throbber
REVIEW
`published: 15 May 2019
`doi: 10.3389/fimmu.2019.01093
`
`Coordination of Immune-Stroma
`Crosstalk by IL-6 Family Cytokines
`
`Nathaniel R. West*
`
`Department of Cancer Immunology, Genentech, South San Francisco, CA, United States
`
`Stromal cells are a subject of rapidly growing immunological interest based on their ability
`to influence virtually all aspects of innate and adaptive immunity. Present in every bodily
`tissue, stromal cells complement the functions of classical
`immune cells by sensing
`pathogens and tissue damage, coordinating leukocyte recruitment and function, and
`promoting immune response resolution and tissue repair. These diverse roles come
`with a price: like classical immune cells, inappropriate stromal cell behavior can lead to
`various forms of pathology, including inflammatory disease, tissue fibrosis, and cancer.
`An important immunological function of stromal cells is to act as information relays,
`responding to leukocyte-derived signals and instructing leukocyte behavior in kind. In
`this regard, several members of the interleukin-6 (IL-6) cytokine family, including IL-6,
`IL-11, oncostatin M (OSM), and leukemia inhibitory factor (LIF), have gained recognition
`as factors that mediate crosstalk between stromal and immune cells, with diverse roles in
`numerous inflammatory and homeostatic processes. This review summarizes our current
`understanding of how IL-6 family cytokines control stromal-immune crosstalk in health
`and disease, and how these interactions can be leveraged for clinical benefit.
`
`Keywords: stromal cells, cytokines, inflammation, fibrosis, immune
`
`THE DIVERSE ROLES OF STROMAL CELLS IN IMMUNITY AND
`INFLAMMATION
`
`The term “stroma” refers to the non-parenchymal components of tissues that form a supportive
`matrix in which parenchymal cells reside (1). While a confusingly broad array of cell types
`have been described as “stromal cells,” in this review they are defined as non-hematopoietic,
`non-epithelial mesenchymal cells, including fibroblasts, myofibroblasts, bone marrow stromal cells,
`and the specialized fibroblast-like stromal cells of secondary lymphoid organs. Other mesenchymal
`populations such as endothelial cells, adipocytes, and muscle cells, while of great interest, are
`largely omitted from this discussion for the sake of brevity and clarity. Long considered to
`be mere structural entities without specialized functions, an explosion of data in the last two
`decades has established stromal cells as key regulators of both protective and pathological immune
`responses (2).
`Regulation of immune function by stromal cells has been most extensively studied in the context
`of secondary lymphoid organs. First identified in 1992, podoplanin (PDPN)+ fibroblastic reticular
`cells (FRC) form a dense reticular network in lymph nodes that facilitates leukocyte migration and
`antigen presentation (2–5). By producing soluble chemokines, cytokines, and other factors—such
`as CCL19 (C-C motif chemokine ligand 19), CCL21, and IL-7 (interleukin 7)—FRC are crucial for
`controlling leukocyte recruitment, survival, and proliferation. FRC-like stromal cells play similar
`roles in other lymphatic tissues, such as in tertiary lymphoid organs of the intestinal mucosa (6, 7).
`
`Edited by:
`
`Teruki Dainichi,
`
`Kyoto University, Japan
`
`Reviewed by:
`
`Carl D. Richards,
`
`McMaster University, Canada
`
`Nathan Karin,
`
`Technion Israel Institute of
`
`Technology, Israel
`
`*Correspondence:
`
`Nathaniel R. West
`
`west.nathaniel@gene.com
`
`Specialty section:
`
`This article was submitted to
`
`Immunological Tolerance and
`
`Regulation,
`
`a section of the journal
`
`Frontiers in Immunology
`
`Received: 28 February 2019
`
`Accepted: 29 April 2019
`
`Published: 15 May 2019
`
`Citation:
`
`West NR (2019) Coordination of
`
`Immune-Stroma Crosstalk by IL-6
`
`Family Cytokines.
`
`Front. Immunol. 10:1093.
`
`doi: 10.3389/fimmu.2019.01093
`
`Frontiers in Immunology | www.frontiersin.org
`
`1
`
`May 2019 | Volume 10 | Article 1093
`
`Lassen - Exhibit 1030, p. 1
`
`

`

`West
`
`IL-6 Family Cytokines and Stromal Cells
`
`In non-lymphoid tissues, stromal cells can exert similar effects
`to those of the secondary lymphoid organs by acting as scaffolds
`for leukocyte migration and by producing a diverse array of
`cytokines and chemokines (2).
