Uploaded by dmasyithahd

Interferon-γ and Systemic Autoimmunity

advertisement
NIH Public Access
Author Manuscript
Discov Med. Author manuscript; available in PMC 2014 September 01.
NIH-PA Author Manuscript
Published in final edited form as:
Discov Med. 2013 September ; 16(87): 123–131.
Interferon-γ and Systemic Autoimmunity
K.M. Pollarda, D.M. Cauvib, C.B Toomeyc, K.V. Morrisa,d, and D.H. Konoe
aDepartment of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla,
CA 92037, USA
bDepartment
of Surgery and Center for Investigations of Health and Education Disparities,
University of California San Diego, La Jolla, CA 92037, USA
cDepartment
of Ophthalmology, Duke University School of Medicine, Albert Eye Research
Institute, Durham, NC 27710, USA
dThe
University of New South Wales, Biotechnology and Biomedical Sciences, Sydney, NSW
2052, Australia
NIH-PA Author Manuscript
eDepartment
of Immunology and Microbial Science, The Scripps Research Institute, La Jolla, CA
92037, USA
Abstract
NIH-PA Author Manuscript
The term interferon describes a family of proteins consisting of three major types (I, II, III) which
differ in their primary protein sequences, cognate receptors, genetic loci, and cell types
responsible for their production. The interferons, especially types I and II, overlap significantly in
the genes they control resulting in a shared spectrum of diverse biological effects which includes
regulation of both the innate and adaptive immune responses. As such, the interferons are major
effectors in the pathogenesis of autoimmunity, especially systemic autoimmunity. The type I IFNs,
because they are produced during the early stages of the innate immune response, are thought to
play the foremost role in autoimmune responses. However, numerous studies have found that the
single type II IFN, IFN-γ, plays an essential role in the development and severity of systemic
autoimmunity, particularly systemic lupus erythematosus. This is supported by animal studies
where IFN-γ is uniformly required in both spontaneous and induced models of lupus. Although
expression of IFN-γ in cells of the innate immune system is almost immediate after activation,
expression in adaptive immunity requires a complex orchestration of cellular interactions,
signaling events and epigenetic modifications. The multifaceted nature of IFN-γ in adaptive
immunity identifies numerous possible therapeutic targets that, because of the essential
contribution of IFN-γ to systemic autoimmunity, have the potential for producing significant
benefits.
Introduction
The term interferon (IFN) was first coined in 1957 to describe a factor with the ability to
interfere with the growth of live influenza virus (Isaacs & Lindenmann, 1957). Since then, it
was found that the IFNs are a family of proteins consisting of three major types; Type I
(IFN-α, -β, -ε, -ω), Type II (IFN-γ), and Type III (IFN-λ1, IFN-λ2 and IFN-λ3, also called
IL-29, IL-28A and IL-28B respectively) (Meyer, 2009). All three major types differ in their
Corresponding Author: K. Michael Pollard, PhD. Department of Molecular and Experimental Medicine MEM125, The Scripps
Research Institute, 10550 North Torrey Pines Road, La Jolla, CA 92037. Tel: 858 784 9124, Fax: 858 784 8836,
[email protected].
Disclosure
The authors report no conflict of interest.
Pollard et al.
Page 2
primary protein sequences, cognate receptors, genetic loci, and cell types responsible for
their production (Table 1).
NIH-PA Author Manuscript
Together the interferons influence a wide variety of biological responses including providing
protection from viral and bacterial infections, anti-tumor effects, and regulation of effector
cells in both innate and adaptive immune responses (Baccala et al., 2005; Hertzog et al.,
2011). Included among this diverse spectrum, are a significant amount of shared biological
effects due to a remarkable overlap in the many hundreds of genes regulated by the different
types of interferon, especially types I and II (Hertzog et al., 2011). Indeed, almost 70% of
genes regulated by IFN-γ are also regulated by type I IFN (Hertzog et al., 2011). Such
redundancy may be explained by the use of some of the same components of the JAK (Janus
kinase)-STAT (signal transducer and activator of transcripton) signaling pathways (Hu &
Ivashkiv, 2009; O’Shea & Plenge, 2012). Of the remaining non redundant genes affected by
IFN-γ, over 2/3 are inducible and the remainder are repressed. Thus, although overlapping
considerably with the type I IFN response, IFN-γ uniquely regulates a significant number of
genes, many of which contribute to the particular actions of IFN-γ in host defense,
inflammation and autoimmune disease (Billiau & Matthys, 2009). In this review we will
address the biology of IFN-γ and the contribution of IFN-γ to the development and
pathogenesis of systemic autoimmunity in both humans and animal models.
NIH-PA Author Manuscript
IFN-γ biology
IFN-γ, is secreted predominantly by T cells and natural killer (NK) cells (Billiau & Matthys,
2009) and, to a lesser extent, by other cell types such as macrophages, dendritic cells (DC)
and B cells (Meyer, 2009) (Table 1). The broad array of IFN-γ responses are mediated by
the cell-specific expression of many hundreds of IFN-γ-regulated genes (Hertzog et al.,
2011), for which functional classification encompasses inflammatory mediators, signaling
molecules, transcriptional activators, mediators of apoptosis and immune modulators (de
Veer et al., 2001). Further adding to the pleiotropic nature of the IFN-γ response are the
variety of cell types that possess the IFN-γ receptor (IFN-γR) and the molecular events that
constitute IFN-γ-dependent signaling pathways (Hu & Ivashkiv, 2009; Stark et al., 1998).
NIH-PA Author Manuscript
During innate immune responses IFN-γ is produced by NK and natural killer T (NKT) cells
(Schoenborn & Wilson, 2007) as well as macrophages and DCs (Meyer, 2009). In adaptive
immunity it is produced by CD8+ T cells in the control of infection, and by the CD4+ T
helper 1 (Th1) subset, which promotes inflammatory responses, clearance of intracellular
pathogens and class-switching to IgG2a, IgG2b and IgG3 (Baudino et al., 2006; Szabo et al.,
2003). IFN-γ is one of the main cytokines that distinguishes Th1 from other CD4+ subsets,
including Th2, Th17, follicular T helper (Tfh) and T regulatory (Treg) cells (Zhu et al.,
2010). Differentiation of CD4+ T cells to the Th1 subset is driven primarily by IL-12 in the
absence of IL-4 and TGF-β (Zhu et al., 2010). Subsequently, IFN-γ production is sustained
in this subset by many factors, including IL-12, IL-18, IL-27, and Stat4 (Murphy &
Stockinger, 2010; Paunovic et al., 2008; Szabo et al., 2003).
