Regulatory T cell heterogeneity and therapy in autoimmune diseases

Regulatory T cell heterogeneity and therapy in autoimmune diseases

Journal Pre-proof Regulatory T cell heterogeneity and therapy in autoimmune diseases Rui Zhang, Jinlin Miao, Ping Zhu PII: S1568-9972(20)30294-9 DO...

910KB Sizes 0 Downloads 33 Views

Journal Pre-proof Regulatory T cell heterogeneity and therapy in autoimmune diseases

Rui Zhang, Jinlin Miao, Ping Zhu PII:

S1568-9972(20)30294-9

DOI:

https://doi.org/10.1016/j.autrev.2020.102715

Reference:

AUTREV 102715

To appear in:

Autoimmunity Reviews

Received date:

20 June 2020

Accepted date:

28 June 2020

Please cite this article as: R. Zhang, J. Miao and P. Zhu, Regulatory T cell heterogeneity and therapy in autoimmune diseases, Autoimmunity Reviews (2020), https://doi.org/ 10.1016/j.autrev.2020.102715

This is a PDF file of an article that has undergone enhancements after acceptance, such as the addition of a cover page and metadata, and formatting for readability, but it is not yet the definitive version of record. This version will undergo additional copyediting, typesetting and review before it is published in its final form, but we are providing this version to give early visibility of the article. Please note that, during the production process, errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

© 2020 Published by Elsevier.

Journal Pre-proof

Regulatory T Cell Heterogeneity and Therapy in Autoimmune Diseases Rui Zhanga,1, Jinlin Miaoa,b,1, Ping Zhua,* a

Department of Clinical Immunology, PLA Specialized Research Institute of Rheumatoid &

Immunology, Xijing Hospital, Fourth Military Medical University, Xi’an, China b

National Translational Science Center for Molecular Medicine, Fourth Military Medical

University, Xi’an, China *

of

Corresponding author: Ping Zhu

ro

Department of Clinical Immunology, PLA Specialized Research Institute of Rheumatoid & Immunology, Xijing Hospital, Fourth Military Medical University, No. 127 West Changle Road,

-p

Xi'an 710032, Shaanxi Province, PR China.

re

Telephone: +86-29-84773951; Fax: +86-29-83240610

Rui Zhang and Jinlin Miao contributed equally to this work.

Highlights

Jo ur

na

1

lP

E-mail: [email protected]



Regulatory T cells are heterogeneous and can be divided into different subsets.



Non-lymphoid tissue resident Treg cells exert special tissue-related functions which go far beyond their immunosuppressive capability.



Treg-targeted therapy, including Treg-based cellular transfer, IL-2 therapy and Treg targeted drugs, will become a promising strategy for autoimmune diseases treatment.

Abstract

1

Journal Pre-proof Regulatory T (Treg) cells are a group of CD4+ T cell with high expression of CD25 and cell linage specific transcription factor forkhead box P3 (Foxp3) and play a vital role in maintaining immune homeostasis. In the last two decades, researchers have shown that Treg cells involved in the occurrence, development and prognosis of many diseases, especially in autoimmune diseases. Treg targeted therapies, such as low-dose interleukin-2 (IL-2) treatment and Treg infusion therapy,

of

which are aimed at restoring the number or function of Treg cells, have become a hot topic in

ro

clinical trials of these diseases. It is believed that Treg cells are heterogeneous. Different subsets of Treg cells have various functions and play different parts in immunomodulatory. Gaining

-p

insights into Treg heterogeneity will help us further understand the function of Treg cells and

re

provide news ideas for the selective therapeutic manipulation of Treg cells. In this review, we

Jo ur

Keywords

na

autoimmune diseases.

lP

mainly summarize the heterogeneity of Treg cells and their potential therapeutic value in

T-Lymphocytes, Regulatory; Subset; Immunotherapy

1. Introduction In 1995, Sakaguchi and his colleagues found that CD4+CD25+ cells contribute to maintaining self-tolerance by down-regulating immune response to self and non-self antigens in mice, which revealed potential involvement of these cells in autoimmune diseases [1]. Then in 2003, the close relationship between the transcription factor Foxp3 and development of CD4+CD25+ Treg cells

2

Journal Pre-proof

was found [2-4]. These pillar researches gave us a general knowledge of Treg cells and initiated new lines of research into pathogenesis and possible treatments of immune-related diseases. Over the past two decades, researchers revealed the important role of Treg cells in the pathogenesis of various diseases. In many autoimmune diseases, including rheumatoid arthritis (RA) [5-10], systemic lupus erythematosus (SLE) [11-13], ankylosing spondylitis (AS) [14],

of

systemic sclerosis [15,16] and so on, the qualitative or quantitative defects of Treg cells have been

ro

widely reported. Several risk factors, including genetics [17] and environmental factors [18], could also involve in the occurrence and development of these diseases by regulating the number

-p

and function of Treg cells. These findings indicate that targeting Treg cells is a promising new

re

therapeutic approach in autoimmune diseases. And several clinical trials of Treg cells infusion

lP

have already obtained affirmative effects on alleviating disease activity [19,20].

na

Since Treg cells are heterogeneous and show functional and phenotypic differentiation within different subsets, in this review, we mainly discuss different subsets of CD4+CD25+Foxp3+ Treg

Jo ur

cells, including tissue Treg. We also summarize Treg-targeted therapies in autoimmune diseases which may provide us new insights into understanding and treatment of these diseases. 2. Function of Treg cells

CD4+CD25+Foxp3+ Treg cells are a component of the immune system with an indispensable role in maintaining immune tolerance and homeostasis. Recent researches reveal that they can also control non-immunological processes. Treg cells resident in tissue have unique tissue-related functions which are critical to safeguarding organismal homeostasis [21,22]. Treg cells exert immunosuppressive function through multiple mechanisms, including secreting inhibitory cytokines such as IL-10, IL-35 and TGF-β [23-25], metabolic regulation via

3

Journal Pre-proof

IL-2 or CD 73 and CD39 [26-29], cytolysis [30] and modulation of antigen presenting cells (APCs) [31,32]. Aside from these classical mechanisms of how Treg cells exert their inhibition function, recent advances uncover other mechanisms of Treg function. Schmidleithner et al. report that Treg cells

highly express

enzyme

hydroxyprostaglandin

dehydrogenase,

which

catabolizes

prostaglandin E2 (PGE2) into the metabolite 15-keto PGE2. And in visceral adipose tissue,

of

Treg-cell-mediated generation of 15-keto PGE2 increase suppressive activity of Treg and impact

ro

conventional T cell proliferation [33]. Akkaya B et al. indicate a novel pathway for Treg-mediated suppression by depleting the complex of cognate peptide and major histocompatibility complex

-p

class II from the dendritic cell (DC) surface in an antigen-specific manner which reduces the

re

capacity of DCs to present antigen and results in the failure to activate naive T cells [34]. These

lP

novel mechanisms give clearer understanding of Treg function and molecules involved may

na

become new targets in Treg-associated treatment. 3. Treg subsets

Jo ur

There are many controversies related to Treg cells in autoimmune diseases, especially the issue of decreased numbers versus dysfunction of Treg cells in the pathogenesis of autoimmune diseases. These controversies may partly be explained by Treg heterogeneity in their phenotype and function. Unfortunately, there is still no systematic and comprehensive classification method for Treg subsets up to now. For all this, making the acquaintance of the existing classification methods will pave the way for a better understanding of Treg heterogeneity as well as the pathogenesis of autoimmune diseases. At present, the following subpopulations of Treg cells are widely investigated by researchers. 3.1 tTreg and pTreg

4

Journal Pre-proof

According to the origin, Treg cells can be divided into two groups: thymus-derived Treg (tTreg) cells or natural Treg (nTreg) cells developing from immature thymocytes and peripheral Treg (pTreg) cells or induced Treg (iTreg) cells developing in periphery from mature CD4+ T cells in certain conditions. It was once believed that transcription factor Helios [35] and cellular surface glycoprotein Neuropilin-1 (Nrp-1) [36,37] which were highly expressed by tTreg cells can be used

of

to distinguish tTreg cells from pTreg cells. However later studies revealed that pTreg cells can

ro

upregulate these two factors under certain conditions [38,39]. So far there is no reliable maker to distinguish tTreg cells from pTreg cells.

-p

3.2 aTreg, rTreg and non Treg

re

Another classification of Treg cells subsets in human is based on the expression of CD45RA and ﹣

lP

Foxp3 [40]: CD45RA+Foxp3lo resting Treg (rTreg) cells, CD45RA Foxp3hi activated Treg (aTreg) ﹣

na

cells and cytokine-secreting CD45RA Foxp3lo non-suppressive T (non-Treg) cells, the majority of which are not Treg cells (Table 1). Further, rTreg cells could acquire an aTreg phenotype and exert

Jo ur

suppression after proliferation upon TCR simulation. These three different subpopulations of Treg cells have been studied in many autoimmune diseases. In RA and Behcet’s disease, researchers reveal that the proportion of total Treg cells and aTreg cells, but not rTreg cells is significantly decreased compared to healthy controls (HCs) [41]. In multiple sclerosis, aTreg cells show a significant reduction and express higher levels of PD-1 compared to HCs which denotes an exhausted and dysfunctional status of aTreg cells [42]. Several studies demonstrate a significant increase in rTreg and non-Treg subsets in patients with active SLE [43,44]. And rTreg cells sorted from peripheral blood mononuclear cells of SLE patients fail to inhibit the responder T cell proliferation. These results indicate that different Treg subsets may

