You are on page 1of 16

Influence of Dietary Components on Regulatory T Cells

Shohreh Issazadeh-Navikas, Roman Teimer, and Robert Bockermann


Biotech Research and Innovation Centre, University of Copenhagen, Copenhagen, Denmark

Common dietary components including vitamins A and D, omega-3 and probiotics are now widely accepted to be essential
to protect against many diseases with an inflammatory nature. On the other hand, high-fat diets are documented to exert multi-
ple deleterious effects, including fatty liver diseases. Here we discuss the effect of dietary components on regulatory T cell (Treg)
homeostasis, a central element of the immune system to prevent chronic tissue inflammation. Accordingly, evidence on the im-
pact of dietary components on diseases in which Tregs play an influential role will be discussed. We will review chronic tissue-spe-
cific autoimmune and inflammatory conditions such as inflammatory bowel disease, type 1 diabetes mellitus, multiple sclerosis,
rheumatoid arthritis and allergies among chronic diseases where dietary factors could have a direct influence via modulation of
Tregs homeostasis and functions.
Online address: http://www.molmed.org
doi: 10.2119/molmed.2011.00311

INTRODUCTION less, a variety of model systems are avail- the influence of dietary components on
Regulatory T cells (Tregs) are a hetero- able to interrogate the characteristics and Tregs, including probiotics and vitamins,
geneous T-cell subpopulation that regu- function of Tregs in health and disease. are discussed. In addition, related studies
lates the immune system in various Although, few studies directly look at on the pathways involved in the modula-
ways. Given the significance that deregu- the effect of common dietary compo- tion of Tregs by dietary components are
lation of the immune system plays in the nents on Treg homeostasis, despite the also examined. In addition to the role of
development and progression of chronic important role shown for the influence of single components such as vitamins and
inflammatory processes, including in- diet on inflammation and immune regu- the consequences of probiotics, gluten
flammatory bowel disease (IBD), type 1 lation. Accordingly, evidence on the im- and fatty acids are also discussed.
diabetes mellitus (T1D), asthma, arthritis pact of dietary components on diseases
and multiple sclerosis (MS), the factors in which Tregs play an influential role DEFINITION OF Tregs
influencing and influenced by Tregs may may provide insight into the role of these Broadly speaking, Tregs have the capa-
offer insight in the mechanisms underly- dietary components on Tregs themselves. bility to suppress the activity of the im-
ing the pathological course of these dis- This report reviews some of the evi- mune system and to regulate self-toler-
eases as well as provide clues to improv- dence showing how dietary components ance. However, it is not possible to
ing their management. influence Tregs. Although dietary com- unambiguously define Tregs as a homo-
The mechanisms involved in the gen- ponents have been reported to influence geneous group, since T cells of different
eration and maintenance of Tregs on the disease outcome via regulating other phenotypes have been shown to exhibit
cellular level and in the host environ- cells, such as helper T cells (Th1, Th2 and immune regulating potential (1). Cur-
ment are just starting to be understood. Th17), important in health and the rently, the most commonly known regu-
It is even less clear how the environ- pathogenesis of immune-related dis- latory T-cell lineage is called
ment-host interaction affects the immune eases, in this review, we restrict our focus CD4+CD25highFoxP3+ regulatory T cells.
balance and the status of Tregs. Nonethe- to Tregs. Recently published results on The forkhead box P3 (FoxP3) transcrip-
tion factor is the cell lineage–determining
master transcription factor of this T-cell
Address correspondence to Shohreh Issazadeh-Navikas, Biotech/Research and Innova- subtype (2). Generation of Tregs is
tion Centre, University of Copenhagen, Copenhagen Biocenter, Ole Maaløes Vej 5, DK- shown to depend on the cytokine trans-
2200 Copenhagen N, Denmark. Phone: +45-353-25649; Fax: +45-3532-5669; E-mail: forming growth factor (TGF)-β (3). A
shohreh.issazadeh@bric.ku.dk. new population of Tregs has been de-
Submitted August 23, 2011; Accepted for publication October 28, 2011; Epub scribed lately that is under the regulation
(www.molmed.org) ahead of print November 16, 2011. of interleukin (IL)-35 (TR35) (4). Although
the first report showed them to be
FoxP3+, a more recent study suggests
that TR35 cells are independent of FoxP3

M O L M E D 1 8 : 9 5 - 1 1 0 , 2 0 1 2 | I S S A Z A D E H - N AV I K A S E T A L . | 9 5
EFFECT OF DIET ON Treg CELLS

(5). Because not much information exists functional Tregs (14). Thus, it is impor- ulating efficient immune responses to
regarding the influences of dietary prod- tant to distinguish if any substance ad- pathogenic bacteria, oral administration
ucts on generation or function of this ministered orally would lead to nonspe- of proteins leads to a state of peripheral
population, within this report, Tregs are cific general Treg expansion or whether (oral) tolerance to ingested proteins. The
defined as CD4+CD25+FoxP3+ T cells un- dietary components suggested in the lit- maintenance of balance between toler-
less otherwise stated. erature have more specific effects leading ance and immunity in the gut is in part
Tregs have been found to suppress to induction of molecular pathways to dictated by specialized gut-resident DCs.
various immune cells such as CD8+ generate or expand Tregs. DCs in the gut-associated lymphoid tis-
T cells, dendritic cells (DCs), monocytes/ sues are unique in their ability to activate
macrophages, B cells, natural killer cells EFFECTS OF PROBIOTIC BACTERIA ON naive T cells to effector T cells in re-
and natural killer T cells (6). Nonfunc- Tregs sponse to pathogens, but under homeo-
tional mutations in the FOXP3 gene in static conditions, gut DCs seem to pro-
humans cause the immune dysfunction, Probiotic Effects in Digestive Tract mote noninflammatory T-cell responses,
polyendocrinopathy, enteropathy, Disorders mainly composed of Tregs via produc-
X-linked (IPEX) syndrome, indicating an Probiotics have been defined by the tion of IL-10 and TGF-β (21).
essential role of FoxP3+ Tregs in the pre- World Health Organization (WHO) as The importance of Tregs in controlling
vention of hyperimmune responses and “live microorganisms which when ad- gut inflammation has long been under-
autoimmune diseases (7,8). On the other ministered in adequate amounts confer a stood. Depletion of Tregs leads to chronic
hand, Tregs at tumor sites or in the pe- health benefit on the host” (15). The ra- inflammation in the experimental models
ripheral blood are suggested to be partly tionale for beneficial effects of probiotics of colitis. Similarly, the transfer of Tregs
responsible for tumor immune escape, is that manipulation of the gut flora is able to control chronic inflammation in
making the inhibition of Treg function a might have an effect on pathogenic bac- the gut. Loss of Tregs in the gut is ob-
favorable treatment intervention in some teria and might also affect the formation served during the IPEX syndrome, which
cases (9). of disease-controlling Tregs (16,17). leads to the manifestation of intestinal le-
Tregs can be further divided into two IBD, which includes Crohn’s disease sions (22).
groups on the basis of their origin. Natu- and ulcerative colitis, involves a variety It is of interest to mention that gut flora
ral Tregs (nTregs) are generated in the of inflammatory manifestations of the have profound effects on the balance of
thymus through major histocompatibility gut. Existing murine models of IBD sup- FoxP3+ Tregs versus proinflammatory
complex class II–dependent T-cell recep- port the concept that host bacteria play Th17 cells in the lamina propria. When
tor interactions resulting in high-avidity an important role in the disease process. the gut flora were manipulated through
selection (10). nTregs are speculated to Therefore, conditions affecting the ho- cohousing of mice from different suppli-
prevent autoimmunity and to generally meostasis of gut bacteria could have a ers, mice from different sources that had
raise the activation threshold to initiate decisive impact on health versus disease marked differences in their basal levels of
an immune response. Adaptive or in- status. Accordingly, specific pathogen- Th17 cells or were lacking Th17 cells ac-
duced Tregs (iTregs), on the other hand, free conditions are protective against quired Th17 cells after introduction of
develop outside the thymus during sub- gut inflammatory disease (18). How- bacteria from Th17 cell–sufficient mice.
immunogenic antigen presentation. ever, mice in gnotobiotic conditions Of note, differentiation of Th17 cells
iTregs are thought to be essential to (that is, exposed to only certain strains correlated with the presence of commen-
maintain a noninflammatory environ- of bacteria), exposed to Bacteroides vulga- sal microbiota, such as Cytophaga-
ment in the gut, to suppress allergic im- tus, are also protected against Escherichia Flavobacterium-Bacteroidetes bacteria in
mune responses to environmental and coli–induced colitis (19). Additionally, the gut. This result was shown to be inde-
food antigens and to decrease chronic in- administration of probiotics leads to re- pendent of Toll-like receptor (TLR) and
flammation (11,12). The discrimination of duced disease in experimental colitis, IL-21 or IL-23 signaling, but it required
nTregs and iTregs is mentioned here which is accompanied by an increase of appropriate TGF-β activation. Treatment
solely for the sake of completeness and is FoxP3+ Tregs (20). with selective antibiotics inhibited Th17
not discussed further in this review. Because the gut hosts normal and po- differentiation. Of note, the absence of
Nevertheless, it is noteworthy to men- tential pathogenic bacteria, the immune Th17 cell–inducing bacteria was accompa-
tion that oral administration of antigens responses in the intestine are tightly reg- nied by an increase in Foxp3+ Tregs in the
is commonly used to induce peripheral ulated to ensure sustained effector re- lamina propria (23). Furthermore, it is
tolerance to subsequent disease induc- sponses to pathogens and to avoid po- also suggested that Tregs regulate intes-
tion by the same antigens (13). It has tentially deleterious inflammatory tinal inflammation induced by pathogenic
been shown that the ensuing oral toler- responses to commensal bacteria or food bacteria. In agreement, CD25+FoxP3+
ance depends on successful expansion of antigens. Therefore, while the gut is reg- Tregs regulate gastric inflammation and

