Skip to main content
Gut logoLink to Gut
. 2006 Oct 15;56(4):584–592. doi: 10.1136/gut.2006.103523

Immunopathogenesis of IBD: insufficient suppressor function in the gut?

I L Huibregtse 1,2, A U van Lent 1,2, S J H van Deventer 1,2
PMCID: PMC1856872  PMID: 17047100

Immune activation and suppression in inflammatory bowel diseases

The intestinal immune system is in a constant state of controlled inflammation, and there is substantial evidence that loss of control is an important pathogenic mechanism in inflammatory bowel diseases (IBD). A major current working hypothesis defines Crohn's disease as a dysregulated immune response towards components of the intestinal flora, leading to chronic intestinal inflammation.1 The causes for this inappropriate response can be attributed to (a combination of) defects in the epithelial barrier, the innate immune response or the adaptive immune response.

Animal experiments as well as clinical data indicate that the immunopathogenesis of Crohn's disease and ulcerative colitis differ at the level of T cell differentiation and activation although the governing mechanisms responsible for these differences have been incompletely defined. In both diseases, activation of T cells is evident but pathogenic T cells in Crohn's disease predominantly produce interferon γ (membrane bound), tumour necrosis factor (TNF)α and interleukin (IL)‐23 whereas ulcerative colitis is characterised by production of IL‐5 and IL‐13.2,3

The increased production of “Th1” type cytokines in Crohn's disease is probably related to increased activation of mucosal dendritic cells and macrophages, and the pivotal function of membrane associated (Toll‐like receptor (TLR)) and intracellular (nucleotide oligomerisation domain (NOD) family) receptors in the activation of these antigen presenting cells (APC) has now been well established. Both receptors are key mediators of innate host defence, crucially involved in maintaining intestinal homeostasis.4 In healthy subjects, the colonic mucosa harbours “non‐inflammatory” dendritic cells, expressing low levels of TLR2 and TLR4 and producing cytokines such as IL‐10, contributing to a non‐inflammatory environment,5,6 but in the mucosa of patients with Crohn's disease the production of IL‐12 is greatly increased.7,8,9 Dendritic cells in both ulcerative colitis and Crohn's disease have an activated phenotype with higher levels of the activation markers CD40 and CD86, and produce more IL‐12 and IL‐6 compared with controls.5,10

The causes for this excessive activation are presumably diverse and have been incompletely defined. A minority of patients with Crohn's disease have inactivating mutations within the susceptibility gene NOD2. The NOD2 protein is normally stimulated by its natural ligand muramyl dipeptide, a degradation product of bacterial peptidoglycan.11,12,13 Some studies have shown increased activation of nuclear factor κB, which in antigen presenting cells causes increased transcription of IL‐12. Indeed, dendritic cells from patients with Crohn's disease with NOD2 mutations produce increased amounts of IL‐12 after stimulation with peptidoglycan, most likely via loss of inhibition of the simultaneously activated TLR2 pathway (Zelinkova et al, submitted). It should be noted that other studies have shown impaired activation of nuclear factor κB in patients with Crohn's disease with NOD2 mutations, suggesting decreased activation.14,15 Hence it remains unclear what mechanism is responsible for the excessive Th1 profile in Crohn's disease and whether the underlying genetic defects lead to initial decreased immune activation with failure to clear pathogens, or whether these mutations directly increase activation of immune cells such as dendritic cells. Abnormal activation and expression of TLR receptors may also be linked to IBD: associations of TLR4 and TLR5 signalling with their bacterial ligands lipopolysaccharide (LPS) and flagellin, respectively, have been reported,16,17 enhanced expression of TLR2 and TLR4 on dendritic cells has been observed5 and recent studies suggest that Crohn's disease is also associated with TLR9 promoter polymorphisms.18

In addition, defective apoptosis of T cells has been suggested to play a role in the pathogenesis and chronic state of inflammation in Crohn's disease. Lamina propria T cells from patients with Crohn's disease were shown to be resistant to activation induced cell death whereas lamina propria T cells from healthy controls readily underwent apoptosis.19,20 The latter would clarify the effective therapeutic action of anti‐TNFα agents such as infliximab in Crohn's disease, as this reagent was shown to induce apoptosis through binding to membrane bound TNFα.21

Although the precise cellular and molecular pathways involved remain to be elucidated, these findings give solid and abundant evidence for increased stimulation or dysregulation of the innate immune system in IBD that, in the case of Crohn's disease, results in induction of hyperactive T cells which is probably necessary for the initiation of chronic mucosal inflammation.

Increased activation of the innate and adaptive mucosal immune systems is tightly controlled by various regulatory circuits, and it is possible that defects in such mechanisms that normally downregulate intestinal inflammation are insufficient in IBD.

This review discusses evidence for abnormal regulation of T cell activation in Crohn's disease, as well as data pertaining to the existence and functional activity of regulatory T cells (Treg) in the intestinal mucosa. We also consider the potential therapeutic application of Treg in IBD.

Regulatory mechanisms in the gut

The immune system controls activation of the innate as well as the adaptive arms through various means, primarily including induction of anergy, apoptosis of activated immune cell and the activities of regulatory CD4+ T cells. In addition, several other regulatory mechanisms are operational in the gut mucosa, including CD8 T cells, γδ T cells and NKT cells, that are highly correlated with their surrounding epithelial cells, and IL‐10 secreting B cells, immature dendritic cells and plasmacytoid dendritic cells.

Intraepithelial CD8 T lymphocytes, γδ T cells and NKT cells are mucosal T cell subsets with a restricted T cell receptor repertoire that are in close contact with mucosal epithelial cells. There is evidence that these interactions can lead to induction of T cells with regulatory capacities: for example, interactions between human intestinal epithelial cells and peripheral blood T cells cause expansion of CD8+CD28 T cells with regulatory activity.22 These cells are present in the lamina propria of healthy individuals but not in the lamina propria of patients with IBD,23 suggesting that these cells prevent the pathogenesis of IBD. Intraepithelial lymphocytes have been reported to downregulate excessive inflammation caused by infection and autoimmunity in epithelial tissues and their protective ability has been shown in several murine models of colitis.24,25,26,27,28

Accumulating evidence implicates additional cell types in immunoregulation. Recent studies have revealed a protective role of IL‐10 producing B cells in murine CD4+ T cell colitis29 and inhibition of antigen specific T cell proliferation by plasmacytoid dendritic cells. This dendritic cell population has a marked presence in mucosal tissue and is, in common with steady state lamina propria immature dendritic cells, able to induce a non‐anergic state of T cell unresponsiveness that involves the differentiation of Treg30,31

These data indicate that mucosal immune activation is regulated at various levels by different cells that downregulate immune responses. A rapidly expanding body of evidence indicates that the most important among these regulatory cells reside within the CD4+ T cell population, and these will be the further focus of this review.

Regulatory CD4+ T cells: phenotype, function and regulation

Once T cells are activated through engagement of the T cell receptor, they do not distinguish between “self” and “non‐self”. It is now clear that the human immune system regards antigens expressed by the normal gut flora as “self”. Because activated T cells that recognise self‐antigens induce significant tissue damage, it is important to either prevent their activation or control proliferation. It has long been known that most high affinity self‐reactive T cells are clonally deleted within the thymus but this system is leaky and by itself insufficient to prevent autoreactivity. Hence prevention of autoreactivity is also continuously controlled outside the thymus, and this “peripheral tolerance” is critically dependent on the presence of Treg.