5

Journal Pre-proof

serve diverse functions in autoimmune diseases and further study in those Treg subsets is likely to lead us to an in-depth understanding of the pathogenesis of these diseases. Table 1 Key properties of rTreg cells, aTreg cells and non-Treg cells rTreg cells

aTreg cells ﹣

non-Treg cells ﹣

CD25++CD45RA+Foxp3lo ﹣ CTLA-4loKi-67

CD25+++CD45RA Foxp3hi CTLA-4hiKi-67+

CD25++CD45RA Foxp3lo CTLA-4int

cytokine production

Poorly produces IL-2 or IFN-γ

Scarcely produces IL-2 or IFN-γ

Produces high amounts of IL-2, IFN-γ and IL-17

methylation status of the FOXP3 gene

CpG methylation sites in 5’ flanking region and the STAT5-responsive region were completely demethylated

The 5’ flanking region was also highly demethylated, the STAT5-responsive region was less demethylated

ro

-p re

Potently suppressed the proliferation of responder cells

Didn’t have suppression function

na

3.3 Th-like Treg

lP

In vitro suppressive activity

of

cell surface molecules

In recent years, Treg cells that phenotypically mirror effector T helper (Th) cells and are known as

Jo ur

Th-like Treg cells have been widely studied in rodent and human. These Treg cells express specific Th-associated transcription factors and play a regulatory or proinflammatory role during different types of immune responses (Table 2). Th1-like Treg cells upregulate Th1 specific transcription factor T-box transcription factor 21 (T-bet) and are thought as a unique Treg subset specific in inhibiting type 1 inflammation [45-47]. Th1-like Treg cells also express C-X-C motif chemokine receptor 3 (CXCR3) in a T-bet-dependent manner which enables them to migrate to sites of type 1 inflammation and inhibit Th1 cell responses. Th17- and Th2-like Treg cells acquire expression of the transcription factor RAR-related orphan receptor γt (RORγt) [48] and GATA-3 [49] and restrain Th17 cells responses and Th2 cells responses, respectively. However, in some

6

Journal Pre-proof

autoimmune diseases and tumors, Th-like Treg cells secrete pro-inflammatory cytokines and display diminished suppressive function [50-56]. Therefore, Th-like Treg cells may exert suppressive capacity or proinflammatory effect in a context-dependent manner and contribute to different prognosis outcomes of varying diseases. And studies in different subsets of Th-like Treg cells will give us further understanding of pathogenic mechanism of various immune-related

of

disorders and potential strategies in the treatments of these diseases.

ro

Table 2 Characters of Th-like Treg cells

Th2-like Treg cells

Th17-like Treg cells

transcription factors

T-bet

GATA-3 and IRF-4

RORγt

cytokine production

IFN-γ

stimulating factors

IL-12, IFN-γ, IL-27

chemokine receptor

CXCR3, CCR5

4. Tissue Treg

re lP

na

Jo ur

tissue specificity

-p

Th1-like Treg cells

-

IL-4, IL-13

IL-17

IL-4, IL-5

IL-1β, IL-6, IL-12

CCR4

CCR4, CCR6

skin

gastrointestinal tract

Aside from different populations of Treg cells in peripheral blood and lymphoid organ, there are also various subsets of Treg cells in non-lymphoid tissue. Non-lymphoid tissue resident Treg cells show unique phenotype, tissue specific transcription factors and distinct T cell receptors (TCR) repertoire. And their special tissue-related functions go far beyond their immunosuppressive capability [21,22,57]. 4.1 Treg cells in adipose tissue

7

Journal Pre-proof

The most widely demonstrated tissue-resident Treg cells are Treg cells in adipose tissue, especially in visceral adipose tissue (VAT) [58]. VAT Treg cells account for more than half of CD4+ cells [59] in adipose. These cells appear to be mostly of thymic origin for they highly express Helios and Nrp1 [60] and have a distinct TCR repertoire [59]. VAT Treg cells express majority of signatures as canonical Treg cells do but display a distinct chemokine/chemokine receptor pattern, produce abundant IL-10 and upregulate genes downstream of IL-10R [59]. They also express peroxisome

of

proliferator-activated receptor γ (PPARγ), an important transcription factor for adipocyte

ro

differentiation [61]. And loss-of-function study reveals that PPARγ is an important factor

-p

controlling Treg cells accumulation and phenotype in adipose tissue [61]. VAT Treg cells play a

re

vital role in the maintenance of immune and metabolic homeostasis of adipose tissue. They not

lP

only act as important negative regulators of VAT inflammation and insulin resistance [62,63], but

na

also involve in adipose tissue function including efficient thermogenesis and lipolysis [64,65]. 4.2 Treg cells in skeletal muscle

Jo ur

In skeletal muscle, Treg cells also show a transcriptome distinct from that of lymphoid-organ Treg cells. Genes encoding IL-10, amphiregulin (Areg) and T-cell immunoglobulin domain and mucin domain 3 (TIM-3) are highly expressed [66]. And just like VAT Treg cells, skeletal muscle Treg cells highly express Helios and Nrp1 and have a unique repertoire of TCRs which indicated a thymic origin [67]. Treg cells in skeletal muscle exert important function in tissue regeneration. They start to accumulate in impaired muscles within a few days after injury [67] and IL-33 receptor ST2 [68] and TCR specificity [69] play a critical role in Treg accumulation in injured muscles. Muscle Treg cells exert their tissue repair function by regulating co-infiltrating conventional T cells, containing the number of myeloid cells and promoting their transition from a

8

Journal Pre-proof

pro- to an anti-inflammatory phenotype and enhancing satellite cells differentiation to promote muscle regeneration [67]. In mouse model of myopathy such as myositis and muscular dystrophy, adoptive transfer of Treg cells or low dose IL-2/anti–IL-2 complexes treatment significantly alleviated muscle inflammation and reduced muscular injury [70,71]. These results indicate a therapeutic potential

of

of targeting Treg cells in myopathy. However, in Toxoplasma gondii infection model, skeletal

ro

muscle Treg cells exert a deleterious effect by promoting proinflammatory macrophages accumulation in muscle, leading to prolonged tissue damage [72]. Therefore, it is essential to

-p

conduct more studies on the consequences of inflammatory environment on Treg physiology.

re

4.3 Treg cells in tumor

lP

In 1999, two Japanese research teams found CD4+CD25+ Treg cells could inhibit tumor rejection

na

and promote tumor growth, revealing that Treg cells were also involved in tumor immune regulation [73,74]. Treg cells may play different roles in different stage of tumorigenesis. A latest

Jo ur

study of pancreatic cancer shows that depletion of Treg cells result in deteriorative pancreatitis and pancreatic intraepithelial neoplasia, and lead to accelerated tumor progression. This study reveals a protective role of Treg cells in precancerous lesion [75]. Nevertheless, tumor-infiltrating Treg cells hinder tumor immune surveillance and inhibit effective host-anti-tumor immune response. In breast cancer [76], melanoma [77] and lung cancer [78], increased frequencies of circulating or tumor-infiltrating Treg cells usually indicate poor prognosis. While in some cancers, especially colon cancer, large infiltrations of Foxp3+ cells suggest a better prognosis [79,80]. However, further study shows that this group of Foxp3+ cells, which indicate a good prognosis, are Foxp3lowCD45RA+ non-Treg cells and increased frequencies of Foxp3hiCD45RA- Treg cells also

9

Journal Pre-proof

lead to a poor prognosis [81]. Tumor infiltrated Treg cells also have different metabolic characteristics from their counterpart in periphery. Intratumoral Treg cells increase their lipid metabolism and have a higher lipid content. Compared with their circulating counterpart, these Treg cells express higher amounts of CD36 which is known to mediate long-chain fatty acid uptake and lipid metabolism. Genetic

of

ablation of Cd36 in Treg cells selectively decrease lipid uptake and content in intratumoral Treg

ro

cells. And loss of CD36 expression perturbs accumulation of Treg cells in tumor [82]. Thus, targeting CD36 and thereby manipulating tumor infiltrating Treg cells may be a promising

-p

anti-tumor therapeutic strategy.

re

4.4 Treg cells in other organs

lP

Treg cells in other nonlymphoid tissue also show their distinct tissue specific functions. Treg cells

na

resident in skin involve in restraining skin inflammation [83,84], regulating fibroblast activation [85], repairing cutaneous injuries [86] and establishing tolerance to commensal microbes [87].

Jo ur

Scientists also uncover that cutaneous Treg cells modulate hair follicle stem cells and promote hair follicle regeneration through Jag1-Notch pathway [88]. Similar to skin Treg cells, Treg cells in intestine play an important role in tissue repair [89,90] and microbiota tolerance [91]. In addition, intestinal Treg cells involve in establishing tolerance towards food antigens [92]. In myocardia, Treg recruitment can inhibit post-myocardial infarction inflammation, excessive matrix degradation and poor remodeling [93]. In bones, Treg cells protect against TNF-α induced bone destruction [94] as well as bone loss caused by ovariectomy [95]. In central nervous system, Treg cells have been proven to promote myelin regeneration [96]. These discoveries about unique functions of Treg cells in different organs demonstrate their ability to adopt to environment and

10

Journal Pre-proof

lay the foundation for optimal apply of Treg cells in the treatment of related diseases. 5. Treg-targeted therapy Alone with the deep studies in the role of Treg cells in autoimmune diseases, Treg-targeted therapy becomes a promising avenue for the treatment of autoimmunity. Among various strategies, Treg-based cellular transfer and low-dose IL-2 modulation have become subjects of intense

of

research currently (Figure). And these strategies have been applied in many clinical trials of a

ro

variety of immune-related disorders [97-104]. 5.1 Treg-based cellular transfer

-p

Treg cells used for adoptive cell therapy usually come from autologous peripheral blood [97-102]

re

or umbilical cord blood [103,104]. To obtain clinically meaningful Treg numbers, Treg cells

lP

derive from autologous peripheral blood or cord blood need to be expanded ex vivo or induced in

na

vitro before transfusion. And several protocols have been developed. Some researches show that cell-based artificial antigen-presenting cells (aAPCs) could massively increase nTreg yield