Regulation of IFN-γ expression
Regulation of IFN-γ expression in innate and adaptive immune responses has been
extensively reviewed elsewhere (Aune et al., 2013; Schoenborn & Wilson, 2007) and only a
basic outline will be presented here. We will discuss in greater detail more recent
information suggesting that long non-coding RNAs (lncRNAs) play a significant role in
IFN-γ transcription.
Discov Med. Author manuscript; available in PMC 2014 September 01.
Pollard et al.
Page 3
Transcription and epigenetics
NIH-PA Author Manuscript
Epigenetic regulation is an important mechanism of transcriptional activation or repression
of the Ifng locus (encoding IFN-γ) in innate and adaptive immune responses (Aune et al.,
2013; Schoenborn & Wilson, 2007). Regulation of IFN-γ involves epigenetic modifications
in the promoter region, conserved non-coding sequences such as CNS-6 and CNS-22 that
help regulate IFN-γ expression in Th1 CD4+ T cells (Schoenborn & Wilson, 2007), and
binding of T-box transcription factor TBX21 (T-bet) which directs Th1 lineage commitment
(Beima et al., 2006; Schoenborn & Wilson, 2007).
IFN-γ expressing cells involved in the innate immune response such as NK and NKT cells
rapidly produce the cytokine due to constitutive expression of transcription factors Eomes
and T-bet (NK cells) or T-bet alone (NKT) which are able to bind to regulatory elements
that are already accessible within the Ifng locus, leading to activation of Ifng transcription.
In contrast, IFN-γ expression by CD4+ and CD8+ T cells in adaptive immune responses
requires induction of T-bet as well as epigenetic modifications within the Ifng gene that
make regulatory elements accessible to T-bet and other transcription factors. This
differentiation process of naïve CD4+ or CD8+ T cells to Th1 or cytotoxic T effector cells
requires T cell receptor (TCR) stimulation and multiple rounds of cell division (Aune et al.,
2013; Schoenborn & Wilson, 2007).
NIH-PA Author Manuscript
lncRNA
Within the last eight years, it has become apparent that anti-sense non-coding RNAs
(ncRNAs) operate as an endogenous cellular mechanism to selectively regulate gene
transcription (Hawkins & Morris, 2008). While there are several distinct mechanisms by
which ncRNAs affect gene expression, one mechanism in particular involves anti-sense long
non-coding RNAs (lncRNAs) that function to specifically recruit epigenetic silencing
complexes to homology containing loci (Hawkins & Morris, 2010; Johnsson et al., 2013;
Morris et al., 2008; Yu et al., 2008). These antisense lncRNAs are distinct from small noncoding RNA effector molecules such as microRNAs (miRNAs) and piwi associated RNAs
(piRNAs) (Hartig et al., 2007), which function predominantly through the RNAi posttranscriptional silencing pathway. Although significant advances have been made in
defining the role of miRNAs and piRNAs in gene regulation, far less is known about the
extent of lncRNAs in regulating gene transcription.
NIH-PA Author Manuscript
Recently, the lncRNA Tmevpg1 (also known as NeST) was shown to positively contribute to
IFN-γ production by CD4+ and CD8+ T cells (Collier et al., 2012; Gomez et al., 2013).
Tmevpg1 is adjacent to the Ifng gene in both mice and humans and is encoded on the DNA
strand opposite to that coding IFN-γ. Tmevpg1 transcription is dependent upon transcription
factors Stat4 and T-bet, which also influence Ifng transcription in CD4+ Th1 T cells.
Significantly, transfection of Tmevpg1 alone cannot increase IFN-γ protein in Th0, Th1 or
Th2 cells (Collier et al., 2012). However, mice transgenic for Tmevpg1 have increased IFNγ and are protected from Salmonella infection arguing, that in vivo, Tmevpg1 can act in
trans to regulate target gene expression (Gomez et al., 2013) similar to other observations of
antisense lncRNA trans directed epigenetic regulation of gene expression (Hawkins &
Morris, 2010; Johnsson et al., 2013; Morris et al., 2008; Yu et al., 2008).
These studies show that IFN-γ expression is essentially unregulated in innate immune
responses resulting in almost immediate production of the cytokine. However, any ensuing
IFN-γ driven adaptive immune response requires a much more complex orchestration of
cellular interactions and signaling events. The recent discovery that the lncRNA Tmevpg1
regulates IFN-γ expression in T cells suggests it could be used as a target to deduce the
Discov Med. Author manuscript; available in PMC 2014 September 01.
Pollard et al.
Page 4
relative importance of innate or adaptive immune system production of IFN--γ in disease
processes including autoimmunity.
NIH-PA Author Manuscript
IFN-γ in autoimmunity
NIH-PA Author Manuscript
IFNs are major effectors in the pathogenesis of autoimmunity (Baccala et al., 2005). In
recent years type I IFN production has been argued to be central to the pathogenesis of
systemic autoimmunity (Baccala et al., 2007; Ronnblom & Eloranta, 2013). This is based on
the observation of increased expression of type I IFN inducible genes in peripheral blood
cells, and is commonly called the type I IFN gene signature (Obermoser & Pascual, 2010;
Ronnblom & Eloranta, 2013). The type I IFN signature is found in patients with systemic
lupus erythematosus (SLE), Sjögren’s syndrome (SS), polymyositis (PM), dermatomyositis
(DM) and systemic sclerosis (SSc), but less frequently in patients with rheumatoid arthritis
(RA) and multiple sclerosis (Ronnblom & Eloranta, 2013). A type I IFN signature, defined
by five genes (IFI44, IFI44L, IFI27, RSAD2 and IFI6) has been shown to correlate with
disease activity and disease-affected tissue in SLE, DM, PM, and SSc (Higgs et al., 2011).
