Co-stimulatory and Co-inhibitory Pathways in Autoimmunity. - PDF Download Free (2024)

HHS Public Access Author manuscript Author Manuscript

Immunity. Author manuscript; available in PMC 2017 May 17. Published in final edited form as: Immunity. 2016 May 17; 44(5): 1034–1051. doi:10.1016/j.immuni.2016.04.017.

Co-stimulatory and co-inhibitory pathways in autoimmunity Qianxia Zhang1 and Dario A.A. Vignali1,2,* 1Department 2Tumor

of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15261

Microenvironment Center, University of Pittsburgh Cancer Institute, Pittsburgh, PA 15232,

USA

Author Manuscript

Abstract

Author Manuscript

The immune system is guided by a series of checks and balances, a major component of which is a large array of co-stimulatory and co-inhibitory pathways that modulate the host response. While co-stimulation is essential to boost and shape the initial response following signaling through the antigen receptor, inhibitory pathways are also critical to modulate the immune response. Excessive co-stimulation and/or insufficient co-inhibition can lead to a breakdown of self-tolerance, leading to autoimmunity. In this review, we will focus on the role of co-stimulatory and co-inhibitory pathways in two systemic (Systemic Lupus Erythematosus and Rheumatoid Arthritis) and two organ-specific (Multiple Sclerosis and Type 1 Diabetes) autoimmune diseases that are emblematic. We will also discuss how mechanistic analysis of these pathways has led to the identification of potential therapeutic targets and initiation of clinical trials for autoimmune diseases, as well as outline some of the challenges that lie ahead.

Introduction

Author Manuscript

Immunological self-tolerance is the unresponsiveness of the immune system to self-antigens (self-Ags). A breakdown in immune homeostasis and self-tolerance leads to autoimmunity, resulting in deleterious inflammation in, and destruction of, self-tissues mediated by autoreactive T cells and autoantibodies (auto-Abs) (Goodnow et al., 2005; Schwartz, 1989). In order to prevent autoimmunity, an intricate series of molecular checks and balances helps to ensure that the immune system produces a measured and appropriate response to foreign threats while avoiding host tissue pathology and destruction. However, emerging observations suggest that these control mechanisms are subverted in autoimmunity, providing underlying mechanistic insight while also pointing to potential avenues for therapeutic intervention. The Two-Signal model proposes that activation of naïve T cells requires both T cell receptor (TCR) stimulation by MHC:peptide complexes [Signal 1] and co-stimulation via costimulatory receptors and their corresponding ligands on antigen presenting cells (APCs)

*

Correspondence to: ; Email: [emailprotected]. Publisher's Disclaimer: 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 citable 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.

Zhang and Vignali

Page 2

Author Manuscript Author Manuscript

[Signal 2] (Lafferty and Cunningham, 1975; Mueller et al., 1989). For instance, one of the most prominent co-stimulatory pathways is the CD28:B7 axis that amplifies TCR signaling and interleukin-2 (IL2) production to promote T cell proliferation and survival. In order to provide a mechanism to turn off T cell activation, co-inhibitory receptors are induced by TCR stimulation and co-stimulation and subsequently transduce feedback signals that dampen the ascending co-stimulatory signals. Therefore, the net outcome of TCR stimulation is modified by both co-stimulatory and co-inhibitory receptors. Both sets of receptors are expressed by all T cell subsets thereby helping to shape the overall immune response. For instance, they are also expressed by, and have critical impact on, regulatory T (Treg) cells, an immunosuppressive population that plays a pivotal role in self-tolerance (Sakaguchi et al., 2008; Vignali et al., 2008). Excessive co-stimulation and/or inadequate coinhibition results in aberrant T cell activation, which can result in a breakdown of selftolerance by activating and expanding autoreactive T cells. Similarly, B cells and other immune cells also require two signals for their activation, maturation and function (Bretscher and Cohn, 1970). Therefore, the immune response is fundamentally shaped and modulated by co-stimulatory and co-inhibitory receptors and their corresponding ligands. Disruption of the balance between co-stimulation and co-inhibition unleashes self-reactivity leading to autoimmune disease.

Author Manuscript

While co-stimulatory and co-inhibitory pathways have a significant impact on all autoimmune diseases, in the interest of brevity, in this review we will focus on their role in two systemic (Systemic Lupus Erythematosus and Rheumatoid Arthritis) and two organspecific (Multiple Sclerosis and Type 1 Diabetes) autoimmune diseases of major importance and interest that are emblematic of other autoimmune diseases. More general aspects of the role of these pathways in T cell development and function, and in other diseases have been discussed in other reviews ([Au: with this statement, do you want to call out all the other pieces? Sharpe, Kuchroo, Bluestone, Ware, Wherry, Ford, Wolchok) We will also discuss how mechanistic analysis of co-stimulatory and co-inhibitory pathways using a wide variety of animal models and human studies has led to the identification of potential therapeutic targets and initiation of clinical trials for autoimmune diseases, as well as outline some of the challenges that lie ahead.

Systemic Lupus Erythematosus

Author Manuscript

Systemic Lupus Erythematosus (SLE) is a systemic autoimmune disorder associated with the presence of anti-nuclear antibodies (Abs) and the combinatorial clinical manifestations of rash, thrombocytopenia, serositis, and nephritis (Lisnevskaia et al., 2014). Lupus nephritis (LN, glomerulonephritis) is a key feature of SLE, marked by inflammation of, and auto-Ab accumulation in, the kidney. The dysregulation of B and T cell activation leads to auto-Ab production, immune complex (IC) formation, and multi-organ damage in SLE (Lisnevskaia et al., 2014). ICs are central players in tissue damage in SLE, and T cells are critical participants in the breakdown of B cell tolerance. Follicular helper T (Tfh) cells are professional helper cells that facilitate germinal center (GC) formation, B cell development, and B cell receptor (BCR) affinity maturation (Crotty, 2014). Aberrant Tfh cell differentiation and activation drives the pathogenesis of many systemic autoimmune diseases including SLE. Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 3

Author Manuscript

The spontaneous murine lupus models, F1 hybrid of New Zealand Black and New Zealand White (NZB/NZW.F1) and MRL mice hom*ozygous for the lymphoproliferation gene (MRL-lpr), are commonly used to study the role of co-stimulatory and co-inhibitory pathways in SLE. NZB/NZW.F1 mice develop IC-mediated glomerulonephritis with elevated anti-nuclear Ab titers, representing a model of the chronic lupus disorder (Crampton et al., 2014). MRL-lpr mice exhibit a more severe lupus-like syndrome with B cell hyperactivity, circulating ICs, and a wide range of auto-Abs (Crampton et al., 2014). Other mouse models of SLE have been developed but will not be discussed here.

Author Manuscript Author Manuscript

Several co-stimulatory pathways have been shown to impact the development of lupus. The co-stimulatory receptor CD28 is activated by its ligands B7.1 (CD80) and B7.2 (CD86), and couples with TCR signaling to promote T cell proliferation and survival during T cell priming. Cd28−/− MRL-lpr mice exhibit delayed and diminished glomerulonephritis and an absence of renal vasculitis and arthritis (Tada et al., 1999a), implying that blocking CD28:B7 interactions may be a potential treatment for autoimmune lupus. However, B7.1 and B7.2 appear to function in a redundant manner, as treatment with both anti-B7.1 and anti-B7.2 significantly reduced auto-Ab production whereas blocking either B7.1 or B7.2 alone had a minimal effect on auto-Ab levels in MRL-lpr mice (Liang et al., 1999). Interestingly, Cd80−/− but not Cd86−/− MRL-lpr mice exhibited more severe glomerulonephritis than wild-type controls (Liang et al., 1999), raising the possibility that B7.1 also has inhibitory activity in this setting which could either be due to preferential interactions with the co-inhibitory receptor CTLA4 (CD152) over B7.2, or due to interactions with another co-inhibitory receptor PD-L1 (B7-H1, CD274) (Butte et al., 2007). CTLA4Ig, a fusion protein composed of the murine CTLA4 extracellular domain and IgG2a Fc, inhibits CD28:B7 interactions by binding to B7 molecules with a higher affinity. CTLA4Ig administration prolonged the survival of NZB/NZW.F1 mice by preventing autoAb production, limiting immunoglobulin class switching and somatic mutation, and reducing CD4+ T cell activation (Finck et al., 1994; Mihara et al., 2000). However, clinical trials of Abatacept [a fusion protein of the human CTLA4 extracellular domain and IgG1 Fc] in SLE patients have yet to exhibit any clear efficacy, although it was well tolerated (Furie et al., 2014; Group, 2014; Merrill et al., 2010).

Author Manuscript

Even though a protective effect was seen in Cd28−/− MRL-lpr mice, the accumulation of autoreactive T cells was unaffected (Tada et al., 1999a), implying that additional T cell costimulatory pathways are exploited in SLE. The co-stimulatory receptor ICOS (CD278) is essential for T cell activation and promotes humoral immunity (Dong et al., 2001). The generation, function and maintenance of Tfh and extrafollicular helper T cells that help GC formation, B cell maturation and IgG production are ICOS-dependent in both NZB/NZW.F1 and MRL-lpr mice (Hu et al., 2009; Odegard et al., 2008; Teichmann et al., 2015). Given the importance of ICOS in promoting SLE pathogenesis, the safety and efficacy of AMG 557, a human Ab that targets the ICOS ligand (ICOSL, B7-H2, CD275), is currently being evaluated in clinical trials with SLE patients. Members of the tumor necrosis factor superfamily (TNFSF) and tumor necrosis factor receptor superfamily (TNFRSF) play a dominant role in the pathogenesis of many autoimmune diseases. The interaction between B and T cells via the CD40

Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 4

Author Manuscript Author Manuscript

(TNFRSF5):CD40L (TNFSF5, CD154) axis provides critical activation signals for thymusdependent humoral immunity in SLE. Loss of CD40:CD40L signaling ameliorated disease severity and prolonged survival of both NZB/NZW.F1 and MRL-lpr mice (Early et al., 1996; Ma et al., 1996). Signals transduced by co-stimulatory receptors and their corresponding ligands-receptors are often reciprocal. Indeed, ligation of CD40L with CD40 not only activates T cells but also promotes B cell class switching and somatic mutation in NZB/ NZW.F1 mice (Wang et al., 2003). However, anti-CD40L in SLE clinical trials has exhibited mixed results thus far (Boumpas et al., 2003; Kalunian et al., 2002). It is possible that redundant pathways are involved in promoting auto-Ab production in SLE, as indicated by the existence of isotype switching in Cd154−/− MRL-lpr mice (Ma et al., 1996). AntiCD40L blockade in combination with CTLA4Ig in NZB/NZW.F1 mice showed synergistic effects in the delay of lupus onset by suppressing both autoreactive B and T cells (Daikh et al., 1997; Wang et al., 2002), suggesting that combinatorial approaches might enhance efficacy.

Author Manuscript

Other co-stimulatory pathways are also involved in the pathogenesis of SLE. Polymorphism at TNFSF4 (OX40L, CD252) is associated with abnormal expression in SLE patients, and has been fine mapped to one of the SLE-susceptibility loci (Cunninghame Graham et al., 2008; Manku et al., 2013). It was suggested that the OX40 (TNFRSF4, CD134):OX40L costimulation axis contributes to SLE pathogenesis by promoting Tfh responses (Jacquemin et al., 2015). Interestingly, another TNFRSF member, 4-1BB (TNFRSF9, CD137), delivers costimulatory signals to T cells, but paradoxically a protective role for 4-1BB has been suggested in murine lupus models. Cd137−/− MRL-lpr mice exhibited pronounced lymphadenopathy and early mortality with increased CD4+ T cell number and B cell function (Vinay et al., 2007). Treatment with 4-1BB agonist Ab resulted in IFNγ-dependent depletion of autoreactive B cells, reduced auto-Ab production and prolonged survival of MRL-lpr mice (Sun et al., 2002a). In contrast, agonistic anti-4-1BB treatment of NZB/ NZW.F1 mice did not alter the frequency of T or B cells, but suppressed GC formation and anti-dsDNA IgG production (Foell et al., 2003). These studies suggest that 4-1BB agonist could be a potential therapeutic agent for SLE, although additional studies will be required to fully understand its impact in different models of SLE.

Author Manuscript

While aberrant co-stimulation promotes the development of SLE, co-inhibitory pathways are required to limit SLE pathogenesis. PD1 (CD279) is the most studied co-inhibitory receptor in SLE and has two ligands, PD-L1 (B7-H1, CD274) and PD-L2 (B7-DC, CD273). Polymorphisms at PDCD1 have been reported to be associated with susceptibility to SLE (Okazaki and Honjo, 2007). It has been suggested that Pdcd1−/− C57BL/6J mice have elevated levels of IgG2b, IgA, and IgG3 in their sera and reduced expression of CD5, a negative regulator of B1 cell activation (Nishimura et al., 1998). Aged Pdcd1−/− C57BL/6J mice spontaneously developed lupus-like autoimmune symptoms and introduction of the lpr mutation facilitated lupus onset (Nishimura et al., 1999). Controversially, while blockade with anti-PD-L1 accelerated LN onset, PD1 blockade limited LN in NZB/NZW.F1 mice and promoted the suppressive capacity of both CD4+ and CD8+ Treg cells (Kasagi et al., 2010; Wong et al., 2013; Wong et al., 2010). Interestingly, it has been suggested that the PD1 pathway regulates Tfh-mediated humoral immunity by suppressing follicular regulatory T

Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 5

Author Manuscript

(Tfr) cells (Sage et al., 2013), raising the possibility that PD1 blockade is mitigating lupus manifestations by preferentially impacting PD1 function on Tfr cells.

Author Manuscript

A protective role for other co-inhibitory receptors of the immunoglobulin superfamily (IgSF) has also been suggested in SLE. For instance, increased serum concentrations of soluble CTLA4 were found in SLE patients (Saverino et al., 2010). A polymorphism at the CTLA4 promoter was also reported to be associated with SLE susceptibility in an Asian population (Taha Khalaf et al., 2011). Overexpression of an alternatively spliced CTLA4 isoform lacking both the ligand binding domain and the transmembrane domain (1/4CTLA4), encoded by exons 2 and 3, precipitated lupus onset in MRL-lpr mice (Ichinose et al., 2013). A recent study has shown that CTLA4 controls humoral responses by modulating Tfh, Tfr and Treg cells (Sage et al., 2014), raising the possibility that expression of CTLA4 on these T cell subsets is essential to prevent SLE. BTLA (CD272) is another IgSF co-inhibitory receptor. Btla−/− MRL-lpr mice exhibited exacerbated lupus-associated disorders with elevated auto-Ab production and massive organ infiltration of inflammatory cells (Oya et al., 2011). These studies suggest that co-inhibitory pathways are utilized to protect against autoimmunity in SLE despite the malfunctioned co-stimulatory pathways.

Author Manuscript

It is noteworthy that co-stimulatory receptors of the signaling lymphocyte activation molecule family (SLAMF) have been of considerable interest recently in SLE due to their critical role in Tfh cell function (Crotty, 2014). Slamf6 (Ly108, CD352) is encoded within the NZW-derived sle1 lupus-susceptibility locus (Crampton et al., 2014). Signaling via SLAMF6 promoted Th1 cytokine production by T cells from SLE patients (Chatterjee et al., 2011). It has been suggested that SLAMF6 transduces co-inhibitory signals by binding with SHP-1, while the Tfh cell help to B cells is dependent on SLAMF6 binding with SLAMassociated protein (SAP) (Kageyama et al., 2012). Interestingly, the protective SLAMF6 isoform was not present in the lupus-prone congenic mice (Keszei et al., 2011), raising the possibility that SLE patients are more likely to express the isoform that preferentially binds SAP. The role of other SLAMF receptors has also been suggested in both murine models and SLE patients (Brown et al., 2011; Koh et al., 2011). However, further studies need to be performed to fully understand the impact of SLAM family receptors in SLE. While the full impact of co-inhibitory pathways in SLE is less well understood, costimulatory signals do appear to be exacerbated in this disease (Figure 1). Given the major contribution of autoreactive B and Tfh cells in SLE pathogenesis, co-stimulatory and coinhibitory pathways that affect B and Tfh cell differentiation and functions (such as the ICOS, CD40 and SLAM pathways) are likely to have a major impact on SLE development, and thus may be potential therapeutic targets in the treatment of SLE.

