Downloads

Song, W., Sun, W., Wang, Z., Teo , K. Y. C., Cheung, C. M. G., & Wang, X. . Targeting Inflammation to Control Tissue Fibrosis. International Journal of Drug Discovery and Pharmacology. 2022, 1(1), 6. doi: https://doi.org/10.53941/ijddp.v1i1.206

Review

Targeting Inflammation to Control Tissue Fibrosis

Weihua Song 1, Wu Sun 2, Zilong Wang 3, Kelvin Yi Chong Teo 2,4,5, Chui Ming Gemmy Cheung 2,4,5, and Xiaomeng Wang 4,5,6,*

1 Innoland Biosciences, 6 West Beijing Road, Taicang 215400, Jiangsu, China.

2 Singapore National Eye Center, 11 Third Hospital Ave 168751, Singapore.

3 Ocean University of China, 5 Yushan Rd, Shinan District, Qingdao 266005, Shandong, China.

4 Singapore Eye Research Institute, 20 College Road 169856, Singapore.

5 Duke-NUS Graduate Medical School, 20 College Road 169856, Singapore.

6 Insitute of Molecular and Cell Biology, 61 Biopolis Dr, Proteos 138673, Singapore.

* Correspondence: xiaomeng.wang@duke-nus.edu.sg

 

 

Received: 17 November 2022

Accepted: 19 November 2022

Published: 21 December 2022

 

Abstract: Remodeling of the extracellular matrix (ECM) is an essential process in host defense against pathogens and tissue repair following injury. However, aberrant inflammatory responses could disturb ECM homeostasis leading to progressive disruption in tissue architecture and organ function. Fibrosis is the common outcome of a wide range of diseases, especially chronic inflammatory disorders, and represents the leading cause of morbidity and mortality globally. This review provides the current understanding of the pathogenesis of fibrosis, with particular emphasis on the role of inflammation in this process and the translational potential of targeting inflammation as a strategy to control fibrotic progression.

Keywords:

inflammation fibrosis cytokines

1. Introduction

Inflammation is the first response mechanism following tissue injury. Various inflammatory cells, especially neutrophils and monocytes, are activated and recruited to the injured tissue, which serves as rich sources of cytokines, chemokines, and growth factors. These damage-associated signals are also essential for activating different effector cells within the tissue microenvironment, such as residential fibroblasts, to promote their proliferation, contraction, and the production of collagenous and non-collagenous extracellular matrix (ECM) components [1]. Furthermore, inflammatory molecules have been reported to induce the myofibroblast transformation through epithelial-mesenchymal transition (EMT) [2] and endothelial-mesenchymal transition (EndMT) [3] process. Recently, infiltrating immune cells have been shown to contribute to the fibrotic progression by directly transforming into myofibroblasts, in a process called macrophage-to-myofibroblast transition (MMT), in the kidney [4] and the eye [5]. The acute inflammatory response is critical for limiting blood loss and preventing the pathogen spread and invasion [6]. Timely resolution of the inflammatory response is essential for restoring homeostasis and preserving tissue integrity and normal organ function. The persistent inflammatory response may trigger uncontrolled ECM remodeling and lead to permanent tissue damage and fibrosis which is associated with many chronic inflammatory diseases [7]. Although numerous factors have been reported to contribute to fibrotic progression, the underlying molecular mechanisms are poorly defined and effective management of fibrotic diseases remains a significant unmet medical need. In recent years, the importance of inflammation in fibrosis has attracted great attention. This review focuses on the role of common inflammatory molecules in fibrotic progression and the potential of targeting these pathways to control tissue fibrosis and restore tissue homeostasis.

2. Transforming Growth Factor-Beta (TGF‑β)

Transforming growth factor-beta (TGF‑β) is a multifunctional growth factor with potent inflammatory activity [8] and has been shown to regulate the function of T cells [9‒13], natural killer (NK) cells [14], and dendritic cells (DC) [15]. At the same time, TGF‑β serves as one of the most potent regulators of tissue fibrosis. Besides inducing the differentiation of fibroblasts into myofibroblasts [16], TGF‑β promotes EMT in epithelial cells originating from the lung, liver, lens, or kidney [17] by regulating the activity of both canonical and non-canonical signaling transducers [18‒20]. Deletion of receptor-mediated Smad [21] or introduction of exogenous inhibitory Smad [22] leads to attenuated EMT and fibrosis in mice. Inhibitors targeting PI3K/Akt [23] and MAPK [24] have also been shown to inhibit fibrosis in preclinical studies and clinical trials. TGF‑β’s role in EndMT has also been extensively investigated and was shown to regulate EndMT through the canonical Smad signalling pathway [25,26]. Connective Tissue Growth Factor (CTGF) was reported to mediate, at least partially, the profibrotic effects of TGF-β. A functional Smad3 binding site has been identified in the CTGF promoter and TGF-β has been shown to induce the expression of CTGF in fibroblasts leading to the subsequent differentiation into myofibroblast [27]. On the other hand, CTGF has been reported to enhance the binding of TGF-β to its type II receptor TβRII leading to subsequent activation of downstream signalling transducers and potentiating the TGF‑β-mediated fibrogenic actions [28]. Despite being recognized as a central pathway of fibrosis, TGF‑β is not an ideal therapeutic targets due to its multifunctional role. Long-term inhibition of TGF‑β has been shown to cause serious adverse effects.

3. Tumor Necrosis Factor Alpha (TNF-α)

Tumor necrosis factor alpha (TNF-α) is a pleiotropic cytokine that is cleaved from the membrane-bound precursors into the soluble mature form by TNF-α-converting enzyme (TACE) [29]. Both precursor and mature TNF‑α can activate downstream signalling transducers, through TNFα receptors. Although being named because of its role in promoting tumour necrosis, TNF-α is now believed to regulate diverse functions, including inflammation [30], angiogenesis [31‒33], tumorigenesis [34] as well as tissue fibrosis [35]. Physiological, TNF-α plays a critical role in normal immune response, however, inappropriate activation of TNF-α contributes to the development of a variety of complications. In terms of tissue fibrosis, TNF-α’s role in liver fibrosis has been studied extensively. Hepatic stellate cells (HSCs) can differentiate into myofibroblasts and serve as the primary contributor to liver fibrosis [36]. Macrophages have been shown to serve as a major source of TNF-α [37] and macrophage-derived TNF-α promotes liver fibrosis by regulating the NF-kB-mediated HSC survival [38]. The synergistic effect of TNF-α and TGF‑β or other inflammatory factors on EMT and EndMT has also been reported [39‒41]. On the other hand, TNF‑α has been shown to reduce ECM deposition by reducing the expression of matrix metalloproteinase-9 (MMP9) [42] or inhibiting the synthesis of ECM components such as collagen I [43‒47], through mediating the TGF‑β-signalling [48‒50]. Therefore, the role of TNF-α remains controversial.

