Accepted Manuscript Title: Sniping the Scout: Targeting the key molecules in dendritic cell functions for treatment of autoimmune diseases Author: Xing Li Yanping Han Erwei Sun PII: DOI: Reference:

S1043-6618(16)30096-2 http://dx.doi.org/doi:10.1016/j.phrs.2016.02.023 YPHRS 3083

To appear in:

Pharmacological Research

Received date: Revised date: Accepted date:

5-2-2016 23-2-2016 23-2-2016

Please cite this article as: Li Xing, Han Yanping, Sun Erwei.Sniping the Scout: Targeting the key molecules in dendritic cell functions for treatment of autoimmune diseases.Pharmacological Research http://dx.doi.org/10.1016/j.phrs.2016.02.023 This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. 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.

Sniping the Scout: Targeting the key molecules in dendritic cell functions for treatment of autoimmune diseases

Authors: Xing Lia,b, Yanping Hanc and Erwei Suna,b,*

a Department of Rheumatology and Immunology, The Third Affiliated Hospital of Southern Medical University, Guangzhou, China. b Institute of Clinical Immunology, Academy of Orthopedics·Guangdong Province, China. c Hospital of South China Normal University, Guangzhou, China.

* Address correspondence to Erwei Sun M.D., Ph.D.: Department of Rheumatology and Immunology, The Third Affiliated Hospital of Southern Medical University, No. 183, Zhongshan Avenue West, Tianhe District, Guangzhou 510630, China. E-mail: [email protected], Tel:+86-20-682784421.

Abstract Dendritic cells (DCs) are a power tool for manipulating immune system. They play important roles in the induction of immunity as well as inducing intrathymic and peripheral tolerance. After generated from stem cells in the bone marrow, DCs traffic to the peripheral tissues, where they capture and process antigens, express lymphocyte co-stimulators, migrate to the secondary lymph organs and present the processed antigen to naive T cells to either activate or tolerize them. These processes are modulated subtly and influenced by various factors. Aberrant regulation of the processes may cause autoimmunity. Investigation into the biology of DCs and the molecules and mechanisms that regulate them helps us understanding the pathogenesis of autoimmune diseases and reveals numerous steps for pharmacological manipulation. In this review, we made a sketch line of the critical events of DC biology that are potential pharmacologic targets for the treatment of autoimmune diseases. Keywords: Autoimmune diseases; Dendritic cells; T helper ; Rheumatoid arthritis; Systemic lupus erythematosus; Chemical compounds studied in this article Dexamethasone (PubChem CID: 5743); Vitamin D3 (PubChem CID: 5280795); rapamycin (PubChem CID: 5284616); Aspirin (PubChem CID: 2244); N-acetyl-L-cysteine (PubChem CID: 12035); mycophenolate mofetil (PubChem CID: 5281078); Butyric acid (PubChem CID: 264); rosiglitazone (PubChem CID: 77999); Sanglifehrin A (PubChem CID: 5388925); Cyclosporin A (PubChem CID: 5284373); Tacrolimus (PubChem CID: 445643); FTY720 (PubChem CID: 107969); Chloroquine (PubChem CID: 2719); Troglitazone (PubChem CID: 5591); Leflunomide (PubChem CID: 3899); Triptolide (PubChem CID: 107985); Apigenin (PubChem CID: 5280443); Quercetin (PubChem CID: 5280343); Chrysin (PubChem CID: 5281607); Fumarates (PubChem CID: 5281069); Auranofin (PubChem CID: 24199313); Glatiramer acetate (PubChem CID: 3081884); Trichostatin A (PubChem CID: 444732); Sunitinib (PubChem CID: 5329102); Oenothein B (PubChem CID: 16132398); Tofacitinib (PubChem CID: 9926791); Fostamatinib (PubChem CID: 11671467); Vasoactive intestinal peptide (PubChem CID: 16129679); TGF-β (PubChem CID: 56842206); IL-10 (PubChem CID: 146070).

1. Introduction The immune system provides protection against infection and maintains immune homeostasis. Dendritic cells (DCs) play a central role in the induction of immunity as well as inducing intrathymic and peripheral tolerance. Clinical autoimmunity arises as a result of aberrant regulation of the immune responses [1]. The development of autoimmune diseases, such as rheumatoid arthritis (RA), systemic lupus erythematosus(SLE), requires three different but related processes: disruption of immune tolerance, development of chronic inflammation in one or several organs, and tissue destruction and their harmful effects [2]. Classical treatment for autoimmune diseases is usually a combination of anti-inflammatory, immunosuppressive drugs and biological agents that are designed to block the activity of pro-inflammatory cytokines and lymphocytes as the primary cellular targets. However, the strategy is not so effective in controlling symptoms and disease progression. Accumulating evidence indicates that the central role of DCs in immunity and tolerance is an indispensable component in the pathogenesis of autoimmune diseases [3]. Therefore, targeting DC functions maybe a potential and valuable therapy for the control of autoimmune diseases. DCs

represent

a

heterogeneous

population

of

uniquely

well-equipped

antigen-presenting cells (APCs) with distinct developmental origins, phenotype markers and immunological functions. After generated from stem cells in the bone marrow, DCs are generally immature and traffic to the peripheral tissues, where they undergo several molecular events (namely antigen uptake, phenotypic and functional maturation, migration to the secondary lymphoid organs and present acquired antigens to naive T lymphocytes). Then, they instruct naive T cells to differentiate toward T helper (Th) -1(Th1), Th2, Th17 or regulatory T cell (Treg) cells as well as to promote maturation of antibody-producing B cells [4, 5]. These processes enable DCs to behave as critical sentinels to eliminate invading pathogens, to induce and regulate of most adaptive immune responses and to maintain tolerance and immune homeostasis [6-9]. Disregulation of these processes may cause imbalance of immune responses and lead to autoimmunity. Animal studies indicate that DCs play important

roles in the initiation and perpetuation of autoimmune diseases [10]. In this review, we present a description of key molecules and mechanisms that regulate DC differentiation, endocytosis, maturation and migration, especially focusing on their significances or potentials as pharmacological drugs or agents in anti-inflammation or immunosuppression for autoimmune diseases.

2. Molecular control of DC biology in autoimmunity 2.1 DC differentiation in autoimmunity After several decades of research, we have known that DCs stem from a hematopoietic lineage distinct from other leukocytes, establishing the DC system as an unique hematopoietic branch [10, 11]. Accumulated evidence suggests different DC lineages play different roles in the maintenance of immune homeostasis via induction of immune tolerance and regulation. Under the mediation of some factors, DC precursors can be instructed to differentiate into immunogenic DCs and tolerogenic DCs. Immunogenic DCs have the ability of up-regulation the expression of co-stimulatory molecules (CD40, CD80 and CD86), MHC class II molecules and proinflammatory cytokines such as interleukin-12 (IL-12), tumor necrosis factor (TNF-α) and IL-6, and presenting antigens to specific T cells to elicit adaptive immune responses. Conversely, although tolerogenic DCs retain the ability of presenting antigens to specific T cells, they downregulate the expression of co-stimulatory molecules and proinflammatory cytokines, upregulate the expression of inhibitory molecules and anti-inflammatory cytokines and are resistant to maturation-inducing signals. The process is crucial for maintenance of immune homeostasis and control of autoimmune disorders. Hence, molecular control of DC differentiation maybe the pharmacological potential targets for the control of autoimmune diseases. Previous reviews have reported that pharmacological effects on DC differentiation and expansion can be achieved by immunomodulatory drugs, such as corticosteroids [12, 13], vitamin D3 [14-16], or corticosteroids plus vitamin D3 [13, 17-22], rapamycin [23-27], aspirin [28-30], N-acetyl-L-cysteine [31], mycophenolate mofetil [32, 33], butyric acid [34-36], rosiglitazone [37, 38], rabeximod [39] , n-3

fatty acid [40] and serial chinese herbal medicine [41]. These agents that have been widely used in the treatment of autoimmune diseases are previously reported to block DC differentiation quantitatively by antagonizing the effects of several key molecules, such as hematopoietic cytokines. Generally different hematopoietic molecules control the differentiation and expansion of bone-marrow(BM) cells toward DC specific lineage and maintain DC homeostasis in the periphery [10]. Here, we will present key hematopoietic cytokines that control DC differentiation and their significance in the treatment of autoimmune diseases.

FMS-like tyrosine kinase-3 Ligand (Flt3L) is a key mediators for DC generation [42]. Loss of Flt3 expression in hematopoietic progenitors correlates with the loss of DC differentiation potential [43, 44]. Treatment with Flt3L tyrosine kinase inhibitors or genetic deletion of Flt3L gene in mice leads to 10-fold decrease of pDCs and DCs in lymphoid organs [45], whereas enforcement of Flt3L expression in progenitors that lack the ability to develop into DC restores some DC developmental potential [46]. Prior studies showed that Flt3L was associated with autoimmune pathogenesis. In rheumatoid arthritis, Flt3L is increased in synovial fluid and correlated with active lesions [47]. In collagen-induced arthritis, Flt3L deficiency mice showed a marked increase in the scores and incidence of arthritis in both acute and chronic phases of CIA model mice as compared with wild type mice [48]. A Flt3-Flt3L pathway inhibitor sunitinib can reduce the intensity of synovitis and the incidence of bone destruction in bovine serum albumin (mBSA)-induced arthritis, which was achieved partly by the inhibition on DC differentiation [49]. Whereas increasing the number of DCs by Flt3L injection leads to an increase in the number of regulatory T cells, which contributes to the protection of mice from developing type 1diabetes(T1D) [50]. Due to the dual effects of Flt3L in both promoting and suppressing autoimmunity, the effects of Flt3L on DC precursors at their different developmental stages during the progress of autoimmune diseases need further investigation.

Granulocyte-macrophage colony stimulating factor(GM-CSF), a hematopoietic growth factor that regulates the differentiation of the myeloid lineage genitor cells, plays a critical role in the differentiation of mouse and human hematopoietic progenitors and monocytes into DCs. Accumulated evidence has shown that GM-CSF affects DCs on multiple levels during the pathogenesis of autoimmune diseases [51-54]. Under the influence of elevated GM-CSF, circulating inflammatory monocytes develops into inflammatory DCs that can present self-antigens to T cells and elicit autoimmune responses [55, 56]. Moreover, after antigen uptake and stimulation by the GM-CSF, DCs acquire enhanced life span and presentation ability to active T cells in the lymph nodes and cause autoimmunity [55]. Prior experiments in vitro showed that the GM-CSF stimulated monocyte-derived DC (moDC) maintained

their

inflammatory

property

and

increased

the

secretion

of

pro-inflammatory cytokines TNF-α and IL-6 [56, 57]. And GM-CSF could stimulate immature myeloid cells to differentiate into DCs in the central nervous system (CNS) and exacerbate experimental autoimmune encephalomyelitis (EAE) [58, 59]. Whereas apart from its key roles in the initiation and progression of DC-mediated autoimmunity, GM-CSF also suppresses the immune responses via its effect on DCs. Previous studies have reported that low-dose GM-CSF prevents the development of various autoimmune diseases in mice, through induction of tolerogenic CD8- myeloid DCs from bone marrow precursors and expansion of Foxp3+Tregs [60, 61]. Due to crucial role of GM-CSF in the differentiation and expansion of DCs, currently three anti-GM-CSF antibodies (CAM-3001, MOR103 and KB003) are tested for the pre-clinical treatment of diseases such as multiple sclerosis(MS), RA and asthma. The results showed rapid and significant beneficial effects on autoimmune diseases with no or less unexpected safety concerns [62-64].

Macrophage colony-stimulating factor (M-CSF), also named CSF-1, is a hematopoietic cytokine that mediates the proliferation and differentiation of macrophages [65]. And, M-CSF binds specifically to its receptor(CSF-1R) and partially regulates the differentiation or survival of nonlymphoid DCs [66, 67].

Epidermal langerhans cell (LCs) are totally absent in Csf-1R deficiency mice [68]. In the presence of M-CSF, circulating inflammatory monocytes develop into tolerogenic DCs that maintain the ability of antigen presentation, whereas downregulate expression of co-stimulatory molecules and proinflammatory cytokines [69]. In addition, injection of M-CSF increases both lymphoid organ pDCs and DCs in mice [70, 71]. Taken together, given their crucial roles in DC and autoimmunity, hematopoietic cytokines(e.g Flt3L, GM-CSF, M-CSF) can serve as potential targets to affect DC genesis as well as functions to prevent the development of autoimmune disease.

2.2 DC endocytosis in autoimmunity Haematopoietic stem cells differentiate into immature DCs that are recruited to peripheral tissues, where they continuously take up and process antigens by different endosomal

pathways

[11].

Previous

studies

have

reported

that

several

immunosuppressive mediators potently manipulate the function of DC endocytosis via distinct mechanisms. Some agents, like corticosteroids [12], vitamin D3 [14, 15] ,aspirin [34] and fumarates [72] inhibit phenotypic and functional DC maturation, and

consequently

enhance

endocytic

capacity.

Whereas

some

other

immunosuppressors suppress DC endocytosis by down-regulating the expression of DC endocytosis receptors in a DC-maturation-independent manner. The main agents are rapamycin, sanglifehrin, cyclosporine A (CsA) [34, 73, 74] and quercetin [75]. In the steady state, immature DCs selectively express plenty of endocytosis receptors, such as Fcγ receptors(FcγRs), mannose receptor(MRs), and heat shock protein receptors, which could be used to target antigens for processing and presentation in vivo [8].  

