidobacteria persists long enough in the gut.
`DCs are the gatekeepers of immune response and are the principal
`stimulators of naive T cells, a property that distinguishes them from
`all other APCs [7]. While a number of
`in vitro studies have
`demonstrated that DCs are the target cells of a variety of probiotic
`strains [8,9,14–16], to date, few studies have been conducted in vivo.
`One of the main purposes of our study w

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