4. Interleukins

Interleukins (ILs) are a group of cytokines that play important immunoregulatory functions. They regulate the immune system by promoting the proliferation and differentiation of immune cells and recruiting them to the injury sites [51‒54]. Recently, ILs have been reported to govern fibrogenesis in serval organ systems.

IL1 family of cytokines consists of members with both proinflammatory and anti-inflammatory properties [55]. Many of them have been implicated in tissue fibrosis. For example, IL-1β is induced in the lungs of mice subjected to bleomycin-induced pulmonary fibrosis [56], in sputum and lung tissues of human patients with chronic obstructive pulmonary disease (COPD) [57], a disease characterized by fibrotic remodeling of the small airways [58], and in bronchoalveolar lavage (BAL) and lung biopsies of patients with idiopathic pulmonary fibrosis (IPF) [59]. Studies showed that IL-1β deficient mice are protected from bleomycin-induced pulmonary fibrosis [56]. Consistent with this observation, administration of IL-1β can lead to pulmonary fibrosis to a comparable magnitude to bleomycin [59]. Transient overexpressing IL-1β in rat lungs has also been reported to cause severe progressive tissue fibrosis [60], whereas neutralizing IL-1β antibody can attenuate the silica-induced lung fibrosis by inhibiting the expression of TGF‑β1 [61]. EMT of alveolar epithelial cells into myofibroblasts contributes to fibrotic progression in the lung [62] and IL‑1β was reported to be actively involved in this process [63]. Besides the lung, IL-1β was reported to regulate renal and hepatic fibrosis by mediating the EMT process, and neutralizing antibody targeting IL-1β prevented the transformation of renal proximal tubular epithelial cells and hepatic stellate cells [64,65]. Therefore, targeting IL-1β may offer an attractive strategy to control tissue fibrosis.

IL-6 is a pleiotropic cytokine with diverse biological functions. Its roles in inflammation and immunity are most well characterized. IL-6 was initially demonstrated to induce immunoglobulin production by directly affecting B cells [66] and STAT3 was reported to play a critical role in this process [67]. Consistent with this observation, IL-6 deficient mice are defective in immunoglobulin production [68,69]. Besides its role in B cells, IL-6 has also been reported in regulating T cell function in a STAT3-dependent manner [70,71]. Interestingly, elevated IL-6 levels are observed in the injured skin tissues [72], patients with pulmonary fibrosis [73], systemic sclerosis [74], and liver cirrhosis [75], suggesting a potential disease-modifying role of IL-6 in tissue fibrosis. Indeed, IL-6 is shown to promote collagen synthesis and tissue fibrosis in the lung[76], kidney [77], heart [78], and skin [79]. Furthermore, increased IL-6 levels as a consequence of repeated inflammation are believed to regulate Th1 cell responses in peritoneal fibrosis, whereas IL-6 deficient mice are resistant to fibrosis [80]. Similarly, the genetic ablation of IL-6 leads to attenuated fibrosis in a bleomycin-induced murine model [81]. It was previously demonstrated that genetic or pharmacologic removal of IL-6 resulted in the attenuation of fibrosis [73]. Tocilizumab is a humanized monoclonal antibody against the IL-6 receptor. Blocking IL-6 trans signalling with Tocilizumab leads to improved skin scores in patients with diffuse systemic sclerosis [82]. These data provide evidence of IL-6 as an attractive therapeutic target for treating fibrosis.

IL-7 is a member of the IL-2 superfamily and signals through IL-7R [83]. IL-7/IL-7R signalling is essential for the development of T cells and mouse B cells, the differentiation and survival of naive T cells, and the generation and maintenance of memory T cells [83]. Unlike IL-1β, IL7’s role in tissue fibrosis is less understood. However, it was reported to downregulate TGF‑β production in macrophages in an IFN-γ-independent manner [84]. Consistent with these in vitro observations, recombinant IL-7 has been shown to reduce bleomycin-induced pulmonary fibrosis in mice by suppressing the expression of TGF-β in a JAK1/STAT1-dependent manner [85]. In addition, IL-7-mediated inhibition of TGF-β signaling was associated with an increase in the expression of an inhibitory Smad, Smad7 [85]. Similarly, IL-7 was also reported to attenuate high-glucose-induced activation of the TGF‑β signaling pathway, EMT of renal proximal tubule epithelial cells and renal fibrosis [86]. Consistent with the observations in pulmonary fibroblasts, high-glucose-induced inhibition of Smad7 is significantly reversed by IL-7 [86]. In liver fibrosis, the IL7RA rs6897932 polymorphism was reported to be associated with an increased risk of liver fibrosis progression in HCV-infected patients [87], and regulating IL7R expression by targeting miR-122-5p can inhibit HBV-related liver fibrosis [88].

5. Chemokines

Chemokines are soluble, membrane-bound proteins that act on the superfamily of G-protein coupled serpentine receptors expressed on various target cells [ 89 ]. They act synergistically with other cytokines to recruit and activate various effectors cells, such as myofibroblasts, endothelial cells, neutrophils, and monocytes, within the tissue microenvironment in response to tissue injury [ 90 ]. Based on the number of amino acids located between the N-terminal cysteine residues, chemokines could be divided into four groups, C, CC, CXC, and CX 3 C [ 91 ].

