Sei sulla pagina 1di 12

Nephrol Dial Transplant (2016) 31: 401–412

doi: 10.1093/ndt/gfv353
Advance Access publication 20 October 2015

Gut Lactobacillus protects against the progression of renal damage


by modulating the gut environment in rats

Ayumi Yoshifuji, Shu Wakino, Junichiro Irie, Takaya Tajima, Kazuhiro Hasegawa, Takeshi Kanda,

Downloaded from http://ndt.oxfordjournals.org/ at Serials Dept / Cornell University Medical Library on September 30, 2016
Hirobumi Tokuyama, Koichi Hayashi and Hiroshi Itoh
Department of Internal Medicine, School of Medicine, Keio University, Tokyo, Japan

Correspondence and offprint requests to: Shu Wakino; E-mail: shuwakino@z8.keio.jp

Keywords: chronic kidney disease, gut microbiota, protein


A B S T R AC T bound uremic retention solutes, tight junction, Toll-like
receptor
Background. The role of gut microbiota in the progression of
chronic kidney disease (CKD) has not been fully elucidated.

ORIGINAL ARTICLE
Methods. Renal failure was induced in 6-week-old spontan-
eously hypertensive rats by 5/6 nephrectomy (Nx). We analyzed INTRODUCTION
the gut microbiota population to identify the relevant species
Chronic kidney disease (CKD) is a worldwide health problem
potentially involved in inducing renal damage. Human colon
that can lead to cardiovascular disease and end-stage kidney
Caco-2 cells were used to delineate the mechanism involved
disease that requires renal replacement therapy. Although its
in the molecular changes in the gut of Nx rats.
pathogenesis and pathophysiology have not been fully eluci-
Results. Nx rats showed an increase in Bacteroides (Bact) and a
dated, the progression of CKD is accompanied by the accumu-
decrease in Lactobacillus (Lact) species compared with sham-
lation of uremic substances that have deleterious effects. For
operated rats. Lact, but not Bact, populations were significantly
example, the protein-bound uremic solutes indoxyl sulfate
associated with urinary protein excretion. Treatment of Nx rats
(IS) and p-cresyl sulfate (PCS), which are normally excreted
with 1 × 1010 CFU/kg/day Lact ameliorated increased urinary
into the urine by the kidneys, accumulate in patients with
protein excretion and higher serum levels of the uremic toxins,
CKD. These substances have been shown to cause damage to
indoxyl sulfate and p-cresyl sulfate, and serum urea nitrogen le-
renal tubular cells or endothelial cells, as well as to other cells,
vels. Lact also attenuated systemic inflammation in Nx rats, as
by increasing cellular oxidative stress [1]. In an in vivo study
evaluated by serum lipopolysaccharide, interleukin-6 and C-
where these substances were administered exogenously, they in-
reactive protein levels. Histologically, renal sclerosis in Nx rats
duced damage to renal or cardiovascular tissues by enhancing
was restored by Lact treatment. A reduction in the expression of
oxidative stress, inflammatory responses or fibrotic changes [2–
tight junction proteins and the Toll-like receptor 2 (TLR2), a
4]. A recent study also demonstrated that another uremic sol-
putative Lact receptor, in the colons of Nx rats were mitigated
ute, indole acetic acid (IAA), caused endothelial cell damage.
by Lact. Treatment of Caco-2 cells with indole downregulated
In addition, IAA serum levels correlated with cardiovascular
tight junction protein expression, which was abolished by ex-
events in CKD patients, suggesting that it may be a cause of car-
posure to Lact. The effects of Lact were reversed by treatment
diovascular complications in CKD [5]. Since the precursors of
with OxPAPC, a TLR inhibitor. Similarly, the increase in the
these substances, indoles or cresols, are produced by bacteria in
permeability of the Caco-2 cell monolayer was reversed by
the intestine from the amino acid tryptophan or tyrosine, re-
the administration of Lact. Lact upregulated TLR2 expression
spectively, and are subsequently absorbed from the colon, the
in Caco-2 cells. Lact also attenuated the increase in serum in-
changes in the influx of these uremic solutes from the intestine
doxyl sulfate and urea levels and urinary protein excretion in
may affect renal function [6]. This kind of interrelationship
Nx rats even in the pseudogerm-free environment.
between the gut and kidney is called ‘the intestinal-renal syn-
Conclusions. Lact supplementation mitigated the systemic in-
drome’[7], and the modulation of this axis, including regulation
flammation and proteinuria associated with renal failure, sug-
of the colon environment or colon barrier, may be a plausible
gesting that in the gut microbiota, Lact plays a protective role
strategy to slow the progression of CKD. A decreased expres-
against the progression of CKD.
sion of tight junction proteins in the colon has been observed

© The Author 2015. Published by Oxford University Press 401


on behalf of ERA-EDTA. All rights reserved.
in CKD, and stabilization of the intestinal barrier by changing levels of interleukin-6 (IL-6) and C-reactive protein (CRP)
the colon environment, using a charcoal absorbent for indole, were measured using commercial ELISA kits (Abcam Japan,
was found to improve the associated endotoxemia, oxidative Tokyo, Japan). At sacrifice, tissue samples of the kidney and as-
stress and inflammatory response [8]. cending colon were obtained, snap frozen and used for subse-
Another factor that affects the intestinal environment is quent experiments. All experiments were performed in
changes in the intestinal microbiota, which have been shown to accordance with the animal experiment guidelines of the Keio
correlate with kidney diseases [9, 10]. Manipulation of micro- University School of Medicine.
biota, such as with the use of probiotics, reduced blood urea ni-
trogen (BUN) levels and enhanced longevity in CKD rats [11, 12]. Morphological examination
Changes in the distribution of intestinal microbiota and reduc- Kidneys were fixed in 10% formaldehyde and embedded in
tions in uremic toxins caused by treatment with probiotics have paraffin blocks. Glomerular sclerosis was assessed by periodic
been observed in hemodialysis patients [13], although the clinical acid-Schiff (PAS) staining and renal fibrosis was assessed by
relevance of these alterations has not been determined [14]. Masson-trichrome staining. Glomerulosclerosis was evaluated

