throbber
NIH Public Access
`Author Manuscript
`J Pathol. Author manuscript; available in PMC 2009 June 9.
`Published in final edited form as:
`J Pathol. 2008 January ; 214(2): 199–210. doi:10.1002/path.2277.
`Cellular and molecular mechanisms of fibrosis
`TA Wynn*
`Immunopathogenesis Section, Laboratory of Parasitic Diseases, National Institute of Allergy and
`Infectious Diseases, National Institutes of Health, Bethesda, MD, USA
`Abstract
`Fibrosis is defined by the overgrowth, hardening, and/or scarring of various tissues and is attributed
`to excess deposition of extracellular matrix components including collagen. Fibrosis is the end result
`of chronic inflammatory reactions induced by a variety of stimuli including persistent infections,
`autoimmune reactions, allergic responses, chemical insults, radiation, and tissue injury. Although
`current treatments for fibrotic diseases such as idiopathic pulmonary fibrosis, liver cirrhosis, systemic
`sclerosis, progressive kidney disease, and cardiovascular fibrosis typically target the inflammatory
`response, there is accumulating evidence that the mechanisms driving fibrogenesis are distinct from
`those regulating inflammation. In fact, some studies have suggested that ongoing inflammation is
`needed to reverse established and progressive fibrosis. The key cellular mediator of fibrosis is the
`myofibroblast, which when activated serves as the primary collagen-producing cell. Myofibroblasts
`are generated from a variety of sources including resident mesenchymal cells, epithelial and
`endothelial cells in processes termed epithelial/endothelial-mesenchymal (EMT/EndMT) transition,
`as well as from circulating fibroblast-like cells called fibrocytes that are derived from bone-marrow
`stem cells. Myofibroblasts are activated by a variety of mechanisms, including paracrine signals
`derived from lymphocytes and macrophages, autocrine factors secreted by myofibroblasts, and
`pathogen-associated molecular patterns (PAMPS) produced by pathogenic organisms that interact
`with pattern recognition receptors (i.e. TLRs) on fibroblasts. Cytokines (IL-13, IL-21, TGF-(cid:533)1),
`chemokines (MCP-1, MIP-1(cid:533)), angiogenic factors (VEGF), growth factors (PDGF), peroxisome
`proliferator-activated receptors (PPARs), acute phase proteins (SAP), caspases, and components of
`the renin–angiotensin–aldosterone system (ANG II) have been identified as important regulators of
`fibrosis and are being investigated as potential targets of antifibrotic drugs. This review explores our
`current understanding of the cellular and molecular mechanisms of fibrogenesis.
`Keywords
`fibrosis; myofibroblast; collagen; wound healing; liver; lung
`IntroductionIn contrast to acute inflammatory reactions, which are characterized by rapidly resolving
`vascular changes, oedema and neutrophilic inflammation, fibrosis typically results from
`chronic inflammation — defined as an immune response that persists for several months and
`in which inflammation, tissue remodelling and repair processes occur simultaneously. Despite
`having distinct aetiological and clinical manifestations, most chronic fibrotic disorders have
`*Correspondence to: TA Wynn, Immunopathogenesis, Section, Laboratory of Parasitic Diseases, National Institute of Allergy and
`Infectious Diseases, National Institutes of Health, 50 South Drive, Room 6154, MSC 8003, Bethesda, MD, 20892, USA. E-mail: E-mail:
`twynn@niaid.nih.gov.
`No conflicts of interest were declared.
`This article is a US government work and is in the public domain in the USA.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`Lassen - Exhibit 1068, p. 1
`
`

