throbber
Available online at www.sciencedirect.com
`
`Cytokine & Growth Factor Reviews 19 (2008) 41–52
`
`www.elsevier.com/locate/cytogfr
`
`Dendritic cells and cytokines in human inflammatory and
`autoimmune diseases
`
`Patrick Blanco a,b,c,*, A. Karolina Palucka d, Virginia Pascual d, Jacques Banchereau d
`a CHU Bordeaux, 33076 Bordeaux, France
`b CNRS-UMR 5164, 33076 Bordeaux, France
`c Universite´ Bordeaux 2, 33076 Bordeaux, France
`d Baylor Institute for Immunology Research and Baylor Research Institute, Dallas, TX, USA
`
`Abstract
`
`Dendritic cells (DCs) produce cytokines and are susceptible to cytokine-mediated activation. Thus, interaction of resting immature DCs
`with TLR ligands, for example nucleic acids, or with microbes leads to a cascade of pro-inflammatory cytokines and skewing of T cell
`responses. Conversely, several cytokines are able to trigger DC activation (maturation) via autocrine, for example TNF and plasmacytoid DCs,
`and paracrine, for example type I IFN and myeloid DCs, pathways. By controlling DC activation, cytokines regulate immune homeostasis and
`the balance between tolerance and immunity. The increased production and/or bioavailability of cytokines and associated alterations in DC
`homeostasis have been implicated in various human inflammatory and autoimmune diseases. Targeting these cytokines with biological agents
`as already is the case with TNF and IL-1 represents a success of immunology and the coming years will expand the range of cytokines as
`therapeutic targets in autoinflammatory and autoimmune pathology.
`# 2007 Elsevier Ltd. All rights reserved.
`
`Keywords: Dendritic cells; IL-1; IL-12; IL-23; TNF-a; IFN-a
`
`1. Introduction
`
`The immune system is composed of a non-antigen-specific
`innate limb and an antigen-specific adaptive limb [1]. Innate
`immunity, borne by cells such as granulocytes and macro-
`phages and proteins such as complement and cytokines,
`includes a variety of prompt reactions in response to infectious
`agents and other challenges. An excessive response results in
`inflammatory processes. The adaptive immunity, borne by
`lymphocytes,
`is acquired in weeks or months.
`It
`is
`characterized by an exquisite specificity for the eliciting
`antigen as well as memory, which allows a faster and stronger
`response upon re-exposure to the antigen. Adaptive responses
`can be immunogenic, leading to resistance to infections and
`possibly cancer, or tolerogenic avoiding response against self.
`
`* Corresponding author. Tel.: +33 5 57 57 14 70; fax: +33 5 57 57 14 72.
`E-mail addresses: patrick.blanco@chu-bordeaux.fr (P. Blanco),
`Virginip@Baylorhealth.edu (V. Pascual).
`
`1359-6101/$ – see front matter # 2007 Elsevier Ltd. All rights reserved.
`doi:10.1016/j.cytogfr.2007.10.004
`
`Indeed, to efficiently protect us from invading microorgan-
`isms, the adaptive immune system must distinguish self from
`non-self as immune responses against self can create a wide
`repertoire of autoimmune diseases. Anti-self
`immune
`responses are prevented through a variety of mechanisms
`occurring at various levels during the development of the
`immune system [2]. Autoreactive lymphocytes can be
`deleted, rendered anergic or rendered suppressive [3–5].
`Suppressor T cells, also called regulatory T cells, suppress
`autoreactive responses both in an antigen-specific and a non-
`antigen-specific fashion. These immunological events happen
`either in the primary lymphoid organs (bone marrow and
`thymus) and are thus collectively called ‘‘central tolerance’’
`or in the periphery and are then called ‘‘peripheral tolerance’’.
`Clinical autoimmunity arises as a result of an altered balance
`between the autoreactive cells and the regulatory mechanisms
`designed to counterbalance them.