9 6 | I S S A Z A D E H - N AV I K A S E T A L . | M O L M E D 1 8 : 9 5 - 1 1 0 , 2 0 1 2
INVITED REVIEW ARTICLE

Helicobacter pylori colonization in vivo (24), ous system (CNS). Other studies also reuteri are more potent than Tregs from
whereas commensal-induced Tregs are in- suggest an important role for commensal mice without previous probiotic expo-
volved in protection against pathogen- bacterial antigens, in particular, Bac- sure and could prevent airway inflam-
induced inflammation (25). teroides fragilis expressing polysaccharide mation caused by allergies (37).
Interventions with probiotics have A, in protecting against CNS demyelina- It is suggested that probiotics have di-
been conducted in several models of di- tion in EAE via modulation of Tregs (31). rect effects on Tregs by interfering with
gestive tract disorders and are summa- Similarly, oral treatment of mice with a nuclear factor (NF)-κB degradation. In
rized in Table 1. broad spectrum of antibiotics induced accordance, Lactobacillus plantarum is able
significant changes in Tregs and reduced to block NF-κB degradation via blockage
Probiotic Effects on Extraintestinal the susceptibility to EAE (32). However, of proteasome function. This effect is
Diseases literature regarding effects of probiotics even maintained in bacteria-free condi-
Probiotics, in addition to having a in the treatment of EAE is controversial. tioned medium but containing the TLR2-
local effect on disease models of the di- Some authors have observed either exac- activating molecule lipoteichoic acid
gestive tract, also operate systemically in erbating effects in rat EAE models, with (38,39). Another chemical proteasome
inflammatory models such as collagen- increased Th1 immune responses, or no blocker, benzyloxycarbonyl-isoleucyl-
induced arthritis (CIA) (Table 2), experi- treatment effect after using probiotics glutamyl(O-tert-butyl)-alanyl-leucinal
mental autoimmune encephalomyelitis (33–35). The plausible reasons for such (PSI), was associated with the generation
(EAE) (Table 3), asthma and delayed- controversies will be discussed later. of FoxP3+ Tregs via PSI-treated APCs
type hypersensitivity reactions (Table 4). (40). Additionally, oral application of the
Lactobacillus casei (L. casei), a widely Possible Mechanisms of Probiotic- probiotic strain B. infantis reduced NF-κB
commercialized dairy product, is re- Induced Tolerance activation and increased the numbers of
ported to reduce arthritis in CIA. Admin- Introducing living bacteria into a host FoxP3+ cells in mucosa and spleen, coun-
istration of this product results in lower likely elicits responses on numerous lev- terbalancing the NF-κB–activating prop-
disease scores and less complement- els affecting the immune response. One erties of pathogenic strains such as Sal-
fixing IgGs. Treatment with L. casei of the major mechanisms of action of liv- monella typhimurium (25).
alone had no effect on FoxP3, but an ing bacteria is reported to occur by in- In support of the indirect affects of
increase was detected with a combina- creasing number of Tregs. Others have probiotics in regulation of Tregs, it was
tion treatment of L. casei and oral shown mechanisms by which bacteria di- shown that tolerogenic DCs are playing a
tolerance–inducing collagen type II rectly induce an increase in FoxP3+ Tregs crucial role in conversion of T cells to
(26,27). Collectively, administration of and/or their suppressive function via Tregs. Activation of DCs by certain
L. casei resulted in disease-reducing ef- production of immunomodulatory cy- species of probiotics such as L. reuteri
fects in several experimental models in- tokines. Moreover, there are several re- and L. casei, but not L. plantarum, leads to
cluding CIA (20,28). This result is also re- ports indicating that the major pathway generation of Tregs. This effect is medi-
ported to be associated with an increase of probiotic-induced tolerance seems to ated by the 3-grabbing nonintegrin mole-
in FoxP3+ Tregs (20). operate via the induction of tolerogenic cule on human DCs (41). Although the
L. casei is also shown to exert protec- antigen-presenting cells (APCs). These effector cells showing suppressive capa-
tive effects on T cell–mediated skin in- tolerogenic APCs consequently con- bilities were called Tregs, no direct evi-
flammation, in contact hypersensitivity tribute to generation of Tregs and hence dence for the presence of Treg markers
to the hapten 2-4-dinitrofluorobenzene, a regulation of inflammatory diseases. was reported. Additionally, tolerogenic
model for allergic contact dermatitis (29), In support of the positive influence of DCs were generated in vitro with specific
on dermatitis (20) and on delayed-type oral probiotics on expansion of Tregs, species of lactobacilli. The in vivo transfer
hypersensitivity responses (28), all asso- administration of L. casei to mice af- of these DCs resulted in generation of
ciated with increases in FoxP3+ Tregs. fected the frequency of CD4+FoxP3+ Tregs and reduction of IBD in mice. The
Taking these results together, L. casei ap- Tregs in the skin. Whereas this amelio- protective effect of the probiotic-matured
pears to be a candidate probiotic agent to rated skin inflammation, it did not DCs induced Tregs depended the pattern
regulate Tregs and thereby act as a potent change the extent of in vivo suppressive recognition receptors TLR2 and non-
immunomodulator of T cell–mediated function of nTregs (29). Accordingly, in obese diabetic (NOD)-2 (42). In support
skin allergies. an experimental model of colitis, live of putative influence of probiotics in reg-
Lactobacillus rhamnosus Goldin-Gorbach probiotics as well as their immunomod- ulation of tolerogenic DC-induced Tregs,
and L. casei Shirota have protective effects ulatory DNA resulted in generation of NOD2/CARD15 (nucleotide-binding
on EAE (30), the most commonly used higher numbers of FoxP3+ Tregs (36). oligomerization domain containing 2,
rodent model for MS, an alleged autoim- Moreover, it has also been shown that also known as the caspase recruitment
mune disease affecting the central nerv- Tregs isolated after oral feeding with L. domain family, member 15) also has a di-

M O L M E D 1 8 : 9 5 - 1 1 0 , 2 0 1 2 | I S S A Z A D E H - N AV I K A S E T A L . | 9 7
Table 1. Probiotic effects on Tregs and disease models (digestive tract).

Treg after Treatment effect Investigated


Disease model Bacterial strains used probiotic treatment of probiotics effector mechanisms Reference

Trinitrobenzene sulfonic (TNBS) Mix 1 (Lactobacillus acidophilus FoxP3 ↑in intraepithelial IBD↓ IL-10↑ 126
acid–induced murine colitis and Bifidobacterium longum) lymphocytes TNF-α↓
Mix 2 (Lactobacillus plantarum, FoxP3 → in Lamina propria Monocyte chemotactic
Streptococcus thermophilus, protein-1 (MCP1)↓
and Bifidobacterium animalis
subspecies Lactis)
TNBS-induced colitis (Balb/c) DCs pulsed with: FoxP3→ IBD↓ indoleamine 42
L. salivarius 2,3-dioxygenase (IDO)↑
L. rhamnosus IL-10↓ (IL-10–independent)
EFFECT OF DIET ON Treg CELLS

TLR2-, NOD2- and


MyD88-dependent
TNBS-induced colitis (SLJ mice) VSL#3 ND IBD↓ IL-10↑ 127
IFN-γ ↓
Leukocyte alkaline
phosphatase+ (LAP+)
cells↑
Pouchitis disease in ulcerative VSL#3 FoxP3↑ Pouchitis↓ IL-1β↓ 128
colitis (humans)
CD4 + CD62L + T-cell–induced CpG FoxP3↑ IBD↓ IL-10↑ 48
IBD in CB17-Prkdcscid IFN-γ↓
129 Ola × C57BL/6-IL 10 KO L. salivarius UCC118, ND IBD↓ TGFβ↑ 129

9 8 | I S S A Z A D E H - N AV I K A S E T A L . | M O L M E D 1 8 : 9 5 - 1 1 0 , 2 0 1 2
Rafamycin-resistant subspecies TNF-α↓
(subcutaneous) IL-12↓
Salmonella typhimurium B. infantis 35624 FoxP3↑ Intestinal disease IL-6↓ 25
LPS-induced intestinal score↓ TNF-α↓
inflammation NF-κB activation↓
H. pylori–induced gastritis H. pylori FoxP3↑ Intestinal disease 24
score↑ after
anti-CD25 treatment

ND, not determined; ↑, increased expression; ↓, decreased expression; →, unchanged.


INVITED REVIEW ARTICLE

Table 2. Probiotic effects on Tregs and disease models (arthritis).

Treg after Treatment


probiotic effect Investigated
Disease model Bacterial strains used treatment of probiotics effector mechanisms Reference

Adjuvant and Lactobacillus rhamnosus GG ND Arthritis↓ ND 130


tropomyosin-induced (ATCC 53103)
arthritis in Lewis rats
CIA in Lewis rats L. casei FoxP3→ Arthritis↓ IL-10↑ 27
Proinflammatory molecules↓
CIA in Lewis rats L. casei FoxP3↑ Arthritis↓ TGF-β ↑ 26
(oral tolerance) IL-10↑
Proinflammatory molecules↓
CIA in dark black agouti L. salivarius UCC118, ND Arthritis↓ ND 129
(DBA)/1 mice Rafamycin-resistant subspecies
(subcutaneous)
CIA in DBA/1 mice L. casei Shirota ND Arthritis↓ IFN-γ↓ 28
Anti–collagen type II (aCII) IgG↓
Delayed-type hypersensitivity↓

ND, not determined; ↑, increased expression; ↓, decreased expression; →, unchanged.

rect effect on the survival of Tregs. In human FoxP3+ T cells (43). Consequently, otics, as suggested in a study using
agreement, the bacterial cell wall product NOD2 deficiency in the mouse also exac- NOD2-deficent mice (45). Such a study,
muramyl dipeptide (MDP), which has a erbates graft versus host disease as well extended to humans, raises a question re-
capacity to bind NOD2, is reported to re- as experimental colitis. The NOD2 defi- garding the efficacy of probiotics in pa-
duce Fas-induced Treg apoptosis (43). ciency was also mirrored by a reduced tients and consumers harboring defects
Moreover, a clear deficiency in the quan- number of FoxP3+ Tregs (44). It would in pattern recognition receptors, as seen
tity of FoxP3+ lymphocytes was reported prove fruitful to test the probiotic-NOD2 in disease-associated NOD2 polymor-
in patients with Crohn’s disease, which axis in vivo and its effect on Treg expan- phisms. For example, the limited efficacy
was associated with disease polymor- sion and function. of lactobacilli in Crohn’s disease could be
phisms in the NOD2 gene. Subsequently, Genetic makeup may be important for related to NOD2 deficiency. This hypoth-
it was found or reported that the NOD2 the colonization of the gut with patho- esis was tested in mice, and it was found
ligand, MDP, activates NF-κB in primary genic bacteria and the effect of probi- that Lactobacillus salivarius rescued mice

Table 3. Probiotic effects on Tregs and disease models (EAE/MS).

Treg after Treatment Investigated


probiotic effect effector
Disease model Bacterial strains used treatment of probiotics mechanisms Reference

Spinal cord homogenate/H37Ra/Freund L. casei strain Shirota ND EAE↑


complete adjuvant (CFA)–induced EAE in Body weight↓ ND 34,35
Lewis rats
Spinal cord homogenate/H37Ra/CFA–induced L. casei strain Shirota (LcS) ND EAE→
EAE in Lewis rats (LEW/CrlCrlj) B. breve (BbY) Body weight→ ND 33
EAE in rats (EAE in SLJ mice) L. casei DN 114-001, ND Rat EAE↓
L. rhamnosus GG Mixed effects
L. casei Shirota (no effect in ND 30
mice)
EAE in C57BL/6 L. paracasei DSM 13434, FoxP3↑ in WT EAE↓ IL-10↑ 131
L. plantarum DSM 15312 not in IL-10 TGF-β↑
KO mice

ND, not determined; ↑, increased expression; ↓, decreased expression; →, unchanged.

M O L M E D 1 8 : 9 5 - 1 1 0 , 2 0 1 2 | I S S A Z A D E H - N AV I K A S E T A L . | 9 9
EFFECT OF DIET ON Treg CELLS

Table 4. Probiotic effects on Tregs and disease models (allergy/asthma).

