This Key Event Relationship is licensed under the Creative Commons BY-SA license. This license allows reusers to distribute, remix, adapt, and build upon the material in any medium or format, so long as attribution is given to the creator. The license allows for commercial use. If you remix, adapt, or build upon the material, you must license the modified material under identical terms.
Relationship: 1706
Title
Activation of Th2 cells leads to Increased cellular proliferation and differentiation
Upstream event
Downstream event
Key Event Relationship Overview
AOPs Referencing Relationship
AOP Name | Adjacency | Weight of Evidence | Quantitative Understanding | Point of Contact | Author Status | OECD Status |
---|---|---|---|---|---|---|
Substance interaction with the pulmonary resident cell membrane components leading to pulmonary fibrosis | adjacent | High | Low | Cataia Ives (send email) | Under development: Not open for comment. Do not cite | EAGMST Under Review |
Taxonomic Applicability
Sex Applicability
Life Stage Applicability
Key Event Relationship Description
The wound healing process involves an inflammatory phase, during which the damage tissue/wound is provisionally filled with extracellular matrix (ECM). This phase is characterised by secretion of cytokines/chemokines, growth factors and recruitment of inflammatory cells, fibroblasts and endothelial cells. The activated T helper (Th)1/Th2 response and increased pool of specific cytokines and growth factors such as Interleukin (IL)-1β, IL-6, IL-13, and Transforming growth factor beta (TGF-β), induce fibroblast proliferation. Th type 2 (Th2) cells can directly stimulate fibroblasts to synthesise collagen with IL-1 and IL-13. Th2 cytokines IL-13 and IL-4, known to mediate the fibrosis process induce phenotypic transition of human fibroblasts (Hashimoto S, 2001). IL-13 is shown to inhibit Matrix metalloproteinases (MMP)-mediated matrix degradation resulting in excessive collagen deposition by downregulating the synthesis and expression of matrix degrading MMPs. IL-13 is also suggested to induce TGF-β1 in macrophages and its absence results in reduced TGF-β1 expression and decrease in collagen deposition (Fichtner-Feigl et al., 2006). These cytokines are suggested to initiate polarisation of macrophages to the alternative phenotype (M2). Th2 cells that synthesise IL-4 and IL-13 induce synthesis of Arginase (Arg)-1 in M2 macrophages. The Arg-1 pathway stimulates synthesis of proline for collagen synthesis required for fibrosis (Barron and Wynn, 2011).
Evidence Collection Strategy
Evidence Supporting this KER
Biological Plausibility
The biological plausibility for this KER is high. There is a widely understood functional relationship between Th2 response related mediators, and their ability to induce proliferation and differentiation of fibroblasts (Shao et al., 2008; Wynn, 2004; Wynn, 2012).
Empirical Evidence
The empirical support for this KER is high. There is a plethora of dose and time response evidence, which show that Th2 cytokines induce the activation and proliferation of fibroblasts (Hashimoto et al., 2001; Lee et al., 2001; additional references can be found in Table 1).
A majority of the weight of evidence studies assess collagen synthesis as a proxy to fibroblast proliferation and myofibroblast differentiation. A few studies have shown that Th2 cytokine IL-4 stimulates fibroblast proliferation (Sempowski et al., 1994) and production of ECM components (Postlethwaite et al., 1992). In human studies, the progression of idiopathic pulmonary fibrosis (IPF) is also associated with a sustained IL-4 production (Ando et al., 1999; Wallace and Howie, 1999). Th2 cytokines induce expression and activity of TGF-β1, levels of which are elevated in bronchoalveolar lavage fluid (BALF) of patients suffering from lung interstitial diseases, is a potent inducer of myofibroblast differentiation and collagen synthesis (Kurosaka et al., 1998; Redington et al., 1997). Exposure of Signal transducer and activator of transcription 6 (STAT6) deficient mice to multi-walled carbon nanotubes (MWCNTs), suppressed acute lung inflammation, expression of Th2-mediated gene expression, reduced vimentin positive cells (marker of fibroblasts), levels of collagen synthesis and reduced the overall fibrotic response to MWCNTs (Nikota et al., 2017). Mice deficient in IL-33r (St2, Th2 response cytokine) or mice treated with anti-IL33 antibody, showed reduced lung inflammation, reduced collagen production and fibrotic pathology induced by bleomycin. IL-33 deficient mice treated with bleomycin showed reduced levels of IL-1 and other pro-inflammatory cytokines. Mice administered exogenously with mature IL-33 enhanced bleomycin-induced lung inflammation, collagen synthesis and fibrotic lesions (Li et al., 2014).