`Importantly, the immunological functions of stromal cells
`can vary substantially depending on their host organ and
`physiological context. For example, lymph node FRC recruit
`CCR7 (C-C chemokine receptor type 7)+ T cells (naïve
`and central memory) and CCR7+ dendritic cells (DC) to
`lymph nodes by producing the chemokines CCL19 and
`CCL21, as well as the pro-survival cytokines IL-7 and IL-
`15, thereby coordinating T cell activation and maintenance
`(4). In contrast, stromal cells in peripheral tissues generally
`lack expression of CCL19 and CCL21; accordingly, naïve
`and central memory T cells are infrequent in the periphery.
`However, expression of various pattern recognition and cytokine
`receptors by non-lymphoid tissue stromal cells allows them
`to sense microbial molecules and endogenous danger signals
`(1, 8, 9). In response, they produce chemokines [including
`CCL20 and CXCL10 (C-X-C motif chemokine ligand 10)] that
`attract effector T cells to sites of inflammation. Furthermore,
`inducible expression of leukocyte adhesion molecules including
`ICAM-1 (intercellular adhesion molecule 1) and VCAM-1
`(vascular cell adhesion molecule 1) allows
`tissue-resident
`stromal cells to further influence the balance between leukocyte
`recruitment, retention, and recirculation (1, 2, 9). Finally,
`stromal cells contribute directly to immune response resolution
`and tissue repair, the latter being one of their best studied
`functions. Examples of “pro-resolution” factors produced by
`stromal cells include NOS2 (nitric oxide synthase 2) and NO
`(nitric oxide), which are released by lymph node FRC to
`constrain T cell proliferation (10–12), and IDO1 (indoleamine
`2,3-dioxygenase 1) produced by peripheral
`stromal cells,
`which similarly limits T cell proliferation by depleting the
`critical T cell metabolite tryptophan (13, 14). Thus, stromal
`cells in different
`tissues collectively regulate the strength,
`quality, and duration of immune responses via diverse and
`complementary mechanisms.
`As with most
`immunological processes, communication
`between stromal and immune cells is highly dependent on
`cytokines. Stromal cells bear receptors to a variety of biologically
`diverse cytokines
`that
`represent virtually all branches of
`innate and adaptive immunity, including innate inflammatory
`cytokines [e.g., TNF (tumor necrosis factor) and IL-1β], Th1
`cytokines [e.g., IFN-γ (interferon gamma)], Th2 cytokines
`(e.g., IL-13), Th17 cytokines (e.g., IL-17A), and tolerogenic
`cytokines [e.g., TGF-β (transforming growth factor beta)]
`(7, 9, 15, 16).
`In turn, stromal cells can be prodigious
`producers of other cytokines and chemokines, such as IL-6
`(1, 2, 7, 9). In recent years, cytokines of the IL-6 family
`have gained increasing attention for their roles in various
`homeostatic and pathological processes, which in many cases
`can be attributed to their ability to co-ordinate immune-
`stroma crosstalk. This review aims to provide a focused update
`on the contributions of IL-6 family members to immune-
`stromal interactions.
`
`AN OVERVIEW OF THE IL-6 CYTOKINE
`FAMILY
`
`The IL-6 family includes IL-6, IL-11, IL-27, IL-31, oncostatin
`M (OSM), leukemia inhibitory factor (LIF), ciliary neurotrophic
`factor (CNTF), cardiotrophin 1 (CT-1), and cardiotrophin-
`like cytokine factor 1 (CLCF1) (17, 18). With the exception
`of IL-27, which is a heterodimeric protein comprised of IL-
`27p28 and EBI3 (Epstein-Barr virus-induced gene 3) (19), IL-6
`family members are compact 4-helix bundle cytokines made
`from a single polypeptide. Glycoprotein 130 (gp130, encoded
`by the IL6ST gene) is a crucial receptor subunit utilized by
`all members of the IL-6 family except IL-31. While gp130
`expression is relatively ubiquitous in a wide variety of tissues and
`organs, cell-type specificity for different IL-6 family members is
`bestowed by the more restricted expression patterns of ligand-
`specific co-receptors, including IL-6R (IL-6 receptor), IL-11R
`(IL-11 receptor), IL-27Rα (IL-27 receptor alpha), OSMR (OSM
`receptor), LIFR (LIF receptor), and CNTFRα (CNTF receptor
`alpha). Three distinct forms of receptor-ligand complexes have
`been described (Figure 1). First characterized was that of
`IL-6, which engages IL-6R along with two subunits of gp130.