Jo ur

[105,106]. In the meantime, these aAPCs could maintain Foxp3 expression and suppressive function of nTreg cells. In other researches, addition of compounds such as rapamycin, retinoic acid, transforming growth factor-β (TGF-β) or combination of IL-2, DNA methyltransferase and histone deacetylase inhibitors could provide us sufficient Treg cells with phenotypic and functional stability [107-109]. Lu Y and his colleagues suggest that tTreg cells treated with miR-146b-5p antagomir show higher expansion and enhanced suppressive function in vitro [110]. Zhang D et al. showed D-mannose can specifically induced Foxp3 expression and Treg cell fate in naive T cells in vitro [111]. In addition, probiotics and prebiotics also exert profound effect on Treg induction and expansion via different mechanisms in animal models or human cells cultured

11

Journal Pre-proof

in vitro [112]. And these discoveries may provide new protocols to obtain sufficient Treg cells for adoptive cell therapy or directly regulate Treg cells in vivo for the treatment of autoimmune diseases. Treg adoptive therapy has been applied in many autoimmune diseases. In SLE, Maria et al. found that autologous adoptive Treg therapy augment activated Treg cells in inflamed skin [97].

of

And this phenomenon may promote the resolution of skin inflammation in SLE. In type 1 diabetes

ro

(T1D), patients treated with autologous expanded ex vivo Treg cells obtain certainty degree of remission with no severe adverse effects observed [98,99]. In these studies, researchers also found

-p

that treatment with Treg cells can prolong survival of β-cells in T1D. Another disease widely

re

treated with Treg transfer is Graft-Versus-Host Disease (GVHD) [102,103]. In these clinical trials

lP

of GVHD, Treg cell therapy has also been proven to be safe and effective.

na

Aside from polyclonal Treg cells infusion, antigen-specific Treg cells have been proven to be vastly superior for therapeutic use in animal models [113,114]. There are various ways to obtain

Jo ur

antigen-specific Treg cells, including antigen-stimulated expansion [115,116], T cell receptor (TCR) overexpression [117,118] and the use of chimeric antigen receptors (CARs) [119]. And antigen-specific Treg cells therapy has already been proved to be a promising clinical application for treating autoimmune diseases [115,120-123]. Regarding to particular Treg subset involved in Treg therapy, there are few studies to date. In Crohn’s disease, in vitro expanded CD4+CD25+CD127loCD45RA+ naïve Treg cells from patients’ blood home to inflammation sites in mouse model and suppress the activation of T cells [124]. In GVHD, researchers uncover that a higher proportion of aTreg cells in hematopoietic stem cells harvested from allogeneic donors correlated with no subsequent development of acute GVHD in

12

Journal Pre-proof

corresponding transplant recipients [125]. And the therapeutic use of Th-like Treg subsets or tissue Treg cells in autoimmune disorders needs further delineate. It remains to be verified next whether using specific subpopulation of Treg cells will be an optimum proposal in Treg adoptive therapy. Treg-based cellular transfer may be a hopeful treatment for various immune-related diseases in the near further. The major bottleneck of the Treg adoptive therapy is how to maintain the

of

phenotypic and functional stability of induced or expanded Treg cells in vivo and how to make

ro

better use specific subset of these fascinating cells in therapy. And further studies need to be conducted with more participates in longer follow-up period.

-p

5.2 Low-dose IL-2 treatment

re

Treg cells constitutively express CD25 (IL-2 receptor α chain) at high level, which, together with

lP

the β and γ chains, forms the high-affinity IL-2 receptor. And IL-2 has been considered an

na

essential growth factor for lymphocytes and is also a requisite for the proliferation and function of Treg cells [126]. Since researchers have shown that low-dose IL-2 (at single doses from 0.33 to

Jo ur

4.5 million IU) could selectively promote the development and maintenance of Treg cells without activation of Teff cells [127,128], utilization of low-dose IL-2 has become a new remedy of autoimmune diseases.

In early studies of lupus mice, researchers showed exogenous recombinant IL-2 (rIL-2) treatment could partially restore Treg : Tconv balance and effectively delay disease progression and decrease mortality [129]. In NOD mice, Johnson MC et al. suggested that β-cell–specific IL-2 therapy not only increase the number of Treg cells in islet but also enhance fitness of islet Treg and suppress T1D in diabetes-free NOD mice [130]. In human T1D, IL-2 is well tolerated at low dose with no serious adverse events, but adverse

13

Journal Pre-proof

events such as injection-site reaction and influenza-like syndrome which are all transient and of mild to moderate grades often occur [131-133]. Although low-dose IL-2 therapy is thought to be safe and can effectively augment Treg cells in T1D, but the numbers of other cells expressing IL-2 receptor – NK cells and eosinophils – are also increased [133], and efficacy of low-does IL-2 treatment needs further studies. As for GVHD, several studies have proven that low-dose IL-2

of

therapy is an effective regimen which obtain well tolerance and may reduce the incidence of

ro

GVHD and infection [134,135]. However, selective expansion of NK cells also occurred during low-dose IL-2 therapy [136]. In SLE, administration of low-dose rIL-2 to active SLE patients

-p

resulted in extensive expansion of Treg cells, enhanced Treg function in peripheral blood and

re

evidently reduced disease activity [137-139]. And a study in primary Sjogren's syndrome come to

lP

the same conclusion and further confirmed the unique therapeutic effect of low-dose IL-2

na

treatment on immune-related disorders [140]. Another recent clinical trial in 11 autoimmune diseases including RA, AS, SLE, psoriasis, Behcet's disease, granulomatosis with polyangiitis,

Jo ur

Takayasu's disease, Crohn's disease, ulcerative colitis, autoimmune hepatitis and sclerosing cholangitis using low-dose IL-2 therapy in 46 patients also confirmed that this treatment selectively activate Treg cells and is safe across different diseases [141]. Furthermore, many clinical trials in different diseases such as RA (NCT02467504, NCT01988506) are still under way. These on-going studies may provide us with stronger evidence that IL-2 maybe the next revolution in immunotherapy. In addition, IL-2 complexed with several different monoclonal antibodies (IL-2/mAb complexes) emerged in recent year for their more selective proliferation of Treg cells and prolonged half-life of IL-2 in vivo. IL-2/mAb complexes, such as JES6–1 and F5111.2, bind to

14

Journal Pre-proof

IL-2 and alter its conformation and binding interaction to IL-2R subunits,result in selective expansion of Treg cells [142]. Injection of IL-2/JES6–1 complexes sufficiently increases Treg proportion and alleviates diseases in animal models such as collagen-induced arthritis (CIA) [143], lupus nephritis [144] and congestive heart failure [145]. F5111.2, a human anti-IL-2 antibody, complexed with IL-2 also plays a protective role in mouse model of NOD, experimental

of

autoimmune encephalomyelitis and GVHD [142]. However, the safety and efficacy of IL-2/mAb

ro

complexes in human need further verification.

In conclusion, there is still a long way to go before this potential therapy can be used in

-p

clinical practice. What is the safe and effective dose range including cumulative dose and whether

re

there will be long-time complication require further researches. And double-blind,

na

indispensable.

lP

placebo-controlled randomized trials and studies of deeper mechanism of IL-2 treatment are also

5.3 Pharmacologic targeting of Treg

Jo ur

As scientists uncovered several signals promoting Treg survival and function, drugs targeting these signaling pathways could preserve Treg numbers and function, becoming potential therapeutic approaches of autoimmune diseases. mTOR kinase signaling plays an important role in T cell differentiation and function. Treg cells inhibited mTOR by rapamycin show improved lineage and functional stability in ex-vivo expansion experiment [109]. And mTOR-deficient T cells fail to differentiate to Th1, Th2 and Th17 cells but divert to Treg cells [146]. In animal model of many autoimmune diseases, such as autoimmune pancreatitis [147], T1D [148] and SLE [149], treatment with rapamycin lead to significant expansion of Treg cells and remission of diseases. In human SLE, rapamycin also

15

Journal Pre-proof

exerts safety and efficiency in treatment of this disease [150]. Immune dysregulation polyendocrinopathy enteropathy X-linked (IPEX) syndrome is a fatal autoimmune disorder caused by loss-of-function mutations in Foxp3, leading to impaired Treg function and autoimmunity [151,152]. A lasted research demonstrates that rapamycin partial restores Treg function in IPEX patients

by

upregulating

immunoregulatory

protein

of

Glucocorticoid-Induced-TNFR-Related-Protein (GITR) and Epstein-Barr-virus-Induced-3 (Ebi3)

ro

and results in a beneficial effect on clinical and histological evolution of IPEX pathology [153]. Histone deacetylases (HDACs) are portent epigenetic regulators of T cell-mediated immunity.

-p

Researches show that HDACs inhibitors act as an anti-inflammatory agent and become potential

re

therapeutic approaches to autoimmune diseases [154]. HDACs inhibitors promote Treg function

lP

both in vivo and in vitro and lead to long-term allograft survival in mice [155]. In human,

na

administration of vorinostat, an HDACs inhibitors, significantly increase the number of Treg cells and expression of Foxp3 in patients with hematologic malignances who have received allogeneic

Jo ur

hematopoietic stem cell transplantation [156]. And there is a significant reduction in the incidence of severe GVHD. HDACs inhibitors could also enhance Treg function [157] or inhibit Th17 cell differentiation in CIA mouse model [158]. Peripheral blood mononuclear cells from RA patients cultured with lipopolysaccharide and HDACs inhibitor in vitro show increased proportion of iTreg cells and higher expression of IL-10 [157]. These results suggest HDACs inhibitors may have a beneficial effect in RA treatment. 6. Conclusion In autoimmune diseases, Treg cells have become currently a subject of intense research in both pathogenesis and treatment. Quantity or quality defects in Treg were found in many diseases and

16

Journal Pre-proof

part of the Treg relevant etiology and mechanisms have been revealed. As a consequence, Treg targeted therapy emerged and became a promising protocol curing these diseases. However, because of Treg heterogenity and plasticity as well as complexity of the immune system, there are still various challenges. How Treg cells regulate immune response in different conditions or in different tissue and how Treg and conventional T cell in different subsets interact with each other

-p re

Compliance with Ethical Standards

ro

comprehensive and thorough understanding of Treg cells.

of

in diverse disease status need further researches. There is still a long way to go for us to get a

lP

Funding This work was supported by grants from the National Key Research and Development

na

Program of China [grant number 2017YFC0909002] and the National Natural Science Foundation

Jo ur

of China [grant number 81801599].