Treg after Treatment Investigated


Bacterial strains probiotic effect effector
Disease Disease model used treatment of probiotics mechanisms Reference

Asthma Ovalbumin (OVA)-induced L. rhamnosus GG FoxP3↑ BAL cells↓ Proliferation↓ 132


asthma model in Balb/c (ATCC 53103) Airway reactivity↓ IL-4, -5, and -10↓
Bifidobacterium IFN-γ↓
lactis (Bb-12) TGF-β→
Anti-OVA Ig↓
OVA-induced asthma L. salivarius FoxP3↑ BAL cells↓ Proliferation↓ 37
model in Balb/c L. reuteri (ATCC (more Airway reactivity↓
23272) potent) IL-10↑ (spleen)
IL-10↓ (BALF)
TGF-β →
Delayed-type OVA and L. casei FoxP3↑ Delayed-type IL-10↑ 29
hypersensitivity 2-4-dinitrofluorobenzene– DN-114 001 hypersensitivity↓
sensitized mice

ND, not determined; ↑, increased expression; ↓, decreased expression; →, unchanged.

from colitis via generation of CD103+ otics, they nonetheless show that bacter- an IL-10–dependent fashion). Accord-
DCs and CD4+FoxP3+ Tregs. This sup- ial components have tolerizing properties ingly, it was reported that L. casei treat-
pressive effect was abolished in NOD2- via induction of tolerogenic DC–medi- ment promoted the activation of antigen-
deficient mice (46). ated Treg cell generation. experienced Tregs and increased their
It is noteworthy to mention that the Additional antiinflammatory mecha- ability to produce IL-10 and hence inhibit
protective effects of probiotics related nism of probiotics could be mediated via skin inflammation (29). Moreover, sup-
to generation of direct Tregs or tolero- the regulation of exogenous bacterial pressive mechanism of Tregs induced by
genic APC-mediated Tregs do not adenosine 5′-triphosphate (ATP) levels as L. casei in vitro was shown to be IL-10 de-
require live bacteria. In agreement, an inflammatory mediator. In support of pendent (41). Nevertheless, L. salivarius
subcutaneous injection of bacterial this, a report described the beneficial role Ls33 induced Tregs, and amelioration of
cytosine–phosphate–guanine (CpG) mo- of exogenous ATP, which increases FoxP3 colitis in mice was correlated with a local
tifs, in the form of synthetic immunos- transcription after A2 adrenergic receptor production of IL-10 (46).
timulatory sequence oligodeoxynu- (A2AR) engagement, also triggering an Although the protective effects of pro-
cleotides (ISS-ODNs), reduced symptoms increase in TGF-β and a decrease of IL-6 biotics by regulating the generation of
in a model of IBD in a TLR9-mediated levels (50). Furthermore, human FoxP3+ Tregs, functions of Tregs and prevention
manner (47). The disease reduction was Tregs are shown to convert ATP via of local inflammation in the gut and gut-
also observed after injection of probiotic CD39 and CD73 to immunosuppressive associated diseases are more rational, it
or E. coli–derived DNA in a murine coli- adenosine (51). In contrast, ATP was re- is an interesting question as to how oral
tis model (36). Similar effects were ported to induce inflammatory Th17 cells administration of probiotics could exert
shown through intraperitoneal injection (52). This finding might shed light on protective effects to modulate inflamma-
of CpG-ODN. Interestingly, this treat- differential effects of probiotics by engag- tory diseases of organs other than the gut
ment induced FoxP3+ Tregs in germ-free ing different subsets of gut-associated (such as the antiinflammatory effects re-
mice, indicating that even without preex- DCs and hence leading to a change in ported in CIA, EAE and allergic airway
istence of bacterial flora, CpG-ODN in- the inflammatory balance depending on inflammation). Such favorable results
duces tolerance, which suggests that the bacterial composition. might not be too surprising in arthritis,
CpG-ODN–induced Tregs are not bacter- Although several different mecha- since it was observed that the gut and
ial antigen specific (48). Particularly, nisms of action have been reported to be joint share lymphocyte homing receptors
CpG-induced CD8+ plasmacytoid DCs attributed to the probiotic-mediated Treg that might redirect regulatory cells from
from the gut have been shown to convert generation, and elevated Treg-suppressive the gut to the joint (53). This scenario is
T cells into Tregs, whereas spleen- functions, there is more consistent agree- interesting and requires further investiga-
derived DCs do not have the same ca- ment on how the probiotic-induced tion to determine if other tissues, such as
pacity (49). Although bacterial CpGs are Tregs exert their antiinflammatory effects brain and skin, share similar Treg homing
not strictly speaking referred to as probi- (that is, via production of IL-10 and or in receptors, allowing gut-generated Tregs

1 0 0 | I S S A Z A D E H - N AV I K A S E T A L . | M O L M E D 1 8 : 9 5 - 1 1 0 , 2 0 1 2
INVITED REVIEW ARTICLE

Table 5. Different mechanisms of regulatory T-cell induction by probiotics.

Direct effects in T cells Reference Indirect effects on T cells Refernce

Higher potency of Tregs 37 Conversion of gut DC to induce Tregs 88


MDP activates NOD2, which prevents Treg from 43 TLR2- and NOD2-dependent induction of maturation 42
Fas-induced apoptosis of tolerogenic DC (IDO)
NOD2 KO mice have increased graft versus host 44 CpG-induced CD8+ plasmacytoid gut DC convert 49
disease and TNBS-induced colitis (less FoxP3) T cells into Tregs
Suppression of bacterial ATP, which prevents 52 CD103+ and CD103– lamina propria DC induce 133
conversion to Th17 cells FoxP3 Tregs
Exogenous ATP activates Treg and FoxP3 expression 50,51 Induction of FoxP3– Tregs by CD103+ ML-DC (ATRA) 134
Proteasome block induces tolerogenic APCs 25,38,39

to home these tissues during the course els discussed (30,35). Such differences the mechanism of action still remains to
of inflammation. Table 5 summarizes could be because of use of different be demonstrated. It will also be impor-
how probiotics directly or indirectly in- strains of probiotics, different doses tant to dissect the quality of a specific
fluence Tregs. and/or different routes of administra- probiotic strain from general properties
tion. Moreover, different genetic back- of bacterial components including DNA
Caveats grounds of the experimental models or cell wall components. In summary, it
Despite the promising signs for benefi- could affect how probiotics exert their seems likely that the generation, survival
cial effects of probiotics, caution is ad- immunomodulatory properties. This re- and homing of FoxP3+ Tregs all play a
vised in light of a study showing that a sult could be influenced by the host’s in- role in probiotic-mediated effects.
commonly used probiotic (Bifidobacterium trinsic immune composition and the pre-
animalis) can actually cause duodenitis dominance of a specific type of immune Vitamin A and D Derivatives in Treg
and mild colonic inflammation in IL-10 response. For example, some strains of Function
knockout mice (54). It is noteworthy to mice are predominantly mounting Th2 Vitamin A (VitA) and vitamin D (VitD)
mention that IL-10 knockout mice are re- type of immune responses, whereas oth- are both inactive precursors of their bio-
ported to develop spontaneous IBD and ers are prone to mount Th1 and/or Th17 logically active derivatives, which need
transfer of Tregs leading to inhibition of type of immune responses. Nevertheless, to be enzymatically modified. These
inflammation. Such findings, extended to the controversies could partly be caused modified bioactive vitamin derivatives
humans, might be especially relevant for by overriding in vitro results by complex- play a fundamental role in many physio-
immunosuppressed or certain IBD pa- ity of in vivo applications. Hence, precau- logical processes such as night vision
tients (55). tions should be considered to avoid gen- and calcium homeostasis. Furthermore,
Furthermore, different strains of lacto- eralization of the protective effects these vitamin derivatives are also in-
bacilli elicit diverse activation of the sup- exerted by probiotics. volved in the regulation of the immune
pressor of cytokine pathway 2 and 3 It might be especially difficult to claim system (61,62) and tissue inflammation
(SOCS2 and SOCS3) on a human gastric a Treg-inducing effect to a unique “probi- (63,64). This section focuses on their role
carcinoma cell line (56). It will be inter- otic” property rather than a general re- in the formation and function of FoxP3+
esting to see if SOCS3 is also activated in sponse toward bacteria. In support of Tregs.
Tregs under these conditions, which this, not only nonpathogenic bacteria
might in this case lead to reduced sup- seem to increase the presence of Tregs. VitA Derivatives and Treg Function
pressor functions as seen during an arti- The expansion of FoxP3+ Tregs also The fat-soluble VitA, found in abun-
ficial SOCS3 increase (57). This result seems to be promoted by the otherwise dance in foods such as liver, carrots and
would fall in line with the observation pathogenic bacterium Listeria monocyto- sweet potatoes, has been shown to have
that some “probiotic” strains (for exam- genes in the mouse (59). Additionally, a variety of immunomodulatory proper-
ple, Bifidobacterium bifidum BI-98, BI-504 gastritis inducing Heliobacter pylori in- ties. Different derivatives of VitA are pro-
and Lactobacillus acidophilus) block the crease TGF-β1 and FoxP3+ Treg popula- vided through food and vitamin supple-
suppressive activity of Tregs in an in vitro tions in the guts of mice and humans ments, such as pro-VitA from plant
suppression model (58). (24,60). sources or as enzymatically modified
Additionally, there are several conflict- Even though in many cases an in- forms from animal products (for exam-
ing reports on the protective role of pro- crease of FoxP3+ T cells could be seen ple, retinol and retinyl ester) (65). Retinol
biotics in the inflammatory disease mod- in probiotic-treated models and diseases, is absorbed by intestinal epithelial cells

M O L M E D 1 8 : 9 5 - 1 1 0 , 2 0 1 2 | I S S A Z A D E H - N AV I K A S E T A L . | 1 0 1
EFFECT OF DIET ON Treg CELLS

and esterified with fatty acids forming,


among other products, less toxic stearate
or oleate derivatives. These retinyl esters
are further transported in chylomicrons
and stored in the liver. Dietary retinol
can also be directly converted to the bio-
logically active all-trans retinoic acid
(ATRA) by gut-associated lymphoid tis-
sue DCs.
The importance of a balanced VitA
concentration for the immune system is
exemplified by the following findings:
VitA deficiency can cause Treg imbal-
ance with excess Th1 and insufficient
Th2 function (66). On the other hand,
high concentrations of VitA enhances in
vitro development of Th2 cells via the
retinoid × receptor (RXR) pathway (67).
The major VitA metabolite, ATRA, was
reported to convert naive FoxP3–CD4+
T cells into a unique, tissue-specific
FoxP3+ Treg subset in both human and
mouse T cells. Histone acetylation at the
FoxP3 gene promoter is induced upon
binding of ATRA to the nuclear retinoic
acid receptor α, which as a consequence,
leads to the prolonged expression of
FoxP3 protein in CD4+ T cells and sta-
bility of Tregs. The resulting Tregs effi-
ciently suppress effector T cells and Figure 1. Overview of the effects of VitA in gut-associated lymphoid tissue. (A) Insuffi-
show an additional upregulation of the cient VitA can cause Treg imbalance with excess Th1 and insufficient Th2 function.
mucosal tissue-homing receptors C-C (B) On the other hand, when there are elevated levels of VitA, enhanced development
of Th2 cells via RXR pathway promotion occurs. (C) When there are normal physiologi-
chemokine receptor type 9 (CCR9) and
cal levels, homeostatic functions of VitA are present in tissue. The pre- or proforms of
integrin β7. These retinoid-induced
VitA found in food are absorbed by intestinal epithelial cells and esterified with fatty
FoxP3+ cells were highly responsive to acids forming less toxic derivatives, which are further transported to liver for storage. Di-
the CCR9 ligand, chemokine (C-C motif) etary retinol is also directly converted to the biologically active ATRA, the major VitA
ligand 25 (CCL25), a chemokine specifi- metabolite, by gut-associated lymphoid tissue DCs. This process is enhanced by TLR2/6-
cally expressed by intestinal epithelial mediated effects of probiotics to induce Raldh2, which is important in conversion of
cells (68). ATRA is also reported to be VitA derivatives (VitAd) to ATRA. ATRA can convert naive FoxP3–CD4+ T cells into unique,
able to prevent the IL-6–induced con- tissue-specific FoxP3+ Tregs. TGF-β exerts synergistic effect on this process. Histone acety-
version of Foxp3+ Tregs into inflamma- lation at the FoxP3 gene promoter is induced upon binding of ATRA to the nuclear
tory Th17 cells. These in vitro TGF-β- retinoic acid receptor α (RARα), which as a consequence, leads to the prolonged ex-
plus ATRA-generated FoxP3+ Tregs pression of FoxP3 protein in CD4+ T cells and stability of Tregs. The resulting Tregs can effi-
ciently suppress effector T cells and show an additional upregulation of the mucosal
were also more protective in a colitis
tissue-homing receptors CCR9 and integrin β7. GALT, gut-associated lymphoid tissue;
model than Tregs generated with TGF-β
RARE, retinoic acid response element.
alone (69). The FoxP3-inducing and
IL-17–suppressing effects of ATRA are
likely mediated through the retinoic sistent with increased FoxP3 mRNA and though they were able to suppress naive
acid receptor α (70) (Figure 1). protein expression (68). CNS-derived 2D2-Tg or splenic effector T cells. The
The increased Treg stability is in line Tregs are capable of suppressing EAE high amounts of IL-6 and tumor necrosis
with findings that ATRA treatment in- (12), whereas in a separate study, CNS- factor (TNF)-α produced by CNS effector
creases histone acetylation of the Foxp3 recovered Tregs did not effectively sup- T cells were suggested to be the probable
promotor region in T cells, a process con- press CNS effector T cells in vitro, even cause of preventing the suppressor func-