`

`Page 2
`in common a persistent irritant that sustains the production of growth factors, proteolytic
`enzymes, angiogenic factors and fibrogenic cytokines, which stimulate the deposition of
`connective tissue elements that progressively remodel and destroy normal tissue architecture
`[1–3].Damage to tissues can result from various stimuli, including infections, autoimmune reactions,
`toxins, radiation and mechanical injury. The repair process typically involves two distinct
`phases: a regenerative phase, in which injured cells are replaced by cells of the same type,
`leaving no lasting evidence of damage; and a phase known as fibroplasia or fibrosis, in which
`connective tissues replaces normal parenchymal tissue. Although initially beneficial, the repair
`process becomes pathogenic when it is not controlled appropriately, resulting in substantial
`deposition of ECM components in which normal tissue is replaced with permanent scar tissue
`[4]. In some diseases, such as idiopathic pulmonary fibrosis, liver cirrhosis, cardiovascular
`fibrosis, systemic sclerosis and nephritis, extensive tissue remodelling and fibrosis can
`ultimately lead to organ failure and death (Table 1).
`Wound healing versus fibrosis
`When epithelial and/or endothelial cells are damaged, they release inflammatory mediators
`that initiate an anti-fibrinolytic coagulation cascade [5], which triggers blood-clot formation
`and formation of a provisional ECM. Platelets are exposed to ECM components, triggering
`aggregation, clot formation and haemostasis. Platelet degranulation also promotes vasodilation
`and increased blood vessel permeability, while myofibroblasts (activated collagen secreting,
`(cid:302)-SMA+ fibroblasts) and epithelial and/or endothelial cells produce MMPs, which disrupt the
`basement membrane, allowing inflammatory cells to be easily recruited to the site of injury.
`Growth factors, cytokines and chemokines are also produced, which stimulates the
`proliferation and recruitment of leukocytes across the provisional ECM. Some of the early
`responders include macrophages and neutrophils, which eliminate tissue debris, dead cells and
`any invading organisms. They also produce cytokines and chemokines, which are mitogenic
`and chemotactic for endothelial cells, which begin to surround the injured site. They also help
`form new blood vessels as epithelial/endothelial cells migrate towards the centre of the wound.
`During this period, lymphocytes and other cells become activated and begin secreting
`profibrotic cytokines and growth factors, such as TGF(cid:533), IL-13 and PDGF [6–8], which further
`activate the macrophages and fibroblasts. Activated fibroblasts transform into (cid:302)-SMA-
`expressing myofibroblasts as they migrate along the fibrin lattice into the wound. Following
`activation, the myofibroblasts promote wound contraction, the process in which the edges of
`the wound migrate towards the centre. Finally, epithelial and/or endothelial cells divide and
`migrate over the basal layers to regenerate the damaged tissue, which completes the wound-
`healing process. However, chronic inflammation and repair can trigger an excessive
`accumulation of ECM components, which leads to the formation of a permanent fibrotic scar.
`Collagen turnover and ECM remodelling is regulated by various MMPs and their inhibitors,
`which include the tissue inhibitors of metalloproteinases (TIMPs). Shifts in synthesis versus
`catabolism of the ECM regulate the net increase or decrease of collagen within the wound
`[9]. Fibrosis occurs when the synthesis of new collagen by myofibroblasts exceeds the rate at
`which it is degraded, such that the total amount of collagen increases over time.
`The cellular origins of myofibroblasts
`Local tissue myofibroblasts were originally believed to be the primary producers of ECM
`components following injury [5]; however, it is now thought that fibroblasts can be derived
`from multiple sources [10]. In addition to resident mesenchymal cells, myofibroblasts are
`derived from epithelial cells in a process termed epithelial–mesenchymal transition (EMT)
`[10–12]. More recently, it was suggested that a similar process occurs with endothelial cells,
`J Pathol. Author manuscript; available in PMC 2009 June 9.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`Wynn
`
`NIH-PA Author Manuscript
`
`Lassen - Exhibit 1068, p. 2
`
`