`DCs are specialized to capture and process antigens to
`present their peptides to lymphocytes. They are found in all
`
`Genome Ex. 1047
`Page 1 of 12
`
`

`

`42
`
`P. Blanco et al. / Cytokine & Growth Factor Reviews 19 (2008) 41–52
`
`tissues including blood and lymphoid organs [6–12]. In
`peripheral tissues, DCs are found in an immature stage
`specialized in the capture of antigens. In response to
`microbes, DCs undergo a complex process of maturation
`into antigen-presenting cells. This happens while the DCs
`migrate from the periphery into the draining lymph node
`through the lymphatics. In the steady state, DCs also migrate
`at a low rate without undergoing activation. Then they
`present self-antigens to lymphocytes in the absence of
`costimulation thereby leading to peripheral
`tolerance.
`Various mouse models have demonstrated that DCs bearing
`self-antigens are able to induce autoimmune diseases [13–
`15]. Furthermore alterations of DC homeostasis have been
`directly implicated in various human autoimmune diseases
`including type I diabetes, multiple sclerosis, and systemic
`lupus erythematosus (SLE) [16,17].
`Here we review our current understanding of dendritic
`cell function in tolerance and how cytokines interfere with
`these processes to generate autoimmunity.
`
`2. Dendritic cells
`
`2.1. Dendritic cell maturation
`
`Dendritic cells (DCs) are a heterogeneous family of cells
`of haematopoietic origin that are specialized in the handling
`of antigens, i.e. those from infectious agents and self, and
`their presentation to lymphocytes. Though most of the
`current knowledge relates to the presentation of peptides to
`T cells in the context of MHC classes I and II molecules,
`DCs can present glycolipids and glycopeptides to T cells and
`NKT cells as well as polypeptides to B cells. DCs undergo a
`complex maturation process from antigen-capturing cells
`into antigen-presenting cells. Numerous agents activate DCs
`including: microbes, dying cells, cells of the innate immune
`system and cells of the adaptive immune system. pathogen-
`associated molecular patterns (PAMPs) from microbes [18]
`signal DCs and other cell types through a variety of pattern-
`recognition receptors (PRR) including toll-like receptors
`(TLRs) [19,20]; cell surface C-type lectins receptors (CLRs)
`[21,22] and intracytoplasmic NOD-like receptors (NLRs)
`[23,24]. TLRs have been given the most attention until now
`and appear to be particularly important in the context of
`autoimmunity and most specifically SLE. Distinct DC
`subsets display different TLRs as will be discussed
`hereunder. Lysates of dying cells induce the maturation of
`DCs [25], and some components involved in dying cells
`enhance antigen presentation by DCs leading to T cell
`immunity [25,26]. These endogenous activating molecules
`are collectively called damage-associated molecular pattern
`molecules (DAMPs) [27]. They include heat shock proteins
`(HSPs) [28], high mobility group box 1 protein (HMGB1)
`[29], b-defensin [30] and uric acid [31].
`DCs can secrete a diversified panel of chemokines that
`attract different cell types at different times of the immune
`
`response [32]. They also express a unique set of
`costimulatory molecules which permit the activation of
`naı¨ve T cells and thus allow the launching of primary
`immune response. Through the cytokines they secrete, e.g.:
`IL-12, IL-23 or IL-10 as well as the surface molecules they
`express, e.g.: OX40-L [33] or ICOS-l [34] DCs can polarize
`naı¨ve T cells into Th1, Th2, Treg or Th17.
`
`2.2. Dendritic cell subsets
`
`There are two main pathways of DC ontogeny from
`hematopoietic progenitor cells
`(HPCs). One pathway
`generates myeloid DCs (mDCs); while another generates
`plasmacytoid DCs (pDCs), a subset capable of secreting
`large amounts of type I IFN in response to viral stimulation
`[35,36] (Fig. 1). At least six DC subsets have been described
`in mouse spleen and lymph nodes, including conventional
`DCs (formerly designated myeloid DCs and lymphoid DCs)
`and plasmacytoid DCs [11,37]. They are distinguished
`according to surface markers such as CD11b, CD8a and
`CD11c, as well as by their functions [38–40]. Myeloid DCs
`are found in three compartments: (1) peripheral tissue, (2)
`secondary lymphoid organ and (3) blood. In the skin, two
`distinct types of mDCs are found in two distinct layers.