Conflict of Interest The authors declare that they have no conflict of interest.

Human and Animal Right and Informed Consent This article does not contain any studies with human and animal subjects performed by any of the authors.

Reference [1] Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol 1995; 17

Journal Pre-proof

155:1151-64. [2] Hori S. Control of Regulatory T Cell Development by the Transcription Factor Foxp3. Science 2003; 299:1057-61. 10.1126/science.1079490. [3] Fontenot JD, Gavin MA, Rudensky AY. Foxp3 programs the development and function of CD4+CD25+ regulatory T cells. Nat Immunol 2003; 4:330-36. 10.1038/ni904.

of

[4] Khattri R, Cox T, Yasayko SA, Ramsdell F. An essential role for Scurfin in CD4+CD25+ T

ro

regulatory cells. Nat Immunol 2003; 4:337-42. 10.1038/ni909.

[5] Sarkar S, Fox DA. Regulatory T cell defects in rheumatoid arthritis. Arthritis & Rheumatism

-p

2007; 56:710-13. 10.1002/art.22415.

re

[6] van Amelsfort JMR, van Roon JAG, Noordegraaf M, Jacobs KMG, Bijlsma JWJ, Lafeber

lP

FPJG, et al. Proinflammatory mediator-induced reversal of CD4+CD25+ regulatory T

10.1002/art.22414.

na

cell-mediated suppression in rheumatoid arthritis. Arthritis and rheumatism 2007; 56:732-42.

Jo ur

[7] Matsuki F, Saegusa J, Miyamoto Y, Misaki K, Kumagai S, Morinobu A. CD45RA–Foxp3high activated/effector regulatory T cells in the CCR7+CD45RA–CD27+CD28+ central memory subset are decreased in peripheral blood from patients with rheumatoid arthritis. Biochem Bioph Res Co 2013; 438:778-83. 10.1016/j.bbrc.2013.05.120. [8] Nie H, Zheng Y, Li R, Guo TB, He D, Fang L, et al. Phosphorylation of FOXP3 controls regulatory T cell function and is inhibited by TNF-α in rheumatoid arthritis. Nat Med 2013; 19:322-28. 10.1038/nm.3085. [9] Komatsu N, Okamoto K, Sawa S, Nakashima T, Oh-hora M, Kodama T, et al. Pathogenic conversion of Foxp3+ T cells into TH17 cells in autoimmune arthritis. Nat Med 2014; 20:62-68.

18

Journal Pre-proof

10.1038/nm.3432. [10] Burmester GR, Feist E, Dörner T. Emerging cell and cytokine targets in rheumatoid arthritis. Nature reviews. Rheumatology 2014; 10:77-88. 10.1038/nrrheum.2013.168. [11] Bonelli M, Savitskaya A, von Dalwigk K, Steiner CW, Aletaha D, Smolen JS, et al. Quantitative and qualitative deficiencies of regulatory T cells in patients with systemic lupus

of

erythematosus (SLE). Int Immunol 2008; 20:861-68. 10.1093/intimm/dxn044.

ro

[12] Bonelli M, Smolen JS, Scheinecker C. Treg and lupus. Ann Rheum Dis 2009; 69:i65-66. 10.1136/ard.2009.117135.

-p

[13] La Cava A. Tregs in SLE: an Update. Curr Rheumatol Rep 2018; 20.

re

10.1007/s11926-018-0714-8.

lP

[14] Guo H, Zheng M, Zhang K, Yang F, Zhang X, Han Q, et al. Functional defects in

10.1038/srep37559.

na

CD4+CD25highFoxP3+ regulatory cells in ankylosing spondylitis. Sci Rep 2016; 6:37559.

Jo ur

[15] Fenoglio D, Bernuzzi F, Battaglia F, Parodi A, Kalli F, Negrini S, et al. Th17 and regulatory T lymphocytes in primary biliary cirrhosis and systemic sclerosis as models of autoimmune fibrotic diseases. Autoimmun Rev 2012; 12:300-04. 10.1016/j.autrev.2012.05.004. [16] Frantz C, Auffray C, Avouac J, Allanore Y. Regulatory T Cells in Systemic Sclerosis. Front Immunol 2018; 9. 10.3389/fimmu.2018.02356. [17] NJ B. How T cells lose FOXP3. Nature reviews. Rheumatology 2019; 15:127. 10.1038/s41584-019-0171-4. [18] CN Z, Z X, GC W, YM M, LN L, Qian-Wu, et al. Emerging role of air pollution in autoimmune diseases. Autoimmun Rev 2019; 18:607-14. 10.1016/j.autrev.2018.12.010.

19

Journal Pre-proof

[19] Miyara M, Ito Y, Sakaguchi S. TREG-cell therapies for autoimmune rheumatic diseases. Nature reviews. Rheumatology 2014; 10:543-51. 10.1038/nrrheum.2014.105. [20] Sharabi A, Tsokos MG, Ding Y, Malek TR, Klatzmann D, Tsokos GC. Regulatory T cells in the treatment of disease. Nat Rev Drug Discov 2018; 17:823-44. 10.1038/nrd.2018.148. [21] Panduro M, Benoist C, Mathis D. Tissue Tregs. Annu Rev Immunol 2016; 34:609-33.

of

10.1146/annurev-immunol-032712-095948.

ro

[22] Sharma A, Rudra D. Emerging Functions of Regulatory T Cells in Tissue Homeostasis. Front Immunol 2018; 9:883. 10.3389/fimmu.2018.00883.

-p

[23] LW C, CJ W, TT K, K B, Y W, KM V, et al. The inhibitory cytokine IL-35 contributes to

re

regulatory T-cell function. Nature 2007; 450:566-69. 10.1038/nature06306.

lP

[24] K S, M W, H J, J K, AH E. Induction of tolerance by IL-10-treated dendritic cells. Journal

na

of immunology (Baltimore, Md. : 1950) 1997; 159:4772-80. Undefined. [25] AG J, J L, S T, KH M. Suppression of antitumor immunity by IL-10 and

Jo ur

TGF-beta-producing T cells infiltrating the growing tumor: influence of tumor environment on the induction of CD4+ and CD8+ regulatory T cells. Journal of immunology (Baltimore, Md. : 1950) 2006; 177:896-904. 10.4049/jimmunol.177.2.896. [26] Chinen T, Kannan AK, Levine AG, Fan X, Klein U, Zheng Y, et al. An essential role for the IL-2 receptor in Treg cell function. Nat Immunol 2016; 17:1322-33. 10.1038/ni.3540. [27] Kobie JJ, Shah PR, Yang L, Rebhahn JA, Fowell DJ, Mosmann TR. T Regulatory and Primed Uncommitted CD4 T Cells Express CD73, Which Suppresses Effector CD4 T Cells by Converting 5'-Adenosine Monophosphate to Adenosine. The Journal of Immunology 2006; 177:6780-86. 10.4049/jimmunol.177.10.6780.

20

Journal Pre-proof

[28] Deaglio S, Dwyer KM, Gao W, Friedman D, Usheva A, Erat A, et al. Adenosine generation catalyzed by CD39 and CD73 expressed on regulatory T cells mediates immune suppression. The Journal of Experimental Medicine 2007; 204:1257-65. 10.1084/jem.20062512. [29] Mandapathil M, Hilldorfer B, Szczepanski MJ, Czystowska M, Szajnik M, Ren J, et al. and

Accumulation

CD4+CD25highFOXP3+

Regulatory

of

Immunosuppressive

T

Cells.

J

Biol

Adenosine Chem

2010;

by

Human

285:7176-86.

of

Generation

ro

10.1074/jbc.M109.047423.

[30] WJ G, JW V, W B, M C, JP A, TJ L. Human T regulatory cells can use the perforin pathway

-p

to cause autologous target cell death. Immunity 2004; 21:589-601. 10.1016/j.immuni.2004.09.002.

on

antigen-presenting

cells.

Eur

J

Immunol

2000;

30:1538-43.

lP

molecules

re

[31] Cederbom L, Hall H, Ivars F. CD4+CD25+ regulatory T cells down-regulate co-stimulatory

na

10.1002/1521-4141(200006)30:6<1538::AID-IMMU1538>3.0.CO;2-X. [32] Oderup C, Cederbom L, Makowska A, Cilio CM, Ivars F. Cytotoxic T lymphocyte

CD4+CD25+

Jo ur

antigen-4-dependent down-modulation of costimulatory molecules on dendritic cells in regulatory

T-cell-mediated

suppression.

Immunology

2006;

118:240-49.

10.1111/j.1365-2567.2006.02362.x.