1 0 2 | I S S A Z A D E H - N AV I K A S E T A L . | M O L M E D 1 8 : 9 5 - 1 1 0 , 2 0 1 2
INVITED REVIEW ARTICLE

tion of CNS Tregs (71). Nevertheless, ad- icance of bacteria in the causation of im- females through estrogen supplementa-
ditional study revealed that the sensitiv- mune modulation also via modification tion (84).
ity of CNS Tregs for inactivation could be of dietary products, as discussed above. An additional pathway for the treat-
modified by ATRA, which makes FoxP3+ ment effect of 1,25-(OH)2D3 has been
Tregs more resistant to IL-6–induced VitD3 Derivatives and Treg Function shown by induction of high numbers of
Th17 conversion (69). This protection VitD3 can be generated in the skin FoxP3+ Tregs in combination with IL-2
from IL-6–induced conversion could in- through processes using ultraviolet B (85). In support of this, dietary adminis-
deed explain in part previously observed (UVB)-mediated photolysis of a choles- tration of 1a-(OH)-D3, an active VitD3
disease-modifying activities of ATRA terol derivative and a spontaneous iso- analog, led to reduced diabetes in non-
and retinoid derivatives in EAE (72–74), merization step. VitD3 needs to be enzy- obese diabetic mice and increased
although these early works did not ad- matically active to its hormonal form FoxP3+ T cells (86). Earlier, it was shown
dress Tregs. However, more recent exper- (78). Because endogenous production of that a 1,25-(OH)2D3 analog also had a
iments that included Treg markers could VitD3 depends on UVB exposure, the therapeutic effect in a spontaneous auto-
show that the treatment resulted in de- concentration is influenced by environ- immune nonobese diabetic model, re-
creased IL-6Rα expression and increased mental factors such as seasonal sunshine sulting in an increased protective CD25+
numbers of FoxP3+ Tregs (75). It would length and latitude, behavioral factors Treg population (87).
be interesting to investigate if a systemic such as time and surface area of exposed A combination of 1,25-(OH)2D3 and the
treatment with retinoids could further in- skin, skin pigmentation and age. antiinflammatory corticosteroid dexa-
crease the stability of the CNS Treg pop- VitD3 is also taken up through the methasone synergistically induced prima-
ulation in the IL-6–rich environment of diet. These factors lead to great varia- rily IL-10–producing FoxP3– Tregs (88,89).
an inflamed brain. tions in circulating VitD3 and calcidiol, Nevertheless, in an experimental IBD
The Treg-inducing effects of ATRA, spurring an ongoing discussion about model, 1,25-(OH)2D3 and dexamethasone
generation of FoxP3+ Tregs and increased “natural,” “normal” and “desired” levels had a FoxP3 expression–promoting effect
gut-homing α4β7-integrin and CCR9 of VitD3 and its derivatives. A daily in- in the gut (90). Not only was the amount
have been shown to be mediated by a take of 100 μg (4,000 IU) is suggested as of FoxP3 expression increased by 1,25-
subpopulation of intestinal DCs. Dietary a safe dose, elevating the 25-(OH)-D3 (OH)2D3 treatment, but the potency of
VitA can be converted to ATRA by levels to desired concentrations in most FoxP3+ Tregs was accentuated, even in a
CD103+ small intestine DCs, a feature not humans (79). This number is higher than Th2-driven in vivo asthma model (91).
shared by splenic DCs. Blockade of the the recommended nutrient intake of be- The 1,25-(OH)2D3 precursor calcidiol
retinoic acid receptor with the retinoid tween 5 and 15 μg/day (200–600 IU) by [25-(OH)D3] is mainly 1α-hydroxylated
inhibitor LE540 in mice on a standard WHO. in the kidney to its active form, but also
diet leads to a strong reduction in FoxP3+ This section deals with the role of it has been shown to be processed in im-
Tregs. Peroxisome proliferator–activated VitD3 and its derivatives on the immune mune cells (92). CD3- and CD28-activated
receptor γ (PPARγ) activation by certain system, focusing on their effects on T cells as well as DCs are able to produce
lipids induces the expression of ATRA- FoxP3+ Treg generation and function in 1,25-(OH)2D3 from its precursor, but
synthesizing enzymes in DCs. Interest- different diseases. UVB light exposure as T cells could not directly produce precur-
ingly, the PPARγ is built up as a het- well as topical calcipotriol treatment can sor from VitD3 under these conditions
erodimer with the RXR. Usage of the lead to vitamin D receptor (VDR)- (93). In support of this, it is shown that
RXR is shared by different receptor het- dependent induction of FoxP3+ Tregs in 1α-hydroxylase is highly upregulated in
erodimers, making it interesting to spec- the skin (80,81). VitD3 supplementation is activated human T cells (94). Further-
ulate whether activation of PPARγ may reported to reduce the CNS autoimmune more, 1,25-(OH)2D3 upregulates VDR ex-
be a mechanism by which dietary com- disease EAE in females but not in males pression, which has been shown to corre-
ponents such as linolenic acids and pro- or ovariectomized female mice. The dis- late with the protective effects of
biotic bacteria affect the ATRA-mediated ease protection correlated with high local 1,25-(OH)2D3 in EAE (95). Nevertheless,
induction of tolerogenic DCs (76). 1,25-(OH)2D3 (active form) levels in the it is reported that 1,25-(OH)2D3 induces
Stimulation of TLR2 and TLR6, the re- inflamed spinal cord, but not in serum. FoxP3+ Treg conversion. The authors also
ceptors involved in recognition of con- The authors suggested a slowed degrada- showed that MS patients have lower lev-
served bacterial molecules, on splenic tion of 1,25-(OH)2D3 process in the CNS, els of calcidiol and 1,25-(OH)2D3 during
DCs leads to induction of the retinol- mediated through a suppression of the active disease, suggesting that VitD3
metabolizing enzyme retinaldehyde de- enzyme CYP24A1, a 24-hydroxylase with metabolites might play a role in control-
hydrogenase type 2 (Raldh2) and hence 1,25-(OH)2D3–inactivating properties ling Treg formation and the inflamma-
stimulates the formation of FoxP3+ Tregs (82,83). The protective effect in females tory process. This role is further sup-
(77). Such findings underscore the signif- could be restored in ovariectomized ported by findings about positive

M O L M E D 1 8 : 9 5 - 1 1 0 , 2 0 1 2 | I S S A Z A D E H - N AV I K A S E T A L . | 1 0 3
EFFECT OF DIET ON Treg CELLS

correlations of serum calcidiol and Treg makes selection of diet for individuals which are reactive toward pancreatic
function in MS patients (94,96). These who suffer from gluten sensitivity a par- β cells (105), it is reasonable that a
studies also received support in a retro- ticularly onerous task. Celiac disease is gluten-free diet can help avoid T1D
spective analysis of serum samples pre- a genetically based autoimmune disor- pathogenesis in genetically predisposed
ceding disease onset and from epidemio- der causing small-intestine injury elic- individuals. The mechanisms by which
logical studies showing an inverted ited by the ingestion of gluten (99). The dietary gluten modification of T1D oc-
correlation of sun exposure and MS standard treatment of celiac disease is a curs are poorly understood, even more
prevalence (97). Although CNS-specific gluten-free diet, which leads to a poor so in how this is related to the regulation
conversion of encephalitogenic T cells response in up to 30% of patients (99). of gut-associated Tregs. However,
into FoxP3+–induced Tregs is described The influence of this therapy on mounting evidence suggests that the gut
(12), its correlation to local VitD levels is CD4+CD25+FoxP3+ Tregs was investi- could play an important role as a defec-
still obscure. The instability of pancreas- gated in a limited number of celiac dis- tive barrier and a possible source of acti-
retrieved Tregs has been suggested (98), ease patients. The percentage of circulat- vated immune cells. Additionally, it is
but it is not reported if this is correlated ing CD4+CD25+FoxP3+ Tregs possible that protective gluten-free diets
to levels of VitD derivatives in pancreas significantly decreased in patients who not only dampen immune activation, but
of diabetic mice. Studying the endoge- had received a gluten-free diet. Al- also enhance islet function (102).
nous levels of VitD derivatives in other though, the mean FoxP3 expression lev- Despite these findings, the aforemen-
target tissues of immune attacks, such as els quantified by fluorescence-activated tioned studies do not resolve whether
in gut, joint and lung, and their role in cell sorter analysis in circulating gluten has a direct effect on Tregs or
induction of tissue-induced Tregs could CD4+CD25+FoxP3+ Tregs were signifi- whether any changes observed in the
be valuable. cantly lower in patients who had ab- Treg population is a result of a gluten-
stained from gluten compared with in- induced inflammation of the intestine.
VitA and VitD Crosstalk to Maintain dividuals on a standard diet, their in One study suggests that gluten directly
Immune Homeostasis vitro suppressive functions were pre- affects the development of Tregs (106).
It is noteworthy to highlight that VitA served (100). These results indicate that Peripheral blood mononuclear cells were
and VitD could be tightly connected to ingested gluten might influence the gen- isolated from healthy children and chil-
regulate immunity, since their signaling eration of Tregs. However, these obser- dren suffering from celiac disease and
receptors are interconnected. It is re- vations could also be interpreted as the stimulated in vitro with gluten. FoxP3
ported that the retinoic acid receptor immune system’s intrinsic attempt to mRNA expression was found to be in-
and the VDR must form heterodimers control the ongoing immune response creased in gluten-stimulated cells, indi-
with RXR to signal. Additionally, an- against gluten and the intestinal inflam- cating that the glutenic stimulation of the
other derivative of VitA, 9-cis retinoic mation by generating Tregs in patients Treg population does not depend on an
acid, increases the affinity of VDR/RXR with a standard diet. intestinal inflammation. Interestingly, the
heterodimers to its DNA recognition In nonobese diabetic mice, a gluten- extent of the FoxP3 induction was signif-
site, induces recruitment of coactivators free diet has been correlated with a de- icantly higher in children with celiac dis-
by the DNA-bound heterodimer and creased incidence of T1D (101–103). A re- ease than in reference children. It was
potentiates VitD-dependent transcrip- cent hypothesis addressing the hypothesized that memory T cells di-
tional responses. Consistent with this possibility of a direct influence of gluten rected against an antigen closely associ-
hypothesis, both VitD and VitA are on the development of Tregs analyzed ated with celiac disease were responsible
shown to be involved in regulation of differential effects of a gluten-free diet in for this effect (106).
TGF-β production and TGF-β signaling an autoimmune genetic model (104). The understanding of the basic molec-
molecules (including Smad3), FoxP3 Nonobese diabetic and BALB/c mice ular mechanisms of gluten action is cur-
and hence Treg generation and suppres- were fed either a standard diet or a rently inadequate. Its influence on gener-
sive functions, inhibition of effector gluten-free diet. As expected, the stan- ation of Tregs and on regulation of other
T helper cells by inhibiting IL-2 produc- dard-fed group showed a significantly inflammatory diseases, except for celiac
tion, as well as inhibiting effector Th1 higher incidence of diabetes than the disease, some allergies and partially T1D,
and Th17. gluten-free–fed group. The levels of is rather minimal and requires further in-
FoxP3+CD4+ Tregs in relation to the over- vestigation to allow conclusions to be
GLUTEN AND TREGS all amount of CD4+ T cells were found to drawn regarding the influences of gluten
Gluten, a protein composite of be higher in the Peyer patches of gluten- on regulation of Tregs directly and on
glutenin and gliadin, is found in grains free–fed mice than in the standard-fed immune regulation in general. Indeed,
such as rye, wheat and barley. Its ubiq- mice. Keeping in mind that Tregs are in- an earlier study investigating the effect
uitous presence in processed food volved in suppressing Th1 effector cells, of gluten-free diet on EAE reported that