`Langerhans cells (LCs), which express CD1a and Langerin
`reside in the epidermis, while interstitial DCs (intDCs),
`which express DC-SIGN and CD14 reside in the dermis
`[41]. Plasmacytoid DCs, circulating in the blood and
`secondary lymphoid organs by crossing high endothelial
`venules, express BDCA2, ILT-7 and CD123, and secrete
`large amounts of type I interferons (IFNs) in response to
`viruses and/or TLR7–9 ligands [9]. Blood plasmacytoid DCs
`express TLR1, 6, 7, 9 and 10, but nor TLR4, while blood
`myeloid DCs express TLR1, 2, 3, 4, 5, 6, 7, 8 and 10, but not
`TLR9 [42,43]. Epidermal Langerhans cells isolated from
`skin lack the expression of TLR4 and TLR5, while dermal
`interstitial DCs express many TLRs including TLR2, 4 and 5
`[44]. In the human, CLRs permit to distinguish DC subsets
`with BDCA2 specifically expressed on plasmacytoid DCs
`[45], Langerin expressed on Langerhans cells [46], and DC-
`SIGN expressed on interstitial DCs [47]. Many other C-type
`lectins are more promiscuous, and are, as is the case with
`TLRs, expressed on various cell types including endothelial
`cells and neutrophils. C-type lectins expressed on DCs act as
`anchors for a large number of microbes including viruses,
`bacteria, parasites and fungi, and allow their internalization,
`but they also act as adhesion molecules between DCs and
`other cell types including endothelial cells, T cells and
`neutrophils. Abnormalities in dendritic cell homeostasis
`have been implicated in various human diseases, including
`cancer, autoimmune diseases, allergy and infections.
`
`2.3. Dendritic cells and immune tolerance
`
`Central tolerance, induced in the thymus or bone marrow,
`plays a pivotal role in the prevention of undesired attacks
`
`Genome Ex. 1047
`Page 2 of 12
`
`

`

`P. Blanco et al. / Cytokine & Growth Factor Reviews 19 (2008) 41–52
`
`43
`
`Fig. 1. Dendritic cells are composed of subsets. DC progenitors originate from bone marrow CD34+FLT3+ hematopoietic progenitor cells (HPCs). A myeloid
`pathway generates both Langerhans cells (LCs), found in stratified epithelia such as the skin, and interstitial (int)DCs, found in all other tissues. It also generates
`mDCs circulating in the blood. Upon inflammation monocytes can yield mDCs. Another pathway generates plasmacytoid DCs (pDCs), which secrete large
`amounts of IFN-a/b after viral infection. Activated (mature) mDCs and pDCs traffic to secondary lymphoid organs either via afferent lymphatics (mDCs) or
`blood (pDCs). Langerhans DCs home to cell zones while interstitial DCs home to follicles consistent with their functional specialization, i.e. generation of
`cellular (Langerhans DCs) and humoral (interstitial DCs) immunity, respectively. The origin of resident lymph node DCs remains to be determined.
`
`against self. Anti-self immune responses are prevented
`through a variety of mechanisms occurring at various levels
`of
`the immune system development
`[2]. Autoreactive
`lymphocytes can be either deleted, or rendered anergic or
`rendered suppressive [3–5]. Suppressor T cells also called
`regulatory T cells are also generated which suppress
`autoreactive responses both in an antigen-specific fashion
`and a non-antigen-specific fashion. Many peripheral auto-
`antigens through their expression in thymic medullary
`epithelial cells (a process regulated by the autoimmune
`regulator AIRE) are known to be responsible for the so-called
`negative selection [48]. Moreover, cytokines like thymic
`stromal lymphopoietin (TSLP) produced by epithelial cells of
`thymic Hassall’s corpuscles promote the conversion of
`CD4+CD25-thymocytes into CD4+CD25+Foxp3+ T regula-
`tory cells (Tregs) [49]. Dendritic cells in the thymus are also
`involved in the process of central tolerance [50].
`It is clear, however, that negative selection in the thymus
`does not eliminate all autoreactive cells. Thus, tolerance
`induced in the periphery becomes a very important
`mechanism to maintain control of emerging autoreactivity.
`The mechanisms involved in peripheral tolerance are not
`entirely understood, but there is evidence that ‘‘resting’’
`immature DCs that capture self-antigens in the steady state
`play an important role in this process. Indeed, under these
`conditions, DCs capture apoptotic bodies and/or cellular
`debris arising from normal cell turnover, migrate to draining
`lymph nodes and silence T cells reacting to these antigens
`[51]. The myeloid DC subset appears to be the most potent
`cell able to capture self-apoptotic bodies.
