Differently from Tr1, Foxp3+ Treg cells require antigen stimulation to expand in vivo but are able to perform their inhibitory tasks also in the absence of TCR stimulation
[67,68,69][37][38][39]. Once activated, Foxp3+ Treg cells are able to suppress Teff cells using all the four mechanisms cited above
[64][34].
In CeD, intestinal Foxp3 expression has been evaluated by RT-PCR in several works, and all of them reported an increased Foxp3 expression in patients with untreated CeD, compared to controls
[18,52,53][40][41][42]. These results were confirmed also by immunohistochemistry
[15,18,21,23,52,53,70][10][40][41][42][43][44][45] and by flow cytometry
[15,18,23,53][10][40][42][44].
These data indicate that the immune system is attempting to control the persistent inflammation either by recruitment of Treg cells from blood to tissue, or through the expansion of such regulatory T cells in the mucosa.
Regardless of Treg cells frequency, some studies have reported that such cells may be impaired in their capacity to downregulate local Teff cell functions or, conversely, that Teff cells may fail to respond to Tregs.
An interesting paper by Serena et al.
[72][46], analyzed the expression of specific Foxp3 isoforms, comparing the full length (FL) to the alternatively spliced isoform D2, since the last isoform cannot properly downregulate the Th17-driven immune response. Intestinal biopsies from patients with active CeD showed increased expression of Foxp3 D2 isoform over FL, while both isoforms were expressed similarly in control subjects, thus suggesting a possible defect in the Foxp3+ Treg function in atrophic celiac mucosa.
In line with the data that IL-15 may interfere with immune regulation,
wescholars have demonstrated that in active CeD patients, IL-15 was able of making Tresp cells resistant to the regulatory effects of CD4+CD25+ Treg cells
[23][44]. In particular, in this study CD4+CD25+ T cells were directly isolated from intestinal biopsies of patients, and their suppressive ability was evaluated on autologous CD4+CD25− Tresp cells. The data showed that intestinal Treg, as well as peripheral blood Treg from celiac patients, are not functionally deficient. Nevertheless, in active CeD patients, IL-15 impaired the functions of Treg cells making Tresp cells refractory to the regulatory effects of Treg cells, in terms of proliferation and production of IFN-γ (
Figure 1b).
3.3. CD8 T Lymphocytes with Regulatory Activity
In addition to CD4 Treg cells, mainly resident in intestinal lamina propria, populations of immune cells can be found within the intestinal epithelial cell layer, such as IELs, which consist mostly of CD8+T cells. Though most of those IELs express T cell receptor (TCR)-alpha beta chains (αβ), CeD is characterized by an increase in TCRγδ+ IELs that remain elevated even after removal of gluten from the diet and whose functional significance in CeD is still under investigation
[74][47]. It has been hypothesized that CD8+TCRγδ + may play a role in the preservation of intestinal homeostasis by regulating the mucosal immune response and/or by contributing to the epithelial cell layer maintenance. Functional properties of TCRγδ+ IELs can be mediated by NK receptors (such as NKGD) expressed on a fraction of these cells
[75][48]. At the same time, CD8+ TCRαβ+ IELs have been suggested to kill intestinal epithelial cells (IECs) in an NKG2D-MICA-dependent manner during CeD
[76][49].
In addition to TCRγδ+ CD8+ cells, a regulatory activity has been recently proposed also for a subpopulation of TCRαβ+ CD8+ cells. A subset of regulatory CD8+ T cells that express Ly49 has been described in mice
[79[50][51],
80], able to reduce autoimmunity in a model of experimental autoimmune encephalomyelitis
[81][52] but also with documented functions in several other disease settings
[79][50]. These cells are Foxp3- but TGF-β is necessary to maintain their regulatory identity. Mechanisms of action involve IL-10 secretion, killing via granzyme/perforin, and induction of apoptosis by FAS/FASL interaction. Ly49 receptors are a family of NK receptors, including some inhibitory ones. Their functional analogues in humans are killer-cell immunoglobulin-like receptor (KIR) genes.
4. Regulatory T Cells in the Context of CeD Pathogenesis
The immune-pathogenesis of CeD is a complex mosaic, where different factors are needed to interplay for promoting the intestinal damage. Briefly, gluten is not completely digested by gastro-intestinal enzymes, and peptides that manage to cross the epithelial barrier are subjected to deamidation by tTG2, into the lamina propria. Here, peptides are taken up by DCs and presented to CD4+ T cells, in the context of HLA-DQ2 or DQ8 molecules, thus promoting the differentiation of naïve gluten-specific CD4+ T cells into Th1 effector T cells, in induction sites.