1 0 4 | I S S A Z A D E H - N AV I K A S E T A L . | M O L M E D 1 8 : 9 5 - 1 1 0 , 2 0 1 2
INVITED REVIEW ARTICLE

gluten-free diet resulted in exacerbated general inhibitory capacity of DHA, crease in hepatic Tregs. In contrast,
chronic EAE (107). Interestingly, a recent likely by affecting the negative T-cell sig- splenic Treg levels were not affected by
study reported that ingestion of a gluten naling via upregulation of cytotoxic this dietary change. The decrease of he-
derivate induced T-cell proliferation pre- T-lymphocyte-associated protein 4 patic Tregs in high-fat diet–fed mice was
dominantly in the spleen but little in (CTLA-4). The effects observed in vitro linked to an increased susceptibility of
mesenteric lymph nodes. The gluten- were confirmed by in vivo studies in these Tregs to apoptosis (122). Treg apo-
reactive T cells secreted much interferon mice. Tregs isolated from mice fed a ptosis in high-fat diet–induced steatosis
(IFN)-γ but also IL-10 and had regulatory DHA diet failed to suppress proliferation is probably induced by local reactive
functions, which could prevent delayed- of Teffs isolated from mice fed a standard oxygen species, which are involved in
type hypersensitivity response (108). diet when cocultured. Surprisingly, this T-cell apoptosis (123). Tregs, in contrast
was not the case if both T-cell types were to CD25– Teffs, were found to exhibit low
DOCOSAHEXAENOIC ACID AND Tregs isolated from mice fed a DHA diet. Fur- expression levels of Bcl-2, a protein that
Over the last two decades, a novel ther effects of DHA treatment both in protects cells from reactive oxygen
group of health-promoting food has been vitro and in vivo were an increased ex- species–induced apoptosis (124).
developed called “functional foods.” pression of FoxP3, CTLA-4 and TGF-β Deregulation of Tregs as a result of ox-
Foods defined as functional were satis- mRNA, whereas IL-10 mRNA expression idative stress and other processes might
factorily shown to beneficially affect one decreased in Tregs. It is noteworthy that be involved in the progression of fatal
or more target functions in the body with classically Treg-suppressive activity was steatohepatitis. Mn(III) tetrakis (4-benzoic
an improvement in health and well- partially attributed to IL-10 and CTLA-4 acid) porphyrin (MnTBAP) is an antioxi-
being and/or reduction in disease risk. (117). In Teffs, DHA induced the expres- dant that has been shown to decrease
Currently, functional foods, for example, sion of CTLA-4, IL-10 and FoxP3, which free radical generation in ob/ob mice
cholesterol-lowering margarines, are part means that these cells obtained a Treg- (125). When high-fat diet–fed mice were
of many leading food companies’ prod- like phenotype. However, these Treg-like treated with MnTBAP, Treg apoptosis
uct lines. A typical group of ingredients cells were still not capable of suppressing was reduced and Treg depletion was re-
of functional foods is omega-3 fatty the proliferation of untreated Teffs. In versed. In untreated mice, Treg depletion
acids, which is supplemented in prod- support of earlier studies, a recent report in high-fat diet–induced steatosis led to
ucts such as bread and margarine to give shows a similar protective effect of DHA increased expression of the proinflamma-
them a health-promoting effect. Docosa- on EAE associated with reduced num- tory cytokine TNF-α, which could be re-
hexaenoic acid (DHA) is an omega-3 bers of IFN-γ– and IL-17–producing Teffs versed upon passive transfer of Tregs.
fatty acid that consists of 22 carbon in both spleen and CNS, as well as in- Briefly described, increased oxidative
atoms that build six cis double bonds. creased Treg markers without increase in stress in fatty liver caused the apoptosis
The panoply of conditions in which the suppressive function of Tregs (118). of Tregs, reduced the number of hepatic
DHA has been investigated run the In summary, the effects of DHA treat- Tregs and led to a lowered suppression
gamut from depression (109), to neural ment seem to be rather complex: on one of inflammatory responses (122).
cell death (110), to cancer (111–114) and hand, this fatty acid exerts an inhibitory The relevance of high-fat diet to
neurodegeneration, including Alz- effect on migratory and Treg function; on obesity-related disorders such as type 2
heimer’s disease. the other hand, it promotes the expres- diabetes mellitus has received major at-
DHA has been reported to reduce sup- sion of the typical Treg markers TGF-β tention. Recently, a distinct subset of
pressive and migratory functions of and FoxP3. Additionally, it should be Tregs was identified as an adipose
Tregs (115). It is shown that coculturing mentioned that earlier studies have tissue–resident population that seems to
CD4+CD25– T effector cells (Teffs) with shown that FoxP3 and CTLA-4 expres- affect metabolic parameters, in particular,
CD4+CD25+ Tregs leads to a suppression sion alone were not sufficient to convert insulin resistance secondary to obesity.
of Teff proliferation (10,116); the in- a T cell into a Treg capable of suppress- Such cells accumulated with age in lean
hibitory capacity of Tregs on Teff prolif- ing Teff proliferation (119). Other factors mice to eventually represent more than
eration was found to be decreased when are required to interact with FoxP3 to as- half of the CD4+ T cells residing in the
Tregs were first incubated with DHA. sign Treg properties to a T cell. visceral fat. Interestingly, Tregs were not
The effect of DHA was shown to be dose enriched in subcutaneous adipose de-
dependent; a concentration of 100 HIGH-FAT DIET AND Tregs pots. Of relevance, the changes in vis-
μmol/L was sufficient to completely re- Tregs are key players in hepatic im- ceral but not in subcutaneous fat were
verse the inhibitory effect of Tregs. Inter- mune regulation (120,121). A high-fat associated with insulin resistance. On the
estingly, the same observations were diet in mice induced hepatic steatosis, a contrary, the percentage and number of
made when Teffs instead of Tregs were model for nonalcoholic steatohepatitis. fat Tregs were substantially reduced in
preincubated with DHA, indicating a The high-fat diet caused a gradual de- obese mice compared with lean controls.

M O L M E D 1 8 : 9 5 - 1 1 0 , 2 0 1 2 | I S S A Z A D E H - N AV I K A S E T A L . | 1 0 5
EFFECT OF DIET ON Treg CELLS

This result was also accompanied by in-


creased proinflammatory innate and
adoptive immune cells (137). The actual
reasons for the decrease in the Treg frac-
tion in abdominal adipose tissue during
obesity are still not well understood;
however, it is interesting to speculate
that increased oxidative stress in abdom-
inal adipose fat could contribute to apo-
ptosis of fat-resident Tregs. This result
consequently could lead to increased at-
traction of proinflammatory cells to
change the immune balance.
Figure 2 and Table 6 summarize how
the above-mentioned dietary compo-
nents could affect Treg properties and
their function.

Figure 2. Overview of dietary components and conditions (represented by red objects)


DO TISSUE-SPECIFIC FACTORS
influencing regulatory T cells. The Treg-modulating effects of dietary components could INFLUENCE Tregs?
be direct by mediating expansion and/or suppressive function of Tregs or indirect via As discussed, over the last few years,
modulation of DCs and their subsequent actions on Tregs. Additionally, the indirect influ- different food components such as vita-
ence of dietary components could be mediated by regulation of naive T cells to convert mins and fatty acids have been identified
to Tregs or by blocking the function of inflammatory Teffs. The positive antiinflammatory ef- to influence the development and char-
fects of Tregs are mediated by expression of FoxP3 and production of IL-10 (indicated by acter of Tregs in vivo and in vitro. This
yellow color). ROS, reactive oxygen species. IL-10hi, high expression of IL-10.

Table 6. Overview of substances and conditions affecting Tregs in different models.

Substance/condition Effects on Tregs Model Reference

Gluten Decrease in % CD4+ FoxP3+ T cells Mouse (NOD) 104


+ + +
Gluten Increase in circulating CD4 CD25 FoxP3 T cells and FoxP3 Human (celiac disease) 100
expression within these cells
Gluten Increased FoxP3 expression in PBMC cultures Human (in vitro) 106
DHA Decrease of Tregs’ inhibitory capacity on Teff proliferation; increased Mouse (in vitro) 115
expression of FoxP3, CTLA-4, TGF-β; decreased IL-10 expression
High-fat diet Hepatic Treg depletion due to increased Treg apoptosis Mouse 122
MnTBAP Decrease in Treg apoptosis due to reduced levels of reactive oxygen Mouse 122
species
High 25(OH)D serum Increase in the ability of Tregs to suppress T responder cells; Human (MS) 96
levels IFN-γ/IL-4 ratio (Th1/Th2 balance) directed toward IL-4 (Th2)
High 25(OH)D serum Low blood Treg percentages Human (MS) 135
levels
Calcipotriol (topical) Induction of Tregs that prevent the priming of cytotoxic T cells in Mouse 80
response to locally administered antigen
VitD3 VitD3-primed DCs can convert T cells to Tregs Human (in vitro) 136
ATRA Conversion of naive CD4+ FoxP3– T cells into gut-homing Human and mouse (in vitro) 68
CD4+ FoxP3+ CCR9+ T cells
VitD3 EAE reduced in females Mouse 82

25(OH)D, 25-hydroxyvitamin D.