However, due to the significant overlap between the genes induced by IFN types I and II it
has proven difficult to differentiate which IFNs are the major contributors to systemic
autoimmune pathogenesis (Gottenberg et al., 2006; Hall et al., 2012; Nguyen & Peck, 2013;
Wong et al., 2012). For example most members of the type I IFN signature described above
are also induced by IFN-γ (Hall et al., 2012). Moreover it must be appreciated that even
genes preferentially induced by IFN-γ (Hall et al., 2012) can also be induced by other
proinflammatory cytokines (Lubeseder-Martellato et al., 2002). However, as type I IFN is
initiated in the early stages of the innate immune response, and is argued to amplify the
adaptive (auto)immune response, it has been suggested that it plays the dominant role
(Baccala et al., 2007). Nonetheless, given the significant overlap in gene expression driven
by IFN types I and II (de Veer et al., 2001; Hall et al., 2012; Hertzog et al., 2011) it is not
surprising that numerous studies have established that type II IFN (IFN-γ) also plays a
pivotal role in the development and severity of autoimmune diseases (Baccala et al., 2005;
Hu & Ivashkiv, 2009; Meyer, 2009).
NIH-PA Author Manuscript
The mechanism whereby IFN-γ leads to systemic autoimmunity remains unclear, however,
the importance of IFN-γ to T cell differentiation and immunoglobulin class switching in B
cells underlines a substantial contribution to adaptive immune responses in autoimmunity.
Increased IFN-γ expression alone can result in the development of systemic autoimmunity
as documented by the finding that epidermal transgenic expression of IFN-γ leads to antidsDNA and anti-histone autoantibodies and glomerulonephritis (Seery, 2000). However,
location of transgenic IFN-γ expression is important as expression in other sites, such as
pancreatic islets and neuromuscular junction, does not lead to systemic autoimmunity
(Seery, 2000). The ability of IFN-γ to promote B cell IgG class switching to more
pathogenic (IgG2a and IgG3 in mice) autoantibodies and activation of IgG Fc receptors and
complement (Baudino et al., 2006) likely also contributes to disease severity. IFN-γ also
plays a significant role in end organ damage due to infiltration of IFN-γ secreting T cells
resulting in macrophage activation, inflammation and tissue damage (Hu & Ivashkiv, 2009;
Schwarting et al., 1998). The Th1 associated chemokine receptor CXCR3 is increased in
lupus nephritis and may contribute to tissue inflammation and autoantibody production
(Lacotte et al., 2013). Mutation of ROQUIN (Rc3h1) in Roquinsan/san mice, another model
of lupus, leads to reduced decay of IFN-γ mRNA resulting in increased IFN-γ signaling and
accumulation of follicular helper T (Tfh) cells, culminating in increased germinal center B
cells and autoantibodies (Lee et al., 2012). Notably in these mutant mice, IFN-γR-deficiency
prevented the development of lupus, whereas deficiency of ICOS, previously thought to be
the cause autoimmunity in this model because of high levels of expression in Tfh cells, did
not. The above findings provide an attractive mechanism to explain the role of IFN-γ in
Discov Med. Author manuscript; available in PMC 2014 September 01.
Pollard et al.
Page 5
NIH-PA Author Manuscript
adaptive (auto)immunity as it directly links expansion of IFN-γ producing CD4+ T cells
with autoantibody production. However, the relevance of this pathway to human SLE or
other murine lupus models will need to investigated.
Human idiopathic systemic autoimmunity
IFN-γ has been shown to be a prominent participant in human systemic autoimmune
responses (Theofilopoulos et al., 2001) (Table 2).This is most convincingly seen in patients
with SLE. IFN-γ mRNA is increased in peripheral blood mononuclear cells (PBMC)
(Csiszar et al., 2000; Harigai et al., 2008) and correlates with PBMC mRNA expression of
its transcription factor T-bet (Lit et al., 2007). Elevated protein levels of IFN-γ have been
found in serum (Funauchi et al., 1991) and following in vitro stimulation of PBMC (Harigai
et al., 2008). Serum IFN-γ correlates with disease activity (Funauchi et al., 1991) and
increased IFN-γ/IL-4 ratio is found in patients with nephritis (Akahoshi et al., 1999;
Masutani et al., 2001).
NIH-PA Author Manuscript
An established role for IFN-γ in other systemic autoimmune diseases is less convincing.
Increased IFN-γ has been found in the PBMC of primary SS patients with Raynaud’s
phenomenon (Willeke et al., 2009) but not those with SS alone (Funauchi et al., 1991;
Willeke et al., 2009). Indeed, although the IFN signature has been found in SS (Ronnblom
& Eloranta, 2013), demonstration of increased IFN-α/β or IFN-γ in primary SS has proven
difficult (Hjelmervik et al., 2005). However, a more recent study has suggested that the IFN
signature in salivary glands of SS shows a predominant IFN-γ response (Hall et al., 2012).
In the idiopathic inflammatory myopathies the IFN-γ/IL-4 ratio is lower in peripheral blood
CD4+ T cells in DM than PM (Ishii et al., 2008) even though IFN-γ levels correlate with
activation of IFN-induced genes in the skin of DM patients (Wong et al., 2012). This
dichotomy may reflect the pronounced infiltration of activated CD4+ T cells in DM skin
(Caproni et al., 2004). A recent study of cytokine expression in systemic sclerosis found
increased IFN-γ positive CD4+ T cells and increased T cell secretion of IFN-γ in a subgroup
of patients with limited cutaneous SSc (Radstake et al., 2009).
Animal models of idiopathic systemic autoimmunity
NIH-PA Author Manuscript
Studies of murine models of spontaneous SLE strongly support the importance of IFN-γ in
systemic autoimmune disease particularly SLE (Table 3). Increases in IFN-γ are found in
lupus-prone mice (Prud’homme et al., 1995) and correlate with disease progression
(Enghard et al., 2006). NZB/W F1 mice treated with IFN-γ have accelerated disease while
treatment with anti-IFN-γ mAb results in disease remission (Jacob et al., 1987). Injection of
cDNA encoding the receptor of IFN-γ (IFN-γR) (Lawson et al., 2000) or treatment with
soluble IFN-γR (Ozmen et al., 1995) reduces serum levels of IFN-γ and manifestations of
disease in MRL-Faslpr and NZB/W F1 mice respectively. Genetic deficiency of IFN-γR in
NZB/W F1 mice reduces production of auto-antibodies and glomerulonephritis (Haas et al.,
1998) and protects MRL-Faslpr mice from lymphadenopathy, autoantibodies and renal
disease (Hron & Peng, 2004). Absence of IFN-γ also dramatically reduces
lymphadenopathy, anti-dsDNA autoantibodies and glomerulonephritis in MRL-Faslpr mice
(Balomenos et al., 1998).
