You are on page 1of 12

Oral Combined Therapy with Probiotics and

Alloantigen Induces B Cell −Dependent


Long-Lasting Specific Tolerance

Downloaded from http://www.jimmunol.org/ at Univ of California-San Diego Serials/Biomed Lib 0699 on September 15, 2014
This information is current as Ana C. T. Mercadante, Suelen M. Perobelli, Ana P. G.
of September 15, 2014. Alves, Triciana Gonçalves-Silva, Wallace Mello, Ana C.
Gomes-Santos, Anderson Miyoshi, Ana M. C. Faria and
Adriana Bonomo
J Immunol 2014; 192:1928-1937; Prepublished online 22
January 2014;
doi: 10.4049/jimmunol.1301034
http://www.jimmunol.org/content/192/4/1928

Supplementary http://www.jimmunol.org/content/suppl/2014/01/22/content.1301034.
Material DCSupplemental.html
References This article cites 54 articles, 23 of which you can access for free at:
http://www.jimmunol.org/content/192/4/1928.full#ref-list-1
Subscriptions Information about subscribing to The Journal of Immunology is online at:
http://jimmunol.org/subscriptions
Permissions Submit copyright permission requests at:
http://www.aai.org/ji/copyright.html
Email Alerts Receive free email-alerts when new articles cite this article. Sign up at:
http://jimmunol.org/cgi/alerts/etoc
Errata An erratum has been published regarding this article. Please see next page
or:
http://www.jimmunol.org/content/192/8/3990.full.pdf

The Journal of Immunology is published twice each month by


The American Association of Immunologists, Inc.,
9650 Rockville Pike, Bethesda, MD 20814-3994.
Copyright © 2014 by The American Association of
Immunologists, Inc. All rights reserved.
Print ISSN: 0022-1767 Online ISSN: 1550-6606.
The Journal of Immunology

Oral Combined Therapy with Probiotics and Alloantigen


Induces B Cell–Dependent Long-Lasting Specific Tolerance

Ana C. T. Mercadante,* Suelen M. Perobelli,† Ana P. G. Alves,* Triciana Gonçalves-Silva,†


Wallace Mello,‡ Ana C. Gomes-Santos,x Anderson Miyoshi,{ Ana M. C. Faria,x and

Downloaded from http://www.jimmunol.org/ at Univ of California-San Diego Serials/Biomed Lib 0699 on September 15, 2014
Adriana Bonomo*,†,‡
Allogeneic hematopietic stem cell transplantation (aHSCT) is widely used for the treatment of hematologic malignancies. Although
aHSCT provides a good response against the malignant cells (graft-versus-leukemia [GVL]), it also leads to the development of
graft-versus-host disease (GVHD), a severe disease with high mortality and morbidity rates. Therapy for GVHD is commonly based
on nonspecific immunosupression of the transplanted recipient, resulting in the concomitant inhibition of the GVL effect. In this
study, we propose an alternative approach to specifically suppress GVHD while sparing the GVL, based on oral treatment of trans-
plant donors with recipient Ags, associated with the intake of probiotic Lactococcus lactis as tolerogenic adjuvant (combined
therapy). We show that treatment of C57BL/6 donor mice with combined therapy before the transplant protects the recipients
F1 (C57BL/6 3 BAL/c) mice from clinical and pathological manifestations of disease, resulting in 100% survival rate. Importantly,
the animals keep the immunological competence maintaining the GVL response as well as the response to third-party Ags. The
protection is specific, long lasting and dependent on donor IL-10–sufficient B cells activity, which induces regulatory T cells in the
host. These data suggest that combined therapy is a promising strategy for prevention of GVHD with preservation of GVL,
opening new possibilities to treat human patients subjected to transplantation. The Journal of Immunology, 2014, 192: 1928–1937.

T
he main limitation of allogeneic hematopoietic stem cell (GVL) effect (4). Because of the close association between GVH
transplantation (aHSCT) for the treatment of malignant and GVL reactions (4), much effort has been put forth to develop
and nonmalignant disease is the development of graft therapies that reduce the first while maintaining the latter (1).
versus host disease (GVHD), a complex multiorgan inflammatory Oral tolerance is classically defined as the suppression of im-
syndrome that results from recognition of genetically disparate mune responses to Ags that have been previously administrated by
tissues of the recipient patient by the incoming allogenic donor the oral route (5). Far from being just a matter of hyporesponsive-
T cells (1). GVHD treatment is based on use of immunosupressive ness, oral tolerance is an active immunologic event influenced by
agents that block T cell function in a nonspecific way, leading to numerous factors such as Ag dose, microbiota, and costimulatory
undesired consequences—because these alloreactive T cells also molecules, resulting from multiple mechanisms of action (6). In-
facilitate engraftment of hematopoietic stem cells, accelerate immune duction of oral tolerance has been shown to be effective in various
reconstitution (1–4), and contribute to the graft-versus-leukemia experimental models of immune disorders (7–9) including GVHD
(10–12). In fact, Nagler et al. (12) demonstrated that it is possible
to alleviate acute GVHD (aGVHD) severity by the induction of
*Department of Experimental Medicine, Brazilian National Cancer Institute, Rio oral tolerance in the recipient, although the maintenance of the
de Janeiro 20231-050, Brazil; †Department of Immunology, Microbiology Institute GVL effect was not addressed in that study.
Prof. Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902,
Brazil; ‡Laboratory of Thymus Research, Oswaldo Cruz Institute, Rio de Janeiro
Administration of probiotics by the oral route also have been
21040-360, Brazil; xDepartment of Biochemistry and Immunology, Biological Sci- shown to modulate the immune response, favoring either pro- (13,
ence Institute, Federal University of Minas Gerais, Belo Horizonte 30161-970, 14) or anti-inflammatory (15, 16) responses. Recent studies sug-
Brazil; and {Department of Biology, Biological Science Institute, Federal Univer-
sity of Minas Gerais, Belo Horizonte 30161-970, Brazil gest that TLRs and NF-kB signaling pathways play a crucial role
Received for publication April 18, 2013. Accepted for publication December 17,
in the outcome of the response (17–21). Also, probiotics can in-
2013. fluence the integrity of the gut epithelial cell barrier through
This work was supported by grants from Conselho Nacional de Pesquisas (57.3806/ modulation of mucus, defensins, and IgA production, reduction of
2008 and 306624/2010-9), Instituto Nacional de Ciência e Tecnologia (306624/2010- bacterial adhesion, and enhancement of tight junction and cell
9), Fundação de Amparo à Pesquisa do Estado do Rio de Janeiro (E-26/111.423/
2010, E-26/110.949/2008, and E-26/110.323/2010), the Swiss Bridge Foundation
survival (22–24).
(2301500), and Fundação de Amparo à Pesquisa do Estado de Minas Gerais The relationship between the endogenous microbiota and de-
(APQ00575-09). velopment of aGVHD was proposed ∼40 y ago in experimental
Address correspondence and reprint requests to Prof. Adriana Bonomo, Laboratory of models (25) and confirmed in humans later on (26). As hypothe-
Thymus Research, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Avenida Brasil
4365, Manguinhos, Rio de Janeiro 21045-900, Brazil. E-mail address: abonomo@
sized by Hill et al. (27), damage of the host intestinal epithelium
micro.ufrj.br by the conditioning regiment results in systemic exposure to mi-
The online version of this article contains supplemental material. crobial Ags that ultimately lead to an inflammatory milieu that is
Abbreviations used in this article: aGVHD, acute graft versus host disease; B6, C57BL6; adequate for donor T cell activation. With those results in mind,
BMT, bone marrow transplantation; Breg, regulatory B cell; GVL, graft versus leuke- intestinal decontamination became a common practice in aHSCT
mia; HSCT, hematopoietic stem-cell transplantation; LAP, latency-associated polypep- (26, 28). More recently, Jenq et al. (29) showed that composition
tide; mLn, mesenteric lymph node; pLN, peripheral lymph node; Treg, regulatory T cell.
of the gut flora of recipient mice that developed GVHD had
Copyright Ó 2014 by The American Association of Immunologists, Inc. 0022-1767/14/$16.00 a dramatic loss of bacterial diversity and a distinct composition