1 0 6 | I S S A Z A D E H - N AV I K A S E T A L . | M O L M E D 1 8 : 9 5 - 1 1 0 , 2 0 1 2
INVITED REVIEW ARTICLE

finding raises not only questions about ble iTregs such as VitA and TGF-β have REFERENCES
the influence food components have on been reported; however, the full under- 1. Maloy KJ, Powrie F. (2001) Regulatory T cells in
the control of immune pathology. Nat. Immunol.
the immune system, but also which role standing is still lacking. Hence, it is valu-
2:816–22.
different tissue environments might play able to understand how Tregs are influ- 2. Ziegler SF. (2006) FOXP3: of mice and men.
in the formation of specific tissue- enced by tissue-specific factors, some of Annu. Rev. Immunol. 24:209–26.
induced Tregs. A Treg generated in the which may include similar dietary deriv- 3. Chen W, Konkel JE. 2010. TGF-beta and ‘adap-
liver that is exposed to high concentra- atives. Understanding the nature of trans tive’ Foxp3(+) regulatory T cells. J. Mol. Cell. Biol.
tions of vitamins, such as VitD, as well as exchange of VitA and VitD derivatives or 2:30–6.
4. Collison LW, et al. (2007) The inhibitory cytokine
free radicals might be different than fatty acids such as DHA between tissue-
IL-35 contributes to regulatory T-cell function.
those generated in other local conditions specific cells such as neurons, β islet cells Nature. 450:566–9.
(for example, in brain or pancreatic tis- and tissue-infiltrating inflammatory 5. Collison LW, et al. (2010) IL-35-mediated induc-
sue). Therefore, these cells might have T cells will prove fruitful to identify if tion of a potent regulatory T cell population. Nat.
different characteristics than iTregs gen- such factors contribute to the generation, Immunol. 11:1093–101.
6. Kim CH. (2006) Migration and function of
erated in the spleen or mesenteric lymph functionality and stability of Tregs to
FoxP3+ regulatory T cells in the hematolym-
nodes. prevent chronic tissue damage. phoid system. Exp. Hematol. 34:1033–40.
It is tempting to speculate that the ex- 7. Bennett CL, et al. (2001) The immune dysregula-
posure of T cells to cell-type specific fatty CONCLUSION tion, polyendocrinopathy, enteropathy, X-linked
acid components, vitamins derivates and CD4+CD25+FoxP3+ Tregs play a cen- syndrome (IPEX) is caused by mutations of
FOXP3. Nat. Genet. 27:20–1.
other factors in different tissues could tral role in maintaining peripheral im-
8. Brunkow ME, et al. (2001) Disruption of a new
contribute to the generation of differen- mune tolerance. As such, it is not sur- forkhead/winged-helix protein, scurfin, results
tial tissue-specific iTregs. Indeed, the prising that proper functioning of Tregs in the fatal lymphoproliferative disorder of the
brain milieu in general and neurons in is important in gut-associated immunity. scurfy mouse. Nat. Genet. 27:68–73.
particular have been shown to support Even more understandable is why the 9. Curiel TJ. (2007) Tregs and rethinking cancer im-
munotherapy. J. Clin. Invest. 117:1167–74.
generation of Tregs from encephalito- critical gut-associated immune cell pop-
10. Sakaguchi S. (2000) Regulatory T cells: key con-
genic Th1 cells, mediated by neuronal ulation, Tregs, adapts itself to the influ- trollers of immunologic self-tolerance. Cell.
production of TGF-β and B7.1 costimula- ence of recurrent ingested dietary com- 101:455–8.
tory signaling (12). Additionally, a dis- ponents such as VitA and VitD, gluten 11. Curotto de Lafaille MA, Lafaille JJ. (2009) Natu-
tinct subset of Tregs was identified to re- and fatty acids. Additionally, as func- ral and adaptive foxp3+ regulatory T cells: more
of the same or a division of labor? Immunity.
side in adipose tissue, and this subset tional food containing probiotics be-
30:626–35.
has an effect on insulin resistance (137). come more widely available and popu- 12. Liu Y, Teige I, Birnir B, Issazadeh-Navikas S.
How these tissue-specific Tregs are gen- lar, understanding the mode of action of (2006) Neuron-mediated generation of regulatory
erated are not yet completely under- these dietary factors on regulation of T cells from encephalitogenic T cells suppresses
stood, and further investigation is Treg homeostasis could prove valuable EAE. Nat. Med. 12:518–25.
needed to understand how tissue- to control many chronic inflammatory 13. Weiner HL, da Cunha AP, Quintana F, Wu H.
(2011) Oral tolerance. Immunol. Rev. 241:241–59.
specific Tregs in general are generated conditions directly affecting the gut
14. Liu Y, Teige I, Ericsson I, Navikas V, Issazadeh-
and controlled and how they influence (IBD, Crohn’s disease) or indirectly in- Navikas S. (2010) Suppression of EAE by oral tol-
homeostasis of different tissues. More fluencing other chronic tissue inflamma- erance is independent of endogenous IFN-beta
importantly, what are the tissue-specific tory conditions (T1D, MS and RA) regu- whereas treatment with recombinant IFN-beta
cells, factors and environmental require- lated by Tregs. ameliorates EAE. Immunol. Cell. Biol. 88:468–76.
15. Food and Agriculture Organization of the United
ments to generate and maintain the func-
Nations [FAO]; WHO. (2001) Health and Nutri-
tion of tissue-specific Tregs? It is also im- ACKNOWLEDGMENTS tional Properties of Probiotics in Food including
portant to point out that lack of stability This work was supported by grants Powder Milk with Live Lactic Acid Bacteria: Re-
of some tissue-specific iTregs has been from the Lundbeck Foundation in Den- port of a Joint FAO/WHO Expert Consultation
reported. It was shown that a population mark, the Novo Nordisk Foundation and on Evaluation of Health and Nutritional Proper-
ties of Probiotics in Food Including Powder Milk
of Tregs expressing Foxp3 are unstable in the Danish Research Council–Medicine.
with Live Lactic Acid Bacteria: Amerian Córdoba
vivo; these so-called exFoxp3 cells with Park Hotel, Córdoba, Argentina: 1-4 October 2001
shared ontogeny with Foxp3+ Tregs and DISCLOSURE [PDF on the Internet]. [Rome]: FAO; [Geneva]:
conventional T cells become significantly The authors declare that they have no WHO; [cited 2012 Jan 19]. Available from:
higher in percentage in the autoimmune competing interests as defined by Molec- www.who.int/entity/foodsafety/publications/
fs_management/en/probiotics.pdf
diabetes setting, but surprisingly they ular Medicine, or other interests that
16. Sartor RB. (2004) Therapeutic manipulation of
are capable of transferring diabetes in- might be perceived to influence the re- the enteric microflora in inflammatory bowel dis-
stead of protecting (98). The tissue fac- sults and discussion reported in this eases: antibiotics, probiotics, and prebiotics. Gas-
tors that facilitate generation of more sta- paper. troenterology. 126:1620–33.

M O L M E D 1 8 : 9 5 - 1 1 0 , 2 0 1 2 | I S S A Z A D E H - N AV I K A S E T A L . | 1 0 7
EFFECT OF DIET ON Treg CELLS