Author Manuscript

Rheumatoid Arthritis Rheumatoid Arthritis (RA) is a systemic autoimmune disease characterized by auto-Ab production, synovial inflammation and hyperplasia, cartilage and bone destruction in multiple joints, and cardiovascular and pulmonary disorders (Burmester et al., 2014). The early stages of RA present with local inflammation in the joints, which subsequently develops into a systemic disorder due to the loss of immune tolerance. Pathogenic T cells

Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 6

Author Manuscript

not only help B cells produce auto-Abs [rheumatoid factor and anti-citrullinated protein], but also actively mediate tissue destruction by secreting proinflammatory cytokines [IL17, IL6 and TNFα] and creating an inflammatory microenvironment that favors macrophage and neutrophil recruitment, and osteoclast activation (Burmester et al., 2014).

Author Manuscript

Numerous murine models of RA have been used to understand the etiology and pathogenesis of this disease (Bevaart et al., 2010; van den Berg, 2009). One of the most commonly used models is collagen-induced arthritis (CIA). Collagen type II-immunized DBA/1 mice develop polyarthritis characterized by synovial inflammation and hyperplasia, and cartilage and bone erosion, similar to the manifestations seen in RA patients. Less frequently utilized models discussed here include the proteoglycan-induced arthritis (PGIA) and Il1ra−/− BALB/c mouse models. The latter exhibit synovial inflammation and articular erosion. IC formation and autoreactive T cells are present in all three models. Other mouse models of RA have been developed but will not be discussed here.

Author Manuscript

Multiple co-stimulatory pathways have been studied in RA, with the CD28:B7 costimulatory pathway playing an indispensible role. Cd28−/− DBA/1 mice were resistant to arthritis after standard collagen II immunization and only a very small percentage of Cd28−/− mice developed CIA after augmented Ag loading (Tada et al., 1999b), suggesting a prerequisite role of CD28 co-stimulation in autoreactive T cell priming in CIA. It was also suggested that the expression of B7 molecules on B cells was essential for autoreactive T cell priming in PGIA (O’Neill et al., 2007). Similar to observations in models of SLE, B7.1 and B7.2 seem to activate CD28 co-stimulation in a redundant manner in CIA (Webb et al., 1996). Blocking CD28:B7 interactions with CTLA4Ig before or at the time of immunization prevented the development of CIA, while administration of CTLA4Ig after arthritis onset was able to ameliorate CIA (Knoerzer et al., 1995; Quattrocchi et al., 2000; Webb et al., 1996). In addition to reduced lymphocyte expansion and pro-inflammatory cytokine production, CTLA4Ig treatment also induced Treg cells by generating tolerogenic dendritic cells (DCs) in the CIA model (Ko et al., 2010). In contrast to SLE, clinical trials with Abatacept (human CTLA4Ig) were more successful in RA patients, and Abatacept was approved by the US FDA (Food and Drug Administration) to treat RA based on long-term safety and efficacy results (Ford et al., 2014). Importantly, Abatacept treatment resulted in clinical benefits in RA patients refractory to other RA treatments (Genovese et al., 2005; Westhovens et al., 2014), and was able to sustain disease remission after withdrawal of therapy in recent-onset RA patients (Emery et al., 2015).

Author Manuscript

Although CD28:B7 co-stimulation plays an essential role in the pathogenesis of RA, autoAb production by B cells remains intact in the absence of CD28:B7 signaling, suggesting that other co-stimulatory receptors contribute to the disease (O’Neill et al., 2007; Tada et al., 1999b). Indeed, the protective effect of CD28:B7 perturbation was diminished by the presence of proinflammatory cytokines, such as IL12 in the CIA model and IL1 in the spontaneous Il1ra−/− RA model (Kotani et al., 2006; Tada et al., 1999b), suggesting that CD28-independent co-stimulatory pathways are provoked by proinflammatory cytokines during RA pathogenesis. Interestingly, ICOSL is induced by TNFα (Swallow et al., 1999), a dominant cytokine in RA, raising the possibility that the ICOS:ICOSL pathway is activated during RA pathogenesis. Blockade with anti-ICOSL resulted in mitigated CIA and reduced

Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 7

Author Manuscript

auto-Ab titers (Iwai et al., 2002). ICOS:ICOSL co-stimulation contributes to CIA induction by promoting Tfh cell generation, GC formation and auto-Ab production (Hu et al., 2009), suggesting that this pathway may be responsible for the maintenance of an auto-Ab response in CIA induced in Cd28−/− mice (O’Neill et al., 2007; Tada et al., 1999b). The CD40:CD40L axis also contributes to auto-Ab production in RA. CD40L was expressed on CD4+ T cells from synovial fluid of RA patients following in vitro activation and promoted auto-Ab production (MacDonald et al., 1997). CD40:CD40L blockade abrogated rheumatoid factor production in transgenic mice overexpressing human rheumatoid factor, whereas a CD40 agonist Ab prevented apoptosis of rheumatoid factor precursor B cells in CIA (Kyburz et al., 1999; Tellander et al., 2000). Thus it is likely that the ICOS:ICOSL and CD40:CD40L pathways contribute to RA pathogenesis by promoting humoral immunity.

Author Manuscript

Other TNFRSF co-stimulatory pathways have also been studied in RA using the CIA model. The expression of OX40 on activated T cells and OX40L predominantly on APCs is restricted to inflamed joints (Gwyer Findlay et al., 2014). Lack of OX40:OX40L signaling resulted in decreased IFNγ and auto-Ab production, and increased tissue integrity in CIA (Gwyer Findlay et al., 2014; Yoshioka et al., 2000). Furthermore, GITR (TNFRSF18, CD357) promoted production of proinflammatory cytokines and chemokines (Cuzzocrea et al., 2005), while blocking the CD27:CD70 pathway ameliorated arthritis by repressing autoAb production (Oflazoglu et al., 2009). In contrast, a 4-1BB agonist Ab substantially ameliorated established arthritis by induction of tolerogenic DCs and subsequent induction of regulatory CD8+ cells (Seo et al., 2004). Thus, while most TNFRSF co-stimulatory pathways promote RA, some may protect. Also, co-stimulation not only affects T cell function but also impacts APCs that express the corresponding ligands.

Author Manuscript

While co-stimulatory signals are exacerbated in RA, defects of co-inhibitory pathways are also implicated in the susceptibility to, and progression of, RA. Polymorphisms within the CTLA4 locus have been described in subgroups of RA patients (Saverino et al., 2010). Treg cells from active RA patients had increased methylation at the CTLA4 promoter, which led to reduction in CTLA4 expression and failure to activate the tolerogenic indoleamine 2,3dioxygenase pathway (Cribbs et al., 2014). These findings raise the possibility that intrinsic defects of co-inhibition contribute to the systemic loss of tolerance in RA.

Author Manuscript

Defects in the PD1 co-inhibitory pathway have also been implicated in RA pathogenesis. Polymorphisms within the PDCD1 locus appear to be associated with RA (Okazaki and Honjo, 2007), while soluble PD1 correlated with increased TNFα levels in RA patients (Wan et al., 2006). Curiously, serum levels of auto-Abs against PD-L1 were found to correlate with active disease in RA patients (Dong et al., 2003), although their functional significance needs to be evaluated further. Pdcd1−/− mice were more susceptible to CIA, whereas activation of the PD1 co-inhibitory pathway by PD-L1.Fc ameliorated disease via reduced IFNγ production (Raptopoulou et al., 2010), suggesting that sufficient PD1 coinhibition is required to control the development of CIA. The role of additional IgSF co-inhibitory receptors in RA has been studied recently. TIGIT (VSIG9, VSTM3) shares the same ligands CD112 and CD155 with another co-stimulatory

Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 8

Author Manuscript

receptor CD226, and exerts suppressive effects on T cell activation and function in CIA (Levin et al., 2011). Another B7 family co-inhibitory receptor, B7-H3 (CD276), is expressed on B7.1−B7.2− fibroblast-like synoviocytes from RA patients (Tran et al., 2008). However, B7-H3 seems to transduce co-stimulatory signals in Th1 and Th17 cells as Cd276−/− mice were more resistant to CIA with decreased IL6, IL17 and TNFα levels (Luo et al., 2015). Studies with the inhibitory ligands HVEM (TNFRSF14, CD270) and Galectin9 have impacted a role for their respective co-inhibitory receptors BTLA and TIM3 (HAVCR2, CD366) in RA pathogenesis. Treatment with HVEMIg augmented CIA (Pierer et al., 2009), raising the possibility that BTLA may impact RA. Galectin 9-deficient mice were more susceptible to CIA induction, while treatment with Galectin 9 ameliorated the disease severity (Seki et al., 2008), implicating a role for TIM3. However, the complexity of these pathways and the paucity of studies on the role of co-inhibitory pathways in RA has limited our understanding of how these pathways might be manipulated therapeutically.

Author Manuscript

While Abatacept has proven to be an effective treatment for RA, it is clear that additional therapies, used in combination, will be required for complete resolution of the disease in all RA patients. In contrast, a protective role of co-inhibitory pathways has also been demonstrated in RA (Figure 2). Although intrinsic defects in these pathways that appear to be associated with susceptibility to RA have been suggested, a clearer understanding of their importance and relevance is needed. As with other autoimmune diseases, it remains to be seen whether boosting coinhibition is a viable therapeutic strategy for RA.

Multiple Sclerosis Author Manuscript

Multiple Sclerosis (MS) is an autoimmune disorder characterized by immune cell infiltration and inflammation within the brain and spinal cord (central nervous system, CNS) that leads to progressive demyelination and neurodegeneration (Dendrou et al., 2015). Macrophages, autoreactive CD8+ T cells, Th1 and Th17 cells and clonally expanded B cells dominate the infiltrate across the blood-brain barrier. The myelin sheaths that surround nerves in the CNS contain myelin basic protein (MBP), myelin oligodendrocyte glycoprotein (MOG), proteolipid protein (PLP), myelin-associated glycoprotein (MAG), and 2′,3′-CNP. Auto-Abs and autoreactive T cells against these auto-Ags have been identified in MS patients. CNSresident cells, microglia and astrocytes are activated during the inflammatory process and also produce proinflammatory mediators, exacerbating the disease.

Author Manuscript

Numerous murine experimental autoimmune encephalomyelitis (EAE) models have provided insightful understanding of MS etiology (Dendrou et al., 2015). Models discussed here include: (a) PLP139-151-immunized SJL/J mice, MOG35-55-immunized C57BL/6J mice or MBP-immunized PLxSJL F1 mice, which exhibit and a chronic EAE course; (b) PLP139-151-immunized SJL/J mice, in which epitope spreading occurs with a relapsingremitting disease course (R-EAE); and (c) MBP-reactive TCR or MOG-reactive TCR (2D2) transgenic mice that exhibit a spontaneous chronic EAE disease course. In these EAE models, IFNγ and IL17 produced by Th1 and Th17 cells, respectively, are dominant

Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 9

Author Manuscript

proinflammatory factors, while GM-CSF production by a subset of pathogenic Th17 cells promotes chronic encephalomyelitis (Dendrou et al., 2015).

Author Manuscript Author Manuscript

Several co-stimulatory pathways have been studied using these EAE models. The CD28:B7 co-stimulatory pathway was found to be essential in lowering the threshold for autoreactive T cell priming during the development of encephalomyelitis. Spontaneous development of EAE was abrogated in Cd28−/− MBP1-17 TCR transgenic mice, but could be reversed by high doses of MBP (Oliveira-dos-Santos et al., 1999). Likewise, C57BL/6J mice lacking both B7.1 and B7.2 were completely resistant to MOG35-55-induced EAE (Chang et al., 1999). However, blockade or deficiency of both B7.1 and B7.2 exacerbated EAE on an SJL/J background (Jabs et al., 2002; Perrin et al., 1996b; Racke et al., 1995), suggesting that the requirement of B7.1 and B7.2 for EAE induction appears to be influenced by the mouse genetic background (Jabs et al., 2002). The contribution of B7.1 and B7.2 is also timedependent as the impact of CD28:B7 blockade on EAE severity is affected by the time and dosage of CTLA4Ig administration (Khoury et al., 1995; Perrin et al., 1996b; Racke et al., 1995; Vogel et al., 2015). B7.1 and B7.2 are dynamically expressed on different cell types during different phases of EAE, and the presentation of myelin epitopes is largely B7.1dependent (Cross et al., 1999; Karandikar et al., 1998; Miller et al., 1995), indicating that co-stimulation via B7.1 might be more dominant in promoting EAE. While blocking with anti-B7.2 had minimal effect, blocking with anti-B7.1 reduced disease severity in both chronic EAE and R-EAE models (Kuchroo et al., 1995; Miller et al., 1995; Perrin et al., 1996b; Racke et al., 1995). However, B7.1 or B7.2 single knockout C57BL/6J mice developed similar disease severity as wild-type mice in the MOG35-55-induced EAE model (Chang et al., 1999), suggesting that B7.1 and B7.2 may function redundantly. Despite uncertainties about the efficacy of CTLA4Ig in mouse EAE models, Abatacept therapy has shown some efficacy in clinical trials and appears to limit MBP-reactive T cells (Viglietta et al., 2008). In contrast, a human CD28 blocking Ab prevented CNS pathology in human MOG-induced EAE in rhesus monkeys (Haanstra et al., 2015), indicating selective blockade of CD28 might be another option to inhibit autoreactive T cell activation.

Author Manuscript

Unlike the co-stimulatory receptor CD28 or the role of ICOS in other autoimmune diseases, ICOS exerts a differential impact during the priming and efferent phases in EAE models. Selective ICOS blockade during the priming phase [1–10 days after immunization] exacerbated disease, whereas selective blockade during the efferent phase [9–20 days after immunization] ameliorated disease (Sporici et al., 2001). Likewise, ICOS blockade promoted effector 2D2 T cell-induced EAE, but mitigated memory 2D2 T cell-induced EAE (Elyaman et al., 2008). Interestingly, Icos−/− mice develop more severe EAE with enhanced Th1 and Th17 cell cytokine production, possibly due to the reduced IL4 and IL10 production that limits Th1 and Th17 cell priming (Dong et al., 2001; Galicia et al., 2009). It has been suggested that CD28-mediated T cell priming is dependent on B7 upregulation on APCs via CD40:CD40L co-stimulation (Yang and Wilson, 1996). Indeed, EAE could not be induced in Cd154−/− mice unless B7 was overexpressed on APCs (Grewal et al., 1996). CD40 is expressed in the CNS of MOG-induced EAE mice as well as active lesions of MS patients (Becher et al., 2001; Gerritse et al., 1996). Treatment with CD40L Ab completely prevented the development of EAE, and also reduced clinical severity in mice with

Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 10

Author Manuscript

established EAE by suppressing T cell activation and IFNγ production (Gerritse et al., 1996; Howard et al., 1999). Preclinical assessment of anti-CD40 in monkeys also provided support for its therapeutic potential in the treatment of MS (Boon et al., 2001; Boon et al., 2002; Laman et al., 2002), but human trials were halted due to unanticipated thromboembolic complications (Ford et al., 2014).