[33] Schmidleithner L, Thabet Y, Schönfeld E, Köhne M, Sommer D, Abdullah Z, et al. Enzymatic Activity of HPGD in Treg Cells Suppresses Tconv Cells to Maintain Adipose Tissue Homeostasis and Prevent Metabolic Dysfunction. Immunity 2019. 10.1016/j.immuni.2019.03.014. [34] Akkaya B, Oya Y, Akkaya M, Al Souz J, Holstein AH, Kamenyeva O, et al. Regulatory T cells mediate specific suppression by depleting peptide–MHC class II from dendritic cells. Nat Immunol 2019; 20:218-31. 10.1038/s41590-018-0280-2.

21

Journal Pre-proof

[35] Thornton AM, Korty PE, Tran DQ, Wohlfert EA, Murray PE, Belkaid Y, et al. Expression of Helios, an Ikaros transcription factor family member, differentiates thymic-derived from peripherally

induced

Foxp3+

T

regulatory

cells.

J

Immunol

2010;

184:3433-41.

10.4049/jimmunol.0904028. [36] Weiss JM, Bilate AM, Gobert M, Ding Y, Curotto De Lafaille MA, Parkhurst CN, et al. Neuropilin 1 is expressed on thymus-derived natural regulatory T cells, but not mucosa-generated

ro

of

induced Foxp3+ T reg cells. The Journal of experimental medicine 2012; 209:1723-42. 10.1084/jem.20120914.

-p

[37] Yadav M, Louvet C, Davini D, Gardner JM, Martinez-Llordella M, Bailey-Bucktrout S, et

in

vivo.

The

Journal

of

Experimental

Medicine

2012;

209:1713-22.

lP

subsets

re

al. Neuropilin-1 distinguishes natural and inducible regulatory T cells among regulatory T cell

na

10.1084/jem.20120822.

[38] Elkord E. Helios Should Not Be Cited as a Marker of Human Thymus-Derived Tregs.

Jo ur

Commentary: Helios+ and Helios- Cells Coexist within the Natural FOXP3+ T Regulatory Cell Subset in Humans. Front Immunol 2016; 7:276. 10.3389/fimmu.2016.00276. [39] Szurek E, Cebula A, Wojciech L, Pietrzak M, Rempala G, Kisielow P, et al. Differences in Expression Level of Helios and Neuropilin-1 Do Not Distinguish Thymus-Derived from Extrathymically-Induced CD4+Foxp3+ Regulatory T Cells. Plos One 2015; 10:e141161. 10.1371/journal.pone.0141161. [40] Miyara M, Yoshioka Y, Kitoh A, Shima T, Wing K, Niwa A, et al. Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity 2009; 30:899-911. 10.1016/j.immuni.2009.03.019.

22

Journal Pre-proof

[41] Kim J, Chae J, Kim S, Ha J. Subpopulations of Regulatory T Cells in Rheumatoid Arthritis, Systemic Lupus Erythematosus, and Behcet's Disease. J Korean Med Sci 2012; 27:1009. 10.3346/jkms.2012.27.9.1009. [42] Sambucci M, Gargano F, De Rosa V, De Bardi M, Picozza M, Placido R, et al. FoxP3 isoforms and PD-1 expression by T regulatory cells in multiple sclerosis. Sci Rep-Uk 2018; 8. 10.1038/s41598-018-21861-5

ro

of

[43] Pan X, Yuan X, Zheng Y, Wang W, Shan J, Lin F, et al. Increased CD45RA+FoxP3low regulatory T cells with impaired suppressive function in patients with systemic lupus

-p

erythematosus. Plos One 2012; 7:e34662. 10.1371/journal.pone.0034662.

re

[44] Silva-Neta HL, Brelaz-de-Castro M, Chagas M, Mariz HA, de Arruda RG, de Vasconcelos

lP

VF, et al. CD4+CD45RA-FOXP3low Regulatory T Cells as Potential Biomarkers of Disease

na

Activity in Systemic Lupus Erythematosus Brazilian Patients. Biomed Res Int 2018; 2018:3419565. 10.1155/2018/3419565.

Jo ur

[45] Koch MA, Tucker-Heard G, Perdue NR, Killebrew JR, Urdahl KB, Campbell DJ. The transcription factor T-bet controls regulatory T cell homeostasis and function during type 1 inflammation. Nat Immunol 2009; 10:595-602. 10.1038/ni.1731. [46] Kitz A, Dominguez-Villar M. Molecular mechanisms underlying Th1-like Treg generation and function. Cell Mol Life Sci 2017; 74:4059-75. 10.1007/s00018-017-2569-y. [47] Littringer K, Moresi C, Rakebrandt N, Zhou X, Schorer M, Dolowschiak T, et al. Common Features of Regulatory T Cell Specialization During Th1 Responses. Front Immunol 2018; 9:1344. 10.3389/fimmu.2018.01344. [48] Voo KS, Wang YH, Santori FR, Boggiano C, Wang YH, Arima K, et al. Identification of

23

Journal Pre-proof IL-17-producing FOXP3+ regulatory T cells in humans. Proc Natl Acad Sci U S A 2009; 106:4793-98. 10.1073/pnas.0900408106. [49] Zheng Y, Chaudhry A, Kas A, DeRoos P, Kim JM, Chu TT, et al. Regulatory T-cell suppressor program co-opts transcription factor IRF4 to control T(H)2 responses. Nature 2009; 458:351-56. 10.1038/nature07674.

of

[50] Arterbery AS, Osafo-Addo A, Avitzur Y, Ciarleglio M, Deng Y, Lobritto SJ, et al.

ro

Production of Proinflammatory Cytokines by Monocytes in Liver-Transplanted Recipients with De Novo Autoimmune Hepatitis Is Enhanced and Induces TH1-like Regulatory T Cells. J

-p

Immunol 2016; 196:4040-51. 10.4049/jimmunol.1502276.

re

[51] Dominguez-Villar M, Baecher-Allan CM, Hafler DA. Identification of T helper type 1–like,

lP

Foxp3+ regulatory T cells in human autoimmune disease. Nat Med 2011; 17:673-75.

na

10.1038/nm.2389.

[52] Piconese S, Timperi E, Pacella I, Schinzari V, Tripodo C, Rossi M, et al. Human OX40

Jo ur

tunes the function of regulatory T cells in tumor and nontumor areas of hepatitis C virus-infected liver tissue. Hepatology 2014; 60:1494-507. 10.1002/hep.27188. [53] Chellappa S, Hugenschmidt H, Hagness M, Line PD, Labori KJ, Wiedswang G, et al. Regulatory T cells that co-express RORγt and FOXP3 are pro-inflammatory and immunosuppressive and expand in human pancreatic cancer. Oncoimmunology 2015; 5:e1102828. 10.1080/2162402X.2015.1102828. [54] Kryczek I, Wu K, Zhao E, Wei S, Vatan L, Szeliga W, et al. IL-17+ regulatory T cells in the microenvironments of chronic inflammation and cancer. J Immunol 2011; 186:4388-95. 10.4049/jimmunol.1003251.

24

Journal Pre-proof

[55] Choi YS, Jung MK, Lee J, Choi SJ, Choi SH, Lee HW, et al. Tumor Necrosis Factor-producing T-regulatory Cells Are Associated With Severe Liver Injury in Patients With Acute Hepatitis A. Gastroenterology 2018; 154:1047-60. 10.1053/j.gastro.2017.11.277. [56] MacDonald KGB, Dawson NAJB, Huang QP, Dunne JVM, Levings MKP, Broady RM. Regulatory T cells produce profibrotic cytokines in the skin of patients with systemic sclerosis.

of

Journal of Allergy and Clinical Immunology, The 2015; 135:946-55. 10.1016/j.jaci.2014.12.1932.

ro

[57] Burzyn D, Benoist C, Mathis D. Regulatory T cells in nonlymphoid tissues. Nat Immunol 2013; 14:1007-13. 10.1038/ni.2683.

-p

[58] Q Z, X S, L X, Z X, M B, T D. A Unique Population: Adipose-Resident Regulatory T Cells.

re

Front Immunol 2018; 9:2075. 10.3389/fimmu.2018.02075.

lP

[59] M F, L H, D C, A N, J W, A N, et al. Lean, but not obese, fat is enriched for a unique

10.1038/nm.2002.

na

population of regulatory T cells that affect metabolic parameters. Nat Med 2009; 15:930-39.

Jo ur

[60] D K, van Panhuys N, C L, AM M, D C, CM M, et al. Antigen- and cytokine-driven accumulation of regulatory T cells in visceral adipose tissue of lean mice. Cell Metab 2015; 21:543-57. 10.1016/j.cmet.2015.03.005. [61] D C, M F, A L, N K, J L, SE S, et al. PPAR-γ is a major driver of the accumulation and phenotype of adipose tissue Treg cells. Nature 2012; 486:549-53. 10.1038/nature11132. [62] US P, TB W, PO C, L J, M P. Female mice are protected against high-fat diet induced metabolic syndrome and increase the regulatory T cell population in adipose tissue. Plos One 2012; 7:e46057. 10.1371/journal.pone.0046057. [63] M Y, L S, Q T, C Z, Y W, J L. Depletion of Regulatory T Cells in Visceral Adipose Tissues

25

Journal Pre-proof

Contributes to Insulin Resistance in Hashimoto's Thyroiditis. Front Physiol 2018; 9:136. 10.3389/fphys.2018.00136. [64] S K, M B, VB O, I S, F H, MMH M, et al. A Stat6/Pten Axis Links Regulatory T Cells with Adipose Tissue Function. Cell Metab 2017; 26:475-92. 10.1016/j.cmet.2017.08.008. [65] D M, TP S, K S, DM R, M D, C S, et al. Brown adipose tissue harbors a distinct

of

sub-population of regulatory T cells. Plos One 2015; 10:e118534. 10.1371/journal.pone.0118534.

ro

[66] D B, C B, D M. Regulatory T cells in nonlymphoid tissues. Nat Immunol 2013; 14:1007-13. 10.1038/ni.2683.