Dose-Response Relationship:
In vivo and in vitro studies have demonstrated a dose-response relationship, at the higher dose of the stressor, Th2 cells leads to increased, fibroblast proliferation, and myofibroblast differentiation.
Lo Re et al. (2011) evaluated the role of regulatory T cells (Treg cells) in a mouse model of lung fibrosis induced by silica (SiO2) particles. SiO2 particles administered 2.5 mg per mouse by pharyngeal instillation induced an increase in the levels of CD4+Foxp3+ regulatory T lymphocytes in lungs after 3 and 15 days of administration. Treg cells, purified from Foxp3-GFP transgenic mice administered with SiO2, stimulated lung fibroblast proliferation in vitro by producing Platelet derived growth factor subunit B (PDGF-B) and Transforming growth factor beta (TGF-b) in a dose-dependent manner. Moreover, these results indicated that the activation of Th2 response (KE4; Event 1499) was needed to activate fibroblast proliferation (KE5; Event 1500). They determined that effector T cells purified from SiO2-treated mice, in the absence of Treg cells, induced fibrosis by producing IL-4, suggesting that many T cell pathways lead to the fibroproliferative process.
Liu et al. (2011) investigated the role of Found in inflammatory zone 2 (FIZZ2) in pulmonary fibrosis in a rodent bleomycin model and the potential role of FIZZ2 in human fibrotic lung disease. FIZZ2 has been found in pulmonary fibrosis after 14 days of exposure to bleomycin in mice (2U and 10 U/Kg) and in lung tissue from patients with IPF and nonspecific interstitial pneumonia. The expression was localized mainly to airway epithelial cells and alveolar epithelial cells (AECs), and to a lesser extent in alveolar macrophages and smooth muscle and endothelial cells. AECs were isolated from rats and humans, and they were exposed to 10 ng/ml rIL-4, rIL-13, rIL-17 and Interferon gamma (INF-g). After 4 h, rIL-4 and rIL-13, induced FIZZ2 mRNA expression in rat lungs. After 8 h, rIL-13 increased FIZZ2 mRNA expression in rat lungs, and rIL-4 and IL-13 induced an expression of FIZZ2 MRNA in human lungs. These results indicate that FIZZ2 mRNA expression is driven by Th2-type cytokines. Mouse lung fibroblasts were isolated and treated with recombinant mouse FIZZ2 at different concentrations. Collagen I deposition was observed at 10 and 25 ng/ml, and a-smooth muscle actin (a-SMA) was induced at 25, 50, and 200 ng/ml; meanwhile, cell proliferation was observed at 10, 25, and 50 ng/ml. These results suggested that FIZZ2 had direct profibrogenic activity. Furthermore, FIZZ2 acts as a chemoattractant for bone marrow cells, especially BM-derived CD11c+ dendritic cells. In knockout mice treated with bleomycin, a decrease in the FIZZ2 expression was seen, and the adverse effects produced by FIZZ2 decreased. The authors concluded that FIZZ2 is a Th2-associated multifunctional mediator which plays a role in fibroblast proliferation mediated via STAT6 signaling.
Temporal Relationship:
In vitro and in vivo studies have demonstrated that Th2 cells are activated prior to fibroblast proliferation and myofibroblast differentiation.
Dong et al. (2016) demonstrated that MWCNTs activated Th2 immune responses. Male C57BL/6J mice were administered with 40 mg/mouse MWCNTs by pharyngeal aspiration for 1, 3, and 7 days. On days 1, 3, and 7, an increase in the expression of Th2 cytokines (IL-4 and IL-3), as well as an induction of STAT6 and GATA Binding Protein 3 (GATA-3) was seen. At day 7, the presence of collagen I fibers was evident.
In another study, male Wistar rats were administered with 5 mg/kg bleomycin by intratracheal instillation. The inflammatory response was evaluated after 7, 14, and 28 days of exposure. Bleomycin increased hydroxyproline levels, total cell counts, and the expression of Nuclear Factor Kappa B (NF-κB) p65 in lung tissue. Collagen type I increased in a time-dependent manner. At day 7, the Th type 1 (Th1) response was suppressed, based on a decrease of IFN-g and an increase of IL-4 levels. Mice treated with hydrogen sulfide showed less intense effects than mice treated with bleomycin (Cao et al., 2014).