`Intriguingly, although this implies the formation of a trimeric
`complex, a series of cooperative interactions can ultimately
`produce an interlocked hexamer comprised of two subunits
`each of IL-6, IL-6R, and gp130 (20). A similar structure is
`likely formed in response to IL-11/IL-11R interaction (21, 22).
`In this arrangement, only gp130 drives signal transduction,
`due to an absence of intracellular signaling motifs in IL-6R
`and IL-11R. In contrast, OSMR, LIFR, and IL-27Rα form
`heterodimers with gp130 in the presence of
`their cognate
`ligands (23–28). Unlike IL-6R and IL-11R, OSMR, LIFR, and
`IL-27Rα are capable of driving signal transduction via their
`own suite of signaling motifs. Finally, CNTF and CLCF1
`drive formation of a trimeric complex that includes gp130,
`LIFR, and CNTFRα (29–31). The gp130-independent outlier
`of the family, IL-31, engages a heterodimeric complex of IL-
`31Rα (previously known as gp130-like receptor) and OSMR
`(18). Notably, while mouse OSM binds with high affinity only
`to the gp130/OSMR heterodimer, human and rat OSM can
`bind with high affinity to either gp130/OSMR or gp130/LIFR
`heterodimers (32–34). Thus, in rats and humans, manipulation
`of LIFR would be expected to affect both OSM and LIF signaling
`(as well as CLCF1, CT-1, and CNTF), while manipulation
`of OSMR would influence OSM and IL-31 signaling. As a
`corollary, changes in human or rat OSM bioavailability would
`influence cells that express OSMR and/or LIFR, while changes
`in LIF or IL-31 would affect only LIFR- or IL-31Rα-expressing
`cells, respectively.
`All members of the IL-6 family drive signal transduction via
`receptor-associated Janus kinases (primarily JAK1 and JAK2),
`which phosphorylate a variety of conserved tyrosine residues in
`the cytoplasmic domains of signaling receptor subunits (gp130,
`OSMR, LIFR, IL-27Rα, and IL-31Rα) (17, 18, 35). Several
`downstream signaling pathways are activated in response,
`including signal
`transducer and activator of
`transcription
`
`Frontiers in Immunology | www.frontiersin.org
`
`2
`
`May 2019 | Volume 10 | Article 1093
`
`Lassen - Exhibit 1030, p. 2
`
`

`

`West
`
`IL-6 Family Cytokines and Stromal Cells
`
`FIGURE 1 | Receptor usage of IL-6 family cytokines. With the exception of IL-31, IL-6 family cytokines transduce signals via receptor complexes that include gp130
`and one or more additional ligand-specific subunits. IL-6 and IL-11 signaling requires IL-6R and IL-11R, respectively. The cytoplasmic domains of these receptor are
`short and lack signaling motifs, making gp130 the sole source of signal transduction downstream of IL-6 and IL-11. The heterodimeric cytokine IL-27 (comprised of
`IL-27p28 and EBI3) requires a complex of gp130 and IL-27RA. LIF and CT-1 use a heterodimeric complex of gp130 and LIFR, while CNTF and CLCF1 signal via a
`trimeric complex of gp130, LIFR, and CNTFRα, a GPI-anchored protein that does not directly contribute to signaling beyond facilitation of ligand binding. OSM
`displays species-specific receptor usage. In humans and rats, OSM signals via either gp130/OSMR or gp130/LIFR complexes, while in mice OSM is primarily
`recognized by OSMR. IL-31 does not require gp130, and instead uses a complex of OSMR and IL-31R. Aside from IL-6R, IL-11R, and CNTFRα, all receptors in the
`IL-6 family are capable of directly activating signal transduction in response to ligand binding. IL-6 family cytokines employ classical JAK-mediated signaling. Major
`downstream mediators include STAT3 (the main STAT for all except IL-27), STAT1 (activated preferentially by IL-27 and to a lesser extent by other IL-6 family
`members), additional STATs that depend on cell type and physiological context (including STATs 4, 5, and 6), the MAPK cascade, PI3K/Akt/mTOR signaling, and
`SRC/YAP/NOTCH signaling. Akt, protein kinase B; CLCF1, cardiotrophin-like cytokine factor 1; CNTF, ciliary neurotrophic factor; CT-1, cardiotrophin 1; EBI3,
`Epstein-Barr virus induced 3; ERK, extracellular signal-regulated kinase; gp130, glycoprotein 130, also known as IL-6 signal transducer; IL, interleukin; IL-6R, IL-6
`receptor; IL-11R, IL-11 receptor; IL-27RA, IL-27 receptor; CNTFRα, CNTF receptor; LIF, leukemia inhibitory factor; LIFR, LIF receptor; MAPK, mitogen activated
`protein kinase; JAK, janus kinase; JNK, c-jun n-terminal kinase; mTOR, mammalian target of rapamycin; OSM, oncostatin M; OSMR, OSM receptor; PI3K,
`phosphatidylinositol-3-kinase; STAT, signal transducer and activator of transcription; SRC, Proto-oncogene tyrosine-protein kinase Src; YAP, yes-associated protein.
