Next Article in Journal
Anti-Inflammatory Activity of 3, 5-Diprenyl-4-hydroxyacetophenone Isolated from Ageratina pazcuarensis
Previous Article in Journal
Crosstalk between microRNA and Oxidative Stress in Heart Failure: A Systematic Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Promising Therapeutic Approaches for Radiation-Induced Pulmonary Fibrosis: Targeting Radiation-Induced Mesenchymal Transition of Alveolar Type II Epithelial Cells

Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, AMMS, Beijing 100850, China
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(23), 15014; https://doi.org/10.3390/ijms232315014
Submission received: 11 October 2022 / Revised: 16 November 2022 / Accepted: 26 November 2022 / Published: 30 November 2022
(This article belongs to the Section Molecular Biology)

Abstract

:
Radiation-induced pulmonary fibrosis (RIPF) is a common consequence of radiation for thoracic tumors, and is accompanied by gradual and irreversible organ failure. This severely reduces the survival rate of cancer patients, due to the serious side effects and lack of clinically effective drugs and methods. Radiation-induced pulmonary fibrosis is a dynamic process involving many complicated and varied mechanisms, of which alveolar type II epithelial (AT2) cells are one of the primary target cells, and the epithelial–mesenchymal transition (EMT) of AT2 cells is very relevant in the clinical search for effective targets. Therefore, this review summarizes several important signaling pathways that can induce EMT in AT2 cells, and searches for molecular targets with potential effects on RIPF among them, in order to provide effective therapeutic tools for the clinical prevention and treatment of RIPF.

Graphical Abstract

1. Introduction

Local normal tissues are also exposed to ionizing radiation during radiation therapy for thoracic cancers such as breast cancer, lung cancer, and esophageal cancer, resulting in serious radiotherapy side effects, including radiation-induced lung injury (RILI). We can divide the process into early acute radiation pneumonitis and late chronic radiation-induced pulmonary fibrosis (RIPF) [1,2,3]. Progressive and irreversible destruction of lung tissue and deterioration of lung function characterizes radiation-induced pulmonary fibrosis, with clinical manifestations of dyspnea, interstitial effusion, and even respiratory insufficiency [3,4]. According to statistics, the incidence of RIPF due to radiotherapy varies widely, but can be as high as 50% [3,4,5], thus severely limiting the application of radiotherapy in the treatment of clinical thoracic tumors. Because of its complex pathogenesis and the lack of effective therapeutic targets, there are a lack of effective treatment strategies in clinical practice [4,5]. General studies have shown that in RIPF, activated myofibroblasts secrete a large number of extracellular matrix (ECM) components in the inflammatory tissues, which greatly favors pulmonary fibrosis [1,3,6,7,8]. Moreover, irradiated alveolar type II epithelial (AT2) cells can acquire a mesenchymal phenotype through epithelial–mesenchymal transition (EMT) and serve as the main source of myofibroblasts in the RIPF [3,6,7]. Therefore, exploring the specific molecular pathway of radiation-induced EMT of AT2 cells is necessary to find new molecular targets and intervention methods for the treatment of RIPF.

2. Radiation-Induced EMT Promotes RIPF

2.1. RIPF

The pathological process of RIPF is mostly characterized by infiltration of inflammatory cells in the lower airways, damage to alveolar epithelial cells and vascular endothelial cells, and the proliferation of fibroblasts and myofibroblasts which secrete large amounts of extracellular matrix proteins (ECM) and collagen—ultimately damaging pulmonary structures [6,9]. Due to its complex pathogeny and poor prognosis, there is no effective treatment for RIPF. Therefore, the clinical approach to the treatment of RIPF focuses on prevention, which means avoiding and minimizing the exposure of normal lung tissue when giving high doses of radiation to the tumor. Until now, FDA-approved drugs for the treatment of pulmonary fibrosis, such as Pirfenidone and Nintedanib, can only alleviate the progression of pulmonary fibrosis to a certain extent, and are associated with serious side effects such as nephrotoxicity [10,11]. Therefore, the search for therapeutic targets of RIPF is of great importance for the clinical development of new treatments.