Downloaded from http://ndt.oxfordjournals.org/ at Serials Dept / Cornell University Medical Library on September 30, 2016
This study assessed the role of the gut environment in CKD by counting sclerotic glomeruli and calculating the glomerulo-
by using 5/6 nephrectomized (Nx) spontaneous hypertensive sclerotic index [18, 19]. The proportion of the fibrotic area was
rats (SHRs), in which we previously observed severe renal dam- measured using Image-Pro Plus 3.0 (Media Cybernetics, Silver
age and inflammation [15]. We found that a decrease in the Spring, MD, USA).
population of Lactobacillus (Lact) in Nx rats, along with disrup-
tion of the intestinal barrier, contributed to the progression of
Analysis of gut bacteria
CKD. We also showed that Lact supplementation ameliorated
renal damage and intestinal changes through activation of a The gut microbiota population was examined as described
Toll-like receptor (TLR) that recognizes Lact. These findings previously [20]. Briefly, DNA was extracted from the bead-
provide compelling evidence for the clinical use of Lact in treated suspension of fecal samples. Amplification of fecal
CKD patients to reverse the inflammatory response and protect 16S rDNA was performed using 25 μM of the labeled primers
against the progression of renal damage. 516f (50 TGCCAGCAGCCGCGGTA 30 ) and 1510r (50 GGTTA
CCTTGTTACGACTT 30 ). For terminal restriction fragment
ORIGINAL ARTICLE

length polymorphism (T-RFLP) analysis of the microbiota,


the resulting amplicons were digested with 2U of Bsl I (Cell Sig-
M AT E R I A L S A N D M E T H O D S naling, Danvers, MA, USA) for 1 h and the fragments were se-
parated using an automated DNA sequencer (ABI PRISM
Animal experiments 3130xl DNA Sequencer, Applied Biosystems, Carlsbad, CA,
Renal failure was induced in 6-week-old male SHRs (Charles USA). The lengths and peak areas of the resulting electrophero-
River, Wilmington, MA, USA) via the Nx model, as described gram were determined using GeneMapper software (Applied
previously [15]. In the first experiment, rats were randomly Biosystems). The relative abundances of operational taxonomic
assigned to two experimental groups, sham-operated SHR units (OTUs) were calculated by dividing the peak area of each
(Sham, n = 8) and Nx rats (n = 8). In the second experiment, fragment by the sum of all peak areas. The T-RFLP patterns of
rats were randomly assigned to three experimental groups: each sample were compared using the dissimilarity index [21].
sham-operated SHR (Sham, n = 8), Nx (n = 8) and Nx rats The amounts of bacteria in the fecal samples of each species
treated with 1 × 1010 CFU/kg/day Lactobacillus acidophilus were quantified by real-time quantitative PCR using the 7500
NT (Lact) (Nx+Lact, n = 8). Lact was kindly provided by Fast Real-time PCR System (Applied Biosystems), as previously
Nitto Pharmaceutical Industries (Kyoto, Japan). Twelve weeks reported [22]. The primers for each species are shown in Table 1
after nephrectomy, body weight and systolic blood pressure [23]. Results with the specific Lactobacillus, Bacteroides and
(SBP) were measured by the tail-cuff method (KN-210, Nat- Prevotella primers were normalized relative to those for the uni-
sume, Tokyo, Japan). Twenty-four-hour urinary protein excre- versal primers.
tion and serum concentrations of BUN, serum creatinine and IS
were measured as described previously [15, 16]. Serum PCS and Cell culture
IAA levels were measured by high-performance liquid chroma- Caco-2 sigmoid colon cancer cells (HTB-37; ATCC, LOT
tography [17]. Serum lipopolysaccharide (LPS) levels were 60143947) were grown in non-fibrin-coated flasks in Dulbecco’s
measured by turbidimetric-kinetic assay using a toxinometer modified eagle’s medium (DMEM, Invitrogen, Auckland, NZ),
MT6500 (Wako Pure Chemical Industries, Osaka). Serum containing glucose, L-glutamine, NaHCO3 and pyridoxine

Table 1. The primers used for bacterial gene analysis

Bacteria Forward Reverse


Lactobacilli AGCAGTAGGGAATCTTCCA CACCGCTACACATGGAG
Bacteroides GAAGGTCCCCCACATTG CAATCGGAGTTCTTCGTG
Prevotella CACCAAGGCGACGATCA GGATAACGCCTGGACCT
Universal primers TCCTACGGGAGGCAGCAGT GACTACCAGGGTATCTAATCCTGTT

402 A. Yoshifuji et al.


(Sigma, St Louis, MO, USA) at 37°C in 5% CO2. To evaluate the Backhamshire, UK), immunoreactive bands were detected
effects of uremic toxins, Caco-2 cells were treated with indole using an ECL detection kit (Amersham Biosciences, Uppsala,
(Sigma), the precursor of IS, at various doses, as well as with Sweden).
2.0 × 109 CFU/plate Lact in antibiotic-free DMEM media con-
taining 0.4% FBS (Invitrogen). The concentration of indole in Real-time reverse transcriptase polymerase chain reaction
the intestinal juice is estimated to be in the range of 0.1–100 μM (RT-PCR)
because the ‘human’ fecal concentration of indole reportedly Total RNA was extracted from the ascending colon tissue
ranges from 250 to 1100 μM [24, 25] and indole is estimated to and Caco-2 cells using TRIzol reagent (Invitrogen). Equal
be highly concentrated in the intestinal juice at least by 10- to amounts (1 μg) of total RNA from each sample were converted
100-fold because of the adsorption of water [26]. Furthermore, to cDNA using the Prime Script RT reagent kit with gDNA Era-
intestinal cells were covered with the mucous layer [26]. Based ser (Takara, Ohtsu, Japan). Real-time RT-PCR was performed
on this evidence, we tested the effects of indole in the preliminary using an ABI Step One Plus sequence detector (Applied Biosys-
experiments and found that 10 μM of indole was toxic to Caco-2 tems). Primers used are shown in Table 2. Levels of mRNA were