`
`(STAT) proteins (including STAT1, STAT3, STAT4, STAT5,
`and STAT6), the mitogen-activated protein kinase (MAPK)
`cascade, the phosphatidylinositol-3-kinase (PI3K)/Akt pathway,
`and the SRC/YAP/NOTCH pathway (Figure 1). While signal
`transduction by individual IL-6 family members is broadly
`similar, the relative strength of activation of specific pathways
`can differ depending on the cytokine, cell type, and physiological
`context. For example, unlike gp130, OSMR efficiently recruits
`the adapter protein SHC, allowing OSM to drive more potent
`activation of the MAPK pathway than IL-6, which signals via
`SHP-2 bound to gp130 (35, 36). Similarly, although STAT3
`is generally considered to be the dominant STAT protein
`activated by the IL-6 family, IL-27 preferentially activates
`STAT1 (37). Further complexity is provided by the capacity
`of IL-6, IL-11, and CNTF to signal via soluble receptor forms
`in a process known as trans signaling. In this process, soluble
`versions of IL-6R, IL-11R, or CNTFRα are produced either
`through proteolytic cleavage of membrane-bound receptors,
`
`in either
`or via expression of alternatively spliced mRNA;
`case, the soluble receptor form can dimerize with its cognate
`ligand in solution, and subsequently produce a functional
`signaling complex in association with membrane-bound gp130
`(18, 38–40). Cells thus require only gp130 to be sensitive
`to trans signaling, which allows many cell types that lack
`IL-6R, IL-11R, or CNTFRα to respond to these cytokines.
`In the case of IL-6, trans signaling is thought to be a critical
`mechanism by which IL-6 promotes disease pathogenesis,
`particularly arthritis and colorectal cancer (18, 41, 42). Thus,
`while many similarities exist between IL-6 family cytokines,
`differences
`in their
`receptor usage,
`signal
`transduction
`profiles, and patterns of
`receptor expression collectively
`foster a substantial degree of functional pleiotropy. Indeed,
`IL-6 family members influence cell survival, proliferation,
`differentiation, metabolism, and migration, thus contributing to
`a plethora of physiological processes that are critical for both
`homeostasis and pathology.
`
`Frontiers in Immunology | www.frontiersin.org
`
`3
`
`May 2019 | Volume 10 | Article 1093
`
`Lassen - Exhibit 1030, p. 3
`
`

`

`West
`
`IL-6 Family Cytokines and Stromal Cells
`
`EXPRESSION OF IL-6 FAMILY CYTOKINES
`BY STROMAL CELLS
`
`Although some members of the IL-6 family are produced
`primarily by hematopoietic cells (notably OSM and IL-27),
`stromal cells can be important sources of several others,
`including IL-6, IL-11, and LIF. Diverse factors appear to regulate
`the expression of these cytokines by stromal cells, including
`microbial sensing, detection of endogenous alarmins, stimulation
`by other cytokines (including those within the IL-6 family
`itself), and cell stress (Figure 2). Although these inputs are
`known drivers of cytokine production, the critical drivers in
`vivo, particularly under physiological conditions, are rarely
`well defined.