`
`This phenomenon needs to be tightly regulated, as an
`unusual
`load of apoptotic bodies can induce systemic
`autoimmune disease [52]. Indeed, dead cells may also
`contribute to DC maturation, as it has been shown for
`necrotic or late apoptotic cells, but no early apoptotic cells.
`As discussed above, endogenous activating molecules are
`collectively called damage-associated molecular pattern
`molecules [27]. There is evidence that plasmacytoid DCs in
`their resting state are involved in tolerance induction [53–
`55]. pDCs stimulated via CD40 induce IL-10-secreting
`regulatory CD4+ T cells [34] as well as suppressor CD8+ T
`cells [56].
`Recently, the concept that ‘‘immature DCs are tolero-
`genic whereas mature DCs are immunogenic’’ has been
`challenged by several studies showing that fully mature DCs
`can induce tolerance and differentiation of regulatory T cells
`[57–59]. In fact, the integration of different signals by the
`DCs,
`including Ag dose, cytokine milieu at sites of
`inflammation, encountered pathogen etc., will determine
`whether DCs will become tolerogenic vs. immunogenic.
`Possibly, peripheral tolerance is actively maintained by
`‘‘tolerogenic’’ DCs [60]. In addition to deleting T cells,
`tolerogenic DCs induce the differentiation and proliferation
`of T cells with regulatory/suppressor functions [3,61]. Some
`pathogens have a capacity to actively render DCs
`tolerogenic [62]. Although the specific markers of tolero-
`genic DCs are yet to be determined, expression of inhibitory
`immunoglobulin like transcript (ILT) receptors might be
`their feature [63]. In vitro-generated DCs exposed to IL-10
`express ILT-3, which is associated with their tolerogenic
`
`Genome Ex. 1047
`Page 3 of 12
`
`

`

`44
`
`P. Blanco et al. / Cytokine & Growth Factor Reviews 19 (2008) 41–52
`
`functions [64]. Studies in mice suggest that DCs might be
`used in the treatment of autoimmunity through their ability
`to induce regulatory T cells. Thus, repetitive injections of
`‘‘semi-mature’’ DCs induce antigen-specific protection of
`mice from experimental autoimmune encephalomyelitis and
`thyroiditis [57,58]. In NOD mice which spontaneously
`develop diabetes, DCs can induce the generation of Tregs in
`vitro which provide a therapeutic benefit even after onset of
`disease [59]. Indeed, Tregs appear to suppress DCs that
`induce autoimmunity by presenting autoantigens [59,65]. In
`keeping with this, animals which are depleted of Tregs show
`autoimmunity that is associated to expansion of activated
`DCs [66,67]. While immunogenic DCs have been used in
`clinical trials to treat mainly patients with cancer, under-
`standing the mechanisms underlying the tolerogenic
`functions of DCs, such as those generated with IL-10
`[68,69] or those infected with RSV [70], opens new avenues
`for the treatment of autoimmunity or the induction of
`specific tolerance in organ transplants.
`Cytokines secreted to induce a specific immune response
`against an invading pathogen might
`interfere with DC
`homeostasis and induce an autoimmune response that can be
`responsible for
`tissue pathology.
`Indeed, clinical and
`epidemiological studies have suggested a link between
`infectious agents and chronic inflammatory disorders,
`including autoimmune diseases [71].
`
`3. Cytokines, inflammation and autoimmunity
`
`Cytokines represent critical mediators of the autoimmune
`process. They may represent products of DCs and/or induce
`the differentiation of immature DCs into mature DCs that
`can select autoreactive lymphocytes (Fig. 2).
`
`Fig. 2. Cytokines and dendritic cell activation. DCs both produce cytokines
`and are susceptible to cytokine-mediated activation. Thus, exposure to DC
`activators, for example TLR ligands or microbes, triggers secretion of pro-
`inflammatory cytokines including type I interferons (IFN), acute phase
`cytokines such as TNF and IL-6, IL-1 as well as IL-12 family (left panel).
`Several cytokines are able to trigger DCs activation (maturation) either in
`autocrine or paracrine fashion including IL-1, TNF, type I IFNs and TSLP
`(right panel).