Downloaded from http://ndt.oxfordjournals.org/ at Serials Dept / Cornell University Medical Library on September 30, 2016
cells. We further examined the effects of indole on the expression normalized to those of β-actin.
of tight junction proteins at the concentrations ranging from 0.1
to 1 μM and observed that indole downregulated ZO-1 and Pseudogerm-free model experiment
occludin between the concentration of 0.1 and 1 nM, although A pseudogerm-free condition was created by administering
claudin-1 was not altered even at 1 μM. Hence the appropriate a broad-spectrum antibiotic cocktail consisting of ampicillin,
concentration used in the experiment was determined to be be- neomycin sulfate, vancomycin and metronidazole as described
tween 0.1 and 1 nM. To investigate the role of the Toll-like recep- previously [28]. Rats were randomly assigned to the following
tor 2 (TLR2), Caco-2 cells were pretreated with a nonselective five groups: (1) sham-operated SHR (Sham, n = 6), (2) Nx
TLR blocker, oxidized 1-palmitoyl-2-arachidonyl-snglycero-3- SHR (Nx, n = 6), (3) Sham plus antibiotics (Sham+Anti, n = 6),
phosphorylcholine (OxPAPC, InvivoGen, San Diego, CA, USA), (4) Nx plus antibiotics (Nx+Anti, n = 6) and (5) Nx+Anti trea-
30 min prior to the administration of Lact or indole. The cells ted with Lact (Nx+Anti+Lact, n = 6). Eight weeks after neph-
were harvested 24 h after the treatment and the expressions of rectomy, 24-h urinary protein excretion levels and SBP were

ORIGINAL ARTICLE
tight junction proteins, including occludin, ZO-1 and claudin-1, measured and the rats were sacrificed. The concentration of
were examined by immunoblotting. BUN and serum concentrations of creatinine and IS were mea-
sured as described [15].
Permeability assay
Permeability was estimated by measuring the paracellular Statistical analysis
transport of FITC-labeled 4-kDa dextran (FD4) as described Data are expressed as mean ± SEM and analyzed by one-way
previously [27]. Briefly, sterilized FD4 was added into the analysis of variance, followed by the Bonferroni’s post hoc test.
apical well at 25 mg/mL with or without indole, Lact or a com- A P-value <0.05 was considered statistically significant.
bination of both for 6 h. The translocation of FD4 across the
cells was determined by sampling the basolateral solution.
FD4 fluorescence at 485/535 nm was measured using a Perkin R E S U LT S
Elmer (Waltham, MS, USA) multimode plate reader (ARVO
X3). All permeability experiments were conducted in triplicate Decreased Lact population in nephrectomized rats
for five sets of cell preparations. FD4 permeability was ex- The microbiota between Nx SHRs, a model of severe ne-
pressed as the apparent permeability coefficient (Papp), calcu- phrosclerosis [15] and Sham-operated SHRs were compared.
lated as described previously [27]. Body weight was significantly lower (286.0 ± 38.2 g versus
366.8 ± 5.4 g, P < 0.05) and SBP was significantly higher
Immunoblotting (Figure 1A) in Nx compared with Sham rats. In addition,
Ascending colon tissues and Caco-2 cells were lysed and so- serum creatinine, IS and BUN levels, as well as urinary protein
nicated in a lysis buffer and centrifuged at 15 000 g for 15 min. excretion, were higher in Nx than in Sham rats (Figure 1B–E),
Aliquots of supernatants were electrophoresed on SDS-PAGE confirming that these rats were an appropriate model of severe
gels and transferred to nitrocellulose membranes, which were renal dysfunction. T-RFLP evaluation of intestinal microbiota
then incubated with primary antibodies against occludin, ZO-1 in feces from the terminal ileum showed that Bacteroides
and claudin-1 (Invitrogen). Following incubation with a second- (Bact) and Prevotella species were more abundant, whereas
ary antibody (HRP-linked anti-rabbit IgG; GE Healthcare, Lact species were less abundant in the Nx rats compared with

Table 2. The primers used for real-time PCR

Gene Forward Reverse


TLR2 (rat) GTACGCAGTGAGTGGTGCAAGT GGCCGCGTCATTGTTCTC
GAPDH (rat) GTTACCAGGGCTGCCTCTC GGGTTTCCCGTTGATGACC
TLR2 (human) GCCAGCAGGTTCAGGATGTC AGGCATCCCGCTCACTGTAA
β-actin (human) GTCAGGTCACTATCGGC CATGGATGCCACAGGATTCC

Effects of Lactobacillus on CKD 403


Downloaded from http://ndt.oxfordjournals.org/ at Serials Dept / Cornell University Medical Library on September 30, 2016
ORIGINAL ARTICLE

F I G U R E 1 : Effects of renal impairment on biochemical parameters and microbiota populations in rats. Spontaneously hypertensive rats
underwent 5/6 nephrectomy (Nx) or sham nephrectomy (Sham). Measurements were taken of (A) SBP, (B) serum creatinine concentrations, (C)
BUN, (D) urinary protein excretion and (E) serum IS concentrations. (F) Relative abundance of microbiota based on the average number of each
subfamily measured by T-RFLP analysis. (G) Populations of Bacteroides species (upper panel) and Lactobacillus (lower panel) in the colons of
Sham and Nx groups were examined by RT-PCR. (H) Relationship between urinary protein excretion levels and Bacteroides (upper panel) and
Lactobacillus (lower panel) populations. *P < 0.05, **P < 0.01 versus Sham (n = 8 per group).

the Sham rats (Figure 1F). Confirmatory real-time PCR using Effects of L. acidophilus on rats with CKD
species-specific primers showed that Bact species were higher Since decreased Lact in the intestine was associated with
and Lact lower in number in Nx compared with Sham rats urinary protein excretion, we investigated the effects of Lact
(Figure 1G), whereas Prevotella species were similar. The Lact supplementation in our rat model of CKD by treating Nx
population was significantly associated with urinary protein ex- rats with 1 × 1010 CFU/kg/day Lact for 12 weeks (Nx+Lact).
cretion levels (Figure 1H; R 2 = 0.425, P < 0.01), whereas the RT-PCR using DNA extracted from fecal samples revealed
Bact population was not.