`

`Page 3
`termed endothelial–mesenchymal transition (EndMT) [13]. Bucala and colleagues also
`identified a unique circulating fibroblast-like cell derived from bone marrow stem cells [14].
`These blood-borne mesenchymal stem cell progenitors have a fibroblast/myofibroblast-like
`phenotype (they express CD34, CD45 and type I collagen) and are now commonly called
`fibrocytes [15–18]. Finally, in some tissues, resident fibroblasts are not the only source of
`myofibroblasts. For example, in liver fibrosis the resident hepatic stellate cell (HSC) appears
`to be the primary source of myofibroblasts, although bone-marrow-derived cells can also
`contribute [18,19]. Because it is now thought that fibrocytes and EMT-derived myofibroblasts
`participate with resident mesenchymal cells in the reparative process, there has been growing
`interest in dissecting the role of the various myofibroblast subpopulations in fibroproliferative
`disease [20]. Because bone marrow-derived fibrocytes must find their way to sites of tissue
`injury to participate in wound healing and fibrosis, there has been a great deal of interest in
`understanding the role of chemokines and acute phase proteins, such as serum amyloid P (SAP),
`in the development and recruitment of myofibroblasts [20–22]. Because fibrocytes and EMT-
`derived myofibroblasts produce a variety of factors that are involved in the fibrotic process
`[10], interrupting their development, recruitment and/or activation could provide a unique
`therapeutic approach to treat a variety of fibrotic diseases.
`Innate and adaptive immune mechanisms regulate myofibroblast activity
`Many fibrotic disorders are thought to have an infectious aetiology, with bacteria, viruses, fungi
`and multicellular parasites driving chronic inflammation and the development of fibrosis. It
`was recently suggested that conserved pathogen-associated molecular patterns (PAMPs) found
`on these organisms help maintain myofibroblasts at a heightened state of activation [23].
`Bacteria living in the gut can also contribute to the activation of myofibroblasts [24]. PAMPs
`are pathogen byproducts, such as lipoproteins, bacterial DNA and double-stranded RNA,
`which are recognized by pattern recognition receptors (PRRs) found on a wide variety of cells,
`including fibroblasts [25]. The interaction between PAMPs and PRRs serves as a first line of
`defence during infection and activates numerous proinflammatory cytokine and chemokine
`responses. In addition, because fibroblasts express a variety of PRRs, including Toll-like
`receptors (TLRs), Toll ligands can directly activate fibroblasts and promote their differentiation
`into collagen-producing myofibroblasts [23,24,26]. Thus, inhibiting TLR signalling might
`represent a novel approach to treat fibrotic disease.
`Nevertheless, pathogenic organisms are not responsible for all fibrotic disorders. Therefore,
`additional mechanisms must also participate in the activation of myofibroblasts. For example,
`in the case of systemic sclerosis (SSc), fibroblasts obtained from lesional skin or fibrotic lungs
`have a constitutively activated myofibroblast-like phenotype, characterized by enhanced ECM
`synthesis, constitutive secretion of cytokines and chemokines and increased expression of cell
`surface receptors [27–29]. Because most of the characteristics of fibroblasts from patients with
`SSc are reproduced in normal human fibroblasts following stimulation with TGF(cid:533), it is thought
`that the SSc fibroblast phenotype is maintained by an autocrine TGF(cid:533) signal. However,
`TGF(cid:533)/SMAD3-independent mechanisms have also been proposed [30,31], including a role for
`viruses such as CMV, which stimulate the production of auto-antibodies and connective tissue
`growth factor (CTGF), both of which are known to participate in the activation of
`myofibroblasts [28,32]. Epigenetic changes may also contribute to the persistent activation of
`myofibroblasts [33]. B cells have also been implicated, either by producing autoanti-bodies or
`by secreting IL-6, a well-known fibroblast growth factor [34]. Still other studies have argued
`that Th2-type cytokines derived from a variety of cellular sources are critically involved in the
`mechanism of fibrosis [35–38]. Therefore, paracrine signals derived from activated
`lymphocytes, autocrine factors produced by fibroblasts, as well as molecules derived from
`pathogenic organisms can cooperate to initiate and maintain myofibroblast activation.
`J Pathol. Author manuscript; available in PMC 2009 June 9.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`Wynn
`
`NIH-PA Author Manuscript
`
`Lassen - Exhibit 1068, p. 3
`
`