In contrast, type I IFN in MRL-Faslpr mice is not required for development of autoimmune
disease. In the absence of type I IFN signaling lymphoproliferation, autoantibodies and end
organ disease were more severe (Hron & Peng, 2004), antagonistic anti-IFNαR antibodies
had minimal effect (Baccala et al., 2012), and in another study, type I IFN was required for
sex (female) dependent proteinuria although not for renal infiltrates or glomerular score
(Nickerson et al., 2013). Notably serum IgG and anti-nucleosome autoantibodies were not
Discov Med. Author manuscript; available in PMC 2014 September 01.
Pollard et al.
Page 6
NIH-PA Author Manuscript
dependent upon type I IFN in the latter study. Although these findings may reflect use of
different substrains of MRL-Faslpr mice (Nickerson et al., 2013) they are clearly different
from the profound reduction in disease in the absence of IFN-γ (Balomenos et al., 1998).
In support of the prominence of IFN-γ in pathogenesis of SS (Hall et al., 2012), NOD/ShiLtJ
mice, a model of Sjögren’s syndrome, have increased levels of IFN-γ and lack of IFN-γ or
IFN-γR inhibits disease including reduction of glandular infiltrates, and autoantibodies
including anti-nuclear autoantibodies (ANA) (Cha et al., 2004; Nguyen & Peck, 2013).
IFN-γ in induced systemic autoimmunity
NIH-PA Author Manuscript
IFN-γ has also been found to be required for animal models of induced systemic
autoimmunity (Table 3). Mercury exposure in mice elicits systemic autoimmunity
characterized by T and B cell activation, hypergammaglobulinemia, ANA, and immune
complex deposits, all of which are abrogated by the absence of IFN-γ (Pollard et al., 2010).
The importance of IFN-γ to murine mercury-induced autoimmunity (mHgIA) is supported
by a study of mice deficient in genes regulating IFN-γ expression (Casp1, Nlrp3, Il12a,
Il12b, Stat4) or function (Ifngr1, Irf1), which revealed that absence of either Ifngr1 or Irf1
resulted in a striking reduction of disease, while deficiency of genes promoting IFN-γ
expression had modest to no effect (Pollard et al., 2012). Our preliminary studies (Pollard &
Kono, 2013) suggest that type I IFN is not required for mHgIA. These findings argue that
IFN-γ signaling pathways predominate in mHgIA.
The naturally occurring hydrocarbon oil TMPD (2,6,10,14-tetramethylpentadecane), more
commonly known as pristane, induces a severe form of SLE in mice which includes female
predominance, ANA, nephritis, arthritis and pulmonary vasculitis (Pollard et al., 2010). In
the absence of IFN-γ, pristane treated mice have significant reduction in serum
immunoglobulin, ANA, kidney immune deposits and proteinuria compared to wild type
mice (Richards et al., 2001). Type I IFN was also required for pristane-induced
autoimmunity, however, in type I IFN receptor (Ifnar) deficient mice renal deposits were not
affected and although levels of ANA were less than wild type mice, all were ANA positive.
(Nacionales et al., 2007). Thus, as for lupus-prone MRL-Faslpr mice and mHgIA, deficiency
of IFN-γ appears to have more profound effects on the spectrum of disease expression than
absence of type I IFN signaling. Attempts to induce/exacerbate disease in lupus mice
deficient in type I IFN with IFN-γ and vice versa would help resolve which IFN type is of
greater importance to disease severity. We are unaware of any studies that address this issue.
NIH-PA Author Manuscript
The role of IFN-γ in animal models of induced RA is controversial. IFN-γ is protective in
collagen-induced arthritis in the presence of CFA but not in its absence (Schurgers et al.,
2011). This protective effect of IFN-γ is most often seen in experimental models which use
complete Freund’s adjuvant such as collage-induced arthritis, experimental autoimmune
encephalomyelitis and experimental autoimmune uveitis. It has been proposed that the
inhibition of development of Th17 cells by IFN-γ contributes to this protective effect
(Kelchtermans et al., 2008). In contrast the earlier onset and more severe arthritis produced
following immunization with glucose-6-phosphate isomerase (G6PI) does require IFN-γ
even in the presence of CFA (Schurgers et al., 2011). Thus the multiple pathways that lead
to experimental arthritis can have different requirements for IFN-γ.
Therapeutic targets
Even though a number of therapeutic approaches have been identified to control IFN-γ and
related cytokines (Chugh, 2012; Hayashi, 2010) little has been done to develop therapies
targeting IFN-γ in systemic autoimmune diseases. This may stem from the fact that human
deficiency of IFN-γ is associated with severe infection (Meyer, 2009). Even so several
Discov Med. Author manuscript; available in PMC 2014 September 01.
Pollard et al.
Page 7
NIH-PA Author Manuscript
efforts have and are being made to determine the effectiveness of anti-IFN-γ therapy.
Fontolizumab, a humanized monoclonal antibody against IFN-γ, was well tolerated and
showed some efficacy in patients with Crohn’s Disease (Reinisch et al., 2010) however a
phase II clinical trial investigating its use in rheumatoid arthritis was terminated because the
first phase did not meet the endpoint (ClinicalTrials.gov). Amgen’s product pipeline
includes AMG 811, a human monoclonal antibody that inhibits IFN-γ. It is being evaluated
in safety trials with subjects with discoid lupus and subjects with systemic lupus
erythematosus with and without glomerulonephritis (ClinicalTrials.gov). Such non-specific
targeting of IFN-γ is likely to impact both innate and adaptive immunity. Given the
important role that IFN-γ plays in adaptive autoimmune responses therapy targeted to
cellular components regulating IFN-γ expression in T cells, such as the lncRNA Tmevpg1,
may provide greater therapeutic benefit without adverse effect on innate responses to
infection.