404 A. Yoshifuji et al.


that the population of Lact was higher in Nx+Lact than in Nx day). Similarly, body weight was lower in Nx (296.4 ± 21.2 g)
rats (Figure 2A). Daily chow intake was lower in Nx than in Sham (361.4 ± 9.4 g) rats, although this reduction im-
(24.3 ± 4.3 g/day) than in Sham (36.0 ± 2.7 g/day) rats, and proved in Nx+Lact rats (322.0 ± 24.9 g). Systolic blood pressure
this decrease was not improved by Lact treatment (28.2 ± 3.1 g/ was similar in Nx and Nx+Lact rats (Figure 2B). Although the

Downloaded from http://ndt.oxfordjournals.org/ at Serials Dept / Cornell University Medical Library on September 30, 2016
ORIGINAL ARTICLE

F I G U R E 2 : Effects of Lactobacillus on nephrectomized rats. Spontaneously hypertensive rats underwent sham nephrectomy (Sham) or 5/6
nephrectomy (Nx) or Nx followed by Lactobacillus treatment (Nx+Lact), as described. (A) Numbers of Lactobacillus species in the colons, as
determined by RT-PCR. Measurements were made of (B) SBP, (C) BUN, (D) serum creatinine level, (E) daily urinary protein excretion, (F) serum
indoxyl sulfate (IS), (G) serum p-cresyl sulfate (PCS), (H) serum indole acetic acid (IAA), (I) serum lipopolysaccharide (LPS), (J) serum
interleukin-6 (IL-6) and (K) serum C-reactive protein (CRP) levels. The evaluation of (L) glomerular sclerosis by PAS staining and (M) interstitial
fibrosis by Masson-trichrome staining are shown. Lower panels represent the quantification of glomerular sclerosis and interstitial fibrosis. Scale
bar, 100 μm. *P < 0.05, **P < 0.01 versus Sham; #P < 0.05 versus Nx (n = 8 per group).

Effects of Lactobacillus on CKD 405


increase in BUN levels caused by Nx improved following Lact intestine by some microorganisms, dose-dependently downre-
treatment (Figure 2C), serum creatinine levels were not altered gulated the expression of occludin and ZO-1, while having no
(Figure 2D), suggesting that supplementation with Lact was not effect on claudin-1 expression (Figure 4A). These indole-
sufficient for the improvement of renal dysfunction in Nx SHRs. induced reductions in occludin and ZO-1 were restored by
The Lact-induced reduction of BUN may have been caused by the coadministration of Lact to Caco-2 cells. These effects of
decreased urea production in the intestine or decreased urea Lact were inhibited by the nonselective TLR blocker OxPAPC
entry from the intestine. However, the increased urinary protein (Figure 4B). Although indole did not alter the expression of
excretion in Nx rats was mitigated in Nx+Lact rats (Figure 2E), TLR2, it was increased by Lact treatment (Figure 4C). These
indicating that Lact had a renoprotective effect. Similarly, serum data implied that, in Nx rats, the expression of tight junction
concentrations of IS (Figure 2F), PCS (Figure 2G), LPS (Fig- proteins was disrupted by the reduction in the Lact population
ure 2I), IL-6 (Figure 2J) and CRP (Figure 2K), which were higher through the reduced activation of TLR2. Finally, the permeabil-
in Nx than in Sham rats, were mitigated in Nx+Lact rats. How- ity of the cell layer was assayed using FD-4 dye as described in
ever, the increase in serum IAA levels (Figure 2H) in Nx rats was the Materials and Methods section. Indole, at a concentration of

Downloaded from http://ndt.oxfordjournals.org/ at Serials Dept / Cornell University Medical Library on September 30, 2016
not altered in Nx+Lact rats. Nx rats showed a significantly high- 1.0 nM, increased the permeability of the Caco-2 cell layer, and
er degree of glomerulosclerosis, as evaluated by the glomerulo- this increase was inhibited by treatment with Lact (Figure 4D).
sclerotic index (Figure 2L) [18, 19] and increased tissue fibrotic This result implied that the increased intestinal permeability in
area on Masson-trichrome staining (Figure 2M) than Sham rats. Nx rats was improved by Lact treatment. It is deduced that Lact
Lact treatment improved the glomerulosclerotic but not the contributes to maintenance of the intestinal environment.
fibrotic index.
Lact-mitigated proteinuria in rats with CKD under
Molecular changes in the intestinal barrier induced pseudogerm-free conditions
by Lact The probiotic effects of Lact on proteinuria in Nx rats were
Disruption of the intestinal barrier has been shown to con- confirmed using pseudogerm-free conditions, where the effects
tribute to increased levels of serum cytokines during renal fail- of microbiota other than Lact are minimized. Treatment of
ure, as well as to the progression of renal impairment [8]. Colon Sham and Nx rats with an antibiotic cocktail consisting of
ORIGINAL ARTICLE

epithelial tight junctions are involved in maintaining the intes- ampicillin, neomycin sulfate, vancomycin and metronidazole
tinal barrier, wherein the proteins occludin, ZO-1 and yielded a pseudogerm-free condition in these animals, whereas
claudin-1 are found to be key components of intestinal tight some of these rats were supplemented with Lact, as confirmed
junctions. The expression of all three proteins was lower in by Gram staining of the feces of each experimental group
the Nx model compared with Sham-operated rats. Although (Figure 5A). Treatment of Sham and Nx rats with antibiotics
Lact supplementation restored the expression of occludin and had no effect on the levels of expression of serum creatinine,
ZO-1, it had no effect on claudin-1 expression (Figure 3A). It BUN, or IS, or on urinary protein excretion (Figure 5B–E).
is possible that the concentrations of uremic toxins or their pre- Treatment of Nx rats with Lact had no effect on serum creatin-
cursors affect the population of microbiota or the expression of ine levels (Figure 5B) but attenuated the increases in BUN and
tight junction proteins [29]. Although the fecal concentrations IS levels and urinary protein excretion even under pseudogerm-
of indole, p-cresol and phenol increased in Nx, Lact treatment free conditions (Figure 5C–E). These findings confirmed the
had no effect on any of these compounds (Figure 3B), indicat- probiotic effects of Lact.
ing that the treatment with Lact had little effect on the produc-
tion or degradation of these substances in the intestine. The
expression of tight junction proteins has been reported to be DISCUSSION
regulated by the activation of pattern recognition receptors,
TLRs [30, 31]. It has also been reported that Lact is recognized It has been strongly suggested that the gut microbiome is an im-
by TLR2 and activates TLR2 in various cells [32, 33]. We found portant factor in the maintenance of a healthy condition [36,
that the expression of TLR2 was lower in the Nx than in 37]. By using the recently developed T-RFLP technique fol-
the Sham group. This decrease was reversed in the Nx+ lowed by real-time PCR, this study analyzed the species present
Lact group (Figure 3C). These results suggested that the restor- in the gut microbiota and found that the Lact population was
ation of intestinal tight junction protein expression and the reduced under CKD conditions. The number of Lact was
concomitant increase in TLR2 expression by Lact leads to the found to be associated with urinary protein excretion levels,
suppression of increased intestinal permeability and to the re- and supplementation with Lact mitigated proteinuria in rats
duction in systemic IS levels as well as in LPS levels, thus with CKD. Furthermore, Lact helped to improve proteinuria
mitigating renal damage and systemic inflammation in Nx by restoring the expression of intestinal barrier proteins and re-
rats [34, 35]. ducing systemic inflammation, even under pseudogerm-free
conditions. Lact had a direct effect on intestinal cells by revers-
Indole- and Lact-modified tight junction molecules ing the indole-induced downregulation of tight junction pro-
in Caco-2 cells teins through activation of the TLR2 pathway. Thus this study
The mechanisms by which Nx and Lact induce molecular identified a specific species of microbiota that may be protective
changes in the intestinal barrier were investigated using the against renal damage and revealed the detailed mechanisms
Caco-2 colonic cell line. Indole, which is produced in the underlying these effects.