In this context, when gluten is ingested, together with expansion of pro-inflammatory lymphocytes, regulatory T cells producing large amount of IL-10 are also increased
[7,12,13,14,15][9][10][23][53][54]. As discussed in the previous sections, at least four different T cells with suppressive activity are present in the intestinal mucosa and/or in the peripheral blood of CeD patients, which could be activated directly by gluten, or indirectly by microenvironmental signals. Such regulatory subsets may expand locally, or may be recruited from the periphery to the inflamed tissue. In spite of their origin and activation, suppressive cells may act directly or indirectly on the main orchestrators of the mucosal inflammation, including CD4+ T cells, cytotoxic IELs and DCs (
Figure 1).
5. Therapeutic Applications of Treg Cells in CeD
The pathogenesis of CeD is triggered by the loss of tolerance towards gluten peptides.
On the other hand, as discussed above, several studies have suggested that the suppressive effect of Treg cells might be impaired in vivo in CeD by the inflammatory microenvironment, and their dysregulated function may contribute to sustain and expand the local inflammatory response.
Among these immunomodulating therapies, there are:
- -
-
Approaches based on in vivo administration of drugs such as rapamycin, or biologicals, such as IL-10 or low-dose IL-2, to suppress Teff cells and promote Tregs.
-
- -
-
Approaches resting on the administration of the autoantigen by using lentiviral vectors or antigen-specific nanoparticles, promoting tolerogenic cells and the expansion of Treg cells.
-
Moreover, cell-based therapies have been developed to enhance Treg cell specificity, function and number. This can be achieved by expanding Tr1 cells expressing IL-10
[88][55], Tregs expressing a natural repertoire of polyclonal TCRs
[89][56] or Tregs that have been ex vivo-engineered to express a specific autoantigen receptor, such as a TCR, or a chimeric antigen receptor (CAR)
[90,91][57][58].
DCs also offer a cell-based therapeutic way to restore tolerance and prevent autoimmunity
[92][59].
Tregs can generate a tolerogenic phenotype in DCs, which can contribute to the rescue of immune tolerance. Tolerogenic DCs (TolDC), generated ex vivo, could be administrated, in vivo, to suppress autoimmunity in diseases such as T1D
[93,94][60][61].
Other approaches have used mature DCs to expand antigen-specific Tregs
[95][62], as for the generation of tolDC by genetic engineering of monocytes (CD14+) with lentiviral vectors co-encoding for immunodominant antigen-derived peptides and IL-10
[96][63].
Therefore, therapeutic approaches aiming to restore tolerance to gluten, and/or to correct Treg functioning, would be a great step forward to protect CeD patients from excessive immune response, to reinstate intestinal homeostasis and, possibly, to allow improvement in patients outcomes.
Other approaches have been developed using in vivo antigen-delivery to induce a tolerogenic inhibition of specific immune response
[97,98][64][65].
This tolerance strategy leads to anergy within Ag-specific Teff cells and activate populations of Ag-specific regulatory T cells
[97,98,99,100,101,102,103][64][65][66][67][68][69][70].
In this context, gliadin encapsulated in nanoparticles, namely TAK-101 (formerly TIMP-GLIA, Tolerogenic Immune Modifying nanoparticles), is under clinical development. Firstly, this approach has been shown to be effective in a mouse model of CeD
[104[71][72],
105], where intravenous infusion of gliadin-encapsulating nanoparticles inhibited the proliferation, and the IFN-γ and IL-17 secretion, of gliadin-specific T cells, while increasing the frequency of FoxP3+ Treg cells.
A possible cellular therapy representing a great step forward in the treatment and, possibly, the cure of chronic inflammatory conditions, is based on the use of stem cells. Both hematopoietic stem cells (HSCs) and mesenchymal stem/stromal cells (MSCs) have been employed in the treatment of refractory cases with promising results. By virtue of the lack of immunogenicity and of the ability to favor tissue regeneration and expansion of T cells with regulatory function, MSCs seem the best candidate for clinical application. MSC are multipotent non-hematopoietic cells present in different tissues, including bone marrow, amniotic fluid, umbilical cord, and placenta, which due to their immunomodulatory characteristics are considered as new therapeutic agents in the cell-based therapy of autoimmune and immune-mediated diseases
[107,108,109,110,111,112,113,114][73][74][75][76][77][78][79][80].
6. Conclusions
Celiac intestinal mucosa harbors two subsets of CD4+ Treg cells, Tr1 and Foxp3+ T cells. Many factors, such as IL-15, largely expressed in the CeD mucosa, may interfere with the function of Treg cells, thus contributing to the loss of intestinal homeostasis and promoting chronic inflammation.
On the other hand, novel T cell subsets with regulatory activity are emerging, and their further characterization is of great interest. As Treg cells exist naturally in the human gut mucosa and maintain intestinal homeostasis, using methods to enhance their numbers and/or function is a possibility worthy of pursuit as a new therapeutical approach to re-establish tolerance to gluten in patients with CeD. Treg immunotherapies based on infusion of autologous TolDC or MSCs, or on enhancement of Treg numbers and function via administration of nanoparticles, remain possible strategies to be implemented in CeD.