5.1. CXCL

The CXC family of chemokines are important mediators of inflammatory response and their main function is to guide the neutrophils to the site of infection [92] and activates them [93]. Besides its important roles in inflammation, CXCLs also participate in tissue fibrosis. CXCL8 is induced in patients with IPF [94,95], pneumoconiosis [96], liver fibrosis [97], and cystic fibrosis [98]. Mesenchymal progenitor cell (MPC)-derived CXCL8 promotes pulmonary fibrosis by increasing MPC proliferation and recruiting activated macrophages in the lung. CXCL8’s role in EMT has also been reported [99], however, how whether it is involved in EMT-mediated tissue fibrosis remains to be investigated. A similar role of CXCL10 has also been reported. CXCL10 is induced in severe hepatitis C virus (HCV)-induced liver fibrosis [100] and the delection of CXCL10 leads to reduced liver fibrosis [101]. CXCL4 is another pro-fibrotic chemokine and it regulates liver fibrosis by inhibiting the migration of CD8+T cells [102]. CXC chemokines are also involved in pulmonary fibrosis, primarily through their action on T helper 1 and Natural Killer T cells [103]. Blocking CXCLs, such as CXCL2, significantly attenuates the development of pulmonary fibrosis [103]. However, other CXCLs, including CXCL10 and CXCL11, serve as potent inhibitors of pulmonary fibrosis [104,105].

5.2. CCL

Hepatic stellate cells (HSC) express a multitude of CCLs, such as CCL2, CCL3, CCL5, and CCL21 [106,107]. Inhibition of CCL2 and CCL21 indeed attenuates liver fibrosis in vivo [107,108]. CCLs have also been implicated in the pathogenesis of pulmonary fibrosis. CCL2 mRNA and protein expression are highly induced in lung epithelial cells and bronchoalveolar fluid from human patients with IPF [109], and the profibrotic role of CCL2 has been demonstrated in various animal models of pulmonary fibrosis [110‒112]. However, clinical trial results on the CCL2 CCL2-neutralizing antibody in patients with IPF was rather disappointing [113]. CCR7 is a cognate receptor of CCL21 and it is expressed in IPF but not normal fibroblasts [114]. Furthermore, CCL21 can activate CCR7 on fibroblasts isolated from patients with IPF to promote their proliferation migration and chemokine expression in a mitogen-activated protein kinase (MAPK) 1/2-dependent manner [114]. Indeed, the CCL21 neutralizing antibody has been shown to attenuate pulmonary fibrosis in vivo [115]. Besides the lung, CCL21 is involved in the fibrotic progression in different organs and tissues, including the lymph nodes [116], heart [117], and kidney [118]. For example, CCL21 is expressed by high endothelial venules in lymph nodes and Peyer’s patches and by stromal cells in the T-cell areas of secondary lymphoid organs [119]. The binding between CCL21 and its receptor CCR7 is essential in the organization of normal lymphoid tissue during development [119]. CCL21 can also stimulate the recruitment of CCR7+ dendritic cells (DCs) and lymphocytes into both renal draining lymph nodes (RDLNs) and spleen, resulting in a systemic lymphocyte expansion, which plays a critical role in driving fibrosis following renal injury. Consistently, Injury-induced intrarenal inflammation and fibrosis could be attenuated by blocking the recruitment of CCR7+ cells into RDLN and spleen or inhibiting lymphangiogenesis [116]. In addition, blockading CCL21/CCR7 signaling using a neutralizing CCL21 antibody effectively attenuates renal fibrosis by reducing the recruitment of macrophages and renal transcripts of monocyte chemoattractant protein-1 (MCP-1/CCL2) [116]. It is worthy highlighting that the CCL21-induced migration of fibrocytes is chemotactic but not chemokinetic [120].

6. Future Directions

Despite the advances in understanding the pathophysiological mechanisms of fibrosis, effective management of fibrotic diseases remains challenging. At the site of tissue injury or infection, recruited and activated inflammatory cells contribute to fibrotic progression by interacting with resident cells within the tissue microenvironment [121]. Targeting inflammatory regulators may serve as an attractive approach for developing novel anti-fibrotic therapeutics. However, the complex role of inflammation molecules and the heterogeneity of different fibrotic disease contexts warrant further investigation in the search for novel therapeutic targets for a specific type of fibrotic disease. Recent single-cell multi-omics approaches looking at molecular changes in distinct cell populations of healthy and diseased tissue samples with unprecedented resolution revolutionized our understanding of disease pathogenesis [122,123]. These approaches offer a powerful exploration of cell states and types at the single-cell level, helping us to generate new insights into the disease mechanisms associated with fibrosis.

Author Contributions: Writing-original draft preparation, W.S., W.S., Z.W., K.Y.C.T, C.M.G.C., and X.W; Writing review and editing, W.S., W.S., Z.W., K.Y.C.T, C.M.G.C., and X.W; Funding acquisition: C.M.G.C, K.Y.C.T and X.W. All authors have read and agreed to the published version of the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding: This research is funded by NMRC-LCG grant Translational Asian Age-related Macular Degeneration Programme (TAAP): NMRC/LCG/2018/004.

Acknowledgments: This work was supported by grants from National Medical Research Council Singapore Large Collaborative Grant TAAP (NMRC/OFLCG/004/2018) to GCMC and XMW.

Conflicts of Interest: The authors declare no conflict of interest.