www.jimmunol.org/cgi/doi/10.4049/jimmunol.1301034
The Journal of Immunology 1929

compared with recipient mice that did not develop GVHD. In ad- Bone marrow transplantation
dition, the authors revealed that antibiotic-treated mice showed an Bone marrow transplantation (BMT) was modified from described previ-
emergence of species that was associated with exacerbated GVHD. ously (32). F1 mice were lethally irradiated with 950 cGy total body ir-
Interestingly, Gerbitz et al. (30) demonstrated that oral adminis- radiation (TH780C irradiator with a cobalt 60 [60Co] source), followed by
tration of Lactobacillus rhamnosus GG before and after transplan- the i.v. transfer of 5 3 106 bone marrow cells from healthy donors along
tation not only improved survival and ameliorated aGVHD clinical with 5 3 106 T cells from total spleen of treated or untreated donors. When
indicated, spleen cells were depleted of CD25+ cells or CD19+ cells. In
scores but also reduced the translocation of enteric bacteria. Both GVL experiments, 105 P815 GFP-expressing mastocytoma cells were
works suggest that manipulation of the gut microbiota might be as injected together with the bone marrow inoculum.
good as decontamination with antibiotics to prevent GVHD.
GVHD clinical and histological score
Although promising, the clinical application of both protocols,
oral tolerance induction (12) and probiotic treatment (30), cannot Mice were monitored daily for survival and weekly for GVHD clinical

Downloaded from http://www.jimmunol.org/ at Univ of California-San Diego Serials/Biomed Lib 0699 on September 15, 2014
be used in transplanted patients. Once the conditioning regimen score. Clinical evaluation was modified from the literature (33, 34) and was
based on six parameters: weight loss, fur texture, activity, posture, fecal
alters the intestinal epithelial barrier, these patients require special consistency, and survival rate. For histopathologic examination, samples of
attention regarding their diet. As so, no live or raw food is skin, liver, and colon collected at day 21 posttransplant were formalin pre-
permitted during the peritransplant phase, precluding oral ad- served, paraffin embedded, sectioned, and stained with H&E. The pathologic
ministration of Ags or probiotics. score system was based on evaluation of individual parameters for each organ
being the final score obtained by the sum of them. For skin sections, in-
In this report, we reasoned if tolerance induction in the donor,
flammatory infiltration, fibrosis, loss of appendices, epidermal chances, and
rather than in the recipient (12, 30), could work as a strategy to ulceration were evaluated. For liver samples, parenchyma suffering, inflam-
dissociate GVH and GVL effects. We propose a new protocol that matory infiltration, and portal space destruction were evaluated. For colons,
combines the induction of oral tolerance in bone marrow trans- lamina propria infiltration, deeper layer infiltration, structural changes, and
plant donors to Ags from the recipient, with the use of the po- damage extension were analyzed.
tential probiotic Lactococcus lactis NCDO2118 as tolerogenic GVL evaluation
adjuvant (combined therapy). Using a haploidentical model of
At day 25 posttransplant, samples of spleen, liver, bone marrow, total lymph
aGVHD, we found that treatment of donor mice with the com- nodes, and blood were analyzed by flow cytometry for the presence of
bined therapy protects recipients from aGVHD while maintaining P815-GFP+ cell. For determination of tumor total elimination, recipients
the GVL effect. This protection is long lasting, Ag specific, and were monitored daily for tumor-related morbidity and mortality. Tumor-
dependent on donor B cells, which induce regulatory T cells related death was determined if mice had paralysis (in that case they were
(Tregs) in the host. euthanized immediately and examined) and if autopsy identified hepat-
osplenomegaly and presence of macroscopic tumor nodules on the liver,
spleen, or bone marrow.
Materials and Methods Cell depletions
Animals
For Treg depletion, spleen cells from donor mice were incubated with anti-
Male 8- to 10-wk-old C57BL/6 (B6) (H-2b), B6 IL-10 knockout, or F1 CD25 (PC61.5) rat anti-mouse IgG Ab and positively selected using goat
(BALB/c 3 B6dXb) mice were used as cell donors for GVHD induction anti-rat IgG Dynabeads (Invitrogen, Carlsbad, CA). B cell depletion was
experiments and female 10- to 12-wk-old F1 (BALB/c 3 B6 2 H-2dXb) done by positive selection using goat anti-mouse IgG Dynabeads (Invi-
mice were used as recipients. For skin transplants, male 12- to 14-wk-old trogen). After depletion, cell suspensions presented ,1% of CD25+ and 3%
BALB/c (H-2d), B10.A (H-2a), or B6 mice were used as donors, and fe- of CD19+ cells. Both procedures were done accordingly to the manu-
male B6 mice or F1 (BALB/c 3 B6) mice were used as recipient. All facturer’s recommendations. For in vivo Treg depletion, recipient chimeras
animals were bred at the Brazilian National Cancer Institute animal facility were injected i.p. with PC61 mAb (250 mg/mouse) at day 3 posttransplant.
(Rio de Janeiro, Brazil), housed in sterilized microisolators, and were On day 14 posttransplant another i.p. dose of PC61 mAb were injected
handled according to the institutional guidelines. Animal care and all (100 mg/mice).
animal procedures were previously approved by Ethical Committee for
Animal Experimentation of the Brazilian National Cancer Institute. Purification and adoptive transfer of B cells
Bacterial growth conditions and administration to donor mice CD19+ B cells were isolated from donor spleen by positive selection using
anti-CD19 MACS microbeads (Miltenyi Biotec, Auburn, CA), according
The wild-type L. lactis ssp lactis NCDO2118 strain (obtained from Lab- to the manufacturer’s recommendations. For adoptive transfer experi-
oratório de Genética Celular e Molecular from Universidade Federal de ments, 1 3 107 purified CD19+ B cells (purity . 95%) from treated or
Minas Gerais, MG/Brazil - taxonomy ID 1117941 at the National Center untreated donor were injected along with the graft. When indicated, the
for Biotechnology Information database). The bacteria were grown in M17 spleen used as T cell source was previously depleted of total B cells and
broth (Difco) supplemented with 0.5% glucose at 30˚C without agitation reconstituted with purified CD19+ B cells.
for 24 h. In the second day, when a 2.0 OD at 600 nm (OD, nm) was
reached, an inoculum with 5 ml was diluted in 25 ml supplemented M17 Flow cytometry staining and analyses
broth and offered to the animals.
For surface staining, cell suspensions were preincubated with 2% normal
Preparation of protein extract from recipient mice mouse serum in PBS (Sigma-Aldrich, St. Louis, MO) on ice, followed by
incubation with appropriate Ab mixture for 15 min. For Foxp3 staining,
To prepare the protein extract, F1 (BALB/c 3 B6) spleen cells were
cells were labeled as specified in the eBioscience kit standard protocol. All
resuspended on ice-cold PBS and filtered through a 40-mm cell strainer, samples were acquired in a FACSCalibur (BD Biosciences, San Jose, CA)
cells were disrupted by freezing and thawing five times and boiled for or in a FACSCanto (BD Biosciences). Acquisitions were performed using
10 min. Soluble fraction was obtained by centrifugation at 14,000 rpm, CellQuest (BD Biosciences) software and BD FACSDiva software, re-
30 min, at 4˚C and used as oral treatment. Total protein was quantified spectively. Analyses were performed using FlowJo software (Tree Star,
with Bio-Rad protein kit assay, according to the manufacturer’s spec- Ashland, OR). For cell staining the following Abs were used: FITC or
ifications. allophycocyanin anti-mouse CD4 (GK1.5); PECy7 CD8a (53-6.7); PE
Treatment of donor mice with combined therapy anti-mouse CD25 (PC61.5, 7D4); allophycocyanin anti-rat/mouse Foxp3
staining kit (FJK-16); FITC or PECy5 anti-mouse CD3e (145-2C11);
Donor B6 mice received 50 mg F1 (BALB/c 3 B6) spleen protein ex- biotinylated anti-mouse CD11c (HL3); FITC anti-mouse CD11b (M1/70);
tract diluted in 100 ml PBS, by gavage, daily for 5 d. During this period, PE, PercP-Cy5.5, allophycocyanin, or FITC anti-mouse CD19 (1D3); PE
ad libitum doses of L. lactis NCDO2118 were offered in replacement anti-mouse B220 (RA3-6B2); FITC anti-mouse IgM (eB121-15F9); allo-
of drinking water. Four days later, the animals were immunized with phycocyanin anti-mouse IgD (11-26c [11-26]); FITC anti-mouse CD5 (53-
106 F1 splenocytes i.p. (31), and donor splenic cells were collected 1 wk 7.3); PE anti-mouse CD1d (1B1); allophycocyanin-eFluor 780 anti-mouse
after. CD21 (eBio8D9); PE anti-mouse CD43 (eBioR2/60); allophycocyanin
1930 ORAL THERAPY INHIBITS aGVHD BUT NOT GVL REACTION