17. Sartor RB. (2006) Mechanisms of disease: patho- encephalomyelitis. Immunopharmacol. Immunotox- port function of T cells with regulatory proper-
genesis of Crohn’s disease and ulcerative colitis. icol. 32:116–24. ties. Immunology. 108:481–92.
Nat. Clin. Pract. Gastroenterol. Hepatol. 3:390–407. 34. Ezendam J, van Loveren H. (2008) Lactobacillus 50. Zarek PE, et al. (2008) A2A receptor signaling
18. Taurog JD, et al. (1994) The germfree state pre- casei Shirota administered during lactation in- promotes peripheral tolerance by inducing T-cell
vents development of gut and joint inflammatory creases the duration of autoimmunity in rats and anergy and the generation of adaptive regulatory
disease in HLA-B27 transgenic rats. J. Exp. Med. enhances lung inflammation in mice. Br. J. Nutr. T cells. Blood. 111:251–9.
180:2359–64. 99:83–90. 51. Mandapathil M, et al. (2010) Generation and accu-
19. Waidmann M, et al. (2003) Bacteroides vulgatus 35. Baken KA, et al. (2006) Evaluation of im- mulation of immunosuppressive adenosine by
protects against Escherichia coli-induced colitis munomodulation by Lactobacillus casei Shirota: human CD4+CD25highFOXP3+ regulatory T cells.
in gnotobiotic interleukin-2-deficient mice. Gas- immune function, autoimmunity and gene ex- J. Biol. Chem. 285:7176–86.
troenterology. 125:162–77. pression. Int. J. Food Microbiol. 112:8–18. 52. Atarashi K, et al. (2008) ATP drives lamina propria
20. Kwon HK, et al. (2010) Generation of regulatory 36. Rachmilewitz D, et al. (2004) Toll-like receptor 9 T(H)17 cell differentiation. Nature. 455:808–12.
dendritic cells and CD4+Foxp3+ T cells by probi- signaling mediates the anti-inflammatory effects 53. Salmi M, Jalkanen S. (2001) Human leukocyte
otics administration suppresses immune disor- of probiotics in murine experimental colitis. Gas- subpopulations from inflamed gut bind to joint
ders. Proc. Natl. Acad. Sci. U. S. A. 107:2159–64. troenterology. 126:520–8. vasculature using distinct sets of adhesion mole-
21. Siddiqui KR, Powrie F. (2008) CD103+ GALT 37. Karimi K, Inman MD, Bienenstock J, Forsythe P. cules. J. Immunol. 166:4650–7.
DCs promote Foxp3+ regulatory T cells. Mucosal (2009) Lactobacillus reuteri-induced regulatory 54. Moran JP, Walter J, Tannock GW, Tonkonogy SL,
Immunol. 1 Suppl 1:S34–8. T cells protect against an allergic airway response Sartor RB. (2009) Bifidobacterium animalis
22. Izcue A, Coombes JL, Powrie F. (2006) Regula- in mice. Am. J. Respir. Crit. Care Med. 179:186–93. causes extensive duodenitis and mild colonic in-
tory T cells suppress systemic and mucosal im- 38. Petrof EO, et al. (2004) Probiotics inhibit nuclear flammation in monoassociated interleukin-10-
mune activation to control intestinal inflamma- factor-kappaB and induce heat shock proteins in deficient mice. Inflamm. Bowel Dis. 15:1022–31.
tion. Immunol. Rev. 212:256–71. colonic epithelial cells through proteasome inhi- 55. Leach MW, Davidson NJ, Fort MM, Powrie F,
23. Ivanov II, et al. (2008) Specific microbiota direct bition. Gastroenterology. 127:1474–87. Rennick DM. (1999) The role of IL-10 in inflam-
the differentiation of IL-17-producing T-helper 39. Petrof EO, et al. (2009) Bacteria-free solution de- matory bowel disease: “of mice and men.” Toxi-
cells in the mucosa of the small intestine. Cell. rived from Lactobacillus plantarum inhibits mul- col. Pathol. 27:123–33.
Host Microbe. 4:337–49. tiple NF-kappaB pathways and inhibits protea- 56. Lee JS, Paek NS, Kwon OS, Hahm KB. (2010)
24. Rad R, et al. (2006) CD25+/Foxp3+ T cells regu- some function. Inflamm. Bowel Dis. 15:1537–47. Anti-inflammatory actions of probiotics through
late gastric inflammation and Helicobacter pylori 40. Cong Y, et al. (2005) Generation of antigen-spe- activating suppressor of cytokine signaling
colonization in vivo. Gastroenterology. 131:525–37. cific, Foxp3-expressing CD4+ regulatory T cells (SOCS) expression and signaling in Helicobacter
25. O’Mahony C, et al. (2008) Commensal-induced by inhibition of APC proteosome function. J. Im- pylori infection: a novel mechanism. J. Gastroen-
regulatory T cells mediate protection against munol. 174:2787–95. terol. Hepatol. 25:194–202.
pathogen-stimulated NF-kappaB activation. PLoS 41. Smits HH, et al. (2005) Selective probiotic bacteria 57. Pillemer BB, Xu H, Oriss TB, Qi Z, Ray A. (2007)
Pathog. 4:e1000112. induce IL-10-producing regulatory T cells in Deficient SOCS3 expression in
26. So JS, et al. (2008) Lactobacillus casei potentiates vitro by modulating dendritic cell function CD4+CD25+FoxP3+ regulatory T cells and
induction of oral tolerance in experimental through dendritic cell-specific intercellular adhe- SOCS3-mediated suppression of Treg function.
arthritis. Mol. Immunol. 46:172–80. sion molecule 3-grabbing nonintegrin. J. Allergy Eur. J. Immunol. 37:2082–9.
27. So JS, et al. (2008) Lactobacillus casei suppresses Clin. Immunol. 115:1260–7. 58. Schmidt EG, Claesson MH, Jensen SS, Ravn P,
experimental arthritis by down-regulating 42. Foligne B, et al. (2007) A key role of dendritic Kristensen NN. (2010) Antigen-presenting cells
T helper 1 effector functions. Mol. Immunol. cells in probiotic functionality. PLoS One. 2:e313. exposed to Lactobacillus acidophilus NCFM, Bi-
45:2690–9. 43. Rahman MK, et al. (2010) The pathogen recogni- fidobacterium bifidum BI-98, and BI-504 reduce
28. Kato I, Endo-Tanaka K, Yokokura T. (1998) Sup- tion receptor NOD2 regulates human FOXP3+ regulatory T cell activity. Inflamm. Bowel Dis.
pressive effects of the oral administration of Lac- T cell survival. J. Immunol. 184:7247–56. 16:390–400.
tobacillus casei on type II collagen-induced 44. Penack O, et al. (2009) NOD2 regulates 59. Ertelt JM, et al. (2009) Selective priming and ex-
arthritis in DBA/1 mice. Life Sci. 63:635–44. hematopoietic cell function during graft-versus- pansion of antigen-specific Foxp3- CD4+ T cells
29. Hacini-Rachinel F, et al. (2009) Oral probiotic con- host disease. J. Exp. Med. 206:2101–10. during Listeria monocytogenes infection. J. Im-
trol skin inflammation by acting on both effector 45. Petnicki-Ocwieja T, et al. (2009) Nod2 is required munol. 182:3032–8.
and regulatory T cells. PLoS One. 4:e4903. for the regulation of commensal microbiota in the 60. Kandulski A, et al. (2008) Naturally occurring
30. Maassen CB, Claassen E. (2008) Strain-dependent intestine. Proc. Natl. Acad. Sci. U. S. A. 106:15813–8. regulatory T cells (CD4+, CD25high, FOXP3+) in
effects of probiotic lactobacilli on EAE autoim- 46. Fernandez EM, et al. (2011) Anti-inflammatory the antrum and cardia are associated with higher
munity. Vaccine. 26:2056–7. capacity of selected lactobacilli in experimental H. pylori colonization and increased gene ex-
31. Ochoa-Reparaz J, et al. (2010) Central nervous sys- colitis is driven by NOD2-mediated recognition pression of TGF-beta1. Helicobacter. 13:295–303.
tem demyelinating disease protection by the of a specific peptidoglycan-derived muropeptide. 61. DeLuca HF. (2004) Overview of general physio-
human commensal Bacteroides fragilis depends on Gut. 60:1050–9. logic features and functions of vitamin D. Am. J.
polysaccharide A expression. J. Immunol.185:4101–8. 47. Rachmilewitz D, et al. (2002) Immunostimulatory Clin. Nutr. 80:1689S–96S.
32. Ochoa-Reparaz J, Mielcarz DW, Haque-Begum S, DNA ameliorates experimental and spontaneous 62. Belkaid Y, Oldenhove G. (2008) Tuning microen-
Kasper LH. (2010) Induction of a regulatory murine colitis. Gastroenterology. 122:1428–41. vironments: induction of regulatory T cells by
B cell population in experimental allergic en- 48. Bleich A, et al. (2009) CpG motifs of bacterial dendritic cells. Immunity. 29:362–71.
cephalomyelitis by alteration of the gut commen- DNA exert protective effects in mouse models of 63. Matheu V, Back O, Mondoc E, Issazadeh-Navikas
sal microflora. Gut Microbes. 1:103–8. IBD by antigen-independent tolerance induction. S. (2003) Dual effects of vitamin D-induced alter-
33. Kobayashi T, et al. (2010) Oral administration of Gastroenterology. 136:278–87. ation of TH1/TH2 cytokine expression: enhanc-
probiotic bacteria, Lactobacillus casei and Bifi- 49. Bilsborough J, George TC, Norment A, Viney JL. ing IgE production and decreasing airway
dobacterium breve, does not exacerbate neuro- (2003) Mucosal CD8alpha+ DC, with a plasmacy- eosinophilia in murine allergic airway disease.
logical symptoms in experimental autoimmune toid phenotype, induce differentiation and sup- J. Allergy Clin. Immunol. 112:585–92.

1 0 8 | I S S A Z A D E H - N AV I K A S E T A L . | M O L M E D 1 8 : 9 5 - 1 1 0 , 2 0 1 2
INVITED REVIEW ARTICLE

64. Matheu V, et al. (2009) Impact on allergic im- violet-irradiated skin is transferable through 95. Meehan TF, DeLuca HF. (2002) The vitamin D
mune response after treatment with vitamin A. CD4+CD25+ T regulatory cells and is dependent receptor is necessary for 1alpha,25-dihydroxyvi-
Nutr. Metab. 6:44. on host-derived IL-10. J. Immunol. 176:2635–44. tamin D(3) to suppress experimental autoim-
65. Penniston KL, Tanumihardjo SA. (2006) The 82. Spach KM, Hayes CE. (2005) Vitamin D3 confers mune encephalomyelitis in mice. Arch. Biochem.
acute and chronic toxic effects of vitamin A. Am. protection from autoimmune encephalomyelitis Biophys. 408:200–4.
J. Clin. Nutr. 83:191–201. only in female mice. J. Immunol. 175:4119–26. 96. Smolders J, et al. (2009) Vitamin D status is posi-
66. Cantorna MT, Nashold FE, Hayes CE. (1994) In 83. Horst RL, Reinhardt TA, Ramberg CF, Koszewski tively correlated with regulatory T cell function
vitamin A deficiency multiple mechanisms estab- NJ, Napoli JL. (1986) 24-Hydroxylation of 1,25- in patients with multiple sclerosis. PLoS One.
lish a regulatory T helper cell imbalance with ex- dihydroxyergocalciferol: an unambiguous deacti- 4:e6635.
cess Th1 and insufficient Th2 function. J. Im- vation process. J. Biol. Chem. 261:9250–6. 97. Munger KL, Levin LI, Hollis BW, Howard NS,
munol. 152:1515–22. 84. Nashold FE, Spach KM, Spanier JA, Hayes CE. Ascherio A. (2006) Serum 25-hydroxyvitamin D
67. Stephensen CB, et al. (2002) Vitamin A enhances (2009) Estrogen controls vitamin D3-mediated re- levels and risk of multiple sclerosis. JAMA.
in vitro Th2 development via retinoid X receptor sistance to experimental autoimmune encephalo- 296:2832–8.
pathway. J. Immunol. 168:4495–503. myelitis by controlling vitamin D3 metabolism 98. Zhou X, et al. (2009) Instability of the transcrip-
68. Kang SG, et al. (2007) Vitamin A metabolites in- and receptor expression. J. Immunol. 183:3672–81. tion factor Foxp3 leads to the generation of
duce gut-homing FoxP3+ regulatory T cells. 85. Jeffery LE, et al. (2009) 1,25-Dihydroxyvitamin D3 pathogenic memory T cells in vivo. Nat. Im-
J. Immunol. 179:3724–33. and IL-2 combine to inhibit T cell production of munol. 10:1000–7.
69. Mucida D, et al. (2007) Reciprocal TH17 and reg- inflammatory cytokines and promote develop- 99. Green PH, Cellier C. (2007) Celiac disease. N. Engl.
ulatory T cell differentiation mediated by retinoic ment of regulatory T cells expressing CTLA-4 J. Med. 357:1731–43.
acid. Science. 317:256–60. and FoxP3. J. Immunol. 183:5458–67. 100. Frisullo G, et al. (2009) Increased
70. Elias KM, et al. (2008) Retinoic acid inhibits Th17 86. Driver JP, Foreman O, Mathieu C, van Etten E, Ser- CD4+CD25+Foxp3+ T cells in peripheral blood
polarization and enhances FoxP3 expression reze DV. (2008) Comparative therapeutic effects of of celiac disease patients: correlation with di-
through a Stat-3/Stat-5 independent signaling orally administered 1,25-dihydroxyvitamin D(3) etary treatment. Hum. Immunol. 70:430–5.
pathway. Blood. 111:1013–20. and 1alpha-hydroxyvitamin D(3) on type-1 dia- 101. Funda DP, Kaas A, Bock T, Tlaskalova-
71. Korn T, et al. (2007) Myelin-specific regulatory betes in non-obese diabetic mice fed a normal- Hogenova H, Buschard K. (1999) Gluten-free
T cells accumulate in the CNS but fail to control calcaemic diet. Clin. Exp. Immunol. 151:76–85. diet prevents diabetes in NOD mice. Diabetes
autoimmune inflammation. Nat. Med. 13:423–31. 87. Gregori S, Giarratana N, Smiroldo S, Uskokovic Metab. Res. Rev. 15:323–7.
72. Racke MK, et al. (1995) Retinoid treatment of ex- M, Adorini L. (2002) A 1alpha,25-dihydroxyvita- 102. Lefebvre DE, Powell KL, Strom A, Scott FW.
perimental allergic encephalomyelitis: IL-4 pro- min D(3) analog enhances regulatory T-cells and (2006) Dietary proteins as environmental modi-
duction correlates with improved disease course. arrests autoimmune diabetes in NOD mice. Dia- fiers of type 1 diabetes mellitus. Annu. Rev.
J. Immunol. 154:450–8. betes. 51:1367–74. Nutr. 26:175–202.
73. Massacesi L, et al. (1991) Immunosuppressive ac- 88. Barrat FJ, et al. (2002) In vitro generation of inter- 103. Schmid S, et al. (2004) Delayed exposure to
tivity of 13-cis-retinoic acid and prevention of ex- leukin 10-producing regulatory CD4(+) T cells is wheat and barley proteins reduces diabetes in-
perimental autoimmune encephalomyelitis in induced by immunosuppressive drugs and inhib- cidence in non-obese diabetic mice. Clin. Im-
rats. J. Clin. Invest. 88:1331–7. ited by T helper type 1 (Th1)- and Th2-inducing munol. 111:108–18.
74. Massacesi L, et al. (1987) Suppression of experi- cytokines. J. Exp. Med. 195:603–16. 104. Ejsing-Duun M, Josephsen J, Aasted B,
mental allergic encephalomyelitis by retinoic 89. Vieira PL, et al. (2004) IL-10-secreting regulatory Buschard K, Hansen AK. (2008) Dietary gluten
acid. J. Neurol. Sci. 80:55–64. T cells do not express Foxp3 but have compara- reduces the number of intestinal regulatory
75. Xiao S, et al. (2008) Retinoic acid increases Foxp3+ ble regulatory function to naturally occurring T cells in mice. Scand. J. Immunol. 67:553–9.
regulatory T cells and inhibits development of CD4+CD25+ regulatory T cells. J. Immunol. 105. Pop SM, Wong CP, Culton DA, Clarke SH, Tisch
Th17 cells by enhancing TGF-beta-driven Smad3 172:5986–93. R. (2005) Single cell analysis shows decreasing
signaling and inhibiting IL-6 and IL-23 receptor 90. Daniel C, Sartory NA, Zahn N, Radeke HH, FoxP3 and TGFbeta1 coexpressing CD4+CD25+
expression. J. Immunol. 181:2277–84. Stein JM. (2008) Immune modulatory treatment regulatory T cells during autoimmune diabetes.
76. Hall JA, Grainger JR, Spencer SP, Belkaid Y. of trinitrobenzene sulfonic acid colitis with cal- J. Exp. Med. 201:1333–46.
(2011) The role of retinoic acid in tolerance and citriol is associated with a change of a T helper 106. Kivling A, et al. (2008) Diverse foxp3 expression
immunity. Immunity. 35:13–22. (Th) 1/Th17 to a Th2 and regulatory T cell pro- in children with type 1 diabetes and celiac dis-
77. Manicassamy S, et al. (2009) Toll-like receptor 2- file. J. Pharmacol. Exp. Ther. 324:23–33. ease. Ann. N. Y. Acad. Sci. 1150:273–7.
dependent induction of vitamin A-metabolizing 91. Gorman S, Judge MA, Burchell JT, Turner DJ, 107. Di Marco R, et al. (2004) Exacerbation of pro-
enzymes in dendritic cells promotes T regulatory Hart PH. (2010) 1,25-dihydroxyvitamin D3 en- tracted-relapsing experimental allergic en-
responses and inhibits autoimmunity. Nat. Med. hances the ability of transferred CD4+ CD25+ cephalomyelitis in DA rats by gluten-free diet.
15:401–9. cells to modulate T helper type 2-driven asth- APMIS. 112:651–5.
78. White JH. (2011) Vitamin D metabolism and sig- matic responses. Immunology. 130:181–92. 108. Du Pre MF, et al. (2011) Tolerance to ingested
naling in the immune system. Rev. Endocr. Metab. 92. Hewison M, et al. (2007) Extra-renal deamidated gliadin in mice is maintained by
Disord. 2011, Aug 16. [Epub ahead of print]. 25-hydroxyvitamin D3–1alpha-hydroxylase in splenic, type 1 regulatory T cells. Gastroenterol-
79. Vieth R. (2004) Why the optimal requirement for human health and disease. J. Steroid. Biochem. ogy. 141:610–620, e611–612.
vitamin D3 is probably much higher than what is Mol. Biol. 103:316–21. 109. McNamara RK, et al. (2007) Selective deficits in
officially recommended for adults. J. Steroid. 93. Sigmundsdottir H, et al. (2007) DCs metabolize the omega-3 fatty acid docosahexaenoic acid in
Biochem. Mol. Biol. 89–90:575–9. sunlight-induced vitamin D3 to ‘program’ T cell the postmortem orbitofrontal cortex of patients
80. Ghoreishi M, et al. (2009) Expansion of antigen- attraction to the epidermal chemokine CCL27. with major depressive disorder. Biol. Psychiatry.
specific regulatory T cells with the topical vita- Nat. Immunol. 8:285–93. 62:17–24.
min D analog calcipotriol. J. Immunol. 182:6071–8. 94. Correale J, Ysrraelit MC, Gaitan MI. (2009) Im- 110. Serhan CN, Gotlinger K, Hong S, Arita M.
81. Ghoreishi M, Dutz JP. (2006) Tolerance induction munomodulatory effects of vitamin D in multi- (2004) Resolvins, docosatrienes, and neuropro-
by transcutaneous immunization through ultra- ple sclerosis. Brain. 132:1146–60. tectins, novel omega-3-derived mediators, and