`
`3.1. IL-1 and its family
`
`The IL-1 family plays an important role in inflammation
`and host defense. Up to 11 members of this family have been
`identified to date [72,73]. Of those, only five have been
`thoroughly studied: IL-1a, IL-1b, IL-18, IL-1RA and the
`recently reported IL-33. The remaining six (IL-1F5; IL-1F6;
`IL-1F7; IL-1F8; IL-1F9; IL-1F10) have been shown to be
`expressed in various cell types or tissues but their functions
`remain to be determined.
`IL-1a and IL-1b are pro-inflammatory cytokines. Both
`are synthesized as precursor molecules (pro-IL-1a and pro-
`IL-1b) by many different cell
`types. Pro-IL-1a is
`biologically active and needs to be cleaved by calpain to
`generate the smaller mature protein. By contrast, pro-IL-1b
`is biologically inactive and requires enzymatic cleavage by
`caspase-1 in order to become active. IL-1a is primarily
`bound to the membrane whereas IL-1b is secreted and thus
`represents the predominant extracellular
`form of
`IL-1
`(reviewed in Ref. [74,75]). IL-33 is a new member of the
`IL-1 family that is produced as a propeptide requiring
`cleavage by caspase-1. It binds to IL-1R4 (ST2) and
`stimulates T helper 2 (Th2) responses [73]. IL-1 is an
`activator of DCs, though it is not yet clear whether such IL-
`1-activated DCs display unique biological functions [73].
`Interleukin-1 is involved in the pathogenesis of numerous
`diseases with an inflammatory component [76]. This is best
`demonstrated by the therapeutic benefits of treatment of
`patients with IL-1 antagonists such as IL-1-RA. These
`diseases include Systemic onset Juvenile Idiopathic Arthritis
`(SoJIA) [77], which represents up to 20% of chronic
`inflammatory arthritis in childhood. IL-1RA has also shown
`therapeutic efficacy in gout [78], type II diabetes [79] as well
`as a series of hereditary diseases causing periodic
`inflammatory symptoms and grouped under
`the term
`‘‘familial autoinflammatory syndromes’’
`[80]. Whether
`the beneficial effects are due to the inhibition of DC
`activation is not demonstrated.
`
`3.2. IL-6 and its family
`
`The IL-6 family is composed of IL-6, IL-11, leukaemia
`inhibitory factor
`(LIF), oncostatin M (OSM), ciliary
`neurotrophic factor (CNTF), cardiotrophin-1 (CT-1) and
`cardiotrophin-like cytokine (CLC). Members of this family
`display pro- but also anti-inflammatory effects and play a
`central role in hemopoiesis as well as in innate and adaptive
`immune responses.
`Activation of IL-6 signalling is mediated through the IL-
`6/sIL-6R complex, a process known as ‘‘trans-signaliza-
`tion’’ and a unique example of a soluble cytokine receptor
`displaying agonistic effects. IL-6 is secreted by many cell
`types,
`including B and T lymphocytes, monocytes,
`fibroblasts, keratinocytes, endothelial cells, mesenchymal
`cells and certain types of tumor cells. IL-6 induces the
`differentiation of B lymphocytes into plasma cells as well as
`
`Genome Ex. 1047
`Page 4 of 12
`
`

`

`P. Blanco et al. / Cytokine & Growth Factor Reviews 19 (2008) 41–52
`
`45
`
`the proliferation of T lymphocytes, differentiation of
`cytotoxic T cells and IL-2 production. IL-6 also induces
`the differentiation of macrophages and megakaryocytes [81].
`Recently, IL-6 has been described to participate in the
`differentiation of a novel T cell subset, Th17, which displays
`pro-inflammatory functions [82,83]. IL-23 is responsible for
`the expansion of Th17 previously differentiated, while IL-6
`and TGF-b are responsible for the differentiation of Th17
`from their naı¨ve precursors. TGF-b induces Foxp3 which
`leads to the formation of Tregs, while IL-6 inhibits Foxp3
`expression induced by TGF-b, and favors the formation of
`Th17 together with TGF-b [84–86].
`IL-6 is likely to be involved in the pathogenesis of
`inflammatory and autoimmune diseases.