anti-mouse CD40 (1C10); FITC anti-mouse CD86 (GL1); biotinylated therapy is used, an elevated survival rate was observed. Also, eval-
anti-mouse B7-H2 (HK5.3); allophycocyanin anti-mouse I-A/I-E (M5/ uation of clinical parameters reveals an important decrease in dis-
114.15.2); biotinylated or allophycocyanin anti-mouse H-2Kb (AF6-
88.5.5.3); PE anti-mouse H-2Kd (SF1-1.1.1) and FITC, PE, PerCP, or
ease severity when compared with all others groups tested (Fig. 1C,
allophycocyanin streptavidins were all purchased from eBioscience (San Supplemental Fig. 1). Donor treatment based on gavage of recipient
Diego, CA). PE anti-mouse latency-associated polypeptide (LAP) (TW7- protein Ags (AgF1) or L. lactis administration ad libitum (Llactis),
16B4) was purchased from BioLegend (San Diego, CA). used separately, shows only a discrete protection (Fig. 1, Supple-
Statistical analysis mental Fig. 1).
To confirm the reduction in aGVHD, we examined histopath-
For grouped analysis, one-way or two-way ANOVA with Bonferroni ologically the skin, colon, and liver of recipient mice 21 d after
posttest were used. Unpaired Student t test was applied for statistical
differences between two groups. Survival data were analyzed with log-rank BMT. The pathologic scores were significantly lower in recipi-
(Mantel–Cox) test. Error bars represent SD. Values of p , 0.05, , 0.01, ents transplanted with spleen cells from treated donors (AgF1/Ll

Downloaded from http://www.jimmunol.org/ at Univ of California-San Diego Serials/Biomed Lib 0699 on September 15, 2014
and , 0.001 are referred to in the figure legends with symbols *, **, and group) when compared with those who received spleen cells from
***, respectively. untreated donors (Ctr+) (Fig. 2), AgF1, or L1actis groups (Sup-
plemental Fig. 2). This reduction might be related to a higher IL-10
Results production found on affected tissues at that same period (Supple-
Treatment of donor mice with combined therapy efficiently mental Fig. 3).
inhibits acute GVHD
Although it has been reported that induction of oral tolerance in the Combined therapy maintains the GVL effect and
recipient can ameliorate aGVHD (12), there are no reports on immunocompetence in the recipient
whether it is possible to induce tolerance to host Ags in the donor The close association between GVHD and the GVL effect reflects
before the transplant is performed. To determine whether donor the need for therapies that target the undesired reaction only (1, 4).
T cells can be rendered tolerant to host tissues through the oral route, To determine whether the combined therapy is able to decrease
donor B6 mice received daily doses of host protein Ags by gavage, GVHD while maintaining the benefic GVL effect, GFP-expressing
whereas L. lactis was offered ad libitum during 5 d (combined P815 mastocytoma cells were injected along with the graft.
therapy). These animals were immunized i.p. with recipient spleen Twenty-five days post-BMT, bone marrow, blood, lymph nodes,
cells after 4 d and 1 wk later used as spleen cell donors along with spleens, and livers of the recipients were examined for the pres-
normal B6 bone marrow to reconstitute lethally irradiated F1 ence of GFP+ cells. As expected by the absence of GVHD, GFP+
(BALB/c 3 B6) mice (Fig. 1A). Fig. 1B shows that when combined cells were found in all tissues analyzed of mice in the negative

FIGURE 1. Donor treatment with combined therapy reduces the clinical signs of acute GVHD. Donor mice (AgF1/Ll) received orally by gavage 50 mg
F1 spleen cell protein extract and ad libitum doses of L. lactis daily for 5 d. Four days later, the animals were immunized with 106 F1 splenic cells i.p. After
6–7 d, BMT was done. Lethally irradiated F1 mice (BALB/c 3 B6) were reconstituted with 5 3 106 bone marrow cells from B6 mice along with 5 3 106
splenic T cells from control B6(Ctr+) or B6 mice orally treated with F1 cell extract (AgF1), with L. lactis (Llactis), or both (AgF1/Ll). (A) Schematic
view of combined therapy treatment. (B) Survival rates. (C) Total clinical score. Data are representative of two experiments with five to eight mice per
group. +p , 0.05, +++p , 0.001 for AgF1/Ll versus Latis group. ***p , 0.001 for AgF1/Ll versus Ctr+ group. ###p , 0.001 for AgF1/Ll versus AgF1
group.
The Journal of Immunology 1931

Downloaded from http://www.jimmunol.org/ at Univ of California-San Diego Serials/Biomed Lib 0699 on September 15, 2014
FIGURE 2. Donor treatment with combined therapy reduces the histopathological manifestations of acute GVHD. Donor mice treatment with combined
therapy and HSCT were performed as described in Fig. 1. Representative micrographs of skin (A), colon (C), and liver (E) from negative control disease
group (left panels), positive control disease group (middle panels), and experimental AgF1/Ll group (right panels) are shown. H&E staining. Total his-
topathological score of skin (B), colon (D), and liver (F). Data were combined from different experiments with 14–18 mice. *p , 0.05, ***p , 0.001.

control disease group. In contrast, in the positive control disease The protected chimeras specifically rejected the unrelated skin
group, where GVHD developed, no GFP+ cells were observed graft but accepted the BALB/c graft (Supplemental Fig. 4A).
(Fig. 3A). However, despite being protected from aGVHD, mice However, after 1 y, we cannot exclude the participation of newly
in the experimental AgF1/Ll group were capable of eliminating generated T cells because the thymic function of the chimeras, at
the tumors cells from all tissues. The protected chimeras remained that posttransplant period, already had been restored and the newly
free of tumor for .15 wk post-BMT (Fig. 3B), whereas chimeras emergent T cells are tolerant to the BALB/c Ags. To test the
from negative and positive control disease groups died with clear specificity of the tolerance induced for host Ags, B6 mice treated or
signals of tumor growth and GVHD development, respectively. not with the combined therapy received skin grafts from third-
Because the therapy proposed in this study combines two treat- party and from F1 donors. As shown in Supplemental Fig. 4B,
ments with immunomodulatory effects, the protection observed both treated or untreated B6 recipients rejected the skin grafts.
could be a result of a general state of immunosuppression that However, when female B6 mice were treated with the combined
would interfere with recipient response to other Ags. To test therapy using male B6 splenic protein extract on gavage, they did
whether the immunosuppression induced by combined therapy not reject male skin graft unlike the untreated females (Supple-
was specific for host Ags, recipients protected for .1 y post-BMT mental Fig. 4C). It suggests that combined therapy induces a more
received skin grafts from third-party and from BALB/c donors. effective tolerance to minor rather than major histocompatibility