`In response to infection or an inflammatory challenge, IL-6
`production is rapidly increased by stromal cells. Depending
`on their location and the nature of the challenge, this could
`be due to direct sensing of danger signals, responses to other
`inflammatory cytokines, or both. As an NF-κB (nuclear factor
`kappa B) response gene (43), IL-6 is induced by stromal cells
`downstream of several pattern recognition receptors including,
`but probably not limited to, toll-like receptor (TLR)2, TLR4, and
`NOD2 (nucleotide binding oligomerization domain 2) (44–46).
`
`The NF-κB activating cytokines IL-1β, IL-17A, and TNF (tumor
`necrosis factor alpha) are also potent inducers of stromal IL-6
`production, and can do so in synergy with one another (43, 47–
`54). Although NF-κB is thought to be the dominant driver of
`IL-6 production downstream of these cytokines, contributions
`by MAPK signaling have also been observed. Indeed, signaling
`by alternative pathways such as the MAPK and PI3K cascades
`may underlie the ability of cytokines like OSM (55, 56), IL-4 (49),
`and TGF-β (54, 57) to promote stromal IL-6 expression, since
`these are not classical activators of NF-κB. Beyond cytokines and
`danger signals, cadherin-11 (CDH11), a mesenchymal cadherin
`that engages in homophilic interactions between adjacent cells,
`has also been shown to drive IL-6 production via NF-κB and
`MAPK signaling (53). Indeed, blockade of CDH11 attenuates
`inflammation in mouse models of arthritis, an effect that may
`be due in part to reduced IL-6 production by CDH11+ synovial
`fibroblasts (53). Finally, IL-6 is a well-known product of the
`senescence-associated secretory phenotype (SASP) in fibroblasts,
`a feature associated with aging and cancer (58). Indeed, IL-6
`produced by prostate tumor fibroblasts in response to metabolic
`stress has been proposed to mediate malignant progression (59).
`Less is known about the regulation of LIF and IL-11 expression
`by stromal cells, but the mechanisms involved may be similar
`
`FIGURE 2 | IL-6 family cytokine production by stromal cells and their biological effects. Stromal cells are important contributors to production of three members of the
`IL-6 family: IL-6, LIF, and IL-11. Expression of these cytokines is regulated by various stimuli including recognition of bacterial products via TLR2, TLR4, or NOD2, and
`diverse cytokines that drive activation of NF-κB, MAPK, PI3K, and STAT3. LIF has been shown to promote IL-6 expression via STAT4 signaling, while IL-4 and IL-13
`can suppress LIF and IL-11 expression through activation of STAT6. Following production by stromal cells, IL-6, LIF, and IL-11 can influence diverse biological
`processes including CD4+ T cell polarization, regulation of chemokine production, promotion of alternative macrophage differentiation, and tissue remodeling through
`effects on stromal and epithelial cells. In this figure, arrows indicate stimulatory effects, and capped lines indicate inhibitory effects. All processes illustrated are
`described further in the main text. CCL, C-C motif chemokine ligand; ECM, extracellular matrix; IL, interleukin; LIF, leukemia inhibitor factor; MAPK, mitogen activated
`protein kinase; NF-κB, nuclear factor kappa B; NOD2, nucleotide-binding oligomerization domain-containing protein 2; OSM, oncostatin M; PI3K,
`phosphatidylinositol-3-kinase; STAT, signal transducer and activator of transcription; TFH, T follicular helper cell; TGFβ, transforming growth factor beta; Th, T helper;
`TLR, toll-like receptor; Treg, regulatory T cell.
`
`Frontiers in Immunology | www.frontiersin.org
`
`4
`
`May 2019 | Volume 10 | Article 1093
`
`Lassen - Exhibit 1030, p. 4
`
`

`

`West
`
`IL-6 Family Cytokines and Stromal Cells
`
`to those of IL-6. Like IL-6, LIF and IL-11 expression by stromal
`cells can be induced by IL-1β, TNF, and TGF-β (60–64). Notably,
`induction of both IL-11 and LIF in response to TGF-β stimulation
`of cancer-associated fibroblasts is thought to promote tumor
`progression (61, 62). Intriguingly, IL-4 and IL-13 were shown
`to counteract TNF and IL-1β-induced expression of LIF and
`IL-11, but not IL-6, by gingival fibroblasts (64). This effect was
`dependent on STAT6, and provides a potential mechanism for
`selective modulation of individual IL-6 family members.