`It plays an
`important role in bone biology by inducing the differentia-
`tion and activation of osteoclasts and it mediates periarti-
`cular destruction of bone and cartilage in experimental
`models of arthritis [87]. IL-6 levels are increased in the
`serum of children with Systemic onset Juvenile Arthritis in a
`disease activity-dependent manner [88], and blocking its
`receptor is emerging as an effective therapy both in SoJIA
`[89] as well as in adult RA [90].
`
`3.3. IL-12 and its family
`
`IL-12, a heterodimeric cytokine produced mainly by
`activated myeloid DCs, plays a pivotal
`role in the
`differentiation and expansion of Th1 cells [91–95]. The
`recent discovery of IL-23 has led to a re-evaluation of
`interleukin-12 biology, as they share a common p40 subunit.
`In animal models, predisposition to autoimmunity can be
`explained by abnormal levels of IL-12 secreted by APCs
`[96]. Furthermore, IL-12 administration has been shown to
`switch tolerance mediated by intravenous or orally
`administered antigens into immunity [97]. In addition
`blocking IL-12 in patients with active Crohn’s disease using
`a specific antibody can induce stable remission [98].
`IL-23 is a cytokine that drives autoimmune diseases,
`including psoriasis and inflammatory bowel diseases
`[99,100]. IL-23 is secreted by human DCs exposed to
`gram-negative bacteria [101]. As mentioned above, IL-23
`promotes the development and expansion of activated CD4+
`T cells that produce IL-17, IL-17F, IL-6 and TNF and are
`called Th17. Their differentiation is inhibited by IFN-
`gamma, IL-4 and IL-2 [102]. Genetic analysis of these Th17
`cells
`identified a unique expression pattern of pro-
`inflammatory cytokines and revealed a unique role in
`different mouse models of autoimmune inflammation [103].
`Given that the levels of IL-23 p19 and IL-17 are elevated in
`human diseases including multiple sclerosis, psoriasis and
`Crohn’s disease, it is possible that these cytokines mediate
`human diseases [104–106]. Indeed, the therapeutic efficacy
`of an IL-12/23 p40 monoclonal antibody in psoriasis has
`recently been established [107]. The Th17 pathway has also
`been implicated in multiple sclerosis (MS) [108]. DCs, i.e.
`monocyte-derived DCs from MS patients, secrete more IL-
`
`23 but equivalent amounts of IL-12 compared to healthy
`controls [104]. Patients with MS also appear to have
`increased numbers of IL-17-expressing cells [109]. Finally,
`a subset of infiltrating T cells express IL-17 in RA synovium
`[110].
`The implication of IL-27 in autoimmunity is less clear as
`this cytokine can have pro- and anti-inflammatory properties
`[111,112].
`
`3.4. TNF-a
`
`TNF-a was among the first cytokines whose dysregula-
`tion was proposed to contribute to the pathogenesis of
`various autoimmune disorders. More importantly, TNF
`blockers have been extensively used and validated as an
`efficacious treatment for RA, Crohn’s disease and psoriasis
`[113,114]. This clearly represents one of the greatest
`successes of immunology though the mechanisms of action
`remain unclear. Inasmuch as TNF induces many cell types,
`including DCs, to secrete pro-inflammatory cytokines, it is
`likely that TNF blocking results in their decreased secretion.
`Alternatively, anti-TNF-a therapy might generate a newly
`differentiated population of Treg cells distinct from natural
`Tregs, which seem to be defective in RA patients [115].
`However, TNF antagonists are not without adverse
`effects, including reactivation of tuberculosis and induction
`of reversible systemic autoimmunity like SLE. In fact TNF
`blockers enhance the production of type I IFNs by pDCs
`exposed to viruses whereas TNF inhibits it. Type I IFNs, as
`described below, have been implicated as
`important
`mediators of autoimmune diseases in humans. Interestingly,
`transcription of type I IFN-inducible genes is observed in
`juvenile arthritis patients treated with TNF blockers. These
`data suggests that TNF represent an endogenous mechanism
`to control IFN production by pDCs [116]. Indeed, TNF
`produced by pDCs in response to viral activation acts as an
`autocrine maturation factor for these cells. Once they
`mature, pDCs are unable to secrete type I IFNs. It is
`therefore conceivable that blocking TNF would keep pDCs
`at an immature stage where they can continue to produce
`type I IFNs. Thus, based on these observations, immunity
`can be viewed as a dynamic system driven by opposite
`vectors, i.e. TNF-type I IFNs. The sum of the vectors yields
`an equilibrium point which allows protective immunity
`when vectors are equal. This dynamic system can
`accommodate the prevalence of either vector to a certain
`extent. However, when one of the vectors prevails beyond a
`certain threshold, the equilibrium point moves into a zone of
`immunopathology, including arthritis when the TNF vector
`prevails and SLE and others when type I IFN production
`prevails (Fig. 3) [16].