Conclusion
NIH-PA Author Manuscript
Numerous studies highlight the pivotal role that both type I and II IFN play in systemic
autoimmunity in humans and spontaneous and induced animal models. While considerable
focus has been placed on the type I IFN signature it is clear that the extensive overlap among
genes induced by IFNs highlights the difficulty in identifying which IFN type has the
predominant activity in inducing disease. Studies of IFN-γ in SLE provide strong evidence
for its role in disease including the adaptive immune response, inflammation, and end organ
damage mediated by infiltrating IFN-γ+ T cells. This is supported by studies in animal
models of SLE which demonstrate the essential role of IFN-γ in both spontaneous and
induced lupus-like disease. These studies suggest that greater thought needs to be given to
therapeutic regulation of IFN-γ in SLE.
Acknowledgments
This work was supported in part by NIH grants ES014847, ES007511, ES020388 (KMP), AI099783 (KVM).
References
NIH-PA Author Manuscript
Akahoshi M, Nakashima H, Tanaka Y, Kohsaka T, Nagano S, Ohgami E, Arinobu Y, Yamaoka K,
Niiro H, Shinozaki M, Hirakata H, Horiuchi T, Otsuka T, Niho Y. Th1/Th2 balance of peripheral T
helper cells in systemic lupus erythematosus. Arthritis Rheum. 1999; 42(8):1644–1648. [PubMed:
10446863]
Aune TM, Collins PL, Collier SP, Henderson MA, Chang S. Epigenetic Activation and Silencing of
the Gene that Encodes IFN-gamma. Frontiers in immunology. 2013; 4:112. [PubMed: 23720660]
Baccala R, Gonzalez-Quintial R, Schreiber RD, Lawson BR, Kono DH, Theofilopoulos AN. AntiIFN-alpha/beta Receptor Antibody Treatment Ameliorates Disease in Lupus-Predisposed Mice. J
Immunol. 2012; 189(12):5976–5984. [PubMed: 23175700]
Baccala R, Hoebe K, Kono DH, Beutler B, Theofilopoulos AN. TLR-dependent and TLR-independent
pathways of type I interferon induction in systemic autoimmunity. Nat Med. 2007; 13(5):543–551.
[PubMed: 17479100]
Baccala R, Kono DH, Theofilopoulos AN. Interferons as pathogenic effectors in autoimmunity.
Immunol Rev. 2005; 204:9–26. [PubMed: 15790347]
Balomenos D, Rumold R, Theofilopoulos AN. Interferon-gamma is required for lupus-like disease and
lymphoaccumulation in MRL-lpr mice. J Clin Invest. 1998; 101(2):364–371. [PubMed: 9435308]
Baudino L, Azeredo Da Silveira S, Nakata M, Izui S. Molecular and cellular basis for pathogenicity of
autoantibodies: lessons from murine monoclonal autoantibodies. Springer Semin Immunopathol.
2006; 28(2):175–184.
Discov Med. Author manuscript; available in PMC 2014 September 01.
Pollard et al.
Page 8
NIH-PA Author Manuscript
NIH-PA Author Manuscript
NIH-PA Author Manuscript
Beima KM, Miazgowicz MM, Lewis MD, Yan PS, Huang TH, Weinmann AS. T-bet binding to newly
identified target gene promoters is cell type-independent but results in variable context-dependent
functional effects. J Biol Chem. 2006; 281(17):11992–12000. [PubMed: 16473879]
Billiau A, Matthys P. Interferon-gamma: a historical perspective. Cytokine Growth Factor Rev. 2009;
20(2):97–113.
Caproni M, Torchia D, Cardinali C, Volpi W, Del Bianco E, D’agata A, Fabbri P. Infiltrating cells,
related cytokines and chemokine receptors in lesional skin of patients with dermatomyositis. Br J
Dermatol. 2004; 151(4):784–791.
Cha S, Brayer J, Gao J, Brown V, Killedar S, Yasunari U, Peck AB. A dual role for interferon-gamma
in the pathogenesis of Sjogren’s syndrome-like autoimmune exocrinopathy in the nonobese
diabetic mouse. Scand J Immunol. 2004; 60(6):552–565.
Chugh PK. Lupus: novel therapies in clinical development. Eur J Intern Med. 2012; 23(3):212–218.
[PubMed: 22385876]
Collier SP, Collins PL, Williams CL, Boothby MR, Aune TM. Cutting edge: influence of Tmevpg1, a
long intergenic noncoding RNA, on the expression of Ifng by Th1 cells. J Immunol. 2012; 189(5):
2084–2088.
Csiszar A, Nagy G, Gergely P, Pozsonyi T, Pocsik E. Increased interferon-gamma (IFN-gamma),
IL-10 and decreased IL-4 mRNA expression in peripheral blood mononuclear cells (PBMC) from
patients with systemic lupus erythematosus (SLE). Clin Exp Immunol. 2000; 122(3):464–470.
De Veer MJ, Holko M, Frevel M, Walker E, Der S, Paranjape JM, Silverman RH, Williams BR.
Functional classification of interferon-stimulated genes identified using microarrays. J Leukoc
Biol. 2001; 69(6):912–920.
Enghard P, Langnickel D, Riemekasten G. T cell cytokine imbalance towards production of IFNgamma and IL-10 in NZB/W F1 lupus-prone mice is associated with autoantibody levels and
nephritis. Scand J Rheumatol. 2006; 35(3):209–216. [PubMed: 16766368]
Funauchi M, Sugishima H, Minoda M, Horiuchi A. Serum level of interferon-gamma in autoimmune
diseases. Tohoku J Exp Med. 1991; 164(4):259–267.
Gomez JA, Wapinski OL, Yang YW, Bureau JF, Gopinath S, Monack DM, Chang HY, Brahic M,
Kirkegaard K. The NeST long ncRNA controls microbial susceptibility and epigenetic activation
of the interferon-gamma locus. Cell. 2013; 152(4):743–754. [PubMed: 23415224]
Gottenberg JE, Cagnard N, Lucchesi C, Letourneur F, Mistou S, Lazure T, Jacques S, Ba N, Ittah M,
Lepajolec C, Labetoulle M, Ardizzone M, Sibilia J, Fournier C, Chiocchia G, Mariette X.
Activation of IFN pathways and plasmacytoid dendritic cell recruitment in target organs of
primary Sjogren’s syndrome. Proc Natl Acad Sci U S A. 2006; 103(8):2770–2775. [PubMed:
16477017]
Haas C, Ryffel B, Le Hir M. IFN-gamma receptor deletion prevents autoantibody production and
glomerulonephritis in lupus-prone (NZB x NZW)F1 mice. J Immunol. 1998; 160(8):3713–3718.