Although, in general, microbes mount strong immune responses, the resident gut flora is unable to activate T cells in healthy individuals. Thus there should be a mechanism by which potentially detrimental immune responses in the gut are prevented.

Regulatory CD4+ T cells represent a population of lymphocytes with the ability to suppress both adaptive and innate immune responses (fig 1),32,33,34 and these characteristics make them important for both maintenance of immunological tolerance and control of antimicrobial responses. Various types of Treg have been identified (table 1), but because specific phenotypic markers have long been lacking, it is uncertain to what extent these Treg constitute separate lineages. Nonetheless, Treg can be divided into two major groups, the so‐called “naturally occurring” Treg and “adaptive” Treg, containing the so‐called Tr1 and Th3 cells.35

graphic file with name gt103523.f1.jpg

Figure 1 Regulatory T cells and their function. Several types of regulatory T cells (Treg) have been identified and the mechanisms of suppression may differ. Thymus derived regulatory T cells, also known as naturally occurring regulatory T cells, are a subset of CD4+CD25+ T cells and are thought to suppress activation of T cells at level of antigen presenting cell. Adaptive peripheral induced regulatory T cells include Tr1, Th3 and CD8. These cells produce the immunosuppressive cytokines interleukin (IL)‐10 and/or transforming growth factor (TGF)β and function in a cytokine dependent manner. IFN‐γ, interferon γ.

Table 1 Characteristics of the different regulatory T cell subsets.

Feature Naturally Treg Adaptive Treg
Subpopulations CD4+CD25+ Tr1 Th3
Site of induction Thymus Periphery
Mechanism of action Cell–cell contact, cytokine independent Cytokine dependent
Characterisation CD25+ and Foxp3+ IL‐10 TGF‐β
Specificity Self‐antigens in the thymus Tissue specific antigens and foreign antigens
Protection demonstrated Transfer colitis, SCID Transfer colitis, SCID Neutralising TGF‐β antibodies

IL, interleukin; TGF‐β, transforming growth factor β; Treg, regulatory T cells.

“Naturally occurring” regulatory T cells

Most CD4+ T cells that recognise autoantigens in the thymus with high affinity are either clonally deleted or differentiate into a “naturally occurring” Treg. This cell is characterised by a unique phenotype and potent suppressive function towards autoreactive peripheral T cells. Thymus derived Treg constitute about 5–10% of mouse and 1–2% of human peripheral CD4+ T cells. Initially, these cells were identified by their CD4+CD25high phenotype but an increasing number of markers has been recently reported (table 2). Several membrane expressed molecular markers such as CD25 (IL‐2 receptor α chain), glucocorticoid induced TNFR family related protein (GITR) and cytotoxic T lymphocyte associated antigen 4 (CTLA‐4) are constitutively expressed on Treg but can also be observed on activated non‐regulatory T cells, and it was not until the discovery of the foxp3 gene (FOXP3 in humans) that a unique marker for murine Treg was identified. Mutations in Foxp3 result in severe autoimmune reactivity in both mice and humans, leading to, respectively, the scurfy or IPEX (immune dysregulation, polyendocrinopathy, enteropathy, x‐linked) syndrome. The foxp3 gene was identified as a master regulatory gene; it is constitutively and specifically expressed in natural Treg and plays an indispensable role in their development and function. Furthermore, forced expression of foxp3 can convert naïve peripheral blood T cells to Treg cells.36 The specificity of foxp3 in mice is clear; it is solely expressed in Treg and the scurfy mutation is always related to defective suppressive function. Conversely, the expression of FOXP3 in humans is not restricted to Treg and can be induced on activation of conventional T cells, albeit at much lower levels than in natural Treg. To add to the confusion, it has been reported that IPEX patients have varying degrees of disease severity and not all patients have dysfunctional Treg.37 Even with these restrictions, there is general consensus that FOXP3 expression is highly correlated to the suppressive function of CD4+ CD25high T cells.

Table 2 Cell surface and intracellular markers constitutively expressed by thymus derived natural regulatory T cells.

Species Cell surface Intracellular
Murine CD25+, CD122+, CD69+, CD44+, CD45RBlow, GITR+, CD103+(αE‐integrin), CD134+(OX‐40), CD54+(ICAM) CTLA‐4+, Foxp3+
Human CD25high, CD122+, HLA‐DR+(50%), CD45RO+(80%), CD95high, CD45RBlow, CD38low, partly CD62low, GITR+,CD127low CTLA‐4+, FOXP3+

It has recently become apparent that expression of the α‐chain of the IL‐7 receptor, CD127, allows an unambiguous flow cytometry based distinction of Treg (CD127low) and non‐regulatory T cells (CD127high) within the CD4+CD25+ populations. CD127low cells were strongly suppressive in functional suppressor assays and expression of FOXP3 protein was highly correlated to a CD127low phenotype.38,39 These findings are important because human Treg can now be accurately identified and isolated.

Treg were originally thought to be anergic when stimulated ex vivo, yet adoptive transfer studies using (DO11.10) T cell receptor (TCR) transgenic CD4+CD25+ cells have clearly demonstrated the ability of these cells to expand in vivo on TCR stimulation.40 Recently, it has been shown that human Treg can be greatly expanded ex vivo by TCR stimulation in the presence of high concentrations of IL‐2 as CD25 is functionally essential as a key component of the high affinity IL‐2 receptor,41,42 largely increasing their potential for therapeutic manipulation. The exact mechanism of suppression by Treg remains uncertain. In vitro, the suppressive function can be assessed by coculturing Treg with conventional CD4 (or CD8) T cells ( “responder” T cells) in a mixed leucocyte reaction. The proliferation of conventional T cells in such assays is induced via TCR stimulation by allogeneic peripheral blood mononuclear cells (PBMCs) or agonistic anti‐CD3 antibodies. In the presence of Treg, in ratios below 1/10, the proliferation of the responder T cells and their cytokine production is strongly suppressed. In these in vitro studies, the suppressive function is cell contact dependent and independent of cytokines. However, mouse studies have proven the suppression to be dependent on cytokines such as transforming growth factor (TGF)β and IL‐10. The mechanisms responsible for these differences between in vivo and in vitro results remain to be fully explained.

Treg execute suppressive functions as soon as they are activated via the TCR, aspecifically (CD28, CD3), by a natural (HLA class II presented) ligand or by foreign antigens that are cross reactive to self‐antigen receptors43 in the periphery. Treg not only suppress proliferation but also downregulate activation, differentiation and even effector function of multiple immune cells, including CD4+ and CD8+ T cells, natural killer cells and dendritic cells.44,45,46

APC are able to regulate Treg activation by differential expressed costimulatory molecules and MHCII molecules. Although the precise mechanism of CTLA‐4 expression and its involvement on Treg is not known,47 it is thought that engagement of CTLA‐4 by CD80/CD86 on dendritic cells activates Treg, whereas interaction with CD28, in the context of TCR activation, downregulates suppression. In addition, activation of GITR by GITR ligand (which is expressed on dendritic cells) downregulates Treg function.48 An integrin that is expressed by dendritic cells, CD103, is also involved in T cell polarisation, promoting a positive balance of Treg over effector T cell activity in the intestine.49 Interestingly, the very same stimuli (ie, LPS) that cause APC such as dendritic cells to become activated and present antigen, have a direct effect on Treg. Murine CD4+CD25+ Treg express TLR1, TLR 2 and TLR 4–8, and activation of TLR‐4 and TLR‐5 by LPS and flagellin, respectively, activates Treg and increases suppressor function in vitro.50,51,52 Therefore, TLR activation of Treg seems to counteract uncontrolled activation of T cell proliferation. Conversely, natural ligands for TLR8 and TLR2 can reverse Treg function.53,54