`

`Wynn
`Page 4
`Chemokines regulate fibrogenesis by controlling myofibroblast recruitment
`Chemokines are leukocyte chemoattractants that cooperate with profibrotic cytokines in the
`development of fibrosis by recruiting myofibroblasts, macrophages and other key effector cells
`to sites of tissue injury. Although a large number of chemokine signalling pathways are
`involved in the mechanism of fibrogenesis, the CC- and CXC-chemokine receptor families
`have consistently exhibited important regulatory roles. Specifically, CCL3 (macrophage
`inflammatory protein 1(cid:302)) and CC-chemokines such as CCL2 (monocyte chemoattractant
`protein-1), which are chemotactic for mononuclear phagocytes, were identified as profibrotic
`mediators. Macrophages and epithelial cells are believed to be the key sources of CCL3, and
`studies in the bleomycin model of pulmonary fibrosis showed that anti-CCL3 antibodies could
`significantly reduce the development of fibrosis [39,40]. Similar results were obtained when
`CCL2 was neutralized, suggesting that a variety of CC-chemokines are involved [41,42].
`Subsequent studies with CC-chemokine receptor 1 (CCR1)- and CCR2-deficient mice
`produced similar results, confirming critical roles for CCL3/CCL2-mediated signalling
`pathways in fibrogenesis [43–47]. Interestingly, in several of these blocking studies, the
`absence of fibrosis was associated with decreased IL-4/IL-13 expression [44,48], suggesting
`a direct link between CC-chemokine activity and the production of profibrotic cytokines such
`as IL-13. IL-13 is a potent inducer of several CC-chemokines, including CCL3, CCL4
`(MIP-1(cid:533)), CCL20 (MIP-3(cid:302)), CCL2, CCL11, CCL22 (macrophage-derived chemokine) and
`CCL6 (C10), among others, suggesting that a positive feedback mechanism exists between
`IL-13 and the CC-chemokine family [49,50]. As seen with anti-CCL3 and anti-CCL2 antibody
`treatment, antibodies to CCL6 significantly attenuated lung remodelling responses in IL-13-
`transgenic mice [50] as well as in mice challenged with bleomycin [49], indicating non-
`redundant roles for a variety of CC-chemokines in the pathogenesis of fibrosis. In mice, CXC
`chemokine receptor 4 (CXCR4), CC chemokine receptor 7 (CCR7) and CCR2 have also been
`shown to regulate the recruitment of fibrocytes to the lung [20,21]. Thus, interrupting specific
`chemokine signalling pathways could have a significant impact on the treatment of a variety
`of fibroproliferative diseases.
`Th1 and Th2 cells differentially regulate organ fibrosis
`Chronic inflammatory reactions are typically characterized by a large infiltrate of mononuclear
`cells, including macrophages, lymphocytes, eosinophils and plasma cells. Lymphocytes are
`mobilized to sites of injury and become activated following contact with various antigens,
`which stimulate the production of lymphokines that further activate macrophages and other
`local inflammatory cells. Thus, there is significant activation of the adaptive immune response
`in many chronic inflammatory diseases. Although inflammation typically precedes the
`development of fibrosis, results from a variety of experimental models suggest that fibrosis is
`not always characterized by persistent inflammation, implying that the mechanisms regulating
`fibrosis are to a certain extend distinct from those controlling inflammation. Findings from our
`own studies of schistosomiasis-induced liver fibrosis support this theory [35]. In this model,
`fibrosis develops progressively in response to schistosome eggs that are deposited in the liver,
`which induce a chronic granulomatous response. As in many other experimental models of
`fibrosis, CD4+ T cells play a prominent role in the progression of the disease. Studies conducted
`with multiple cytokine-deficient mice have demonstrated that liver fibrosis is strongly linked
`with the development of a CD4+ Th2 cell response (involving IL-4, IL-5, IL-13 and IL-21)
`[51–55].
`Several experimental models of fibrosis in addition to our own have also documented potent
`antifibrotic activities for the Th1-associated cytokines IFN(cid:534) and IL-12. In schistosomiasis,
`while treatment with IFN(cid:534) or IL-12 has no effect on the establishment of infection, collagen
`deposition associated with chronic granuloma formation is substantially decreased [51].
`J Pathol. Author manuscript; available in PMC 2009 June 9.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`Lassen - Exhibit 1068, p. 4
`
`