FIGURE 3. Combined therapy preserves the graft versus tumor effect. Donor mice treatment with combined therapy and HSCT were performed as
described previously. Along with hematopoietic stem cells, 105 mastocitoma tumor cells (P815) expressing GFP were injected. (A) Twenty-five days
posttransplant, spleen, liver, bone marrow (BM), total lymph nodes (tLn), and blood from the chimeras were analyzed by flow cytometry for the presence of
GFP+ cells. (B) Survival rate of chimeras that also received tumor cells. Data are representative of two experiments, with five mice individually analyzed per
group. ***p , 0.001 for AgF1/Ll (+P815) versus Ctr2 (+P815) group. ###p , 0.001 for AgF1/Ll (+P815) versus Ctr+ (+P815) group.
1932 ORAL THERAPY INHIBITS aGVHD BUT NOT GVL REACTION

Ags, which may explain how it is able to protect from GVHD pre- combined therapy protocol. Spleen cells from combined therapy–
serving the GVL effect. treated donors were depleted of B lymphocytes before transplan-
Altogether, the data show that treatment of donor mice with com- tation. As indicated in Fig. 5A, elimination of B cells completely
bined therapy efficiently reduces clinical and pathological aGVHD abrogated the protection from aGVHD, leading to a higher total
development on a specific and long-lasting fashion, without immuno- clinical score and an elevated mortality rate in this group (AgF1/
suppressing the recipient and preserving the GVL effect. Ll [-LØB]) when compared with non-B cell–depleted one (AgF1/
Ll). Because the role of B cells in GVHD pathophysiology is still
Protection against aGVHD by the combined therapy is not
controversial (37), one may argue that loss of protection could be
mediated by donor Foxp3+ Tregs
related not only to the absence of B cells tolerized by treatment
The phenomenon of oral tolerance can occur by different mech- but also being exacerbated by the absence of B lymphocytes at all.
anisms being the expansion of regulatory-suppressive T cells gen- To overcome this issue, spleen cells from combined therapy–

Downloaded from http://www.jimmunol.org/ at Univ of California-San Diego Serials/Biomed Lib 0699 on September 15, 2014
erally related to repeated lower doses of Ag (5, 6). To investigate the treated donors depleted of B cells were reconstituted with purified
mechanism by which the combined therapy exerts the protection splenic CD19+ B cells from untreated donors. The reconstitution
against aGVHD, spleen cells from treated and untreated donors had no effect, and the recipients still developed aGVHD. However,
were analyzed. No significant differences were observed in absolute when CD19+ B cells from a treated donor were used to recon-
number or frequency of splenic cell subpopulations, including stitute the combined therapy B-depleted spleen, protection was
Foxp3+ T cells (Fig. 4A, 4B). To further exclude the participation of completely restored (Fig. 5A).
donor Tregs, CD25+ T cells were depleted from spleen cells of We next tested whether tolerized B cells were sufficient to inhibit
treated donors before transplantation. Depletion of donor Tregs did aGVHD by transferring CD19+ B cells from a treated donor along
not abrogate the protection, and the elevated survival rate was with spleen cells (and bone marrow) from a control untreated B6
maintained (Fig. 4C, 4D), confirming that these cells are not nec- donor (Fig. 5B). Of interest, depletion of B lymphocytes from
essary for the protective effect. untreated donor spleen cells ameliorated the GVHD, suggesting
that B cells from untreated mice, at least in this model, have a
Donor B cells tolerized by the combined therapy are
pathogenic role in GVHD development. Taken together, the data
responsible for GVHD protection
show that protection of recipient mice against aGVHD is mediated
Several recent studies in both mice and humans have showed the by B lymphocytes with regulatory functions generated by the
participation of B cell on immunoregulation and protection against combined therapy treatment of donor.
acquired inflammatory syndromes (35). In models of oral tolerance
induction, B cells seem to play an important role in tolerance Oral administration of recipient protein Ag with L. lactis
induced with low doses of Ag (36). Because the combined therapy induces an increase in splenic B cells compatible with Bregs
is based on induction of oral tolerance with small amounts of To better characterize the B cells involved in the regulatory activity
recipient Ag, we evaluated the participation of donor B cells in the over aGVHD development, donor mice were treated or not with the

FIGURE 4. Depletion of donor CD25+ T cells before transplantation does not affect the protective effect. Donor mice were treated with combined
therapy as described previously. (A and B) Evaluation of leukocyte populations in treated donor spleens. Splenic cells were analyzed 6 d after the end of the
oral therapy. Nontreated mice were used as controls. (A) Frequency of spleen cell subpopulations. (B) Absolute number of spleen cell subpopulations.
(C and D) Analysis of GVHD development in absence of donor Tregs. Before transplantation, CD25+ T cells were depleted from combined therapy–treated
donor splenocytes. (C) Total clinical score. (D) Survival rates. Data are representative of five pools of three mice per group in (A) and (B), and a total of
10 mice was individually analyzed per group in (C) and (D). ***p , 0.001 for AgF1/Ll (2CD25) versus Ctr+. #p , 0.05, ##p , 0.01, ###p , 0.001 for
AgF1/Ll versus Ctr+.
The Journal of Immunology 1933

Downloaded from http://www.jimmunol.org/ at Univ of California-San Diego Serials/Biomed Lib 0699 on September 15, 2014
FIGURE 5. Protection of recipient mice against GVHD is dependent on donor B cells generated by combined therapy. Donor mice were treated with the
combined therapy as described previously. Before transplantation, B cells were depleted from combined therapy treated or untreated donor spleen. (A) Total
clinical score and survival rates of mice reconstituted with untouched splenocytes from treated donors (AgF1/Ll group), splenocytes depleted of B cells
(AgF1/Ll[2LøB] group), splenocytes depleted of B cells, and reconstituted with B cells from treated (AgF1/Ll[2LøB]+[LøBAg/Ll] group) or untreated
donors (AgF1/Ll[2LøB]+[LøB B6] group). (B) Total clinical score and survival rates of mice reconstituted with untouched splenocytes from untreated
donors added (Ctr+[2LøB]+[LøBAg/Ll]) or not with B cells from treated donors (Ctr+) and spleen depleted of B cells (Ctr+[2LøB]). Data are repre-
sentative of two experiments with 6–10 mice/group. *p , 0.05, ***p , 0.001 for (AgF1/Ll) versus (AgF1/Ll[2LøB]). #p , 0.05, ##p , 0.01, ###p , 0.001
for (AgF1/Ll) versus (AgF1/Ll[2LøB] + [LøB B6]) in (A). *p , 0.05, **p , 0.01, ***p , 0.001 for (Ctr+) versus (Ctr+[2LøB] + [LøBAg/Ll]). #p , 0.05,
###
p , 0.001 (Ctr+) versus (Ctr+[2LøB]) in (B).