The functional importance of Treg is underscored by many observations in mice where depletion of the CD4+CD25+ population precipitates diseases characterised by autoreactive T cells.55,56,57,58,59

“Adaptive” regulatory T cells

Apart from the CD4+CD25+ thymus derived Treg, there is evidence for the existence of Treg that are induced in the periphery, the so‐called “adaptive” Treg. In mice and humans, peripheral conventional CD4+ T cells were shown to differentiate into CD4+CD25+ Treg under the influence of TGFβ in addition to TCR mediated signals.60,61,62 Alternatively, adaptive Treg which are phenotypically distinct from Treg from intrathymic origin have been identified, and known as the Th3 and Tr1 T cell subsets. These generally do not express CD25 or foxp3 and are characterised by the secretion of the immunosuppressive cytokines TGFβ and IL‐10, respectively. Although their functions are complex and incompletely understood, it seems that their suppressive activity is critically dependent on the production of regulatory cytokines.

A classical example of peripheral regulatory cells is the Th3 Treg that secretes predominantly TGFβ together with varying amounts of IL‐4 and IL‐10, and mediates oral tolerance.63,64 The main immunosuppressive effect of TGFβ is inhibition of Th1 responses via downregulation of IL‐12β2 chain expression, and TGFβ itself is required for differentiation of TGFβ producing cells. Th3‐like cells have been shown to be important in some cases of allergy and in autoimmune diseases.65,66

Tr1 cells were initially isolated from human SCID chimera and subsequently derived by culturing naïve T cells in the presence of high concentrations of IL‐10. They secrete high levels of IL‐10, a cytokine that inhibits Th1 induction by downregulation of IL‐12, and suppress the production of proinflammatory effector cytokines. Tr‐1 cells are anergic, functionally suppressive in vitro, generally produce low levels of TGFβ and IL‐5 but no IL‐4, and are critically dependent on IL‐10 for their function and development.67 In SCID patients transplanted with HLA mismatched haematopoietic stem cells, the number of Tr1 cells correlated with tolerance of the host to the graft.37

Proliferation of murine Tr1 cells in vivo is induced by plasmacytoid dendritic cells that express low numbers of CD11c and costimulatory molecules, and secrete large amounts of IL‐10.68 Human Tr1 cells can be induced ex vivo with the pharmacological immunosuppressant vitamin D3 and dexamethasone69 and by immature (CD83) dendritic cells.70 In contrast with the latter observations, we have demonstrated that induction of Treg that result from activation of monocyte derived CD11c+ dendritic cells by probiotic bacteria requires full maturation of the dendritic cells (Braat H et al, submitted). Induction of these Treg is dependent on production of IL‐10 by the mature dendritic cells, and although these Treg also secrete IL‐10, this is not required for their regulatory function.

In summary, peripheral Treg comprise a heterogeneous group of T cells that secrete immunomodulatory cytokines that have been implicated in various inflammatory conditions.

Regulatory T cells in experimental colitis

It is well known that adoptive transfer of T cells depleted of CD4+CD25+ cells in immunodeficient mice causes multiorgan autoimmunity in the recipient animals71 and many studies have demonstrated that depletion of CD4+CD25+ T cells in mice aggravates T cell mediated models of inflammation,72 including colitis.73 Conversely, Treg clearly have anti‐inflammatory effects in various murine models of IBD. For example, the induction of colitis that results from transfer of CD4+CD45RBhigh T cells into immunodeficient mice can be prevented by cotransfer of the antigen experienced CD4+CD45RBlow T cells. Thymus derived Treg are CD45RBlow, and it is now thought that the CD4+CD25+ Treg present in the CD4+CD45RBlow subset are responsible for this regulatory activity.74,75 Cotransfer with isolated CD4+CD25+ T cells prevents the induction of colitis which is reverted by the addition of monoclonal anti‐CTLA‐4, anti‐IL‐10R or anti‐TGFβ antibodies. Not only do CD4+CD25+ T cells prevent the induction of colitis, they can also reverse established colitis and wasting disease, indicating their importance in controlling ongoing immune mediated inflammation.76

Peripheral Treg with a Tr1 phenotype also have the capacity to control colitis. Chronic activation of OVA specific naïve CD4+ T cells in the presence of IL‐10 induced Tr1 cells that produced large amounts of IL‐10 after exposure to OVA. These cells were able to control colitis induced by pathogenic CD4+CD45RBhigh T cells in immunodeficient mice, and this function was dependent on activation of the T cell receptor by OVA.77,78 Mouse strains deficient in IL‐10 spontaneously develop chronic enterocolitis, underlining the importance of IL‐10 in controlling responses against the commensal flora.79,80,81 Treatment with recombinant IL‐10 in the T cell transfer model prevents but does not cure established colitis. In contrast, local mucosal delivery of recombinant IL‐10 by the genetically modified bacteria of the strain L lactis seemed to be effective when disease activity was well established, and ameliorated DSS induced colitis and colitis in IL‐10 deficient mice.82

TGFβ is also recognised for its ability to downregulate immune responses. TGFβ1 deficient mice develop a systemic inflammatory response, and blockade of TGFβ signalling in T cells results in T cell activation and induction of IBD in mice.83 CD4+ Th3 cells protect against uncontrolled inflammation in the gut, and in models of intestinal inflammation TGFβ producing mucosal T cells were shown to reduce disease activity.84,85,86

The role of regulatory T cells in inflammatory bowel diseases

Because it has long been difficult to reliably characterise Treg, all data about their presence and functional characteristics in humans are recent, and some of these data require confirmation. Several clinical observations indicate that the CD4+CD25+ population in patients with “autoimmune” diseases such as multiple sclerosis, uveitis and autoimmune polyglandular syndromes II is functionally defective.87 In patients with Crohn's disease, approximately 6% of both peripheral blood and lamina propria T cells were found to be CD4+CD25+, and the fraction with a high expression of CD25 (CD25bright) expressed CTLA‐4 and GITR. In contrast with peripheral blood T cells, some expression of CTLA‐4 and GITR was found on lamina propria CD4+CD25 T cells. In agreement with this finding, Foxp3 was predominantly transcribed by CD4+CD25bright lamina propria T cells, and to a lesser extent by CD4+CD25 T cells. When tested for functional properties, it was found that lamina propria CD4+CD25bright T cells, but not CD4+CD25 T cells, suppressed the proliferation (as well as cytokine production) of peripheral blood CD4+CD25 T cells88,89 but not of lamina propria CD4+CD25 T cells. The inability of lamina propria Treg to suppress proliferation of lamina propria T cells may be related to the relative anergic and memory phenotype of the latter.85 Likewise, CD4+CD25+ T cells isolated from human colonic mesenterial lymph node in ulcerative colitis display typical features of Treg cells and possess potent suppressor activity in vitro in spite of persistent mucosal inflammation {unpublished results, Saruta M et al, DDW 2006, abstract 599}.