M O L M E D 1 8 : 9 5 - 1 1 0 , 2 0 1 2 | I S S A Z A D E H - N AV I K A S E T A L . | 1 0 9
EFFECT OF DIET ON Treg CELLS

their aspirin-triggered endogenous epimers: an molecules in murine steatohepatitis. Hepatology.


overview of their protective roles in catabasis. 39:1277–85.
Prostaglandins Other Lipid Mediat. 73:155–72. 126. Roselli M, et al. (2009) Prevention of TNBS-
111. El-Mesery M, Al-Gayyar M, Salem H, Darweish induced colitis by different Lactobacillus and
M, El-Mowafy A. (2009) Chemopreventive and Bifidobacterium strains is associated with an
renal protective effects for docosahexaenoic expansion of gammadeltaT and regulatory
acid (DHA): implications of CRP and lipid per- T cells of intestinal intraepithelial lymphocytes.
oxides. Cell Div. 4:6. Inflamm. Bowel Dis. 15:1526–36.
112. Kato T, et al. (2002) Influence of omega-3 fatty 127. Di Giacinto C, Marinaro M, Sanchez M, Strober
acids on the growth of human colon carcinoma W, Boirivant M. (2005) Probiotics ameliorate re-
in nude mice. Cancer Lett. 187:169–77. current Th1-mediated murine colitis by induc-
113. Schonberg SA, et al. (2006) Closely related colon ing IL-10 and IL-10-dependent TGF-beta-bear-
cancer cell lines display different sensitivity to ing regulatory cells. J. Immunol. 174:3237–46.
polyunsaturated fatty acids, accumulate different 128. Pronio A, et al. (2008) Probiotic administration
lipid classes and downregulate sterol regulatory in patients with ileal pouch-anal anastomosis
element-binding protein 1. FEBS J. 273:2749–65. for ulcerative colitis is associated with expan-
114. Shaikh IA, Brown I, Schofield AC, Wahle KW, sion of mucosal regulatory cells. Inflamm. Bowel
Heys SD. (2008) Docosahexaenoic acid en- Dis. 14:662–8.
hances the efficacy of docetaxel in prostate can- 129. Sheil B, et al. (2004) Is the mucosal route of ad-
cer cells by modulation of apoptosis: the role of ministration essential for probiotic function?
genes associated with the NF-kappaB pathway. Subcutaneous administration is associated with
Prostate. 68:1635–46. attenuation of murine colitis and arthritis. Gut.
115. Yessoufou A, Ple A, Moutairou K, Hichami A, 53:694–700.
Khan NA. (2009) Docosahexainoic acid HA re- 130. Baharav E, Mor F, Halpern M, Weinberger A.
duces suppressive and migratory functions of (2004) Lactobacillus GG bacteria ameliorate
CD4+CD25+ regulatory T-cells. J Lipid Res. arthritis in Lewis rats. J. Nutr. 134:1964–9.
50:2377–88. 131. Lavasani S, et al. (2010) A novel probiotic mix-
116. Shevach EM. (2002) CD4+ CD25+ suppressor ture exerts a therapeutic effect on experimental
T cells: more questions than answers. Nat. Rev. autoimmune encephalomyelitis mediated by
Immunol. 2:389–400. IL-10 producing regulatory T cells. PLoS One.
117. Sebastiani S, et al. (2001) Chemokine receptor ex- 5:e9009.
pression and function in CD4+ T lymphocytes 132. Feleszko W, et al. (2007) Probiotic-induced sup-
with regulatory activity. J. Immunol. 166:996–1002. pression of allergic sensitization and airway in-
118. Kong W, Yen JH, Ganea D. (2011) Docosa- flammation is associated with an increase of
hexaenoic acid prevents dendritic cell matura- T regulatory-dependent mechanisms in a
tion, inhibits antigen-specific Th1/Th17 differ- murine model of asthma. Clin. Exp. Allergy.
entiation and suppresses experimental 37:498–505.
autoimmune encephalomyelitis. Brain Behave. 133. Sun CM, et al. (2007) Small intestine lamina pro-
Immun. 25:872–82. pria dendritic cells promote de novo generation
119. Tran DQ, Ramsey H, Shevach EM. (2007) Induc- of Foxp3 T reg cells via retinoic acid. J. Exp.
tion of FOXP3 expression in naive human Med. 204:1775–85.
CD4+FOXP3 T cells by T-cell receptor stimula- 134. Coombes JL, et al. (2007) A functionally special-
tion is transforming growth factor-beta depen- ized population of mucosal CD103+ DCs in-
dent but does not confer a regulatory pheno- duces Foxp3+ regulatory T cells via a TGF-beta
type. Blood. 110:2983–90. and retinoic acid-dependent mechanism. J. Exp.
120. Chang KM. (2005) Regulatory T cells and the Med. 204:1757–64.
liver: a new piece of the puzzle. Hepatology. 135. Royal W 3rd, Mia Y, Li H, Naunton K. (2009)
41:700–2. Peripheral blood regulatory T cell measure-
121. Winer S, et al. (2009) Normalization of obesity- ments correlate with serum vitamin D levels in
associated insulin resistance through im- patients with multiple sclerosis. J. Neuroim-
munotherapy. Nat. Med. 15:921–9. munol. 213:135–41.
122. Ma X, et al. (2007) A high-fat diet and regulatory 136. Unger WW, Laban S, Kleijwegt FS, van der Slik
T cells influence susceptibility to endotoxin-in- AR, Roep BO. (2009) Induction of Treg by
duced liver injury. Hepatology. 46:1519–29. monocyte-derived DC modulated by vitamin
123. Hildeman DA, et al. (1999) Reactive oxygen D(3) or dexamethasone: differential role for PD-
species regulate activation-induced T cell apo- L1. Eur. J. Immunol. 39:3147–59.
ptosis. Immunity. 10:735–44. 137. Cipolletta D, Kolodin D, Benoist C, Mathis D.
124. Hockenbery DM, Oltvai ZN, Yin XM, Milliman (2011) Tissular Tregs: a unique population of
CL, Korsmeyer SJ. (1993) Bcl-2 functions in an adipose-tissue-resident Foxp3+CD4+ T cells
antioxidant pathway to prevent apoptosis. Cell. that impacts organismal metabolism. Semin. Im-
75:241–51. munol. 23:431–7.
125. Laurent A, et al. (2004) Pivotal role of superox-
ide anion and beneficial effect of antioxidant

1 1 0 | I S S A Z A D E H - N AV I K A S E T A L . | M O L M E D 1 8 : 9 5 - 1 1 0 , 2 0 1 2

You might also like