combined therapy and their splenic B cells analyzed by flow Convincing data have attributed the suppressive capacity of
cytometry. Bregs to their IL-10 production (41–43). To check whether the
When the B lymphocytes were grouped according to the ex- protection observed in this study is due to B cell–derived IL-10,
pression of IgM and IgD, we found a higher frequency of transitional CD19+ B cells from B6 IL-10 knockout mice treated with the
B cells (CD19+IgM+IgDlo) in treated animals. No differences were combined therapy were transferred to GVHD developing mice
observed among the immature (CD19+IgMhiIgD2) and mature (irradiated F1 receiving spleen and bone marrow cells from control
(CD19+IgMloIgD+) B cells subpopulations (Fig. 6A), suggesting that B6 mice). The results showed that B cells from IL-10–deficient
combined therapy induced an increase in transitional Bregs. How- mice were not able to protect the recipient from GVHD devel-
ever, several different phenotypes have been attributed to B cells opment, unlike B cells from IL-10–sufficient donors pretreated
with regulatory functions and at least two subpopulations were de- with the combined therapy (Fig. 7A, 7B).
scribed: transitional Breg, mentioned above, and the B10 cells (37). Altogether, the above results indicate that the protection ob-
In common, both express CD21 and CD1d molecules, being that the served after oral combined therapy administration depends on IL-
B10 is also positive for CD5 (38). Despite the increase in overall 10–producing Bregs.
CD21 expression in splenic B cells from treated donors (Fig. 6B),
we observed that CD19+CD21+CD1d+ and CD19+CD21+CD1d+ Splenic B cells from combined therapy–treated donor mice
CD5+ subpopulations were reduced on these animals (Fig. 6E, 6F). induce Foxp3+ Tregs on recipient mice
Although there is not a unique marker, or set of markers, that A growing body of evidence has demonstrated the involvement of
exclusively identifies regulatory B cells (Bregs), expression of Bregs in the generation or recruitment of regulatory T lymphocytes
molecules such as CD86 (39) and class II MHC (40) are necessary (41, 44). To test whether B cells from combined therapy–treated
for most of their regulatory functions, regardless of the specific donors were inducing Tregs, chimeras transplanted with control
B cell immunophenotype. In fact, splenic B lymphocytes from spleen cells or spleens depleted of B lymphocytes from both,
combined therapy–treated donor express high amounts of both treated or untreated donors, were evaluated for the presence of
molecules (Fig. 6C, 6D). donor regulatory Foxp3+ cells in spleens and mesenteric and pe-
Recently, B cells with strong suppressive functions in vitro and ripheral lymph nodes 4 d post-BMT. As shown in Fig. 8A and 8B,
in vivo were generated by the incubation of naive B cells with recipients reconstituted with spleen cells from treated donors had
a relevant Ag conjugated to cholera toxin B subunit. In this sce- higher frequencies and absolute numbers of Foxp3+ cells in the
nario, tolerance was shown to be at least, partially dependent on spleen and lymph nodes. In addition, depletion of B lymphocytes
B cell expressing the LAP/TGF-b (41). On the basis of these data, from the graft completely abrogated this increase. Of note, the
we looked at LAP expression in CD19+ cells and found a higher increase of donor Foxp3+ cells happens in the recipient mice,
expression of this molecule in B cells from treated when com- indicating that the regulatory phenotype is induced during the
pared with untreated mice (Fig. 6G, 6H). alloresponse in the host and not before because spleen cells from
1934 ORAL THERAPY INHIBITS aGVHD BUT NOT GVL REACTION

Downloaded from http://www.jimmunol.org/ at Univ of California-San Diego Serials/Biomed Lib 0699 on September 15, 2014
FIGURE 6. B cells generated by combined therapy in the donors are phenotypically compatible with Bregs. Donor mice were treated with combined
therapy as previously described, and splenocytes were analyzed at the end of treatment. (A) Frequency of immature (CD19+IgMhiIgD2), transitional
(CD19+IgM+IgDlo), and mature (CD19+IgMloIgD+) B cells. (B) CD21 expression on CD19+ B cells (mean fluorescence intensity [MFI]). (C) CD86
expression on CD19+ B cells (MFI). (D) Class II MHC expression on CD19+ B cells (MFI). (E) Frequency of splenic CD19+CD21+CD1d+ subpopulation.
(F) Frequency of splenic CD19+CD21+CD1d+CD5+ subpopulation. (G) Frequency of LAP+ B cells. (H) Quantification of (G). Data are representative of
10 mice individually analyzed per group. *p , 0.05, **p , 0.01, ***p , 0.001.

donor mice do not have higher numbers of Tregs nor do the spleen the immune system by induction of oral tolerance (12) and ma-
cells depend on the Tregs to exert their inhibitory effect (Fig. 4). nipulation of gut microbiota by ingestion of probiotic bacteria (30)
Reinforcing these data, in vivo depletion of CD25+cells from re- represent rationally viable and noninvasive therapies to reduce the
cipient reconstituted with combined therapy–treated donors totally development of GVHD. Nonetheless, their clinical application has
abrogated the protection (Fig. 9A, 9B). been limited because the HSCT recipients are subjected to con-
Taken together, the results show that tolerized B cells from the ditioning regimens that require a strict diet in which live or raw
donors mediate the protection against GVHD by inducing Foxp3+ food is precluded.
cells in the transplant recipients. In this study, we demonstrate that treatment of donor mice with
combined therapy reduces clinical and pathological manifestations
Discussion of aGVHD, improving the survival rates to 100%. This phenom-
HSCT is an immunotherapy for treatment of malignant and non- enon is specific and long lasting, and it is not a result of a non-
malignant diseases that is seriously limited by GVHD, an immune specific immunosuppressive milieu. Maintenance of beneficial
syndrome with high morbidity and mortality rates (1–3). Pro- GVL effect is as important as the reduction of GVHD when HSCT
phylaxis and treatment of GVHD is based on nonspecific immu- is used to treat a malignant disease (45–47). In humans, although it
nosupressive agents that, among other complications, eliminate has been suggested that GVL and GVHD are temporally separate
GVLr, which elevates the risk of tumor relapse (4). Education of (48) and that the severity of GVHD does not implicate in im-

FIGURE 7. Protection of recipient mice against GVHD is dependent on IL-10 produced by tolerized donor B cells. Lethally irradiated F1 mice (BALB/c 3
B6) were reconstituted with 5 3 106 bone marrow cells from B6 mice along with 5 3 106 splenic T cells from control B6 (Ctr+). Splenic CD19+ cells (LøB)
from untreated mice (Ctr+LøB B6), from combined therapy treated wild-type (Ctr+LøB AgF1/Ll) or IL-10–deficient (Ctr+LøB IL10KO) donors were trans-
ferred along with the bone marrow and spleen cells to assess the role of IL-10 in GVHD protection. (A) Total clinical score. (B) Survival rates. A total of
10 mice was individually analyzed per group. *p , 0.05, **p , 0.01, ***p , 0.001. *(Ctr+) versus (Ctr+LøB AgF1/Ll).
The Journal of Immunology 1935

Downloaded from http://www.jimmunol.org/ at Univ of California-San Diego Serials/Biomed Lib 0699 on September 15, 2014
FIGURE 8. B cells generated by the combined therapy increase the frequency of donor Foxp3+ cells in the recipient mice. Donor mice were treated with
the combined therapy as described previously. Before transplantation, B cells were depleted from combined therapy treated or untreated donor spleen. Four
days posttransplant, the recipients were euthanized, and the presence of H-2Kb+H-2Kd2Foxp3+ was analyzed in the spleen and mesenteric and peripheral
lymph nodes. Representative data of Foxp3+ cells on peripheral and mesenterical lynph nodes (A) and spleen (C) gated on H-2Kb+H-2Kd2 cells. Frequency
and absolute number of H-2Kb+H-2Kd2Foxp3+ in peripheral or mesenteric lymph nodes (B) and in the spleen (D). Data are representative of the individual
analysis of six to eight mice per group for the spleen and analysis of three to four pools of five animals for lymph nodes. *p , 0.05, **p , 0.01, ***p , 0.001.

proved GVL effect (49), both reactions are clinically associated tained. This dual capacity may be related with the fact that
(4), and there is no specific therapeutic approach to target the combined therapy induces a more effective tolerance to minor
undesired reaction only. In our approach, recipients reconstituted histocompatibility Ags. Actually, it is expected because the Ags
with spleen cells from combined therapy–treated donors develop are administered in the form of soluble proteins that will be
less severe aGVHD and are still capable of eliminating tumor cells captured and processed, losing the entire MHC peptide com-
injected along with the graft, showing that the GVLr is main- plexes.