-p

[67] Dalia B, Wilson K, Dmitriy K, L SJ, Massimiliano C, Young J, et al. A special population

re

of regulatory T cells potentiates muscle repair. Cell 2013;155.

lP

[68] W K, D B, M P, KK W, YC J, AJ W, et al. Poor Repair of Skeletal Muscle in Aging Mice

na

Reflects a Defect in Local, Interleukin-33-Dependent Accumulation of Regulatory T Cells. Immunity 2016; 44:355-67. 10.1016/j.immuni.2016.01.009.

Jo ur

[69] J C, W K, C B, D M. T cell receptor specificity drives accumulation of a reparative population of regulatory T cells within acutely injured skeletal muscle. P Natl Acad Sci Usa 2019. 10.1073/pnas.1914848116.

[70] NA Y, R S, AK F, BH K, B B, WN J. Aberrant muscle antigen exposure in mice is sufficient to cause myositis in a Treg cell-deficient milieu. Arthritis and rheumatism 2013; 65:3259-70. 10.1002/art.38184. [71] SA V, W R, L M, A K, T S, JG T, et al. Regulatory T cells suppress muscle inflammation and

injury

in

muscular

dystrophy.

Sci

10.1126/scitranslmed.3009925.

26

Transl

Med

2014;

6:142r-258r.

Journal Pre-proof

[72] RM J, SJ B, J W, RR H, IJ B, EA W. Regulatory T Cells Promote Myositis and Muscle Damage in Toxoplasma gondii Infection. Journal of immunology (Baltimore, Md. : 1950) 2017; 198:352-62. 10.4049/jimmunol.1600914. [73] J S, S Y, S S. Induction of tumor immunity by removing CD25+CD4+ T cells: a common basis between tumor immunity and autoimmunity. Journal of immunology (Baltimore, Md. : 1950)

of

1999; 163:5211-18.

ro

[74] S O, I T, J S, S S, T F, E N. Tumor rejection by in vivo administration of anti-CD25 (interleukin-2 receptor alpha) monoclonal antibody. Cancer Res 1999; 59:3128-33.

-p

[75] Y Z, J L, NG S, W Y, HJ L, ZC N, et al. Regulatory T-cell Depletion Alters the Tumor

re

Microenvironment and Accelerates Pancreatic Carcinogenesis. Cancer Discov 2020; 10:422-39.

lP

10.1158/2159-8290.CD-19-0958.

na

[76] Ehara WMA, Maeda OJM, Karine AM, Julio CV. Regulatory T cells and breast cancer: implications for immunopathogenesis. Cancer metastasis reviews 2010; 29:569-79.

Jo ur

[77] Luis V, Karolina PA, W FJ, Tomoki I, Yong-Jun L, Jacques B, et al. Circulating tumor antigen-specific regulatory T cells in patients with metastatic melanoma. P Natl Acad Sci Usa 2007; 104:20884-89.

[78] Tatiana A, Tianyi Z, Dmitri N, Sunil S, Jason S, Abhishek R, et al. Human lung tumor FOXP3+ Tregs upregulate four "Treg-locking" transcription factors. JCI insight 2017;2. [79] M FD, A DR, T VC, Inti Z, Alessandro L, Urs Z, et al. High frequency of tumor-infiltrating FOXP3+ regulatory T cells predicts improved survival in mismatch repair-proficient colorectal cancer patients. Int J Cancer 2010; 126:2635-43. [80] A SF, L RR, M AS, L KK, R FN, J SD. Intraepithelial effector (CD3+)/regulatory (FoxP3+)

27

Journal Pre-proof

T-cell ratio predicts a clinical outcome of human colon carcinoma. Gastroenterology 2009; 137:1270-79. [81] Takuro S, Hiroyoshi N, Hisashi W, Yuji N, Daisuke S, Koji A, et al. Two FOXP3 +CD4+ T cell subpopulations distinctly control the prognosis of colorectal cancers. Nat Med 2016; 22:679-84.

of

[82] H W, F F, YC T, X X, MP T, R Z, et al. CD36-mediated metabolic adaptation supports

ro

regulatory T cell survival and function in tumors. Nat Immunol 2020; 21:298-308. 10.1038/s41590-019-0589-5.

-p

[83] S H, A K, R S, T D, T N, T H, et al. Percutaneous sensitization is limited by in situ

re

inhibition of cutaneous dendritic cell migration through skin-resident regulatory T cells. The

lP

Journal of allergy and clinical immunology 2019; 144:1343-53. 10.1016/j.jaci.2019.05.033.

na

[84] N M, JM L, U J, O B, C K, NK O, et al. RORα-expressing T regulatory cells restrain allergic skin inflammation. Science immunology 2018; 3. 10.1126/sciimmunol.aao6923.

to

suppress

Jo ur

[85] LA K, JN C, N P, PM S, AN M, JM M, et al. Regulatory T cells in skin are uniquely poised profibrotic

immune

responses.

Science

immunology

2019;

4.

10.1126/sciimmunol.aaw2910.

[86] A N, N P, HA T, P M, M E, TC S, et al. Cutting Edge: Regulatory T Cells Facilitate Cutaneous Wound Healing. Journal of immunology (Baltimore, Md. : 1950) 2016; 196:2010-14. 10.4049/jimmunol.1502139. [87] TC S, KS V, HA T, SV G, ML P, A N, et al. A Wave of Regulatory T Cells into Neonatal Skin

Mediates

Tolerance

to

Commensal

10.1016/j.immuni.2015.10.016.

28

Microbes.

Immunity

2015;

43:1011-21.

Journal Pre-proof

[88] N A, B Z, RS R, ML P, HA T, K L, et al. Regulatory T Cells in Skin Facilitate Epithelial Stem Cell Differentiation. Cell 2017; 169:1119-29. 10.1016/j.cell.2017.05.002. [89] C S, T K, A C, A F, K A, EA W, et al. The alarmin IL-33 promotes regulatory T-cell function in the intestine. Nature 2014; 513:564-68. 10.1038/nature13577. [90] DM Z, van Loosdregt J, A G, CP B, A G, M S, et al. Amphiregulin enhances regulatory T

of

cell-suppressive function via the epidermal growth factor receptor. Immunity 2013; 38:275-84.

ro

10.1016/j.immuni.2012.09.023.

[91] Mo X, Maria P, Yi D, Ren Y, Christy A, J HO, et al. c-MAF-dependent regulatory T cells

-p

mediate immunological tolerance to a gut pathobiont. Nature 2018; 554:373-77.

re

[92] Soon KK, Sung-Wook H, Daehee H, Jaeu Y, Jisun J, Bo-Gie Y, et al. Dietary antigens limit

na

2016; 351:858-63.

lP

mucosal immunity by inducing regulatory T cells in the small intestine. Science (New York, N.Y.)

[93] M D, Y X, M B, C G, NG F. CCR5 signaling suppresses inflammation and reduces adverse

Jo ur

remodeling of the infarcted heart, mediating recruitment of regulatory T cells. The American journal of pathology 2010; 176:2177-87. 10.2353/ajpath.2010.090759. [94] MM Z, B F, A H, J Z, J L, F N, et al. Regulatory T cells protect from local and systemic bone destruction in arthritis. Journal of immunology (Baltimore, Md. : 1950) 2010; 184:7238-46. 10.4049/jimmunol.0903841. [95] MM Z, K S, A H, K E, C B, F N, et al. Increased bone density and resistance to ovariectomy-induced bone loss in FoxP3-transgenic mice based on impaired osteoclast differentiation. Arthritis and rheumatism 2010; 62:2328-38. 10.1002/art.27535. [96] Y D, T O, M D, R P, SR M, P B, et al. Regulatory T cells promote myelin regeneration in

29

Journal Pre-proof

the central nervous system. Nat Neurosci 2017; 20:674-80. 10.1038/nn.4528. [97] Dall'Era M, Pauli ML, Remedios K, Taravati K, Sandova PM, Putnam AL, et al. Adoptive Treg Cell Therapy in a Patient With Systemic Lupus Erythematosus. Arthritis Rheumatol 2019; 71:431-40. 10.1002/art.40737. [98] Marek-Trzonkowska N, Myśliwiec M, Dobyszuk A, Grabowska M, Derkowska I, Juścińska J, et al. Therapy of type 1 diabetes with CD4+CD25highCD127- regulatory T cells prolongs survival

ro

of

of pancreatic islets — Results of one year follow-up. Clin Immunol 2014; 153:23-30. 10.1016/j.clim.2014.03.016.

-p

[99] Bluestone JA, Buckner JH, Fitch M, Gitelman SE, Gupta S, Hellerstein MK, et al. Type 1

re

diabetes immunotherapy using polyclonal regulatory T cells. Sci Transl Med 2015; 7:189r-315r.

lP

10.1126/scitranslmed.aad4134.

na

[100] Marek-Trzonkowska N, Mysliwiec M, Iwaszkiewicz-Grzes D, Gliwinski M, Derkowska I, Zalinska M, et al. Factors affecting long-term efficacy of T regulatory cell-based therapy in type 1

Jo ur

diabetes. J Transl Med 2016; 14:332. 10.1186/s12967-016-1090-7. [101] Todo S, Yamashita K, Goto R, Zaitsu M, Nagatsu A, Oura T, et al. A pilot study of operational tolerance with a regulatory T-cell-based cell therapy in living donor liver transplantation. Hepatology 2016; 64:632-43. 10.1002/hep.28459. [102] Brunstein CG, Miller JS, Cao Q, McKenna DH, Hippen KL, Curtsinger J, et al. Infusion of ex vivo expanded T regulatory cells in adults transplanted with umbilical cord blood: safety profile and detection kinetics. Blood 2011; 117:1061-70. 10.1182/blood-2010-07-293795. [103] Brunstein CG, Miller JS, McKenna DH, Hippen KL, DeFor TE, Sumstad D, et al. Umbilical cord blood-derived T regulatory cells to prevent GVHD: kinetics, toxicity profile, and clinical

30

Journal Pre-proof

effect. Blood 2016; 127:1044-51. 10.1182/blood-2015-06-653667. [104] Voskens CJ, Fischer A, Roessner S, Lorenz C, Hirschmann S, Atreya R, et al. Characterization and Expansion of Autologous GMP-ready Regulatory T Cells for TREG-based Cell Therapy in Patients with Ulcerative Colitis. Inflamm Bowel Dis 2017; 23:1348-59. 10.1097/MIB.0000000000001192. [105] Hoffmann P, Eder R, Kunz-Schughart LA, Andreesen R, Edinger M. Large-scale in vitro

ro

of

expansion of polyclonal human CD4+CD25high regulatory T cells. Blood 2004; 104:895-903. 10.1182/blood-2004-01-0086.