`
`RESPONSIVENESS OF STROMAL CELLS
`TO IL-6 FAMILY CYTOKINES
`
`Stromal cells express the necessary receptor subunits to respond
`to the majority of gp130-dependent IL-6 family cytokines. In
`general, gp130 and OSMR are ubiquitously expressed by stromal
`cells from essentially all anatomical locations studied thus far.
`OSM is therefore a major activating factor of stromal cells,
`as well as various other mesenchymal populations including
`endothelial cells, muscle cells, adipocytes, and osteoblasts (34,
`56, 65). Expression of other ligand-specific receptor subunits
`is more variable and depends on the cell type, anatomical
`location, and physiological context. IL-6R, for example, tends
`to be expressed at relatively low levels, and stromal cells are
`correspondingly less sensitive to classical IL-6 signaling than
`OSM. Indeed, expression of OSMR mRNA in human colon
`fibroblasts is roughly 10x higher than that of IL-6R (55).
`However, inflammatory conditions that yield soluble IL-6R can
`increase stromal cell sensitivity to IL-6 due to trans signaling.
`Responsiveness of stromal cells to LIF appears to vary widely
`depending on anatomical location. For example, LIF induces
`contractile and inflammatory phenotypes in dermal and synovial
`fibroblasts, respectively, but has little effect on colon fibroblasts
`(55, 62, 63, 66). Sensitivity of stromal cells to IL-11 and IL-27
`has also been documented (67–73). In contrast, IL-31Rα does
`not seem to be expressed by most stromal cells at physiologically
`relevant levels (74, 75).
`
`CONTROL OF INFLAMMATION AND
`ADAPTIVE IMMUNITY BY THE
`IL-6-STROMA AXIS
`
`Exposure of stromal cells to factors such as microbial ligands or
`inflammatory cytokines can drive IL-6 production during both
`acute and chronic inflammation. Following infection of mice
`by Toxoplasma gondii, for example, IL-6 expression was shown
`to be elevated in a population of bone marrow stromal cells
`characterized by high VCAM-1 and low CD146 expression, and
`stroma-derived IL-6 was required for the increased myelopoiesis
`that occurs as part of the host response to infection (76).
`Bone marrow stromal cells also induce IL-6 in response to
`viral infections such as CMV (cytomegalovirus) (77). During
`Helicobacter hepaticus-driven colitis in mice, non-hematopoietic
`stromal cells are the dominant intestinal producers of IL-6, with
`expression levels that substantially exceed those of MHC-II+
`myeloid cells (55). Interestingly, IL-6 expression may be a feature
`
`of specific intestinal stromal cell subsets with distinct ontogeny
`or activation states. For example, human OSMRhigh intestinal
`stromal cells were found to be enriched in IL-6 expression
`relative to their OSMRlow counterparts (55), consistent with
`the well-established ability of OSM to induce IL-6 expression
`in mesenchymal cells (78–86). Single-cell RNA-sequencing has
`similarly revealed substantial heterogeneity in the intestinal
`stromal cell compartment. High IL-6 expression is enriched
`in a stromal cell subset that is rare in healthy individuals,
`but dramatically expanded in patients with inflammatory bowel
`disease (IBD) (87). Notably, these cells were further characterized
`by expression of a variety of additional immunostimulatory
`molecules, including IL-33 and the FRC-associated chemokines
`CCL19 and CCL21,
`implying a specialized role in immune
`regulation (87). Notably, a disease-associated single nucleotide
`polymorphism (SNP) in the human IL6 promoter was shown
`to control production of IL-6 by fibroblasts, but had no effect
`on IL-6 expression by CD14+ monocytes, suggesting that host
`genetics can also play an important role in determining stromal
`IL-6 output (88).
`Following initiation of acute inflammation, IL-6 can act on
`several cell types to shape the quality of the ensuing immune
`response. For example, IL-6 controls the balance between
`inducible regulatory T cell (Treg) and Th17 differentiation
`following activation of naïve CD4+ T cells (41). Although stromal
`cells have not conclusively been demonstrated to contribute to
`this process, FRC-derived IL-6 has been suggested to support
`the development and maintenance of B cell responses. Medullary
`FRC were shown to be important regulators of plasma cell
`homeostasis, in part by producing the plasma cell survival factor
`IL-6 (89, 90). IL-6 is also necessary for the differentiation of
`follicular helper T cells (TFH), which drive the maturation of B
`cells and the generation of protective antibody responses (91, 92).