406 A. Yoshifuji et al.


Downloaded from http://ndt.oxfordjournals.org/ at Serials Dept / Cornell University Medical Library on September 30, 2016
ORIGINAL ARTICLE
F I G U R E 3 : Intestinal conditions in nephrectomized rats. Rats underwent sham nephrectomy (Sham), 5/6 nephrectomy (Nx) or Nx plus Lact
treatment (Nx+Lact), as described in the legend to Figure 2. (A) Expression levels of the tight junction proteins occludin (left panel), ZO-1 (middle
panel) and claudin-1 (right panel) in the colon. Each upper subpanel shows an immunoblot of a tight junction protein and each lower subpanel
shows the densitometric analysis of expression. (B) Fecal concentrations of indole (upper panel), p-cresol (middle panel) and phenol (lower panel).
(C) Levels of expressions of TLR2 in the colon, as shown by real-time RT-PCR. *P < 0.05, **P < 0.01 versus Sham; #P < 0.05 versus Nx (n = 8 per
group).

Colon microbiota are altered under uremic conditions. with uremia compared with healthy subjects [38]. It was
For example, the populations of certain microbes, such as reported that in CKD patients, probiotics with the combin-
Clostridiaceae, Enterobacteriacea and Verrucomicrobiacea, ation of L. acidophilus, Bifidobacterium longum and Strepto-
which produce indole or p-cresol, are higher, whereas the po- coccus thermophiles produced a significant reduction of BUN
pulations of microbial families with butyrate-producing en- and enhanced well-being [39]. Although several other studies
zymes, including Lact and Prevotellae, are lower in subjects also revealed that probiotics or prebiotics in CKD patients

Effects of Lactobacillus on CKD 407


Downloaded from http://ndt.oxfordjournals.org/ at Serials Dept / Cornell University Medical Library on September 30, 2016
ORIGINAL ARTICLE

F I G U R E 4 : Expression of intestinal barrier proteins in Caco-2 cells. (A) Caco-2 cells were incubated with various concentrations of indole and the
levels of expression of the tight junction proteins occludin (left panel), ZO-1 (middle panel) and claudin-1 (right panel) were assessed by im-
munoblotting. Each upper subpanel shows an immunoblot and each lower subpanel shows the densitometric analysis of that blot. *P < 0.05 versus
unstimulated cells (n = 5). (B) Effects of Lactobacillus (Lact) and the nonselective TLR blocker OxPAPC (Ox) on the expression of occludin (upper
panel), ZO-1 (middle panel) and β-actin (lower panel) in indole-treated (1.0 nmol/L) Caco-2 cells. Each upper subpanel shows an immunoblot
and each lower subpanel shows the densitometric analysis of that blot. – denotes no treatment. *P < 0.05 versus unstimulated cells, #P < 0.05 versus
indole-stimulated cells, ¶P < 0.05 versus indole-stimulated cells treated with Lact (n = 5). (C) Effects of Lact and indole on the expression of TRL2
mRNA in indole-treated Caco-2 cells. *P < 0.05 versus control (n = 5). (D) Effects of Lact and indole on the permeability of the Caco-2 cell
monolayer. *P < 0.05 versus unstimulated cells, #P < 0.05 versus indole-stimulated cells (n = 5).

reduce BUN levels or serum IS levels, some papers have to develop more efficient therapeutic strategies or to
reported no beneficial effects of probiotics, prompting a elucidate more precise mechanisms for dysbiosis in CKD
controversy regarding probiotic therapy [6, 14, 40]. In this [6, 14]. Our data provide relevant information for these fu-
sense, it has been proposed that future studies are necessary ture studies.

408 A. Yoshifuji et al.


Downloaded from http://ndt.oxfordjournals.org/ at Serials Dept / Cornell University Medical Library on September 30, 2016
ORIGINAL ARTICLE
F I G U R E 5 : The effects of Lactobacillus on nephrectomized rats in a pseudogerm-free environment. (A) Gram staining of stool samples of SHRs
(Sham), 5/6 nephrectomized SHRs (Nx), Sham rats treated with antibiotics (Sham+Anti), Nx rats treated with antibiotics (Nx+Anti) and Nx rats
treated with antibiotics and Lactobacillus (Nx+Anti+Lact). Measurements of (B) serum creatinine, (C) BUN (D) and serum IS levels and (E) daily
urinary protein excretion in the five rat groups. *P < 0.05, **P < 0.01 versus Sham+Anti; #P < 0.05 versus Nx+Anti (n = 6 per group).