FIGURE 9. Posttransplant in vivo depletion of CD25+ T cells abrogates the protection. Donor mice treatment with combined therapy and HSCT was
performed as described previously. On third day posttransplant, chimeras reconstituted with combined therapy treated donor spleen received an i.p. dose of
PC61 mAb (250 mg/mice) (54) for depletion of CD25+ cells. On day 14 posttransplant, a reinforced i.p. dose of PC61 mAb was injected in recipient
chimeras (100 mg/mice). (A) Total clinical score. (B) Survival rates. A total of 10 mice was individually analyzed per group. ***p , 0.001 for AgF1/Ll
versus AgF1/Ll (2CD25). ##p , 0.01, ###p , 0.001 for AgF1/Ll versus Ctr+. ++p , 0.01 for AgF1/Ll (2CD25+) versus Ctr+.
1936 ORAL THERAPY INHIBITS aGVHD BUT NOT GVL REACTION

Recently, B lymphocytes subsets able to modulate inflammatory generating Tregs. Activity of these alloreactive Tregs would dampen
responses were described previously (35). These Bregs seem to the response to recipient Ags but preserve the response to Ags de-
play an essential role in oral tolerance induced with low amounts rived from the tumor cells (51, 52).
of Ags (36). In accordance to that, protection obtained with com- Our results suggest that oral tolerance induction by the use of
bined therapy is totally abrogated in the absence of B lymphocytes. a probiotic bacteria and the recipient protein extract, on HSCT
Moreover, CD19+ cells from treated donors, but not from untreated donors before transplantation, might prove safe, simple, and ef-
ones, are sufficient to inhibit aGVHD in recipients reconstituted with fective for preventing GVHD in human patients.
control spleen cells, showing that combined therapy induces the
generation of a B cell subpopulation with regulatory functions.
Acknowledgments
Generation of specific Foxp3+ Tregs by allogeneic primary
We thank Ligia Peçanha for critical reading of the manuscript and sugges-
B cells (50) and by LAP/TGF-b+ B cells (41) have been reported

Downloaded from http://www.jimmunol.org/ at Univ of California-San Diego Serials/Biomed Lib 0699 on September 15, 2014
tions, João P. Monteiro for discussions and critical reading and Jane Littrell
previously. Indeed, our data show that combined therapy not only for English review.
increases the CD19+LAP+ population but also induces donor
Foxp3+ Tregs. Interestingly, in our model, the expansion of Tregs
Disclosures
did not occur in treated donors because we did not observe dif-
The authors have no financial conflicts of interest.
ferences in frequency or absolute number of donor splenic Foxp3+
cells. Moreover, elimination of CD25+ population in the graft
before the transplant does not abrogate the protective effect from References
combined therapy. The induction of donor Foxp3+ cells, possibly 1. Blazar, B. R., W. J. Murphy, and M. Abedi. 2012. Advances in graft-versus-host
disease biology and therapy. Nat. Rev. Immunol. 12: 443–458.
from effector alloreactive T cells, happens on recipient after the 2. Shlomchik, W. D. 2007. Graft-versus-host disease. Nat. Rev. Immunol. 7: 340–352.
transplant and is dependent on those B cells generated by the 3. Monteiro, J. P., A. Benjamin, E. S. Costa, M. A. Barcinski, and A. Bonomo.
treatment, indicating that Ag-specific presentation and recognition 2005. Normal hematopoiesis is maintained by activated bone marrow CD4+
T cells. Blood 105: 1484–1491.
are important steps for acquisition of a regulatory phenotype by 4. Appelbaum, F. R. 2003. The current status of hematopoietic cell transplantation.
allo-T cells and for their following activation. The abrogation of Annu. Rev. Med. 54: 491–512.
protection after in vivo depletion of CD25+ cells on recipient 5. Weiner, H. L., and A. M. C. Faria. 2006. Oral tolerance: therapeutic implications
for autoimmune diseases. Clin. Dev. Immunol. 13: 143‑157.
supports this hypothesis. On the basis of these data, we propose 6. Weiner, H. L., A. P. da Cunha, F. Quintana, and H. Wu. 2011. Oral tolerance.
a model in which B cells from orally treated mice present cognate Immunol. Rev. 241: 241–259.
7. Higgins, P. J., and H. L. Weiner. 1988. Suppression of experimental autoimmune
Ags to allospecific responder T cells responsible for the GVH encephalomyelitis by oral administration of myelin basic protein and its frag-
reaction, explaining the Ag-specific suppression toward aGVHD ments. J. Immunol. 140: 440–445.
only. These results are in accordance with previous reports 8. Maron, R., M. Guerau-de-Arellano, X. Zhang, and H. L. Weiner. 2001. Oral
administration of insulin to neonates suppresses spontaneous and cyclophos-
showing that the generation of specific Tregs after allostimulation phamide induced diabetes in the NOD mouse. J. Autoimmun. 16: 21–28.
in vitro inhibits aGVHD and spare GVL/GVT responses in some 9. Verhasselt, V., V. Milcent, J. Cazareth, A. Kanda, S. Fleury, D. Dombrowicz,
experimental models (51, 52). N. Glaichenhaus, and V. Julia. 2008. Breast milk-mediated transfer of an antigen
induces tolerance and protection from allergic asthma. Nat. Med. 14: 170–175.
The heterogeneity among Bregs and the absence of a master 10. Nagler, A., M. Pines, U. Abadi, O. Pappo, M. Zeira, E. Rabbani, D. Engelhardt,
transcriptional factor associated with this subpopulation strongly M. Ohana, N. R. Chowdhury, J. R. Chowdhury, and Y. Ilan. 2000. Oral toler-
ization ameliorates liver disorders associated with chronic graft versus host
suggest that, like their T cell counterpart, they can be divided into disease in mice. Hepatology 31: 641–648.
different regulatory subsets originated from ontogenically distinct 11. Ilan, Y., M. Margalit, M. Ohana, I. Gotsman, E. Rabbani, D. Engelhardt, and
subsets (53). However, some features such as the production of IL- A. Nagler. 2005. Alleviation of chronic GVHD in mice by oral immuner-
egulation toward recipient pretransplant splenocytes does not jeopardize the graft
10 (35) and expression of molecules like CD86 (39) and class II versus leukemia effect. Hum. Immunol. 66: 231–240.
MHC (40) along with an immature phenotype are common among 12. Nagler, A., M. Ohana, R. Alper, V. Doviner, Y. Sherman, E. Rabbani,
the different subsets. In fact, we observed increased IL-10 in D. Engelhardt, and Y. Ilan. 2003. Induction of oral tolerance in bone marrow
transplantation recipients suppresses graft-versus-host disease in a semi-
protected recipients. Moreover, combined therapy induces a high allogeneic mouse model. Bone Marrow Transplant. 32: 363–369.
frequency of transitional B lymphocytes, with high expression of 13. Tejada-Simon, M. V., and J. J. Pestka. 1999. Proinflammatory cytokine and nitric
oxide induction in murine macrophages by cell wall and cytoplasmic extracts of
CD21, CD86, and class II MHC, a phenotype compatible with lactic acid bacteria. J. Food Prot. 62: 1435–1444.
Bregs. In addition, we demonstrated that IL-10 production by 14. Won, T. J., B. Kim, E. S. Oh, J. S. Bang, Y. J. Lee, J. S. Yoo, H. Yu, J. Yoon,
splenic B cells from treated donors is necessary for the protection K. E. Hyung, S. Y. Park, and K. W. Hwang. 2011. Immunomodulatory activity of
Lactobacillus strains isolated from fermented vegetables and infant stool. Can. J.
observed after combined therapy, indicating the B10 cells, the IL- Physiol. Pharmacol. 89: 429–434.
10–producing Breg (42), is in fact the key player in this setting. 15. Round, J. L., and S. K. Mazmanian. 2010. Inducible Foxp3+ regulatory T-cell
The contribution of a distinct cell type in the immediate induction development by a commensal bacterium of the intestinal microbiota. Proc. Natl.
Acad. Sci. USA 107: 12204–12209.
of B cell with regulatory function cannot be excluded, although to 16. Jeon, S. G., H. Kayama, Y. Ueda, T. Takahashi, T. Asahara, H. Tsuji,
our knowledge there is no report showing the dependence of other N. M. Tsuji, H. Kiyono, J. S. Ma, T. Kusu, et al. 2012. Probiotic Bifidobacterium
breve induces IL-10‑producing Tr1 cells in the colon. PLoS Pathog. 8: e1002714
cells, such as dendritic cells, on Breg activation. 10.1371/journal.ppat.1002714.
Finally, B cell differentiation into a regulatory subtype was 17. Neish, A. S., A. T. Gewirtz, H. Zeng, A. N. Young, M. E. Hobert, V. Karmali,
described by Lampropoulou et al. (43) as being dependent on two A. S. Rao, and J. L. Madara. 2000. Prokaryotic regulation of epithelial responses
by inhibition of IkB-a ubiquitination. Science 289: 1560–1563.
signals. A first signal delivered by TLR initiates IL-10 production 18. Rakoff-Nahoum, S., J. Paglino, F. Eslami-Varzaneh, S. Edberg, and
by B cells. In a second phase, engagement of receptors classically R. Medzhitov. 2004. Recognition of commensal microflora by Toll-like receptors
involved with B cell survival and expansion, such as the BCR, is required for intestinal homeostasis. Cell 118: 229–241.
19. Kelly, D., J. I. Campbell, T. P. King, G. Grant, E. A. Jansson, A. G. Coutts,
amplifies the initial population of IL-10–producing B cells, allow- S. Pettersson, and S. Conway. 2004. Commensal anaerobic gut bacteria attenuate
ing the effective suppression. Altogether, these data might support inflammation by regulating nuclear-cytoplasmic shuttling of PPAR-g and RelA.
Nat. Immunol. 5: 104–112.
the idea that the ingestion of L. lactis gives the innate stimulus 20. Iyer, C., A. Kosters, G. Sethi, A. B. Kunnumakkara, B. B. Aggarwal, and
necessary to the acquisition of a regulatory phenotype by the J. Versalovic. 2008. Probiotic Lactobacillus reuteri promotes TNF-induced
B cells, which is dependent on IL-10 in our model. The recipient apoptosis in human myeloid leukemia-derived cells by modulation of NF-kB
and MAPK signalling. Cell. Microbiol. 10: 1442–1452.
Ags in the protein extract would then provide BCR stimuli that 21. van Baarlen, P., F. J. Troost, S. van Hemert, C. van der Meer, W. M. de Vos,
would enable specific B cells to present the cognate Ags to T cells, P. J. de Groot, G. J. Hooiveld, R. J. Brummer, and M. Kleerebezem. 2009.
The Journal of Immunology 1937