`

`Page 5
`Similar results have been obtained in models of pulmonary, liver and kidney fibrosis [56–
`59]. These findings suggest that it might be possible to develop an antifibrosis vaccine based
`on immune deviation [51,60], in which the profibrotic effects of the Th2 response are switched
`off in favour of an antifibrotic Th1 response. Indeed, similar approaches have been proposed
`for individuals suffering from allergic airway inflammation [61], which is also driven by Th2-
`type responses. Studies investigating the gene expression patterns of fibrotic tissues found that
`markedly different gene expression profiles are induced during Th1 and Th2 polarized
`responses [62,63]. As might be expected, a large number of IFN(cid:534)-induced genes are
`upregulated in the tissues of mice exhibiting Th1-polarized responses, with no evidence of
`significant activation of the fibrosis-associated genes in this setting [62–64]. Instead, two major
`groups of genes were identified in Th1-polarized mice: those associated with the acute-phase
`reaction and apoptosis (cell death), findings which may explain the extensive tissue damage
`that is commonly observed when Th1 responses continue unchecked [65]. By contrast, several
`genes known to be involved in the mechanisms of wound healing and fibrosis were upregulated
`in animals exhibiting Th2-polarized inflammation [62,63]. The regulation and function of a
`few of the genes, including procollagens I, III and VI, arginase-1 [66], lysyl oxidase [67,68],
`matrix metalloproteinase-2 (MMP-2) [69,70], MMP-9 [71,72] and tissue inhibitor of matrix
`metalloproteinase-1 (TIMP-1) [73,74], have been investigated in some detail. Several
`additional Th2-linked genes [62,63], including haem oxygenase, procollagen III, secreted
`phosphoprotein 1, procollagen V, reticulocalbin and fibrillin 1 have also been reported in the
`fibrotic lungs of bleomycin-treated mice [75] and in CCl4-stimulated rat hepatic stellate cells
`(collagen-producing cells in the liver) [76], providing further evidence that fibrosis is often
`associated with the development of Th2-type responses.
`Unique roles for the Th2 cytokines IL-4, IL-5, IL-13 and IL-21 in fibrosis
`The Th2 cytokines IL-4, IL-5, IL-13 and IL-21 each have distinct roles in the regulation of
`tissue remodelling and fibrosis. IL-4 is found at increased levels in the bronchoalveolar lavage
`fluids of patients with idiopathic pulmonary fibrosis (IPF) [77], in the pulmonary interstitium
`of individuals with cryptogenic fibrosing alveolitis [78] and in peripheral blood mononuclear
`cells (PBMCs) of those suffering from periportal fibrosis [79]. Development of post-irradiation
`fibrosis is also associated with increased production of IL-4 [80]. Although the extent to which
`IL-4 participates in fibrosis varies in different diseases, it has long been considered a potent
`profibrotic mediator. In fact, studies have suggested that IL-4 is nearly twice as effective as
`TGF(cid:533) [81], another potent profibrotic cytokine that has been extensively studied [82].
`Receptors for IL-4 are found on many mouse [83] and human fibroblast subtypes [84] and in
`vitro studies showed the synthesis of the extracellular matrix proteins, types I and III collagen
`and fibronectin, following IL-4 stimulation. One of the first in vivo reports to investigate the
`contribution of IL-4 was a study of schistosomiasis in mice, in which neutralizing antibodies
`to IL-4 were shown to significantly reduce the development of hepatic fibrosis [52]. Inhibitors
`of IL-4 were also found to reduce dermal fibrosis in a chronic skin graft rejection model and
`in a mouse model of scleroderma [85,86].
`IL-13 shares many functional activities with IL-4 because both cytokines exploit the same
`IL-4R(cid:302)/Stat6 signalling pathways [87]. However, with the development of IL-13 transgenic
`and knockout mice [88,89], as well as IL-13 antagonists [53,90], unique and non-redundant
`roles for IL-13 and IL-4 have been revealed in numerous models. When IL-4 and IL-13 were
`inhibited independently, IL-13 was identified as the dominant effector cytokine of fibrosis in
`several experimental models of fibrosis [38,53,91–94]. In schistosomiasis, although the egg-
`induced inflammatory response was unaffected by IL-13 blockade, collagen deposition
`decreased by more than 85% [53,95], despite continued and undiminished production of IL-4
`[53,96]. Related studies have also shown a dominant role for IL-13 in the pathogenesis of
`pulmonary fibrosis. Over-expression of IL-13 in the lung triggered significant subepithelial
`J Pathol. Author manuscript; available in PMC 2009 June 9.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`Wynn
`
`NIH-PA Author Manuscript
`
`Lassen - Exhibit 1068, p. 5
`
`