In our experiment using the pseudogerm-free condition, al- serum levels of IS as well as LPS. Our findings show that the
though the number of Lact should be decreased by antibiotics, gut contains various types of microbiota that may have an effect
the antibiotic cocktail used in this study had no effects on pro- on renal damage, either positively or negatively. Both a decrease
teinuria. These data imply that antibiotic therapy also reduces in renoprotective Lact and an increase in renal pathogenic Bact
other microbiota with unfavorable effects on the kidney, such as contributed to the progression or maintenance of renal damage
Clostridium species or Bact species, which are the microbiota in Nx rats. Collectively, it is suggested that the balance between
producing indole. Our analysis showed an increase in the popu- these bacteria may have some role in CKD.
lation of Bact, and it was reported that ∼20% of Bact species In the present study, Lact treatment salvaged the expression
produce indole [41]. Consistently, fecal concentrations of of tight junction proteins in the intestine of Nx rats. Our study
indole in the intestine were higher in Nx compared with provided the molecular mechanism and evidence for the direct
Sham rats. Because indole directly downregulates the expres- action of Lact in intestinal cells. In the previous study, the
sion of tight junction proteins that form a barrier of intestinal downregulation of tight junction proteins by infection of
cells, and also because indole is a direct precursor of the uremic Caco-2 cells with Escherichia coli or Salmonella was reversed
toxin IS, the increase in the population of Bact in the Nx model by the addition of Lactobacillus amylophilus via modulation
could lead to increased intestinal permeability and increased of the extracellular signal-regulated kinase (ERK) and c-Jun

Effects of Lactobacillus on CKD 409


NH2-terminal kinase (JNK) pathways [42]. In addition, activa- cytokines, endotoxins, uremic toxins and microorganisms [9,
tion of the TLR2 pathway has been reported to lead to ERK or 32]. Tight junction proteins form an effective barrier against
JNK activation [43, 44]. TLRs are mucosal pattern recognition the influx of these noxious substances from the gastrointestinal
receptors that recognize microbes, leading to the preservation of lumen to the internal milieu. In a uremic state, intestinal barrier
tight junction integrity [30, 31]. Taken together, our findings function is impaired, resulting in increased circulating endo-
suggest that Lact regulates the expression of tight junction pro- toxin levels and enhanced systemic inflammation [48]. This
teins through the direct activation of TLR2. Consistent with this systemic inflammation contributes to the progression of renal
hypothesis, our experiments using Caco-2 cells showed that the impairment [10]. In the previous report, Nx rats exhibited low-
restoration of indole-induced downregulation of ZO-1 and grade inflammation, leading to impairment of kidney structure
occludin by Lact was inhibited by pretreatment with the TLR and function [49]. In this study, Lact restored intestinal tight
antagonist OxPAPC. In CKD, reduction in the Lact population junction protein expression, which possibly reduced intestinal
and the subsequent downregulation of TLR2 also contributed permeability as shown in the experiment described in Fig-
to downregulation of tight junction proteins (Figure 6). Supple- ure 4D. These effects stabilized intestinal barrier function,

Downloaded from http://ndt.oxfordjournals.org/ at Serials Dept / Cornell University Medical Library on September 30, 2016
mentation with Lact successfully reversed these changes. We blocked the translocation of LPS and attenuated systemic in-
also found that tight junction proteins were downregulated by flammation, as indicated by the reduction in both serum IL-6
the direct action of indole. Indole has been shown to activate and CRP levels. These effects ultimately improved renal sclerot-
arylhydroxycarbon (AhR) and the pregnane X receptors ic changes and proteinuria in Nx rats.
(PXRs) expressed in cell nuclei [45, 46]. Our results suggest a Moreover, the increase in uremic solutes IS and PCS has
possible cross-talk between these nuclear receptors and TLR been reported to induce renal inflammatory responses [4, 50,
signaling in the intestinal cells (Figure 6) [10, 47]. 51]. Our study implied that in Nx rats, besides an impaired
The intestine plays an important role in regulating systemic clearance, the increase in intestinal indole and p-cresol produc-
inflammation by serving as a barrier against the entry of various tion, presumably by an increase in the population of indole-
ORIGINAL ARTICLE

F I G U R E 6 : Scheme depicting the effects of CKD on microbiota populations and systemic inflammation. Intestinal indole downregulates the
expression of tight junction proteins, presumably through binding to the arylhydroxycarbon receptor (AhR) or PXR. In contrast, Lactobacillus
maintains protein expression through the activation of TLR2. In CKD, indole concentrations increase and the population of Lactobacillus de-
creases, which both result in the downregulation of expression of tight junction proteins. These mechanisms enhance intestinal permeability, thus
allowing various uremic toxins such as indole and p-cresol to enter the systemic circulation, consequently elevating the systemic indoxylsulfate (IS)
and p-cresylsulfate (PCS) levels. Increased permeability also allows bacterial lipopolysaccharide (LPS) to translocate to the systemic circulation,
leading to systemic inflammation along with increases in serum cytokine levels such as IL-6 and CRP. These changes ultimately cause renal cell
damage. Lactobacillus supplementation reverses these conditions and mitigates renal damage.

410 A. Yoshifuji et al.


and p-cresol-producing microbiota, and the disruption of the 7. Ritz E. Intestinal-renal syndrome: mirage or reality? Blood Purif 2011; 31:
intestinal barrier due to the increased levels of the indole itself 70–76
8. Vaziri ND, Yuan J, Khazaeli M et al. Oral activated charcoal adsorbent
or the reduction in the Lact population could be responsible for (AST-120) ameliorates chronic kidney disease-induced intestinal epithelial
the observed increase in serum IS and PCS levels. Treatment of barrier disruption. Am J Nephrol 2013; 37: 518–525
Nx rats with Lact stabilized barrier function and reduced the 9. Anders HJ, Andersen K, Stecher B. The intestinal microbiota, a leaky gut,
serum levels of IS and PCS, resulting in reductions in renal in- and abnormal immunity in kidney disease. Kidney Int 2013; 83: 1010–1016
flammation and in renal sclerosis. Because Lact treatment did 10. Ramezani A, Raj DS. The gut microbiome, kidney disease, and targeted in-
terventions. J Am Soc Nephrol 2014; 25: 657–670
not alter fecal concentrations of indole, as well as p-cresol, or 11. Ranganathan N, Patel B, Ranganathan P et al. Probiotic amelioration of
renal excretory function, Lact-induced reductions in systemic azotemia in 5/6th nephrectomized Sprague-Dawley rats. Sci World J
IS and PCS were considered to be primarily due to blocking 2005; 5: 652–660
of indole and p-cresol entry from the intestine. These mechan- 12. Ranganathan N, Patel BG, Ranganathan P et al. In vitro and in vivo assess-
isms may explain the renoprotective effects of Lact (Figure 6). ment of intraintestinal bacteriotherapy in chronic kidney disease. ASAIO J
2006; 52: 70–79
This barrier-stabilizing effect may also explain why Lact re- 13. Vaziri ND, Wong J, Pahl M et al. Chronic kidney disease alters intestinal