Differential NF-kB pathways induction by Lactobacillus plantarum in the du- 37. Shimabukuro-Vornhagen, A., M. J. Hallek, R. F. Storb, and M. S. von Bergwelt-
odenum of healthy humans correlating with immune tolerance. Proc. Natl. Acad. Baildon. 2009. The role of B cells in the pathogenesis of graft-versus-host dis-
Sci. USA 106: 2371–2376. ease. Blood 114: 4919–4927.
22. Garcı́a-Lafuente, A., M. Antolı́n, F. Guarner, E. Crespo, and J. R. Malagelada. 38. Mauri, C., and P. A. Blair. 2010. Regulatory B cells in autoimmunity: devel-
2001. Modulation of colonic barrier function by the composition of the com- opments and controversies. Nat Rev Rheumatol 6: 636–643.
mensal flora in the rat. Gut 48: 503–507. 39. Mann, M. K., K. Maresz, L. P. Shriver, Y. Tan, and B. N. Dittel. 2007. B cell
23. Madsen, K., A. Cornish, P. Soper, C. McKaigney, H. Jijon, C. Yachimec, regulation of CD4+CD25+ T regulatory cells and IL-10 via B7 is essential for
J. Doyle, L. Jewell, and C. De Simone. 2001. Probiotic bacteria enhance murine recovery from experimental autoimmune encephalomyelitis. J. Immunol. 178:
and human intestinal epithelial barrier function. Gastroenterology 121: 580–591. 3447–3456.
24. Mennigen, R., K. Nolte, E. Rijcken, M. Utech, B. Loeffler, N. Senninger, and 40. Saraiva, M., and A. O’Garra. 2010. The regulation of IL-10 production by im-
M. Bruewer. 2009. Probiotic mixture VSL#3 protects the epithelial barrier by mune cells. Nat. Rev. Immunol. 10: 170–181.
maintaining tight junction protein expression and preventing apoptosis in a mu- 41. Sun, J. B., C. F. Flach, C. Czerkinsky, and J. Holmgren. 2008. B lymphocytes
rine model of colitis. Am. J. Physiol. Gastrointest. Liver Physiol. 296: G1140– promote expansion of regulatory T cells in oral tolerance: powerful induction by
G1149. antigen coupled to cholera toxin B subunit. J. Immunol. 181: 8278–8287.