Hall JC, Casciola-Rosen L, Berger AE, Kapsogeorgou EK, Cheadle C, Tzioufas AG, Baer AN, Rosen
A. Precise probes of type II interferon activity define the origin of interferon signatures in target
tissues in rheumatic diseases. Proc Natl Acad Sci U S A. 2012; 109(43):17609–17614. [PubMed:
23045702]
Harigai M, Kawamoto M, Hara M, Kubota T, Kamatani N, Miyasaka N. Excessive production of IFNgamma in patients with systemic lupus erythematosus and its contribution to induction of B
lymphocyte stimulator/B cell-activating factor/TNF ligand superfamily-13B. J Immunol. 2008;
181(3):2211–2219. [PubMed: 18641361]
Hartig JV, Tomari Y, Forstemann K. piRNAs--the ancient hunters of genome invaders. Genes Dev.
2007; 21(14):1707–1713. [PubMed: 17639076]
Hawkins PG, Morris KV. RNA and transcriptional modulation of gene expression. Cell Cycle. 2008;
7(5):602–607. [PubMed: 18256543]
Hawkins PG, Morris KV. Transcriptional regulation of Oct4 by a long non-coding RNA antisense to
Oct4-pseudogene 5. Transcription. 2010; 1(3):165–175.
Hayashi T. Therapeutic strategies for SLE involving cytokines: mechanism-oriented therapies
especially IFN-gamma targeting gene therapy. J Biomed Biotechnol. 2010; 2010
Discov Med. Author manuscript; available in PMC 2014 September 01.
Pollard et al.
Page 9
NIH-PA Author Manuscript
NIH-PA Author Manuscript
NIH-PA Author Manuscript
Hertzog P, Forster S, Samarajiwa S. Systems biology of interferon responses. J Interferon Cytokine
Res. 2011; 31(1):5–11. [PubMed: 21226606]
Higgs BW, Liu Z, White B, Zhu W, White WI, Morehouse C, Brohawn P, Kiener PA, Richman L,
Fiorentino D, Greenberg SA, Jallal B, Yao Y. Patients with systemic lupus erythematosus,
myositis, rheumatoid arthritis and scleroderma share activation of a common type I interferon
pathway. Ann Rheum Dis. 2011; 70(11):2029–2036.
Hjelmervik TO, Petersen K, Jonassen I, Jonsson R, Bolstad AI. Gene expression profiling of minor
salivary glands clearly distinguishes primary Sjogren’s syndrome patients from healthy control
subjects. Arthritis Rheum. 2005; 52(5):1534–1544. [PubMed: 15880807]
Hron JD, Peng SL. Type I IFN protects against murine lupus. J Immunol. 2004; 173(3):2134–2142.
[PubMed: 15265950]
Hu X, Ivashkiv LB. Cross-regulation of signaling pathways by interferon-gamma: implications for
immune responses and autoimmune diseases. Immunity. 2009; 31(4):539–550. [PubMed:
19833085]
Isaacs A, Lindenmann J. Virus interference. I. The interferon. Proceedings of the Royal Society of
London. Series B, Containing papers of a Biological character. Royal Society. 1957; 147(927):
258–267.
Ishii W, Matsuda M, Shimojima Y, Itoh S, Sumida T, Ikeda S. Flow cytometric analysis of
lymphocyte subpopulations and TH1/TH2 balance in patients with polymyositis and
dermatomyositis. Intern Med. 2008; 47(18):1593–1599. [PubMed: 18797118]
Jacob CO, Van Der Meide PH, Mcdevitt HO. In vivo treatment of (NZB X NZW)F1 lupus-like
nephritis with monoclonal antibody to gamma interferon. J Exp Med. 1987; 166(3):798–803.
Johnsson P, Ackley A, Corcoran M, Vidarsdottir L, Grander D, Morris KV. A novel pseudogene noncoding RNA network regulates PTEN transcription and translation in human cells. Nature
Structural & Molecular Biology. 2013 In Press.
Kelchtermans H, Billiau A, Matthys P. How interferon-gamma keeps autoimmune diseases in check.
Trends Immunol. 2008; 29(10):479–486. [PubMed: 18775671]
Lacotte S, Decossas M, Le Coz C, Brun S, Muller S, Dumortier H. Early differentiated CD138(high)
MHCII+ IgG+ plasma cells express CXCR3 and localize into inflamed kidneys of lupus mice.
PLoS One. 2013; 8(3):e58140. [PubMed: 23520491]
Lawson BR, Prud’homme GJ, Chang Y, Gardner HA, Kuan J, Kono DH, Theofilopoulos AN.
Treatment of murine lupus with cDNA encoding IFN-gammaR/Fc. J Clin Invest. 2000; 106(2):
207–215.
Lee SK, Silva DG, Martin JL, Pratama A, Hu X, Chang PP, Walters G, Vinuesa CG. Interferongamma excess leads to pathogenic accumulation of follicular helper T cells and germinal centers.
Immunity. 2012; 37(5):880–892. [PubMed: 23159227]
Lit LC, Wong CK, Li EK, Tam LS, Lam CW, Lo YM. Elevated gene expression of Th1/Th2
associated transcription factors is correlated with disease activity in patients with systemic lupus
erythematosus. J Rheumatol. 2007; 34(1):89–96. [PubMed: 17117487]
Lubeseder-Martellato C, Guenzi E, Jorg A, Topolt K, Naschberger E, Kremmer E, Zietz C, Tschachler
E, Hutzler P, Schwemmle M, Matzen K, Grimm T, Ensoli B, Sturzl M. Guanylate-binding
protein-1 expression is selectively induced by inflammatory cytokines and is an activation marker
of endothelial cells during inflammatory diseases. Am J Pathol. 2002; 161(5):1749–1759.
[PubMed: 12414522]
Masutani K, Akahoshi M, Tsuruya K, Tokumoto M, Ninomiya T, Kohsaka T, Fukuda K, Kanai H,
Nakashima H, Otsuka T, Hirakata H. Predominance of Th1 immune response in diffuse
proliferative lupus nephritis. Arthritis Rheum. 2001; 44(9):2097–2106. [PubMed: 11592372]
Meyer O. Interferons and autoimmune disorders. Joint Bone Spine. 2009; 76(5):464–473. [PubMed:
19773191]
Morris KV, Santoso S, Turner AM, Pastori C, Hawkins PG. Bidirectional transcription directs both
transcriptional gene activation and suppression in human cells. PLoS Genet. 2008;
4(11):e1000258. [PubMed: 19008947]
Murphy KM, Stockinger B. Effector T cell plasticity: flexibility in the face of changing circumstances.
Nat Immunol. 2010; 11(8):674–680. [PubMed: 20644573]
Discov Med. Author manuscript; available in PMC 2014 September 01.
Pollard et al.
Page 10
NIH-PA Author Manuscript
NIH-PA Author Manuscript
NIH-PA Author Manuscript
Nacionales DC, Kelly-Scumpia KM, Lee PY, Weinstein JS, Lyons R, Sobel E, Satoh M, Reeves WH.
Deficiency of the type I interferon receptor protects mice from experimental lupus. Arthritis
Rheum. 2007; 56(11):3770–3783. [PubMed: 17968932]
Nguyen CQ, Peck AB. The Interferon-Signature of Sjogren’s Syndrome: How Unique Biomarkers Can
Identify Underlying Inflammatory and Immunopathological Mechanisms of Specific Diseases.
Frontiers in immunology. 2013; 4:142.
Nickerson KM, Cullen JL, Kashgarian M, Shlomchik MJ. Exacerbated autoimmunity in the absence of
TLR9 in MRL.Fas(lpr) mice depends on Ifnar1. J Immunol. 2013; 190(8):3889–3894.
O’shea JJ, Plenge R. JAK and STAT signaling molecules in immunoregulation and immune-mediated
disease. Immunity. 2012; 36(4):542–550.
Obermoser G, Pascual V. The interferon-alpha signature of systemic lupus erythematosus. Lupus.
2010; 19(9):1012–1019. [PubMed: 20693194]
Ozmen L, Roman D, Fountoulakis M, Schmid G, Ryffel B, Garotta G. Experimental therapy of
systemic lupus erythematosus: the treatment of NZB/W mice with mouse soluble interferongamma receptor inhibits the onset of glomerulonephritis. Eur J Immunol. 1995; 25(1):6–12.
Paunovic V, Carroll HP, Vandenbroeck K, Gadina M. Signalling, inflammation and arthritis: crossed
signals: the role of interleukin (IL)-12, -17, -23 and -27 in autoimmunity. Rheumatology (Oxford).
2008; 47(6):771–776. [PubMed: 18238793]
Pollard KM, Hultman P, Kono DH. Toxicology of autoimmune diseases. Chem Res Toxicol. 2010;
23(3):455–466.
Pollard KM, Hultman P, Toomey CB, Cauvi DM, Hoffman HM, Hamel JC, Kono DH. Definition of
IFN-gamma-related pathways critical for chemically-induced systemic autoimmunity. J
Autoimmun. 2012; 39(4):323–331. [PubMed: 22578563]
Pollard KM, Kono DH. Requirements for innate immune pathways in environmentally induced
autoimmunity. BMC medicine. 2013; 11:100. [PubMed: 23557436]
Prud’homme GJ, Kono DH, Theofilopoulos AN. Quantitative polymerase chain reaction analysis
reveals marked overexpression of interleukin-1 beta, interleukin-1 and interferon-gamma mRNA
in the lymph nodes of lupus-prone mice. Mol Immunol. 1995; 32(7):495–503. [PubMed: 7783752]
Radstake TR, Van Bon L, Broen J, Hussiani A, Hesselstrand R, Wuttge DM, Deng Y, Simms R,
Lubberts E, Lafyatis R. The pronounced Th17 profile in systemic sclerosis (SSc) together with
intracellular expression of TGFbeta and IFNgamma distinguishes SSc phenotypes. PLoS One.
2009; 4(6):e5903.
Reinisch W, De Villiers W, Bene L, Simon L, Racz I, Katz S, Altorjay I, Feagan B, Riff D, Bernstein
CN, Hommes D, Rutgeerts P, Cortot A, Gaspari M, Cheng M, Pearce T, Sands BE. Fontolizumab
in moderate to severe Crohn’s disease: a phase 2, randomized, double-blind, placebo-controlled,
multiple-dose study. Inflamm Bowel Dis. 2010; 16(2):233–242. [PubMed: 19637334]
Richards HB, Satoh M, Jennette JC, Croker BP, Yoshida H, Reeves WH. Interferon-gamma is required
for lupus nephritis in mice treated with the hydrocarbon oil pristane. Kidney Int. 2001; 60(6):
2173–2180. [PubMed: 11737591]
Ronnblom L, Eloranta ML. The interferon signature in autoimmune diseases. Curr Opin Rheumatol.
2013; 25(2):248–253. [PubMed: 23249830]
Schoenborn JR, Wilson CB. Regulation of interferon-gamma during innate and adaptive immune
responses. Adv Immunol. 2007; 96:41–101. [PubMed: 17981204]
Schurgers E, Billiau A, Matthys P. Collagen-induced arthritis as an animal model for rheumatoid
arthritis: focus on interferon-gamma. J Interferon Cytokine Res. 2011; 31(12):917–926. [PubMed:
21905879]
Schwarting A, Wada T, Kinoshita K, Tesch G, Kelley VR. IFN-gamma receptor signaling is essential
for the initiation, acceleration, and destruction of autoimmune kidney disease in MRL-Fas(lpr)
mice. J Immunol. 1998; 161(1):494–503. [PubMed: 9647261]
Seery JP. IFN-gamma transgenic mice: clues to the pathogenesis of systemic lupus erythematosus?
Arthritis Res. 2000; 2(6):437–440. [PubMed: 11094455]
Stark GR, Kerr IM, Williams BR, Silverman RH, Schreiber RD. How cells respond to interferons.
Annu Rev Biochem. 1998; 67:227–264.
Discov Med. Author manuscript; available in PMC 2014 September 01.
Pollard et al.