Downloaded from http://ndt.oxfordjournals.org/ at Serials Dept / Cornell University Medical Library on September 30, 2016
duced BUN levels without affecting renal function, as serum microbial flora. Kidney Int 2013; 83: 308–315
urea levels are also determined by urea production by intestinal 14. Rossi M, Klein K, Johnson DW et al. Pre-, pro-, and synbiotics: do they have
microbiota and by urea entry into the intestine [52]. Several a role in reducing uremic toxins? A systematic review and meta-analysis. Int
other metabolic pathways, including uptake via the intestinal J Nephrol 2012; 2012: 631–673
15. Sugano N, Wakino S, Kanda T et al. T-type calcium channel blockade as a
wall, and metabolic conjugation reactions, such as sulfotrans- therapeutic strategy against renal injury in rats with subtotal nephrectomy.
feration of indole to IS or that of p-cresol to PCS by the intes- Kidney Int 2008; 73: 826–834
tinal wall, may be involved in uremic toxin generation [6]. The 16. Saito S, Shimizu H, Yisireyili M et al. Indoxyl sulfate-induced activation of
effects of Nx and Lact treatment on these pathways remain to be ( pro)renin receptor is involved in expression of TGF-β1 and α-smooth
investigated. muscle actin in proximal tubular cells. Endocrinology 2014; 155:
1899–1907
In conclusion, Lact exhibited renal protective effects in CKD 17. Itoh Y, Ezawa A, Kikuchi K et al. Protein-bound uremic toxins in hemodi-
rats by modulating the gut environment and regulating system- alysis patients measured by liquid chromatography/tandem mass spec-
ic inflammation. These effects suggest a novel mechanism by trometry and their effects on endothelial ROS production. Anal Bioanal

ORIGINAL ARTICLE
which renoprotective Lact can improve systemic inflammation Chem 2012; 403: 1841–1850
and slow the progression of renal damage. 18. Wu L, Cox A, Roe C et al. Transforming growth factor β1 and renal injury
following subtotal nephrectomy in the rat: role of the renin-angiotensin sys-
tem. Kidney Int 1997; 51: 1553–1567
19. Kelly DJ, Zhang Y, Gow R et al. Tranilast attenuates structural and function-
al aspects of renal injury in the remnant kidney model. J Am Soc Nephrol
AC K N O W L E D G E M E N T S
2004; 15: 2619–2629
20. Nagashima K, Hisada T, Sato M et al. Application of new primer-enzyme
This work was supported by the Scientific Research Fund of the combinations to terminal restriction fragment length polymorphism profil-
Ministry of Education, Culture, Sports, Science, and Technol- ing of bacterial populations in human feces. Appl Environ Microbiol 2003;
ogy of Japan (grant no. 70573286). 69: 1251–1262
21. Hiraishi A, Iwasaki M, Shinjo H. Terminal restriction pattern analysis of
16S rRNA genes for the characterization of bacterial communities of acti-
vated sludge. J Biosci Bioeng 2000; 90: 148–156
C O N F L I C T O F I N T E R E S T S TAT E M E N T 22. Takayama F, Taki K, Niwa T. Bifidobacterium in gastro-resistant seamless
capsule reduces serum levels of indoxyl sulfate in patients on hemodialysis.
Am J Kidney Dis 2003; 41(Suppl): S142–S145
None declared.
23. Larsen N, Vogensen FK, van den Berg FW et al. Gut microbiota in human
adults with type 2 diabetes differs from non-diabetic adults. PLoS One
2010; 5: e9085
24. Karlin DA, Mastromarino AJ, Jones RD et al. Fecal skatole and indole and
REFERENCES breath methane and hydrogen in patients with large bowel polyps or cancer.
J Cancer Res Clin Oncol 1985; 109: 135–141
1. Vanholder R, Schepers E, Pletinck A et al. The uremic toxicity of indoxyl
25. Zuccato E, Venturi M, Di Leo G et al. Role of bile acids and metabolic ac-
sulfate and p-cresyl sulfate: a systematic review. J Am Soc Nephrol 2014; 25:
tivity of colonic bacteria in increased risk of colon cancer after cholecystec-
1897–1907
tomy. Dig Dis Sci 1993; 38: 514–519
2. Muteliefu G, Shimizu H, Enomoto A et al. Indoxyl sulfate promotes vascu-
26. Hall JE. Digestion and absorption in the gastrointestinal tract. In: Hall JE
lar smooth muscle cell senescence with upregulation of p53, p21, and pre-
(ed). Guyton and Hall Textbook of Medical Physiology, 13th edn. Philadel-
lamin A through oxidative stress. Am J Physiol Cell Physiol 2012; 303:
phia, PA: Elsevier, 2015, pp. 833–842
C126–C134.
3. Shimizu H, Yisireyili M, Higashiyama Y et al. Indoxyl sulfate upregulates 27. Vllasaliu D, Casettari L, Fowler R et al. Absorption-promoting effects of
renal expression of ICAM-1 via production of ROS and activation of chitosan in airway and intestinal cell lines: a comparative study. Int J
NF-κB and p53 in proximal tubular cells. Life Sci 2013; 92: 143–148. Pharm 2012; 430: 151–160
4. Watanabe H, Miyamoto Y, Honda D et al. p-Cresyl sulfate causes renal 28. Emmelot CH, van der Waaij D. The dose at which neomycin and poly-
tubular cell damage by inducing oxidative stress by activation of NADPH myxin B can be applied for selective decontamination of the digestive
oxidase. Kidney Int 2013; 83: 582–592 tract in mice. J Hyg (Lond) 1980; 84: 331–340
5. Dou L, Sallée M, Cerini C et al. The cardiovascular effect of the uremic sol- 29. McCall IC, Betanzos A, Weber DA et al. Effects of phenol on barrier
ute indole-3 acetic acid. J Am Soc Nephrol 2015; 26: 876–887 function of a human intestinal epithelial cell line correlate with altered
6. Vanholder R, Glorieux G. The intestine and the kidneys: a bad marriage can tight junction protein localization. Toxicol Appl Pharmacol 2009; 241:
be hazardous. Clin Kidney J 2015; 8: 168–179 61–70