References

  1. Duffield J.S.; Lupher M.; Thannickal V.J.; et al. Host responses in tissue repair and fibrosis. Annu. Rev. Pathol., 2013, 8, 241-276.
  2. Suarez-Carmona M.; Lesage J.; Cataldo D.; et al. EMT and inflammation: inseparable actors of cancer progression. Mol. Oncol., 2017, 11(7): 805-823.
  3. Cho J. G.; Lee A.; Chang W.; et al. Endothelial to Mesenchymal Transition Represents a Key Link in the Interaction between Inflammation and Endothelial Dysfunction. Front. Immunol., 2018, 9, 294.
  4. Meng X. M.; Wang S.; Huang X. R.; et al. Inflammatory macrophages can transdifferentiate into myofibroblasts during renal fibrosis. Cell Death Dis., 2016, 7(12): e2495.
  5. Little K.; Llorián-Salvador M.; Tang M.; et al. Macrophage to myofibroblast transition contributes to subretinal fibrosis secondary to neovascular age-related macular degeneration. J. Neuroinflammation, 2020, 17(1): 355.
  6. Medzhitov R. Inflammation 2010: new adventures of an old flame. Cell, 2010, 140(6): 771-776, doi:10.1016/j.cell.2010.03.006.
  7. Wynn T. A. Cellular and molecular mechanisms of fibrosis. J. Pathol., 2008, 18(3): 199-210, doi:10.1002/path.2277.
  8. Li M.O.; Flavell R.A. Contextual regulation of inflammation: a duet by transforming growth factor-beta and interleukin-10. Immunity 2008, 28(4): 468-476, doi:10.1016/j.immuni.2008.03.003 .
  9. Li M.O.; Sanjabi S.; Flavell R.A. Transforming growth factor-beta controls development, homeostasis, and tolerance of T cells by regulatory T cell-dependent and -independent mechanisms. Immunity, 2006, 25(3): 455-471, doi:10.1016/j.immuni.2006.07.011.
  10. Marie J.C.; Liggitt D.; Rudensky A.Y. Cellular mechanisms of fatal early-onset autoimmunity in mice with the T cell-specific targeting of transforming growth factor-beta receptor. Immunity, 2006, 25 (3): 441-454, doi:10.1016/j.immuni.2006.07.012.
  11. Li M.O.; Flavell R.A. TGF-beta: a master of all T cell trades. Cell, 2008, 134(3): 392-404, doi:10.1016/j.cell.2008.07.025.
  12. Zhou L.; Lopes J.E.; Chong M.M.; et al. TGF-beta-induced Foxp3 inhibits T(H)17 cell differentiation by antagonizing RORgammat function. Nature, 2008, 453(7192): 236-240, doi:10.1038/nature06878.
  13. Filippi C.M.; Juedes A.E.; Oldham J.E.; et al. Transforming growth factor-beta suppresses the activation of CD8+ T-cells when naive but promotes their survival and function once antigen experienced: a two-faced impact on autoimmunity. Diabetes, 2008, 57(10): 2684-2692, doi:10.2337/db08-0609.
  14. Laouar Y.; Sutterwala F.S.; Gorelik L.; et al. Transforming growth factor-beta controls T helper type 1 cell development through regulation of natural killer cell interferon-gamma. Nat. Immunol., 2005, 6(6): 600-607, doi:10.1038/ni1197.
  15. Laouar Y.; Town T.; Jeng D.; et al. TGF-beta signaling in dendritic cells is a prerequisite for the control of autoimmune encephalomyelitis. Proc. Natl. Acad. Sci., 2008, 105(31): 10865-10870, doi:10.1073/pnas.0805058105.
  16. Frangogiannis N. Transforming growth factor-beta in tissue fibrosis. J. Exp. Med., 2020, 217(3): e20190103, doi:10.1084/jem.20190103.
  17. Miettinen P.J.; Ebner R.; Lopez A.R.; et al. TGF-beta induced transdifferentiation of mammary epithelial cells to mesenchymal cells: involvement of type I receptors. J. Cell Biol., 1994, 127(6 Pt 2): 2021-2036, doi:10.1083/jcb.127.6.2021.
  18. Derynck R.; Zhang Y. E. Smad-dependent and Smad-independent pathways in TGF-beta family signalling. Nature, 2003, 425(6958): 577-584, doi:10.1038/nature02006.
  19. Wendt M.K.; Allington T.M.; SchiemannW. P. Mechanisms of the epithelial-mesenchymal transition by TGF-beta. Future Oncol., 2009, 5(8): 1145-1168, doi:10.2217/fon.09.90.
  20. Kriz W.; Kaissling B.; Le Hir M. Epithelial-mesenchymal transition (EMT) in kidney fibrosis: fact or fantasy? J. Clin. Invest. 2011, 214 (2): 468-474, doi:10.1172/jci44595.
  21. Meng F.; Li J.; Yang X.; et al. Role of Smad3 signaling in the epithelialmesenchymal transition of the lens epithelium following injury. Int. J. Mol. Med., 2018, 42(2): 851-860, doi:10.3892/ijmm.2018.3662.
  22. Zeisberg M.; Hanai J.; Sugimoto H.; et al. BMP-7 counteracts TGF-beta1-induced epithelial-to-mesenchymal transition and reverses chronic renal injury. Nat. Med., 2003 9(7): 964-968, doi:10.1038/nm888.
  23. Wang J.; Hu K.; Cai X.; et al. Targeting PI3K/AKT signaling for treatment of idiopathic pulmonary fibrosis. Acta Pharm. Sin. B 2022, 12(1): 18-32, doi:10.1016/j.apsb.2021.07.023.
  24. Dolivo D.M.; Larson S.A.; Dominko T. Crosstalk between mitogen-activated protein kinase inhibitors and transforming growth factor-beta signaling results in variable activation of human dermal fibroblasts. Int J Mol Med 2019, 43(1): 325-335, doi:10.3892/ijmm.2018.3949.
  25. Ma J.; van der Zon G.; Sanchez-Duffhues G.; et al. TGF-beta-mediated Endothelial to Mesenchymal Transition (EndMT) and the Functional Assessment of EndMT Effectors using CRISPR/Cas9 Gene Editing. J. Vis. Exp., 2021, 168, doi:10.3791/62198.
  26. Song S.; Zhang R.; Cao W.; et al . Foxm1 is a critical driver of TGF-beta-induced EndMT in endothelial cells through Smad2/3 and binds to the Snail promoter. J. Cell Physiol., 2019, 234(6): 9052-9064, doi:10.1002/jcp.27583.
  27. Ihn H. Pathogenesis of fibrosis: role of TGF-beta and CTGF. Curr. Opin. Rheumatol., 2002, 14(6): 681-685, doi:10.1097/00002281-200211000-00009.
  28. Abreu J.G.; Ketpura N.I.; Reversade B.; et al. Connective-tissue growth factor (CTGF) modulates cell signalling by BMP and TGF-beta. Nat. Cell Biol., 2002, 4(8): 599-604, doi:10.1038/ncb826.
  29. Idriss H.T.; Naismith J. H. TNF alpha and the TNF receptor superfamily: structure-function relationship(s). Microsc. Res. Tech., 2000, 50(3): 184-195, doi:10.1002/1097-0029(20000801)50:3<184::AID-JEMT2>3.0.CO;2-H.
  30. Bradley J.R. TNF-mediated inflammatory disease. J. Pathol., 2008, 214(2): 149-160, doi:10.1002/path.2287.
  31. Montrucchio G.; Lupia E.; Battaglia E.; et al. Tumor necrosis factor alpha-induced angiogenesis depends on in situ platelet-activating factor biosynthesis. J. Exp. Med., 1994, 180(1): 377-382, doi:10.1084/jem.180.1.377.
  32. Hammam O.; Mahmoud O.; Zahran M.; et al. A Possible Role for TNF-alpha in Coordinating Inflammation and Angiogenesis in Chronic Liver Disease and Hepatocellular Carcinoma. Gastrointest. Cancer Res., 2013, 6(4): 107-114.
  33. Pandey A.; Shao H.; Marks R.M.; et al. Role of B61, the ligand for the Eck receptor tyrosine kinase, in TNF-alpha-induced angiogenesis. Science, 1995, 268(5210): 567-569, doi:10.1126/science.7536959.
  34. Wang X.; Lin Y. Tumor necrosis factor and cancer, buddies or foes? Acta. Pharmacol. Sin., 2008,29 (11): 1275-1288, doi:10.1111/j.1745-7254.2008.00889.x.
  35. Theiss A.L.; Simmons J.G.; Jobin C.; et al. Tumor necrosis factor (TNF) alpha increases collagen accumulation and proliferation in intestinal myofibroblasts via TNF receptor 2. J. Biol. Chem., 2005 280(43): 36099-36109, doi:10.1074/jbc.M505291200.
  36. Mederacke I.; Hsu C.C.; Troeger J.S.; et al. Fate tracing reveals hepatic stellate cells as dominant contributors to liver fibrosis independent of its aetiology. Nat. Commun., 2013, 4, 2823, doi:10.1038/ncomms3823.
  37. TraceyK. J.; Cerami A. Tumor necrosis factor: an updated review of its biology. Crit. Care Med., 1993, 21(10 Suppl): S415-422.
  38. Pradere J. P.; Kluwe J.; De Minicis S.; et al. Hepatic macrophages but not dendritic cells contribute to liver fibrosis by promoting the survival of activated hepatic stellate cells in mice. Hepatology, 2013, 58(4): 1461-1473, doi:10.1002/hep.26429.
  39. Bates R.C.; Mercurio A.M. Tumor necrosis factor-alpha stimulates the epithelial-to-mesenchymal transition of human colonic organoids. Mol. Biol. Cell, 2003, 14(5): 1790-1800, doi:10.1091/mbc.e02-09-0583.
  40. Liao S.J.; Luo J.; Li D.; et al. TGF-beta1 and TNF-alpha synergistically induce epithelial to mesenchymal transition of breast cancer cells by enhancing TAK1 activation. J. Cell Commun. Signal, 2019, 13(3): 369-380, doi:10.1007/s12079-019-00508-8.
  41. Dong W.; Sun S.; Cao X.; et al. Exposure to TNFalpha combined with TGFbeta induces carcinogenesis in vitro via NF-kappaB/Twist axis. Oncol. Rep. 2017, 37(3): 1873-1882, doi:10.3892/or.2017.5369.
  42. Knittel T.; Mehde M.; Kobold D.; et al. Expression patterns of matrix metalloproteinases and their inhibitors in parenchymal and non-parenchymal cells of rat liver: regulation by TNF-alpha and TGF-beta1. J. Hepatol., 1999, 30(1): 48-60, doi:10.1016/s0168-8278(99)80007-5.
  43. Solis-Herruzo J.A.; Brenner D.A.; Chojkier M. Tumor necrosis factor alpha inhibits collagen gene transcription and collagen synthesis in cultured human fibroblasts. J. Biol. Chem., 1988, 263(12): 5841-5845.
  44. Mauviel A.; Lapiere J.C.; Halcin C.; et al. Differential cytokine regulation of type I and type VII collagen gene expression in cultured human dermal fibroblasts. J. Biol. Chem., 1994, 269(1): 25-28.
  45. Verrecchia F.; Wagner E.F.; Mauviel A. Distinct involvement of the Jun-N-terminal kinase and NF-kappaB pathways in the repression of the human COL1A2 gene by TNF-alpha. EMBO Rep., 2002, 3(11): 1069-1074, doi:10.1093/embo-reports/kvf219.
  46. Hernandez-Munoz I.; de la Torre P.; Sánchez-Alcázar J. A.; et al. Tumor necrosis factor alpha inhibits collagen alpha 1(I) gene expression in rat hepatic stellate cells through a G protein. Gastroenterology, 1997, 113(2): 625-640, doi:10.1053/gast.1997.v113.pm9247485.
  47. Iraburu M.J.; Domínguez-Rosales J. A.; Fontana L.; et al. Tumor necrosis factor alpha down-regulates expression of the alpha1(I) collagen gene in rat hepatic stellate cells through a p20C/EBPbeta- and C/EBPdelta-dependent mechanism. Hepatology, 2000, 31(5): 1086-1093, doi:10.1053/he.2000.5981.
  48. Verrecchia F.; Tacheau C.; Wagner E.F.; et al. A central role for the JNK pathway in mediating the antagonistic activity of pro-inflammatory cytokines against transforming growth factor-beta-driven SMAD3/4-specific gene expression. J. Biol. Chem., 2003, 278(3): 1585-1593, doi:10.1074/jbc.M206927200.
  49. Verrecchia F.; Mauviel A. TGF-beta and TNF-alpha: antagonistic cytokines controlling type I collagen gene expression. Cell Signal, 2004, 16(8): 873-880, doi:10.1016/j.cellsig.2004.02.007 .
  50. Yamane K.; Ihn H.; Asano Y.; et al. Antagonistic effects of TNF-alpha on TGF-beta signaling through down-regulation of TGF-beta receptor type II in human dermal fibroblasts. J. Immunol., 2003, 171(7): 3855-3862, doi:10.4049/jimmunol.171.7.3855.
  51. Fickenscher H.; Hör S.; Küpers H.; et al. The interleukin-10 family of cytokines. Trends Immunol., 2002, 23(2): 89-96, doi:10.1016/s1471-4906(01)02149-4.
  52. Richmond J.; Tuzova M.; Cruikshank W.; et al. Regulation of cellular processes by interleukin-16 in homeostasis and cancer. J. Cell. Physiol., 2014, 229(2): 139-147, doi:10.1002/jcp.24441.
  53. Dinarello C. A. Overview of the IL-1 family in innate inflammation and acquired immunity. Immunol. Rev., 2018, 281(1): 8-27, doi:10.1111/imr.12621.
  54. Sims J.E.; Smith D.E. The IL-1 family: regulators of immunity. Nat. Rev. Immunol., 2010, 10(2): 89-102, doi:10.1038/nri2691.
  55. Borthwick L.A. The IL-1 cytokine family and its role in inflammation and fibrosis in the lung. Semin. Immunopathol., 2016, 38(4): 517-534, doi:10.1007/s00281-016-0559-z.
  56. Gasse P.; Mary C.; Guenon I.; et al. IL-1R1/MyD88 signaling and the inflammasome are essential in pulmonary inflammation and fibrosis in mice. J. Clin. Invest., 2007, 117(12): 3786-3799, doi:10.1172/JCI32285.
  57. Pauwels N.S.; Bracke K.R.; Dupont L.; et al. Role of IL-1alpha and the Nlrp3/caspase-1/IL-1beta axis in cigarette smoke-induced pulmonary inflammation and COPD. Eur. Respir. J., 2011, 38(5): 1019-1028, doi:10.1183/09031936.00158110.
  58. Hogg J.C.; Chu F.; Utokaparch S.; et al. The nature of small-airway obstruction in chronic obstructive pulmonary disease. N. Engl. J. Med., 2004, 350(26): 2645-2653, doi:10.1056/NEJMoa032158.
  59. Wilson M.S.; Madala S.K.; Ramalingam T.; et al. Bleomycin and IL-1beta-mediated pulmonary fibrosis is IL-17A dependent. J. Exp. Med., 2010, 207(3): 535-552, doi:10.1084/jem.20092121 .
  60. Kolb M.; Margetts P.J.; Anthony D.C.; et al. Transient expression of IL-1beta induces acute lung injury and chronic repair leading to pulmonary fibrosis. J. Clin. Invest., 2001, 107(12): 1529-1536, doi:10.1172/JCI12568.
  61. Guo J.; Gu N.; Chen J.; et al. Neutralization of interleukin-1 beta attenuates silica-induced lung inflammation and fibrosis in C57BL/6 mice. Arch. Toxicol., 2013, 87(11): 1963-1973, doi:10.1007/s00204-013-1063-z.
  62. Hill C.; Jones M.G.; Davies D.E.; et al. Epithelial-mesenchymal transition contributes to pulmonary fibrosis via aberrant epithelial/fibroblastic cross-talk. J. Lung Health Dis., 2019, 3(2): 31-35.
  63. Li R.; Ong S.L.; Tran L.M.; et al. Chronic IL-1beta-induced inflammation regulates epithelial-to-mesenchymal transition memory phenotypes via epigenetic modifications in non-small cell lung cancer. Sci. Rep., 2020, 10(1): 377, doi:10.1038/s41598-019-57285-y.
  64. Masola V.; Carraro A.; Granata S.; et al. In vitro effects of interleukin (IL)-1 beta inhibition on the epithelial-to-mesenchymal transition (EMT) of renal tubular and hepatic stellate cells. J. Transl. Med., 2019, 17(1): 12, doi:10.1186/s12967-019-1770-1.
  65. Gieling R.G.; Wallace K.; Han Y. P. Interleukin-1 participates in the progression from liver injury to fibrosis. Am. J. Physiol. Gastrointest. Liver Physiol., 2009, 296(6): G1324-1331, doi:10.1152/ajpgi.90564.2008.
  66. Hirano T.; Yasukawa K.; Harada H.; et al. Complementary DNA for a novel human interleukin (BSF-2) that induces B lymphocytes to produce immunoglobulin. Nature, 1986, 324(6092): 73-76, doi:10.1038/324073a0.
  67. Rodriguez-Bayona B.; Ramos-Amaya A.; Lopez-Blanco R.; et al. STAT-3 activation by differential cytokines is critical for human in vivo-generated plasma cell survival and Ig secretion. J. Immunol., 2013, 191(10): 4996-5004, doi:10.4049/jimmunol.1301559.
  68. Kopf M.; Baumann H.; Freer G.; et al. Impaired immune and acute-phase responses in interleukin-6-deficient mice. Nature, 1994, 368(6469): 339-342, doi:10.1038/368339a0.
  69. Ramsay A.J.; Husband A.J.; Ramshaw I.A.; et al. The role of interleukin-6 in mucosal IgA antibody responses in vivo. Science, 1994, 264(5158): 561-563, doi:10.1126/science.8160012.
  70. Tsukamoto H.; Fujieda K.; Hirayama M.; et al. Soluble IL6R Expressed by Myeloid Cells Reduces Tumor-Specific Th1 Differentiation and Drives Tumor Progression. Cancer Res., 2017, 77(9): 2279-2291, doi:10.1158/0008-5472.CAN-16-2446.
  71. Rincon M.; Anguita J.; Nakamura T.; et al. Interleukin (IL)-6 directs the differentiation of IL-4-producing CD4+ T cells. J. Exp. Med., 1997, 185(3): 461-469, doi:10.1084/jem.185.3.461.
  72. Johnson B.Z.; Stevenson A.W.; Prele C.M.; et al. The Role of IL-6 in Skin Fibrosis and Cutaneous Wound Healing. Biomedicines, 2020, 8(5): doi:10.3390/biomedicines8050101.
  73. Pedroza M.; Alcorn J.L.; Galas D.; et al. Interleukin-6 contributes to inflammation and remodeling in a model of adenosine mediated lung injury. PLoS One, 2011, 6(7): e22667, doi:10.1371/journal.pone.0022667.
  74. Sato S.; Hasegawa M.; Takehara K. Serum levels of interleukin-6 and interleukin-10 correlate with total skin thickness score in patients with systemic sclerosis. J. Dermatol. Sci., 2001, 27(2): 140-146, doi:10.1016/s0923-1811(01)00128-1.
  75. Migita K.; Abiru S.; Maeda Y.; et al. Serum levels of interleukin-6 and its soluble receptors in patients with hepatitis C virus infection. Hum. Immunol. 2006, 67(1-2): 27-32, doi:10.1016/j.humimm.2006.02.025.
  76. Zhong H.; Belardinelli L.; Maa T.; et al. Synergy between A2B adenosine receptors and hypoxia in activating human lung fibroblasts. Am. J. Respir. Cell Mol. Biol., 2005, 32(1): 2-8, doi:10.1165/rcmb.2004-0103OC.
  77. Dai Y.; Zhang W.; Wen J.; et al. A2B adenosine receptor-mediated induction of IL-6 promotes CKD. J. Am. Soc. Nephrol., 2011, 22(5): 890-901, doi:10.1681/ASN.2010080890.
  78. Melendez G.C.; McLarty J.L.; Levick S.P.; et al. Interleukin 6 mediates myocardial fibrosis, concentric hypertrophy, and diastolic dysfunction in rats. Hypertension, 2010, 56(2): 225-231, doi:10.1161/HYPERTENSIONAHA.109.148635.
  79. Tanaka T.; Narazaki M.; Kishimoto T. Therapeutic targeting of the interleukin-6 receptor. Annu. Rev. Pharmacol. Toxicol., 2012, 52, 199-219, doi:10.1146/annurev-pharmtox-010611-134715.
  80. Fielding C.A.; Jones G.W.; McLoughlin R.M. ; et al. Interleukin-6 signaling drives fibrosis in unresolved inflammation. Immunity, 2014, 40(1): 40-50, doi:10.1016/j.immuni.2013.10.022.
  81. O’Donoghue R.J.; Knight D.A.; Richards C.D.; et al. Genetic partitioning of interleukin-6 signalling in mice dissociates Stat3 from Smad3-mediated lung fibrosis. EMBO Mol. Med., 2012, 4(9): 939-951, doi:10.1002/emmm.201100604.
  82. Shima Y.; Kuwahara Y.; Murota H.; et al. The skin of patients with systemic sclerosis softened during the treatment with anti-IL-6 receptor antibody tocilizumab. Rheumatology (Oxford), 2010, 49(12): 2408-2412, doi:10.1093/rheumatology/keq275.
  83. Barata J.T.; Durum S.K.; Seddon B. Flip the coin: IL-7 and IL-7R in health and disease. Nat. Immunol., 2019, 20(12): 1584-1593, doi:10.1038/s41590-019-0479-x.
  84. Dubinett S.M.; Huang M.; Dhanani S.; et al. Down-regulation of macrophage transforming growth factor-beta messenger RNA expression by IL-7. J. Immunol., 1993, 151(12): 6670-6680.
  85. Huang M.; Sharma S.; Zhu L.X.; et al. IL-7 inhibits fibroblast TGF-beta production and signaling in pulmonary fibrosis. J. Clin. Invest., 2002, 109(7): 931-937, doi:10.1172/JCI14685.
  86. Hsieh P.F.; Liu S.F.; Lee T.C.; et al. The role of IL-7 in renal proximal tubule epithelial cells fibrosis. Mol. Immunol., 2012, 50(1-2): 74-82, doi:10.1016/j.molimm.2011.12.004.
  87. Jimenez-Sousa M.A.; Gómez-Moreno A.Z.; Pineda-Tenor D.; et al. The IL7RA rs6897932 polymorphism is associated with progression of liver fibrosis in patients with chronic hepatitis C: Repeated measurements design. PLoS One, 2018, 13(5): e0197115, doi:10.1371/journal.pone.0197115.
  88. Li B.; Li Y.; Li S.; et al. Circ_MTM1 knockdown inhibits the progression of HBV-related liver fibrosis via regulating IL7R expression through targeting miR-122-5p. Am. J. Transl. Res., 2022, 14(4): 2199-2211.
  89. Zlotnik A.; Yoshie O. Chemokines: a new classification system and their role in immunity. Immunity, 2000, 12(2): 121-127, doi:10.1016/s1074-7613(00)80165-x.
  90. Cyster J. G. Leukocyte migration: scent of the T zone. Curr. Biol., 2000, 10(1): R30-33, doi:10.1016/s0960-9822(99)00253-5.
  91. Bonecchi R.; Galliera E.; Borroni E.M.; et al. Chemokines and chemokine receptors: an overview. Front. Biosci., (Landmark Ed) 2009, 14(2): 540-551, doi:10.2741/3261.
  92. Baggiolini M.; Moser B.; Clark-Lewis I. Interleukin-8 and related chemotactic cytokines. The Giles Filley Lecture. Chest, 1994, 105(3): 95S-98S, doi:10.1378/chest.105.3_supplement.95s .
  93. Luster A.D.; Weinshank R.L.; Feinman R.; et al. Molecular and biochemical characterization of a novel gamma-interferon-inducible protein. J. Biol. Chem., 1988, 263(24): 12036-12043.
  94. Yang L.; Herrera J.; Gilbertsen A.; et al. IL-8 mediates idiopathic pulmonary fibrosis mesenchymal progenitor cell fibrogenicity. Am. J. Physiol. Lung Cell Mol. Physiol., 2018, 314(1): L127-L136, doi:10.1152/ajplung.00200.2017.
  95. Carre P.C.; et al. Increased expression of the interleukin-8 gene by alveolar macrophages in idiopathic pulmonary fibrosis. A potential mechanism for the recruitment and activation of neutrophils in lung fibrosis. J. Clin. Invest., 1991, 88(6): 1802-1810, doi:10.1172/JCI115501.
  96. Lee J.S.; Shin J.H.; Choi B. S. Serum levels of IL-8 and ICAM-1 as biomarkers for progressive massive fibrosis in coal workers' pneumoconiosis. J. Korean Med. Sci., 2015, 30(2): 140-144, doi:10.3346/jkms.2015.30.2.140.
  97. Zimmermann H.W.; Seidler S.; Gassler N.; et al. Interleukin-8 is activated in patients with chronic liver diseases and associated with hepatic macrophage accumulation in human liver fibrosis. PLoS One, 2011, 6(6): e21381, doi:10.1371/journal.pone.0021381.
  98. Dai Y.; Dean T.P.; Church M.K.; et al. Desensitisation of neutrophil responses by systemic interleukin 8 in cystic fibrosis. Thorax., 1994, 49(9): 867-871, doi:10.1136/thx.49.9.867 .
  99. Brysse A.; Mestdagt M.; Polette M.; et al. Regulation of CXCL8/IL-8 expression by zonula occludens-1 in human breast cancer cells. Mol. Cancer Res., 2012, 10(1): 121-132, doi:10.1158/1541-7786.MCR-11-0180.
  100. Zeremski M.; Petrovic L.M.; Chiriboga L.; et al. Intrahepatic levels of CXCR3-associated chemokines correlate with liver inflammation and fibrosis in chronic hepatitis C. Hepatology, 2008, 48(5): 1440-1450, doi:10.1002/hep.22500.
  101. Hintermann E.; Bayer M.; Pfeilschifter J.M.; et al. CXCL10 promotes liver fibrosis by prevention of NK cell mediated hepatic stellate cell inactivation. J. Autoimmun., 2010, 35(4): 424-435, doi:10.1016/j.jaut.2010.09.003.
  102. von Hundelshausen P.; Koenen R.R.; Sack M.; et al. Heterophilic interactions of platelet factor 4 and RANTES promote monocyte arrest on endothelium. Blood, 2005, 105(3): 924-930, doi:10.1182/blood-2004-06-2475.
  103. Strieter R.M.; Gomperts B.N.; Keane M.P. The role of CXC chemokines in pulmonary fibrosis. J. Clin. Invest., 2007, 117 (3): 549-556, doi:10.1172/JCI30562.
  104. Burdick M.D.; Murray L. A.; Keane M. P.; et al. CXCL11 attenuates bleomycin-induced pulmonary fibrosis via inhibition of vascular remodeling. Am. J. Respir. Crit. Care Med., 2005, 171(3): 261-268, doi:10.1164/rccm.200409-1164OC.
  105. Tager A.M.; Kradin R.L.; LaCamera P.; et al. Inhibition of pulmonary fibrosis by the chemokine IP-10/CXCL10. Am. J. Respir. Cell Mol. Biol., 2004, 31(4): 395-404, doi:10.1165/rcmb.2004-0175OC.
  106. Holt A.P.; Haughton E.L.; Lalor P.F.; et al. Liver myofibroblasts regulate infiltration and positioning of lymphocytes in human liver. Gastroenterology, 2009, 136(2): 705-714, doi:10.1053/j.gastro.2008.10.020.
  107. Bonacchi A.; Petrai I.; Defranco R.M.; et al. The chemokine CCL21 modulates lymphocyte recruitment and fibrosis in chronic hepatitis C. Gastroenterology, 2003, 125(4): 1060-1076, doi:10.1016/s0016-5085(03)01194-6.
  108. Nellen A.; Heinrichs D.; Berres M.L.; et al. Interference with oligomerization and glycosaminoglycan binding of the chemokine CCL5 improves experimental liver injury. PLoS One, 2012, 7(5): e36614, doi:10.1371/journal.pone.0036614.
  109. Capelli A.; Di Stefano A.; Gnemmi I.; et al. CCR5 expression and CC chemokine levels in idiopathic pulmonary fibrosis. Eur. Respir. J., 2005, 25(4): 701-707, doi:10.1183/09031936.05.00082604.
  110. Moore B.B.; Kolodsick J.E.; Thannickal V.J.; et al. CCR2-mediated recruitment of fibrocytes to the alveolar space after fibrotic injury. Am. J. Pathol., 2005, 166(3): 675-684, doi:10.1016/S0002-9440(10)62289-4.
  111. Osterholzer J.J.; Olszewski M.A.; Murdock B.J.; et al. Implicating exudate macrophages and Ly-6C(high) monocytes in CCR2-dependent lung fibrosis following gene-targeted alveolar injury. J. Immunol., 2013, 190(7): 3447-3457, doi:10.4049/jimmunol.1200604.
  112. Osterholzer J.J.; Christensen P.J.; Lama V.; et al. PAI-1 promotes the accumulation of exudate macrophages and worsens pulmonary fibrosis following type II alveolar epithelial cell injury. J. Pathol., 2012, 228(2): 170-180, doi:10.1002/path.3992.
  113. Raghu G.; Martinez F.J.; Brown K.K.; et al. CC-chemokine ligand 2 inhibition in idiopathic pulmonary fibrosis: a phase 2 trial of carlumab. Eur. Respir. J. 2015, 46(6): 1740-1750, doi:10.1183/13993003.01558-2014.
  114. Pierce E.M.; Carpenter K.; Jakubzick C.; et al. Idiopathic pulmonary fibrosis fibroblasts migrate and proliferate to CC chemokine ligand 21. Eur. Respir. J., 2007, 29(6): 1082-1093, doi:10.1183/09031936.00122806.
  115. Pierce E.M.; Carpenter K.; Jakubzick C.; et al. Therapeutic targeting of CC ligand 21 or CC chemokine receptor 7 abrogates pulmonary fibrosis induced by the adoptive transfer of human pulmonary fibroblasts to immunodeficient mice. Am. J. Pathol., 2007, 170(4): 1152-1164, doi:10.2353/ajpath.2007.060649.
  116. Pei G.; Yao Y.; Yang Q.; et al. Lymphangiogenesis in kidney and lymph node mediates renal inflammation and fibrosis. Sci. Adv., 2019, 5(6): eaaw5075, doi:10.1126/sciadv.aaw5075 .
  117. Nakayama Y.; Bromberg J.S. Lymphotoxin-beta receptor blockade induces inflammation and fibrosis in tolerized cardiac allografts. Am. J. Transplant, 2012, 12(9): 2322-2334, doi:10.1111/j.1600-6143.2012.04090.x.
  118. Wada T.; Sakai N.; Matsushima K.; et al. Fibrocytes: a new insight into kidney fibrosis. Kidney Int., 2007, 72(3): 269-273, doi:10.1038/sj.ki.5002325.
  119. Baggiolini M. Chemokines in pathology and medicine. J. Intern. Med., 2001, 250(2): 91-104, doi:10.1046/j.1365-2796.2001.00867.x.
  120. Abe R.; Donnelly S.C.; Peng T.; et al. Peripheral blood fibrocytes: differentiation pathway and migration to wound sites. J. Immunol., 2001, 166(12): 7556-7562, doi:10.4049/jimmunol.166.12.7556.
  121. Wynn T.A.; Vannella K. M. Macrophages in tissue repair, regeneration, and fibrosis. Immunity, 2016, 44(3): 450-462, doi:10.1016/j.immuni.2016.02.015.
  122. Xie T.; Wang Y.; Deng N.; et al. Single-Cell Deconvolution of Fibroblast Heterogeneity in Mouse Pulmonary Fibrosis. Cell Rep., 2018, 22(13): 3625-3640, doi:10.1016/j.celrep.2018.03.010.
  123. Peyser R.; MacDonnell S.; Gao Y.; et al. Defining the Activated Fibroblast Population in Lung Fibrosis Using Single-Cell Sequencing. Am. J. Respir. Cell Mol. Biol., 2019, 61(1): 74-85, doi:10.1165/rcmb.2018-0313OC.