Yin et al. (2013) studied the role of IL-33 in cutaneous wound healing. Male BALB/c mice were injured on the dorsal skin and administered with murine recombinant IL-33 (1.0 mg/mouse). After 5 days of injury, M2 macrophages accumulation and mRNA expression of M2-associated genes increased (fibronectin and collagen). Collagen deposition increased in a time-dependent manner, and the percentage of wound closure and re-epithelization increased over 14 days. This study indicates that macrophage polarization, which is associated with KE4 (Event 1499) preceded the KE5 (Event 1500: fibroblast proliferation). IL-33 is essential for homeostasis and wound healing.
Wynes et al. (2004) evaluated the proliferative response associated with Th2 activation. Insulin-like growth factor-I (IGF-I) is a fibroblast growth and survival factor that has been implicated in the pathogenesis of IPF. The authors observed that mouse bone marrow-derived macrophages from C3H/HeJ mice treated with IL-4 2 ng/ml for 26 h released IGF-I. CCL39 myofibroblasts, cultured with conditioned media from IL-4-treated macrophages, consumed IGF-1 and avoided apoptosis (caspase-3 activity reduced and pro-survival kinases Protein kinase B [Akt] and extracellular signal-regulated kinase [ERK] were activated). The survival effect was lost when IGF was immunodepleted from macrophage-condition media with IGF-I-specific antibodies. These results indicate that a Th2 response conditions macrophages to release mediators which induce persistence of fibroblasts in a fibrotic setting.
Meziani et al. (2018) studied the role of large doses of radiation in promoting M2 macrophage polarization. In patients with thoracic malignancies and preoperative radiotherapy between 25 and 60 Gray (Gy), an infiltration of CD163+ macrophages was found in fibrotic areas. The pulmonary infiltration was characterized during radiation-induced lung fibrosis in a murine model. Female C57BL/6J mice were locally irradiated at the thorax with a dose rate of 1.08 Gy min-1. A single dose of 16 Gy was locally administered to the whole thorax. Infiltrating macrophages (IMs) and alveolar macrophages were isolated post-irradiation. They observed the number of Icam1+ IMs transiently increased at day 6, and an increase number of CD206+ IMs at week 20 post-irradiation. At this time, Th1 cytokines decreased, and Metalloproteinase inhibitor 1 (TIMP-1) increased. Moreover, IMs express high levels of Arg-1. IMs were co-cultured with normal fibroblasts and increased the expression of a-SMA and TGF-b1. They found that IMs isolated from normal mouse lungs, activated in vitro with IL-13 and IL-4 for 24 h, were able to increase a-SMA levels. After 20 weeks, irradiation induced collagen deposition and an increase in the expression of TGF-b1, Plasminogen activator inhibitor 1 (PAI-1), and SMAD family member (Smad)2/3 phosphorylation in lung tissue. Depletion of tissue IMs by anti-Colony stimulating factor 1 receptor (CSF1R) after thoracic irradiation, blocks the observed effects.
Gibbons et al. (2011) studied the role of circulating monocytes and lung macrophages in the pathogenesis of lung fibrosis and the importance of M2 macrophages and Ly6Chi monocyte phenotype. Female C57Bl/6 mice were given 0.05 U bleomycin or 1X108 plaque forming unit (pfu) Adenoviral TGF-b (AdTGF-b) intratracheally. Bleomycin induced early fibrosis at day 18, progressive fibrosis at day 32, or resolving fibrosis at day 56. Bleomycin increased the expression of a-SMA and Collagen type I alpha 1 chain (Col1A1) at day 25. At this time point, they also found an increase in the level of Arg activity and Ym1, and at day 32 an increase in the number of cells per field (markers of M2 macrophages). Macrophages isolated from patients with IPF showed CD163, a human marker of M2 macrophages. AdTGF-b induced an increase in BALF TGF-b at day 5, and collagen deposition at day 14. The administration of liposomal clodronate intratracheally (100 ml) at 10, 21-23 after the exposure to bleomycin or AdTGF-b decreased fibrosis, collagen deposition and M2 macrophages. The depletion of circulating monocytes reduced fibrosis and M2 macrophages. It was found that Ly6Chi inflammatory monocytes were the direct precursor of the M2 lung macrophages.