Downloaded from http://www.jimmunol.org/ at Univ of California-San Diego Serials/Biomed Lib 0699 on September 15, 2014
25. van Bekkum, D. W., J. Roodenburg, P. J. Heidt, and D. van der Waaij. 1974. 42. Vitale, G., F. Mion, and C. Pucillo. 2010. Regulatory B cells: evidence, devel-
Mitigation of secondary disease of allogeneic mouse radiation chimeras by opmental origin and population diversity. Mol. Immunol. 48: 1–8.
modification of the intestinal microflora. J. Natl. Cancer Inst. 52: 401–404. 43. Lampropoulou, V., K. Hoehlig, T. Roch, P. Neves, E. Calderón Gómez,
26. Vossen, J. M., P. J. Heidt, H. van den Berg, E. J. Gerritsen, J. Hermans, and C. H. Sweenie, Y. Hao, A. A. Freitas, U. Steinhoff, S. M. Anderton, and
L. J. Dooren. 1990. Prevention of infection and graft-versus-host disease by S. Fillatreau. 2008. TLR-activated B cells suppress T cell-mediated autoimmu-
suppression of intestinal microflora in children treated with allogeneic bone nity. J. Immunol. 180: 4763–4773.
marrow transplantation. Eur. J. Clin. Microbiol. Infect. Dis. 9: 14–23. 44. Sun, J. B., C. Czerkinsky, and J. Holmgren. 2012. B lymphocytes treated in vitro
27. Hill, G. R., J. M. Crawford, K. R. Cooke, Y. S. Brinson, L. Pan, and J. L. Ferrara. with antigen coupled to cholera toxin B subunit induce antigen-specific Foxp3+
1997. Total body irradiation and acute graft-versus-host disease: the role of regulatory T cells and protect against experimental autoimmune encephalomy-
gastrointestinal damage and inflammatory cytokines. Blood 90: 3204–3213. elitis. J. Immunol. 188: 1686–1697.
28. Storb, R., R. L. Prentice, C. D. Buckner, R. A. Clift, F. Appelbaum, J. Deeg, 45. Reddy, P., T. Teshima, G. Hildebrandt, U. Duffner, Y. Maeda, K. R. Cooke, and
K. Doney, J. A. Hansen, M. Mason, J. E. Sanders, et al. 1983. Graft-versus-host J. L. Ferrara. 2002. Interleukin 18 preserves a perforin-dependent graft-versus-
disease and survival in patients with aplastic anemia treated by marrow grafts leukemia effect after allogeneic bone marrow transplantation. Blood 100: 3429–3431.
from HLA-identical siblings: beneficial effect of a protective environment. 46. Hill, G. R., T. Teshima, A. Gerbitz, L. Pan, K. R. Cooke, Y. S. Brinson,
N. Engl. J. Med. 308: 302–307. J. M. Crawford, and J. L. Ferrara. 1999. Differential roles of IL-1 and TNF-a on
29. Jenq, R. R., C. Ubeda, Y. Taur, C. C. Menezes, R. Khanin, J. A. Dudakov, C. Liu, graft-versus-host disease and graft versus leukemia. J. Clin. Invest. 104: 459–
M. L. West, N. V. Singer, M. J. Equinda, et al. 2012. Regulation of intestinal 467.
inflammation by microbiota following allogeneic bone marrow transplantation. 47. Hanash, A. M., L. W. Kappel, N. L. Yim, R. A. Nejat, G. L. Goldberg,
J. Exp. Med. 209: 903–911. O. M. Smith, U. K. Rao, L. Dykstra, I. K. Na, A. M. Holland, et al. 2011.
30. Gerbitz, A., M. Schultz, A. Wilke, H. J. Linde, J. Schölmerich, R. Andreesen, Abrogation of donor T-cell IL-21 signaling leads to tissue-specific modulation of
and E. Holler. 2004. Probiotic effects on experimental graft-versus-host disease: immunity and separation of GVHD from GVL. Blood 118: 446–455.
let them eat yogurt. Blood 103: 4365–4367. 48. Hari, P., B. Logan, and W. R. Drobyski. 2004. Temporal discordance between
31. Russo, M., M. A. Nahori, J. Lefort, E. Gomes, A. de Castro Keller, D. Rodriguez, graft-versus-leukemia and graft-versus-host responses: a strategy for the sepa-
O. G. Ribeiro, S. Adriouch, V. Gallois, A. M. de Faria, and B. B. Vargaftig. 2001. ration of graft-versus-leukemia/graft-versus-host reactivity? Biol. Blood Marrow
Suppression of asthma-like responses in different mouse strains by oral toler- Transplant. 10: 743–747.
ance. Am. J. Respir. Cell Mol. Biol. 24: 518–526. 49. Weisdorf, D. J. 2010. How closely related is graft-vs-leukemia to donor/recipient
32. Vasconcelos, Z. F., B. M. Dos Santos, J. Farache, T. S. Palmeira, R. B. Areal, disparity? Best Pract. Res. Clin. Haematol. 23: 525–528.
J. M. Cunha, M. A. Barcinski, and A. Bonomo. 2006. G-CSF‑treated gran- 50. Chen, X., and P. E. Jensen. 2007. Cutting edge: primary B lymphocytes pref-
ulocytes inhibit acute graft-versus-host disease. Blood 107: 2192–2199. erentially expand allogeneic FoxP3+ CD4 T cells. J. Immunol. 179: 2046–2050.
33. Cooke, K. R., L. Kobzik, T. R. Martin, J. Brewer, J. Delmonte, Jr., 51. Trenado, A., F. Charlotte, S. Fisson, M. Yagello, D. Klatzmann, B. L. Salomon,
J. M. Crawford, and J. L. Ferrara. 1996. An experimental model of idiopathic and J. L. Cohen. 2003. Recipient-type specific CD4+CD25+ regulatory T cells
pneumonia syndrome after bone marrow transplantation. I. The roles of minor H favor immune reconstitution and control graft-versus-host disease while main-
antigens and endotoxin. Blood 88: 3230–3239. taining graft-versus-leukemia. J. Clin. Invest. 112: 1688–1696.
34. Brok, H. P., P. J. Heidt, P. H. van der Meide, C. Zurcher, and J. M. Vossen. 1993. 52. Edinger, M., P. Hoffmann, J. Ermann, K. Drago, C. G. Fathman, S. Strober, and
Interferon-g prevents graft-versus-host disease after allogeneic bone marrow R. S. Negrin. 2003. CD4+CD25+ regulatory T cells preserve graft-versus-tumor
transplantation in mice. J. Immunol. 151: 6451–6459. activity while inhibiting graft-versus-host disease after bone marrow transplan-
35. Mauri, C., and A. Bosma. 2012. Immune regulatory function of B cells. Annu. tation. Nat. Med. 9: 1144–1150.
Rev. Immunol. 30: 221–241. 53. Mizoguchi, A., and A. K. Bhan. 2006. A case for regulatory B cells. J. Immunol.
36. Gonnella, P. A., H. P. Waldner, and H. L. Weiner. 2001. B cell-deficient (mu MT) 176: 705–710.
mice have alterations in the cytokine microenvironment of the gut-associated 54. Setiady, Y. Y., J. A. Coccia, and P. U. Park. 2010. In vivo depletion of CD4+
lymphoid tissue (GALT) and a defect in the low dose mechanism of oral tol- FOXP3+ Treg cells by the PC61 anti-CD25 monoclonal antibody is mediated by
erance. J. Immunol. 166: 4456–4464. FcgRIII+ phagocytes. Eur. J. Immunol. 40: 780–786.
The Journal of Immunology

Corrections
Mercadante, A. C. T., S. M. Perobelli, A. P. G. Alves, T. Gonçalves-Silva, W. Mello, A. C. Gomes-Santos, A. Miyoshi, A. M. C. Faria, and
A. Bonomo. 2014. Oral combined therapy with probiotics and alloantigen induces B cell–dependent long-lasting specific tolerance.
J. Immunol. 192: 1928–1937.

The eighth author’s name was omitted from the article. The corrected author and affiliation lines are shown below.

Ana C. T. Mercadante*, Suelen M. Perobelli†, Ana P. G. Alves*, Triciana Gonçalves-Silva†, Wallace Mello‡, Ana C. Gomes-Santosx,
Anderson Miyoshi{, Vasco Azevedo{, Ana M. C. Fariax, and Adriana Bonomo*,†,‡

*Department of Experimental Medicine, Brazilian National Cancer Institute, Rio de Janeiro 20231-050, Brazil; †Department of Im-
munology, Microbiology Institute Prof. Paulo de Góes, Federal University of Rio de Janeiro, Rio de Janeiro 21941-902, Brazil;

Laboratory of Thymus Research, Oswaldo Cruz Institute, Rio de Janeiro 21040-360, Brazil; xDepartment of Biochemistry and Im-
munology, Biological Science Institute, Federal University of Minas Gerais, Belo Horizonte 30161-970, Brazil; and {Department of
Biology, Biological Science Institute, Federal University of Minas Gerais, Belo Horizonte 30161-970, Brazil

www.jimmunol.org/cgi/doi/10.4049/jimmunol.1490007

Copyright Ó 2014 by The American Association of Immunologists, Inc. 0022-1767/14/$16.00

You might also like