Effects of Lactobacillus on CKD 411


30. Cario E, Gerken G, Podolsky DK. Toll-like receptor 2 controls mucosal in- 42. Yu Q, Wang Z, Yang Q. Lactobacillus amylophilus D14 protects tight junc-
flammation by regulating epithelial barrier function. Gastroenterology tion from enteropathogenic bacteria damage in Caco-2 cells. J Dairy Sci
2007; 132: 1359–1374 2012; 95: 5580–5587
31. Rakoff-Nahoum S, Paglino J, Eslami-Varzaneh F et al. Recognition of com- 43. Ahmad R, Shihab PK, Jasem S et al. FSL-1 induces MMP-9 production
mensal microflora by toll-like receptors is required for intestinal homeosta- through TLR-2 and NF-κB/AP-1 signaling pathways in monocytic
sis. Cell 2004; 118: 229–241 THP-1 cells. Cell Physiol Biochem 2014; 34: 929–942
32. Karczewski J, Troost FJ, Konings I et al. Regulation of human epithelial tight 44. Xu C, Wang Y, Sun R et al. Modulatory effects of vasoactive intestinal pep-
junction proteins by Lactobacillus plantarum in vivo and protective effects tide on intestinal mucosal immunity and microbial community of weaned
on the epithelial barrier. Am J Physiol Gastrointest Liver Physiol 2010; 298: piglets challenged by an enterotoxigenic Escherichia coli (K88). PLoS One
G851–G859 2014; 9: e104183
33. Sakai F, Hosoya T, Ono-Ohmachi A et al. Lactobacillus gasseri SBT2055 in- 45. Schroeder JC, Dinatale BC, Murray IA et al. The uremic toxin 3-indoxyl
duces TGF-β expression in dendritic cells and activates TLR2 signal to pro- sulfate is a potent endogenous agonist for the human aryl hydrocarbon re-
duce IgA in the small intestine. PLoS One 2014; 9: e105370 ceptor. Biochemistry 2010; 49: 393–400
34. Vaziri ND, Goshtasbi N, Yuan J et al. Uremic plasma impairs barrier func- 46. Heath-Pagliuso S, Rogers WJ, Tullis K et al. Activation of the Ah receptor by
tion and depletes the tight junction protein constituents of intestinal epithe- tryptophan and tryptophan metabolites. Biochemistry 1998; 37: 11508–11515
lium. Am J Nephrol 2012; 36: 438–443 47. Venkatesh M, Mukherjee S, Wang H et al. Symbiotic bacterial metabolites

Downloaded from http://ndt.oxfordjournals.org/ at Serials Dept / Cornell University Medical Library on September 30, 2016
35. Vaziri ND, Yuan J, Norris K. Role of urea in intestinal barrier dysfunction regulate gastrointestinal barrier function via the xenobiotic sensor PXR and
and disruption of epithelial tight junction in CKD. Am J Nephrol 2013; 37: Toll-like receptor 4. Immunity 2014; 41: 296–310
1–6 48. Vaziri ND. CKD impairs barrier function and alters microbial flora of the
36. Sekirov I, Russell SL, Antunes LC et al. Gut microbiota in health and dis- intestine: a major link to inflammation and uremic toxicity. Curr Opin Ne-
ease. Physiol Rev 2010; 90: 859–904 phrol Hypertens 2012; 21: 587–592
37. Cénit MC, Matzaraki V, Tigchelaar EF et al. Rapidly expanding knowledge 49. Kondo M, Tahara A, Hayashi K et al. Renoprotective effects of novel
on the role of the gut microbiome in health and disease. Biochim Biophys interleukin-1 receptor-associated kinase 4 inhibitor AS2444697 through
Acta 2014; 1842: 1981–1992 anti-inflammatory action in 5/6 nephrectomized rats. Naunyn Schmiede-
38. Wong J, Piceno YM, Desantis TZ et al. Expansion of urease and uricase- bergs Arch Pharmacol 2014; 387: 909–919
contaning, indole- and p-cresol-forming and contraction of short chain 50. Shimizu H, Bolati D, Adijiang A et al. NF-kappaB plays an important role in
fatty acid-producing intestinal bacteria in ESRD. Am J Nephrol 2014; 39: indoxyl sulfate-induced cellular senescence, fibrotic gene expression, and
230–237 inhibition of proliferation in proximal tubular cells. Am J Physiol Cell Phy-
39. Ranganathan N, Ranganathan P, Friedman EA et al. Pilot study of probiotic siol 2011; 301: C1201–C1212
dietary supplementation for promoting healthy kidney function in patients 51. Yisireyili M, Shimizu H, Saito S et al. Indoxyl sulfate promotes cardiac fi-
ORIGINAL ARTICLE

with chronic kidney disease. Adv Ther 2010; 27: 634–647 brosis with enhanced oxidative stress in hypertensive rats. Life Sci 2013; 92:
40. Schulman G, Berl T, Beck GJ et al. Randomized placebo-controlled EPPIC 1180–1185
trials of AST-120 in CKD. J Am Soc Nephrol 2015; 26: 1732–1746 52. Cummings JH. Absorption and secretion by the colon. Gut 1975; 16:
41. Jenkins SG, Birk RJ, Zabransky RJ. Differences in susceptibilities of species 323–329
of the Bacteroides fragilis group to several beta-lactam antibiotics: indole
production as an indicator of resistance. Antimicrob Agents Chemother Received for publication: 4.4.2015; Accepted in revised form: 8.9.2015
1982; 22: 628–634

412 A. Yoshifuji et al.

Potrebbero piacerti anche