2.2. Radiation-Induced EMT

Radiation-induced EMT of alveolar epithelial cells has been considered to be an important source of pulmonary fibroblasts, which are responsible for the expression and secretion of ECM during RIPF [1]. Additionally, EMT is a complex process in which radiation activates various signaling molecules to trigger irradiation-induced EMT by enhancing reactive oxygen species (ROS) [1,7,9]. Then, the epithelial cells lose intercellular adhesion and gradually transform into mesenchymal cells, which occurs primarily during tissue injury, wound healing, organic fibrosis, and tumor metastasis [12,13,14,15,16,17]. Type I EMT participates in embryo implantation and organ development, Type II EMT favors tissue regeneration and fibrosis of organs, and type III EMT is involved in tumor metastasis [18,19,20,21]. The transcription regulation and gene expression models of epithelial cells are modified, in which transcription factors such as Snail, Slug, ZEB1, Twist, and TCF/LEF are implicated in cell reprogramming as key regulators of EMT, leading to decreased epithelial markers E-cadherin and ZO-1 as well as increased mesenchymal markers N-cadherin, vimentin, and α-SMA [12,13,14,17,18,19]. In addition, epithelial cells move from oval cells to fibroblasts, which then differentiate into myofibroblasts secreting extracellular matrix [13,14,15,16,17], including collagen (particularly types I and III), glycoproteins, and proteoglycans (fibronectin, laminin), which exacerbate fibrosis in tissue organs [12,13,19].
Alveolar type II epithelial (AT2) cells function as progenitors or stem cells that participate in the repair of pulmonary injury through self-renewal and differentiation into alveolar type I epithelial cells (AT1) [22,23,24]. In idiopathic pulmonary fibrosis (IPF), AT2 cells can acquire a mesenchymal phenotype through EMT, followed by differentiation into myofibroblasts to produce large amounts of ECM, leading to pulmonary fibrosis [6,25,26,27]. The EMT of AT2 cells leads to the exhaustion of alveolar epithelial stem cells, which reduces the repair capacity of the alveolar epithelium and speeds up pulmonary fibrosis in mice [28]. One study showed that in mouse models of IPF with only alveolar epithelial cells expressing β-gal, cells with increased vimentin were nearly always positive β-gal, suggesting that alveolar epithelial EMT is an important source of fibroblasts in pulmonary fibrosis [29]. Similarly, AT2 cells are considered to be part of an extremely important biological process during the RIPF (Figure 1) [6,9,30]. In studies of radiation-induced pulmonary fibrosis, the radiation dose at 6 Gy significantly inhibited the expression of E-cadherin and promoted the expression of vimentin in AT2 cells [9,25,26]. More powerfully, S1PR3 gene deficiency in the lung significantly ameliorated RIPF in recombinant adeno-associated virus-mediated S1PR3 (AAV-S1PR3) gene-deficient mice, as evidenced by reduced collagen deposition and elimination of α-SMA; additionally, irradiation-induced EMT and fibrosis were significantly suppressed in S1PR3-deficient alveolar epithelial cells [30].
During the development of RIPF, triggering ionizing radiation (IR)-induced EMT in AT2 cells requires appropriate extracellular signals in addition to IR-induced ROS, which include ECM components (integrins, MMPs, collagen) and several polypeptide growth factors such as TGF-β, Connective tissue growth factor (CTGF), Platelet-derived growth factor (PDGF), hepatocyte growth factor (HGF), fibroblast growth factor (FGF), Epidermal Growth Factor (EGF), and vascular endothelial growth factor (VEGF) that can interact with cognate receptors (tyrosine kinase receptors or serine/threonine kinase receptors) outside the cell membrane [1,13,17,22,23]. In addition, many complex and crosstalk intracellular signaling pathways are involved in the occurrence of EMT in AT2 cells. Recent studies suggest that ROS, Polypeptide growth factor, NF-κB, PI3K/AKT, epigenetic factors (MicroRNAs), Wnt/β-catenin, and Notch are all triggers involved in EMT in radiation-induced pulmonary fibrosis [1,2,3,4,5,6,7,8,9,24]. Although there are many studies involving the molecular mechanisms of RIPF, these studies have not yet been effectively translated into clinical applications.