`

`Page 6
`airway fibrosis in mice in the absence of any additional inflammatory stimulus [89], while
`treatment with anti-IL-13 antibody markedly reduced collagen deposition in the lungs of
`animals challenged with A. fumigatus conidia [91] or bleomycin [49]. In contrast, transgenic
`mice that over-expressed IL-4 displayed little evidence of subepithelial airway fibrosis, despite
`developing an intense inflammatory response in the lung [97]. Interestingly, two recent studies
`suggested that IL-13-regulated responses [98], including lung fibrosis [99], could develop in
`the absence of IL-4R(cid:302) or Stat6-mediated signalling, suggesting that IL-13 can exploit an
`additional signalling mechanism that is distinct from the IL-4R(cid:302)/Stat6-signalling pathway.
`Indeed, a recent report suggested that TGF(cid:533)1-driven pulmonary fibrosis might in some cases
`be dependent on IL-13-mediated signalling through the IL-13R(cid:302)2 chain [100], which was
`originally thought to operate exclusively as a decoy receptor for IL-13 and as an inhibitor of
`fibrosis [53,101].
`IL-5 and eosinophils have also been shown to regulate tissue fibrogenesis. The differentiation,
`activation and recruitment of eosinophils is highly dependent on IL-5, and eosinophils are an
`important source of fibrogenic cytokines, including TGF(cid:533)1 and IL-13. IL-5 and tissue
`eosinophils have been observed in a variety of diseases, including skin allograft rejection and
`pulmonary fibrosis [86,102,103]. However, studies with neutralizing anti-IL-5 antibodies and
`IL-5 knockout mice have often yielded conflicting results [104]. Early experiments with
`neutralizing anti-IL-5 monoclonal antibodies showed no reduction in liver fibrosis following
`S. mansoni infection, even though tissue eosinophil responses were markedly reduced [105].
`Although negative findings were also reported in some of the skin and lung fibrosis models
`[105,106], other studies observed significant reductions in fibrosis when IL-5 activity was
`neutralized [86,107–110]. A recent study demonstrated that although excessive amounts of
`IL-5 can exacerbate bleomycin-induced fibrosis, IL-5(cid:237)/(cid:237) mice showed no impairment in
`fibrosis [111], suggesting that IL-5 and/or eosinophils act as amplifiers rather than as direct
`mediators of fibrosis. In mice deficient in IL-5 and/or CCL11 (eotaxin), tissue eosinophilia
`was abolished and the ability of CD4+ Th2 cells to produce the profibrotic cytokine IL-13 was
`significantly impaired [112]. Eosinophils were also found to be an important source of IL-13
`in the schistosomiasis-induced model of liver fibrosis [55]. IL-5 and eosinophils can also
`regulate the TGF(cid:533) response in the lungs of mice [109,113]. Thus, one of the key roles of IL-5
`and eosinophils may be to facilitate production of important profibrotic cytokines like IL-13
`and/or TGF(cid:533), which function as the key mediators of fibrosis.
`Finally, similar to IL-5 [55], IL-21/IL-21R signalling was recently shown to promote fibrosis
`by facilitating the development of the CD4+ Th2 response [54]. IL-21R-signalling was also
`critical for Th2-cell survival and for the migration Th2 cells to the peripheral tissues [114]. In
`addition to supporting the development of Th2 responses, IL-21 also increased IL-4 and IL-13
`receptor expression on macrophages [54], which enhances the development of alternatively
`activated macrophages that are believed to be important regulators of fibrosis [66,115].
`Distinct and overlapping roles for TGF(cid:533) and Th2-type cytokines in fibrosis
`TGF(cid:533) has been the most intensively studied regulator of the ECM and has been linked with
`the development of fibrosis in a number of diseases [116–119]. There are three isotypes of
`TGF(cid:533) in mammals, TGF(cid:533)1, -2 and -3, all exhibiting similar biological activity [120]. Although
`a variety of cell types produce and respond to TGF(cid:533) [82], tissue fibrosis is primarily attributed
`to the TGF(cid:533)1 isoform, with circulating monocytes and tissue macrophages being the
`predominant cellular sources. In macrophages, the primary level of control is not in the
`regulation of TGF(cid:533)1 mRNA expression, but in the regulation of both the secretion and
`activation of latent TGF(cid:533)1. TGF(cid:533)1 is stored inside the cell as a disulphide-bonded homodimer,
`non-covalently bound to a latency-associated protein (LAP), which keeps TGF(cid:533) inactive.
`Binding of the cytokine to its receptors requires dissociation of the LAP, a process that is
`J Pathol. Author manuscript; available in PMC 2009 June 9.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`Wynn
`
`NIH-PA Author Manuscript
`
`Lassen - Exhibit 1068, p. 6
`
`