Uncertainties and Inconsistencies
Due to multifarious functions of several cytokines involved in the process of inflammation and repair, the timing of when a pathway is intervened in an experiment is important in the assessment of the KER studies. For example, exposure to pro-fibrotic bleomycin stimulates IL-4 production during the acute inflammatory phase, which is suggested to limit the recruitment of T lymphocytes and production of damaging cytokines such as Tumor necrosis factor alpha (TNF-α), IFN-γ, and nitric oxide, playing a tissue protective role. However, production of IL- 4 during the chronic phase of tissue repair and healing, favors fibrosis manifestation. Treatment of IL4 -/- mice with low doses of bleomycin induced fewer fibrotic lesions compared to IL-4 +/+ mice. However, treatment of high doses of bleomycin induced more lethality in IL-4 -/- mice compared to the wild type mice (Huaux et al., 2003). Moreover, the KEs represented in AOP 173 can function in parallel in a positive feedback loop, perpetuating and magnifying the response at each stage. The resulting microenvironment may contain the same molecules in different proportions exhibiting different functions. Thus, the complexity of the process and the functional heterogeneity of the molecular players involved, makes it nearly impossible to establish KERs using a targeted deletion of one single gene or a pathway in a study, which is how most of the studies are designed.
Known modulating factors
Quantitative Understanding of the Linkage
Response-response Relationship
Time-scale
Known Feedforward/Feedback loops influencing this KER
Domain of Applicability
References
- Ando M, Miyazaki E, Fukami T, Kumamoto T, Tsuda T. Interleukin-4-producing cells in idiopathic pulmonary fibrosis: an immunohistochemical study. Respirology. 1999 Dec;4(4):383-91. doi: 10.1046/j.1440-1843.1999.00209.x.
- Barron L, Wynn TA. Fibrosis is regulated by Th2 and Th17 responses and by dynamic interactions between fibroblasts and macrophages. Am J Physiol Gastrointest Liver Physiol. 2011 May;300(5):G723-8. doi: 10.1152/ajpgi.00414.2010.
- Cao H, Zhou X, Zhang J, Huang X, Zhai Y, Zhang X, Chu L. Hydrogen sulfide protects against bleomycin-induced pulmonary fibrosis in rats by inhibiting NF-κB expression and regulating Th1/Th2 balance. Toxicol Lett. 2014 Jan 30;224(3):387-94. doi: 10.1016/j.toxlet.2013.11.008.
- Dong J, Ma Q. In vivo activation of a T helper 2-driven innate immune response in lung fibrosis induced by multi-walled carbon nanotubes. Arch Toxicol. 2016 Sep;90(9):2231-2248. doi: 10.1007/s00204-016-1711-1.
- Fichtner-Feigl S, Strober W, Kawakami K, Puri RK, Kitani A. IL-13 signaling through the IL-13alpha2 receptor is involved in induction of TGF-beta1 production and fibrosis. Nat Med. 2006 Jan;12(1):99-106. doi: 10.1038/nm1332.
- Gibbons MA, MacKinnon AC, Ramachandran P, Dhaliwal K, Duffin R, Phythian-Adams AT, van Rooijen N, Haslett C, Howie SE, Simpson AJ, Hirani N, Gauldie J, Iredale JP, Sethi T, Forbes SJ. Ly6Chi monocytes direct alternatively activated profibrotic macrophage regulation of lung fibrosis. Am J Respir Crit Care Med. 2011 Sep 1;184(5):569-81. doi: 10.1164/rccm.201010-1719OC.
- Hashimoto S, Gon Y, Takeshita I, Maruoka S, Horie T. IL-4 and IL-13 induce myofibroblastic phenotype of human lung fibroblasts through c-Jun NH2-terminal kinase-dependent pathway. J Allergy Clin Immunol. 2001 Jun;107(6):1001-8. doi: 10.1067/mai.2001.114702.
- Huaux F, Liu T, McGarry B, Ullenbruch M, Phan SH. Dual roles of IL-4 in lung injury and fibrosis. J Immunol. 2003 Feb 15;170(4):2083-92. doi: 10.4049/jimmunol.170.4.2083.
- Kurosaka H, Kurosaka D, Kato K, Mashima Y, Tanaka Y. Transforming growth factor-beta 1 promotes contraction of collagen gel by bovine corneal fibroblasts through differentiation of myofibroblasts. Invest Ophthalmol Vis Sci. 1998 Apr;39(5):699-704.
- Lee CG, Homer RJ, Zhu Z, Lanone S, Wang X, Koteliansky V, Shipley JM, Gotwals P, Noble P, Chen Q, Senior RM, Elias JA. Interleukin-13 induces tissue fibrosis by selectively stimulating and activating transforming growth factor beta(1). J Exp Med. 2001 Sep 17;194(6):809-21. doi: 10.1084/jem.194.6.809.
- Li D, Guabiraba R, Besnard AG, Komai-Koma M, Jabir MS, Zhang L, Graham GJ, Kurowska-Stolarska M, Liew FY, McSharry C, Xu D. IL-33 promotes ST2-dependent lung fibrosis by the induction of alternatively activated macrophages and innate lymphoid cells in mice. J Allergy Clin Immunol. 2014 Dec;134(6):1422-1432.e11. doi: 10.1016/j.jaci.2014.05.011.
- Liu T, Baek HA, Yu H, Lee HJ, Park BH, Ullenbruch M, Liu J, Nakashima T, Choi YY, Wu GD, Chung MJ, Phan SH. FIZZ2/RELM-β induction and role in pulmonary fibrosis. J Immunol. 2011 Jul 1;187(1):450-61. doi: 10.4049/jimmunol.1000964.
- Lo Re S, Lecocq M, Uwambayinema F, Yakoub Y, Delos M, Demoulin JB, Lucas S, Sparwasser T, Renauld JC, Lison D, Huaux F. Platelet-derived growth factor-producing CD4+ Foxp3+ regulatory T lymphocytes promote lung fibrosis. Am J Respir Crit Care Med. 2011 Dec 1;184(11):1270-81. doi: 10.1164/rccm.201103-0516OC.
- Meziani L, Mondini M, Petit B, Boissonnas A, Thomas de Montpreville V, Mercier O, Vozenin MC, Deutsch E. CSF1R inhibition prevents radiation pulmonary fibrosis by depletion of interstitial macrophages. Eur Respir J. 2018 Mar 1;51(3):1702120. doi: 10.1183/13993003.02120-2017.
- Nikota J, Banville A, Goodwin LR, Wu D, Williams A, Yauk CL, Wallin H, Vogel U, Halappanavar S. Stat-6 signaling pathway and not Interleukin-1 mediates multi-walled carbon nanotube-induced lung fibrosis in mice: insights from an adverse outcome pathway framework. Part Fibre Toxicol. 2017 Sep 13;14(1):37. doi: 10.1186/s12989-017-0218-0.
- Postlethwaite AE, Holness MA, Katai H, Raghow R. Human fibroblasts synthesize elevated levels of extracellular matrix proteins in response to interleukin 4. J Clin Invest. 1992 Oct;90(4):1479-85. doi: 10.1172/JCI116015.
- Redington AE, Madden J, Frew AJ, Djukanovic R, Roche WR, Holgate ST, Howarth PH. Transforming growth factor-beta 1 in asthma. Measurement in bronchoalveolar lavage fluid. Am J Respir Crit Care Med. 1997 Aug;156(2 Pt 1):642-7. doi: 10.1164/ajrccm.156.2.9605065.
- Sempowski GD, Beckmann MP, Derdak S, Phipps RP. Subsets of murine lung fibroblasts express membrane-bound and soluble IL-4 receptors. Role of IL-4 in enhancing fibroblast proliferation and collagen synthesis. J Immunol. 1994 Apr 1;152(7):3606-14.
- Shao DD, Suresh R, Vakil V, Gomer RH, Pilling D. Pivotal Advance: Th-1 cytokines inhibit, and Th-2 cytokines promote fibrocyte differentiation. J Leukoc Biol. 2008 Jun;83(6):1323-33. doi: 10.1189/jlb.1107782.
- Wallace WA, Howie SE. Immunoreactive interleukin 4 and interferon-gamma expression by type II alveolar epithelial cells in interstitial lung disease. J Pathol. 1999 Mar;187(4):475-80. doi: 10.1002/(SICI)1096-9896(199903)187:4<475::AID-PATH268>3.0.CO;2-N.
- Wynes MW, Frankel SK, Riches DW. IL-4-induced macrophage-derived IGF-I protects myofibroblasts from apoptosis following growth factor withdrawal. J Leukoc Biol. 2004 Nov;76(5):1019-27. doi: 10.1189/jlb.0504288.
- Wynn TA. Fibrotic disease and the T(H)1/T(H)2 paradigm. Nat Rev Immunol. 2004 Aug;4(8):583-94. doi: 10.1038/nri1412.
- Wynn TA, Ramalingam TR. Mechanisms of fibrosis: therapeutic translation for fibrotic disease. Nat Med. 2012 Jul 6;18(7):1028-40. doi: 10.1038/nm.2807.
- Yin H, Li X, Hu S, Liu T, Yuan B, Gu H, Ni Q, Zhang X, Zheng F. IL-33 accelerates cutaneous wound healing involved in upregulation of alternatively activated macrophages. Mol Immunol. 2013 Dec;56(4):347-53. doi: 10.1016/j.molimm.2013.05.225.