At present, these sparse data on Treg in IBD suggest that the inflammatory pathology in patients with Crohn's disease does not result from an absence or altered functionality of the Treg population although the increase in Treg numbers and activity may be insufficient to suppress the inflammatory condition.90

There are no reliable data on the existence of Tr1 or Th3 cells in the human mucosa, or on their functional properties. IL‐10 deficient mice develop IBD but patients with IBD do not have deficient IL‐10 production.91 Remarkably, isolated T cells from patients with IBD were found to express high levels of SMAD7, a negative regulator of TGFβ signalling, suggesting that impaired responsiveness to TGFβ may be involved in IBD.92,93 The functional role of lamina propria Treg may be more complex than that of peripheral regulatory cells because of the necessity to specifically suppress immune responses to endogenous bacteria but not to bacterial pathogens.

Therapeutic potential of regulatory T cells

As described above, Treg can prevent and even cure various experimental colitis models. Although their therapeutic potential is without dispute, translation of these data into therapeutic strategies is not straightforward. Furthermore, the ability to apply this therapeutic strategy in a human clinical setting will depend on techniques to isolate and transfer adequate numbers of cells. In most mouse models of autoimmune diseases, the antigens that induce T cell activation are known, and antigen specific Treg are able to potently suppress activation in an antigen dependent manner. For example, in the NOD model of autoimmune diabetes, islet antigen specific BDC2.5 Treg completely prevent diabetes. However, polyclonal Treg are at least 50‐fold less potent than antigen specific Treg and can only be a viable therapeutic option in this context when sufficient numbers are applied.94

Although there is evidence for a role of peptidoglycan and flagellin at the level of dendritic cell stimulation, it is not known what antigens are involved in the pathogenesis of IBD, excluding the possibility to using antigen specific Treg. It is now clear that Treg do not need to be antigen specific in order to suppress immune responses as a result of so‐called bystander suppression (fig 2). A clear example of bystander suppression was demonstrated in the SCID transfer model where OVA specific Tr1 cells suppressed the occurrence of IBD after administration of OVA although OVA is not involved in the immune mediated inflammation in this model.95 Therefore, the OVA specific Tr1 cells were able to suppress responses induced by other antigens, very likely derived from intestinal bacteria, and this is known as “bystander” suppression. In various situations CD4+CD25+ Treg, once activated by their TCR, have been shown to be capable of such antigen‐non‐specific bystander suppression.96

graphic file with name gt103523.f2.jpg

Figure 2 Bystander suppression. Presentation of bacterial antigens to naive T cells by dendritic cells results in the generation of Th1 effector cells that migrate into the intestine and cause an inflammatory response. Because the antigens that are involved in inflammatory bowel disease are unknown, therapeutic application of Treg requires antigen‐non‐specific suppression. Bystander suppression is the capacity of Treg to suppress immune responses that are caused by a different antigen. The mechanisms involved include the production of regulatory cytokines, deactivation of dendritic cells that attempt to stimulate effector T cells, or direct contact with the responding T cell. The concept of bystander suppression has been shown for Tr1, CD4+CD25+, Th3 and CD8+ T cells. IFNγ, interferon γ; IL, interleukin; TGFβ, transforming growth factor β; TNF, tumour necrosis factor.

Apart from bystander suppression, Treg inhibit the response of conventional CD4 T cells in a contact dependent manner and can even confer suppressive properties to such T cells. This process is known as “infectious tolerance” and results in the conversion of conventional T cells into IL‐10 producing Tr1‐like cells and TGFβ producing Th3‐like cells.97 These concepts of “infectious tolerance” and bystander suppression are instrumental in providing a context for using Treg as a potential therapy.

It has now become possible to produce and expand sufficient CD4+CD25+ and Tr1‐like cells for therapeutic application, and clinical studies have been initiated. For example, in a currently ongoing clinical trial, Roncarolo et al (personal communication) used ex vivo induced Tr1 cells as post‐transplant cellular therapy in haematological cancer patients undergoing HLA‐haploidentical HSC transplantation. After a 10 day ex vivo culture of donor PBMC in the presence of irradiated host PBMC and IL‐10, the IL‐10 anergised donor T cells are infused into the host. The ultimate goal is to provide immune reconstitution with donor T cells that are anergic towards host antigens and contain precursors of host specific Tr1 cells. Although promising, the clinical usage of Tr1 cells for the cure of T cell mediated diseases is still in a developmental stage.77

A second important observation has been that regulatory functions can be imprinted in mouse and human T cells by genetic engineering. Unselected peripheral blood naïve mouse T cells become regulatory following transfection with a retroviral vector encoding IL‐10, and these cells are able to suppress inflammation by a bystander mechanism.98 Such cells can also be generated from human peripheral T cells.99 Using similar techniques, FOXP3 can be overexpressed in human CD4+ T cells but the data on the functional efficacy of these generated suppressor T cells are conflicting.100,101

Finally, it may be possible to induce Treg in vivo by directing APC such as dendritic cells. It appears that the capacity of dendritic cells to induce regulatory T cells depends on the dendritic cell instruction and maturation state.102 Different approaches to render “regulatory” dendritic cells103,104 include ex vivo genetic manipulation, anti‐inflammatory cytokine exposure or by direct instruction with tolerogenic compounds. We have recently demonstrated that injection of the Bordetella pertussis derived filamentous haemagglutinin A reduced inflammation in a mouse model of IBD.105 The experimental data of T cell as well as dendritic cell manipulation, along with future investigations, needs to determine the exact value of both approaches but recent advances are very promising.

When considering ex vivo manipulation or induction of Treg, two major hurdles need to be overcome.

Firstly, sufficient numbers of the manipulated T cells need to be directed to the gut mucosa, a process known as “homing”, which is directed by specific integrins and by chemokines. It has been reported that cultured CD4+ gut derived T cells that express high levels of the pivotal gut homing receptor α4β7 did not home to the gut following injection in healthy individuals.89 However, we have reported that homing, at least in mice and rats, is greatly increased in inflammatory conditions. Also, it has become apparent that the isolation and expansion of CD45RA+ naïve (instead of CD45RO+) CD4+CD25+ T cells is the best strategy for adoptive Treg cell therapies.106

Secondly, it might be a problem that the bystander suppression of adoptively transferred Treg cannot be controlled. Treg may worsen inflammatory disease because they may interfere with the immune mechanisms that are necessary for clearance of microbial pathogens.107 The non‐specific immunosuppressive effects of Treg are a concern when considering therapeutic application. On the other hand, such effects may be limited because effective pathogen specific immune responses are shown to be Treg resistant.108 However, from this perspective the use of natural occurring CD4+CD25high Treg may be preferred, as in these cells, at least in vitro, TLR2 triggering results in a temporal loss of the suppressive Treg phenotype.54

Conclusions

Regulatory T cells are key players of immune regulation, and they have important functions in suppressing unwanted inflammatory responses towards self antigens, and the antigens of endogenous intestinal bacteria. Patients with IBD do not seem to have a primary absolute defect in regulatory cells but, apparently, the regulatory capacity of these cells is insufficient to downregulate inflammation.

None of the current therapies for IBD directly targets Treg function or generation but drugs that are used widely clinically may influence Treg function. For example, corticosteroids may enhance Treg function in asthma and allergic diseases109,110,111 but future research needs to determine the exact role of corticosteroids on regulatory T cell function. Interestingly, it was recently shown that anti‐TNF antibodies increased the FOXP3 mRNA and protein levels in the CD4+CD25high compartment and restored their tolerogenic function.112 In mice and humans with diabetes, treatment with non‐agonistic anti‐CD3 antibodies resulted in prevention of progression of loss of islet cell function by immunosuppressive mechanisms that included the induction of Treg.113

None of the previously discussed possible clinical approaches uses a strategy that allows control over Treg activity. Are such approaches feasible?