Author Manuscript

Additional co-stimulatory pathways appear to complement CD28:B7 co-stimulation in promoting MS pathogenesis. For instance, expansion of MBP-reactive T cells from MS patients was CD28:B7 co-stimulation-independent (Scholz et al., 1998), while double immunization reversed the protection against EAE in Cd28−/− mice (Chitnis et al., 2001). Interestingly, OX40L blockade ameliorated EAE in double immunized Cd28−/− mice (Chitnis et al., 2001). OX40 is up-regulated on autoreactive T cells within active lesions in both EAE mice and MS patients, and anti-OX40:toxin conjugate-mediated depletion of OX40+ T cells reduced EAE (Carboni et al., 2003; Weinberg et al., 1996). The OX40:OX40L co-stimulation axis mediates autoreactive T cell priming, migration and function in EAE (Ndhlovu et al., 2001; Nohara et al., 2001; Weinberg et al., 1999). However, this axis does not appear to be a primary co-stimulatory pathway promoting EAE as deficiency in either OX40 or OX40L, or blockade of OX40:OX40L interactions led to a reduction of, but not complete protection against, EAE (Carboni et al., 2003; Ndhlovu et al., 2001; Nohara et al., 2001; Weinberg et al., 1999). In addition, transgenic mice overexpressing OX40L developed enhanced EAE, but were protected against EAE in the absence of CD28 or CD40 (Ndhlovu et al., 2001), suggesting a prerequisite for the CD28:B7 or CD40:CD40L co-stimulatory pathways in OX40-mediated autoreactive T cell priming.

Author Manuscript

The role of other TNFRSF co-stimulatory receptors has also been studied in EAE. Deficiency of 4-1BB ligand (TNFSF9, CD137L) resulted in resistance to MOG35-55-induced EAE, reduction in Th1 and Th17 cell cytokine production and autoreactive T cell infiltration (Martinez Gomez et al., 2012). However, agonistic 4-1BB Abs ameliorated disease severity in MOG35-55 induced EAE mice by promoting activation-induced cell death of autoreactive T cells and inhibiting Th17 cell differentiation (Kim et al., 2011; Sun et al., 2002b). Thus, negated or exacerbated 4-1BB signaling limits EAE, suggesting that a defined, optimal window of 4-1BB co-stimulation is required to promote EAE.

Author Manuscript

Analysis of the impact of the CD27:CD70 co-stimulation axis in EAE has added more complexity. While CD70 blockade inhibited EAE induction in PLP139-151-immunized SJL/J mice by repressing TNFα production (Nakajima et al., 22000), MOG35-55-induced EAE was exacerbated in Cd27−/− or Cd70−/− mice and ameliorated in transgenic mice overexpressing CD70, where IL17 and CXCR6 expression was suppressed in differentiating Th17 cells (Coquet et al., 2013). These studies provide another example of a co-stimulatory pathway that seems to have a differential impact depending on the type of EAE model and/or mouse genetic backgrounds. While aberrant co-stimulation leads to MS pathogenesis, insufficient co-inhibition can also promote the development and progression of MS as is seen in other autoimmune diseases. The co-inhibitory receptor CTLA4 was found to be dysregulated in MBP-reactive T cells from MS patients compared with healthy controls (Oliveira et al., 2003). CTLA4 blockade

Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 11

Author Manuscript Author Manuscript

resulted in exacerbated EAE, increased cytokine production, and enhanced epitope spreading (Hurwitz et al., 1997; Karandikar et al., 2000; Karandikar et al., 1996; Perrin et al., 1996a). Lack of CTLA4 co-inhibition also induced severe disease in EAE-resistant BALB/c mice (Hurwitz et al., 2002), highlighting a key role for CTLA4 in limiting selfreactivity and preventing EAE development. However, a recent study suggests that CTLA4 may also intrinsically limit Treg cell activation and expansion, and deletion of Ctla4 in Treg cells during adulthood was necessary and sufficient to provide protection from EAE (Paterson et al., 2015). CTLA4 appeared to negatively select Treg cells and narrow the conventional T cell repertoire by restricting TCRα chain usage in an MBP-reactive Vβ8.2 transgenic model, and therefore these transgenic mice were protected from spontaneous EAE in the absence of CTLA4 expression in the thymus (Verhagen et al., 2009; Verhagen et al., 2013). These studies highlight the differential impact of CTLA4 co-inhibition in the thymus versus the periphery, as well as its inhibitory effect on both autoreactive T cells and Treg cells.

Author Manuscript

Other inhibitory receptors also limit MS and EAE. Polymorphisms in the PDCD1 locus appear to be associated with disease progression in MS patients (Okazaki and Honjo, 2007), while Pdcd1−/− mice are more susceptible to MOG35-55-induced EAE (Carter et al., 2007). PD1 and its ligands PD-L1 and PD-L2 are expressed on CNS-infiltrating cells, and PD-L1 but not PD-L2 is substantially upregulated on endothelium and microglial in the CNS of EAE mice and MS patients (Liang et al., 2003; Magnus et al., 2005; Schreiner et al., 2008). Consistent with these observations, PD-L1 plays a more critical role than PD-L2 in controlling EAE as demonstrated using Cd274−/− versus Cd273−/− mice, respectively (Bodhankar et al., 2013; Carter et al., 2007; Latchman et al., 2004; Schreiner et al., 2008). However, it is notable that the relative contribution of PD-L1 and PD-L2 in controlling EAE varies in different mouse strains and may also depend on the phase of disease (Zhu et al., 2006). Interestingly, B7.1 has been identified as another ligand for PD-L1, and interactions between B7.1 and PD-L1 inhibit T cell responses (Butte et al., 2007). The differential role of B7.1 and B7.2, and PD-L1 and PD-L2 observed in different mouse strains may reflect a preferential usage of the B7.1:PD-L1 axis in certain strains.

Author Manuscript

Newly identified IgSF co-inhibitory receptors have also been implicated in modulating the disease course of MS. While transgenic mice overexpressing TIGIT or mice activated by a TIGIT agonist Ab showed reduced EAE symptoms, Tigit−/− mice were more susceptible to EAE (Joller et al., 2011; Levin et al., 2011). TIGIT cell-intrinsically suppressed T cell activation and function in the 2D2 TCR transgenic mouse model (Joller et al., 2011). The expression of another co-inhibitory receptor, TIM3 was found to be impaired in MS patients (Koguchi et al., 2006; Yang et al., 2008). Interaction between TIM3 and its ligand Galectin-9 negatively regulates Th1 cell responses in PLP139-151-immunized SJL/J mice (Monney et al., 2002; Sabatos et al., 2003; Zhu et al., 2005). While blockade of B7-H4, another B7 family co-inhibitory receptor, exacerbated EAE disease, activation of its unidentified ligand on T cells by B7-H4Ig provided protection against EAE (Podojil et al., 2013; Prasad et al., 2003). Btla−/− T cells showed increased proliferation and mediated attenuated EAE (Watanabe et al., 2003), suggesting that BTLA may also regulate autoreactive T cells. However, the role of B7-H3 in MOG35-55-induced EAE seems controversial. B7-H3 blockade resulted in enhanced EAE, whereas mice deficient in B7-H3 showed reduced disease severity and Th1 Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 12

Author Manuscript

and Th17 cell cytokine production (Luo et al., 2015; Prasad et al., 2004), suggesting that more analysis of this pathway is required. Collectively, these studies suggest that multiple co-inhibitory receptors can limit EAE inferring that boosting co-inhibitory signals might be a valid therapeutic approach for MS. T cells contribute a critical and central role in the pathogenesis and progression of EAE and MS. Consequently, there are multiple co-stimulatory and co-inhibitory receptors that impact disease and thus offer opportunities for therapeutic intervention (Figure 3). Interestingly, the role of several co-stimulatory and co-inhibitory receptors (B7.1 and B7.2, CD27 and CD70, and PD-L1 and PD-L2) appears to be affected by the EAE model and/or the genetic background used. Whether this reflects subtle nuances between these different model systems or a potential heterogeneous outcome in targeting these pathways in MS patients remains to be determined.

Author Manuscript

Type 1 Diabetes Type 1 Diabetes (T1D), also referred as Juvenile Diabetes, is a chronic autoimmune disorder associated with the destruction of insulin-secreting β-cells by autoreactive T cells and subsequent uncontrolled hyperglycemia (Atkinson et al., 2014). Around 60 T1Dsusceptibility regions have been mapped thus far, with the strongest association (HLA locus) and several additional prominent associations (eg. PTPN22, CTLA4, IL2RA) impacting T cell activation. Although multiple immune cell types participate in this disease, autoreactive CD4+ and CD8+ T cells are key drivers in the propagation of T1D, while Treg cells limit diabetogenic T cell function (Herold et al., 2013).

Author Manuscript

The non-obese diabetic (NOD) mouse is the T1D model of choice for the analysis of immune mechanisms that modulate the onset and progression of autoimmune diabetes (Reed and Herold, 2015). Autoimmune diabetes in NOD mice shares various common features with T1D in humans. NOD mice spontaneously develop chronic autoimmune diabetes, with insulitis starting at 3–4 weeks-of-age and subsequent diabetes onset occurring between 12– 25 weeks-of-age in female mice, while male NOD mice exhibit reduced onset and incidence. Similarly, various insulin-dependent diabetes susceptibility regions (Idd) have also been mapped in NOD mice. Variants of this model include the adoptive transfer of diabetic NOD splenocytes or islet-Ag specific TCR transgenic T cells (eg. BDC2.5) into NOD.scid mice.

Author Manuscript

Several co-stimulatory receptors have been shown to be critical for the development of autoimmune diabetes in NOD mice. Even though the CD28:B7 axis provides essential costimulation for autoreactive T cell priming, unlike other autoimmune diseases discussed above, it is also critical for Treg cell development and homeostasis in autoimmune diabetes. Both Cd28−/− and B7.1-B7.2 double-deficient NOD mice had a profoundly reduced number of CD4+CD25+ Treg cells, and thus exhibited accelerated autoimmune diabetes (Salomon et al., 2000). In addition, defects in CD27:B7 co-stimulation broke the Th1-Th2 balance with enhanced IFNγ production in NOD mice (Lenschow et al., 1996). The protective effect of CD28:B7 co-stimulation seems to be mainly provided by B7.1 as blocking both B7.1 and B7.2 or blocking B7.1 alone in NOD mice had a similar effect (Lenschow et al., 1995), while the phenotype in B7.1-deficient versus B7.1-B7.2-double deficient NOD mice appears

Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 13

Author Manuscript Author Manuscript

comparable (Salomon et al., 2000). However, B7.2-deficient NOD mice were free of autoimmune diabetes (Bour-Jordan et al., 2004), and treatment of young NOD mice (2 week-of-age) with anti-B7.2 resulted in delayed diabetes onset as well as reduced insulitis and T cell activation, but the protective effect that was abolished if anti-B7.2 was administrated in older NOD mice (Lenschow et al., 1995). Thus, B7.1 and B7.2 may have different functions depending on the timing and context of their involvement (Butte et al., 2007; Paterson et al., 2011). NOD mice ectopically expressing CTLA4Ig or treated with murine CTLA4Ig at 2–4 weeks-of-age lacked CD4+CD25+ Treg cells and exhibited exacerbated autoimmune diabetes, consistent with observations in Cd28−/− NOD mice (Lenschow et al., 1996; Salomon et al., 2000). Despite this unexpected phenotype in NOD mice, Abatacept (human CTLA4Ig) treatment showed some efficacy in recent-onset T1D patients, with ~9.5 month delay in C-peptide decline and lower levels of HbA1c compared to the placebo group (Orban et al., 2014). Nevertheless, the generally limited impact of Abatacept suggested that other co-stimulatory pathways independent of the CD28:B7 axis are involved in T1D pathogenesis.

Author Manuscript

ICOS:ICOSL co-stimulation is also exploited by both autoreactive T cells and Treg cells in autoimmune diabetes. Blockade with anti-ICOS resulted in delayed autoimmune diabetes in NOD mice, and Icos−/− NOD mice were free of autoimmune diabetes with reduced IFNγ production (Ansari et al., 2008; Hawiger et al., 2008). Conversely, other studies using the BDC2.5 TCR transgenic NOD model demonstrated that ICOS is preferentially expressed on Treg cells and mediates enhanced suppressive capacity of Treg cells (Herman et al., 2004; Kornete et al., 2015; Kornete et al., 2012), with ICOS blockade in BDC2.5 NOD mice resulting in rapid diabetes onset (Ansari et al., 2008; Herman et al., 2004). Thus, in contrast to the role of ICOS in other autoimmune diseases, the impact of ICOS blockade depends on the model used and its differential impact on diabetogenic T cells versus Treg cells.

Author Manuscript

Unlike the CD28:B7.1 and ICOS:ICOSL co-stimulatory pathways that are important for both autoreactive T cell activation and Treg cell homeostasis, the CD40:CD40L axis appears to have less impact on Treg cells in autoimmune diabetes. CD40:CD40L co-stimulation is required for the initiation of insulitis and autoreactive T cell priming in autoimmune diabetes, which also differs from its canonical role in activating APCs and/or humoral immunity in other autoimmune diseases. Blockade with anti-CD40L in neonates or deficiency of CD40L prevented autoimmune diabetes in NOD mice (Baker et al., 2008; Balasa et al., 1997; Bour-Jordan et al., 2004; Green et al., 2000). Cd154−/− autoreactive CD4+ T cells appeared to lose their diabetogenic potential even though APCs were systemically activated through CD40 ligation (Amrani et al., 2002), suggesting that either CD40L is essential for diabetogenic T cell activation or there may be another receptor for CD40L that activates APCs. In contrast to the CD40:CD40L axis, OX40:OX40L co-stimulation appears to be more important at later stages of autoimmune diabetes. OX40 is expressed on primed (CD44hi) T cells and OX40L (encoded by Tnfsf4) is upregulated on DCs after insulitis initiation but prior to islet destruction (11–13 week-of-age). Thus, interruption of OX40:OX40L interactions had no effect on diabetes onset in young NOD mice but exhibited the most significant delay in diabetes onset in NOD mice at 12 week-of-age (Pakala et al., 2004).

Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 14

Author Manuscript

Interestingly, Tnfsf4−/− NOD mice exhibited normal (or even accelerated) insulitis but did not develop autoimmune diabetes (Martin-Orozco et al., 2003), supporting the idea that different co-stimulatory axes participate at different stages to promote autoimmune diabetes.

Author Manuscript

Another TNFRSF member, 4-1BB (TNFRSF9), has also been studied in autoimmune diabetes. Tnfrsf9 has been mapped to Idd9.3, which mediates susceptibility to autoimmune diabetes in NOD mice. However, the autoimmune diabetes-resistant NOD.B10 Idd9.3 locus facilitates enhanced Treg cell induction and secretion of soluble 4-1BB that blocks the 4-1BB co-stimulatory signal on autoreactive T cells (Cannons et al., 2005; Kachapati et al., 2012; Lyons et al., 2000). Interestingly, while treatment with 4-1BB agonist Ab prevented autoimmune diabetes in NOD mice by inducing Treg cells, overexpression of a 4-1BB agonist induced severe autoimmune diabetes (Irie et al., 2007; Kachapati et al., 2013; Sytwu et al., 2003). 4-1BB is constitutively expressed on Treg cells, but transiently upregulated on effector T cells after activation. Therefore, there might be functionally and/or temporally distinct usage of 4-1BB on Treg cells versus autoreactive T cells in autoimmune diabetes. Collectively, these studies highlight the importance of multiple co-stimulatory receptors in modulating autoimmune diabetes via their differential impact on diabetogenic T cell function and Treg cell homeostasis.

Author Manuscript

In addition to Treg cell-mediated suppression, co-inhibitory receptors also provide cell intrinsic regulation of autoreactive T cells in NOD mice. For instance, CTLA4 is critical for controlling autoimmune diabetes. Polymorphisms within the CTLA4 locus have been identified that appear to associate with susceptibility to T1D and map to Idd5.1 in NOD mice (Saverino et al., 2010; Wicker et al., 2004). NOD mice are genetically deficient in expression of a CTLA4 splice variant that cannot bind to its B7 ligand (ligand-independent CTLA4; li-CTLA4) (Ueda et al., 2003; Vijayakrishnan et al., 2004). Curiously, restoration of li-CTLA4 expression in NOD mice limits insulitis and autoimmune diabetes in full-length CTLA4-dependent manner (Araki et al., 2009; Stumpf et al., 2013). Genetic deletion or blockade of CTLA4 showed dramatic acceleration of diabetes onset in NOD mice (Luhder et al., 2000; Luhder et al., 1998), whereas APC-directed CTLA4 engagement delayed autoimmune diabetes onset by inhibition of B cell maturation (Fife et al., 2006). CTLA4 engagement dampens diabetogenic T cell activity but in a very restricted time window that coincides with islet Ag re-encounter (Luhder et al., 2000; Luhder et al., 1998). Interestingly, CTLA4 upregulation is prevented by low CD86 expression and impaired T cell priming in NOD mice (Dahlen et al., 2000), suggesting that optimal T cell priming is also required to induce cell-intrinsic negative signals.

Author Manuscript

PD1:PD-L1 or :PD-2 interactions mediate co-inhibitory effects that appear to be distinct from CTLA4. Polymorphisms at the PDCD1 locus are associated with susceptibility to T1D (Okazaki and Honjo, 2007). Pdcd1−/− NOD mice developed autoimmune diabetes by 11 weeks-of-age with 100% penetrance in both females and males (Wang et al., 2005). In contrast to CTLA4 blockade, treatment with anti-PD1 or anti-PD-L1, but not anti-PD-L2, precipitated insulitis and autoimmune diabetes onset in NOD mice regardless of when the mice are treated [1–10 weeks-of-age] (Ansari et al., 2003), suggesting PD1:PD-L1 interactions impact all stages of autoimmune diabetes onset and progression. PD1 appears to prevent stable interactions between T cells and DCs in pancreatic lymph nodes (Fife et al.,

Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 15

Author Manuscript

2009). Interestingly, PD-L1 may also be expressed on pancreatic islets (Ansari et al., 2003; Liang et al., 2003), providing a regulatory signal before autoreactive T cells enter the islets. Interaction between PD-L1 and another of its known ligands B7.1 induces negative signals in diabetogenic T cells at the late phase of autoimmune diabetes (Paterson et al., 2011). Curiously, while PD-L1 overexpression on β-cells exhibited a protective role in NOD mice (Wang et al., 2008), its overexpression on β-cells in B6 mice provoked autoreactive T cells (Subudhi et al., 2004). The basis for these contradictory observations remains to be defined. Besides its regulation on peripheral autoreactive T cells, it is noteworthy that PD1 may also impacts thymic selection of diabetogenic T cells (Zucchelli et al., 2005).

Author Manuscript

Like PD1, the inhibitory receptor LAG3 (CD223) is also required to restrain the expansion of diabetogenic T cells in the islets at both early and late stages of autoimmune diabetes. LAG3 is a hom*olog to CD4 that also binds to MHC class II molecules, but with a higher affinity, and utilizes a unique, conserved intracellular KIEELE motif to mediate its regulatory function (Workman et al., 2002). Both female and male Lag3−/− NOD mice develop accelerated autoimmune diabetes with 100% penetrance and exhibit increased accumulation of islet Ag-specific T cells in the islets (Bettini et al., 2011). LAG3 blockade after the initiation of insulitis also precipitated diabetes onset (Bettini et al., 2011), synonymous with PD1 blockade but distinct from CTLA4 blockade. Interestingly, LAG3 is highly upregulated on CD4+CD25+ Treg cells and appears to mediate their suppressive function (Huang et al., 2004; Liang et al., 2008). This raises the possibility that the exacerbated autoimmune diabetes observed in Lag3−/− NOD mice is partially due to impaired Treg cell suppression in the absence of LAG3.

Author Manuscript

Co-inhibitory receptors TIM3 and B7-H4 may also play a role in dampening β-cell destruction at this later stage. Treatment with either anti-TIM3 or TIM3Ig that blocks interactions between endogenous TIM3 and its ligand augmented autoimmune diabetes but did not impact insulitis (Sanchez-Fueyo et al., 2003). Likewise, early treatment with B7H4Ig that provides co-inhibitory signals via its unknown ligand on T cells did not block insulitis but delayed aggressive β-cell destruction at the later stage (Wang et al., 2011). Thus, the analysis of co-inhibitory receptors in NOD mice suggests that these pathways are broadly utilized but at different stages during the pathogenesis of autoimmune diabetes.

Author Manuscript

The most prominent feature of co-stimulatory or co-inhibitory pathway utilization in autoimmune diabetes is the differential temporal utilization observed. CD28:B7 and CD40:CD40L costimulation, and CTLA4-mediated inhibition appear to be more important during disease onset and establishment of insulitis. In contrast, other co-stimulatory and coinhibitory receptors are utilized more broadly or at later stages of disease progression after insulitis onset (Figure 4). In addition, co-stimulation is also essential for the maintenance of Treg homeostasis in NOD mice, potentially complicating the therapeutic potential of some modalities.

Hierarchical usage of co-stimulatory and co-inhibitory pathways It is clear from the four disease-focused sections above that a wide variety of co-stimulatory and co-inhibitory pathways impact and shape systemic and organ-specific autoimmune

Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 16

Author Manuscript

disease. This evaluation not only provides an opportunity to compare and contrast their utilization in four distinct but nevertheless emblematic autoimmune diseases, but also offers potential approaches to address some unanswered questions. What appears to be clear feature is the hierarchical utilization of both co-stimulatory and coinhibitory pathways (Figure 1–4). This notion is highlighted by six general observations.

Author Manuscript Author Manuscript

First, there are “first-line” and “second-line” pathways impacting autoimmune disease. The CD28 co-stimulation is a “first-line” pathway essential for autoreactive T cell priming, whereas PD1 has a dominant “first-line” inhibitory impact on all autoimmune disease models discussed. However, the relative contributions of their ligands B7.1, B7.2 and PDL1, PD-L2, respectively, seem to be affected by the genetic background, and occasionally, the model system used, which may be explained by the timing of ligand expression and interaction, cell types that preferentially express one ligand versus the other, and/or the addition of B7.1:PD-L1 interactions in different mouse strains or models. This is probably further complicated by differential interactions between B7 molecules with CTLA4, as a lack of CTLA4 co-inhibition precipitates autoimmune manifestations under autoimmuneprone conditions, albeit to differing degrees. Studies with Abs that specifically block CD28:B7.1, CD28:B7.2, CTLA4:B7.1, CTLA4:B7.2, PD1:PD-L1, PD1:PD-L2 and PDL1:B7.1 interactions would help to clarify the relative contribution of these receptors and ligands. There may also be “second-line” pathways that do not impact all autoimmune disease all the time. For instance, while many IgSF co-inhibitory receptors are upregulated on autoreactive T cells and reinforce co-inhibitory signals after T cell priming, their contribution may not impact all autoimmune diseases equally. Inhibitory receptors such as LAG3, TIM3, TIGIT, BTLA4, B7-H3 and B7-H4 are either not consistently utilized between diseases or they need to be studied further to fully assess their true impact. Second, there are also temporal implications in the use of different co-stimulatory and coinhibitory pathways. For instance unlike the CD28:B7 or CD40:CD40L axes, the other TNFRSF co-stimulatory pathways tend to participate at later stages, probably because these receptors are upregulated on activated T cells and/or their corresponding ligands may be restricted to inflamed tissues. From studies in autoimmune diabetes, it would appear that CTLA4 functions in a more restricted time window during the priming phase, while TIM3 and B7-H4 play a role in the later stage. This contrasts with PD1 and LAG3 that appear to be involved over a broader time period. Whether this applies to other autoimmune diseases remains to be determined.

Author Manuscript

Third, while there is differential usage of each co-stimulatory pathway, compensatory mechanisms also exist when some co-stimulatory pathways are absent to promote T and B cell self-reactivity. For instance, CD28:B7.1 but not CD28:B7.2 co-stimulation predominantly provides T cell priming signals, but blockade of B7.1 alone does not always result in significant reduction in disease severity, especially in systemic autoimmune diseases (eg. SLE, RA). The loss of CD28:B7.1 co-stimulation can be compensated for by either increased utilization of CD28:B7.2 co-stimulation and/or exploiting the use of other receptor-mediated co-stimulation, such as the CD40:CD40L, ICOS:ICOSL and

Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 17

Author Manuscript

OX40:OX40L axes. Therefore, combinatorial blocking approaches could be exploited to enhance efficacy. Fourth, while TNFRSF co-stimulatory pathways predominantly promote T cell selfreactivity, they do not do so uniformly or to the same extent and occasionally exhibit an unexpected regulatory capacity. For instance, the OX40:OX40L pathway promotes all four autoimmune diseases to some extent, while the CD27:CD70 and GITR:GITRL pathways contribute to a variable degree. Interestingly, in some settings the 4-1BB:4-1BBL pathway can promote or inhibit immunity. For instance, the co-stimulation of 4-1BB via its endogenous ligand 4-1BBL can provide stimulatory signals, but agonist 4-1BB Abs promote activation-induced cell death and/or the generation of tolerogenic DCs or Treg cells in multiple autoimmune settings. Determining whether there are any other unknown binding partners may help to clarify some of these ambiguities.

Author Manuscript Author Manuscript

Fifth, the utilization of specific co-stimulatory and co-inhibitory axes is likely impacted by the cellular mechanisms that promote pathology in each disease. For instance, TIM3 is preferentially expressed on Th1 cells, so it may have more impact on Th1-mediated autoimmune diseases (eg. MS, T1D). Conversely, both the CD40:CD40L and the ICOS:ICOSL axes facilitate crosstalk between autoreactive T and B cells, but to differing extents. CD40:CD40L co-stimulation promotes B cell activation, maturation and upregulation of B7 molecules, and thus provides strong signals for T cell activation via the CD28:B7 axis. Therefore, CD40:CD40L co-stimulation is required to initiate the development of autoimmune diseases. However, ICOS:ICOSL co-stimulation is required for the generation, function and maintenance of Tfh cells that help GC formation and auto-Ab production. Therefore, the ICOS:ICOSL axis has a greater impact on systemic autoimmune disorders that are dominated by thymus-dependent humoral immune responses. Likewise, the SLAMF receptors affect Tfh cell development and function, so thus appear to have a greater role in IC-mediated autoimmune diseases (eg. SLE).

Author Manuscript

Sixth, while co-stimulation is essential to prime autoreactive T cells and sustain their selfreactivity, it is also critical for the maintenance of Treg cell homeostasis and function, as is demonstrated in autoimmune diabetes, which further complicates data interpretation and a full understanding of their role in shaping autoimmune disease. For instance, while it is clear that CD28 provides a dominant co-stimulatory contribution in promoting autoimmunity, it can also impact Treg cell development and thereby limit autoimmunity. Also, while coinhibitory receptors are highly upregulated on Treg cells, it is unclear whether they mediate the cell-intrinsic regulation of Treg cell function or the cell-extrinsic Treg cell-mediated control of autoreactive T cells. Studies with cell type-restricted deletion of co-inhibitory molecules may help to clarify their roles and define populations that could be targeted therapeutically.

Concluding remarks Although remarkable progress has been made in the past few decades in defining how costimulatory and co-inhibitory pathways impact autoimmune diseases, many questions and challenges still remained. Four are particularly noteworthy.

Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 18

Author Manuscript

First, what additional co-stimulatory and co-inhibitory pathways impact autoimmune disease? It is evident from the discussion above that there remain gaps in our understanding of the role of certain co-stimulatory and co-inhibitory receptors and ligands in many autoimmune diseases. Filling in these gaps will provide a greater appreciation for the global impact of each pathway. Although this review only focused on four autoimmune diseases, it is clear that expanding this sphere of knowledge to all autoimmune disease would be highly valuable, and will be essential for shaping future therapeutic strategy. We should also not rule out the possibility that there are additional co-stimulatory and co-inhibitory receptors and ligands that remain to be discovered or novel receptors/ligands for known molecules. Indeed, the receptor for B7-H4 remain elusive and we have yet to fully understand the impact of more recently discovered B7 family co-modulatory molecules VISTA (B7-H5, Gi24, Dies1), B7-H6 and B7-H7 .

Author Manuscript

Second, can effective therapeutic strategies be developed for the treatment of autoimmune disease by modulating co-stimulatory and co-inhibitory pathways? The impact of blocking the inhibitory receptors CTLA4 and PD1 in the treatment of cancer has been spectacular, with justifiable excitement about the potential for a long pipeline of blocking and agonist Abs that are in early stage trials or in development (Topalian et al., 2015). The key challenge is whether the same therapeutic impact can be achieved for autoimmune diseases by modulating co-stimulatory and coinhibitory pathways? This is likely to be a greater challenge and will inevitably require a combinatorial approach that effectively block co-stimulatory pathways while enhancing coinhibition. Indeed, in light of the redundant use of many co-stimulatory pathways and the inadequacy of host co-inhibitory signals to suppress autoreactive T and B cells, modulating a single or even multiple pathways is not likely to be sufficient.

Author Manuscript

Third, how can we minimize the adverse side effects of targeting co-stimulatory pathways in patients with autoimmune disease? As autoimmune disease tends not to be immediately life threatening, unlike cancer, the risk tolerance in therapeutic development is lower. Thus an additional challenge to the one cited above is to also ensure that therapies have relatively high safety profile. For instance, blocking CD40:CD40L co-stimulation exhibited some efficacy in earlier preclinical and clinical trials for autoimmune disease, but have been halted due reported adverse thromboembolic events (Ford et al., 2014). While this creates unique challenges for the treatment of systemic autoimmune diseases, it might be feasible to target specific tissues, and thus organ specific autoimmune diseases, using targeted therapies such as engineered viral vector or nanoparticle delivery of co-stimulatory or co-inhibitory modulators.

Author Manuscript

Fourth, what immune cell population should be targeted? Many of the co-stimulatory and co-inhibitory pathways discussed above impact both the pathogenic effector and regulatory populations. Thus the global targeting of any pathway can have mixed and/or unexpected outcomes. This raises the question of whether it will be possible to develop therapies that target specific cell types. Given the significant impact of co-stimulatory and co-inhibitory pathways in autoimmunity, there is clearly significant justification and momentum to expand our understanding, and

Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 19

Author Manuscript

determine the full impact of, these pathways in all autoimmune diseases. While the development of effective therapies for autoimmune diseases remains a major challenge, the impact of targeting these pathways in cancer should serve as a source of encouragement.

Acknowledgments The authors wish to thank the Vignali Lab helpful discussions. This work was supported by the National Institutes of Health (AI108545, AI091977 and DK089125 to D.A.A.V), and the NCI Comprehensive Cancer Center Support CORE grant (CA047904, to D.A.A.V.).