`
`3.5. Type I interferons (IFNs)
`
`IFNs (IFN-a/b), major controllers of viral
`Type I
`infections, play a role in several human autoimmune
`
`Genome Ex. 1047
`Page 5 of 12
`
`

`

`46
`
`P. Blanco et al. / Cytokine & Growth Factor Reviews 19 (2008) 41–52
`
`found between the lupus nephritis activity score and the
`number of periglomerular infiltrating CD8+ T lymphocytes.
`No correlation was found, however, with the chronicity
`score. In a similar way, autoreactive CD8+ T lymphocytes
`directed against myelin epitopes are expanded in patients
`with CNS lupus involvement. None of those autoreactive
`cells were found in a control population without neurologic
`involvement or in SLE patients in whom thrombosis was
`responsible for the neurologic symptoms. IFN also appears
`to be associated to other autoimmune diseases including
`myositis [124,125], Sjogren’s syndrome [126,127] and the
`initial phase of psoriasis [128].
`
`3.5.2. IFN-a and B cell activation
`The key role of B lymphocytes in SLE has been known
`for a long time and it has recently been reinforced by the
`observation that
`treatment of patients with the B cell
`depleting CD20 antibody leads to disease improvement
`[129]. Through their direct effect on B cells, type I IFNs
`enhance primary antibody responses to soluble proteins and
`induce the production of all subclasses of IgG in mice [130].
`IFN-a up-regulates CD38, a germinal center B cell and
`plasma cell marker, on B lymphocytes and BAFF (B cell
`activating factor) on monocytes and mDCs. BAFF in turn
`contributes to the survival of autoreactive B lymphocytes
`[131]. In addition, IFN-a promotes the differentiation of
`activated B lymphocytes into plasmablasts. pDCs activated
`with viruses secrete IFN-a and IL-6, which permits
`plasmablasts to become antibody-secreting plasma cells
`[132]. The same effect is observed when pDCs are activated
`with SLE immune complexes containing nucleic acids that
`bind TLRs [133,134]. This could contribute to amplify the
`production of type I IFNs and subsequently the differentia-
`tion of autoreactive plasma cells that would further secrete
`autoantibodies, thus perpetuating this pathogenic loop.
`
`4. Toll-like receptor (TLR) ligands and
`autoimmunity
`
`Infections frequently precede the occurrence of either
`organ-specific or systemic autoimmune diseases. Molecular
`mimicry, however, cannot account for all the autoimmune
`responses that have been linked to infectious diseases. TLRs
`are key components of the innate immune system. These
`receptors activate multiple pathways of inflammation that
`eventually permit to eradicate invading pathogens [135].
`Microbial-derived TLR ligands include a wide range of
`molecules with strong adjuvant activity that can activate
`DCs, macrophages and other APCs [136]. TLRs are
`involved in the pathogenesis of autoimmune disorders
`[14], and endogenous ligands also activate these receptors
`[137–139]. Exposure to TLR3 or TLR7 ligands is required,
`for example, to induce autoimmune diabetes in transgenic
`mice that harbour large numbers of pancreatic islet-reactive
`cytotoxic T cells.
`In this model, TLR-induced local
`
`Fig. 3. Cross-regulation of TNF and IFN-a in autoimmune diseases. TNF
`and IFN-a represent opposite vectors (paths) of immune responses. The sum
`of the vectors yields an equilibrium point, which allows protective immunity
`when vectors are equal. When one of the vectors prevails beyond a certain
`threshold, the equilibrium point moves into a zone of autoimmunity: an
`excess of IFN-a/b is pathogenic in SLE, Sjogren’s, dermatomyositis and
`early stages of psoriasis while excess of TNF is pathogenic in rheumatoid
`arthritis, inflammatory bowel disease (IBD), Crohn’s disease and psoriasis.