Intravenous administration of relatively large doses of regulatory cytokines is not effective (human recombinant IL‐10 administration in IBD patients114) or found to be toxic (TGFβ). Only a small fraction of the total administered dose of such cytokines reaches the mucosa and results may be better when such cytokines are locally administered. Mucosal delivery of recombinant IL‐10 by genetically modified bacteria such as L lactis addresses this problem and was indeed shown to ameliorate DSS colitis and colitis in IL‐10−/− mice. Recently, we have demonstrated in a clinical phase 1 trial that this engineered cytokine excreting organism can be safely administered to patients with Crohn's disease and is biologically contained.115 TGFβ can also be locally expressed, for example by delivery of TGFβ encoding plasmids to mucosal surfaces and this approach is in preclinical trials.116

Summary

  • Activation of mucosal T cells occurs in IBD and is inhibited by various subsets of “regulatory” T cells, which can be functionally and phenotypically characterised.

  • Regulatory T cells can be generated within the thymus or in peripheral tissues.

  • In mice, loss of regulatory T cell activity results in IBD. Regulatory T cell activity has not been thoroughly studied in IBD but currently available data do not provide evidence for a loss of regulatory T cell activity in human IBD.

  • Regulatory T cells can be generated or activated in vivo and in vitro using a variety of approaches, including specific small molecules, cytokine mediated activation, and gene therapy. These cells have therapeutic activity in the preclinical setting.

Contained immunosuppression can also be accomplished by modifying or expanding T cells with regulatory properties ex vivo. This strategy involves a harvesting step that yields peripheral blood T lymphocytes (eg, by apheresis) followed by forced differentiation by exposure to cytokines and tolerogenic compounds, or by genetic engineering with a “regulatory” gene. On readministration to the patient, such cells can downregulate inflammation by a bystander mechanism, following specific integrin mediated homing. It is technically feasible to specifically expand T cells with a predefined TCR that can be specifically activated by an orally administered antigen, allowing for control of the immune suppression. These strategies are attractive in view of the long lifespan of T cells, and are expected to have long term effects.

Treg are extremely potent and, with production of low amounts of IL‐10 by a very small fraction of mucosal T cells (IL‐10 engineered T cells that comprised only ∼ 0.001% of all mucosal T cells were effective), a sufficient therapeutic effect can be achieved.117 With the identification of more genes that determine Treg development, the ability to identify Treg using cell surface markers, and with improving transduction methods over time, the possibilities for such approaches will be significantly expanded.

Acknowledgements

We would like to thank Professor Dr H Spits for critical reading of the manuscript.

Footnotes

Competing interests: None.