Page 11
NIH-PA Author Manuscript
Szabo SJ, Sullivan BM, Peng SL, Glimcher LH. Molecular Mechanisms Regulating Th1 Immune
Responses. Annu Rev Immunol. 2003; 21:713–758. [PubMed: 12500979]
Theofilopoulos AN, Koundouris S, Kono DH, Lawson BR. The role of IFN-gamma in systemic lupus
erythematosus: a challenge to the Th1/Th2 paradigm in autoimmunity. Arthritis Res. 2001; 3(3):
136–141. [PubMed: 11299053]
Willeke P, Schluter B, Schotte H, Domschke W, Gaubitz M, Becker H. Interferon-gamma is increased
in patients with primary Sjogren’s syndrome and Raynaud’s phenomenon. Semin Arthritis Rheum.
2009; 39(3):197–202.
Wong D, Kea B, Pesich R, Higgs BW, Zhu W, Brown P, Yao Y, Fiorentino D. Interferon and biologic
signatures in dermatomyositis skin: specificity and heterogeneity across diseases. PLoS One.
2012; 7(1):e29161. [PubMed: 22235269]
Yu W, Gius D, Onyango P, Muldoon-Jacobs K, Karp J, Feinberg AP, Cui H. Epigenetic silencing of
tumour suppressor gene p15 by its antisense RNA. Nature. 2008; 451(7175):202–206. [PubMed:
18185590]
Zhu J, Yamane H, Paul WE. Differentiation of effector CD4 T cell populations (*). Annu Rev
Immunol. 2010; 28:445–489. [PubMed: 20192806]
NIH-PA Author Manuscript
NIH-PA Author Manuscript
Discov Med. Author manuscript; available in PMC 2014 September 01.
NIH-PA Author Manuscript
III
1(H), 4(M)
21(H), 16(M)
IL10R2
21(H), 16(M)
IFNLR1
6(H), 10(M)
IFNGR2
21(H), 16(M)
21(H), 16(M)
Gene Loci Chr. (Species)
IFNGR1
IFNAR2
IFNAR1
Receptor
pDCs
CD4 and CD8 T cells, NK cells, NKT cells, Macrophages, DCs, B cells
Fibroblasts, pDCs
pDCs**
Cellular Source
Abbreviations: DCs dendritic cells, pDCs plasmacytoid dendritic cells, NK natural killer, NKT natural killer T, H human, M mouse
Most cells can secrete IFN-α, but pDCs have the greatest capacity
**
IFN-ε (1 gene) and IFN-γ (1 gene present in humans, but not mice) are not included.
*
12(H), 10(M)
γ (1)
19(H), 7(M)
9(H), 4(M)
β (1)
λ (3)
cluster on
α (13)
I*
II
Gene Loci Chr.(Species)
Class (no. of genes)
IFN Type
NIH-PA Author Manuscript
The three major types of interferon (IFN)
NIH-PA Author Manuscript
Table 1
Pollard et al.
Page 12
Discov Med. Author manuscript; available in PMC 2014 September 01.
Pollard et al.
Page 13
Table 2
IFN-γ in human systemic autoimmune diseases
NIH-PA Author Manuscript
Disease
IFN-γ Phenotype
Reference
SLE
Increased IFN-γ mRNA in PBMC.
Csiszár et al, 2000
Increased IFN-γ in serum of patients with active disease.
Funauchi et al, 1991
Increased IFN-γ RNA and protein after anti-CD3/CD28 stimulation of PBMC.
Harigai et al, 2008
Increased IFN-γ mRNA in PBMC correlated with T-bet mRNA. IFN-γ/IL-4 ratio correlated with disease
activity.
Lit et al, 2007
Increased IFN-γ/IL-4 ratio by intracellular flow cytometry in PBMC of patients with diffuse proliferative
glomerulonephritis.
Akahoshi et al, 1999
Increased IFN-γ/IL-4 ratio in PBMC and increased IFN-γ+ cells in renal biopsy of patients with diffuse
proliferative glomerulonephritis .
Masutani et al, 2001
Increased IFN-γ secretion of PBMC from SS patients with Raynaud’s phenomenon stimulated with T cell
mitogen phytohemagglutinin.
Willeke et al, 2009
SS
Prominent IFN-γ transcriptional response in SS salivary glands.
Hall et al, 2012
DM
Increased IFN-γ mRNA in skin of DM patients correlates with IFN transcriptional response in DM skin
Wong et al, 2012
SSc
Increased IFN-γ by intracellular flow cytometry in T cells of patients with limited SSc. Increased in vitro
IFN-γ secretion by T cells of patients with limited SSc.
Radstake et al, 2009
NIH-PA Author Manuscript
NIH-PA Author Manuscript
Discov Med. Author manuscript; available in PMC 2014 September 01.
Pollard et al.
Page 14
Table 3
IFN-γ in animal models of systemic autoimmunity
NIH-PA Author Manuscript
Model (Strain)
Disease
IFN-γ Manipulation
Disease Severity
Reference
CBAxC57/BL10 F1
SLE
IFN-γ transgene
Increased
Seery, 2000
NZBW F1
SLE
IFN-γ treatment
Increased
Jacob et al, 1987
Anti-IFN-γ treatment
Reduced
Jacob et al, 1987
Soluble IFN-γR
Reduced
Ozmen et al, 1995
IFN-γR null
Reduced
Haas et al, 1998
IFN-γR cDNA
Reduced
Lawson et al, 2000
IFN-γR null
Reduced
Hron & Peng, 2004
IFN-γR null
Reduced
Schwarting et al, 1998
IFN-γ null
Reduced
Balomenos et al, 1998
IFN-γR null
Reduced
Nguyen & Peck, 2013
IFN-γ null
Reduced
Nguyen & Peck, 2013
IFN-γR null
Reduced
Pollard et al, 2012
IFN-γ null
Reduced
Pollard et al, 2010
Spontaneous
MRL-Faslpr
NOD/ShiLtJ
SLE
SS
Induced
NIH-PA Author Manuscript
Mercury (B10.S)
SLE
Pristane (BALB/cJ)
SLE
IFN-γ null
Reduced
Richards et al, 2001
Collagen (CFA)(DBA/1)
RA
IFN-γR null
Increased
Schurgers et al, 2011
Collagen (IFA) (DBA/1)
RA
IFN-γR null
Reduced
Schurgers et al, 2011
G6PI (CFA) (DBA/1)
RA
IFN-γR null
Reduced
Schurgers et al, 2011
NIH-PA Author Manuscript
Discov Med. Author manuscript; available in PMC 2014 September 01.
Download