-p

[106] Hippen KL, Merkel SC, Schirm DK, Sieben CM, Sumstad D, Kadidlo DM, et al. Massive

re

ex Vivo Expansion of Human Natural Regulatory T Cells (Tregs) with Minimal Loss of in Vivo

lP

Functional Activity. Sci Transl Med 2011; 3:41r-83r. 10.1126/scitranslmed.3001809.

na

[107] Angelika S, Matilda E, Ming-Mei S, Heiko W, Jesper T. Comparative Analysis of Protocols to Induce Human CD4+Foxp3+ Regulatory T Cells by Combinations of IL-2, TGF-beta, Retinoic

Jo ur

Acid, Rapamycin and Butyrate. Plos One 2016; 11:e148474. [108] Makoto M, Driss C, Aude B, Jérémie G, Delphine S, Françoise N, et al. Combination of IL-2, rapamycin, DNA methyltransferase and histone deacetylase inhibitors for the expansion of human regulatory T cells. Oncotarget 2017; 8:104733-44. [109] Maura R, Roberto S, Suzan S, Camillus C, Maryam M, Yao LJ, et al. Ex vivo-expanded but not in vitro-induced human regulatory T cells are candidates for cell therapy in autoimmune diseases thanks to stable demethylation of the FOXP3 regulatory T cell-specific demethylated region. Journal of immunology (Baltimore, Md. : 1950) 2015; 194:113-24. [110] Lu Y, Hippen KL, Lemire AL, Gu J, Wang W, Ni X, et al. miR-146b antagomir-treated

31

Journal Pre-proof

human Tregs acquire increased GVHD inhibitory potency. Blood 2016; 128:1424-35. 10.1182/blood-2016-05-714535. [111] Zhang D, Chia C, Jiao X, Jin W, Kasagi S, Wu R, et al. D-mannose induces regulatory T cells and suppresses immunopathology. Nat Med 2017; 23:1036-45. 10.1038/nm.4375. [112] M D, P K, NC L, EH K. Induction of regulatory T cells: A role for probiotics and prebiotics

of

to suppress autoimmunity. Autoimmun Rev 2016; 15:379-92. 10.1016/j.autrev.2016.01.002.

ro

[113] Q T, KJ H, M B, EB F, G S, J Y, et al. In vitro-expanded antigen-specific regulatory T cells suppress autoimmune diabetes. The Journal of experimental medicine 2004; 199:1455-65.

-p

10.1084/jem.20040139.

re

[114] GP W, CA N, SA X, GM B, A H, TN S, et al. Adoptive therapy with redirected primary

lP

regulatory T cells results in antigen-specific suppression of arthritis. P Natl Acad Sci Usa 2009;

na

106:19078-83. 10.1073/pnas.0907396106.

[115] Izquierdo C, Ortiz AZ, Presa M, Malo S, Montoya A, Garabatos N, et al. Treatment of T1D

Jo ur

via optimized expansion of antigen-specific Tregs induced by IL-2/anti-IL-2 monoclonal antibody complexes and peptide/MHC tetramers. Sci Rep 2018; 8:8106. 10.1038/s41598-018-26161-6. [116] Xiang YJ, Ren M, Jiang H, Yang TT, He Y, Ao DH, et al. Ex vivo expansion of antigen-specific CD4+CD25+ regulatory T cells from autologous naive CD4+ T cells of multiple sclerosis patients as a potential therapeutic approach. Eur Rev Med Pharmacol Sci 2016; 20:5261-70. [117] Hull CM, Nickolay LE, Estorninho M, Richardson MW, Riley JL, Peakman M, et al. Generation of human islet-specific regulatory T cells by TCR gene transfer. J Autoimmun 2017; 79:63-73. 10.1016/j.jaut.2017.01.001.

32

Journal Pre-proof

[118] Brusko TM, Koya RC, Zhu S, Lee MR, Putnam AL, McClymont SA, et al. Human antigen-specific regulatory T cells generated by T cell receptor gene transfer. Plos One 2010; 5:e11726. 10.1371/journal.pone.0011726. [119] MacDonald KG, Hoeppli RE, Huang Q, Gillies J, Luciani DS, Orban PC, et al. Alloantigen-specific regulatory T cells generated with a chimeric antigen receptor. J Clin Invest

of

2016; 126:1413-24. 10.1172/JCI82771.

ro

[120] Kasahara H, Kondo T, Nakatsukasa H, Chikuma S, Ito M, Ando M, et al. Generation of allo-antigen-specific induced Treg stabilized by vitamin C treatment and its application for

-p

prevention of acute graft versus host disease model. Int Immunol 2017; 29:457-69.

re

10.1093/intimm/dxx060.

lP

[121] Sun G, Hou Y, Gong W, Liu S, Li J, Yuan Y, et al. Adoptive Induced Antigen-Specific Treg

na

Cells Reverse Inflammation in Collagen-Induced Arthritis Mouse Model. Inflammation 2018; 41:485-95. 10.1007/s10753-017-0704-4.

Jo ur

[122] Kasagi S, Zhang P, Che L, Abbatiello B, Maruyama T, Nakatsukasa H, et al. In vivo-generated antigen-specific regulatory T cells treat autoimmunity without compromising antibacterial immune response. Sci Transl Med 2014; 6:241r-278r. 10.1126/scitranslmed.3008895. [123] Desreumaux P, Foussat A, Allez M, Beaugerie L, Hebuterne X, Bouhnik Y, et al. Safety and efficacy of antigen-specific regulatory T-cell therapy for patients with refractory Crohn's disease. Gastroenterology 2012; 143:1207-17. 10.1053/j.gastro.2012.07.116. [124] Canavan JB, Scottà C, Vossenkämper A, Goldberg R, Elder MJ, Shoval I, et al. Developing in vitro expanded CD45RA+ regulatory T cells as an adoptive cell therapy for Crohn's disease. Gut 2016; 65:584-94. 10.1136/gutjnl-2014-306919.

33

Journal Pre-proof

[125] Roschupkina T, Juliusson G. Subpopulations of T Regulatory Cells in Blood Stem Cell Harvests Influence Development of Acute Graft Versus Host Disease in Allogeneic Transplant Recipients. Cytometry Part B: Clinical Cytometry 2018; 94:264-69. 10.1002/cyto.b.21404. [126] Boyman O, Sprent J. The role of interleukin-2 during homeostasis and activation of the immune system. Nat Rev Immunol 2012; 12:180-90. 10.1038/nri3156. [127] Hirakawa M, Matos T, Liu H, Koreth J, Kim HT, Paul NE, et al. Low-dose IL-2 selectively

of

activates subsets of CD4+ Tregs and NK cells. JCI Insight 2016; 1. 10.1172/jci.insight.89278.

ro

[128] Zhao Z, Zhang X, Su L, Xu L, Zheng Y, Sun J. Fine tuning subsets of CD4+ T cells by

-p

low-dosage of IL-2 and a new therapeutic strategy for autoimmune diseases. Int

re

Immunopharmacol 2018; 56:269-76. 10.1016/j.intimp.2018.01.042.

lP

[129] Humrich JY, Morbach H, Undeutsch R, Enghard P, Rosenberger S, Weigert O, et al.

lupus.

Proceedings

of

the

National

Academy

of

Sciences

2010;

107:204-09.

Jo ur

10.1073/pnas.0903158107.

na

Homeostatic imbalance of regulatory and effector T cells due to IL-2 deprivation amplifies murine

[130] Johnson MC, Garland AL, Nicolson SC, Li C, Samulski RJ, Wang B, et al. beta-cell-specific IL-2 therapy increases islet Foxp3+ Treg and suppresses type 1 diabetes in NOD mice. Diabetes 2013; 62:3775-84. 10.2337/db13-0669. [131] Rosenzwajg M, Churlaud G, Mallone R, Six A, Dérian N, Chaara W, et al. Low-dose interleukin-2 fosters a dose-dependent regulatory T cell tuned milieu in T1D patients. J Autoimmun 2015; 58:48-58. 10.1016/j.jaut.2015.01.001. [132] Hartemann A, Bensimon G, Payan CA, Jacqueminet S, Bourron O, Nicolas N, et al. Low-dose interleukin 2 in patients with type 1 diabetes: a phase 1/2 randomised, double-blind,

34

Journal Pre-proof

placebo-controlled

trial.

Lancet

Diabetes

Endocrinol

2013;

1:295-305.