References

  • 1.Bouma G, Strober W. The immunological and genetic basis of inflammatory bowel disease. Nat Rev Immunol 20033521–533. [DOI] [PubMed] [Google Scholar]
  • 2.Krajina T, Leithauser F, Moller P.et al Colonic lamina propria dendritic cells in mice with CD4+ T cell‐induced colitis. Eur J Immunol 2003331073–1083. [DOI] [PubMed] [Google Scholar]
  • 3.Vainer B, Nielsen O H, Hendel J.et al Colonic expression and synthesis of interleukin 13 and interleukin 15 in inflammatory bowel disease. Cytokine 2000121531–1536. [DOI] [PubMed] [Google Scholar]
  • 4.Cario E. Bacterial interactions with cells of the intestinal mucosa: Toll‐like receptors and NOD2. Gut 2005541182–1193. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Hart A L, Al Hassi H O, Rigby R J.et al Characteristics of intestinal dendritic cells in inflammatory bowel diseases. Gastroenterology 200512950–65. [DOI] [PubMed] [Google Scholar]
  • 6.Rimoldi M, Chieppa M, Salucci V.et al Intestinal immune homeostasis is regulated by the crosstalk between epithelial cells and dendritic cells. Nat Immunol 20056507–514. [DOI] [PubMed] [Google Scholar]
  • 7.Matsuoka K, Inoue N, Sato T.et al T‐bet upregulation and subsequent interleukin 12 stimulation are essential for induction of Th1 mediated immunopathology in Crohn's disease. Gut 2004531303–1308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Monteleone G, Biancone L, Marasco R.et al Interleukin 12 is expressed and actively released by Crohn's disease intestinal lamina propria mononuclear cells. Gastroenterology 19971121169–1178. [DOI] [PubMed] [Google Scholar]
  • 9.Parrello T, Monteleone G, Cucchiara S.et al Up‐regulation of the IL‐12 receptor beta 2 chain in Crohn's disease. J Immunol 20001657234–7239. [DOI] [PubMed] [Google Scholar]
  • 10.Kelsall B L, Leon F. Involvement of intestinal dendritic cells in oral tolerance, immunity to pathogens, and inflammatory bowel disease. Immunol Rev 2005206132–148. [DOI] [PubMed] [Google Scholar]
  • 11.Cho J H. The Nod2 gene in Crohn's disease: implications for future research into the genetics and immunology of Crohn's disease. Inflamm Bowel Dis 20017271–275. [DOI] [PubMed] [Google Scholar]
  • 12.Ogura Y, Bonen D K, Inohara N.et al A frameshift mutation in NOD2 associated with susceptibility to Crohn's disease. Nature 2001411603–606. [DOI] [PubMed] [Google Scholar]
  • 13.Hugot J P, Chamaillard M, Zouali H.et al Association of NOD2 leucine‐rich repeat variants with susceptibility to Crohn's disease. Nature 2001411599–603. [DOI] [PubMed] [Google Scholar]
  • 14.Inohara N, Ogura Y, Fontalba A.et al Host recognition of bacterial muramyl dipeptide mediated through NOD2. Implications for Crohn's disease. J Biol Chem 20032785509–5512. [DOI] [PubMed] [Google Scholar]
  • 15.Li J, Moran T, Swanson E.et al Regulation of IL‐8 and IL‐1beta expression in Crohn's disease associated NOD2/CARD15 mutations. Hum Mol Genet 2004131715–1725. [DOI] [PubMed] [Google Scholar]
  • 16.Franchimont D, Vermeire S, El Housni H.et al Deficient host–bacteria interactions in inflammatory bowel disease? The toll‐like receptor (TLR)‐4 Asp299gly polymorphism is associated with Crohn's disease and ulcerative colitis. Gut 200453987–992. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Lodes M J, Cong Y, Elson C O.et al Bacterial flagellin is a dominant antigen in Crohn disease. J Clin Invest 20041131296–1306. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Torok H P, Glas J, Tonenchi L.et al Crohn's disease is associated with a toll‐like receptor‐9 polymorphism. Gastroenterology 2004127365–366. [DOI] [PubMed] [Google Scholar]
  • 19.Boirivant M, Pica R, DeMaria R.et al Stimulated human lamina propria T cells manifest enhanced Fas‐mediated apoptosis. J Clin Invest 1996982616–2622. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.De Maria R, Boirivant M, Cifone M G.et al Functional expression of Fas and Fas ligand on human gut lamina propria T lymphocytes. A potential role for the acidic sphingomyelinase pathway in normal immunoregulation. J Clin Invest 199697316–322. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Peppelenbosch M P, van Deventer S J. T cell apoptosis and inflammatory bowel disease. Gut 2004531556–1558. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Allez M, Brimnes J, Dotan I.et al Expansion of CD8+ T cells with regulatory function after interaction with intestinal epithelial cells. Gastroenterology 20021231516–1526. [DOI] [PubMed] [Google Scholar]
  • 23.Brimnes J, Allez M, Dotan I.et al Defects in CD8+ regulatory T cells in the lamina propria of patients with inflammatory bowel disease. J Immunol 20051745814–5822. [DOI] [PubMed] [Google Scholar]
  • 24.Hayday A, Tigelaar R. Immunoregulation in the tissues by gammadelta T cells. Nat Rev Immunol 20033233–242. [DOI] [PubMed] [Google Scholar]
  • 25.Chen Y, Chou K, Fuchs E.et al Protection of the intestinal mucosa by intraepithelial gamma delta T cells. Proc Natl Acad Sci U S A 20029914338–14343. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Guy‐Grand D, Cerf‐Bensussan N, Malissen B.et al Two gut intraepithelial CD8+ lymphocyte populations with different T cell receptors: a role for the gut epithelium in T cell differentiation. J Exp Med 1991173471–481. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Menachem Y, Trop S, Kolker O.et al Adoptive transfer of NK 1. 1+ lymphocytes in immune‐mediated colitis: a pro‐inflammatory or a tolerizing subgroup of cells, Microbes Infect 20057825–835. [DOI] [PubMed] [Google Scholar]
  • 28.Saubermann L J, Beck P, De Jong Y P.et al Activation of natural killer T cells by alpha‐galactosylceramide in the presence of CD1d provides protection against colitis in mice. Gastroenterology 2000119119–128. [DOI] [PubMed] [Google Scholar]
  • 29.Dalwadi H, Wei B, Schrage M.et al B cell developmental requirement for the G alpha i2 gene. J Immunol 20031701707–1715. [DOI] [PubMed] [Google Scholar]
  • 30.Bilsborough J, George T C, Norment A.et al Mucosal CD8alpha+ DC, with a plasmacytoid phenotype, induce differentiation and support function of T cells with regulatory properties. Immunology 2003108481–492. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Turnbull E L, Yrlid U, Jenkins C D.et al Intestinal dendritic cell subsets: differential effects of systemic TLR4 stimulation on migratory fate and activation in vivo. J Immunol 20051741374–1384. [DOI] [PubMed] [Google Scholar]
  • 32.Lim H W, Hillsamer P, Banham A H.et al Cutting edge: direct suppression of B cells by CD4+ CD25+ regulatory T cells. J Immunol 20051754180–4183. [DOI] [PubMed] [Google Scholar]
  • 33.Maloy K J, Antonelli L R, Lefevre M.et al Cure of innate intestinal immune pathology by CD4+CD25+ regulatory T cells. Immunol Lett 200597189–192. [DOI] [PubMed] [Google Scholar]
  • 34.Sakaguchi S. Naturally arising CD4+ regulatory t cells for immunologic self‐tolerance and negative control of immune responses. Annu Rev Immunol 200422531–562. [DOI] [PubMed] [Google Scholar]
  • 35.Bluestone J A, Abbas A K. Natural versus adaptive regulatory T cells. Nat Rev Immunol 20033253–257. [DOI] [PubMed] [Google Scholar]
  • 36.Hori S, Nomura T, Sakaguchi S. Control of regulatory T cell development by the transcription factor Foxp3. Science 20032991057–1061. [PubMed] [Google Scholar]