`Importantly, IL-6 induces production of IL-21 by TFH cells,
`which is critical for both TFH maintenance and plasma cell
`differentiation in germinal centers (93, 94). Publicly available data
`provided by the ImmGen project suggest that FRC constitutively
`express IL-6, and do so at levels that far exceed those of other
`lymph node-resident cell types (95). Thus, FRC-derived IL-6 is
`likely to be a central linchpin in the regulation of both T cell and
`B cell responses in secondary lymphoid organs.
`In inflamed peripheral tissues, IL-6 controls the temporal
`switch from recruitment of granulocytes
`to preferential
`recruitment of mononuclear cells by modulating chemokine and
`cytokine production in local mesenchymal cells, including the
`suppression of TNF and IL-1β production, possibly via STAT3-
`mediated repression of NF-κB signaling (96, 97). IL-6 promotes
`the differentiation of monocytes into macrophages rather than
`dendritic cells in vitro, but genetic IL-6 deficiency does not
`affect dendritic cell frequencies in vivo (98–101). However,
`IL-6 appears to mediate alternative macrophage differentiation
`in vivo and inhibits inflammatory cytokine production and
`microbicidal activity by macrophages (102–105). IL-6 can also
`promote survival and regeneration of damaged epithelia during
`inflammatory challenges, a feature that can be subverted to
`promote cancer progression (106). Thus, while IL-6 is important
`for initiation of immune responses, it also promotes resolution
`
`Frontiers in Immunology | www.frontiersin.org
`
`5
`
`May 2019 | Volume 10 | Article 1093
`
`Lassen - Exhibit 1030, p. 5
`
`

`

`West
`
`IL-6 Family Cytokines and Stromal Cells
`
`of inflammation and tissue repair (54, 62). Notably, IL-6 protects
`mice from the lethal inflammatory effects of Staphylococcal
`enterotoxin B (SEB; a model of toxic shock), in direct contrast
`with TNF (107).
`Although IL-6 is an important regulator of physiological
`immune responses, excess or chronic IL-6 production can
`promote inflammatory or fibrotic pathology. IL-6 has been
`implicated in a variety of inflammatory diseases, but is perhaps
`best studied in the context of arthritis, a condition that can be
`effectively treated via blockade of IL-6 signaling (42). Synovial
`fibroblasts in inflamed joints are thought to be the major
`source of IL-6, which is likely produced in response to a
`variety of inflammatory factors including TNF, IL-1β, LIF, and
`CDH11 (47, 53, 63). IL-6 is necessary for pathology in pre-
`clinical models of antigen-induced and spontaneous arthritis,
`in which it orchestrates a variety of inflammatory processes
`including activation of CCL2 production by synovial fibroblasts,
`differentiation of autoinflammatory Th17 cells, and bone erosion
`via increased osteoclastogenesis (108–111). IL-6 has also been
`shown to promote CCL20 production by fibroblasts, which may
`further promote recruitment of inflammatory Th17 cells (112).
`IL-6 appears to promote arthritis primarily via trans signaling,
`likely because synovial fibroblasts and activated CD4+ T cells do
`not express sufficient IL-6R to respond to IL-6 alone (108, 109).
`Indeed, CCL2 production following IL-6 stimulation of synovial
`fibroblasts requires the presence of soluble IL-6R, or a chimeric
`IL-6/IL-6R protein known as “hyper-IL-6” (108). IL-6 has also
`been shown to mediate fibrosis in the skin, lung, and heart (113–
`116). Notably, in a phase 2 clinical trial of patients with systemic
`sclerosis, a disease characterized by skin fibrosis, treatment with
`actemra (tocilizumab; anti-IL6R) dramatically attenuated fibrotic
`behavior and transcriptional signatures in dermal fibroblasts,
`along with significant attenuation of disease severity (113).