10.1016/S2213-8587(13)70113-X. [133] Todd JA, Evangelou M, Cutler AJ, Pekalski ML, Walker NM, Stevens HE, et al. Regulatory T Cell Responses in Participants with Type 1 Diabetes after a Single Dose of Interleukin-2: A Non-Randomised, Open Label, Adaptive Dose-Finding Trial. Plos Med 2016; 13:e1002139.

of

10.1371/journal.pmed.1002139.

ro

[134] Koreth J, Kim HT, Jones KT, Lange PB, Reynolds CG, Chammas MJ, et al. Efficacy, durability, and response predictors of low-dose interleukin-2 therapy for chronic graft-versus-host

-p

disease. Blood 2016; 128:130-37. 10.1182/blood-2016-02-702852.

re

[135] Kennedy-Nasser AA, Ku S, Castillo-Caro P, Hazrat Y, Wu MF, Liu H, et al. Ultra

lP

Low-Dose IL-2 for GVHD Prophylaxis after Allogeneic Hematopoietic Stem Cell Transplantation

na

Mediates Expansion of Regulatory T Cells without Diminishing Antiviral and Antileukemic Activity. Clin Cancer Res 2014; 20:2215-25. 10.1158/1078-0432.CCR-13-3205.

Jo ur

[136] Hirakawa M, Matos TR, Liu H, Koreth J, Kim HT, Paul NE, et al. Low-dose IL-2 selectively activates subsets of CD4+ Tregs and NK cells. JCI Insight 2016; 1:e89278. 10.1172/jci.insight.89278.

[137] von Spee-Mayer C, Siegert E, Abdirama D, Rose A, Klaus A, Alexander T, et al. Low-dose interleukin-2 selectively corrects regulatory T cell defects in patients with systemic lupus erythematosus. Ann Rheum Dis 2016; 75:1407-15. 10.1136/annrheumdis-2015-207776. [138] Humrich JY, von Spee-Mayer C, Siegert E, Alexander T, Hiepe F, Radbruch A, et al. Rapid induction of clinical remission by low-dose interleukin-2 in a patient with refractory SLE. Ann Rheum Dis 2015; 74:791-92. 10.1136/annrheumdis-2014-206506.

35

Journal Pre-proof

[139] He J, Zhang X, Wei Y, Sun X, Chen Y, Deng J, et al. Low-dose interleukin-2 treatment selectively modulates CD4+ T cell subsets in patients with systemic lupus erythematosus. Nat Med 2016; 22:991-93. 10.1038/nm.4148. [140] Miao M, Hao Z, Guo Y, Zhang X, Zhang S, Luo J, et al. Short-term and low-dose IL-2 therapy restores the Th17/Treg balance in the peripheral blood of patients with primary Sjogren's

of

syndrome. Ann Rheum Dis 2018; 77:1838-40. 10.1136/annrheumdis-2018-213036.

ro

[141] Rosenzwajg M, Lorenzon R, Cacoub P, Pham HP, Pitoiset F, El Soufi K, et al. Immunological and clinical effects of low-dose interleukin-2 across 11 autoimmune diseases in a

-p

single, open clinical trial. Ann Rheum Dis 2019; 78:209-17. 10.1136/annrheumdis-2018-214229.

cells

by a

mechanism.

Nat

Med

2018;

24:1005-14.

na

10.1038/s41591-018-0070-2.

structure-based

lP

regulatory T

re

[142] E T, PH B, SL S, LK E, KM J, Le DT, et al. A human anti-IL-2 antibody that potentiates

[143] SY L, ML C, HJ O, JG R, MJ P, JY J, et al. Interleukin-2/anti-interleukin-2 monoclonal

Jo ur

antibody immune complex suppresses collagen-induced arthritis in mice by fortifying interleukin-2/STAT5 signalling pathways. Immunology 2012; 137:305-16. 10.1111/imm.12008. [144] JJ Y, JG L, JY J, TY K, C A, J Y. IL-2/anti-IL-2 complexes ameliorate lupus nephritis by expansion

of

CD4+CD25+Foxp3+

regulatory

T cells.

Kidney

Int

2017;

91:603-15.

10.1016/j.kint.2016.09.022. [145] H W, L H, D K, J F, X X, A C, et al. Increasing Regulatory T Cells With Interleukin-2 and Interleukin-2 Antibody Complexes Attenuates Lung Inflammation and Heart Failure Progression. Hypertension (Dallas, Tex. : 1979) 2016; 68:114-22. 10.1161/HYPERTENSIONAHA.116.07084. [146] GM D, TP K, Y Z, PE Z, KL M, B X, et al. The mTOR kinase differentially regulates

36

Journal Pre-proof

effector

and

regulatory

T

cell

lineage

commitment.

Immunity

2009;

30:832-44.

10.1016/j.immuni.2009.04.014. [147] T S, van den Brandt C, B F, S Z, F K, A D, et al. Autoimmune pancreatitis in MRL/Mp mice is a T cell-mediated disease responsive to cyclosporine A and rapamycin treatment. Gut 2014; 63:494-505. 10.1136/gutjnl-2012-303635.

of

[148] JN M, CH W. Combination Therapy Using IL-2/IL-2 Monoclonal Antibody Complexes,

ro

Rapamycin, and Islet Autoantigen Peptides Increases Regulatory T Cell Frequency and Protects against Spontaneous and Induced Type 1 Diabetes in Nonobese Diabetic Mice. Journal of

-p

immunology (Baltimore, Md. : 1950) 2015; 195:5203-14. 10.4049/jimmunol.1402540.

re

[149] Z G, W T, J J, G F, Y M, Z D, et al. Rapamycin reverses the senescent phenotype and

lP

improves immunoregulation of mesenchymal stem cells from MRL/lpr mice and systemic lupus

na

erythematosus patients through inhibition of the mTOR signaling pathway. Aging 2016; 8:1102-14. 10.18632/aging.100925.

Jo ur

[150] ZW L, R K, T W, I M, A S, J Y, et al. Sirolimus in patients with clinically active systemic lupus erythematosus resistant to, or intolerant of, conventional medications: a single-arm, open-label,

phase

1/2

trial.

Lancet

(London,

England)

2018;

391:1186-96.

10.1016/S0140-6736(18)30485-9. [151] CL B, J C, F R, ME B, PJ F, L W, et al. The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3. Nat Genet 2001; 27:20-21. 10.1038/83713. [152] JH P, KH L, B J, HD O, JS L, HY G, et al. Immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome: A systematic review. Autoimmun Rev 2020; 19:102526.

37

Journal Pre-proof

10.1016/j.autrev.2020.102526. [153] L P, F B, R C, C S, G B, F T, et al. Treatment with rapamycin can restore regulatory T-cell function in IPEX patients. The Journal of allergy and clinical immunology 2020; 145:1262-71. 10.1016/j.jaci.2019.11.043. [154] KJ F, RW J. Histone deacetylases and their inhibitors in cancer, neurological diseases and

of

immune disorders. Nature reviews. Drug discovery 2014; 13:673-91. 10.1038/nrd4360.

ro

[155] J H, L W, S D, UH B, R H, A S, et al. Histone/protein deacetylase 11 targeting promotes Foxp3+ Treg function. Sci Rep-Uk 2017; 7:8626. 10.1038/s41598-017-09211-3.

-p

[156] SW C, T B, L C, JL F, A P, JM M, et al. Vorinostat plus tacrolimus and mycophenolate to

re

prevent graft-versus-host disease after related-donor reduced-intensity conditioning allogeneic

na

10.1016/S1470-2045(13)70512-6.

lP

haemopoietic stem-cell transplantation: a phase 1/2 trial. The Lancet. Oncology 2014; 15:87-95.

[157] BR O, DH S, D B, N H, YI C, HJ Y, et al. Therapeutic effect of a novel histone deacetylase

Jo ur

6 inhibitor, CKD-L, on collagen-induced arthritis in vivo and regulatory T cells in rheumatoid arthritis in vitro. Arthritis Res Ther 2017; 19:154. 10.1186/s13075-017-1357-2. [158] DS K, HK M, EK K, SC Y, HS N, SY L, et al. Suberoylanilide Hydroxamic Acid Attenuates Autoimmune Arthritis by Suppressing Th17 Cells through NR1D1 Inhibition. Mediat Inflamm 2019; 2019:5648987. 10.1155/2019/5648987.

38

re

-p

ro

of

Journal Pre-proof

lP

Figure. Treg-targeted therapy Treg cells used for adoptive cell therapy usually come from autologous peripheral blood or umbilical cord blood. In order to obtain clinically meaningful Treg

na

numbers, Treg cells need to be expanded ex vivo or induced in vitro with artificial

Jo ur

antigen-presenting cells or Treg inducers (such as rapamycin, retinoic acid, transforming growth factor-β, DNA methyltransferase and histone deacetylase inhibitors) before transfusion. Artificial antigen-specific Treg cells are generated by introducing synthetic receptors (CARs or engineered TCR). Treg-targeted drugs, such as rapamycin and HDACs inhibitors, promote Treg survival and function by targeting specific signal pathways which are of crucial importance for Treg development. Low-dose IL-2 modulation is another well-studied treatment which could selectively promote the development and maintenance of Treg cells without activation of Teff cells as well as correct the imbalance of Treg and Teff cells. aAPC, artificial antigen-presenting cells; CAR, chimeric antigen receptor; HDACs, Histone deacetylases; RA, retinoic acid; TCR, T cell receptor; UCB, umbilical cord blood. 39

Journal Pre-proof

Outline Part 1:This part is a brief introduction of the function and subsets of regulatory T (Treg) cells, including some recent advances in this area. Part 2:This part mainly elaborates non-lymphoid tissue subsets of Treg cells, including visceral adipose tissue Treg (VAT Treg) cells, skeletal muscle Treg cells, intratumoral Treg cells and Treg

of

cells in other organs. The characteristics and special tissue-related functions of different tissue

ro

Treg subsets as well as their status in different diseases are also been expounded.

-p

Part 3:This part is mainly about Treg-targeted therapy, including Treg-based cellular transfer,

Jo ur

na

lP

re

IL-2 therapy and Treg-targeted drugs in autoimmune diseases.

40