`
`diseases, most particularly SLE. Indeed, SLE is the first
`autoimmune disorder where alterations in the type I IFN
`system were reported. In 1979, Notkins and colleagues
`described the presence of IFN activity in the serum of
`patients with SLE [117]. More importantly, induction of
`autoimmunity, including appearance of anti-nuclear anti-
`bodies and occasionally clinical symptoms of SLE, were
`reported during repeated administration of recombinant
`IFN-a to patients with various malignancies or chronic viral
`infection [118]. Recent studies have identified an ‘‘IFN
`signature’’ in the majority of patients with active SLE [119-
`121]. IFN-a in SLE patients is mainly secreted by pDCs and
`understanding what drives its unabated secretion in SLE
`patients remains an area of intense investigation. Type I IFNs
`can contribute to the breaking of tolerance through different
`mechanisms including direct effect on APCs, T cells and B
`cells.
`
`3.5.1. IFN-a and DC alteration
`We have shown that SLE blood constitutes a DC-
`inducing environment, as it promotes the differentiation of
`healthy monocytes into mDCs. The DC-inducing property
`of SLE sera is mainly mediated through IFN-a [122].
`Indeed, blood SLE monocytes display DC-like functions as
`they capture antigens and autoantigens and present them to
`CD4+ and CD8+ T cells. Thus, type I IFN-induced unabated
`DC activation could promote the expansion of autoreactive
`T cells. SLE DCs are characterized by their unique in vitro
`ability to promote the differentiation of CD8+ T lympho-
`cytes in CTLs able to generate nucleosomes and granzyme
`B-dependent autoantigens. Interestingly, terminally differ-
`entiated effector CD8+ T lymphocytes (CCR7, CD45RA+)
`are expanded in the blood of SLE patients and this expansion
`correlates with disease activity as assessed by the SLE
`Disease Activity Index (SLEDAI) [123]. These cells can
`induce direct tissue damage as they represent the main cell
`subset infiltrating the kidney in lupus nephritis, where they
`adopt a periglomerular localization. A direct correlation is
`
`Genome Ex. 1047
`Page 6 of 12
`
`

`

`P. Blanco et al. / Cytokine & Growth Factor Reviews 19 (2008) 41–52
`
`47
`
`production of IFN-a triggers the recruitment of autoreactive T
`cells into the pancreatic islets [140]. TLR3 and TLR9 ligation
`are also key events in the development of autoimmune
`myocarditis by inducing the maturation of DCs pulsed with
`heart-specific self-peptide [14]. In humans, TLR activation
`has been reported as a pathogenic event mainly in the context
`of systemic autoimmune diseases such as SLE. Early studies
`demonstrated that immune complexes were potent stimuli for
`IFN-a secretion by pDCs in an Fc receptor (CD32)-dependent
`manner [141,142]. Indeed, chromatin and/or ribonucleopro-
`tein-containing immune complexes are internalized by pDCs
`via FcgRIIa, reach the endosomal compartment and activate
`IFN-a secretion through TLR9 and/or 7-dependent pathways
`[143,144]. Sera from SLE patients can also induce IFN-a
`secretion in a TLR7/8-dependent manner [133]. Accordingly,
`there is a correlation between the presence of an IFN gene
`signature in blood leukocytes and the detection of auto-
`antibodies directed against ribonucleoproteins in the sera of
`SLE patients [145]. Since INF-a induces the transcription of
`TLR7 itself, a self-amplifying loop could take place at this
`stage as well, thus explaining the above-described correlation.
`The contribution of
`immune complexes and TLR
`signalling to the generation of autoantibodies characteristic
`of SLE has been the purpose of several studies in murine SLE
`models. Chromatin-containing IC activate transgenic auto-
`reactive B cells via sequential engagement of the B cell
`antigen receptor (BCR) and TLR9. In vivo, TLR9 contributes
`to the development of anti-dsDNA antibodies, as lupus-prone
`(Fas-deficient) mice that lack TLR9 on the mixed MRL-B6-
`129 background fail to generate these antibodies [146].
`
`Unexpectedly, in another mouse model (MRL/lpr), TLR9
`deficiency leads to an increased production of autoantibodies
`and a more severe lupus-like disease [147]. Thus, depending
`on the genetic background, TLR9 seems to deliver a
`

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket