Open Access

TGF‑β1: Gentlemanly orchestrator in idiopathic pulmonary fibrosis (Review)

  • Authors:
    • Zhimin Ye
    • Yongbin Hu
  • View Affiliations

  • Published online on: May 18, 2021     https://doi.org/10.3892/ijmm.2021.4965
  • Article Number: 132
  • Copyright: © Ye et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Idiopathic pulmonary fibrosis (IPF) is a worldwide disease characterized by the chronic and irreversible decline of lung function. Currently, there is no drug to successfully treat the disease except for lung transplantation. Numerous studies have been devoted to the study of the fibrotic process of IPF and findings showed that transforming growth factor‑β1 (TGF‑β1) plays a central role in the development of IPF. TGF‑β1 promotes the fibrotic process of IPF through various signaling pathways, including the Smad, MAPK, and ERK signaling pathways. There are intersections between these signaling pathways, which provide new targets for researchers to study new drugs. In addition, TGF‑β1 can affect the fibrosis process of IPF by affecting oxidative stress, epigenetics and other aspects. Most of the processes involved in TGF‑β1 promote IPF, but TGF‑β1 can also inhibit it. This review discusses the role of TGF‑β1 in IPF.

1. Introduction

Idiopathic pulmonary fibrosis (IPF) is a chronic, lethal and irreversible disease, which is characterized by fibroblast proliferation and excessive deposition of extracellular matrix in the lung (1,2). It was reported that the overall survival of the patients who were diagnosed with IPF was 3-5 years (3). The annual incidence of IPF is between 0.22 and 7.4 per 100,000 individuals in Europe and North America, but is lower in East Asia and South American (4). The incidence and prevalence of IPF increase with age and are higher in men (Tables I and II), which have been on the increase in recent years (1,5,6). Smoking, silica, and lampblack may be high risk factors for IPF (7). IPF can cause many symptoms such as dyspneal breathlessness, and chest discomfort, which does great harm to human and induces tremendous economic burden (8).

Table I

The association between IPF incidence with age.

Table I

The association between IPF incidence with age.

Studies<50 years50-59 years (%)60-69 years (%)>70 years (%)(Refs.)
Miyake2.9%14.754.927.5(117)
KimNA17.125.757.2(118)

Table II

The association between IPF incidence with sex.

Table II

The association between IPF incidence with sex.

StudiesMale (%)Female (%)(Refs.)
Baumgartner6040(119)
Miyake90.29.8(117)
García-Sancho Figueroa73.226.8(120)
Awadalla47.342.7(121)
Kim75.724.3(118)
Koo70.529.5(122)
Paolocci72.527.5(123)

At present, many studies have focused on the pathogenesis mechanisms, which mainly include the Smad, MAPK, and ERK signaling pathways (9). Of these mechanisms TGF-β1 is of critical significance (10). Researchers have conducted pharmacological studies on TGF-β1 in IPF, and some new drugs targeting TGF-β1-relevant signaling pathways have been developed. Such drugs include Nimbolide (11), Tanshinone IIA (Tan IIA) (12), methylsulfonylmethane (13) and Isoliquiritigenin (ISL) (14). However, since none of these medicines can successfully treat IPF, lung transplantation remains the primary method of treatment (15).

Both basic research and clinical research have proven that TGF-β1 plays an important role in the pathogenesis of IPF (Table III). However, no review systematically summarizing and discussing the role of TGF-β1 and relevant pathways in IPF has currently been published. The aim of the present review was to summarize the studies concerning the role of TGF-β1 in the development of IPF in recent decades (16) (Fig. 1). The findings may help researchers to grasp the latest progress in the pathogenesis of IPF related to TGF-β1 and to provide novel targets and a theoretical basis for the development of IPF clinical drugs.

Table III

Targeting molecules and signaling pathways initiated by TGF-β1 in IPF.

Table III

Targeting molecules and signaling pathways initiated by TGF-β1 in IPF.

Author, yearCell/tissue typeTarget genePotential signaling pathwaysBiological effect(Refs.)
Canonical TGF-β1/Smad signaling pathway
 Gu et al, 2007Human fetal lung fibroblastsSmad3 TGF-β1/Smad3/α-SMAPromoting myofibroblast differentiation(22)
 Ramirez et al, 2012Murine lung fibroblastsSmad3 TGF-β1/Smad3/PPARγPromoting pulmonary fibrogenesis(38)
 Li et al, 2016Human embryonic lung fibroblastsSmad3 TGF-β1/Smad3/CTGFPromoting pulmonary fibrogenesis(39)
 Huang et al, 2020Human lung fibroblastsSmad3 TGF-β1/Smad3/miR-424/Slit2Promoting myofibroblast differentiation(26)
 Zheng et al, 2017Mouse pulmonary fibroblastsSmad3 TGF-β1/Smad3/c-Jun/FstlPromoting fibrogenesis(40)
 Hecker et al, 2009Human fetal lung mesenchymal cellsSmad3 TGF-β1/Smad3/NOX4/H2O2Promoting myofibroblast differentiation(27)
 Guo et al, 2017Normal human lung fibroblastsSmad3 TGF-β1/Smad3/NOX4/ROSPromoting myofibroblast differentiation(29)
 Fierro-Fernández et al, 2015Human fetal lung fibroblastsSmad3 TGF-β1/Smad3/NOX4/ROS/miR-9-5p/NOX4Attenuating myofibroblast(28)
 Huang et al, 2020Mouse lung fibroblastsSmad3 TGF-β1/Smad3/FENDRRPromoting pulmonary fibrogenesis(41)
 Kadoya et al, 2019Human lung fibroblastsSmad3 TGF-β1/Smad3/ERK5Promoting pulmonary fibrogenesis(42)
 Cushing et al, 2011;Human fetal lung fibroblastSmad3 TGF-β1/Smad3/miR-29Promoting pulmonary fibrogenesis(43)
 Yang et al, 2013;(44)
 Xiao et al, 2012(45)
 Kang et al, 2007Murine lungSmad3TGF-β1/Smad3/SEMA 7APromoting pulmonary fibrogenesis(47)
 Selvarajah et al, 2019Primary human lung fibroblastsSmad3 TGF-β1/Smad3/mTORC1/4E-BP1/ATF4Promoting collagen biosynthesis(49)
 Jiang et al, 2018Human endothelial cellsSmad2/3/4 TGF-β1/Smad2/3/4/RELM-βAttenuating EndMT(36)
 Câmara and Jarai, 2010Human bronchial epithelial cellsSmad2/3TGF-β1/Smad2/3Promoting EMT(32)
 Li et al, 2015Human alveolar epithelial cell (A549)Smad2/3TGF-β1/Smad2/3Promoting EMT(34)
 Guan and Zhou, 2017Mice lung endothelial cellsSmad2/3 TGF-β1/Smad2/3/CXCR7/TGF-β1/Jag1-NotchAttenuating EndMT(35)
 Chen et al, 2020Human embryonic lung fibroblastsSmad2/3 TGF-β1/Smad2/3/miR-182-5p/Smad7Promoting pulmonary fibrogenesis(46)
 Kasai et al, 2005Human alveolar epithelial cell (A549)Smad2TGF-β1/Smad2Promoting EMT(33)
 Ji et al, 2014Human embryonic lung fibroblastsSmad2 TGF-β1/Smad2/RhoAPromoting myofibroblast differentiation(31)
PI3K relevant signaling pathway
 Shi et al, 2016Human alveolar epithelial cellsPI3K TGF-β1/PI3K/CTGFPromoting EMT and fibrogenesis(56)
 Wygrecka et al, 2012Human lung fibroblastsPI3K TGF-β1/PI3K/JNK/AKT/TFPromoting pulmonary fibrogenesis(57)
MAPK relevant signaling pathway
 JNK pathway
 Chen et al, 2013Human alveolar epithelialJNK-p38TGF-β1/JNK-p38Promoting EMT(63)
 Khalil et al, 2005(64)
 Jablonska et al, 2010Human lung fibroblastsJNK TGF-β1/JNK/Smad3/FXIIPromoting pulmonary fibrogenesis(62)
MAPK relevant signaling pathway
 Hashimoto et al, 2001Human lung fibroblastsJNKTGF-β1/JNKPromoting myofibroblast differentiation(65)
 Cui et al, 2014Human lung fibroblastsJNK TGF-β1/JNK/VEGF-DPromoting pulmonary fibrogenesis(66)
p38 signaling pathway
 Kulasekaran et al, 2009Human lung fibroblastsp38 TGF-β1/p38/PI3K/AKTAttenuates apoptosis(68)
 Deng et al, 2015Human lung fibroblastsp38 TGF-β1/p38/α-SMAPromoting pulmonary fibrogenesis(24)
 García-Alvarez et al, 2006Human lung fibroblastsp38 TGF-β1/p38/TIMP3/VEGFPromoting pulmonary fibrogenesis(69)
 Gu et al, 2014Human small airway epithelial cellsp38 TGF-β1//p38/CIPs/complementPromoting epithelial injury in IPF(70)
ERK signaling pathway
 Caraci et al, 2008Human lung fibroblastsERK1/2 TGF-β1/ERK1/2/GSK-3β/β-cateninPromoting myofibroblast differentiation(72)
 Ghatak et al, 2017Human lung fibroblastsERK TGFβ1/ERK/EGR1-AP-1/CD44v6Promoting myofibroblast differentiation(73)
Wnt/β-catenin relevant signaling pathway
 Lu et al, 2019Lung resident mesenchymal stem cellsβ-catenin TGF-β1/β-cateninPromoting myofibroblast differentiation(79)
 Zhou et al, 2012Human alveolar epithelial cellβ-catenin TGF-β1/β-catenin/CBPPromoting EMT(83)
 Wang et al, 2015Human embryonic lung fibroblasts Wnt3a/β-catenin TGF-β1/Wnt3a/β-catenin/miR-29Promoting cell proliferation(84)
Other signaling pathway
 Arsalane et al, 1997Human alveolar epithelialγ-GCS TGF-β/γ-GCS/ROSPromoting pulmonary fibrogenesis(101)
 Jardine et al, 2002cell (A549)(102)
 Boustani et al, 1997(103)
 Yu et al, 2020Mouse alveolar epithelial cellsTRB3 TGF-β/TRB3/Wnt/β-cateninPromoting EMT(97)
 Yamasaki et al, 2008Murine lung epithelial cellsTNF-α TGF-β/TNF-α/p21Attenuating fibrosis, and alveolar remodeling(88)
 Zhang et al, 2019Human fetal lung fibroblastsSIRT6 TGF-β1/SIRT6/TGF-β1/Smad2Attenuating myofibroblast differentiation(30)
 Kang et al, 2007Murine lungSEMA 7ATGF-β1/SEMA 7A/PI3K/PKB/AKTPromoting pulmonary fibrogenesis(47)
 Kolosionek et al, 2009Human alveolar epithelial cellsRho TGF-β1/Rho/PDE4Promoting EMT(37)
 Wei et al, 2019Human lung fibroblastsmiR-133a TGF-β1/miR-133a/CTGF-Col1a1Attenuating myofibroblast differentiation and pulmonary fibrosis(87)
 Lu et al, 2002Alveolar interstitial cellsIntegrin α8β1 TGF-β1-LAPT/integrin α8β1/ERKPromoting cell adhesion(75)
 Lim et al, 2014Fibroblast cell linesGal-1 TGF-β1/Gal-1/Smad2Promoting myofibroblast differentiation(25)
 Xiao et al, 2012Human alveolar epithelial cellFGF-2 TGFβ1/FGF-2/ERK1/2Promoting fibroblast proliferation and fibrogenesis(74)
 Noskovičová et al, 2018Human lung fibroblastsCDCP1TGF-β1/CDCP1Attenuating myofibroblast differentiation(85)
 Hagimoto et al, 2002Human bronchiolar epithelial cellscaspase-3 TGF-β/caspase-3/FasPromoting cell apoptosis and lung injury(96)
 Finlay et al, 2000Human lung fibroblastsbFGF TGF-β1/bFGF/ERK-AP1Promoting pulmonary fibrogenesis(71)
 Uhal et al, 2007Primary human lung fibroblastsANGTGF-β1/ANGPromoting development of IPF(86)
 Zhou et al, 2012Human alveolar epithelial cell (A549)Amphiregulin TGF-β1/amphiregulin/EGFR/TGF-β1Promoting pulmonary fibrosis(91)

2. TGF-β1-involved pathway in IPF

Canonical TGF-β1/Smad signaling pathway

The Smads family comprises three subfamilies, including five receptor-activated Smads (R-Smads), one common mediator Smad (Co-Smad) and two inhibitory Smads (I-Smads). Smad6 and Smad7 are the third type of Smads known as 'inhibitory Smads' or 'anti-Smads'. They are structurally different from other members of the family, and have been proven to be inhibitors of the Smad signaling pathway by disturbing the activation of R-Smads (17). Usually, TGF-β1 activates Smads through the transmembrane receptor serine/threonine kinase, successively regulating the transcription of target genes (18).

When TGF-β type I receptor kinase was activated by TGF-β1 signal, R-Smads (Smad2 and Smad3) were phosphorylated; of note is that Smad3 is more sensitive to TGF-β1 than Smad2 (19). Activated Smad2 and Smad3 form a complex, which combines with the Co-Smad (Smad4) and transfers into the nucleus to regulate the expression of target genes (20). The contribution of TGF-β1/Smad signaling pathway to IPF is mainly dependent on the following three processes: Myofibroblast differentiation, EMT/EndMT, and fibrogenesis.

TGF-β1-involved myofibroblast differentiation

TGF-β1 regulates the terminal differentiation of human lung fibroblasts (HLF) and promotes the synthesis of fibroblast extracellular matrix (21). Additionally, TGF-β1/Smad3 is the chief signaling pathway that regulates fibroblast differentiation (22,23). Transcription of α-smooth muscle actin (α-SMA), a target of myofibroblasts, was stimulated by TGF-β1 via a Smad3-, but not Smad2, dependent manner, resulting in the increased expression of α-SMA protein in human fetal lung fibroblasts (HFLF) (22). However, Deng et al (24) demonstrated that although Smad3 can be activated by TGF-β1 in HLF, the former did not affect the expression of collagen I or α-SMA. Treating fibroblasts with TGF-β1 could increase the expression of galectin-1 (Gal-1), which phosphorylated Smad2 and enhanced the nuclear retention of Smad2, promoting myofibroblast differentiation and accelerating fibrosis (25). TGF-β1 induced upregulation of miR-424 through the Smad3-denpendent signaling pathway, which inhibited the expression of Slit2, an inhibitory protein on TGF-β1 profibrogenic signaling. As a result, miR-424 acts as a positive feedback regulator of the TGF-β1 signaling pathway, promoting the myofibroblast differentiation of HLF (26). Interestingly, with the treatment of miR-424 inhibitor, Smad3 phosphorylation by TGF-β1 was reduced in HLFs, indicating miR-424 as a positive feedback regulator of TGF-β1/Smad3 synergistically (26). Previous findings demonstrated TGF-β1/Smad3-induced NADPH oxidase 4 (NOX4) mediated the production of H2O2, which was necessary for myofibroblast differentiation of lung mesenchymal cells, providing novel insight into the therapeutic targeting in IPF (27,28). In addition, TGF-β1 was reported to accelerate lung fibrosis by stimulating the production of ROS depending on NOX-4, and the produced ROS promoted the nuclear export of histone deacetylase 4 (HDAC4) and formation of α-SMA fiber in normal human lung fibroblasts (NHLFs) (29). Furthermore, following exposure to ROS, the expression of miR-9-5p, which inhibits the transformation from mesothelial cells to myofibroblast and reduces fibrogenesis via targeting TGF-β receptor type II (TGFBR2) and NOX4, was also upregulated, demonstrating that there may be a self-limiting homeostatic mechanism (28). Moreover, TGF-β1 can upregulate the level of Sirtuin 6 (SIRT6) protein in HFLF. The overexpression of SIRT6 inhibits TGF-β1-induced myofibroblast differentiation by suppressing TGF-β1/Smad2 and NF-κB signaling pathways (30). Inhibition of TGF-β1/Smad signal downregulated the expression of Rock1, RhoC and RhoA, demonstrating Rho kinase was a key mediator in myofibroblast differentiation induced by TGF-β1/Smad (31).

TGF-β1-involved EMT/EndMT

It was also reported that TGF-β1 stimulated primary human bronchial epithelial cells (HBEC) to the status of EMT in vitro mainly through Smad2/3-dependent mechanism (32). TGF-β1 induces alveolar epithelial cells (AEC) to EMT in a time- and concentration-dependent manner through Smad2 activation, and this event induced by TGF-β1 was not relevant to the ERK1/2 signaling pathway (33). In addition, TGF-β1/Smad2/3 signaling mediated the EMT induced by the high mobility group box 1 (HMGB1) released from injured lung in A549 cells (34). There was a negative feedback mechanism in the TGF-β1/Smad-involved pulmonary fibrosis. TGF-β1 upregulates the expression of CXCR7, a seven transmembrane G protein-coupled receptor in endothelial cells, in a Smad2/3-dependent pattern. Overexpression of CXCR7 impeded endothelial-to-mesenchymal transition (EndMT) and lung fibrosis induced by TGF-β1 through inhibition of the Jag1-Notch pathway (35). TGF-β1 stimulation significantly upregulated the expression of Resistin-like molecule-β (RELM-β) through the Smad2/3/4 pathway, which was reported to enhance TGF-β1-induced cell proliferation and EndMT (36). Rho kinase signal transduction activated by TGF-β1 in EMT was a positive regulator of phosphodiesterase 4 (PDE4), which promoted EMT of AEC (37).

TGF-β1-involved pulmonary fibrogenesis

The expression of peroxisome proliferator-activated receptor γ (PPAPγ), a negative regulator of TGF-β1-induced fibrosis, is mainly controlled by TGF-β1. Cells lacking Smad3 showed that the down-regulation effect of TGF-β1 on PPARγ was weakened, suggesting that TGF-β1 regulates the PPARγ in a Smad3-dependent manner (38). TGF-β1 exerted a pro-fibrosis effect by regulating the expression of connective tissue growth factor (CTGF), which was attributed to activation of the TGF-β1/Smad3 signaling pathway (39). Follistatin-like protein 1 (Fstl1) was a glycoprotein that plays a crucial role in promoting fibrogenesis. At the transcriptional and translational level, the expression of Fstl1 was upregulated by TGF-β1 via the Smad3-c-Jun signaling pathway in mouse pulmonary fibroblasts, suggesting that TGF-β1 may contribute to the IPF through a Smad3/c-Jun/Fstl1 axis (40). Huang et al (41) reported that TGF-β1/Smad3 signal inhibited the expression of long noncoding RNA fetal-lethal noncoding developmental regulatory RNA (FENDRR) which can reduce fibrogenesis and inhibit the process of pulmonary fibrosis. The TGF-β1/Smad3 signal upregulates the phosphorylation level of ERK5 and further leads to the contraction and migration of collagen gel induced by TGF-β1 (42). miR-29, a downstream target gene of TGF-β/Smad, was capable of inhibiting numerous fibrosis-related genes upregulated by TGF-β1 including CTGF, Smad3 and TGF-β1 (43). However, in fibroblasts, the expression of miR-29 was negatively regulated by TGF-β1/Smad3 signal (43-45). Similarly, Smad7, a negative regulator of TGF-β1, is suppressed by miR-182-5p which is induced by TGF-β1, resulting in the development of IPF (46). TGF-β1 activates Semaphorin (SEMA) 7A and its receptors through a Smad3-independent and Smad 2/3-independent mechanism, respectively, promoting pulmonary fibrosis (47) Activating transcription factor 4 (ATF4) was a pivotal transcriptional regulator for the metabolism of amino acid (48). TGF-β1/Smad3 signaling could increase the expression of the ATF4 through initiating the mechanistic target of rapamycin complex 1 (mTORC1) and its downstream translation initiation factor 4E binding protein 1 (4E-BP1), promoting collagen biosynthesis (49). This is one of the key pathways through which TGF-β1 stimulates collagen synthesis and IPF in HLF (50) (Fig. 2).

PI3K-relevant signaling pathway

A great number of studies indicated that phosphatidylinositol-3-kinase (PI3K) was involved in the pathomechanism of pulmonary fibrosis (51-54). It was also revealed that PI3K may play an important role in TGF-β1-relevant IPF.

As mentioned previously, CTGF is a functional intermediate product between TGF-β1 and ECM protein. CTGF derived from epithelial cells can activate fibroblasts and further accelerate the fibrosis process in an autocrine manner (55). It was reported that TGF-β1 may induce the EMT and synthesis of ECM in lung epithelial cells through the TGF-β1/PI3K/CTGF signaling pathway (56). Treating human lung epithelial cells with PI3K inhibitor can, not only inhibit the synthesis of CTGF and type I collagen, but also reverse the EMT and fibrogenesis stimulated by TGF-β1. TGF-β1 activated PI3K and protein kinase B (PKB)/AKT via SEMA 7A-dependent mechanisms. SEMA 7A plays a central role in the PI3K/PKB/AKT pathway, which contributes to TGF-β1-induced fibrosis and remodeling (47). TGF-β1 activated the PI3K/Jun-NH2-terminal kinase (JNK)/AKT and AP-1 synergistically to induce tissue factor (TF) expression in HLF, promoting the process of IPF (57) (Fig. 3).

MAPK-relevant signaling pathway

Mitogen-activated protein kinase (MAPK), mainly consisting of three distinctive cascades, the JNK, p38 and ERK pathways, is a well-known and crucial signaling pathway in multiple diseases (58-61). In the past decades, the role of MAPK cascade in the TGF-β1-relevant IPF has been gradually elucidated.

JNK pathway

Coagulation factor XII (FXII) is a serine protease relevant to fibrinolysis, it was demonstrated that the production of FXII induced by TGF-β1 in HLF was mediated with JNK/Smad3 signaling pathways (62). With the stimulation of TGF-β1, the expression of phosphorylated p38, phosphorylated JNK, and interstitial phenotypic markers including desmin, vimentin and a-SMA were significantly increased (63). TGF-β1-induced primary lung fibroblasts immediately release extracellular fibroblast growth factor-2 (FGF-2), p38 MAPK and JNK phosphorylation. As a result, lung fibroblasts proliferated in response to TGF-β1 indirectly (64). TGF-β1 can induce the phenotype of HLF to myofibroblasts in a dose- and time-dependent manner. Although the activity and phosphorylation of c-JNK, p38 MAPK, and ERK increased in response to TGF-β1, phenotypic modulation from HLF to myofibroblast was only regulated by c-JNK, suggesting that TGF-β1 induced HLF to myofibroblast via a c-JNK-mediated pathway (65). TGF-β1 was also reported to contribute to pulmonary fibrosis through down-regulation of the expression of vascular endothelial growth factor-D (VEGF-D) in HLF via the JNK signaling pathway, providing a speculative mechanism in the tissue remodeling of IPF (66). Notably, this protective effect of TGF-β1 on fibroblasts was independent on endothelin (ET)-1, which also endows fibroblast resistance to apoptosis. TGF-β1 could induce the deposition of extracellular matrix derived from tracheal basal cells, and the latter promoted EMT via a c-JNK1 involved pathway, which impairs the homeostasis of epithelial cell and the occurrence of IPF (67).

p38 signaling pathway

Notably, TGF-β1/MAPK signal not only contributed to the phenotypic modulation to myofibroblast, but also showed a protective effect on myofibroblasts. For example, TGF-β1 attenuates the apoptosis of fibroblast by inducing the production of a p38-dependent growth factor, which activates PI3K/AKT successively (68). It is noteworthy that activation of p38 MAPK induced by TGFβ1 was able to induce α-SMA but not collagen I in HLF (24). Tissue inhibitors of matrix metalloproteinases 3 (TIMP3), an effective angiogenesis inhibitor blocking the binding of VEGF to VEGF receptor 2, may be an important mediator of TGF-β1-mediated IPF (69). As TGF-β1 strongly upregulates the expression of TIMP3 in HLF, this process is relevant to p38 but not ERK pathway. The p38-mediated loss of epithelial complement inhibitory protein (CIP) caused by TGF-β1 led to the expansion of IPF epithelial damage, which in turn led to complement activation, further downregulated CIPs and induced the expression of TGF-β1 in feedback (70).

ERK signaling pathway

TGF-β1 regulates the autocrine of basic fibroblast growth factor (bFGF) in HLF, which activated the expression of ERK pathway and the induction of activator protein-1 (AP-1), accelerating pulmonary fibrogenesis (71). It was also reported that TGF-β1 induces GSK-3β inhibition and nuclear β-catenin translocation in HLF through ERK1/2 activation, which successively led to the production of γ-SMA and collagen (72). CD44v6 regulates the synthesis of COL1 and α-SMA in fibroblasts, and it is a potential activation target of TGF-β1 in lung fibroblasts (73). The induction of CD44v6 by TGF-β1 not only depends on ERK-induced early growth response-1 (EGR1) signaling, but also requires abundant AP-1 involvement, suggesting that there is a TGFβ1-ERK-EGR1/AP-1-CD44v6 axis (73). TGF-β1 can induce the expression of FGF-2 and its release from type II AEC. In addition, the FGF-2 signaling is responsible for the fibroblast proliferation and fibrotic activation through the ERK pathway (74). TGF-β1 binds non-covalently to the latency-related peptide (LAP) to form a complex. Consequently, the interaction of integrin α8β1 and LAPT-TGF-β1 complex induces FAK and ERK phosphorylation and promotes cell proliferation (75) (Fig. 4).

Wnt/β-catenin relevant signaling pathway

The Wnt/β-catenin pathway is the canonical Wnt signaling pathway, also known as the 'β-catenin-dependent' Wnt pathway. Wnt/β-catenin has been proven to play an important role in body development and growth, tumor, cardiovascular disease, musculoskeletal diseases, and also respiratory disease (76-78). In normal conditions, the glycogen synthase kinase-3β (GSK-3β) combines with the β-catenin, axis inhibition protein (Axin) and adenomatous polyposis coli (APC) to form a complex. When the Wnt/β-catenin was activated, the complex degraded, while β-catenin was not degraded and translocated into the nucleus (77).

Increasing evidence suggested that Wnt/β-catenin was involved in the TGF-β1-relevant IPF. TGF-β1 initiated the Wnt/β-catenin cascade via upregulating β-catenin and GSK-3β, promoting the fibrotic differentiation of lung resident mesenchymal stem cells (LR-MSCs) (79). In addition, it was found that, Wnt/β-catenin was required for the initiation of Smad2/3 induced by TGF-β1, suggesting that there may be a crosstalk between the two mechanisms in the myofibroblast differentiation (80). GSK-3 signaling decreases the phosphorylation of cAMP-response element binding protein (CREB) and attenuates its antagonism function on TGF-β/Smad signaling, promoting the myofibroblast differentiation in HLF (81). However, Liu et al suggested that in the transition of human normal skin fibroblast to myofibroblast induced by TGF-β1, Wnt/β-catenin played the role of negative regulator (82). TGF-β1 was capable of inducing the accumulation of β-catenin in the nuclear, facilitating EMT in a CREB-binding protein (CBP)-depending pattern in AEC (83). This revealed a potential cascade of TGF-β1/β-catenin/CBP. miR-29 negatively regulated the proliferation of IMR-90 cells induced by TGF-β1, but TGF-β1 inhibited the expression of all three members of the miR-29 family via Wnt3a/β-catenin pathway (84) (Fig. 5).

Feedback regulation mechanism

Feedback regulation is a crucial aspect in molecule cascades. Both positive and negative feedback are revealed in TGF-β1-involved pathway in IPF.

TGF-β1 strongly downregulated Cub domain-containing protein 1 (CDCP1), which promoted myofibroblast differentiation through inhibition of the potential negative feedback effect of CDCP1 expression on TGF-β1 stimulation (85). Similarly, TGF-β1 activated the autocrine mechanism of angiotensin (ANG) and angiotensinogen (AGT) peptide, which upregulated the expression of TGF-β1 to form an 'autocrine loop', promoting the development of IPF (86). miR-133a was reported to attenuate the differentiation of myofibroblasts by targeting many components of the TGF-β1 pro-fibrosis pathway, including α-SMA, CTGF and collagen. There seems to be a negative-feedback loop in the TGF-β1 pro-fibrogenesis pathway, because TGF-β1 upregulates the expression of miR-133a (87). Additionally, p21, a key regulator of apoptosis induced by TGF-β1 through tumor necrosis factor-α (TNF-α) signaling pathway, negatively regulates TNF-α expression induced by TGF-β1, participating in the fibrosis and alveolar remodeling induced by TGF-β1 (88). TNF-α could enhance the process of EMT induced by TGF-β1 in A549 cells through combination with TGF-β1 (89). However, TGF-β1 was also reported to inhibit the release of TNF-α from mast cells (90). TGF-β1 stimulates the EGFR ligand, amphiregulin, which regulates the classical and non-classical TGF-β1 signaling pathway through the activation of EGFR (91) (Fig. 6).

Other signaling pathways

Besides the signaling pathways discussed above, other molecules cascades were also revealed to be involved in the TGF-β1 relevant mechanisms of IPF.

The proliferation of fibroblasts is mainly mediated by platelet-derived growth factor (PDGF) isoforms, whose activity was potentially regulated by TGF-β1 (92). It was reported that TGF-β1 downregulated the expression of PDGF-α receptor (PDGF-Rα) transcript. However, TGF-β1 facilitated the transcription of PDGF-Rα in HLF, suggesting that TGF-β1 may contribute to IPF through a PDGF-Rα-involved complex network (92). It was reported that the IL-11 secreted by fibroblasts in the lungs of patients with IPF was significantly upregulated (93), and results demonstrated that TGF-β1 significantly increases IL-11 receptor expression in mouse fibroblasts (94), suggesting that IL-11 may be an important mediator of TGF-β1 involved IPF. Fas pathway-mediated apoptosis of lung epithelial cells is involved in the pathogenesis of pulmonary fibrosis (95). In lung tissues of patients with IPF, Fas- and FasL-induced apoptosis occurs in AEC and infiltrated inflammatory cells. TGF-β1 enhances the Fas-mediated pulmonary epithelial cell apoptosis through caspase-3, resulting in lung injury and pulmonary fibrosis (96). TGF-β1 induces the expression of exogenous tribbles homolog 3 (TRB3), which stimulates EMT and promotes the onset of IPF. In addition, TRB3 may participate in the regulation of EMT in MLE-12 cells induced by TGF-β1 through the Wnt/β-catenin signaling pathway (97). Insulin-like growth factor-1 (IGF-I) can co-operate with TGF-β1 to enhance the proliferation of lung fibroblast (98).

Currently, findings have shown that TGF-β1 may contribute to the development of IPF through epigenetic regulation. In fibroblasts from patients with IPF, TGF-β1 induces the upregulation of DNA methyltransferase (DNMT3a) and tetmethylcytosine dioxygenase 3 (TET3) (99). TGF-β1 inhibits Caveolin (Cav)-1 gene via histone modifications, contributing to fibroblast proliferation and apoptosis resistance (100).

TGF-β1 may promote IPF by reducing the production of antioxidant substance and inducing oxidative stress. TGF-β1 disturbs the homeostasis of the messenger RNA (mRNA) of the γ-glutamylcysteine synthase (γ-GCS) gene and downregulates the transcription of the gene, inducing the production of ROS in epithelial cells (101,102). It was also reported that TGF-β1 reduced the production of glutathione by downregulating precursor amino acid transport and synthesis rate (103). These results are consistent with previous reports of Guo et al (29) and Hecker et al (27) (Fig. 7).

3. Discussion

IPF is an irreversible lung disease, and there is no exact cause (1). In recent years, the incidence of IPF has gradually increased. There are numerous reasons for the increasing incidence of IPF. Firstly, IPF susceptibility is closely related to aging, which may lead to telomeres shortening and mitochondrial dysfunction. At present, the aging population is on the rise, resulting in an increasing incidence of IPF (104). Secondly, the development of medical technology has led to easy, convenient, and precise diagnosis of IPF, resulting in increasing incidence of IPF (105). Additionally, accumulating exposures to numerous risk factors such as smoking, occupational dust, drug stimulation, bacterial and virus infection, also play a role (106). The increased incidence of IPF has had a significant impact on the economic development of human society and the physical and mental health of people (4). The drugs currently studied can only delay the progression of the disease and maintain lung function but cannot cure the disease (107). In the pathogenesis of IPF, there are many mechanisms, of which TGF-β1 plays an important role (16). The IPF incidence of male was higher than that of female; this may be because of exposure to smoking, which is an acknowledged risk factor (106). Regarding the association between the IPF incidence and age, as mentioned previously, IPF is an age-associated disorder (1). Accumulated environmental exposures and cellular functional alteration with aging, for example, telomeres shortening, would facilitate the injury of lung (104). Although lung transplantation is the single most effective way to treat IPF, age is an influencing factor as older patients are less tolerant to surgery. According to the current study, age has become a limiting condition for lung transplantation in IPF patients, and the survival rate after lung transplantation in elderly patients older than 65 years is relatively low (108). Therefore, it is ofgreat significance to develop effective early diagnostic methods and innovative therapeutic strategies, such as applications of mesenchymal stem cells (109).

TGF-β1 activates Smads through the transmembrane receptor serine/threonine kinase, thereby continuously regulating the transcription of target genes (110), The TGF-β1/Smad signaling pathway functions in IPF mainly through the following three processes: Myofibroblast differentiation, EMT/EndMT and fibrogenesis (111). TGF-β1 activates PI3K and protein kinase B (PKB)/AKT through a SEMA 7A-dependent mechanism, thereby inducing the formation of EMT and ECM in lung epithelial cells (47). TGF-β1 mediates the production of FXII through the JNK/Smad3 signaling pathway (62). It also attenuates the apoptosis of fibroblasts by inducing the production of p38-dependent growth factor, which continuously activates PI3K/AKT. At the same time, it also initiates the Wnt/β-catenin cascade by upregulating β-catenin and GSK-3β (79). TGF-β1, not only regulates various mechanism pathways, but also affects IPF by regulating epigenetics, oxidative stress, and miRNA (112-115). Some research suggested that Smad3 activation has no effect on collagen I or α-SMA (24). However, Liu et al suggested that in the transition of human normal skin fibroblast to myofibroblast induced by TGF-β1, Wnt/β-catenin played a role of negative regulator, but had different functions in the lung, thereby promoting the hypothesis that Wnt/β-catenin is tissue-specific (82).

There are crosstalks and self-regulating loop in different pathways involved in TGF-β1-induced IPF. The Rho/Rock and Smad signaling pathways may cross talk in lung fibroblast differentiation (31). The Rho/Rock inhibitor downregulated Smad2 expression and the TGF-β/Smad inhibitor down-regulated RhoA, RhoC and Rock1 expression. There may be a complex network between the Rho/Rock pathway and Smad signaling in the process of lung fibroblasts to myofibroblasts induced by TGF-β1. TGF-β1 mainly promotes IPF, but there are also some self-regulating mechanisms that can induce miR-133a expression which acts as an antifibrosis regulator of TGF-β1, which induces IPF (87). Activation of the MAPK family is mediated by TGF-β1, which affects Smad signaling. ERK1/2 activation directly phosphorylates and activates p90RSK, which is a set of serine/threonine kinases that play a key role in the MAPK signaling pathway (116).

However, some mechanisms and pathways involved in TGF-β1 have not been clarified; thus, greater efforts to identify these should be made with regard to TGF-β1. Although some pathways have been proven, fewer drugs are actually converted into clinical applications. As for further studies on TGF-β1 in IPF, the focus should be on the intersection of various pathways, to facilitate the development of more effective drugs. At the same time, in addition to study on the various signal pathways involved in TGF-β1, an in-depth study of its role in epigenetics, and oxidative stress should also be conducted. After all, the purpose of research is to serve the clinic and solve the problem of clinical IPF treatment.

4. Conclusion

TGF-β1 plays a crucial role in the development of IPF as it regulates the pathomechanism of IPF through a number of signaling pathways, including Smad, MAPK, Wnt, and ERK pathways. The effect of TGF-β1 on IPF is one of stimulation. Nevertheless, there are some self-limiting mechanisms. Furthermore, some TGF-β1-relevant mechanisms in IPF remain to be elucidated.

Availability of data and materials

Not applicable.

Authors' contributions

ZY substantially contributed to the conception and design of the work and wrote the manuscript. YH revised the manuscript critically for important intellectual content. Both authors approved the final version of the manuscript.

Ethics approval and consent to participate

Not applicable.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

Acknowledgments

Not applicable.

Funding

This review was funded by the National Natural Science Foundation of China (grant no. 81673120).

References

1 

Martinez FJ, Collard HR, Pardo A, Raghu G, Richeldi L, Selman M, Swigris JJ, Taniguchi H and Wells AU: Idiopathic pulmonary fibrosis. Nat Rev Dis Primers. 3:170742017. View Article : Google Scholar : PubMed/NCBI

2 

George PM, Spagnolo P, Kreuter M, Altinisik G, Bonifazi M, Martinez FJ, Molyneaux PL, Renzoni EA, Richeldi L, Tomassetti S, et al: Progressive fibrosing interstitial lung disease: Clinical uncertainties, consensus recommendations, and research priorities. Lancet Respir Med. 8:925–934. 2020. View Article : Google Scholar : PubMed/NCBI

3 

Chanda D, Otoupalova E, Smith SR, Volckaert T, De Langhe SP and Thannickal VJ: Developmental pathways in the pathogenesis of lung fibrosis. Mol Aspects Med. 65:56–69. 2019. View Article : Google Scholar :

4 

Hutchinson J, Fogarty A, Hubbard R and McKeever T: Global incidence and mortality of idiopathic pulmonary fibrosis: A systematic review. Eur Respir J. 46:795–806. 2015. View Article : Google Scholar : PubMed/NCBI

5 

Nalysnyk L, Cid-Ruzafa J, Rotella P and Esser D: Incidence and prevalence of idiopathic pulmonary fibrosis: Review of the literature. Eur Respir Rev. 21:355–361. 2012. View Article : Google Scholar : PubMed/NCBI

6 

Park Y, Ahn C and Kim TH: Occupational and environmental risk factors of idiopathic pulmonary fibrosis: A systematic review and meta-analyses. Sci Rep. 11:43182021. View Article : Google Scholar : PubMed/NCBI

7 

Lv M, Liu Y, Ma S and Yu Z: Current advances in idiopathic pulmonary fibrosis: The pathogenesis, therapeutic strategies and candidate molecules. Future Med Chem. 11:2595–2620. 2019. View Article : Google Scholar : PubMed/NCBI

8 

Hadjicharalambous MR and Lindsay MA: Idiopathic pulmonary fibrosis: Pathogenesis and the emerging role of long non-coding RNAs. Int J Mol Sci. 21:5242020. View Article : Google Scholar :

9 

Hewlett JC, Kropski JA and Blackwell TS: Idiopathic pulmonary fibrosis: Epithelial-mesenchymal interactions and emerging therapeutic targets. Matrix Biol. 71-72:112–127. 2018. View Article : Google Scholar : PubMed/NCBI

10 

Hu HH, Chen DQ, Wang YN, Feng YL, Cao G, Vaziri ND and Zhao YY: New insights into TGF-β/Smad signaling in tissue fibrosis. Chem Biol Interact. 292:76–83. 2018. View Article : Google Scholar : PubMed/NCBI

11 

Prashanth Goud M, Bale S, Pulivendala G and Godugu C: Therapeutic effects of Nimbolide, an autophagy regulator, in ameliorating pulmonary fibrosis through attenuation of TGF-β1 driven epithelial-to-mesenchymal transition. Int Immunopharmacol. 75:1057552019. View Article : Google Scholar

12 

Feng F, Cheng P, Xu S, Li N, Wang H, Zhang Y and Wang W: Tanshinone IIA attenuates silica-induced pulmonary fibrosis via Nrf2-mediated inhibition of EMT and TGF-β1/Smad signaling. Chem Biol Interact. 319:1090242020. View Article : Google Scholar

13 

Moustafa EM, Ibrahim SI and Salem FAF: Methylsulfonylmethane inhibits lung fibrosis progression, inflammatory response, and epithelial-mesenchymal transition via the transforming growth factor-Beta 1/SMAD2/3 pathway in rats exposed to both γ-radiation and Bisphenol-A. Toxin Rev. 1–10. 2020.

14 

He J, Peng H, Wang M, Liu Y, Guo X, Wang B, Dai L, Cheng X, Meng Z, Yuan L, et al: Isoliquiritigenin inhibits TGF-β1-induced fibrogenesis through activating autophagy via PI3K/AKT/mTOR pathway in MRC-5 cells. Acta Biochim Biophys Sin (Shanghai). 52:810–820. 2020. View Article : Google Scholar

15 

Sgalla G, Iovene B, Calvello M, Ori M, Varone F and Richeldi L: Idiopathic pulmonary fibrosis: Pathogenesis and management. Respir Res. 19:322018. View Article : Google Scholar : PubMed/NCBI

16 

Kim KK, Sheppard D and Chapman HA: TGF-beta 1 signaling and tissue fibrosis. Cold Spring Harb Perspect Biol. 10:a0222932018. View Article : Google Scholar

17 

Werner F, Jain MK, Feinberg MW, Sibinga NE, Pellacani A, Wiesel P, Chin MT, Topper JN, Perrella MA and Lee ME: Transforming growth factor-beta 1 inhibition of macrophage activation is mediated via Smad3. J Biol Chem. 275:36653–36658. 2000. View Article : Google Scholar : PubMed/NCBI

18 

Flanders KC: Smad3 as a mediator of the fibrotic response. Int J Exp Pathol. 85:47–64. 2004. View Article : Google Scholar : PubMed/NCBI

19 

Zheng R, Xiong Q, Zuo B, Jiang S, Li F, Lei M, Deng C and Xiong Y: Using RNA interference to identify the different roles of SMAD2 and SMAD3 in NIH/3T3 fibroblast cells. Cell Biochem Funct. 26:548–556. 2008. View Article : Google Scholar : PubMed/NCBI

20 

Roberts AB, Piek E, Bottinger EP, Ashcroft G, Mitchell JB and Flanders KC: Is Smad3 a major player in signal transduction pathways leading to fibrogenesis? Chest. 120(1 Suppl): 43S–47S. 2001. View Article : Google Scholar : PubMed/NCBI

21 

Evans RA, Tian YC, Steadman R and Phillips AO: TGF-beta1-mediated fibroblast-myofibroblast terminal differentiation-the role of Smad proteins. Exp Cell Res. 282:90–100. 2003. View Article : Google Scholar : PubMed/NCBI

22 

Gu L, Zhu YJ, Yang X, Guo ZJ, Xu WB and Tian XL: Effect of TGF-beta/Smad signaling pathway on lung myofibroblast differentiation. Acta Pharmacol Sin. 28:382–391. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Kobayashi T, Liu X, Wen FQ, Kohyama T, Shen L, Wang XQ, Hashimoto M, Mao L, Togo S, Kawasaki S, et al: Smad3 mediates TGF-beta1-induced collagen gel contraction by human lung fibroblasts. Biochem Biophys Res Commun. 339:290–295. 2006. View Article : Google Scholar

24 

Deng X, Jin K, Li Y, Gu W, Liu M and Zhou L: Platelet-derived growth factor and transforming growth factor β1 Regulate ARDS-associated lung fibrosis through distinct signaling pathways. Cell Physiol Biochem. 36:937–946. 2015. View Article : Google Scholar

25 

Lim MJ, Ahn J, Yi JY, Kim MH, Son AR, Lee SL, Lim DS, Kim SS, Kang MA, Han Y and Song JY: Induction of galectin-1 by TGF-β1 accelerates fibrosis through enhancing nuclear retention of Smad2. Exp Cell Res. 326:125–135. 2014. View Article : Google Scholar : PubMed/NCBI

26 

Huang Y, Xie Y, Abel PW, Wei P, Plowman J, Toews ML, Strah H, Siddique A, Bailey KL and Tu Y: TGF-β1-induced miR-424 promotes pulmonary myofibroblast differentiation by targeting Slit2 protein expression. Biochem Pharmacol. 180:1141722020. View Article : Google Scholar

27 

Hecker L, Vittal R, Jones T, Jagirdar R, Luckhardt TR, Horowitz JC, Pennathur S, Martinez FJ and Thannickal VJ: NADPH oxidase-4 mediates myofibroblast activation and fibrogenic responses to lung injury. Nat Med. 15:1077–1081. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Fierro-Fernández M, Busnadiego Ó, Sandoval P, Espinosa-Díez C, Blanco-Ruiz E, Rodríguez M, Pian H, Ramos R, López-Cabrera M, García-Bermejo ML and Lamas S: miR-9-5p suppresses pro-fibrogenic transformation of fibroblasts and prevents organ fibrosis by targeting NOX4 and TGFBR2. EMBO Rep. 16:1358–1377. 2015. View Article : Google Scholar : PubMed/NCBI

29 

Guo W, Saito S, Sanchez CG, Zhuang Y, Gongora Rosero RE, Shan B, Luo F and Lasky JA: TGF-β1 stimulates HDAC4 nucleus-to-cytoplasm translocation and NADPH oxidase 4-derived reactive oxygen species in normal human lung fibroblasts. Am J Physiol Lung Cell Mol Physiol. 312:L936–L944. 2017. View Article : Google Scholar

30 

Zhang Q, Tu W, Tian K, Han L, Wang Q, Chen P and Zhou X: Sirtuin 6 inhibits myofibroblast differentiation via inactivating transforming growth factor-β1/Smad2 and nuclear factor-κB signaling pathways in human fetal lung fibroblasts. J Cell Biochem. 120:93–104. 2019. View Article : Google Scholar

31 

Ji H, Tang H, Lin H, Mao J, Gao L, Liu J and Wu T: Rho/Rock cross-talks with transforming growth factor-β/Smad pathway participates in lung fibroblast-myofibroblast differentiation. Biomed Rep. 2:787–792. 2014. View Article : Google Scholar : PubMed/NCBI

32 

Câmara J and Jarai G: Epithelial-mesenchymal transition in primary human bronchial epithelial cells is Smad-dependent and enhanced by fibronectin and TNF-alpha. Fibrogenesis Tissue Repair. 3:22010. View Article : Google Scholar : PubMed/NCBI

33 

Kasai H, Allen JT, Mason RM, Kamimura T and Zhang Z: TGF-beta1 induces human alveolar epithelial to mesenchymal cell transition (EMT). Respir Res. 6:562005. View Article : Google Scholar : PubMed/NCBI

34 

Li LC, Li DL, Xu L, Mo XT, Cui WH, Zhao P, Zhou WC, Gao J and Li J: High-mobility group box 1 mediates epithelial-to-mesenchymal transition in pulmonary fibrosis involving transforming growth factor-β1/Smad2/3 signaling. J Pharmacol Exp Ther. 354:302–309. 2015. View Article : Google Scholar : PubMed/NCBI

35 

Guan S and Zhou J: CXCR7 attenuates the TGF-β-induced endothelial-to-mesenchymal transition and pulmonary fibrosis. Mol Biosyst. 13:2116–2124. 2017. View Article : Google Scholar : PubMed/NCBI

36 

Jiang Y, Zhou X, Hu R and Dai A: TGF-β1-induced SMAD2/3/4 activation promotes RELM-β transcription to modulate the endothelium-mesenchymal transition in human endothelial cells. Int J Biochem Cell Biol. 105:52–60. 2018. View Article : Google Scholar : PubMed/NCBI

37 

Kolosionek E, Savai R, Ghofrani HA, Weissmann N, Guenther A, Grimminger F, Seeger W, Banat GA, Schermuly RT and Pullamsetti SS: Expression and activity of phosphodiesterase isoforms during epithelial mesenchymal transition: The role of phosphodiesterase 4. Mol Biol Cell. 20:4751–4765. 2009. View Article : Google Scholar : PubMed/NCBI

38 

Ramirez A, Ballard EN and Roman J: TGFβ1 controls PPARγ expression, transcriptional potential, and activity, in part, through Smad3 signaling in murine lung fibroblasts. PPAR Res. 2012:3758762012. View Article : Google Scholar

39 

Li HH, Cai Q, Wang YP, Liu HR and Huang M: The role of transforming growth factor-β1/connective tissue growth factor signaling pathway in paraquat-induced pulmonary fibrosis. Zhonghua Lao Dong Wei Sheng Zhi Ye Bing Za Zhi. 34:484–488. 2016.In Chinese. PubMed/NCBI

40 

Zheng X, Qi C, Zhang S, Fang Y and Ning W: TGF-β1 induces Fstl1 via the Smad3-c-Jun pathway in lung fibroblasts. Am J Physiol Lung Cell Mol Physiol. 313:L240–L251. 2017. View Article : Google Scholar

41 

Huang C, Liang Y, Zeng X, Yang X, Xu D, Gou X, Sathiaseelan R, Senavirathna LK, Wang P and Liu L: Long noncoding RNA FENDRR exhibits antifibrotic activity in pulmonary fibrosis. Am J Respir Cell Mol Biol. 62:440–453. 2020. View Article : Google Scholar :

42 

Kadoya K, Togo S, Tulafu M, Namba Y, Iwai M, Watanabe J, Okabe T, Jin J, Kodama Y, Kitamura H, et al: Specific features of fibrotic lung fibroblasts highly sensitive to fibrotic processes mediated via TGF-β-ERK5 interaction. Cell Physiol Biochem. 52:822–837. 2019. View Article : Google Scholar

43 

Cushing L, Kuang PP, Qian J, Shao F, Wu J, Little F, Thannickal VJ, Cardoso WV and Lü J: miR-29 is a major regulator of genes associated with pulmonary fibrosis. Am J Respir Cell Mol Biol. 45:287–294. 2011. View Article : Google Scholar :

44 

Yang T, Liang Y, Lin Q, Liu J, Luo F, Li X, Zhou H, Zhuang S and Zhang H: miR-29 mediates TGFβ1-induced extracellular matrix synthesis through activation of PI3K-AKT pathway in human lung fibroblasts. J Cell Biochem. 114:1336–1342. 2013. View Article : Google Scholar

45 

Xiao J, Meng XM, Huang XR, Chung AC, Feng YL, Hui DS, Yu CM, Sung JJ and Lan HY: miR-29 inhibits bleomycin-induced pulmonary fibrosis in mice. Mol Ther. 20:1251–1260. 2012. View Article : Google Scholar : PubMed/NCBI

46 

Chen Y, Zhang Q, Zhou Y, Yang Z and Tan M: Inhibition of miR-182-5p attenuates pulmonary fibrosis via TGF-β/Smad pathway. Hum Exp Toxicol. 39:683–695. 2020. View Article : Google Scholar

47 

Kang HR, Lee CG, Homer RJ and Elias JA: Semaphorin 7A plays a critical role in TGF-beta1-induced pulmonary fibrosis. J Exp Med. 204:1083–1093. 2007. View Article : Google Scholar : PubMed/NCBI

48 

Mukherjee D, Bercz LS, Torok MA and Mace TA: Regulation of cellular immunity by activating transcription factor 4. Immunol Lett. 228:24–34. 2020. View Article : Google Scholar : PubMed/NCBI

49 

Selvarajah B, Azuelos I, Platé M, Guillotin D, Forty EJ, Contento G, Woodcock HV, Redding M, Taylor A, Brunori G, et al: mTORC1 amplifies the ATF4-dependent de novo serine-glycine pathway to supply glycine during TGF-β1-induced collagen biosynthesis. Sci Signal. 12:eaav30482019. View Article : Google Scholar

50 

Woodcock HV, Eley JD, Guillotin D, Platé M, Nanthakumar CB, Martufi M, Peace S, Joberty G, Poeckel D, Good RB, et al: The mTORC1/4E-BP1 axis represents a critical signaling node during fibrogenesis. Nat Commun. 10:62019. View Article : Google Scholar : PubMed/NCBI

51 

Cong LH, Li T, Wang H, Wu YN, Wang SP, Zhao YY, Zhang GQ and Duan J: IL-17A-producing T cells exacerbate fine particulate matter-induced lung inflammation and fibrosis by inhibiting PI3K/Akt/mTOR-mediated autophagy. J Cell Mol Med. 24:8532–8544. 2020. View Article : Google Scholar : PubMed/NCBI

52 

Fang L, Chen H, Kong R and Que J: Endogenous tryptophan metabolite 5-methoxytryptophan inhibits pulmonary fibrosis by downregulating the TGF-β/SMAD3 and PI3K/AKT signaling pathway. Life Sci. 260:1183992020. View Article : Google Scholar

53 

Hettiarachchi SU, Li YH, Roy J, Zhang F, Puchulu-Campanella E, Lindeman SD, Srinivasarao M, Tsoyi K, Liang X, Ayaub EA, et al: Targeted inhibition of PI3 kinase/mTOR specifically in fibrotic lung fibroblasts suppresses pulmonary fibrosis in experimental models. Sci Transl Med. 12:eaay37242020. View Article : Google Scholar : PubMed/NCBI

54 

Hu X, Xu Q, Wan H, Hu Y, Xing S, Yang H, Gao Y and He Z: PI3K-Akt-mTOR/PFKFB3 pathway mediated lung fibroblast aerobic glycolysis and collagen synthesis in lipopolysaccharide-induced pulmonary fibrosis. Lab Invest. 100:801–811. 2020. View Article : Google Scholar : PubMed/NCBI

55 

Graves DT and Milovanova TN: Mucosal immunity and the FOXO1 transcription factors. Front Immunol. 10:25302019. View Article : Google Scholar : PubMed/NCBI

56 

Shi L, Dong N, Fang X and Wang X: Regulatory mechanisms of TGF-β1-induced fibrogenesis of human alveolar epithelial cells. J Cell Mol Med. 20:2183–2193. 2016. View Article : Google Scholar : PubMed/NCBI

57 

Wygrecka M, Zakrzewicz D, Taborski B, Didiasova M, Kwapiszewska G, Preissner KT and Markart P: TGF-β1 induces tissue factor expression in human lung fibroblasts in a PI3K/JNK/Akt-dependent and AP-1-dependent manner. Am J Respir Cell Mol Biol. 47:614–627. 2012. View Article : Google Scholar : PubMed/NCBI

58 

Bengal E, Aviram S and Hayek T: p38 MAPK in glucose metabolism of skeletal muscle: Beneficial or harmful? Int J Mol Sci. 21:64802020. View Article : Google Scholar :

59 

Guo YJ, Pan WW, Liu SB, Shen ZF, Xu Y and Hu LL: ERK/MAPK signalling pathway and tumorigenesis. Exp Ther Med. 19:1997–2007. 2020.PubMed/NCBI

60 

He X and Wang C, Wang H, Li L and Wang C: The function of MAPK cascades in response to various stresses in horticultural plants. Front Plant Sci. 11:9522020. View Article : Google Scholar : PubMed/NCBI

61 

Magnelli L, Schiavone N, Staderini F, Biagioni A and Papucci L: MAP kinases pathways in gastric cancer. Int J Mol Sci. 21:28932020. View Article : Google Scholar :

62 

Jablonska E, Markart P, Zakrzewicz D, Preissner KT and Wygrecka M: Transforming growth factor-β1 induces expression of human coagulation factor XII via Smad3 and JNK signaling pathways in human lung fibroblasts. J Biol Chem. 285:11638–11651. 2010. View Article : Google Scholar : PubMed/NCBI

63 

Chen HH, Zhou XL, Shi YL and Yang J: Roles of p38 MAPK and JNK in TGF-β1-induced human alveolar epithelial to mesenchymal transition. Arch Med Res. 44:93–98. 2013. View Article : Google Scholar : PubMed/NCBI

64 

Khalil N, Xu YD, O'Connor R and Duronio V: Proliferation of pulmonary interstitial fibroblasts is mediated by transforming growth factor-beta1-induced release of extracellular fibroblast growth factor-2 and phosphorylation of p38 MAPK and JNK. J Biol Chem. 280:43000–43009. 2005. View Article : Google Scholar

65 

Hashimoto S, Gon Y, Takeshita I, Matsumoto K, Maruoka S and Horie T: Transforming growth factor-beta1 induces phenotypic modulation of human lung fibroblasts to myofibroblast through a c-Jun-NH2-terminal kinase-dependent pathway. Am J Respir Crit Care Med. 163:152–157. 2001. View Article : Google Scholar : PubMed/NCBI

66 

Cui Y, Osorio JC, Risquez C, Wang H, Shi Y, Gochuico BR, Morse D, Rosas IO and El-Chemaly S: Transforming growth factor-β1 downregulates vascular endothelial growth factor-D expression in human lung fibroblasts via the Jun NH2-terminal kinase signaling pathway. Mol Med. 20:120–134. 2014. View Article : Google Scholar : PubMed/NCBI

67 

van der Velden JL, Wagner DE, Lahue KG, Abdalla ST, Lam YW, Weiss DJ and Janssen-Heininger YMW: TGF-β1-induced deposition of provisional extracellular matrix by tracheal basal cells promotes epithelial-to-mesenchymal transition in a c-Jun NH2-terminal kinase-1-dependent manner. Am J Physiol Lung Cell Mol Physiol. 314:L984–L997. 2018. View Article : Google Scholar

68 

Kulasekaran P, Scavone CA, Rogers DS, Arenberg DA, Thannickal VJ and Horowitz JC: Endothelin-1 and transforming growth factor-beta1 independently induce fibroblast resistance to apoptosis via AKT activation. Am J Respir Cell Mol Biol. 41:484–493. 2009. View Article : Google Scholar : PubMed/NCBI

69 

García-Alvarez J, Ramirez R, Checa M, Nuttall RK, Sampieri CL, Edwards DR, Selman M and Pardo A: Tissue inhibitor of metalloproteinase-3 is up-regulated by transforming growth factor-beta1 in vitro and expressed in fibroblastic foci in vivo in idiopathic pulmonary fibrosis. Exp Lung Res. 32:201–214. 2006. View Article : Google Scholar : PubMed/NCBI

70 

Gu H, Mickler EA, Cummings OW, Sandusky GE, Weber DJ, Gracon A, Woodruff T, Wilkes DS and Vittal R: Crosstalk between TGF-β1 and complement activation augments epithelial injury in pulmonary fibrosis. FASEB J. 28:4223–4234. 2014. View Article : Google Scholar : PubMed/NCBI

71 

Finlay GA, Thannickal VJ, Fanburg BL and Paulson KE: Transforming growth factor-beta 1-induced activation of the ERK pathway/activator protein-1 in human lung fibroblasts requires the autocrine induction of basic fibroblast growth factor. J Biol Chem. 275:27650–27656. 2000. View Article : Google Scholar : PubMed/NCBI

72 

Caraci F, Gili E, Calafiore M, Failla M, La Rosa C, Crimi N, Sortino MA, Nicoletti F, Copani A and Vancheri C: TGF-beta1 targets the GSK-3beta/beta-catenin pathway via ERK activation in the transition of human lung fibroblasts into myofibroblasts. Pharmacol Res. 57:274–282. 2008. View Article : Google Scholar : PubMed/NCBI

73 

Ghatak S, Markwald RR, Hascall VC, Dowling W, Lottes RG, Baatz JE, Beeson G, Beeson CC, Perrella MA, Thannickal VJ and Misra S: Transforming growth factor β1 (TGFβ1) regulates CD44V6 expression and activity through extracellular signal-regulated kinase (ERK)-induced EGR1 in pulmonary fibrogenic fibroblasts. J Biol Chem. 292:10465–10489. 2017. View Article : Google Scholar : PubMed/NCBI

74 

Xiao L, Du Y, Shen Y, He Y, Zhao H and Li Z: TGF-beta 1 induced fibroblast proliferation is mediated by the FGF-2/ERK pathway. Front Biosci (Landmark Ed). 17:2667–2674. 2012. View Article : Google Scholar

75 

Lu M, Munger JS, Steadele M, Busald C, Tellier M and Schnapp LM: Integrin alpha8beta1 mediates adhesion to LAP-TGFbeta1. J Cell Sci. 115:4641–4648. 2002. View Article : Google Scholar : PubMed/NCBI

76 

Bugter JM, Fenderico N and Maurice MM: Mutations and mechanisms of WNT pathway tumour suppressors in cancer. Nat Rev Cancer. 21:5–21. 2021. View Article : Google Scholar

77 

Rapetti-Mauss R, Berenguier C, Allegrini B and Soriani O: Interplay between ion channels and the Wnt/β-catenin signaling pathway in cancers. Front Pharmacol. 11:5250202020. View Article : Google Scholar

78 

Söderholm S and Cantù C: The WNT/β-catenin dependent transcription: A tissue-specific business. Wiley Interdiscip Rev Syst Biol Med. Oct 21–2020.Epub ahead of print. View Article : Google Scholar

79 

Lu Y, Zhang T, Shan S, Wang S, Bian W, Ren T and Yang D: MiR-124 regulates transforming growth factor-β1 induced differentiation of lung resident mesenchymal stem cells to myofibroblast by repressing Wnt/β-catenin signaling. Dev Biol. 449:115–121. 2019. View Article : Google Scholar : PubMed/NCBI

80 

Xu L, Cui WH, Zhou WC, Li DL, Li LC, Zhao P, Mo XT, Zhang Z and Gao J: Activation of Wnt/β-catenin signalling is required for TGF-β/Smad2/3 signalling during myofibroblast proliferation. J Cell Mol Med. 21:1545–1554. 2017. View Article : Google Scholar : PubMed/NCBI

81 

Baarsma HA, Engelbertink LH, van Hees LJ, Menzen MH, Meurs H, Timens W, Postma DS, Kerstjens HA and Gosens R: Glycogen synthase kinase-3 (GSK-3) regulates TGF-β1-induced differentiation of pulmonary fibroblasts. Br J Pharmacol. 169:590–603. 2013. View Article : Google Scholar : PubMed/NCBI

82 

Liu J, Wang Y, Pan Q, Su Y, Zhang Z, Han J, Zhu X, Tang C and Hu D: Wnt/β-catenin pathway forms a negative feedback loop during TGF-β1 induced human normal skin fibroblast-to-myofibroblast transition. J Dermatol Sci. 65:38–49. 2012. View Article : Google Scholar

83 

Zhou B, Liu Y, Kahn M, Ann DK, Han A, Wang H, Nguyen C, Flodby P, Zhong Q, Krishnaveni MS, et al: Interactions between β-catenin and transforming growth factor-β signaling pathways mediate epithelial-mesenchymal transition and are dependent on the transcriptional co-activator cAMP-response element-binding protein (CREB)-binding protein (CBP). J Biol Chem. 287:7026–7038. 2012. View Article : Google Scholar : PubMed/NCBI

84 

Wang Y, Liu J, Chen J, Feng T and Guo Q: MiR-29 mediates TGFβ 1-induced extracellular matrix synthesis through activation of Wnt/β-catenin pathway in human pulmonary fibroblasts. Technol Health Care. 23(Suppl 1): S119–S125. 2015. View Article : Google Scholar

85 

Noskovičová N, Heinzelmann K, Burgstaller G, Behr J and Eickelberg O: Cub domain-containing protein 1 negatively regulates TGF-β signaling and myofibroblast differentiation. Am J Physiol Lung Cell Mol Physiol. 314:L695–L707. 2018. View Article : Google Scholar

86 

Uhal BD, Kim JK, Li X and Molina-Molina M: Angiotensin-TGF-beta 1 crosstalk in human idiopathic pulmonary fibrosis: Autocrine mechanisms in myofibroblasts and macrophages. Curr Pharm Des. 13:1247–1256. 2007. View Article : Google Scholar : PubMed/NCBI

87 

Wei P, Xie Y, Abel PW, Huang Y, Ma Q, Li L, Hao J, Wolff DW, Wei T and Tu Y: Transforming growth factor (TGF)-β1-induced miR-133a inhibits myofibroblast differentiation and pulmonary fibrosis. Cell Death Dis. 10:6702019. View Article : Google Scholar

88 

Yamasaki M, Kang HR, Homer RJ, Chapoval SP, Cho SJ, Lee BJ, Elias JA and Lee CG: P21 regulates TGF-beta1-induced pulmonary responses via a TNF-alpha-signaling pathway. Am J Respir Cell Mol Biol. 38:346–353. 2008. View Article : Google Scholar

89 

Yamauchi Y, Kohyama T, Takizawa H, Kamitani S, Desaki M, Takami K, Kawasaki S, Kato J and Nagase T: Tumor necrosis factor-alpha enhances both epithelial-mesenchymal transition and cell contraction induced in A549 human alveolar epithelial cells by transforming growth factor-beta 1. Exp Lung Res. 36:12–24. 2010. View Article : Google Scholar : PubMed/NCBI

90 

Bissonnette EY, Enciso JA and Befus AD: TGF-beta1 inhibits the release of histamine and tumor necrosis factor-alpha from mast cells through an autocrine pathway. Am J Respir Cell Mol Biol. 16:275–282. 1997. View Article : Google Scholar : PubMed/NCBI

91 

Zhou Y, Lee JY, Lee CM, Cho WK, Kang MJ, Koff JL, Yoon PO, Chae J, Park HO, Elias JA and Lee CG: Amphiregulin, an epidermal growth factor receptor ligand, plays an essential role in the pathogenesis of transforming growth factor-β-induced pulmonary fibrosis. J Biol Chem. 287:41991–42000. 2012. View Article : Google Scholar : PubMed/NCBI

92 

Bonner JC, Badgett A, Lindroos PM and Osornio-Vargas AR: Transforming growth factor beta 1 downregulates the platelet-derived growth factor alpha-receptor subtype on human lung fibroblasts in vitro. Am J Respir Cell Mol Biol. 13:496–505. 1995. View Article : Google Scholar : PubMed/NCBI

93 

Ng B, Dong J, D'Agostino G, Viswanathan S, Widjaja AA, Lim WW, Ko NSJ, Tan J, Chothani SP, Huang B, et al: Interleukin-11 is a therapeutic target in idiopathic pulmonary fibrosis. Sci Transl Med. 11:eaaw12372019. View Article : Google Scholar : PubMed/NCBI

94 

Zhang L, Zhang J, Zhang Y and Yi Z: Expression of interleukin-11 and its receptor in lung of mice with idiopathic pulmonary fibrosis. Zhong Nan Da Xue Xue Bao Yi Xue Ban. 43:1083–1088. 2018.In Chinese. PubMed/NCBI

95 

Otsuki T, Hayashi H, Nishimura Y, Hyodo F, Maeda M, Kumagai N, Miura Y, Kusaka M and Uragami K: Dysregulation of autoimmunity caused by silica exposure and alteration of Fas-mediated apoptosis in T lymphocytes derived from silicosis patients. Int J Immunopathol Pharmacol. 24(Suppl): 11S–16S. 2011.PubMed/NCBI

96 

Hagimoto N, Kuwano K, Inoshima I, Yoshimi M, Nakamura N, Fujita M, Maeyama T and Hara N: TGF-beta 1 as an enhancer of Fas-mediated apoptosis of lung epithelial cells. J Immunol. 168:6470–6478. 2002. View Article : Google Scholar : PubMed/NCBI

97 

Yu W, Mi L and Wang F: Effect of the alteration of Tribbles homologue 3 expression on epithelial-mesenchymal transition of transforming growth factor β1-induced mouse alveolar epithelial cells through the Wnt/β-catenin signaling pathway. Mol Med Rep. 21:615–622. 2020.PubMed/NCBI

98 

Andonegui G, Ni A, Leger C, Kelly MM, Wong JF, Jalloul A and Winston BW: Sequential expression of IGF-IB followed by active TGF-β1 induces synergistic pulmonary fibroproliferation in vivo. Am J Physiol Lung Cell Mol Physiol. 303:L788–L798. 2012. View Article : Google Scholar : PubMed/NCBI

99 

Negreros M, Hagood JS, Espinoza CR, Balderas-Martinez YI, Selman M and Pardo A: Transforming growth factor beta 1 induces methylation changes in lung fibroblasts. PLoS One. 14:e02235122019. View Article : Google Scholar : PubMed/NCBI

100 

Sanders YY, Liu H, Scruggs AM, Duncan SR, Huang SK and Thannickal VJ: Epigenetic regulation of caveolin-1 gene expression in lung fibroblasts. Am J Respir Cell Mol Biol. 56:50–61. 2017. View Article : Google Scholar :

101 

Arsalane K, Dubois CM, Muanza T, Bégin R, Boudreau F, Asselin C and Cantin AM: Transforming growth factor-beta1 is a potent inhibitor of glutathione synthesis in the lung epithelial cell line A549: Transcriptional effect on the GSH rate-limiting enzyme gamma-glutamylcysteine synthetase. Am J Respir Cell Mol Biol. 17:599–607. 1997. View Article : Google Scholar : PubMed/NCBI

102 

Jardine H, MacNee W, Donaldson K and Rahman I: Molecular mechanism of transforming growth factor (TGF)-beta1-induced glutathione depletion in alveolar epithelial cells. Involvement of AP-1/ARE and Fra-1. J Biol Chem. 277:21158–21166. 2002. View Article : Google Scholar : PubMed/NCBI

103 

Boustani MR, Hertig IA, Maloney EK, Fanburg BL and White AC: Transforming growth factor B1 decreases uptake of glutathione precursor amino acids in bovine pulmonary artery endothelial cells. Endothelium. 5:1–10. 1997. View Article : Google Scholar : PubMed/NCBI

104 

Cho SJ and Stout-Delgado HW: Aging and lung disease. Annu Rev Physiol. 82:433–459. 2020. View Article : Google Scholar

105 

Wakwaya Y and Brown KK: Idiopathic pulmonary fibrosis: Epidemiology, diagnosis and outcomes. Am J Med Sci. 357:359–369. 2019. View Article : Google Scholar : PubMed/NCBI

106 

Abramson MJ, Murambadoro T, Alif SM, Benke GP, Dharmage SC, Glaspole I, Hopkins P, Hoy RF, Klebe S, Moodley Y, et al: Occupational and environmental risk factors for idiopathic pulmonary fibrosis in Australia: Case-control study. Thorax. 75:864–869. 2020. View Article : Google Scholar : PubMed/NCBI

107 

Somogyi V, Chaudhuri N, Torrisi SE, Kahn N, Müller V and Kreuter M: The therapy of idiopathic pulmonary fibrosis: What is next? Eur Respir Rev. 28:1900212019. View Article : Google Scholar : PubMed/NCBI

108 

Amor MS, Rosengarten D, Shitenberg D, Pertzov B, Shostak Y and Kramer MR: Lung transplantation in idiopathic pulmonary fibrosis: Risk factors and outcome. Isr Med Assoc J. 22:741–746. 2020.

109 

Yang S, Liu P, Jiang Y, Wang Z, Dai H and Wang C: Therapeutic applications of mesenchymal stem cells in idiopathic pulmonary fibrosis. Front Cell Dev Biol. 9:6396572021. View Article : Google Scholar : PubMed/NCBI

110 

Massagué J, Seoane J and Wotton D: Smad transcription factors. Genes Dev. 19:2783–2810. 2005. View Article : Google Scholar : PubMed/NCBI

111 

Chang X, Tian M, Zhang Q, Gao J, Li S and Sun Y: Nano nickel oxide promotes epithelial-mesenchymal transition through transforming growth factor β1/smads signaling pathway in A549 cells. Environ Toxicol. 35:1308–1317. 2020. View Article : Google Scholar : PubMed/NCBI

112 

Rosell-García T, Palomo-Álvarez O and Rodríguez-Pascual F: A hierarchical network of hypoxia-inducible factor and SMAD proteins governs procollagen lysyl hydroxylase 2 induction by hypoxia and transforming growth factor β1. J Biol Chem. 294:14308–14318. 2019. View Article : Google Scholar

113 

Ko J, Mills T, Huang J, Chen NY, Mertens TCJ, Collum SD, Lee G, Xiang Y, Han L, Zhou Y, et al: Transforming growth factor β1 alters the 3′-UTR of mRNA to promote lung fibrosis. J Biol Chem. 294:15781–15794. 2019. View Article : Google Scholar : PubMed/NCBI

114 

Senavirathna LK, Huang C, Pushparaj S, Xu D and Liu L: Hypoxia and transforming growth factor β1 regulation of long non-coding RNA transcriptomes in human pulmonary fibroblasts. Physiol Rep. 8:e143432020. View Article : Google Scholar

115 

Neveu WA, Mills ST, Staitieh BS and Sueblinvong V: TGF-β1 epigenetically modifies Thy-1 expression in primary lung fibroblasts. Am J Physiol Cell Physiol. 309:C616–C626. 2015. View Article : Google Scholar : PubMed/NCBI

116 

Kim S, Han JH, Kim S, Lee H, Kim JR, Lim JH and Woo CH: p90RSK inhibition ameliorates TGF-β1 signaling and pulmonary fibrosis by inhibiting Smad3 transcriptional activity. Cell Physiol Biochem. 54:195–210. 2020. View Article : Google Scholar : PubMed/NCBI

117 

Miyake Y, Sasaki S, Yokoyama T, Chida K, Azuma A, Suda T, Kudoh S, Sakamoto N, Okamoto K, Kobashi G, et al: Occupational and environmental factors and idiopathic pulmonary fibrosis in Japan. Ann Occup Hyg. 49:259–265. 2005.PubMed/NCBI

118 

Kim SY, Kang DM, Lee HK, Kim KH and Choi J: Occupational and environmental risk factors for chronic fibrosing idiopathic interstitial pneumonia in South Korea. J Occup Environ Med. 59:e221–e226. 2017. View Article : Google Scholar : PubMed/NCBI

119 

Baumgartner KB, Samet JM, Coultas DB, Stidley CA, Hunt WC, Colby TV and Waldron JA: Occupational and environmental risk factors for idiopathic pulmonary fibrosis: A multicenter case-control study. Collaborating centers. Am J Epidemiol. 152:307–315. 2000. View Article : Google Scholar : PubMed/NCBI

120 

García-Sancho Figueroa MC, Carrillo G, Pérez-Padilla R, Fernández-Plata MR, Buendía-Roldán I, Vargas MH and Selman M: Risk factors for idiopathic pulmonary fibrosis in a Mexican population. A case-control study. Respir Med. 104:305–309. 2010. View Article : Google Scholar

121 

Awadalla NJ, Hegazy A, Elmetwally RA and Wahby I: Occupational and environmental risk factors for idiopathic pulmonary fibrosis in Egypt: A multicenter case-control study. Int J Occup Environ Med. 3:107–116. 2012.PubMed/NCBI

122 

Koo JW, Myong JP, Yoon HK, Rhee CK, Kim Y, Kim JS, Jo BS, Cho Y, Byun J, Choi M, et al: Occupational exposure and idiopathic pulmonary fibrosis: A multicentre case-control study in Korea. Int J Tuberc Lung Dis. 21:107–112. 2017. View Article : Google Scholar : PubMed/NCBI

123 

Paolocci G, Folletti I, Torén K, Ekström M, Dell'Omo M, Muzi G and Murgia N: Occupational risk factors for idiopathic pulmonary fibrosis in Southern Europe: A case-control study. BMC Pulm Med. 18:752018. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

July-2021
Volume 48 Issue 1

Print ISSN: 1107-3756
Online ISSN:1791-244X

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ye Z and Ye Z: TGF‑β1: Gentlemanly orchestrator in idiopathic pulmonary fibrosis (Review). Int J Mol Med 48: 132, 2021
APA
Ye, Z., & Ye, Z. (2021). TGF‑β1: Gentlemanly orchestrator in idiopathic pulmonary fibrosis (Review). International Journal of Molecular Medicine, 48, 132. https://doi.org/10.3892/ijmm.2021.4965
MLA
Ye, Z., Hu, Y."TGF‑β1: Gentlemanly orchestrator in idiopathic pulmonary fibrosis (Review)". International Journal of Molecular Medicine 48.1 (2021): 132.
Chicago
Ye, Z., Hu, Y."TGF‑β1: Gentlemanly orchestrator in idiopathic pulmonary fibrosis (Review)". International Journal of Molecular Medicine 48, no. 1 (2021): 132. https://doi.org/10.3892/ijmm.2021.4965