`
`REGULATION OF INFLAMMATION AND
`HEMATOPOIESIS BY THE OSM-STROMAL
`CELL AXIS
`
`OSM is a pleiotropic cytokine with roles reported in a plethora
`of homeostatic and disease settings (34, 56, 65). Unlike IL-6,
`OSM is not generally produced at significant levels by stromal
`cells, but is instead a product of various hematopoietic cell types,
`including monocytes, macrophages, dendritic cells, neutrophils,
`eosinophils, mast cells, and T cells (34, 56, 65). OSM is further
`distinguished from IL-6 by the cellular distribution of its specific
`receptors (OSMR and LIFR in humans; OSMR in mice), which
`are largely restricted to non-hematopoietic cell types, notably
`epithelial cells, fibroblasts, endothelial cells, adipocytes, and
`neurons (34, 56, 65). OSM thus provides a means for leukocytes
`to deliver information to non-hematopoietic cells in inflamed or
`damaged tissues.
`While little is known about the role of OSM in infectious
`disease or other host-defense settings, OSM can clearly
`influence hematopoietic homeostasis. OSM is necessary
`for the maintenance of granulocyte-macrophage, erythroid,
`megakaryocyte, and multipotential hematopoietic progenitor
`
`populations in the bone marrow, an effect that likely involves
`stimulation of bone marrow stromal cells by OSM (117–120).
`The ability of OSM to drive expression of CXCL12 (SDF1) in
`stromal cells may partly explain its effects on hematopoiesis
`(119, 121–123). However, aging studies have shown that OSM-
`deficient mice develop progressive hematological defects that
`include reduced numbers of circulating leukocytes, erythrocytes,
`and platelets, along with pronounced bone marrow adiposity.
`OSM was shown to suppress adipose differentiation of murine
`PDGFRα+ Sca1+ mesenchymal stem cells, thereby preventing
`the development of “fatty” marrow and safeguarding the
`hematopoietic niche (120). Several additional studies have
`confirmed that OSM acts on stromal progenitors to suppress
`adipocyte differentiation in favor of osteoblast development
`(124–130), suggesting that OSM plays a fundamental role
`in regulating the bone marrow microenvironment. Notably,
`overexpression of OSM in bone marrow stromal cells promotes
`the development of lethal myeloproliferative neoplasms and
`bone marrow fibrosis in mice (131, 132).
`Numerous studies have implicated OSM in the pathogenesis
`of inflammatory conditions, including arthritis, inflammatory
`bowel disease, psoriasis, and allergic airway inflammation.
`Intra-articular adenoviral delivery of OSM causes arthritis-
`like pathology characterized by robust leukocyte infiltration,
`synovial hyperplasia, and erosion of bone and cartilage (133–
`135). Consistent with these findings, antibody blockade of OSM
`can reduce pathology in the collagen-induced and pristane-
`challenge pre-clinical models of inflammatory arthritis (136).
`Synovial fibroblasts respond to OSM by producing a wide
`variety of inflammatory factors including cytokines (e.g., IL-
`6), chemokines, (e.g., CCL2, CCL13, CXCL1), and leukocyte
`adhesion factors such as ICAM-1 (78, 82, 83, 134, 137–139).
`Furthermore, cytokine receptors such as IL1R1 (IL-1 receptor,
`type 1), gp130, and OSMR are induced by OSM, suggesting that
`OSM can sensitize synovial fibroblasts to additional cytokine
`stimulation. OSM can synergize with TNF and IL-1β to promote
`increased cytokine and chemokine expression, as well as high
`MMP (matrix metalloprotease) to TIMP1 (tissue inhibitor of
`metalloproteases) ratios to promote tissue damage. Remarkably,
`OSM alone drives high TIMP1 expression (134, 135, 138, 140,
`141), and OSM only promotes net tissue degradation when
`acting in synergy with TNF, IL-1β, or IL-17A, suggesting that its
`pathogenicity may depend on the presence of other inflammatory
`factors (135, 137, 138, 142).
`Emerging data suggest an important role for OSM-stromal cell
`interactions in barrier tissues, such as skin and intestinal mucosa.
`Dermal fibroblasts express extracellular matrix components such
`as collagens and glycosaminoglycans in response to OSM, and
`display an interferon-like response featuring upregulation of
`the viral RNA sensors RIG-I and MDA5 (143–146). While
`sufficient
`to induce skin inflammation, OSM may not be
`required for psoriasis-like pathology, as it is dispensable in
`the aldara (imiquimod) challenge model of psoriasis (55, 147,
`148). OSM and OSMR are also overexpressed in the lesional
`skin of patients with atopic dermatitis, but whether OSM
`signaling is required for pathogenesis of
`this condition is
`unclear (147). A potentially non-redundant inflammatory role
`
`Frontiers in I

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