Fcγ receptors(FcγRs), a family of endocytotic receptors for the Fc portion of immunoglobulin, confer the protective effects of the immune system by recognition of IgG bound pathogens and play a critical role in autoimmune diseases [76]. Both human and mouse immature DCs express several FcγRs, and the activating

FcγRs(FcγR and FcγRⅢ) enhanc DC activation via immunoreceptor tyrosine-based activation motif (ITAM) ,while the inhibitory FcR (FcγR ) regulates DC function to promote Treg induction but inhibit effector T cell responses via immunoreceptor tyrosine-based inhibitory motif (ITIM) [77, 78]. Prior studies suggest that activating FcγR(FcγR and FcγRⅢ) deficiency protects from autoimmune diseases, such as arthritis and lupus [79]. TG19320, a tetrameric peptide interfere with IgG/FcγR interaction, attenuates kidney damage and prolongs the survival rate of lupus-prone MRL/lpr mice [80]. Moreover, mouse anti-human FcγRⅢ mAb 3G8 has been used to successfully treat immune thrombocytopenia (ITP) [81, 82]. Conversely, inhibitory FcR(FcγR Ⅱ ) suppresses the expression of proinflammatory cytokines of DCs stimulated by toll-like receptor (TLR) 4 via lipid kinase phosphoinositied 3-OH kinase(PI3K)/AKT signaling pathway [83]. And FcγR Ⅱ -overexpressing DCs dramatically prevents glomerulonephritis and enhances the survival rate of lupus-prone MRL/lpr mice [84]. Additionally, FcγRⅡspecific small chemical entities suppress arthritis development in a collagen-induced arthritis(CIA) model, an suppressive effect even stronger than methotrexate(MTX), a classical drug for RA in human [85]. Currently, FcγRs are appealing targets in the treatment of inflammatory autoimmune diseases. Targeting approaches include blocking activating FcγR and activating inhibitory Fc γ RⅡ.

DEC205 is an endocytotic receptor and highly expressed on several subsets of DCs [86]. Accumulating evidence suggests that DEC205 plays an important role for the induction of immunity and tolerance [87]. Prior studies have suggested that selective delivery of antigens to DEC205 on steady-state DCs confers a powerful method to induce antigen-specific T cell tolerance [88]. And recombinant anti-DEC205 Abs fused to disease-relevant autoantigens have been developed and used successfully for tolerogenic protection from autoimmunity in mouse models of autoimmune diabetes, encephalomyelitis and arthritis [89-91].

Dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN) is a typeⅡC-type lectin receptor that is abundantly expressed on DCs, especially immature DCs. Apart from being an adhesion molecule, DC-SIGN has emerged as a key player in the regulation of immune responses by recognition with several endogenous and exogenous antigens [92, 93]. As DC-specific phenotypic marker, DC-SIGN commonly recognizes intestinal commensal, which promotes DC induced generation of Tregs and prevents the development of T cell-dependent colitis [94]. Hence, DC-SIGN maybe a potential and valuable target on DC endocytosis in autoimmunity.

2.3 DC maturation in autoimmunity DCs are specialized sentinels responsible for coordinating innate and adaptive immunity. DC function is dependent upon their sensitivity to environmental inflammatory signs that promotes cellular maturation, phenotype alteration, activation and migration. We now appreciate that various molecules, signals, and mechanisms control the maturation of steady-state DC. Corruption of these steady-state operatives has diverse immunological consequences and pinpoints DCs as potent therapy targets of autoimmune diseases [95]. 2.3.1 Pattern recognition receptors Pattern-recognition receptors (PRRs) consist of Toll-like receptors (TLRs), C-type lectin receptors (CLRs), nucleotide oligomerization domain (NOD)-like receptors (NLRs), and retinoic acid–inducible gene (RIG)-I-like receptors (RLRs) [95, 96]. Although best known as probes employed by DCs to sense pathogens and other environmental stimuli, many PRRs also transduce potent DC maturation signals and play a critical roles in autoimmunity.

Toll-Like Receptors(TLRs) are ligand-dependent detection modules that facilitate key aspects of innate and adaptive immunity. TLR signals within DCs are critical for induction of immune responses, with some exceptions given the nature of the

immunogens. Multiple lines of evidence indicate that activating TLR signals induce DC phenotypic and functional maturation and systemic autoimmunity [97]. Psoriasis in human patients is associated with upregulation of host molecules that enhance nucleic acid sensing by TLRs of plasmacytoid dendritic cells (pDCs) [98]. Duplication of the TLR7 gene causes DC expansion, maturation, and systemic autoimmunity. TLR9 over expression to the cell surface of hematopoietic stem cells (HSCs) results in CD11c+ DC expansion and activation, which induces the secretion of multiple pro-inflammatory cytokines (TNF-α, IL-1β) and fatal inflammation in irradiated mouse recipients. Anti-malarial drugs chloroquine and hydroxychloroquine that have been widely used in treatment of autoimmune diseases block DC activation through TLR7 and TLR9 [99, 100]. Myeloid differentiation factor 88(MyD88), the common TLR adaptor, mediates activation of peripheral myeloid dendritic cells and directs differentiation of T cells toward autoimmune Th17 cells [101]. MyD88 deficiency protects MRL.Faslpr mice from systemic lupus erythematosus (SLE), which is associated with down-regulation of expansion, activation, and cytokine secretion by DCs [102]. However, not all TLR signals induce DC maturation and autoimmune responses. Certain microbial products (e.g yeast zymosan, yersinia pestis, schistosomamansoni, bordetella pertussis) promote unexpected tolerogenic responses in DCs by binding TLR2, TLR4 or TLR6 [103]. The mechanisms that commensal sampling by DCs confers protection from autoimmune diseases and the intracellular signaling cascades that drive those tolerogenic DC functions require further investigation. Therefore, TLRs and their agonists maybe another potential targets for the treatment of autoimmune diseases.

C-Type Lectin Receptors (CLRs) are a variety family of transmembrane molecules containing the C-type lectin protein domain that enables binding of Ca2+ or carbohydrate ligands of commensal sampling. In most cases, CLRs down-regulate DC functions. For example, BDCA-2-Syk signals in human pDCs restrict interferon (IFN)-α

production

[104,

inhibition motif (ITIM)-containing

105]. CLRs

Immunoreceptor tyrosine-based that

activate

Src-homology

phosphatase(SHP) phosphatases can transduce negative maturation signaling in DCs. SHP-1 deficiency specifically up-regulates the expression of co-stimulatory molecules and MHC class

on DCs and leads to autoimmunity [106, 107]. Dendritic cell

immunoreceptor 1(DCIR1) is the only CLR whose genetic deletion in mice elicits spontaneous

autoimmune

disease

and

break

of

immune

homeostasis.

DCIR1-deficient mice develop IgM rheumatoid factor and antinuclear antibodies by 4-6 months of age, and arthritis develops in 30% of mice. In monocyte-derived DCs deprived from human, DCIR inhibits TLR-induced cytokine production [108, 109]. Those potential roles of CLR ligands in DCs may constitute an exciting prospect that may yield disease modulatory treatments with therapeutic value in autoimmune diseases.

Nucleotide oligomerization domain-like Receptors(NLRs) NLRs are cytoplasmic receptors that use their caspase recruitment domains to activate NF-κB or inflammasome to control the production of IL-1β and IL-18 [110, 111]. Nucleotide oligomerization domain (Nod) 2, which recognizes bacterial peptidoglycans, regulates some aspects of DC biology [112]. In graft versus host disease (GVHD), Nod2 knockout-DCs show enhanced phenotypic and functional maturation [113]. And Nod2 deficiency in mice displays gain-of-function and increased inflammatory cytokine secretion in DCs that causes increased susceptibility to DSS colitis [114]. These findings indicate a potential value for down-regulated DC Nod2 signals in the pathogenesis of autoimmune disease. Collectively, those potential mechanisms add intrigue to our understanding of the identity of PRRs. As the steady-state functions of those receptors may confer protection from autoimmune diseases, further pharmalogical development may constitute an exciting future with therapeutic value [115]. 2.3.2 Transcription factors Transcription factors, such as nuclear factor-kappa B (NF-κB), class O of forkhead box transcription factor(Foxo)3 and T-bet, potently manipulate DC phenotypic and functional maturation via upstream activating signals, and also play

critical role in autoimmunity. NF-κB is a signal transduction pathway molecule crucially involved in the inflammatory responses. Previous study reported that a common feature of drugs targeting DCs was their capacity to inhibit NF-κB, such as corticosteroids [12], CsA [73, 74], vitamin D3 analogs [35]. The NF-κB family member RelB is required for myeloid DC differentiation, and controls DC function via regulation of CD40 and MHC class II molecule expression [35]. Whereas another NF-κB family member NF-κB1, crucial for maintaining the resting state of DCs, restricts DC production of TNF-α [116, 117]. NF-κB inhibitor Bay11-7082 suppresses both CD40 and MHCⅡ expression and cytokine production on NiSO4 stimulated DCs [118, 119]. Bay11-7082 treated DCs prevent the development of collagen-induce arthritis (CIA) and inhibit CII specific T lymphocyte proliferation and antibody production [120]. Currently, a phase I clinical trial with tolerogenic DCs generated with Bay11-7082 is carried out in patients with RA. It has been found that DCs modified by Bay11-7082 were well tolerated and no major adverse effects were observed in this trial [121]. FOXO3, a member of Foxo family, was previously reported to play critical roles in

DC

function

through

a

“reverse

signaling”

process

mediated

cytotoxic T lymphocyte-associated antigen-4

(CTLA-4)

interaction

with

by B7

molecules resulting in increased indoleamine 2,3-dioxygenase (IDO) levels [95]. And siRNA-silenced Foxo3 expression leads to a decrease of DC tolerogenicity and expression of suppressive factors transforming growth factor (TGF)-β, and a concomitant increase in expression of CD80 and IL-6 [122]. And Foxo3 deficiency in steady state leads to 2-fold cellular expansion of splenic DCs and modest up-regulation of CD80 and CD86 in DCs [123]. Also Foxo3 restricts DC production of IL-6 and to a lesser extent of TNF-α and CCL2 [124]. In autoimmunity, prior study reported that Foxo3 activity was a predictive indicator of disease severity in Crohn's disease (CD) and rheumatoid arthritis [125]. And genetic manipulation in Foxo3 led to milder CD and lesser inflammatory responses, and was also associated with a milder course of RA but with increased susceptibility to more severe malaria [126]. In summary, those data suggest that Foxo3 may be part of the regulatory mechanisms

that program DC maturation and immune responses and can be a useful target in immune based therapies. T-bet is a transcription factor mostly noted for its role in T effector cell differentiation, but its role in DCs seems negative. The effect of T-bet on DC immunobiology depends mainly on the relationship between T-bet-mediated suppression of DC-produced TNF-α and TNF-α-dependent progression of colitis. T-bet-deficiency in DCs confers TNF-α over-express and triggers spontaneous ulcerative colitis with progressive development to colorectal cancer. And patients suffering inflammatory bowel disease are often successfully treated with anti-TNF-α therapy [127-129]. 2.3.3 Immune mediators DC maturation encompasses the down regulation of endocytic capacity, the up-regulation of surface T-cell co-stimulatory (CD40, CD80 and CD86) and MHC class II molecules, the production of bioactive pro-inflammatory cyokines such as IL-12, TNF-α and IL-6 and the inhibition of anti-inflammatory cyokines such as IL-10 and TGF-β. These immune mediators control steady-state DC maturation and disruption of these factors leads to diverse immunological consequences and pinpoints DCs as potent drivers of autoimmune disease. Thus, targeting on these immune mediators may result in therapeutic values in autoimmune diseases. Most anti-inflammatory and immunosuppressive mediators suppressed immune responses are coupled with inhibition in DC phenotypic and functional maturation. These mediators include immunosuppressive drugs such as corticosteroids [12, 13], vitamin D3 [14-17], rapamycin [23-27], CsA [73, 74], tacrolimus [130], aspirin [28-30], mycophenolate [32, 33], sanglifehrin A [131], leflunomide [132], intravenous immunoglobulin(IVIG) [133] rosiglitazone [37, 38] and troglitazone [18]; the mucolytic antioxidant N-acetyl-L-cysteine [31]; anti-malarial drugs such as chloroquine and hydroxychloroquine [134, 135]; pro-inflammatory factor blockers such as TNF-α blocker (adalimumab [136], infliximab [137]), and IL-6 blocker(tocilizumab [138]); anti-inflammatory factors such as IL-10 [139-144],TGF-β [145-148]; vascular endothelial growth factor (VEGF), PGE2, retinoids, HLAG,

flavonoid  [9, 35, 103, 149-151], and several Chinese herbal medicine and their components [41, 72, 132] [152](for detail seen Tab.1)  Co-stimulatory and MHC class II molecules

DCs have a dual ability to either

activate or suppress immunity, which is mainly associated with the expression of co-stimulatory and MHC class II molecules [153]. In addition to chemical or tissue culture induced tolerogenic DCs, genetic engineering methods can also be used for the creation of tolerogenic DCs. Recent studies indicate that gene silencing of CD40, CD80,and CD86 through administering short interfering RNA (siRNA) achieves optimal protection from disease and induced antigen-specific Tregs in CIA mice [154, 155]. Apart from co-stimulatory molecules, MHC class II molecules are indispensable for DC immune biology. In the steady state, MHC class II molecules connect with a single lysine by ubiquitin chains. And once DC activated, deubiquitination will happen in MHC class II molecules, which leads to accumulation of MHC class II on DC cell surface [155-158]. Thus, blocking the deubiquitination and maintaining ubiquitination of MHC class II will be a potential target on preventing immune responses in autoimmune diseases. The ubiquitin-conjugating molecule MARCH1 is primarily expressed in immature DCs, and has the effect to down regulate DC activation. MARCH1-deficient DCs express 10- fold higher levels of MHC class II molecules and CD86, but with significantly reduced cytokine responses to LPS and CD40 ligand and diminished capacity to present MHC class II restricted antigens [156, 159]. The mechanisms and significances of the contradictory effects of MARCH1-deficient DCs require further investigated. Collectively, those data offer a new insight in targeting DC phenotypic maturation to manipulate the treatment of autoimmunity. Cyokines. Once activated, DCs undergo functional maturation and produce multiple cytokines, including pro-inflammatory cytokines such as TNF-α, IL-6, IL-12 and IL-1β, and anti -inflammatory cytokines such as IL-10, TGF-β [1, 160]. Those cytokines play critical roles in autoimmune responses. As the products of DCs, they also induce the differentiation of immature DCs into mature DCs that can instruct naive T cells differentiating toward Th1, Th2, Th17 or Treg cells [160]. Due to their

potential on regulating the balance between immunity and tolerance, the bioavailability of those cytokines and their effect on DC functions have been implicated in inflammatory and autoimmune diseases. Currently, biological agents targeting on cytokines TNF-α, IL-6 and IL-1 are widely used in autoimmune diseases, such as RA, Crohn’s disease, psoriatic arthritis (PsA), and ankylosing spondylitis(AS) [161-167]. This represents a success of immunology in clinical application and the coming years will witness the expanding benefit of cytokines as therapeutic targets in autoinflammatory and autoimmune pathology. Apart from pro-inflammatory cytokines, anti-inflammatory cytokines IL-10, TGF-β directly targeting on DC functions, are powerful tolerogenic agents which induce tolerogenic DCs to secrete even higher levels of anti-inflammatory cytokines. IL-10 interferes with differentiation of peripheral blood monocytes to DCs and induces cells with a tolerogenic phenotype [168]. Addition of IL-10 to differentiation cocktail induces a sustained inhibitory effect on subsequent maturation of monocyte-derived DCs And exogenous IL-10 reduced the expression of MHCⅡand CD80, decreased IL-12p40 level, and increased IL-10 level in DC, which resulted in impaired capacity to stimulate allogenic T-cell proliferation [169]. IT9302, a nonameric peptide homologous to the C-terminal domain of human IL-10, can promote monocyte differentiation to tolerogenic DCs through mechanisms involving STAT3 inactivation and NF-κB intracellular pathway blockade [170]. And IL-10 induced tolerogenic DCs were previously reported to prolong allograft survival by blocking the expression of CD80 and CD86, leading to apoptosis of allospecific T cells [171]. Similar to IL-10, TGF-β utilizes distinct pathways to reduce the expression of CD80 and CD86 by DCs and suppresses IL-12 production, giving rise to a reduced capacity to prime T cells [172]. Moreover, tolerogenic DCs stimulated by TGF-β delayed corneal allograft rejection by up-regulation the number of Foxp3+ Tregs [173]. And treatment of grafted β-cells islets with TGF-β-generated tolerogenic DCs prolonged graft survival [174]. Besides, genetic manipulation of DC maturation has allowed DC up-regulation of IL-10 and TGF-β that inhibit antigen presenting process. And those genetically programmed DCs display tolerogenic function through various mechanisms and some

of them even show therapeutic potential for graft rejection and autoimmune diseases [103]. 2.3.4 Other regulators Several other molecules have been found to intrinsically regulate DC maturation, and importantly, absence of any one of these regulators results in evident development of autoimmune diseases in mouse models. These intrinsic controllers have different protein characters, including αvβ8 integrin [175]; signal transducer and activator of transcription 3 (STAT3) [176] and A20 [95]. αvβ8 integrin expressed by DCs activates latent TGF-β and is critical for immune homeostasis [177]. Mice with DCs lacking αvβ8 and those lacking αvβ8 on all immune cells displayed similar pathologies of splenomegaly, lymphadenopathy, spontaneous T cell activation, autoantibodies, and colitis [175]. STAT3-mediated transcription was required to suppress activation markers on DCs [178]. Moreover, DCs can be categorized by the levels of cellular STAT3 phosphorylation/activation [178], the highest of which correlates with low expression of MHC class II. Thus, those molecules serve as ‘domestic police’ that inhibit DC dysfunction and the subsequent T cell disorder, and eventually contribute to the prevention of autoimmune diseases. A20, the product of the TNFAIP3 gene, restricts a number of innate immune signaling cascades that regulate DC functions, including TLR, NOD2, CD40, and TNF signals [179-181].

2.4 DC migration in autoimmunity The ability of DCs to initiate and orchestrate adaptive immune responses is a consequence of their localization within tissues and their specialized capacity for migration. Accumulating evidence indicates that trafficking molecule matrix metalloproteinases (MMPs) [182, 183], chemokines and their receptors [184] and adhesion molecules [185] play critical roles in orchestrating DC migration. Once a mobilization signal has triggered detachment from parenchymal tissues, maturing DC secrete MMPs to disintegrate extracellular matrix (ECM) barriers. Then under the regulation of chemokines and adhesion molecules, DCs traffic to secondary lymphoid organs to present the antigen to T cells and elicit the adaptive immune responses, and

consequently result in autoimmune diseases. Hence, pharmacological inhibition on DC migration will contribute to the prevention of autoimmune diseases. Prior studies have reported that pharmacological effects on DC migration can be achieved by immunomodulatory agents, including immunosuppressant such as corticosteroids [34, 186], vitamin D3 [34], CsA [34, 73, 74], mycophenolate [187] and leflunomide [188, 189]; Chinese herbal medicine such as triptolide [190], apigenin [191] and quercetin [75]; cytokines such as IL-10 [192], IL-21 [193], M-CSF [194]. These agents modulate DC migration primarily via control of trafficking molecules. Here, we review the key molecular traffic signals that govern the migration of DCs and provide novel insights into the significance of DC migration in the treatment of autoimmune diseases.

2.4.1 Matrix metalloproteinases Matrix metalloproteinases (MMPs), particularly MMP-2 and MMP-9 serve to disintegrate extracellular matrix(ECM) barriers, such as collagen types I-IV, fibronectin, and laminin [182, 195], which enables DCs to traffick through interstitial space to afferent lymph vessels. MMPs have been found to be key molecules in DC migration. Neutralizing or suppressing antibodies to MMP activity inhibits DC migration in Matrigel assays as well as langerhans cell(LC) emigration from skin explants [196, 197]. And MMP-9-deficient DCs in vitro are markedly defective in transepithelial migration and move poorly to LNs in vivo [198]. Inhibitors of metalloproteinases (TIMPs) and endogenous regulators of MMP activity have been suggested to block interstitial DC migration [182]. Furthermore, previous study showed that over-expression of genes encoding MMPs, such as MMP-1, -10 and -12 correlated with enhanced migratory capacity of DCs measured in Matrigel assays [199]. Besides, studies have suggested that MMPs(especially MMP-9) play a critical role in autoimmune disease. Over production of MMP-9 have been found in the sera and synovial fluid (SF) from patients with RA [200]. And the increased level of MMP-9 activity in the synovial fluid of RA and the severity of the disease has been correlated [201]. MMP-9 deficient mice showed inhibition of the development of

antibody-induced arthritis, one of the murine models of RA. The mechanisms that MMP-9 promote arthritis development may result from the effect of MMP-9 on migration of inflammatory cells, such as DCs [202]. Those findings indicate a potential role of MMPs in DC migration in the pathogenesis of autoimmune diseases and pharmacological inhibition on MMPs may constitute an exciting future for the treatment of autoimmune diseases. 2.4.2 Chemokines and the receptors Chemokines are small-secreted chemotactic cytokines that regulate the migration of leukocytes under steady state and inflammatory conditions. The migratory pathway of DCs start with recognition of a mobilizing signal induced by a variety of maturation factors, such as IL-1β, TNF-α and LPS [203]. Administration of these molecules leads to the loss of DCs from the periphery within a few hours. Pharmacological inhibitors or neutralizing antibodies of IL-1β or TNF-α impair mobilization and migration of DCs [203, 204]. Corticosteroids inhibit DC migration to lymphoid tissues through suppressing the inflammatory cytokine production. DC activation initiates a significant change in the repertoire of chemokine receptors (CCRs) [186]. The dominant chemokine pathway for DC migration to lymph nodes via lymphatics is CCR7 and its ligands, CCL19 and CCL21. CCR7-deficient mice show defective DC migration to LNs, which results in fewer putative migratory DCs in LNs after contact sensitization [205, 206]. Mobilizing signal induced by TNF-α significantly up-regulates the expression of CCL21 secreted by lymphatic endothelial cells, thus making the reactive vessels even more attractive to migrating DCs [207, 208]. Neutralizing antibodies to CCL21 has been suggested to inhibit the migration of skin-derived DCs into skin-draining LNs [209]. The cyclophilin-binding agent sanglifehrin A can suppress DC migration by suppressing CCL5, CCL17, CCL19, CXCL9 and CXCL10 expression in human monocyte-derived DCs [210]. IFN-β inhibits DC migration through STAT-1–mediated transcriptional suppression of CCR7 [211]. Chinese herbal derived immunosuppressive medicine triptolide impairs DC migration by inhibiting CCR7 and COX-2 expression [190] . Some flavonoid compounds like quercetin and apigenin can also inhibit DC migration through

down-regulation of chemokines and their receptors [75, 191]. Several other chemokine receptors play a role in DC traffic from the periphery to draining lymph nodes but appear less important. Migration of plasmacytoid DCs (pDCs ) activated with CpG for 24 h from peripheral blood into the lymph nodes has been indicated to depend on the CXCR3 and ligand CXCL9 [212]. CXCR4 inhibition impairs migration of LCs and dermal DCs to draining LNs [213]. Moreover, emigration of skin-DCs and lung-DCs [214] to the draining lymph node was partially blocked in CCR8-deficient mice. Hence, chemokine receptors are essential in lymph node homing of endogenous DCs. And pharmacological therapy on DC migration may target on down-regulation the expression of chemokine receptors on DCs or local chemokine expression. 2.4.3 Adhesion molecules In addition to acquiring the capacity to across ECM barriers, maturing DCs must develop the ability to entry into lymphatic system. Accumulating evidence indicates that multiple traffic molecules play a role in DC migration through the lymphatic system [4]. The best-validated molecules for DC migration into afferent lymphatics are adhesion molecules. After pro-inflammation-cytokine stimulation, lymphatic endothelial cells up regulate adhesion molecules and form tight and adherens junctions, resulting in DC trafficking within the afferent lymphatics [215, 216]. The key adhesion molecules for DC migration included selectins, intercellular adhesion molecule-1 (ICAM-1), vascular cell adhesion molecule-1 (VCAM-1) and their respective receptors [216]. Related study showed that antibody neutralization or suppression on VCAM-1 and ICAM-1expression can block DC adhesion to lymphatic endothelium and migration into afferent lymph vessels both in vitro and in vivo [217, 218]. And ICAM-1 deficiency mice also showed impaired langerhans cell migration to lymph nodes(LNs) [219]. P-selectin glycoprotein ligand-1 (PSGL-1) is the ligand for P-selectin and highly expressed on DCs. Prior study suggests that antibody against PSGL-1 block DC migration into LNs [220]. Similarly, integrin α6 is upregulated and contributes to migration of LCs [221]. Currently, the inhibition of adhesion molecules has been found to prevent or ameliorate disease severity in animal

models and clinical studies [216, 222]. Treatment with anti-ICAM-1 monoclonal antibody(mAb) have been found to reduce the severity of collagen- induced arthritis in mice and adjuvant induced arthritis in rats and to prolong the survival of cardiac allografts in mice [222-224]. Whereas pre-clinical studies of an antisense oligonucleotide to human ICAM-1 in Crohn’s disease and RA demonstrated no significant differences in disease severity between the placebo and treated patients [216, 225]. Similarly, related studies showed that anti-E-selectin mAb treatment protected against adjuvant-induced arthritis and colitis in animal models [216, 226], but not effective in the treatment of psoriasis in a randomized clinical trial [227]. However, not all the adhesion blockade studies show disappointing results. The anti-CD11a (the receptor of ICAM-1) mAb, efalizumab, has shown promising results in psoriasis patients. And the anti-α4 mAb, natalizumab, has proven to be safe and efficacious in patients with multiple sclerosis [228] or Crohn’s disease [229]. Taken together, adhesive interaction is a most important aspect of DC migration and understanding the underlying mechanisms may provide new potential therapeutic targets to modulate immune responses in autoimmune diseases. 2.4.4 S1P and receptors Sphingosine-1-phosphate (S1P), a potent bioactive sphingolipid metabolite that regulates many cellular processes including cell survival, cytoskeletal rearrangements, cell migration, and production of cytokines and chemokines , is involved in many kinds of diseases such as atherosclerosis, cancer, asthma and autoimmune diseases [230, 231]. S1P and its receptors also play a role in the interstitial migration of fully differentiated DCs. The novel immune-modulator FTY720 is a chemical derivative of myriocin, and its phosphorylated metabolite FTY720-P is a structural homolog of S1P. FTY720 inhibits S1P function through internalizing of S1P receptors, making cells unresponsive to S1P [232]. Researchers have demonstrated FTY720 could alleviate the clinical signs of experimental asthma, allergic contact dermatitis, and skin allograft transplantation through impairing DC trafficking to draining LNs [233-235]. Our recent study showed that FTY720 treatment also significantly suppressed the incidence and severity of collagen-induced arthritis (CIA) in DBA/1J mice via the

modulation of DC migration. CCR7 down-regulation on DCs and decreased CCL19 secretion are responsible for the impairment of DC migration by FTY720. The results indicate inhibition on migration by FTY720 may provide a novel approach in treating autoimmune disease such as RA[236]. And FTY720 has been shown to be a useful agent for the prevention of transplant rejection and has been proved by FDA to treat MS [237]. 2.4.5 Others molecules Factors present in inflammatory sites, such as lipid mediators, particularly prostaglandin E2 (PGE2) [238], and the high mobility group box-1 (HMGB1) protein [239], have been shown to induce migration of maturing DCs. The role of PGE2 in DC migration is strongly supported by the fact that mice deficient in the PGE2 receptor EP4 or wild-type mice treated with an EP4 antagonist exhibit reduced DC migration to lymph nodes [240]. The PGE2-induced DC migration is mediated through up-regulates MMP-9 expression, including both secreted and membranebound MMP-9 [183]. Besides, Faure-Andre and colleagues recently noted that DC migration was regulated by Ii, a key intracellular chaperone of MHC class II molecules that directs MHC class II endosomal localization and enables peptide loading. Ii-mediated association and activation of myosin light chain, which mediates cell migration, may enable DCs to effectively probe the tissue microenvironment [241]. DCs express cannabinoid receptor 2 (CBR2) and CBR2 agonists decrease autoimmune inflammation and DC production of pro-inflamamtory cytokines and capacity to stimulate CD4+ T cells. CBR2 agonists were found to inhibit DC migration from peripheral tissues by blocking MMP9 expression [242] .

3. Conclusion As the most powerful APC, DCs serve as the key bridge between innate and adaptive immunity. DCs also function as a critical switcher between immune activation and immune tolerance. To be a truly effective immune system manipulator, DCs must endure many steps of evolution. And each step was regulated subtly by a

variety of molecules. These steps, from differentiation, endocytosis to maturation and migration, are closely associated with the function of DCs and the outcome of immune response. Impaired endocytosis of dead cells by DCs is implicated in the pathogenesis of SLE; Different DC maturation state determines the immune system to be activated or tolerant; Migration is the prerequisite step for DCs to intact with T cells/B cells in LNs and orchestrate them subsequently. The complexity of DC evolution process and the flexibility of their property determine the biological basis of their variety functions and the ability to react with different environments efficiently. And also provide us with plenty therapeutic targets to manipulate them. Further studies in the mechanisms of current drugs or novel molecules in DC functions will be of great significance for developing unpreceding protocols for the treatment of autoimmune diseases.

Conflict of interest The authors have no conflict of interest. Acknowledgments We thank the many researchers who have contributed to our current understanding of DC functions. This work was supported by grants from National Natural Science Foundation of China (Grant No.81202359,81471613)

References [1] Torres-Aguilar H. Tolerogenic dendritic cells in autoimmune diseases: crucial players in induction and prevention of autoimmunity. Autoimmun Rev, 2010; 10(1): 8-17. [2] Lutzky V, Hannawi S and Thomas R. Cells of the synovium in rheumatoid arthritis. Dendritic cells. Arthritis Res Ther, 2007; 9(4): p. 219. [3] Walsh NC, et al. Rheumatic diseases: the effects of inflammation on bone. Immunol Rev, 2005; 208: 228-51. [4] Alvarez D, Vollmann EH and Andrian UH. Mechanisms and consequences of dendritic cell migration. Immunity, 2008; 29(3): 325-42. [5] Khan S, Greenberg JD and Bhardwaj N. Dendritic cells as targets for therapy in rheumatoid arthritis. Nat Rev Rheumatol, 2009; 5(10): p. 566-71. [6] Firestein GS. Evolving concepts of rheumatoid arthritis. Nature, 2003; 423(6937): p. 356-61. [7] Ganguly D, et al. The role of dendritic cells in autoimmunity. Nat Rev Immunol, 2013; 13(8): 566-77. [8] Steinman RM, Hawiger D and Nussenzweig MC. Tolerogenic dendritic cells. Annu Rev Immunol, 2003; 21: 685-711. [9] Torres-Aguilar H, et al. Tolerogenic dendritic cells in autoimmune diseases: crucial players in induction and prevention of autoimmunity. Autoimmun Rev, 2010; 10(1): 8-17. [10] Merad M, et al. The dendritic cell lineage: ontogeny and function of dendritic cells and their subsets in the steady state and the inflamed setting. Annu Rev Immunol, 2013; 31: 563-604. [11] Banchereau J, et al. Immunobiology of dendritic cells. Annu Rev Immunol, 2000; 18: 767-811. [12] Rea D, et al. Glucocorticoids transform CD40-triggering of dendritic cells into an alternative activation pathway resulting in antigen-presenting cells that secrete IL-10. Blood, 2000; 95(10): 3162-7. [13] Moser MM, et al. Glucocorticoids down-regulate dendritic cell function in vitro and in vivo. Eur J Immunol, 1995; 25(10): 2818-24. [14] Berer A, et al. 1,25-Dihydroxyvitamin D3 inhibits dendritic cell differentiation and maturation in vitro. Exp. Hematol, 2000; 28:575–583. [15] Farias AS, et al. Vitamin D3 induces IDO+ tolerogenic DCs and enhances Treg, reducing the severity of EAE. CNS Neurosci Ther, 2013; 19(4): 269-77.

[16] Raiotach-Regue D, et al. Stable antigen-specific T-cell hyporesponsiveness induced by tolerogenic dendritic cells from multiple sclerosis patients. Eur J Immunol, 2012; 42(3): 771-82. [17] Ferreira GB, et al. Differential protein pathways in 1, 25-dihydroxyvitamin d(3) and dexamethasone modulated tolerogenic human dendritic cells. J Proteome Res, 2012;11(2): p. 941-71. [18] Volchenkov R, et al. Type 1 regulatory T cells and regulatory B cells induced by tolerogenic dendritic cells. Scand J Immunol, 2013; 77(4): 246-54. [19] Volchenkov R, et al. In vitro suppression of immune responses using monocyte-derived tolerogenic dendritic cells from patients with primary Sjogren's syndrome. Arthritis Res Ther, 2013; 15(5): R114. [20] Xing N, et al. Distinctive dendritic cell modulation by vitamin D(3) and glucocorticoid pathways. Biochem Biophys Res Commun, 2002; 297(3): 645-52. [21] Unger WW, et al. Induction of Treg by monocyte-derived DC modulated by vitamin D3 or dexamethasone: differential role for PD-L1. Eur J Immunol, 2009; 39(11): 3147-59. [22] Garcia-Gonzalez P, et al. A short protocol using dexamethasone and monophosphoryl lipid A generates tolerogenic dendritic cells that display a potent migratory capacity to lymphoid chemokines. J Transl Med, 2013; 11: 128. [23] Turnquist HR, et al. Rapamycin-conditioned dendritic cells are poor stimulators of allogeneic CD4+ T cells, but enrich for antigen-specific Foxp3+ T regulatory cells and promote organ transplant tolerance. J Immunol, 2007; 178(11): 7018-31. [24] Taner T, et al. Rapamycin-treated, alloantigen-pulsed host dendritic cells induce ag-specific T cell regulation and prolong graft survival. Am J Transplant, 2005; 5(2): 228-36. [25] Hackstein H, et al. Rapamycin inhibits IL-4--induced dendritic cell maturation in vitro and dendritic cell mobilization and function in vivo. Blood, 2003. 101(11): p. 4457-63. [26] Fischer RT, et al. Rapamycin-conditioned, alloantigen-pulsed myeloid dendritic cells present donor MHC class I/peptide via the semi-direct pathway and inhibit survival of antigen-specific CD8(+) T cells in vitro and in vivo. Transpl Immunol, 2011; 25(1): 20-6. [27] Fedoric B and Krishnan R. Rapamycin downregulates the inhibitory receptors ILT2, ILT3, ILT4 on human dendritic cells and yet induces T cell hyporesponsiveness independent of FoxP3 induction. Immunol Lett, 2008; 120(1-2): 49-56. [28] Hackstein H, et al. Aspirin inhibits in vitro maturation and in vivo immunostimulatory function of

murine myeloid dendritic cells. J Immunol, 2001; 166(12): 7053-62. [29] Buckland M, et al. Aspirin modified dendritic cells are potent inducers of allo-specific regulatory T-cells. Int Immunopharmacol, 2006; 6(13-14): 1895-901. [30] Cai DT, et al. Aspirin regulates SNARE protein expression and phagocytosis in dendritic cells. Mol Membr Biol, 2011; 28(2): 90-102. [31] Verhasselt V, et al. N-acetyl-L-cysteine inhibits primary human T cell responses at the dendritic cell level: association with NF-kappaB inhibition. J Immunol, 1999; 162(5): 2569-74. [32] Mehling A, et al. Mycophenolate mofetil impairs the maturation and function of murine dendritic cells. J Immunol, 2000; 165(5): 2374-81. [33] Colic M, et al. Mycophenolate mofetil inhibits differentiation, maturation and allostimulatory function of human monocyte-derived dendritic cells. Clin Exp Immunol, 2003; 134(1): 63-9. [34] Hackstein H and Thomson AW. Dendritic cells: emerging pharmacological targets of immunosuppressive drugs. Nat Rev Immunol, 2004; 4(1): 24-34. [35] Adorini L, Giarratana N and Penna G. Pharmacological induction of tolerogenic dendritic cells and regulatory T cells. Semin Immunol, 2004; 16(2): 127-34. [36] Millard A, et al. Butyrate affects differentiation, maturation and function of human monocyte-derived dendritic cells and macrophages. Clin Exp Immunol, 2002; 130(2): p. 245-55. [37] Iruretagoyena MI, et al. Inhibition of nuclear factor-kappa B enhances the capacity of immature dendritic cells to induce antigen-specific tolerance in experimental autoimmune encephalomyelitis. J Pharmacol Exp Ther, 2006; 318(1): 59-67. [38] Szatmari I, et al. PPARgamma regulates the function of human dendritic cells primarily by altering lipid metabolism. Blood; 2007. 110(9): 3271-80. [39] Giusti P, et al. The novel anti-rheumatic compound Rabeximod impairs differentiation and function of human pro-inflammatory dendritic cells and macrophages. Immunobiology, 2011; 216(1-2): 243-50. [40] Rajnavolgyi , et al. Effects of RAMEA-complexed polyunsaturated fatty acids on the response of human dendritic cells to inflammatory signals. Beilstein J Org Chem, 2014; 10: 3152-60. [41] Li J and Zhang F. The immunoregulatory effects of Chinese herbal medicine on the maturation and function of dendritic cells. J Ethnopharmacol, 2015; 171: 184-95. [42] Onai N, et al. Flt3 in regulation of type I interferon-producing cell and dendritic cell development.

Ann N Y Acad Sci, 2007; 1106: 253-61. [43] Karsunky H, et al. Flt3 ligand regulates dendritic cell development from Flt3+ lymphoid and myeloid-committed progenitors to Flt3+ dendritic cells in vivo. J Exp Med, 2003; 198(2): 305-13. [44] McKenna H, et al. Mice lacking flt3 ligand have deficient hematopoiesis affecting hematopoietic progenitor cells, dendritic cells, and natural killer cells. Blood, 2000; 95(11): 3489-97. [45] Tussiwand R, et al. Inhibition of natural type I IFN-producing and dendritic cell development by a small molecule receptor tyrosine kinase inhibitor with Flt3 affinity. J Immunol, 2005; 175(6): p. 3674-80. [46] Adolfsson J, et al. Upregulation of Flt3 expression within the bone marrow Lin(-)Sca1(+)c-kit(+) stem cell compartment is accompanied by loss of self-renewal capacity. Immunity, 2001; 15(4): 659-69. [47] Ramos MI, et al. FMS-related tyrosine kinase 3 ligand (Flt3L)/CD135 axis in rheumatoid arthritis. Arthritis Res Ther, 2013; 15(6): R209. [48] Ramos MI, et al. Absence of Fms-like tyrosine kinase 3 ligand (Flt3L) signalling protects against collagen-induced arthritis. Ann Rheum Dis, 2015; 74(1): 211-9. [49] Dehlin M, et al. Inhibition of fms-like tyrosine kinase 3 alleviates experimental arthritis by reducing formation of dendritic cells and antigen presentation. J Leukoc Biol, 2011; 90(4): 811-7. [50] Lee CN, et al. NOD mice are functionally deficient in the capacity of cross-presentation. Immunol Cell Biol, 2015; 93(6): 548-57. [51] Sallusto F and Lanzavecchia A. Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor alpha. J Exp Med, 1994; 179(4): 1109-18. [52] Inaba K, et al. Generation of large numbers of dendritic cells from mouse bone marrow cultures supplemented with granulocyte/macrophage colony-stimulating factor. J Exp Med, 1992; 176(6): 1693-702. [53] Caux C, et al. CD34+ hematopoietic progenitors from human cord blood differentiate along two independent dendritic cell pathways in response to GM-CSF+TNF alpha. J Exp Med, 1996; 184(2): 695-706. [54] Banchereau J and Palucka AK. Dendritic cells as therapeutic vaccines against cancer. Nat Rev Immunol, 2005; 5(4): 296-306.

[55]. Conti L and Gessani S. GM-CSF in the generation of dendritic cells from human blood monocyte precursors: recent advances. Immunobiology, 2008; 213(9-10): 859-70. [56] Lebre MC and Tak PP. Dendritic cell subsets: their roles in rheumatoid arthritis. Acta Reumatol Port, 2008; 33(1): 35-45. [57] MacDonald KP, et al. Resistance of rheumatoid synovial dendritic cells to the immunosuppressive effects of IL-10. J Immunol, 1999; 163(10): p. 5599-607. [58] King IL, Dickendesher TL and Segal BM. Circulating Ly-6C+ myeloid precursors migrate to the CNS and play a pathogenic role during autoimmune demyelinating disease. Blood, 2009; 113(14): 3190-7. [59] Mausberg

AK,

Jander

S

and

Reichmann

G.

Intracerebral

granulocyte-macrophage

colony-stimulating factor induces functionally competent dendritic cells in the mouse brain. Glia, 2009; 57(12): 1341-50. [60] Gopisetty A, et al. OX40L/Jagged1 cosignaling by GM-CSF-induced bone marrow-derived dendritic cells is required for the expansion of functional regulatory T cells. J Immunol, 2013; 190(11): 5516-25. [61] Rutell S, et al. Hepatocyte growth factor favors monocyte differentiation into regulatory interleukin (IL)-10++IL-12low/neg accessory cells with dendritic-cell features. Blood, 2006; 108(1): p. 218-27. [62] Burmester GR, et al. Mavrilimumab, a human monoclonal antibody targeting GM-CSF receptor-alpha, in subjects with rheumatoid arthritis: a randomised, double-blind, placebo-controlled, phase I, first-in-human study. Ann Rheum Dis, 2011; 70(9): 1542-9. [63] Burmester GR, et al. Efficacy and safety of mavrilimumab in subjects with rheumatoid arthritis. Ann Rheum Dis, 2013; 72(9): 1445-52. [64] Li BZ, et al. GM-CSF alters dendritic cells in autoimmune diseases. Autoimmunity, 2013; 46(7): 409-18. [65] Pixley FJ and Stanley ER. CSF-1 regulation of the wandering macrophage: complexity in action. Trends Cell Biol, 2004; 14(11): 628-38. [66] Bogunovic M, et al. Origin of the lamina propria dendritic cell network. Immunity, 2009; 31(3): 513-25. [67] Ginhoux F, et al. The origin and development of nonlymphoid tissue CD103+ DCs. J Exp Med,

2009; 206(13): 3115-30. [68] Ginhoux F, et al. Langerhans cells arise from monocytes in vivo. Nat Immunol, 2006; 7(3): 265-73. [69] Li G, Kim YJ and Broxmeyer HE. Macrophage colony-stimulating factor drives cord blood monocyte differentiation into IL-10(high)IL-12absent dendritic cells with tolerogenic potential. J Immunol, 2005; 174(8): 4706-17. [70] Onai N, et al. Identification of clonogenic common Flt3+M-CSFR+ plasmacytoid and conventional dendritic cell progenitors in mouse bone marrow. Nat Immunol, 2007; 8(11): 1207-16. [71] Fancke B, et al. M-CSF: a novel plasmacytoid and conventional dendritic cell poietin. Blood, 2008; 111(1): 150-9. [72] Ghoreschi K, et al. Fumarates improve psoriasis and multiple sclerosis by inducing type II dendritic cells. J Exp Med, 2011; 208(11): 2291-303. [73] Woltman AM, et al. The effect of calcineurin inhibitors and corticosteroids on the differentiation of human dendritic cells. Eur J Immunol, 2000; 30(7): p. 1807-12. [74] Chen T, et al. Cyclosporin A impairs dendritic cell migration by regulating chemokine receptor expression and inhibiting cyclooxygenase-2 expression. Blood, 2004; 103(2): 413-21. [75] Huang RY, et al. Immunosuppressive effect of quercetin on dendritic cell activation and function. J Immunol, 2010; 184(12): 6815-21. [76] Li X and Kimberly RP. Targeting the Fc receptor in autoimmune disease. Expert Opin Ther Targets, 2014; 18(3): 335-50. [77] Samsom JN, et al. Fc gamma RIIB regulates nasal and oral tolerance: a role for dendritic cells. J Immunol, 2005; 174(9): 5279-87. [78] Desai DD, et al. Fc gamma receptor IIB on dendritic cells enforces peripheral tolerance by inhibiting effector T cell responses. J Immunol, 2007; 178(10): 6217-26. [79] Ravetch JV and Bolland S. IgG Fc receptors. Annu Rev Immunol, 2001; 19: 275-90. [80] Marino M, et al. Prevention of systemic lupus erythematosus in MRL/lpr mice by administration of an immunoglobulin-binding peptide. Nat Biotechnol, 2000; 18(7): 735-9. [81] Clarkson SB, et al. Blockade of clearance of immune complexes by an anti-Fc gamma receptor monoclonal antibody. J Exp Med, 1986; 164(2): 474-89. [82] Clarkson SB, et al. Treatment of refractory immune thrombocytopenic purpura with an anti-Fc

gamma-receptor antibody. N Engl J Med, 1986. 314(19): 1236-9. [83] Wenink MH, et al. The inhibitory Fc gamma IIb receptor dampens TLR4-mediated immune responses and is selectively up-regulated on dendritic cells from rheumatoid arthritis patients with quiescent disease. J Immunol, 2009; 183(7): 4509-20. [84] Zhang Y, et al. Immune complex enhances tolerogenecity of immature dendritic cells via FcgammaRIIb and promotes FcgammaRIIb-overexpressing dendritic cells to attenuate lupus. Eur J Immunol, 2011; 41(4): 1154-64. [85] Marino M, et al. Prevention of systemic lupus erythematosus in MRL/lpr mice by administration of an immunoglobulin-binding peptide. Nat Biotechnol, 2000; 18(7): 735-9. [86] Jiang W, et al. The receptor DEC-205 expressed by dendritic cells and thymic epithelial cells is involved in antigen processing. Nature, 1995; 375(6527): 151-5. [87] Niezold T, et al. DNA vaccines encoding DEC205-targeted antigens: immunity or tolerance? Immunology, 2015; 145(4): 519-33. [88] Silva-Sanchez A, et al. ESAT-6 Targeting to DEC205+ Antigen Presenting Cells Induces Specific-T Cell Responses against ESAT-6 and Reduces Pulmonary Infection with Virulent Mycobacterium tuberculosis. PLoS One, 2015; 10(4): 0124828. [89] Ring S, et al. Targeting of autoantigens to DEC205 (+) dendritic cells in vivo suppresses experimental allergic encephalomyelitis in mice. J Immunol, 2013; 191(6): p. 2938-47. [90] Katakowski JA, et al. Delivery of siRNAs to Dendritic Cells Using DEC205-Targeted Lipid Nanoparticles to Inhibit Immune Responses. Mol Ther, 2015. [91] Spiering R, et al. DEC205+ Dendritic Cell-Targeted Tolerogenic Vaccination Promotes Immune Tolerance in Experimental Autoimmune Arthritis. J Immunol, 2015; 194(10): 4804-13. [92] Svajger U, et al. C-type lectin DC-SIGN: an adhesion, signalling and antigen-uptake molecule that guides dendritic cells in immunity. Cell Signal, 2010; 22(10): 1397-405. [93] Geijtenbeek TB. Dunnen J and Gringhuis SI. Pathogen recognition by DC-SIGN shapes adaptive immunity. Future Microbiol, 2009; 4(7): 879-90. [94] Foligne B, et al. A key role of dendritic cells in probiotic functionality. PLoS One, 2007; 2(3): e313. [95] Hammer GE and Ma A. Molecular control of steady-state dendritic cell maturation and immune homeostasis. Annu Rev Immunol, 2013; 31: 743-91.

[96] Takeuchi O and Akira S. Pattern recognition receptors and inflammation. Cell, 2010; 140(6): p. 805-20. [97] Deane JA, et al. Control of toll-like receptor 7 expression is essential to restrict autoimmunity and dendritic cell proliferation. Immunity, 2007; 27(5): 801-10. [98] Lande R, et al. Plasmacytoid dendritic cells sense self-DNA coupled with antimicrobial peptide. Nature, 2007; 449(7162): 564-9. [99] Lee J, et al. Molecular basis for the immunostimulatory activity of guanine nucleoside analogs: activation of Toll-like receptor 7. Proc Natl Acad Sci USA, 2003; 100(11): 6646-51. [100] Hacker H, et al. CpG-DNA-specific activation of antigen-presenting cells requires stress kinase activity and is preceded by non-specific endocytosis and endosomal maturation. EMBO J, 1998; 17(21): 6230-40. [101] Marta M, Meier UC and Lobell A. Regulation of autoimmune encephalomyelitis by toll-like receptors. Autoimmun Rev, 2009; 8(6): 506-9. [102] Sadanaga A, et al. Protection against autoimmune nephritis in MyD88-deficient MRL/lpr mice. Arthritis Rheum, 2007; 56(5): 1618-28. [103] Liu J and Cao XT. Regulatory dendritic cells in autoimmunity: A comprehensive review. J Autoimmun, 2015; 63: 1-12. [104] Cao W, et al. BDCA2/Fc epsilon RI gamma complex signals through a novel BCR-like pathway in human plasmacytoid dendritic cells. PLoS Biol, 2007; 5(10): e248. [105] Rock J, et al. CD303 (BDCA-2) signals in plasmacytoid dendritic cells via a BCR-like signalosome involving Syk, Slp65 and PLCgamma2. Eur J Immunol, 2007; 37(12): p. 3564-75. [106] Zhang J, Somani AK and Siminovitch KA. Roles of the SHP-1 tyrosine phosphatase in the negative regulation of cell signalling. Semin Immunol, 2000; 12(4): 361-78. [107] Kaneko T, et al. Dendritic cell-specific ablation of the protein tyrosine phosphatase Shp1 promotes Th1 cell differentiation and induces autoimmunity. J Immunol, 2012; 188(11): 5397-407. [108] Meyer-Wentrup F, et al. Targeting DCIR on human plasmacytoid dendritic cells results in antigen presentation and inhibits IFN-alpha production. Blood, 2008; 111(8): 4245-53. [109] Meyer-Wentrup F, et al. DCIR is endocytosed into human dendritic cells and inhibits TLR8-mediated cytokine production. J Leukoc Biol, 2009; 85(3): 518-25. [110] Franchi L, Munoz-Planillo R and Nunez G. Sensing and reacting to microbes through the

inflammasomes. Nat Immunol, 2012; 13(4): 325-32. [111] Rathinam VA, Vanaja SK and Fitzgerald KA. Regulation of inflammasome signaling. Nat Immunol, 2012; 13(4): 333-42. [112] Strober W and Watanabe T. NOD2, an intracellular innate immune sensor involved in host defense and Crohn's disease. Mucosal Immunol, 2011; 4(5): 484-95. [113] Penack O, et al. NOD2 regulates hematopoietic cell function during graft-versus-host disease. J Exp Med, 2009; 206(10): 2101-10. [114] Maeda S, et al. Nod2 mutation in Crohn's disease potentiates NF-kappaB activity and IL-1beta processing. Science, 2005; 307(5710): 734-8. [115] Lorentzen JC, et al. Association of arthritis with a gene complex encoding C-type lectin-like receptors. Arthritis Rheum, 2007; 56(8): 2620-32. [116] Kastenbauer S and Ziegler-Heitbrock HW. NF-kappaB1 (p50) is upregulated in lipopolysaccharide tolerance and can block tumor necrosis factor gene expression. Infect Immun, 1999. 67(4): 1553-9. [117] Dissanayake D, et al. Nuclear factor-kappaB1 controls the functional maturation of dendritic cells and prevents the activation of autoreactive T cells. Nat Med, 2011; 17(12): 1663-7. [118] Ade N et al. NF-kappaB plays a major role in the maturation of human dendritic cells induced by NiSO(4) but not by DNCB. Toxicol Sci, 2007; 99(2): 488-501. [119] Martin E, et al. Antigen-specific suppression of established arthritis in mice by dendritic cells deficient in NF-kappaB. Arthritis Rheum, 2007; 56(7): 2255-66. [120] Popov I, et al. Preventing autoimmune arthritis using antigen-specific immature dendritic cells: a novel tolerogenic vaccine. Arthritis Res Ther, 2006; 8(5): R141. [121] Hilkens CM and Isaacs JD. Tolerogenic dendritic cell therapy for rheumatoid arthritis: where are we now? Clin Exp Immunol, 2013; 172(2): 148-57. [122] Watkins SK, et al. FOXO3 programs tumor-associated DCs to become tolerogenic in human and murine prostate cancer. J Clin Invest, 2011; 121(4): 1361-72. [123] Dejean AS, et al. Transcription factor Foxo3 controls the magnitude of T cell immune responses by modulating the function of dendritic cells. Nat Immunol, 2009; 10(5): 504-13. [124] Fallarino F, et al. CTLA-4-Ig activates forkhead transcription factors and protects dendritic cells from oxidative stress in nonobese diabetic mice. J Exp Med, 2004; 200(8): 1051-62.

[125] Lee JC and Smith KG. Prognosis in autoimmune and infectious disease: new insights from genetics. Clin Transl Immunology, 2014; 3(5): e15. [126] Lee JC, et al. Human SNP links differential outcomes in inflammatory and infectious disease to a FOXO3-regulated pathway. Cell, 2013. 155(1): p. 57-69. [127] Garrett WS, et al. Communicable ulcerative colitis induced by T-bet deficiency in the innate immune system. Cell, 2007; 131(1): p. 33-45. [128] Garrett WS, et al. Colitis-associated colorectal cancer driven by T-bet deficiency in dendritic cells. Cancer Cell, 2009; 16(3): 208-19. [129] Garrett WS, et al. Enterobacteriaceae act in concert with the gut microbiota to induce spontaneous and maternally transmitted colitis. Cell Host Microbe, 2010; 8(3): 292-300. [130] Ren Y, et al. Tolerogenic dendritic cells modified by tacrolimus suppress CD4(+) T-cell proliferation and inhibit collagen-induced arthritis in mice. Int Immunopharmacol, 2014. 21(1): 247-54. [131] Steinschulte C, et al. Cutting edge: sanglifehrin A, a novel cyclophilin-binding immunosuppressant blocks bioactive IL-12 production by human dendritic cells. J Immunol, 2003; 171(2): 542-6. [132] Wu WL, et al. Immunosuppressive effects and mechanisms of leflunomide in dengue virus infection of human dendritic cells. J Clin Immunol, 2011; 31(6): 1065-78. [133] Bayry J, et al. Inhibition of maturation and function of dendritic cells by intravenous immunoglobulin. Blood, 2003; 101(2): 758-65. [134] Schultz KR, et al. Chloroquine treatment affects T-cell priming to minor histocompatibility antigens and graft-versus-host disease. Blood, 1995; 86(11): 4344-52. [135] Newton CA, et al.

Role of Toll-like receptor 9 in Legionella pneumophila-induced

interleukin-12 p40 production in bone marrow-derived dendritic cells and macrophages from permissive and nonpermissive mice. Infect Immun, 2007; 75(1): 146-51. [136] Dombrecht EJ, et al. Influence of anti-tumor necrosis factor therapy (Adalimumab) on regulatory T cells and dendritic cells in rheumatoid arthritis. Clin Exp Rheumatol, 2006; 24(1): 31-7. [137] Balanescu A, et al. Early and late effect of infliximab on circulating dendritic cells phenotype in rheumatoid arthritis patients. Int J Clin Pharmacol Res, 2005; 25(1): 9-18. [138] Richez C, et al. Tocilizumab treatment decreases circulating myeloid dendritic cells and monocytes, 2 components of the myeloid lineage. J Rheumatol, 2012; 39(6): 1192-7.

[139] Sato K, et al. Extracellular signal-regulated kinase, stress-activated protein kinase/c-Jun N-terminal kinase, and p38mapk are involved in IL-10-mediated selective repression of TNF-alpha-induced activation and maturation of human peripheral blood monocyte-derived dendritic cells. J Immunol, 1999; 162(7): 3865-72. [140] Knodler A, et al. Post-transcriptional regulaSztion of adapter molecules by IL-10 inhibits TLR-mediated activation of antigen-presenting cells. Leukemia, 2009; 23(3): 535-44. [141] Velten FW, et al. A gene signature of inhibitory MHC receptors identifies a BDCA3(+) subset of IL-10-induced dendritic cells with reduced allostimulatory capacity in vitro. Eur J Immunol, 2004; 34(10): 2800-11. [142] Li X, et al. Induction of type 2 T helper cell allergen tolerance by IL-10-differentiated regulatory dendritic cells. Am J Respir Cell Mol Biol, 2010; 42(2): 190-9. [143] Kubsch S, et al. Suppressor activity of anergic T cells induced by IL-10-treated human dendritic cells: association with IL-2- and CTLA-4-dependent G1 arrest of the cell cycle regulated by p27Kip1. Eur J Immunol, 2003; 33(7): 1988-97. [144] Steinbrink K, et al. CD4(+) and CD8(+) anergic T cells induced by interleukin-10-treated human dendritic cells display antigen-specific suppressor activity. Blood, 2002; 99(7): 2468-76. [145] Torres-Aguilar H, et al. Tolerogenic dendritic cells inhibit antiphospholipid syndrome derived effector/memory CD4(+) T cell response to beta2GPI. Ann Rheum Dis, 2012; 71(1): 120-8. [146] Yan F, et al. Tolerogenic dendritic cells suppress murine corneal allograft rejection by modulating CD28/CTLA-4 expression on regulatory T cells. Cell Biol Int, 2014; 38(7): 835-48. [147] Thomas DC, et al. Protection of islet grafts through transforming growth factor-beta-induced tolerogenic dendritic cells. Diabetes, 2013; 62(9): 3132-42. [148]

Segovia-Gamboa

N,

et

al.

Tolerogenic

dendritic

cells

induce

antigen-specific

hyporesponsiveness in insulin- and glutamic acid decarboxylase 65-autoreactive T lymphocytes from type 1 diabetic patients. Clin Immunol, 2014; 154(1): 72-83. [149] Stoop JN, Robinson KG and Hilkens CM. Developing tolerogenic dendritic cell therapy for rheumatoid arthritis: what can we learn from mouse models? Ann Rheum Dis, 2011; 70(9): 1526-33. [150] Mackern-Oberti JP, et al. Targeting dendritic cell function during systemic autoimmunity to restore tolerance. Int J Mol Sci, 2014; 15(9): 16381-417. [151] Zhang K, et al. Chrysin suppresses human CD14(+) monocyte-derived dendritic cells and

ameliorates experimental autoimmune encephalomyelitis. J Neuroimmunol, 2015; 288: p. 13-20. [152] Jung S, et al. Induction of IL-10 in rat peritoneal macrophages and dendritic cells by glatiramer acetate. J Neuroimmunol, 2004; 148(1-2): 63-73. [153] Kim IK, et al. Impact of innate and adaptive immunity on rejection and tolerance. Transplantation, 2008; 86(7): 889-94. [154]. Kalantari T, et al. Tolerogenic dendritic cells produced by lentiviral-mediated CD40- and interleukin-23p19-specific shRNA can ameliorate experimental autoimmune encephalomyelitis by suppressing T helper type 17 cells. Clin Exp Immunol, 2014; 176(2): 180-9. [155] Zheng X, et al. Treatment of autoimmune arthritis using RNA interference-modulated dendritic cells. J Immunol, 2010; 184(11): 6457-64. [156] Matsuki Y, et al. Novel regulation of MHC class II function in B cells. EMBO J, 2007; 26(3): p. 846-54. [157] Walseng E, et al. Ubiquitination regulates MHC class II-peptide complex retention and degradation in dendritic cells. Proc Natl Acad Sci USA, 2010; 107(47): 20465-70. [158] Shin JS, et al. Surface expression of MHC class II in dendritic cells is controlled by regulated ubiquitination. Nature, 2006; 444(7115): 115-8. [159] Ohmura-Hoshino M, et al. Cutting edge: requirement of MARCH-I-mediated MHC II ubiquitination for the maintenance of conventional dendritic cells. J Immunol, 2009; 183(11): 6893-7. [160] Blanco P, et al. Dendritic cells and cytokines in human inflammatory and autoimmune diseases. Cytokine Growth Factor Rev, 2008; 19(1): 41-52. [161] Gilbert KE, Manalo IF and Wu JJ. Efficacy and safety of etanercept and adalimumab with and without a loading dose for psoriasis: A systematic review. J Am Acad Dermatol, 2015; 73(2): 329-31. [162] Scott LJ. Etanercept: a review of its use in autoimmune inflammatory diseases. Drugs, 2014; 74(12): 1379-410. [163] Fenix-Caballero S, et al. Direct and indirect comparison of the efficacy and safety of adalimumab, etanercept, infliximab and golimumab in psoriatic arthritis. J Clin Pharm Ther, 2013; 38(4): 286-93. [164] Ogata A, et al. Subcutaneous formulation of tocilizumab for treatment of rheumatoid arthritis. Ther Deliv, 2015; 6(3): 283-95. [165] Tanaka T, Hishitani Y and Ogata A. Monoclonal antibodies in rheumatoid arthritis: comparative effectiveness of tocilizumab with tumor necrosis factor inhibitors. Biologics, 2014; 8: 141-53.

[166] Maxwell LJ, et al. TNF-alpha inhibitors for ankylosing spondylitis. Cochrane Database Syst Rev, 2015; 4: CD005468. [167] McDermott MF. Rilonacept in the treatment of chronic inflammatory disorders. Drugs Today (Barc), 2009; 45(6): 423-30. [168] Ruffner MA, et al. B7-1/2, but not PD-L1/2 molecules, are required on IL-10-treated tolerogenic DC and DC-derived exosomes for in vivo function. Eur J Immunol, 2009; 39(11): 3084-90. [169] Schwarz AM, et al. Impact of interleukin-10 on phenotype and gene expression during early monocyte differentiation into dendritic cells. Anticancer Res, 2013; 33(11): 4791-8. [170] Lopez MN, et al. A synthetic peptide homologous to IL-10 functional domain induces monocyte differentiation to TGF-beta+ tolerogenic dendritic cells. Immunobiology, 2011; 216(10): 1117-26. [171] Jiang H, et al. Administration of tolerogenic dendritic cells induced by interleukin-10 prolongs rat splenic allograft survival. Transplant Proc, 2004; 36(10): 3255-9. [172] Eljaafari A, Li YP and Miossec P. IFN-gamma, as secreted during an alloresponse, induces differentiation of monocytes into tolerogenic dendritic cells, resulting in FoxP3+ regulatory T cell promotion. J Immunol, 2009; 183(5): 2932-45. [173] Yan F, et al. Tolerogenic dendritic cells suppress murine corneal allograft rejection by modulating CD28/CTLA-4 expression on regulatory T cells. Cell Biol Int, 2014; 38(7): 835-48. [174] Thomas DC, et al. Protection of islet grafts through transforming growth factor-beta-induced tolerogenic dendritic cells. Diabetes, 2013; 62(9): 3132-42. [175] Travis MA, et al. Loss of integrin alpha(v)beta8 on dendritic cells causes autoimmunity and colitis in mice. Nature, 2007; 449(7160): 361-5. [176] Melillo JA, et al. Dendritic cell (DC)-specific targeting reveals Stat3 as a negative regulator of DC function. J Immunol, 2010; 184(5): 2638-45. [177] Travis MA, et al. Loss of integrin alpha(v)beta8 on dendritic cells causes autoimmunity and colitis in mice. Nature, 2007; 449(7160): 361-5. [178] Park SJ, et al. IL-6 regulates in vivo dendritic cell differentiation through STAT3 activation. J Immunol, 2004; 173(6): 3844-54. [179] Lee EG, et al. Failure to regulate TNF-induced NF-kappaB and cell death responses in A20-deficient mice. Science, 2000; 289(5488): 2350-4. [180] Wertz IE, et al. De-ubiquitination and ubiquitin ligase domains of A20 downregulate NF-kappaB

signalling. Nature, 2004; 430(7000): 694-9. [181] Boone DL, et al. The ubiquitin-modifying enzyme A20 is required for termination of Toll-like receptor responses. Nat Immunol, 2004; 5(10): 1052-60. [182] Ratzinger G, et al. Matrix metalloproteinases 9 and 2 are necessary for the migration of Langerhans cells and dermal dendritic cells from human and murine skin. J Immunol, 2002; 168(9): p. 4361-71. [183] Yen JH, Khayrullina T and Ganea D. PGE2-induced metalloproteinase-9 is essential for dendritic cell migration. Blood, 2008; 111(1): 260-70. [184] Gunn MD, et al. Mice lacking expression of secondary lymphoid organ chemokine have defects in lymphocyte homing and dendritic cell localization. J Exp Med, 1999; 189(3): 451-60. [185] Johnson LA, et al. An inflammation-induced mechanism for leukocyte transmigration across lymphatic vessel endothelium. J Exp Med, 2006; 203(12): 2763-77. [186] Cumberbatch M, Dearman RJ and Kimber I. Inhibition by dexamethasone of Langerhans cell migration: influence of epidermal

cytokine signals. Immunopharmacology, 1999; 41(3): 235-43.

[187] Mehling A, et al. Mycophenolate mofetil impairs the maturation and function of murine dendritic cells. J Immunol, 2000; 165(5): 2374-81. [188] Kirsch BM, et al. The active metabolite of leflunomide, A77 1726, interferes with dendritic cell function. Arthritis Res Ther, 2005; 7(3): R694-703. [189] Wu WL, et al. Immunosuppressive effects and mechanisms of leflunomide in dengue virus infection of human dendritic cells. J Clin Immunol, 2011; 31(6): 1065-78. [190] Chen X, et al. Triptolide, a constituent of immunosuppressive Chinese herbal medicine, is a potent suppressor of dendritic-cell maturation and trafficking. Blood, 2005; 106(7): 2409-16. [191] Li X, et al. Apigenin, a potent suppressor of dendritic cell maturation and migration, protects against collagen-induced arthritis. J Cell Mol Med, 2016; 20(1): 170-80. [192] Takayama T, et al. Mammalian and viral IL-10 enhance C-C chemokine receptor 5 but down-regulate C-C chemokine receptor 7 expression by myeloid dendritic cells: impact on chemotactic responses and in vivo homing ability. J Immunol, 2001; 166(12): 7136-43. [193] Brandt K, et al. Interleukin-21 inhibits dendritic cell activation and maturation. Blood, 2003; 102(12): 4090-8. [194] Li G, Kim YJ and Broxmeyer HE. Macrophage colony-stimulating factor drives cord blood

monocyte differentiation into IL-10(high)IL-12absent dendritic cells with tolerogenic potential. J Immunol, 2005; 174(8): 4706-17. [195] Yen JH, Khayrullina T and Ganea D. PGE2-induced metalloproteinase-9 is essential for dendritic cell migration. Blood, 2008; 111(1): 260-70. [196] Kobayashi Y, et al. Possible involvement of matrix metalloproteinase-9 in Langerhans cell migration and maturation. J Immunol, 1999; 163(11): 5989-93. [197] Lebre MC, et al. Inhibition of contact sensitizer-induced migration of human Langerhans cells by matrix metalloproteinase inhibitors. Arch Dermatol Res, 1999; 291(7-8): 447-52. [198] Ichiyasu H, et al. Matrix metalloproteinase-9-deficient dendritic cells have impaired migration through tracheal epithelial tight junctions. Am J Respir Cell Mol Biol, 2004; 30(6): 761-70. [199] Fulcher JA, et al. Galectin-1 co-clusters CD43/CD45 on dendritic cells and induces cell activation and migration through Syk and protein kinase C signaling. J Biol Chem, 2009; 284(39): 26860-70. [200] de Rooy DP, et al. A genetic variant in the region of MMP-9 is associated with serum levels and progression of joint damage in rheumatoid arthritis. Ann Rheum Dis, 2014; 73(6): 1163-9. [201] Gruber BL, et al. Markedly elevated serum MMP-9 (gelatinase B) levels in rheumatoid arthritis: a potentially useful laboratory marker. Clin Immunol Immunopathol, 1996; 78(2): 161-71. [202] Itoh T, et al. The role of matrix metalloproteinase-2 and matrix metalloproteinase-9 in antibody-induced arthritis. J Immunol, 2002; 169(5): p. 2643-7. [203] Sozzani S. Dendritic cell trafficking: more than just chemokines. Cytokine Growth Factor Rev, 2005; 16(6): 581-92. [204] De Smedt T, et al. Regulation of dendritic cell numbers and maturation by lipopolysaccharide in vivo. J Exp Med, 1996; 184(4): 1413-24. [205] Luther SA, et al. Coexpression of the chemokines ELC and SLC by T zone stromal cells and deletion of the ELC gene in the plt/plt mouse. Proc Natl Acad Sci USA, 2000; 97(23): 12694-9. [206] Nakano H and Gunn MD. Gene duplications at the chemokine locus on mouse chromosome 4: multiple strain-specific haplotypes and the deletion of secondary lymphoid-organ chemokine and EBI-1 ligand chemokine genes in the plt mutation. J Immunol, 2001; 166(1): 361-9. [207] MartIn-Fontecha A, et al. Regulation of dendritic cell migration to the draining lymph node: impact on T lymphocyte traffic and priming. J Exp Med, 2003; 198(4): 615-21.

[208] Ohl L, et al. CCR7 governs skin dendritic cell migration under inflammatory and steady-state conditions. Immunity, 2004; 21(2): 279-88. [209] Saeki H, et al. Cutting edge: secondary lymphoid-tissue chemokine (SLC) and CC chemokine receptor 7 (CCR7) participate in the emigration pathway of mature dendritic cells from the skin to regional lymph nodes. J Immunol, 1999; 162(5): 2472-5. [210] Yoneyama H, et al. Evidence for recruitment of plasmacytoid dendritic cell precursors to inflamed lymph nodes through high endothelial venules. Int Immunol, 2004; 16(7): 915-28. [211] Yen JH, Kong W and Ganea D. IFN-beta inhibits dendritic cell migration through STAT-1-mediated transcriptional suppression of CCR7 and matrix metalloproteinase 9. J Immunol, 2010; 184(7): 3478-86. [212] Yoneyama H, et al. Evidence for recruitment of plasmacytoid dendritic cell precursors to inflamed lymph nodes through high endothelial venules. Int Immunol, 2004; 16(7): 915-28. [213] Kabashima K, et al. CXCL12-CXCR4 engagement is required for migration of cutaneous dendritic cells. Am J Pathol, 2007; 171(4): 1249-57. [214] Qu , et al. Role of CCR8 and other chemokine pathways in the migration of monocyte-derived dendritic cells to lymph nodes. J Exp Med, 2004; 200(10): 1231-41. [215] Rodriguez-Fernandez JL and Corbi AL. Adhesion molecules in human dendritic cells. Curr Opin Investig Drugs, 2005;

6(11): 1103-11.

[216] Dedrick RL, Bodary S and Garovoy MR. Adhesion molecules as therapeutic targets for autoimmune diseases and transplant rejection. Expert Opin Biol Ther, 2003; 3(1): 85-95. [217] Jakubzick C, et al. Modulation of dendritic cell trafficking to and from the airways. J Immunol, 2006; 176(6): 3578-84. [218] Johnson LA, et al. An inflammation-induced mechanism for leukocyte transmigration across lymphatic vessel endothelium. J Exp Med, 2006; 203(12): 2763-77. [219] Xu H, et al. The role of ICAM-1 molecule in the migration of Langerhans cells in the skin and regional lymph node. Eur J Immunol, 2001; 31(10): 3085-93. [220] Ren XX, et al. Antibodies to P-selectin glycoprotein ligand-1 block dendritic cell-mediated enterovirus 71 transmission and prevent virus-induced cells death. Virulence, 2015; 6(8): 802-8. [221] Lammermann T, et al. Rapid leukocyte migration by integrin-independent flowing and squeezing. Nature, 2008; 453(7191): 51-5.

[222] Kakimoto K, et al. The effect of anti-adhesion molecule antibody on the development of collagen-induced arthritis. Cell Immunol, 1992; 142(2): 326-37. [223] Willenborg DO, et al. ICAM-1-dependent pathway is not critically involved in the inflammatory process of autoimmune encephalomyelitis or in cytokine-induced inflammation of the central nervous system. J Neuroimmunol, 1993; 45(1-2): 147-54. [224] Isobe M, et al. Specific acceptance of cardiac allograft after treatment with antibodies to ICAM-1 and LFA-1. Science, 1992; 255(5048): 1125-7. [225] Yacyshyn BR, et al. A placebo-controlled trial of ICAM-1 antisense oligonucleotide in the treatment of Crohn's disease. Gastroenterology, 1998; 114(6): 1133-42. [226] Issekutz AC, et al. E-selectin, but not P-selectin, is required for development of adjuvant-induced arthritis in the rat. Arthritis Rheum, 2001; 44(6): 1428-37. [227] Bhushan M, et al. Anti-E-selectin is ineffective in the treatment of psoriasis: a randomized trial. Br J Dermatol, 2002; 146(5): 824-31. [228] Tubridy N, et al. The effect of anti-alpha4 integrin antibody on brain lesion activity in MS. The UK Antegren Study Group. Neurology, 1999; 53(3): p. 466-72. [229] Podolsky DK, et al. Attenuation of colitis in the cotton-top tamarin by anti-alpha 4 integrin monoclonal antibody. J Clin Invest, 1993; 92(1): 372-80. [230] Spiegel S and Milstien S. The outs and the ins of sphingosine-1-phosphate in immunity. Nat Rev Immunol, 2011; 11(6): 403-15. [231] Sekiguchi M, et al. Role of sphingosine 1-phosphate in the pathogenesis of Sjogren's syndrome. J Immunol, 2008; 180(3): 1921-8. [232] Graler MH and Goetzl EJ. The immunosuppressant FTY720 down-regulates sphingosine 1-phosphate G-protein-coupled receptors. FASEB J, 2004; 18(3): 551-3. [233] Idzko M, et al. Local application of FTY720 to the lung abrogates experimental asthma by altering dendritic cell function. J Clin Invest, 2006; 116(11): 2935-44. [234] Lan YY, et al. Sphingosine 1-phosphate receptor agonism impairs skin dendritic cell migration and homing to secondary lymphoid tissue: association with prolonged allograft survival. Transpl Immunol, 2008; 20(1-2): 88-94. [235] Reines I, et al. Topical application of sphingosine-1-phosphate and FTY720 attenuate allergic contact dermatitis reaction through inhibition of dendritic cell migration. J Invest Dermatol, 2009;

129(8): 1954-62. [236] Han Y, Li X and Sun E. FTY720 Abrogates Collagen-Induced Arthritis by Hindering Dendritic Cell Migration to Local Lymph Nodes. J Immunol, 2015; 195:4126-4135. [237] Lan YY, et al. Sphingosine 1-phosphate receptor agonism impairs skin dendritic cell migration and homing to secondary lymphoid tissue: association with prolonged allograft survival. Transpl Immunol, 2008; 20(1-2): 88-94. [238] Randolph GJ, Angeli V and Swartz MA. Dendritic-cell trafficking to lymph nodes through lymphatic vessels. Nat Rev Immunol, 2005; 5(8): 617-28. [239] Dumitriu IE, et al. The secretion of HMGB1 is required for the migration of maturing dendritic cells. J Leukoc Biol, 2007; 81(1): 84-91. [240] Legler DF, et al. Prostaglandin E2 is generally required for human dendritic cell migration and exerts its effect via EP2 and EP4 receptors. J Immunol, 2006; 176(2): 966-73. [241] Faure-Andre G, et al. Regulation of dendritic cell migration by CD74, the MHC class II-associated invariant chain. Science, 2008; 322(5908): 1705-10. [242] Adhikary S, et al. Signaling through cannabinoid receptor 2 suppresses murine dendritic cell migration by inhibiting matrix metalloproteinase 9 expression. Blood, 2012; 120(18): 3741-9. [243] Han S, et al. Auranofin, an immunosuppressive drug, inhibits MHC class I and MHC class II pathways of antigen presentation in dendritic cells. Arch Pharm Res, 2008; 31(3):370-6. [244] Salvi V, et al. Trichostatin A blocks type I interferon production by activated plasmacytoid dendritic cells. Immunobiology, 2010; 215(9-10):756-61. [245] Yoshimura M, et al. Immunological effects of Oenothein B, an ellagitannin dimer, on dendritic cells. nt J Mol Sci, 2012 20;14(1):46-56. [246] Kubo S, et al. The JAK inhibitor, tofacitinib, reduces the T cell stimulatory capacity of human monocyte-derived dendritic cells. Ann Rheum dis, 2014; 73:2192-2198. [247] Platt AM, et al. The active metabolite of spleen tyrosine kinase inhibitor fostamatinib abrogates the CD4+T cell-priming capacity of dendritic cells. Rheumatology (Oxford), 2015;54(1):169-77. [248] Chorny A, et al. Vasoactive intestinal peptide induces regulatory dendritic cells with therapeutic effects on autoimmune disorders. Proc Natl Acad Sci USA,2005; 102(38):13562-7. [249] Li G, Kim YJ, and Broxmeyer HE. Macrophage colony-stimulating factor drives cord blood monocyte differentiation into IL-10(high)IL-12absent dendritic cells with tolerogenic potential. J

Immunol, 2005;174:4706 -17. [250] Brandt K, Bulfone-Paus S, Foster DC and Ruckert R. Interleukin-21 inhibits dendritic cell activation and maturation. Blood, 2003;102:4090-08. [251] Menges M, et al. Repetitive injections of dendritic cells matured with tumor necrosis factor alpha induce antigen-specific protection of mice from autoimmunity. J Exp Med, 2002;195:15-21. [252] Yen JH, Kong W and Ganea D IFN-beta inhibits dendritic cell migration through STAT-1-mediated transcriptional suppression of CCR7 and matrix metalloproteinase 9. J Immunol, 2010; 184:3478-3486.

Figure legend Fig. 1. Targeting dendritic cell (DC) functions for treatment of autoimmune diseases. After generated from haematopoietic stem cell in the bone marrow, DCs are generally immature and traffic to the peripheral tissues, where they undergo several molecular events, namely differentiation, endocytosis, phenotypic and functional maturation, migration to the secondary lymphoid organs and present acquired antigens to naive T lymphocytes. And each event was regulated subtly by various molecules, signals, and mechanisms. HSC, haematopoietic stem cell; iDCs, immature dendritic cells; mDCs mature dendritic cells; LNs, Lymph nodes; ECM, extracellular matrixc, ECM.

Table 1. Representative immunomodulatory targets and their effects on DC functions. Agents

Species

DC origin

Vitro/vivo

DC functions

Targeted Diseases

References

[12, 22]

Study Differentiation

Dexamethasone

human

moDCs

in vitro

low levels of CD1a

Phenotypic

Cytokine

Migration

T Cell

maturation

production

MHC-II↓

TNF-α↓

CCL3↓

Less Th1/Th2

Immune-mediated diseases

stimulation

And other

CD40↓

IL-12↓

CCL2↓

cell generation,

Corticosteroids

CD86↓

IL-1β↓

CCL5↓

More Treg cell

CCL28↓

generation

CD54↓

CCL22↓ Vitamin D3

mouse

human

Dexamethasone +

mouse

BMDCs

moDCs

BMDCs

in vivo

in vitro

in vitro

(-)

low levels of CD1a

(-)

Vitamin D3

MHC-II↓

TNF-α↓

CCL3↑

More Treg cell

EAE,

CD80↓

IL-10↑

CCL2↑

generation

Autoimmunity

MHC-II↓

IL-10↑

CCL5↓

Less Th1/Th2

MS,

CD40↓

IL-12↓

CCL18↑

cell generation,

Autoimmunity

CD83↓

TNF-α↓

CCL22↓

More Treg cell

CD86↓

IL-6↓

[13, 14]

[15]

generation

MHC-II↓

IL-12↓

ICAM-1↓

Inhibit

CD40↓

INF-γ↓

CCL3↑

stimulation,

diseases

CD80↓

CCL2↑

CD86↓

CCL5↓

T-cell

Immune-mediated

[19,22]

CCR1,2,5,7↓ human

moDCs

in vitro pre-clinical

(-)

MHC-II↓

IL-10↑

Inhibit T-cell

graft rejection;

CD40↓

IL-12↓

stimulation,

Rheumatoid

CD83↓

TNF-α↓

arthitis; Sjogren

CD86↓

IL-6↓

syndrome

[16-21]

Agents

Species

DC origin

Vitro/vivo

DC functions

Targeted Diseases

References

[21]

Study Differentiation

Phenotypic

Cytokine

maturation

production

Migration

T Cell stimulation

MHC-II↓

IL-10↑

Less Th1

Rheumatoid

Dexamethasone

CD40↓

IL-12↓

cell generation,

arthritis;

+monophosphoryl

CD80↓

TNF-α↓

More Treg cell

Immune-mediated

lipid A

CD83↓

IL-23↓

generation

diseases;

human

moDCs

in vitro

(-)

CCL19↓,

Prevention of graft

CD86↓

rejection Rapamycin

mouse

BMDCs

in vitro

GM-CSF↓,

MHC-II↓

IL-12↓

IL-4R↓,

CD40↓

TNF-α↓

FLT3L↓

CD80↓

(-)

More Treg cell

Prevention of graft

generation

rejection

Reduce survival

Prevention of graft

of alloantigen

rejection

[23-25]

CD86↓ mouse

BMDCs

in vivo

(-)

(-)

(-)

(-)

[26]

specific CD8+ T cells human

moDCs

in vitro

(-)

CD40↓

(-)

(-)

CD80↓

More Treg cell

Immune-mediated

generation

diseases

[27]

CD86↓ Sanglifehrin A

human

moDCs

in vitro

(-)

(-)

IL-12↓ IL-23↓

(-)

(-)

Immune-mediated diseases

[131]

Agents

Species

DC origin

Vitro/vivo

DC functions

Targeted Diseases

References

Inhibit T-cell

Immune-mediated

[74]

TNF-α↓

stimulation

diseases

IL-12↓

Lower Th1

Immune-mediated

TNF-α↓

cell generation,

diseases

Study Differentiation

Cyclosporin A

mouse

human

BMDCs

moDCs

in vitro

in vitro

(-)

(-)

Phenotypic

Cytokine

maturation

production

(-)

IL-12↓

(-)

Migration

T Cell stimulation

CCR7↓

[73]

More Treg cell generation Mycophenolate

mouse

BMDCs

in vitro

(-)

CD40↓

IL-12↓

ICAM-1↓

CD80↓

Inhibit T-cell

Immune-mediated

stimulation

diseases

[32]

CD86↓ human

Tacrolimus

mouse

moDCs

BMDCs

in vitro

in vivo

(-)

(-)

CD54↓

IL-10↓

Inhibit T-cell

Immune-mediated

CD80↓

IL-12↓

stimulation

diseases

CD83↓

TNF-α↓

CD86↓

IL-18↓

(-)

(-)

More Treg cell

CIA

[130]

More Treg cell

Rheumatoid

[130]

generation

arthritis

More Treg cell

CIA

generation

MS

(-)

(FK-506)

[33]

generation human

moDCs

in vitro

(-)

MHC-II↓

TNF-α↓

CD40↓

IL-10↑

(-)

CD80↓ CD86↓ FTY720

mouse

BMDCs

in vivo

(-)

(-)

IL-12↓

CCR7↓

[236, 237]

Agents

Species

DC origin

Vitro/vivo

DC functions

Targeted Diseases

References

Inhibit T-cell

Immune-mediated

[134, 135]

stimulation

diseases

(-)

Immune-mediated

Study Differentiation

chloroquine

mouse

BMDCs

in vitro

(-)

Phenotypic

Cytokine

maturation

production

CD80↓

IL-10↓

CD86↓

IL-12↓

Migration

T Cell stimulation

(-)

TNF-α↓ Aspirin

human

moDCs

in vitro

(-)

CD83↓

IL-10↓

(-)

IL-12↓

[30]

diseases

TNF-α↑ mouse

Butyric acid

human

BMDCs

moDCs

in vitro

in vitro

promoted the

MHC-II↓

generation of CD11c+

CD40↓

DC;Phagocytosis

CD80↓

inhibition;

CD86↓

Differentiation

MHC-II↓

Inhibition;

CD40↓

reducing the phagocytic

CD86↓

capacity

CD83↓

IL-12↓

(-)

(-)

(-)

Inhibit T-cell

Immune-mediated

stimulation

diseases

Inhibit T-cell

Immune-mediated

stimulation

diseases

Inhibit T-cell

Immune-mediated

stimulation

diseases

Reduce T cell

Type-1 Diabetes

[28, 29]

[36]

CD86↓ N-acetyl-L-cystei

human

moDCs

in vitro

ne

(-)

MHC-II↓

IL-12↓

CD40↓

IL-6↓

CD86↓

IL-8↓

(-)

[31]

TNF-α↓ Troglitazone

human

moDCs

in vitro

(-)

priming

[17]

Agents

Species

DC origin

Vitro/vivo

DC functions

Targeted Diseases

References

Reduce T cell

Autoimmunity;

[37]

priming

EAE Type-1 Diabetes

[38]

Immune-mediated

[132]

Study Differentiation

Rosiglitazone

mouse

BMDCs

in vitro

(-)

Phenotypic

Cytokine

maturation

production

(-)

(-)

Migration

T Cell stimulation

(-)

human

moDCs

in vitro

CD1a↓

CD80↓

(-)

(-)

Reduce T cell

human

moDCs

in vitro

(-)

CD80↓

(-)

CCR7↓

(-)

priming leflunomide

CD86↓ Triptolide

Apigenin

human

mouse

mouse

moDCs

BMDCs

BMDCs

in vitro

in vitro

In vivo

CD1a↓

(-)

(-)

diseases

MHC-II↓

IL-12↓

CCR7↓,

Reduce T cell

Immune-mediated

CD40↓

TNF-α↓

CCR5↑,

priming;

diseases

CD80↓

IL-10↓

COX-2↓,

More Treg cell

CD86↓

TGF-β↓

MHC-II↓

IL-12↓

CD40↓,

TNF-α↓

CD80↓,

IL-10↓

CD86↓,

IL-1β↓

MHC-II↓

TNF-α↓

CD40↓

IL-6↓

CD80↓

IL-1β↓

[190]

generation CXCR4↓

(-)

(-)

[191]

CXCR4↓

(-)

CIA

[191]

[75]

CD86↓ Quercetin

mouse

BMDCs

in vitro

reducing the phagocytic

MHC-II↓

IL-12↓

CCL3↓

Reduce T cell

Immune-mediated

capacity

CD40↓

TNF-α↓

CCL2↓

priming

diseases

CD80↓

IL-10↓

CCL5↓

CD86↓

IL-1β↓

Agents

Species

DC origin

Vitro/vivo

DC functions

Targeted Diseases

References

EAE

[151]

EAE

[72]

[242]

Study Differentiation

Chrysin

human

moDCs

In vivo

(-)

Phenotypic

Cytokine

maturation

production

MHC-II↓

IFN-γ↓

CD83↓

IL-12↓

Migration

T Cell stimulation

(-)

Less Th1 cell generation,

CD86↓

More Treg cell generation

Fumarates

mouse

BMDCs

in vitro

(-)

(-)

IL-12↓

More Treg cell

More Treg cell

IL-23↓

generation

generation

(-)

Reduce T cell

Immune-mediated

priming;

diseases

Reduce T cell

rheumatoid

priming;

arthritis;

IL-10↑ Auranofin

mouse

BMDCs

in vitro

inhibition of the

MHC-I↓

phagocytic activity of

MHC-II↓

(-)

DCs Rabeximod

human

moDCs

in vitro

CD1a↓,

MHC-I↓

IL-6↓

(-)

MHC-II↓

n-3 fatty acid

human

moDCs

in vitro

CD1a↓,

CD80↓

Immune-mediated

CD40↓

diseases

(-)

IL-6↓

(-)

(-)

Immune-mediated

[39]

[40]

diseases Glatiramer acetate

mouse

BMDCs

In vivo

(-)

(-)

IL-10↑

CCL2↓ CCL3↓ CCL5↓ IP-10↓

(-)

EAE

[152]

Agents

Species

DC origin

Vitro/vivo

DC functions

Targeted Diseases

References

Reduce T cell

Immune-mediated

[133]

priming;

diseases,

Study Differentiation

IVIg

human

moDCs

in vitro

CD1a↓

Phenotypic

Cytokine

maturation

production

MHC-II↓

TNF-α↓

CD83↓

IL-12↓

CD86↓

IL-10↑

Migration

T Cell stimulation

(-)

transplantation

CD40↓ Bay 11-7082

human

(NF-κB inhibitor)

human

in vitro

(-)

umbilical

CD83↓

IL-8↓

(-)

(-)

CD86↓

Immune-mediated

[121]

diseases

cord blood-DCs mouse

BMDCs

In vivo

(-)

(-)

(-)

(-)

Reduce T cell

CIA

[118-120]

Reduce T cell

SKG mice from

[243]

priming;

getting arthritis

response MHC-II↓

IL-6↓

(histone

CD40↓

IL-12↓

deacetylase

CD80↓

Trichostatin A

mouse

BMDCs

in vitro

(-)

mouse

BMDCs

In vivo

(Flt3 inhibitor) Oenothein B

Lupus

CD86↓

inhibitor) Sunitinib

(-)

Reduce CD11c+

(-)

(-)

(-)

(-)

population mouse

BMDCs

in vitro

CD1a↓,

antigen-induced

[49]

arthritis CD83↓

IL-6↓ IL-1β↓

(-)

(-)

inflammatory bowel disease, celiac disease, rheumatoid arthritis.

[244]

Agents

Species

DC origin

Vitro/vivo

DC functions

Targeted Diseases

References

Reduce T cell

rheumatoid

[137]

priming;

arthritis

Reduce T cell

rheumatoid

priming;

arthritis

Study Differentiation

Infliximab

human

moDCs

In vivo

(TNF-α blocker)

Less CD11c+ DCs

Phenotypic

Cytokine

maturation

production

(-)

Migration

T Cell stimulation

(-)

CD123+ DCs percentages

Tocilizumab

human

(-)

In vivo

(IL-6 blocker) Tofacitinib

reduction of circulating

(-)

IL-6↓,\

(-)

myeloid DCs human

moDCs

in vitro

(-)

CD80↓ CD86↓

IL-1β↓

mouse

BMDCs

in vitro

(-)

(-)

IFN-α↓

Chemokines

Reduce T cell

Immune-mediated

(spleen tyrosine

IL-2↓

IP-10↓

co-stimulator

diseases

kinase inhibitor)

IL-5↓

CCL3↓

(ICOS) and

( JAK inhibitor) Fostamatinib

IL-6↓

rheumatoid

[138] [245]

arthritis [246]

PD-1

IL-6↓ IL-13↓ IL-17↓ Vasoactive

mouse

BMDCs

in vitro;

endocytosis↑,

intestinal peptide

MHCⅡ↓

TNF-α↓

More Treg cell

Immune-mediated

CD40↓

IL-12↓

Generation, and

diseases

CD80↓

IL-6↓

Reduce T cell

CD86↓

IL-10↑

priming

(-)

[247]

CD83↓ TGF-β

mouse

BMDCs

In vivo

(-)

MHCⅡ↓

TNF-α↓

CD40↓

IL-12↓

CD80↓

IL-6↓

CD86↓

IL-1β↓

(-)

Reduce T cell

Type 1 Diabetes;

priming

Prevention of graft rejection

[146, 147]

Agents

Species

DC origin

Vitro/vivo

DC functions

Targeted Diseases

References

Reduce T cell

Systemic Lupus

[139-144]

priming;

Erythematosus;

Study Differentiation

IL-10

human

moDCs

Phenotypic

Cytokine

maturation

production

in vitro

CD1a↓,

MHC-Ⅱ↓

IL-12↓

pre-clinical

DC-SIGN↑,

MHC- ↓

IL-6↓

CD83↓

TNF-α↓

Migration

T Cell stimulation

CCR7↓

Type 1 Diabetes;

CD80↓

Immune-mediated

CD86↓

diseases; Asthma and allergy

rat

BMDCs

in vivo

(-)

CD86↓

(-)

(-)

Reduce T cell

Prevention of graft

priming and

rejection

[171]

allospecific T cell response mouse

BMDCs

(-)

MHCⅡ↓

IL-6↓

MHCⅠ↓

IL-12↓

B7-1↓

IL-23↓

(-)

Reduce T cell

delayed-type

priming

hypersensitivity

Reduce T cell

Anti-phospholipid

priming

syndrome;

[168]

B7-2↓ IL-10+TGF-β

human

moDCs

in vitro;

CD1a↑

pre-clinical

MHCⅡ↓

IL-12↓

CD40↓

IL-10↑

(-)

[145, 148]

Type 1 Diabetes

CD83↓ CD86↓ M-CSF

human

moDCs

in vitro;

CD1a↓,

MHCⅡ↓

TNF-α↓

CD40↓

IL-12↓ IL-10↑

CXCR4↓

Reduce T cell

Immune-mediated

priming

diseases

[248]

Agents

Species

DC origin

Vitro/vivo

DC functions

Targeted Diseases

References

Reduce T cell

Immune-mediated

[249]

priming

diseases

More Treg cell

EAE

Study Differentiation

IL-21

human

moDCs

in vitro;

endocytosis↑,

Phenotypic

Cytokine

maturation

production

MHCⅡ↓

TNF-α↓

CD80↓

IL-12↓

Migration

T Cell stimulation

CCR7↓

IL-6↓ IL-1β↓ TNF-α

IFN-β

mouse

mouse

BMDCs

BMDCs

In vivo

in vitro

(-)

(-)

CD40↑

IL-12↓

CD80↑

IL-6↓

Generation, and

CD86↑

IL-1β↓

Reduce T cell

IL-10↑

priming

CD40↑ CD80↑

(-)

(-)

CCR7↓

[250]

[251]

MMP-9↓

CD86↑

moDC, monocyte-derived DC; BMDC, bone marrow-derived DC; MHC II, major histocompatibility class II molecules; IL: interleukin; TNF: tumor necrosis factor; IFN: interferon; TGF:transforming growth factor; ICAM, intercellular cell adhesion molecule; CIA: collagen-induced arthritis; EAE: experimental autoimmune encephalomyelitis; MS: multiple sclerosis.

Sniping the scout: Targeting the key molecules in dendritic cell functions for treatment of autoimmune diseases.

Dendritic cells (DCs) are a power tool for manipulating immune system. They play important roles in the induction of immunity as well as inducing intr...
526KB Sizes 0 Downloads 9 Views