References

Author Manuscript Author Manuscript Author Manuscript

Amrani A, Serra P, Yamanouchi J, Han B, Thiessen S, Verdaguer J, Santamaria P. CD154-dependent priming of diabetogenic CD4(+) T cells dissociated from activation of antigen-presenting cells. Immunity. 2002; 16:719–732. [PubMed: 12049723] Ansari MJ, Fiorina P, Dada S, Guleria I, Ueno T, Yuan X, Trikudanathan S, Smith RN, Freeman G, Sayegh MH. Role of ICOS pathway in autoimmune and alloimmune responses in NOD mice. Clin Immunol. 2008; 126:140–147. [PubMed: 17889619] Ansari MJ, Salama AD, Chitnis T, Smith RN, Yagita H, Akiba H, Yamazaki T, Azuma M, Iwai H, Khoury SJ, et al. The programmed death-1 (PD-1) pathway regulates autoimmune diabetes in nonobese diabetic (NOD) mice. J Exp Med. 2003; 198:63–69. [PubMed: 12847137] Araki M, Chung D, Liu S, Rainbow DB, Chamberlain G, Garner V, Hunter KM, Vijayakrishnan L, Peterson LB, Oukka M, et al. Genetic evidence that the differential expression of the ligandindependent isoform of CTLA-4 is the molecular basis of the Idd5.1 type 1 diabetes region in nonobese diabetic mice. J Immunol. 2009; 183:5146–5157. [PubMed: 19783679] Atkinson MA, Eisenbarth GS, Michels AW. Type 1 diabetes. Lancet. 2014; 383:69–82. [PubMed: 23890997] Baker RL, Wagner DH Jr, Haskins K. CD40 on NOD CD4 T cells contributes to their activation and pathogenicity. J Autoimmun. 2008; 31:385–392. [PubMed: 18951762] Balasa B, Krahl T, Patstone G, Lee J, Tisch R, McDevitt HO, Sarvetnick N. CD40 ligand-CD40 interactions are necessary for the initiation of insulitis and diabetes in nonobese diabetic mice. J Immunol. 1997; 159:4620–4627. [PubMed: 9379064] Becher B, Durell BG, Miga AV, Hickey WF, Noelle RJ. The clinical course of experimental autoimmune encephalomyelitis and inflammation is controlled by the expression of CD40 within the central nervous system. J Exp Med. 2001; 193:967–974. [PubMed: 11304557] Bettini M, Szymczak-Workman AL, Forbes K, Castellaw AH, Selby M, Pan X, Drake CG, Korman AJ, Vignali DA. Cutting edge: accelerated autoimmune diabetes in the absence of LAG-3. J Immunol. 2011; 187:3493–3498. [PubMed: 21873518] Bevaart L, Vervoordeldonk MJ, Tak PP. Evaluation of therapeutic targets in animal models of arthritis: how does it relate to rheumatoid arthritis? Arthritis Rheum. 2010; 62:2192–2205. [PubMed: 20506322] Bodhankar S, Galipeau D, Vandenbark AA, Offner H. PD-1 Interaction with PD-L1 but not PD-L2 on B-cells Mediates Protective Effects of Estrogen against EAE. J Clin Cell Immunol. 2013; 4:143. [PubMed: 24009988] Boon L, Brok HP, Bauer J, Ortiz-Buijsse A, Schellekens MM, Ramdien-Murli S, Blezer E, van Meurs M, Ceuppens J, de Boer M, et al. Prevention of experimental autoimmune encephalomyelitis in the common marmoset (Callithrix jacchus) using a chimeric antagonist monoclonal antibody against human CD40 is associated with altered B cell responses. J Immunol. 2001; 167:2942–2949. [PubMed: 11509643] Boon L, Laman JD, Ortiz-Buijsse A, den Hartog MT, Hoffenberg S, Liu P, Shiau F, de Boer M. Preclinical assessment of anti-CD40 Mab 5D12 in cynomolgus monkeys. Toxicology. 2002; 174:53–65. [PubMed: 11972992] Boumpas DT, Furie R, Manzi S, Illei GG, Wallace DJ, Balow JE, Vaishnaw A, Group BGLNT. A short course of BG9588 (anti-CD40 ligand antibody) improves serologic activity and decreases

Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 20

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

hematuria in patients with proliferative lupus glomerulonephritis. Arthritis Rheum. 2003; 48:719– 727. [PubMed: 12632425] Bour-Jordan H, Salomon BL, Thompson HL, Szot GL, Bernhard MR, Bluestone JA. Costimulation controls diabetes by altering the balance of pathogenic and regulatory T cells. J Clin Invest. 2004; 114:979–987. [PubMed: 15467837] Bretscher P, Cohn M. A theory of self-nonself discrimination. Science. 1970; 169:1042–1049. [PubMed: 4194660] Brown DR, Calpe S, Keszei M, Wang N, McArdel S, Terhorst C, Sharpe AH. Cutting edge: an NK cell-independent role for Slamf4 in controlling humoral autoimmunity. J Immunol. 2011; 187:21– 25. [PubMed: 21622868] Burmester GR, Feist E, Dorner T. Emerging cell and cytokine targets in rheumatoid arthritis. Nat Rev Rheumatol. 2014; 10:77–88. [PubMed: 24217582] Butte MJ, Keir ME, Phamduy TB, Sharpe AH, Freeman GJ. Programmed death-1 ligand 1 interacts specifically with the B7–1 costimulatory molecule to inhibit T cell responses. Immunity. 2007; 27:111–122. [PubMed: 17629517] Cannons JL, Chamberlain G, Howson J, Smink LJ, Todd JA, Peterson LB, Wicker LS, Watts TH. Genetic and functional association of the immune signaling molecule 4-1BB (CD137/TNFRSF9) with type 1 diabetes. J Autoimmun. 2005; 25:13–20. [PubMed: 15998581] Carboni S, Aboul-Enein F, Waltzinger C, Killeen N, Lassmann H, Pena-Rossi C. CD134 plays a crucial role in the pathogenesis of EAE and is upregulated in the CNS of patients with multiple sclerosis. J Neuroimmunol. 2003; 145:1–11. [PubMed: 14644025] Carter LL, Leach MW, Azoitei ML, Cui J, Pelker JW, Jussif J, Benoit S, Ireland G, Luxenberg D, Askew GR, et al. PD-1/PD-L1, but not PD-1/PD-L2, interactions regulate the severity of experimental autoimmune encephalomyelitis. J Neuroimmunol. 2007; 182:124–134. [PubMed: 17182110] Chang TT, Jabs C, Sobel RA, Kuchroo VK, Sharpe AH. Studies in B7-deficient mice reveal a critical role for B7 costimulation in both induction and effector phases of experimental autoimmune encephalomyelitis. J Exp Med. 1999; 190:733–740. [PubMed: 10477557] Chatterjee M, Kis-Toth K, Thai TH, Terhorst C, Tsokos GC. SLAMF6-driven co-stimulation of human peripheral T cells is defective in SLE T cells. Autoimmunity. 2011; 44:211–218. [PubMed: 21231893] Chitnis T, Najafian N, Abdallah KA, Dong V, Yagita H, Sayegh MH, Khoury SJ. CD28-independent induction of experimental autoimmune encephalomyelitis. J Clin Invest. 2001; 107:575–583. [PubMed: 11238558] Coquet JM, Middendorp S, van der Horst G, Kind J, Veraar EA, Xiao Y, Jacobs H, Borst J. The CD27 and CD70 costimulatory pathway inhibits effector function of T helper 17 cells and attenuates associated autoimmunity. Immunity. 2013; 38:53–65. [PubMed: 23159439] Crampton SP, Morawski PA, Bolland S. Linking susceptibility genes and pathogenesis mechanisms using mouse models of systemic lupus erythematosus. Dis Model Mech. 2014; 7:1033–1046. [PubMed: 25147296] Cribbs AP, Kennedy A, Penn H, Read JE, Amjadi P, Green P, Syed K, Manka SW, Brennan FM, Gregory B, Williams RO. Treg cell function in rheumatoid arthritis is compromised by ctla-4 promoter methylation resulting in a failure to activate the indoleamine 2,3-dioxygenase pathway. Arthritis Rheumatol. 2014; 66:2344–2354. [PubMed: 24891289] Cross AH, Lyons JA, San M, Keeling RM, Ku G, Racke MK. T cells are the main cell type expressing B7-1 and B7-2 in the central nervous system during acute, relapsing and chronic experimental autoimmune encephalomyelitis. Eur J Immunol. 1999; 29:3140–3147. [PubMed: 10540325] Crotty S. T follicular helper cell differentiation, function, and roles in disease. Immunity. 2014; 41:529–542. [PubMed: 25367570] Cunninghame Graham DS, Graham RR, Manku H, Wong AK, Whittaker JC, Gaffney PM, Moser KL, Rioux JD, Altshuler D, Behrens TW, Vyse TJ. Polymorphism at the TNF superfamily gene TNFSF4 confers susceptibility to systemic lupus erythematosus. Nat Genet. 2008; 40:83–89. [PubMed: 18059267]

Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 21

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Cuzzocrea S, Ayroldi E, Di Paola R, Agostini M, Mazzon E, Bruscoli S, Genovese T, Ronchetti S, Caputi AP, Riccardi C. Role of glucocorticoid-induced TNF receptor family gene (GITR) in collagen-induced arthritis. FASEB J. 2005; 19:1253–1265. [PubMed: 16051692] Dahlen E, Hedlund G, Dawe K. Low CD86 expression in the nonobese diabetic mouse results in the impairment of both T cell activation and CTLA-4 up-regulation. J Immunol. 2000; 164:2444– 2456. [PubMed: 10679081] Daikh DI, Finck BK, Linsley PS, Hollenbaugh D, Wofsy D. Long-term inhibition of murine lupus by brief simultaneous blockade of the B7/CD28 and CD40/gp39 costimulation pathways. J Immunol. 1997; 159:3104–3108. [PubMed: 9317105] Dendrou CA, Fugger L, Friese MA. Immunopathology of multiple sclerosis. Nat Rev Immunol. 2015; 15:545–558. [PubMed: 26250739] Dong C, Juedes AE, Temann UA, Shresta S, Allison JP, Ruddle NH, Flavell RA. ICOS co-stimulatory receptor is essential for T-cell activation and function. Nature. 2001; 409:97–101. [PubMed: 11343121] Dong H, Strome SE, Matteson EL, Moder KG, Flies DB, Zhu G, Tamura H, Driscoll CL, Chen L. Costimulating aberrant T cell responses by B7-H1 autoantibodies in rheumatoid arthritis. J Clin Invest. 2003; 111:363–370. [PubMed: 12569162] Early GS, Zhao W, Burns CM. Anti-CD40 ligand antibody treatment prevents the development of lupus-like nephritis in a subset of New Zealand black x New Zealand white mice. Response correlates with the absence of an anti-antibody response. J Immunol. 1996; 157:3159–3164. [PubMed: 8816428] Elyaman W, Kivisakk P, Reddy J, Chitnis T, Raddassi K, Imitola J, Bradshaw E, Kuchroo VK, Yagita H, Sayegh MH, Khoury SJ. Distinct functions of autoreactive memory and effector CD4+ T cells in experimental autoimmune encephalomyelitis. Am J Pathol. 2008; 173:411–422. [PubMed: 18583313] Emery P, Burmester GR, Bykerk VP, Combe BG, Furst DE, Barre E, Karyekar CS, Wong DA, Huizinga TW. Evaluating drug-free remission with abatacept in early rheumatoid arthritis: results from the phase 3b, multicentre, randomised, active-controlled AVERT study of 24 months, with a 12-month, double-blind treatment period. Ann Rheum Dis. 2015; 74:19–26. [PubMed: 25367713] Fife BT, Griffin MD, Abbas AK, Locksley RM, Bluestone JA. Inhibition of T cell activation and autoimmune diabetes using a B cell surface-linked CTLA-4 agonist. J Clin Invest. 2006; 116:2252–2261. [PubMed: 16886063] Fife BT, Pauken KE, Eagar TN, Obu T, Wu J, Tang Q, Azuma M, Krummel MF, Bluestone JA. Interactions between PD-1 and PD-L1 promote tolerance by blocking the TCR-induced stop signal. Nat Immunol. 2009; 10:1185–1192. [PubMed: 19783989] Finck BK, Linsley PS, Wofsy D. Treatment of murine lupus with CTLA4Ig. Science. 1994; 265:1225– 1227. [PubMed: 7520604] Foell J, Strahotin S, O’Neil SP, McCausland MM, Suwyn C, Haber M, Chander PN, Bapat AS, Yan XJ, Chiorazzi N, et al. CD137 costimulatory T cell receptor engagement reverses acute disease in lupus-prone NZB x NZW F1 mice. J Clin Invest. 2003; 111:1505–1518. [PubMed: 12750400] Ford ML, Adams AB, Pearson TC. Targeting co-stimulatory pathways: transplantation and autoimmunity. Nat Rev Nephrol. 2014; 10:14–24. [PubMed: 24100403] Furie R, Nicholls K, Cheng TT, Houssiau F, Burgos-Vargas R, Chen SL, Hillson JL, MeadowsShropshire S, Kinaszczuk M, Merrill JT. Efficacy and safety of abatacept in lupus nephritis: a twelve-month, randomized, double-blind study. Arthritis Rheumatol. 2014; 66:379–389. [PubMed: 24504810] Galicia G, Kasran A, Uyttenhove C, De Swert K, Van Snick J, Ceuppens JL. ICOS deficiency results in exacerbated IL-17 mediated experimental autoimmune encephalomyelitis. J Clin Immunol. 2009; 29:426–433. [PubMed: 19291374] Genovese MC, Becker JC, Schiff M, Luggen M, Sherrer Y, Kremer J, Birbara C, Box J, Natarajan K, Nuamah I, et al. Abatacept for rheumatoid arthritis refractory to tumor necrosis factor alpha inhibition. N Engl J Med. 2005; 353:1114–1123. [PubMed: 16162882]

Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 22

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Gerritse K, Laman JD, Noelle RJ, Aruffo A, Ledbetter JA, Boersma WJ, Claassen E. CD40-CD40 ligand interactions in experimental allergic encephalomyelitis and multiple sclerosis. Proc Natl Acad Sci U S A. 1996; 93:2499–2504. [PubMed: 8637903] Goodnow CC, Sprent J, Fazekas de St Groth B, Vinuesa CG. Cellular and genetic mechanisms of self tolerance and autoimmunity. Nature. 2005; 435:590–597. [PubMed: 15931211] Green EA, Wong FS, Eshima K, Mora C, Flavell RA. Neonatal tumor necrosis factor alpha promotes diabetes in nonobese diabetic mice by CD154-independent antigen presentation to CD8(+) T cells. J Exp Med. 2000; 191:225–238. [PubMed: 10637268] Grewal IS, Foellmer HG, Grewal KD, Xu J, Hardardottir F, Baron JL, Janeway CA Jr, Flavell RA. Requirement for CD40 ligand in costimulation induction, T cell activation, and experimental allergic encephalomyelitis. Science. 1996; 273:1864–1867. [PubMed: 8791592] Group AT. Treatment of lupus nephritis with abatacept: the Abatacept and Cyclophosphamide Combination Efficacy and Safety Study. Arthritis Rheumatol. 2014; 66:3096–3104. [PubMed: 25403681] Gwyer Findlay E, Danks L, Madden J, Cavanagh MM, McNamee K, McCann F, Snelgrove RJ, Shaw S, Feldmann M, Taylor PC, et al. OX40L blockade is therapeutic in arthritis, despite promoting osteoclastogenesis. Proc Natl Acad Sci U S A. 2014; 111:2289–2294. [PubMed: 24469824] Haanstra KG, Dijkman K, Bashir N, Bauer J, Mary C, Poirier N, Baker P, Scobie L, t Hart BA, Vanhove B. Selective blockade of CD28-mediated T cell costimulation protects rhesus monkeys against acute fatal experimental autoimmune encephalomyelitis. J Immunol. 2015; 194:1454– 1466. [PubMed: 25589073] Hawiger D, Tran E, Du W, Booth CJ, Wen L, Dong C, Flavell RA. ICOS mediates the development of insulin-dependent diabetes mellitus in nonobese diabetic mice. J Immunol. 2008; 180:3140–3147. [PubMed: 18292537] Herman AE, Freeman GJ, Mathis D, Benoist C. CD4+CD25+ T regulatory cells dependent on ICOS promote regulation of effector cells in the prediabetic lesion. J Exp Med. 2004; 199:1479–1489. [PubMed: 15184501] Herold KC, Vignali DA, Cooke A, Bluestone JA. Type 1 diabetes: translating mechanistic observations into effective clinical outcomes. Nat Rev Immunol. 2013; 13:243–256. [PubMed: 23524461] Howard LM, Miga AJ, Vanderlugt CL, Dal Canto MC, Laman JD, Noelle RJ, Miller SD. Mechanisms of immunotherapeutic intervention by anti-CD40L (CD154) antibody in an animal model of multiple sclerosis. J Clin Invest. 1999; 103:281–290. [PubMed: 9916140] Hu YL, Metz DP, Chung J, Siu G, Zhang M. B7RP-1 blockade ameliorates autoimmunity through regulation of follicular helper T cells. J Immunol. 2009; 182:1421–1428. [PubMed: 19155489] Huang CT, Workman CJ, Flies D, Pan X, Marson AL, Zhou G, Hipkiss EL, Ravi S, Kowalski J, Levitsky HI, et al. Role of LAG-3 in regulatory T cells. Immunity. 2004; 21:503–513. [PubMed: 15485628] Hurwitz AA, Sullivan TJ, Krummel MF, Sobel RA, Allison JP. Specific blockade of CTLA-4/B7 interactions results in exacerbated clinical and histologic disease in an actively-induced model of experimental allergic encephalomyelitis. J Neuroimmunol. 1997; 73:57–62. [PubMed: 9058759] Hurwitz AA, Sullivan TJ, Sobel RA, Allison JP. Cytotoxic T lymphocyte antigen-4 (CTLA-4) limits the expansion of encephalitogenic T cells in experimental autoimmune encephalomyelitis (EAE)resistant BALB/c mice. Proc Natl Acad Sci U S A. 2002; 99:3013–3017. [PubMed: 11867713] Ichinose K, Zhang Z, Koga T, Juang YT, Kis-Toth K, Sharpe AH, Kuchroo V, Crispin JC, Tsokos GC. Brief report: increased expression of a short splice variant of CTLA-4 exacerbates lupus in MRL/lpr mice. Arthritis Rheum. 2013; 65:764–769. [PubMed: 23203389] Irie J, Wu Y, Kachapati K, Mittler RS, Ridgway WM. Modulating protective and pathogenic CD4+ subsets via CD137 in type 1 diabetes. Diabetes. 2007; 56:186–196. [PubMed: 17192481] Iwai H, Kozono Y, Hirose S, Akiba H, Yagita H, Okumura K, Kohsaka H, Miyasaka N, Azuma M. Amelioration of collagen-induced arthritis by blockade of inducible costimulator-B7 hom*ologous protein costimulation. J Immunol. 2002; 169:4332–4339. [PubMed: 12370365] Jabs C, Greve B, Chang TT, Sobel RA, Sharpe AH, Kuchroo VK. Genetic background determines the requirement for B7 costimulation in induction of autoimmunity. Eur J Immunol. 2002; 32:2687– 2697. [PubMed: 12207354]

Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 23

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Jacquemin C, Schmitt N, Contin-Bordes C, Liu Y, Narayanan P, Seneschal J, Maurouard T, Dougall D, Davizon ES, Dumortier H, et al. OX40 Ligand Contributes to Human Lupus Pathogenesis by Promoting T Follicular Helper Response. Immunity. 2015; 42:1159–1170. [PubMed: 26070486] Joller N, Hafler JP, Brynedal B, Kassam N, Spoerl S, Levin SD, Sharpe AH, Kuchroo VK. Cutting edge: TIGIT has T cell-intrinsic inhibitory functions. J Immunol. 2011; 186:1338–1342. [PubMed: 21199897] Kachapati K, Adams DE, Wu Y, Steward CA, Rainbow DB, Wicker LS, Mittler RS, Ridgway WM. The B10 Idd9.3 locus mediates accumulation of functionally superior CD137(+) regulatory T cells in the nonobese diabetic type 1 diabetes model. J Immunol. 2012; 189:5001–5015. [PubMed: 23066155] Kachapati K, Bednar KJ, Adams DE, Wu Y, Mittler RS, Jordan MB, Hinerman JM, Herr AB, Ridgway WM. Recombinant soluble CD137 prevents type one diabetes in nonobese diabetic mice. J Autoimmun. 2013; 47:94–103. [PubMed: 24145149] Kageyama R, Cannons JL, Zhao F, Yusuf I, Lao C, Locci M, Schwartzberg PL, Crotty S. The receptor Ly108 functions as a SAP adaptor-dependent on-off switch for T cell help to B cells and NKT cell development. Immunity. 2012; 36:986–1002. [PubMed: 22683125] Kalunian KC, Davis JC Jr, Merrill JT, Totoritis MC, Wofsy D, Group ILS. Treatment of systemic lupus erythematosus by inhibition of T cell costimulation with anti-CD154: a randomized, double-blind, placebo-controlled trial. Arthritis Rheum. 2002; 46:3251–3258. [PubMed: 12483729] Karandikar NJ, Eagar TN, Vanderlugt CL, Bluestone JA, Miller SD. CTLA-4 downregulates epitope spreading and mediates remission in relapsing experimental autoimmune encephalomyelitis. J Neuroimmunol. 2000; 109:173–180. [PubMed: 10996219] Karandikar NJ, Vanderlugt CL, Eagar T, Tan L, Bluestone JA, Miller SD. Tissue-specific up-regulation of B7-1 expression and function during the course of murine relapsing experimental autoimmune encephalomyelitis. J Immunol. 1998; 161:192–199. [PubMed: 9647224] Karandikar NJ, Vanderlugt CL, Walunas TL, Miller SD, Bluestone JA. CTLA-4: a negative regulator of autoimmune disease. J Exp Med. 1996; 184:783–788. [PubMed: 8760834] Kasagi S, Kawano S, Okazaki T, Honjo T, Morinobu A, Hatachi S, Shimatani K, Tanaka Y, Minato N, Kumagai S. Anti-programmed cell death 1 antibody reduces CD4+PD-1+ T cells and relieves the lupus-like nephritis of NZB/W F1 mice. J Immunol. 2010; 184:2337–2347. [PubMed: 20139271] Keszei M, Detre C, Rietdijk ST, Munoz P, Romero X, Berger SB, Calpe S, Liao G, Castro W, Julien A, et al. A novel isoform of the Ly108 gene ameliorates murine lupus. J Exp Med. 2011; 208:811– 822. [PubMed: 21422172] Khoury SJ, Akalin E, Chandraker A, Turka LA, Linsley PS, Sayegh MH, Hanco*ck WW. CD28-B7 costimulatory blockade by CTLA4Ig prevents actively induced experimental autoimmune encephalomyelitis and inhibits Th1 but spares Th2 cytokines in the central nervous system. J Immunol. 1995; 155:4521–4524. [PubMed: 7594447] Kim YH, Choi BK, Shin SM, Kim CH, Oh HS, Park SH, Lee DG, Lee MJ, Kim KH, Vinay DS, Kwon BS. 4-1BB triggering ameliorates experimental autoimmune encephalomyelitis by modulating the balance between Th17 and regulatory T cells. J Immunol. 2011; 187:1120–1128. [PubMed: 21715692] Knoerzer DB, Karr RW, Schwartz BD, Mengle-Gaw LJ. Collagen-induced arthritis in the BB rat. Prevention of disease by treatment with CTLA-4-Ig. J Clin Invest. 1995; 96:987–993. [PubMed: 7543497] Ko HJ, Cho ML, Lee SY, Oh HJ, Heo YJ, Moon YM, Kang CM, Kwok SK, Ju JH, Park SH, et al. CTLA4-Ig modifies dendritic cells from mice with collagen-induced arthritis to increase the CD4+CD25+Foxp3+ regulatory T cell population. J Autoimmun. 2010; 34:111–120. [PubMed: 19665867] Koguchi K, Anderson DE, Yang L, O’Connor KC, Kuchroo VK, Hafler DA. Dysregulated T cell expression of TIM3 in multiple sclerosis. J Exp Med. 2006; 203:1413–1418. [PubMed: 16754722] Koh AE, Njoroge SW, Feliu M, Cook A, Selig MK, Latchman YE, Sharpe AH, Colvin RB, Paul E. The SLAM family member CD48 (Slamf2) protects lupus-prone mice from autoimmune nephritis. J Autoimmun. 2011; 37:48–57. [PubMed: 21561736]

Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 24

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Kornete M, Mason ES, Girouard J, Lafferty EI, Qureshi S, Piccirillo CA. Th1-Like ICOS+ Foxp3+ Treg Cells Preferentially Express CXCR3 and Home to beta-Islets during Pre-Diabetes in BDC2.5 NOD Mice. PLoS One. 2015; 10:e0126311. [PubMed: 25946021] Kornete M, Sgouroudis E, Piccirillo CA. ICOS-dependent homeostasis and function of Foxp3+ regulatory T cells in islets of nonobese diabetic mice. J Immunol. 2012; 188:1064–1074. [PubMed: 22227569] Kotani M, Hirata K, Ogawa S, Habiro K, Ishida Y, Tanuma S, Horai R, Iwakura Y, Kishimoto H, Abe R. CD28-dependent differentiation into the effector/memory phenotype is essential for induction of arthritis in interleukin-1 receptor antagonist-deficient mice. Arthritis Rheum. 2006; 54:473–481. [PubMed: 16453283] Kuchroo VK, Das MP, Brown JA, Ranger AM, Zamvil SS, Sobel RA, Weiner HL, Nabavi N, Glimcher LH. B7-1 and B7-2 costimulatory molecules activate differentially the Th1/Th2 developmental pathways: application to autoimmune disease therapy. Cell. 1995; 80:707–718. [PubMed: 7534215] Kyburz D, Corr M, Brinson DC, Von Damm A, Tighe H, Carson DA. Human rheumatoid factor production is dependent on CD40 signaling and autoantigen. J Immunol. 1999; 163:3116–3122. [PubMed: 10477577] Lafferty KJ, Cunningham AJ. A new analysis of allogeneic interactions. Aust J Exp Biol Med Sci. 1975; 53:27–42. [PubMed: 238498] Laman JD, t Hart BA, Brok H, Meurs M, Schellekens MM, Kasran A, Boon L, Bauer J, Boer M, Ceuppens J. Protection of marmoset monkeys against EAE by treatment with a murine antibody blocking CD40 (mu5D12). Eur J Immunol. 2002; 32:2218–2228. [PubMed: 12209634] Latchman YE, Liang SC, Wu Y, Chernova T, Sobel RA, Klemm M, Kuchroo VK, Freeman GJ, Sharpe AH. PD-L1-deficient mice show that PD-L1 on T cells, antigen-presenting cells, and host tissues negatively regulates T cells. Proc Natl Acad Sci U S A. 2004; 101:10691–10696. [PubMed: 15249675] Lenschow DJ, Herold KC, Rhee L, Patel B, Koons A, Qin HY, Fuchs E, Singh B, Thompson CB, Bluestone JA. CD28/B7 regulation of Th1 and Th2 subsets in the development of autoimmune diabetes. Immunity. 1996; 5:285–293. [PubMed: 8808683] Lenschow DJ, Ho SC, Sattar H, Rhee L, Gray G, Nabavi N, Herold KC, Bluestone JA. Differential effects of anti-B7-1 and anti-B7-2 monoclonal antibody treatment on the development of diabetes in the nonobese diabetic mouse. J Exp Med. 1995; 181:1145–1155. [PubMed: 7532678] Levin SD, Taft DW, Brandt CS, Bucher C, Howard ED, Chadwick EM, Johnston J, Hammond A, Bontadelli K, Ardourel D, et al. Vstm3 is a member of the CD28 family and an important modulator of T-cell function. Eur J Immunol. 2011; 41:902–915. [PubMed: 21416464] Liang B, Gee RJ, Kashgarian MJ, Sharpe AH, Mamula MJ. B7 costimulation in the development of lupus: autoimmunity arises either in the absence of B7.1/B7.2 or in the presence of anti-b7.1/B7.2 blocking antibodies. J Immunol. 1999; 163:2322–2329. [PubMed: 10438978] Liang B, Workman C, Lee J, Chew C, Dale BM, Colonna L, Flores M, Li N, Schweighoffer E, Greenberg S, et al. Regulatory T cells inhibit dendritic cells by lymphocyte activation gene-3 engagement of MHC class II. J Immunol. 2008; 180:5916–5926. [PubMed: 18424711] Liang SC, Latchman YE, Buhlmann JE, Tomczak MF, Horwitz BH, Freeman GJ, Sharpe AH. Regulation of PD-1, PD-L1, and PD-L2 expression during normal and autoimmune responses. Eur J Immunol. 2003; 33:2706–2716. [PubMed: 14515254] Lisnevskaia L, Murphy G, Isenberg D. Systemic lupus erythematosus. Lancet. 2014; 384:1878–1888. [PubMed: 24881804] Luhder F, Chambers C, Allison JP, Benoist C, Mathis D. Pinpointing when T cell costimulatory receptor CTLA-4 must be engaged to dampen diabetogenic T cells. Proc Natl Acad Sci U S A. 2000; 97:12204–12209. [PubMed: 11035773] Luhder F, Hoglund P, Allison JP, Benoist C, Mathis D. Cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) regulates the unfolding of autoimmune diabetes. J Exp Med. 1998; 187:427–432. [PubMed: 9449722]

Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 25

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Luo L, Zhu G, Xu H, Yao S, Zhou G, Zhu Y, Tamada K, Huang L, Flies AD, Broadwater M, et al. B7H3 Promotes Pathogenesis of Autoimmune Disease and Inflammation by Regulating the Activity of Different T Cell Subsets. PLoS One. 2015; 10:e0130126. [PubMed: 26065426] Lyons PA, Hanco*ck WW, Denny P, Lord CJ, Hill NJ, Armitage N, Siegmund T, Todd JA, Phillips MS, Hess JF, et al. The NOD Idd9 genetic interval influences the pathogenicity of insulitis and contains molecular variants of Cd30, Tnfr2, and Cd137. Immunity. 2000; 13:107–115. [PubMed: 10933399] Ma J, Xu J, Madaio MP, Peng Q, Zhang J, Grewal IS, Flavell RA, Craft J. Autoimmune lpr/lpr mice deficient in CD40 ligand: spontaneous Ig class switching with dichotomy of autoantibody responses. J Immunol. 1996; 157:417–426. [PubMed: 8683147] MacDonald KP, Nishioka Y, Lipsky PE, Thomas R. Functional CD40 ligand is expressed by T cells in rheumatoid arthritis. J Clin Invest. 1997; 100:2404–2414. [PubMed: 9410920] Magnus T, Schreiner B, Korn T, Jack C, Guo H, Antel J, Ifergan I, Chen L, Bischof F, Bar-Or A, Wiendl H. Microglial expression of the B7 family member B7 hom*olog 1 confers strong immune inhibition: implications for immune responses and autoimmunity in the CNS. J Neurosci. 2005; 25:2537–2546. [PubMed: 15758163] Manku H, Langefeld CD, Guerra SG, Malik TH, Alarcon-Riquelme M, Anaya JM, Bae SC, Boackle SA, Brown EE, Criswell LA, et al. Trans-ancestral studies fine map the SLE-susceptibility locus TNFSF4. PLoS Genet. 2013; 9:e1003554. [PubMed: 23874208] Martin-Orozco N, Chen Z, Poirot L, Hyatt E, Chen A, Kanagawa O, Sharpe A, Mathis D, Benoist C. Paradoxical dampening of anti-islet self-reactivity but promotion of diabetes by OX40 ligand. J Immunol. 2003; 171:6954–6960. [PubMed: 14662903] Martinez Gomez JM, Croxford JL, Yeo KP, Angeli V, Schwarz H, Gasser S. Development of experimental autoimmune encephalomyelitis critically depends on CD137 ligand signaling. J Neurosci. 2012; 32:18246–18252. [PubMed: 23238738] Merrill JT, Burgos-Vargas R, Westhovens R, Chalmers A, D’Cruz D, Wallace DJ, Bae SC, Sigal L, Becker JC, Kelly S, et al. The efficacy and safety of abatacept in patients with non-lifethreatening manifestations of systemic lupus erythematosus: results of a twelve-month, multicenter, exploratory, phase IIb, randomized, double-blind, placebo-controlled trial. Arthritis Rheum. 2010; 62:3077–3087. [PubMed: 20533545] Mihara M, Tan I, Chuzhin Y, Reddy B, Budhai L, Holzer A, Gu Y, Davidson A. CTLA4Ig inhibits T cell-dependent B-cell maturation in murine systemic lupus erythematosus. J Clin Invest. 2000; 106:91–101. [PubMed: 10880052] Miller SD, Vanderlugt CL, Lenschow DJ, Pope JG, Karandikar NJ, Dal Canto MC, Bluestone JA. Blockade of CD28/B7-1 interaction prevents epitope spreading and clinical relapses of murine EAE. Immunity. 1995; 3:739–745. [PubMed: 8777719] Monney L, Sabatos CA, Gaglia JL, Ryu A, Waldner H, Chernova T, Manning S, Greenfield EA, Coyle AJ, Sobel RA, et al. Th1-specific cell surface protein Tim-3 regulates macrophage activation and severity of an autoimmune disease. Nature. 2002; 415:536–541. [PubMed: 11823861] Mueller DL, Jenkins MK, Schwartz RH. Clonal expansion versus functional clonal inactivation: a costimulatory signalling pathway determines the outcome of T cell antigen receptor occupancy. Annu Rev Immunol. 1989; 7:445–480. [PubMed: 2653373] Nakajima A, Oshima H, Nohara C, Morimoto S, Yoshino S, Kobata T, Yagita H, Okumura K. Involvement of CD70-CD27 interactions in the induction of experimental autoimmune encephalomyelitis. J Neuroimmunol. 2000; 109:188–196. [PubMed: 10996221] Ndhlovu LC, Ishii N, Murata K, Sato T, Sugamura K. Critical involvement of OX40 ligand signals in the T cell priming events during experimental autoimmune encephalomyelitis. J Immunol. 2001; 167:2991–2999. [PubMed: 11509650] Nishimura H, Minato N, Nakano T, Honjo T. Immunological studies on PD-1 deficient mice: implication of PD-1 as a negative regulator for B cell responses. Int Immunol. 1998; 10:1563– 1572. [PubMed: 9796923] Nishimura H, Nose M, Hiai H, Minato N, Honjo T. Development of lupus-like autoimmune diseases by disruption of the PD-1 gene encoding an ITIM motif-carrying immunoreceptor. Immunity. 1999; 11:141–151. [PubMed: 10485649]

Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 26

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Nohara C, Akiba H, Nakajima A, Inoue A, Koh CS, Ohshima H, Yagita H, Mizuno Y, Okumura K. Amelioration of experimental autoimmune encephalomyelitis with anti-OX40 ligand monoclonal antibody: a critical role for OX40 ligand in migration, but not development, of pathogenic T cells. J Immunol. 2001; 166:2108–2115. [PubMed: 11160262] O’Neill SK, Cao Y, Hamel KM, Doodes PD, Hutas G, Finnegan A. Expression of CD80/86 on B cells is essential for autoreactive T cell activation and the development of arthritis. J Immunol. 2007; 179:5109–5116. [PubMed: 17911596] Odegard JM, Marks BR, DiPlacido LD, Poholek AC, Kono DH, Dong C, Flavell RA, Craft J. ICOSdependent extrafollicular helper T cells elicit IgG production via IL-21 in systemic autoimmunity. J Exp Med. 2008; 205:2873–2886. [PubMed: 18981236] Oflazoglu E, Boursalian TE, Zeng W, Edwards AC, Duniho S, McEarchern JA, Law CL, Gerber HP, Grewal IS. Blocking of CD27-CD70 pathway by anti-CD70 antibody ameliorates joint disease in murine collagen-induced arthritis. J Immunol. 2009; 183:3770–3777. [PubMed: 19710474] Okazaki T, Honjo T. PD-1 and PD-1 ligands: from discovery to clinical application. Int Immunol. 2007; 19:813–824. [PubMed: 17606980] Oliveira EM, Bar-Or A, Waliszewska AI, Cai G, Anderson DE, Krieger JI, Hafler DA. CTLA-4 dysregulation in the activation of myelin basic protein reactive T cells may distinguish patients with multiple sclerosis from healthy controls. J Autoimmun. 2003; 20:71–81. [PubMed: 12604314] Oliveira-dos-Santos AJ, Ho A, Tada Y, Lafaille JJ, Tonegawa S, Mak TW, Penninger JM. CD28 costimulation is crucial for the development of spontaneous autoimmune encephalomyelitis. J Immunol. 1999; 162:4490–4495. [PubMed: 10201986] Orban T, Bundy B, Becker DJ, Dimeglio LA, Gitelman SE, Goland R, Gottlieb PA, Greenbaum CJ, Marks JB, Monzavi R, et al. Costimulation modulation with abatacept in patients with recentonset type 1 diabetes: follow-up 1 year after cessation of treatment. Diabetes Care. 2014; 37:1069–1075. [PubMed: 24296850] Oya Y, Watanabe N, Kobayashi Y, Owada T, Oki M, Ikeda K, Suto A, Kagami S, Hirose K, Kishimoto T, Nakajima H. Lack of B and T lymphocyte attenuator exacerbates autoimmune disorders and induces Fas-independent liver injury in MRL-lpr/lpr mice. Int Immunol. 2011; 23:335–344. [PubMed: 21521881] Pakala SV, Bansal-Pakala P, Halteman BS, Croft M. Prevention of diabetes in NOD mice at a late stage by targeting OX40/OX40 ligand interactions. Eur J Immunol. 2004; 34:3039–3046. [PubMed: 15368274] Paterson AM, Brown KE, Keir ME, Vanguri VK, Riella LV, Chandraker A, Sayegh MH, Blazar BR, Freeman GJ, Sharpe AH. The programmed death-1 ligand 1:B7-1 pathway restrains diabetogenic effector T cells in vivo. J Immunol. 2011; 187:1097–1105. [PubMed: 21697456] Paterson AM, Lovitch SB, Sage PT, Juneja VR, Lee Y, Trombley JD, Arancibia-Carcamo CV, Sobel RA, Rudensky AY, Kuchroo VK, et al. Deletion of CTLA-4 on regulatory T cells during adulthood leads to resistance to autoimmunity. J Exp Med. 2015; 212:1603–1621. [PubMed: 26371185] Perrin PJ, Maldonado JH, Davis TA, June CH, Racke MK. CTLA-4 blockade enhances clinical disease and cytokine production during experimental allergic encephalomyelitis. J Immunol. 1996a; 157:1333–1336. [PubMed: 8759711] Perrin PJ, Scott D, Davis TA, Gray GS, Doggett MJ, Abe R, June CH, Racke MK. Opposing effects of CTLA4-Ig and anti-CD80 (B7-1) plus anti-CD86 (B7-2) on experimental allergic encephalomyelitis. J Neuroimmunol. 1996b; 65:31–39. [PubMed: 8642061] Pierer M, Schulz A, Rossol M, Kendzia E, Kyburz D, Haentzschel H, Baerwald C, Wagner U. Herpesvirus entry mediator-Ig treatment during immunization aggravates rheumatoid arthritis in the collagen-induced arthritis model. J Immunol. 2009; 182:3139–3145. [PubMed: 19234211] Podojil JR, Liu LN, Marshall SA, Chiang MY, Goings GE, Chen L, Langermann S, Miller SD. B7H4Ig inhibits mouse and human T-cell function and treats EAE via IL-10/Treg-dependent mechanisms. J Autoimmun. 2013; 44:71–81. [PubMed: 23683881] Prasad DV, Nguyen T, Li Z, Yang Y, Duong J, Wang Y, Dong C. Murine B7-H3 is a negative regulator of T cells. J Immunol. 2004; 173:2500–2506. [PubMed: 15294965]

Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 27

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Prasad DV, Richards S, Mai XM, Dong C. B7S1, a novel B7 family member that negatively regulates T cell activation. Immunity. 2003; 18:863–873. [PubMed: 12818166] Quattrocchi E, Dallman MJ, Feldmann M. Adenovirus-mediated gene transfer of CTLA-4Ig fusion protein in the suppression of experimental autoimmune arthritis. Arthritis Rheum. 2000; 43:1688–1697. [PubMed: 10943858] Racke MK, Scott DE, Quigley L, Gray GS, Abe R, June CH, Perrin PJ. Distinct roles for B7-1 (CD-80) and B7-2 (CD-86) in the initiation of experimental allergic encephalomyelitis. J Clin Invest. 1995; 96:2195–2203. [PubMed: 7593605] Raptopoulou AP, Bertsias G, Makrygiannakis D, Verginis P, Kritikos I, Tzardi M, Klareskog L, Catrina AI, Sidiropoulos P, Boumpas DT. The programmed death 1/programmed death ligand 1 inhibitory pathway is up-regulated in rheumatoid synovium and regulates peripheral T cell responses in human and murine arthritis. Arthritis Rheum. 2010; 62:1870–1880. [PubMed: 20506224] Reed JC, Herold KC. Thinking bedside at the bench: the NOD mouse model of T1DM. Nat Rev Endocrinol. 2015; 11:308–314. [PubMed: 25623120] Sabatos CA, Chakravarti S, Cha E, Schubart A, Sanchez-Fueyo A, Zheng XX, Coyle AJ, Strom TB, Freeman GJ, Kuchroo VK. Interaction of Tim-3 and Tim-3 ligand regulates T helper type 1 responses and induction of peripheral tolerance. Nat Immunol. 2003; 4:1102–1110. [PubMed: 14556006] Sage PT, Francisco LM, Carman CV, Sharpe AH. The receptor PD-1 controls follicular regulatory T cells in the lymph nodes and blood. Nat Immunol. 2013; 14:152–161. [PubMed: 23242415] Sage PT, Paterson AM, Lovitch SB, Sharpe AH. The coinhibitory receptor CTLA-4 controls B cell responses by modulating T follicular helper, T follicular regulatory, and T regulatory cells. Immunity. 2014; 41:1026–1039. [PubMed: 25526313] Sakaguchi S, Yamaguchi T, Nomura T, Ono M. Regulatory T cells and immune tolerance. Cell. 2008; 133:775–787. [PubMed: 18510923] Salomon B, Lenschow DJ, Rhee L, Ashourian N, Singh B, Sharpe A, Bluestone JA. B7/CD28 costimulation is essential for the homeostasis of the CD4+CD25+ immunoregulatory T cells that control autoimmune diabetes. Immunity. 2000; 12:431–440. [PubMed: 10795741] Sanchez-Fueyo A, Tian J, Picarella D, Domenig C, Zheng XX, Sabatos CA, Manlongat N, Bender O, Kamradt T, Kuchroo VK, et al. Tim-3 inhibits T helper type 1-mediated auto- and alloimmune responses and promotes immunological tolerance. Nat Immunol. 2003; 4:1093–1101. [PubMed: 14556005] Saverino D, Simone R, Bagnasco M, Pesce G. The soluble CTLA-4 receptor and its role in autoimmune diseases: an update. Auto Immun Highlights. 2010; 1:73–81. [PubMed: 26000110] Scholz C, Patton KT, Anderson DE, Freeman GJ, Hafler DA. Expansion of autoreactive T cells in multiple sclerosis is independent of exogenous B7 costimulation. J Immunol. 1998; 160:1532– 1538. [PubMed: 9570577] Schreiner B, Bailey SL, Shin T, Chen L, Miller SD. PD-1 ligands expressed on myeloid-derived APC in the CNS regulate T-cell responses in EAE. Eur J Immunol. 2008; 38:2706–2717. [PubMed: 18825752] Schwartz RH. Acquisition of immunologic self-tolerance. Cell. 1989; 57:1073–1081. [PubMed: 2525422] Seki M, Oomizu S, Sakata KM, Sakata A, Arikawa T, Watanabe K, Ito K, Takesh*ta K, Niki T, Saita N, et al. Galectin-9 suppresses the generation of Th17, promotes the induction of regulatory T cells, and regulates experimental autoimmune arthritis. Clin Immunol. 2008; 127:78–88. [PubMed: 18282810] Seo SK, Choi JH, Kim YH, Kang WJ, Park HY, Suh JH, Choi BK, Vinay DS, Kwon BS. 4-1BBmediated immunotherapy of rheumatoid arthritis. Nat Med. 2004; 10:1088–1094. [PubMed: 15448685] Sporici RA, Beswick RL, von Allmen C, Rumbley CA, Hayden-Ledbetter M, Ledbetter JA, Perrin PJ. ICOS ligand costimulation is required for T-cell encephalitogenicity. Clin Immunol. 2001; 100:277–288. [PubMed: 11513541]

Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 28

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Stumpf M, Zhou X, Bluestone JA. The B7-independent isoform of CTLA-4 functions to regulate autoimmune diabetes. J Immunol. 2013; 190:961–969. [PubMed: 23293354] Subudhi SK, Zhou P, Yerian LM, Chin RK, Lo JC, Anders RA, Sun Y, Chen L, Wang Y, Alegre ML, Fu YX. Local expression of B7-H1 promotes organ-specific autoimmunity and transplant rejection. J Clin Invest. 2004; 113:694–700. [PubMed: 14991067] Sun Y, Chen HM, Subudhi SK, Chen J, Koka R, Chen L, Fu YX. Costimulatory molecule-targeted antibody therapy of a spontaneous autoimmune disease. Nat Med. 2002a; 8:1405–1413. [PubMed: 12426559] Sun Y, Lin X, Chen HM, Wu Q, Subudhi SK, Chen L, Fu YX. Administration of agonistic anti-4-1BB monoclonal antibody leads to the amelioration of experimental autoimmune encephalomyelitis. J Immunol. 2002b; 168:1457–1465. [PubMed: 11801689] Swallow MM, Wallin JJ, Sha WC. B7h, a novel costimulatory hom*olog of B7.1 and B7.2, is induced by TNFalpha. Immunity. 1999; 11:423–432. [PubMed: 10549624] Sytwu HK, Lin WD, Roffler SR, Hung JT, Sung HS, Wang CH, Cheng TL, Tsou SC, Hsi SC, Shen KL. Anti-4-1BB-based immunotherapy for autoimmune diabetes: lessons from a transgenic nonobese diabetic (NOD) model. J Autoimmun. 2003; 21:247–254. [PubMed: 14599849] Tada Y, Nagasawa K, Ho A, Morito F, Koarada S, Ushiyama O, Suzuki N, Ohta A, Mak TW. Role of the costimulatory molecule CD28 in the development of lupus in MRL/lpr mice. J Immunol. 1999a; 163:3153–3159. [PubMed: 10477582] Tada Y, Nagasawa K, Ho A, Morito F, Ushiyama O, Suzuki N, Ohta H, Mak TW. CD28-deficient mice are highly resistant to collagen-induced arthritis. J Immunol. 1999b; 162:203–208. [PubMed: 9886387] Taha Khalaf A, Song JQ, Gao TT, Yu XP, Lei TC. CTLA-4 gene polymorphism and the risk of systemic lupus erythematosus in the Chinese population. J Biomed Biotechnol. 2011; 2011:167395. [PubMed: 21915163] Teichmann LL, Cullen JL, Kashgarian M, Dong C, Craft J, Shlomchik MJ. Local triggering of the ICOS coreceptor by CD11c(+) myeloid cells drives organ inflammation in lupus. Immunity. 2015; 42:552–565. [PubMed: 25786178] Tellander AC, Michaelsson E, Brunmark C, Andersson M. Potent adjuvant effect by anti-CD40 in collagen-induced arthritis. Enhanced disease is accompanied by increased production of collagen type-II reactive IgG2a and IFN-gamma. J Autoimmun. 2000; 14:295–302. [PubMed: 10882055] Topalian SL, Drake CG, Pardoll DM. Immune checkpoint blockade: a common denominator approach to cancer therapy. Cancer Cell. 2015; 27:450–461. [PubMed: 25858804] Tran CN, Thacker SG, Louie DM, Oliver J, White PT, Endres JL, Urquhart AG, Chung KC, Fox DA. Interactions of T cells with fibroblast-like synoviocytes: role of the B7 family costimulatory ligand B7-H3. J Immunol. 2008; 180:2989–2998. [PubMed: 18292521] Ueda H, Howson JM, Esposito L, Heward J, Snook H, Chamberlain G, Rainbow DB, Hunter KM, Smith AN, Di Genova G, et al. Association of the T-cell regulatory gene CTLA4 with susceptibility to autoimmune disease. Nature. 2003; 423:506–511. [PubMed: 12724780] van den Berg WB. Lessons from animal models of arthritis over the past decade. Arthritis Res Ther. 2009; 11:250. [PubMed: 19849822] Verhagen J, Gabrysova L, Minaee S, Sabatos CA, Anderson G, Sharpe AH, Wraith DC. Enhanced selection of FoxP3+ T-regulatory cells protects CTLA-4-deficient mice from CNS autoimmune disease. Proc Natl Acad Sci U S A. 2009; 106:3306–3311. [PubMed: 19218450] Verhagen J, Genolet R, Britton GJ, Stevenson BJ, Sabatos-Peyton CA, Dyson J, Luescher IF, Wraith DC. CTLA-4 controls the thymic development of both conventional and regulatory T cells through modulation of the TCR repertoire. Proc Natl Acad Sci U S A. 2013; 110:E221–230. [PubMed: 23267099] Viglietta V, Bourcier K, Buckle GJ, Healy B, Weiner HL, Hafler DA, Egorova S, Guttmann CR, Rusche JR, Khoury SJ. CTLA4Ig treatment in patients with multiple sclerosis: an open-label, phase 1 clinical trial. Neurology. 2008; 71:917–924. [PubMed: 18794494] Vignali DA, Collison LW, Workman CJ. How regulatory T cells work. Nat Rev Immunol. 2008; 8:523–532. [PubMed: 18566595]

Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 29

Author Manuscript Author Manuscript Author Manuscript Author Manuscript

Vijayakrishnan L, Slavik JM, Illes Z, Greenwald RJ, Rainbow D, Greve B, Peterson LB, Hafler DA, Freeman GJ, Sharpe AH, et al. An autoimmune disease-associated CTLA-4 splice variant lacking the B7 binding domain signals negatively in T cells. Immunity. 2004; 20:563–575. [PubMed: 15142525] Vinay DS, Choi JH, Kim JD, Choi BK, Kwon BS. Role of endogenous 4-1BB in the development of systemic lupus erythematosus. Immunology. 2007; 122:394–400. [PubMed: 17608689] Vogel I, Kasran A, Cremer J, Kim YJ, Boon L, Van Gool SW, Ceuppens JL. CD28/CTLA-4/B7 costimulatory pathway blockade affects regulatory T-cell function in autoimmunity. Eur J Immunol. 2015; 45:1832–1841. [PubMed: 25727069] Wan B, Nie H, Liu A, Feng G, He D, Xu R, Zhang Q, Dong C, Zhang JZ. Aberrant regulation of synovial T cell activation by soluble costimulatory molecules in rheumatoid arthritis. J Immunol. 2006; 177:8844–8850. [PubMed: 17142787] Wang CJ, Chou FC, Chu CH, Wu JC, Lin SH, Chang DM, Sytwu HK. Protective role of programmed death 1 ligand 1 (PD-L1)in nonobese diabetic mice: the paradox in transgenic models. Diabetes. 2008; 57:1861–1869. [PubMed: 18420489] Wang J, Yoshida T, Nakaki F, Hiai H, Okazaki T, Honjo T. Establishment of NOD-Pdcd1−/− mice as an efficient animal model of type I diabetes. Proc Natl Acad Sci U S A. 2005; 102:11823–11828. [PubMed: 16087865] Wang X, Hao J, Metzger DL, Mui A, Ao Z, Akhoundsadegh N, Langermann S, Liu L, Chen L, Ou D, et al. Early treatment of NOD mice with B7-H4 reduces the incidence of autoimmune diabetes. Diabetes. 2011; 60:3246–3255. [PubMed: 21984581] Wang X, Huang W, Mihara M, Sinha J, Davidson A. Mechanism of action of combined short-term CTLA4Ig and anti-CD40 ligand in murine systemic lupus erythematosus. J Immunol. 2002; 168:2046–2053. [PubMed: 11823542] Wang X, Huang W, Schiffer LE, Mihara M, Akkerman A, Hiromatsu K, Davidson A. Effects of antiCD154 treatment on B cells in murine systemic lupus erythematosus. Arthritis Rheum. 2003; 48:495–506. [PubMed: 12571860] Watanabe N, Gavrieli M, Sedy JR, Yang J, Fallarino F, Loftin SK, Hurchla MA, Zimmerman N, Sim J, Zang X, et al. BTLA is a lymphocyte inhibitory receptor with similarities to CTLA-4 and PD-1. Nat Immunol. 2003; 4:670–679. [PubMed: 12796776] Webb LM, Walmsley MJ, Feldmann M. Prevention and amelioration of collagen-induced arthritis by blockade of the CD28 co-stimulatory pathway: requirement for both B7-1 and B7-2. Eur J Immunol. 1996; 26:2320–2328. [PubMed: 8898940] Weinberg AD, Bourdette DN, Sullivan TJ, Lemon M, Wallin JJ, Maziarz R, Davey M, Palida F, Godfrey W, Engleman E, et al. Selective depletion of myelin-reactive T cells with the anti-OX-40 antibody ameliorates autoimmune encephalomyelitis. Nat Med. 1996; 2:183–189. [PubMed: 8574963] Weinberg AD, Wegmann KW, Funatake C, Whitham RH. Blocking OX-40/OX-40 ligand interaction in vitro and in vivo leads to decreased T cell function and amelioration of experimental allergic encephalomyelitis. J Immunol. 1999; 162:1818–1826. [PubMed: 9973447] Westhovens R, Kremer JM, Emery P, Russell AS, Alten R, Barre E, Dougados M. Long-term safety and efficacy of abatacept in patients with rheumatoid arthritis and an inadequate response to methotrexate: a 7-year extended study. Clin Exp Rheumatol. 2014; 32:553–562. [PubMed: 25005467] Wicker LS, Chamberlain G, Hunter K, Rainbow D, Howlett S, Tiffen P, Clark J, Gonzalez-Munoz A, Cumiskey AM, Rosa RL, et al. Fine mapping, gene content, comparative sequencing, and expression analyses support Ctla4 and Nramp1 as candidates for Idd5.1 and Idd5.2 in the nonobese diabetic mouse. J Immunol. 2004; 173:164–173. [PubMed: 15210771] Wong M, La Cava A, Hahn BH. Blockade of programmed death-1 in young (New Zealand Black x New Zealand White)F1 mice promotes the suppressive capacity of CD4+ regulatory T cells protecting from lupus-like disease. J Immunol. 2013; 190:5402–5410. [PubMed: 23636058] Wong M, La Cava A, Singh RP, Hahn BH. Blockade of programmed death-1 in young (New Zealand black x New Zealand white)F1 mice promotes the activity of suppressive CD8+ T cells that protect from lupus-like disease. J Immunol. 2010; 185:6563–6571. [PubMed: 21041733]

Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 30

Author Manuscript Author Manuscript

Workman CJ, Dugger KJ, Vignali DA. Cutting edge: molecular analysis of the negative regulatory function of lymphocyte activation gene-3. J Immunol. 2002; 169:5392–5395. [PubMed: 12421911] Yang L, Anderson DE, Kuchroo J, Hafler DA. Lack of TIM-3 immunoregulation in multiple sclerosis. J Immunol. 2008; 180:4409–4414. [PubMed: 18354161] Yang Y, Wilson JM. CD40 ligand-dependent T cell activation: requirement of B7-CD28 signaling through CD40. Science. 1996; 273:1862–1864. [PubMed: 8791591] Yoshioka T, Nakajima A, Akiba H, Ishiwata T, Asano G, Yoshino S, Yagita H, Okumura K. Contribution of OX40/OX40 ligand interaction to the pathogenesis of rheumatoid arthritis. Eur J Immunol. 2000; 30:2815–2823. [PubMed: 11069062] Zhu B, Guleria I, Khosroshahi A, Chitnis T, Imitola J, Azuma M, Yagita H, Sayegh MH, Khoury SJ. Differential role of programmed death-ligand 1 [corrected] and programmed death-ligand 2 [corrected] in regulating the susceptibility and chronic progression of experimental autoimmune encephalomyelitis. J Immunol. 2006; 176:3480–3489. [PubMed: 16517716] Zhu C, Anderson AC, Schubart A, Xiong H, Imitola J, Khoury SJ, Zheng XX, Strom TB, Kuchroo VK. The Tim-3 ligand galectin-9 negatively regulates T helper type 1 immunity. Nat Immunol. 2005; 6:1245–1252. [PubMed: 16286920] Zucchelli S, Holler P, Yamagata T, Roy M, Benoist C, Mathis D. Defective central tolerance induction in NOD mice: genomics and genetics. Immunity. 2005; 22:385–396. [PubMed: 15780994]

Author Manuscript Author Manuscript Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 31

Author Manuscript Author Manuscript Figure 1. Co-stimulatory and co-inhibitory pathways in Systemic Lupus Erythematosus (SLE)

Author Manuscript

The hierarchical utilization of co-stimulatory and co-inhibitory receptors and their corresponding ligands is compared in SLE. The extent to which a co-stimulatory or coinhibitory molecule is utilized is indicted by dark (dominant utilization - red or blue) or light shading (partial utilization - pink or cyan). Molecules that appear to exhibit differential or controversial function (mixed bluered) in certain diseases, or for which there are no data (grey shading) are also noted.

Author Manuscript Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 32

Author Manuscript Author Manuscript Figure 2. Co-stimulatory and co-inhibitory pathways in Rheumatoid Arthritis (RA)

Author Manuscript

The hierarchical utilization of co-stimulatory and co-inhibitory receptors and their corresponding ligands is compared in RA. The extent to which a co-stimulatory or coinhibitory molecule is utilized is indicted by dark (dominant utilization - red or blue) or light shading (partial utilization - pink or cyan). Molecules that appear to exhibit differential or controversial function (mixed bluered) in certain diseases, or for which there are no data (grey shading) are also noted.

Author Manuscript Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 33

Author Manuscript Author Manuscript Author Manuscript

Figure 3. Co-stimulatory and co-inhibitory pathways in Multiple Sclerosis (MS)

The hierarchical utilization of co-stimulatory and co-inhibitory receptors and their corresponding ligands is compared in MS. The extent to which a co-stimulatory or coinhibitory molecule is utilized is indicted by dark (dominant utilization - red or blue) or light shading (partial utilization - pink or cyan). Molecules that appear to exhibit differential or controversial function (mixed bluered) in certain diseases, or for which there are no data (grey shading) are also noted.

Author Manuscript Immunity. Author manuscript; available in PMC 2017 May 17.

Zhang and Vignali

Page 34

Author Manuscript Author Manuscript Author Manuscript

Figure 4. Co-stimulatory and co-inhibitory pathways in Type 1 Diabetes (T1D)

The hierarchical utilization of co-stimulatory and co-inhibitory receptors and their corresponding ligands is compared in T1D. The extent to which a co-stimulatory or coinhibitory molecule is utilized is indicted by dark (dominant utilization - red or blue) or light shading (partial utilization - pink or cyan). Molecules that appear to exhibit differential or controversial function (mixed bluered) in certain diseases, or for which there are no data (grey shading) are also noted.

Author Manuscript Immunity. Author manuscript; available in PMC 2017 May 17.

Co-stimulatory and Co-inhibitory Pathways in Autoimmunity. - PDF Download Free (2024)

FAQs

What are co-stimulatory and co-inhibitory molecules? ›

a | Co-stimulatory molecules deliver positive signals to T cells following their engagement by ligands and counter-receptors on antigen-presenting cells (APCs). Several co-stimulatory molecule interactions are bidirectional. b | Co-inhibitory molecules deliver negative signals into T cells.

What is autoimmune disorder pdf? ›

Autoimmune is a pathological condition in which there is malfunctioning of immune system and the immune system is activated causing the destruction of its own cells since it serve its own cells as foreign cells.

What is autoimmune disease slideshare? ›

8.1K views•15 slides. Autoimmunity is the system of immune responses of an organism against its own healthy cells and tissues. Any disease that results from such an aberrant immune response is termed an "autoimmune disease".

What is the introduction of autoimmune disease? ›

Autoimmune disease occurs when the immune system attacks self-molecules as a result of a breakdown of immunologic tolerance to autoreactive immune cells. Many autoimmune disorders have been strongly associated with genetic, infectious, and/or environmental predisposing factors.

Why is co-stimulation important? ›

The co-stimulatory signal is necessary for T cell proliferation, differentiation and survival. Activation of T cells without co-stimulation may lead to the unresponsiveness of the T cell (also called anergy), apoptosis or the acquisition of the immune tolerance.

What are co-inhibitory receptors? ›

Co-inhibitory receptors play an important role in several T-cell subsets including activated T cells, regulatory T cells, and exhausted T cells. In activated T cells, co-inhibitory receptors control and contract the expanded T-cell population.

What is the root cause of autoimmune disease? ›

On a basic level, autoimmune disease occurs because the body's natural defenses — the immune system — attack the body's own healthy tissue. Researchers have several ideas about why this happens. When the body senses danger from a virus or infection, the immune system kicks into gear and attacks it.

What are the three main autoimmune diseases? ›

While there are more than 100 different kinds of autoimmune disorders, the most common ones include:
  • Inflammatory bowel disease (Crohn's disease, ulcerative colitis)
  • Rheumatoid arthritis (RA)
  • Lupus.
  • Type 1 diabetes.
  • Multiple sclerosis (MS)
  • Scleroderma.
  • Psoriasis.
  • Psoriatic arthritis.
Oct 3, 2023

What is the most autoimmune disease? ›

The most common autoimmune diseases are psoriatic arthritis, rheumatoid arthritis, connective tissue diseases, multiple sclerosis, autoimmune thyroid diseases such as Hashimoto's, celiac disease, and inflammatory bowel disease.

What is the difference between autoimmunity and autoimmune disease? ›

Autoimmunity is present in all individuals and increases with age; however, autoimmune disease occurs only in those individuals in whom the breakdown of one or more of the basic mechanisms regulating immune tolerance results in self-reactivity that can cause tissue damage.

What is the pathophysiology of autoimmune disease? ›

1.2 Pathogenesis of autoimmune diseases

Autoimmune diseases may be a result of genetic and environmental triggers that progress through a series of preclinical phases, involving dysregulation of the immune system and subsequent tissue inflammation prior to clinical signs and symptoms.

What is the mechanism of autoimmunity? ›

Autoimmunity refers to an aberration in the body's normal development such that the immune system mounts an attack against its own cells. [1] The etiology behind autoimmune diseases is multifactorial, with genetic, hormonal, and environmental factors all playing a role.

What does autoimmune fatigue feel like? ›

It's a feeling of constant exhaustion that makes it hard to get through the day, let alone participate in activities you enjoy.

What drugs trigger autoimmune disease? ›

  • Sulfasalazine.
  • Antithyroid.
  • Anticonvulsants: Ethosuximide Phenytoin Primidone Valproate Zonisamide Carbamazepine.
  • Statins: Lovastatin Simvastatin Fluvastatin Pravastatin Atorvastatin.
  • Terbinafine.
  • Penicillamine.
  • Fluorouracil agents.
  • Hydrochlorothiazide.
Jan 20, 2016

What kind of doctor is best for autoimmune disease? ›

While rheumatologists treat diseases of your musculoskeletal system, immunologists focus on your immune system. Both rheumatologists and immunologists can help if you're dealing with an autoimmune disease that affects your muscles, bones, or joints.

What are inhibitory molecules? ›

Small molecule inhibitors include essential primary metabolites that inhibit upstream enzymes that produce those metabolites. This provides a negative feedback loop that prevents over production of metabolites and thus maintains cellular homeostasis (steady internal conditions).

What is the role of co-stimulatory molecules in T helper cell differentiation? ›

Costimulatory molecules are a heterogenous group of cell surface molecules that act to amplify or counteract the initial activating signals provided to T cells from the T cell receptor following its interaction with an antigen/major histocompatibility complex, thereby influencing T cell differentiation and fate.

What is co inhibition? ›

Co-inhibition, a controller of homeostasis, antigen specific responses or both? The engagement of a co-inhibitory receptor with its ligand could influence the homeostasis of T cells. Blockade or absence of co-inhibitory molecules induced expansion of antigen-specific reactive T cells.

What are co receptors in immunology? ›

In immunology in particular, the term "co-receptor" often describes a secondary receptor used by a pathogen to gain access to the cell, or a receptor that works alongside T cell receptors such as CD4, CD8, or CD28 to bind antigens or regulate T cell activity in some way.

References

Top Articles
Latest Posts
Article information

Author: Duncan Muller

Last Updated:

Views: 6242

Rating: 4.9 / 5 (59 voted)

Reviews: 82% of readers found this page helpful

Author information

Name: Duncan Muller

Birthday: 1997-01-13

Address: Apt. 505 914 Phillip Crossroad, O'Konborough, NV 62411

Phone: +8555305800947

Job: Construction Agent

Hobby: Shopping, Table tennis, Snowboarding, Rafting, Motor sports, Homebrewing, Taxidermy

Introduction: My name is Duncan Muller, I am a enchanting, good, gentle, modern, tasty, nice, elegant person who loves writing and wants to share my knowledge and understanding with you.