`

`Page 7
`catalysed by several agents, including cathepsins, plasmin, calpain, thrombospondin, integrin-
`(cid:302)v(cid:533)6 and matrix metalloproteinases [82,120,121], many of which have become potential
`targets of antifibrotic drugs. Once activated, TGF(cid:533) signals through transmembrane receptors
`that trigger signalling intermediates known as Smad proteins, which modulate transcription of
`important target genes, including procollagen I and III [122]. Dermal fibrosis following
`irradiation [123] and renal interstitial fibrosis induced by unilateral ureteral obstruction [116]
`are both reduced in Smad3-deficient mice, confirming an important role for the TGF(cid:533)
`signalling pathway. Macrophage-derived TGF(cid:533)1 is thought to promote fibrosis by directly
`activating resident mesenchymal cells including epithelial cells, which differentiate into
`collagen-producing myofibroblasts via EMT. Interestingly, a recent paper showed that the loss
`of TGF(cid:533) signalling in fibroblasts triggers intraepithelial neoplasia, suggesting that TGF(cid:533)1
`signalling critically regulates the activity of fibroblasts as well as the oncogenic potential of
`neighbouring epithelial cells [124]. In the bleomycin model of fibrosis, alveolar macrophages
`are thought to produce nearly all of the active TGF(cid:533) that promotes pulmonary fibrosis [125].
`Nevertheless, Smad3/TGF(cid:533)1-independent mechanisms of fibrosis have also been
`demonstrated in the lung and other tissues [30,126,127], suggesting that profibrotic mediators
`such as IL-4, IL-5, IL-13 and IL-21 can act separately from the TGF(cid:533)/Smad-signalling pathway
`to stimulate collagen deposition.
`There is also evidence that Th2 cytokines cooperate with TGF(cid:533) to induce fibrosis. IL-13
`induces the production of latent TGF(cid:533)1 in macrophages and can also serve as an indirect
`activator of TGF(cid:533) by upregulating expression of proteins that cleave the LAP [128,129].
`Indeed, IL-13 is a potent stimulator of both MMP and cathepsin-based proteolytic pathways
`that activate TGF(cid:533) [74,129]. Thus, the significant tissue remodelling associated with polarized
`Th2 responses may involve a pathway wherein IL-13-expressing CD4+ Th2 cells trigger
`macrophage production of TGF(cid:533)1, which then serves as the major stimulus for fibroblast
`activation and collagen deposition [100,128,130]. In support of this hypothesis, when TGF(cid:533)1
`activity was neutralized in the lungs of IL-13-transgenic mice, development of subepithelial
`fibrosis was significantly reduced [128]. However, related studies observed enhanced
`pulmonary pathology when the TGF(cid:533)/Smad signalling pathway was blocked [131,132],
`suggesting that TGF(cid:533) suppresses rather than induces tissue remodelling in some settings. The
`source of TGF(cid:533)1 appears to be critical, since macrophage-derived TGF(cid:533)1 is often profibrotic
`[128], while T cell-derived TGF(cid:533)1 appears to play a suppressive role [133]. Some studies
`investigating the mechanisms of IL-13-driven fibrosis also reported no reduction in fibrosis in
`MMP-9-, Smad3- and TGF(cid:533)1-deficient mice, suggesting that IL-13 can operate independently
`from TGF(cid:533)1 [30]. This may explain the unexpected failure of Smad/TGF(cid:533) inhibitors in some
`blocking studies [126,127]. Thus, it remains unclear to what extent IL-13 must act through
`TGF(cid:533)1 to trigger fibrosis. Given that numerous antifibrotic therapies are focused on inhibiting
`the TGF(cid:533)1 signalling pathway [82,134], it will be important to determine whether the collagen-
`inducing activity of IL-13 is dependent on TGF(cid:533)1 or whether IL-13 and other profibrotic
`mediators [135] can also operate independently, as has been suggested in some studies [30,
`53,135].
`Vascular changes often accompany the development of fibrosis
`In addition to fibroproliferation and deposition of ECM components, the pathogenesis of IPF,
`systemic sclerosis (SSc), liver fibrosis and many other fibrotic diseases, including many fibrotic
`diseases of the eye, are characterized by substantial vascular remodelling, which often occurs
`prior to the development of fibrosis. In the case of systemic sclerosis, vascular changes are a
`prominent and early manifestation of the disease, with impaired angiogenesis leading to the
`progressive disappearance of blood vessels [28,29]. It has been suggested that reduced numbers
`of circulating bone marrow-derived CD34+ endothelial progenitor cells, as well as their
`impaired differentiation into mature endothelial cells, might be contributing to the early
`J Pathol. Author manuscript; available in PMC 2009 June 9.
`
`NIH-PA Author Manuscript
`
`NIH-PA Author Manuscript
`
`Wynn
`
`NIH-PA Author Manuscript
`
`Lassen - Exhibit 1068, p. 7
`
`

`

`Page 8
`vascular defects in SSc [136]. In contrast to SSc, where fibrosis is associated with the loss of
`blood vessels, fibrosis and traction retinal detachments associated with advanced diabetic
`retinopathy (DR) are characterized by uncontrolled vascular proliferation [137]. Indeed, the
`common pathway for many fibrotic eye diseases, including age-related macular degeneration
`(ARMD) [138], is injury to the cornea and/or retina, which results in inflammatory changes,
`tissue oedema, hypoxia-driven neovascularization and ultimately fibrosis. Once new blood
`vessels begin to grow in the eye, they are prone to haemorrhage, leading to further activation
`of the wound-healing response, and ultimately development of severe fibrosis [139]. Therefore,
`prevention of the primary vascular abnormality has been the most promising therapeutic
`strategy for many diseases of the eye. Because various members of the CXC-chemokine family
`exhibit potent angiogenic or angiostatic activity [140], targeting the CXC-chemokine family
`might offer a unique approach to regulate angiogenesis and fibrosis.
`Angiotensin II plays a critical role in fibrosis
`Although all major components of the renin–angiotensin–aldosterone system exhibit
`profibrotic activity, ANG II appears to be the dominant hormone responsible for cardiac
`fibrosis in hypertensive heart disease [141]. ANG II also plays an important role in the
`development of renal and hepatic fibrosis [142]. ANG II, produ

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