  • 37.Bacchetta R, Passerini L, Gambineri E.et al Defective regulatory and effector T cell functions in patients with FOXP3 mutations. J Clin Invest 20061161713–1722. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Liu W, Putnam A L, Xu‐Yu Z.et al CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4(+) T reg cells. J Exp Med 20062031701–1711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Seddiki N, Santner‐Nanan B, Martinson J.et al Expression of interleukin (IL)‐2 and IL‐7 receptors discriminates between human regulatory and activated T cells. J Exp Med 20062031693–1700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Walker L S, Chodos A, Eggena M.et al Antigen‐dependent proliferation of CD4+ CD25+ regulatory T cells in vivo. J Exp Med 2003198249–258. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Earle K E, Tang Q, Zhou X.et al In vitro expanded human CD4+CD25+ regulatory T cells suppress effector T cell proliferation. Clin Immunol 20051153–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Setoguchi R, Hori S, Takahashi T.et al Homeostatic maintenance of natural Foxp3(+) CD25(+) CD4(+) regulatory T cells by interleukin (IL)‐2 and induction of autoimmune disease by IL‐2 neutralization. J Exp Med 2005201723–735. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Suffia I J, Reckling S K, Piccirillo C A.et al Infected site‐restricted Foxp3+ natural regulatory T cells are specific for microbial antigens. J Exp Med 2006203777–788. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Ghiringhelli F, Menard C, Terme M.et al CD4+CD25+ regulatory T cells inhibit natural killer cell functions in a transforming growth factor‐beta‐dependent manner. J Exp Med 20052021075–1085. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45.Sakaguchi S. Naturally arising Foxp3‐expressing CD25+CD4+ regulatory T cells in immunological tolerance to self and non‐self. Nat Immunol 20056345–352. [DOI] [PubMed] [Google Scholar]
  • 46.von Boehmer H. Mechanisms of suppression by suppressor T cells. Nat Immunol 20056338–344. [DOI] [PubMed] [Google Scholar]
  • 47.Izcue A, Coombes J L, Powrie F. Regulatory T cells suppress systemic and mucosal immune activation to control intestinal inflammation. Immunol Rev 2006212256–271. [DOI] [PubMed] [Google Scholar]
  • 48.Kim J D, Choi B K, Bae J S.et al Cloning and characterization of GITR ligand. Genes Immun 20034564–569. [DOI] [PubMed] [Google Scholar]
  • 49.Annacker O, Coombes J L, Malmstrom V.et al Essential role for CD103 in the T cell‐mediated regulation of experimental colitis. J Exp Med 20052021051–1061. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Caramalho I, Lopes‐Carvalho T, Ostler D.et al Regulatory T cells selectively express toll‐like receptors and are activated by lipopolysaccharide. J Exp Med 2003197403–411. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Netea M G, Sutmuller R, Hermann C.et al Toll‐like receptor 2 suppresses immunity against Candida albicans through induction of IL‐10 and regulatory T cells. J Immunol 20041723712–3718. [DOI] [PubMed] [Google Scholar]
  • 52.Crellin N K, Garcia R V, Hadisfar O.et al Human CD4+ T cells express TLR5 and its ligand flagellin enhances the suppressive capacity and expression of FOXP3 in CD4+CD25+ T regulatory cells. J Immunol 20051758051–8059. [DOI] [PubMed] [Google Scholar]
  • 53.Peng G, Guo Z, Kiniwa Y.et al Toll‐like receptor 8‐mediated reversal of CD4+ regulatory T cell function. Science 20053091380–1384. [DOI] [PubMed] [Google Scholar]
  • 54.Sutmuller R P, den Brok M H, Kramer M.et al Toll‐like receptor 2 controls expansion and function of regulatory T cells. J Clin Invest 2006116485–494. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 55.Morgan M E, Sutmuller R P, Witteveen H J.et al CD25+ cell depletion hastens the onset of severe disease in collagen‐induced arthritis. Arthritis Rheum 2003481452–1460. [DOI] [PubMed] [Google Scholar]
  • 56.Morgan M E, Witteveen H J, Sutmuller R P.et al CD25+ regulatory cells from HLA‐DQ8 transgenic mice are capable of modulating collagen‐induced arthritis. Hum Immunol 2004651319–1327. [DOI] [PubMed] [Google Scholar]
  • 57.Mqadmi A, Zheng X, Yazdanbakhsh K. CD4+CD25+ regulatory T cells control induction of autoimmune hemolytic anemia. Blood 20051053746–3748. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Sugihara S, Fujiwara H, Shearer G M. Autoimmune thyroiditis induced in mice depleted of particular T cell subsets. Characterization of thyroiditis‐inducing T cell lines and clones derived from thyroid lesions. J Immunol 1993150683–694. [PubMed] [Google Scholar]
  • 59.Takeuchi M, Keino H, Kezuka T.et al Immune responses to retinal self‐antigens in CD25(+)CD4(+) regulatory T‐cell‐depleted mice. Invest Ophthalmol Vis Sci 2004451879–1886. [DOI] [PubMed] [Google Scholar]
  • 60.Chen W, Jin W, Hardegen N.et al Conversion of peripheral CD4+. J Exp Med 20031981875–1886. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Fu S, Zhang N, Yopp A C.et al TGF‐beta induces Foxp3 + T‐regulatory cells from CD4 + C. Am J Transplant 200441614–1627. [DOI] [PubMed] [Google Scholar]
  • 62.Yamagiwa S, Gray J D, Hashimoto S.et al A role for TGF‐beta in the generation and expansion of CD4+CD25+ regulatory T cells from human peripheral blood. J Immunol 20011667282–7289. [DOI] [PubMed] [Google Scholar]
  • 63.Weiner H L. Oral tolerance: immune mechanisms and the generation of Th3‐type TGF‐beta‐secreting regulatory cells. Microbes Infect 20013947–954. [DOI] [PubMed] [Google Scholar]
  • 64.Chen Y, Kuchroo V K, Inobe J.et al Regulatory T cell clones induced by oral tolerance: suppression of autoimmune encephalomyelitis. Science 19942651237–1240. [DOI] [PubMed] [Google Scholar]
  • 65.Andersson P O, Olsson A, Wadenvik H. Reduced transforming growth factor‐beta1 production by mononuclear cells from patients with active chronic idiopathic thrombocytopenic purpura. Br J Haematol 2002116862–867. [DOI] [PubMed] [Google Scholar]
  • 66.Perez‐Machado M A, Ashwood P, Thomson M A.et al Reduced transforming growth factor‐beta1‐producing T cells in the duodenal mucosa of children with food allergy. Eur J Immunol 2003332307–2315. [DOI] [PubMed] [Google Scholar]
  • 67.Groux H, O'Garra A, Bigler M.et al A CD4+ T‐cell subset inhibits antigen‐specific T‐cell responses and prevents colitis. Nature 1997389737–742. [DOI] [PubMed] [Google Scholar]
  • 68.Wakkach A, Fournier N, Brun V.et al Characterization of dendritic cells that induce tolerance and T regulatory 1 cell differentiation in vivo. Immunity 200318605–617. [DOI] [PubMed] [Google Scholar]
  • 69.Barrat F J, Cua D J, Boonstra A.et al In vitro generation of interleukin 10‐producing regulatory CD4(+) T cells is induced by immunosuppressive drugs and inhibited by T helper type 1 (Th1)‐ and Th2‐inducing cytokines. J Exp Med 2002195603–616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Jonuleit H, Schmitt E, Schuler G.et al Induction of interleukin 10‐producing, nonproliferating CD4(+) T cells with regulatory properties by repetitive stimulation with allogeneic immature human dendritic cells. J Exp Med 20001921213–1222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Sakaguchi S, Takahashi T, Nishizuka Y. Study on cellular events in post‐thymectomy autoimmune oophoritis in mice. II. Requirement of Lyt‐1 cells in normal female mice for the prevention of oophoritis. J Exp Med 19821561577–1586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Morgan M E, Witteveen H J, Sutmuller R P.et al CD25+ regulatory cells from HLA‐DQ8 transgenic mice are capable of modulating collagen‐induced arthritis. Hum Immunol 2004651319–1327. [DOI] [PubMed] [Google Scholar]
  • 73.Veltkamp C, Ruhwald R, Giesem T.et al CD4+CD25+ cell depletion from the normal CD4+ T cell pool prevents tolerance toward the intestinal flora and leads to chronic colitis in immunodeficient mice. Inflamm Bowel Dis 200612437–446. [DOI] [PubMed] [Google Scholar]
  • 74.Morrissey P J, Charrier K, Braddy S.et al CD4+ T cells that express high levels of CD45RB induce wasting disease when transferred into congenic severe combined immunodeficient mice. Disease development is prevented by cotransfer of purified CD4+ T cells. J Exp Med 1993178237–244. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Powrie F, Leach M W, Mauze S.et al Phenotypically distinct subsets of CD4+ T cells induce or protect from chronic intestinal inflammation in C. B‐17 scid mice. Int Immunol 199351461–1471. [DOI] [PubMed] [Google Scholar]
  • 76.Mottet C, Uhlig H H, Powrie F. Cutting edge: cure of colitis by CD4+CD25+ regulatory T cells. J Immunol 20031703939–3943. [DOI] [PubMed] [Google Scholar]
  • 77.Battaglia M, Gregori S, Bacchetta R.et al Tr1 cells: from discovery to their clinical application. Semin Immunol 200618120–127. [DOI] [PubMed] [Google Scholar]
  • 78.Groux H, O'Garra A, Bigler M.et al A CD4+ T‐cell subset inhibits antigen‐specific T‐cell responses and prevents colitis. Nature 1997389737–742. [DOI] [PubMed] [Google Scholar]
  • 79.Fahlen L, Read S, Gorelik L.et al T cells that cannot respond to TGF‐beta escape control by CD4(+)CD25(+) regulatory T cells. J Exp Med 2005201737–746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Kuhn R, Lohler J, Rennick D.et al Interleukin‐10‐deficient mice develop chronic enterocolitis. Cell 199375263–274. [DOI] [PubMed] [Google Scholar]
  • 81.Shull M M, Ormsby I, Kier A B.et al Targeted disruption of the mouse transforming growth factor‐beta 1 gene results in multifocal inflammatory disease. Nature 1992359693–699. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Steidler L, Hans W, Schotte L.et al Treatment of murine colitis by Lactococcus lactis secreting interleukin–10. Science 20002891352–1355. [DOI] [PubMed] [Google Scholar]
  • 83.Gorelik L, Flavell R A. Abrogation of TGFbeta signaling in T cells leads to spontaneous T cell differentiation and autoimmune disease. Immunity 200012171–181. [DOI] [PubMed] [Google Scholar]
  • 84.Boirivant M, Fuss I J, Chu A.et al Oxazolone colitis: A murine model of T helper cell type 2 colitis treatable with antibodies to interleukin 4. J Exp Med 19981881929–1939. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Fuss I J, Boirivant M, Lacy B.et al The interrelated roles of TGF‐beta and IL‐10 in the regulation of experimental colitis. J Immunol 2002168900–908. [DOI] [PubMed] [Google Scholar]
  • 86.Neurath M F, Fuss I, Kelsall B L.et al Experimental granulomatous colitis in mice is abrogated by induction of TGF‐beta‐mediated oral tolerance. J Exp Med 19961832605–2616. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Baecher‐Allan C, Hafler D A. Human regulatory T cells and their role in autoimmune disease. Immunol Rev 2006212203–216. [DOI] [PubMed] [Google Scholar]
  • 88.Makita S, Kanai T, Oshima S.et al CD4+CD25bright T cells in human intestinal lamina propria as regulatory cells. J Immunol 20041733119–3130. [DOI] [PubMed] [Google Scholar]
  • 89.Kelsen J, Agnholt J, Falborg L.et al Indium‐labelled human gut‐derived T cells from healthy subjects with strong in vitro adhesion to MAdCAM‐1 show no detectable homing to the gut in vivo. Clin Exp Immunol 200413866–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Maul J, Loddenkemper C, Mundt P.et al Peripheral and intestinal regulatory CD4+ CD25(high) T cells in inflammatory bowel disease. Gastroenterology 20051281868–1878. [DOI] [PubMed] [Google Scholar]
  • 91.Kuhn R, Lohler J, Rennick D.et al Interleukin‐10‐deficient mice develop chronic enterocolitis. Cell 199375263–274. [DOI] [PubMed] [Google Scholar]
  • 92.Fahlen L, Read S, Gorelik L.et al T cells that cannot respond to TGF‐beta escape control by CD4(+)CD25(+) regulatory T cells. J Exp Med 2005201737–746. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Monteleone G, Kumberova A, Croft N M.et al Blocking Smad7 restores TGF‐beta1 signaling in chronic inflammatory bowel disease. J Clin Invest 2001108601–609. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Tang Q, Henriksen K J, Bi M.et al In vitro‐expanded antigen‐specific regulatory T cells suppress autoimmune diabetes. J Exp Med 20041991455–1465. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Groux H, O'Garra A, Bigler M.et al A CD4+ T‐cell subset inhibits antigen‐specific T‐cell responses and prevents colitis. Nature 1997389737–742. [DOI] [PubMed] [Google Scholar]
  • 96.Thornton A M, Shevach E M. Suppressor effector function of CD4+CD25+ immunoregulatory T cells is antigen nonspecific. J Immunol 2000164183–190. [DOI] [PubMed] [Google Scholar]
  • 97.Stassen M, Schmitt E, Jonuleit H. Human CD(4+)CD(25+) regulatory T cells and infectious tolerance. Transplantation 200477(Suppl)S23–S25. [DOI] [PubMed] [Google Scholar]
  • 98.van Montfrans C, Rodriguez Pena M S, Pronk I.et al Prevention of colitis by interleukin 10‐transduced T lymphocytes in the SCID mice transfer model. Gastroenterology 20021231865–1876. [DOI] [PubMed] [Google Scholar]
  • 99.van Montfrans C, Hooijberg E, Rodriguez Pena M S.et al Generation of regulatory gut‐homing human T lymphocytes using ex vivo interleukin 10 gene transfer. Gastroenterology 20021231877–1888. [DOI] [PubMed] [Google Scholar]
  • 100.Allan S E, Passerini L, Bacchetta R.et al The role of 2 FOXP3 isoforms in the generation of human CD4+ Tregs. J Clin Invest 20051153276–3284. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Yagi H, Nomura T, Nakamura K.et al Crucial role of FOXP3 in the development and function of human CD25+CD4+ regulatory T cells. Int Immunol 2004161643–1656. [DOI] [PubMed] [Google Scholar]
  • 102.Steinman R M, Hawiger D, Nussenzweig M C. Tolerogenic dendritic cells. Annu Rev Immunol 200321685–711. [DOI] [PubMed] [Google Scholar]
  • 103.Rutella S, Lemoli R M. Regulatory T cells and tolerogenic dendritic cells: from basic biology to clinical applications. Immunol Lett 20049411–26. [DOI] [PubMed] [Google Scholar]
  • 104.Smits H H, de Jong E C, Wierenga E A.et al Different faces of regulatory DCs in homeostasis and immunity. Trends Immunol 200526123–129. [DOI] [PubMed] [Google Scholar]
  • 105.Braat H, McGuirk P, Kate F T.et al Prevention of experimental colitis by parenteral administration of a pathogen‐derived immunomodulatory molecule. Gut. 2006 (4 Sep, Epub ahead of print). [DOI] [PMC free article] [PubMed]
  • 106.Hoffmann P, Eder R, Boeld T J.et al Only the CD45RA+ subpopulation of CD4+CD25high T cells gives rise to homogeneous regulatory T cell lines upon in vitro expansion. Blood 20061084260–4267. [DOI] [PubMed] [Google Scholar]
  • 107.McGuirk P, McCann C, Mills K H. Pathogen‐specific T regulatory 1 cells induced in the respiratory tract by a bacterial molecule that stimulates interleukin 10 production by dendritic cells: a novel strategy for evasion of protective T helper type 1 responses by Bordetella pertussis. J Exp Med 2002195221–231. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Pasare C, Medzhitov R. Toll pathway‐dependent blockade of CD4+CD25+ T cell‐mediated suppression by dendritic cells. Science 20032991033–1036. [DOI] [PubMed] [Google Scholar]
  • 109.Goleva E, Cardona I D, Ou L S.et al Factors that regulate naturally occurring T regulatory cell‐mediated suppression. J Allergy Clin Immunol 20051161094–1100. [DOI] [PubMed] [Google Scholar]
  • 110.Karagiannidis C, Akdis M, Holopainen P.et al Glucocorticoids upregulate FOXP3 expression and regulatory T cells in asthma. J Allergy Clin Immunol 20041141425–1433. [DOI] [PubMed] [Google Scholar]
  • 111.Fattorossi A, Battaglia A, Buzzonetti A.et al Circulating and thymic CD4 CD25 T regulatory cells in myasthenia gravis: effect of immunosuppressive treatment. Immunology 2005116134–141. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Valencia X, Stephens G, Goldbach‐Mansky R.et al TNF downmodulates the function of human CD4+CD25hi T‐regulatory cells. Blood 2006108253–261. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 113.Bisikirska B C, Herold K C. Use of anti‐CD3 monoclonal antibody to induce immune regulation in type 1 diabetes. Ann N Y Acad Sci 200410371–9. [DOI] [PubMed] [Google Scholar]
  • 114.Tilg H, van Montfrans C, van den E A.et al Treatment of Crohn's disease with recombinant human interleukin 10 induces the proinflammatory cytokine interferon gamma. Gut 200250191–195. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 115.Braat H, Rottiers P, Hommes D W.et al A phase I trial with transgenic bacteria expressing interleukin‐10 in Crohn's disease. Clin Gastroenterol Hepatol 20064754–759. [DOI] [PubMed] [Google Scholar]
  • 116.Kitani A, Fuss I J, Nakamura K.et al Treatment of experimental (trinitrobenzene sulfonic acid) colitis by intranasal administration of transforming growth factor (TGF)‐beta1 plasmid: TGF‐beta1‐mediated suppression of T helper cell type 1 response occurs by interleukin (IL)‐10 induction and IL‐12 receptor beta2 chain downregulation. J Exp Med 200019241–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Van Montfrans C, Rodriguez Pena M S, Pronk I.et al Prevention of colitis by interleukin 10‐transduced T lymphocytes in the SCID mice transfer model. Gastroenterology 20021231865–1876. [DOI] [PubMed] [Google Scholar]

Articles from Gut are provided here courtesy of BMJ Publishing Group

RESOURCES