3. Signaling Pathway Involved in EMT of AT2 Cell in RIPF

3.1. TGF-β Signaling Pathway

Transforming growth factor β (TGF-β) is a pleiotropic cytokine with three distinct isoforms, including TGF-β1, TGF-β2, and TGF-β3, that regulate cellular proliferation, differentiation, EMT, and immune regulation [3,25,26,27]. TGF-β is generally present in the ECM as a potential complex and can be separated from the potential complex to bind to its receptors for action, including the TGF-β type I receptor (TβR1) and the TGF-β type II receptor (TβR2) [3,25,26]. It has been noted that IR-induced ROS can promote the separation of TGF-β from the latent complex (Figure 2), so that activated TGF-β can bind to TGF-β type II receptor (TβR2) on the AT2 cell membrane, finally inducing ECM production and myofibroblast proliferation to promote RIPF by triggering the IR-induced EMT signal transduction pathway [22,28,29,30]. Additionally, in the TGF-β/Smad signaling pathway, TGF-β attaches to TβR2 to recruit TβR1 into the plasmatic membrane to form a heterotrimer. The heterotrimer phosphorylates Smad2/3 proteins (a process inhibited by Smad7) to form a complex with Smad4, which is subsequently transferred to the nucleus to interact with transcription factors to regulate transcription, translation, and protein synthesis of target genes [25,26]. In addition, the non-Smad pathway is associated with TGFβ-induced EMT through activation of multiple signaling pathways (PI3K-AKT, ERK-MAPK, p38-MAPK, JNK, RhoA, and NF-κB) [2,9,31].
The TGFβ-induced EMT in AT2 cells plays a key role in the development of RIPF. As the critical cytokine in pulmonary fibrosis, radiation-activated TGF-β triggers the EMT in AT2 cells (Figure 2), which increases mesenchymal genes N-cadherin and vimentin, as well as inhibiting epithelial genes ZO-1 and E-cadherin; it also transforms the morphology of epithelial cells into mesenchymal cells to produce a large amount of ECM [27,32]. Activated TGF-β carried by adenovirus may promote fibrosis in rodent models [6], and blocking TGF-β may attenuate the development of RIPF [3]. In addition, alveolar cells can be a source of TGF-β in pulmonary fibrosis. Similarly, irradiated mouse alveolar epithelial cells may release large amounts of chemokine to recruit M2 macrophages, and TGF-β secreted by M2 macrophages can induce EMT in MLE-12 to increase the expression of N-cadherin and promote the progression of RIPF [6]. In conclusion, the TGF-β pathway is the most promising therapeutic target for RIPF. Given that TGF-β is activated in ECM before it can bind to receptors on the AT2 cell membrane, and then regulate transcription factors in a Smad and non-Smad-dependent manner to promote the mesenchymal phenotype, clinical studies have mainly focused on inhibiting the synthesis and activation of TGF-β and blocking the signaling pathway.

3.2. Tyrosine Kinases Pathway

In RIPF, damaged alveolar epithelial cells, resident macrophages, and endothelial cells all secrete a variety of proinflammatory and profibrotic growth factors [1,33]. Previous studies have confirmed that the occurrence of chronic lung diseases is related to the overexpression of the growth factor ligands, or the increased expression of the receptor tyrosine kinases (RTKs) [34]. When related RTKs interact with polypeptide growth factor ligands including FGF, VEGF, EGF, HGF, and PDGF (Figure 2), these ligands can promote the dimerization of RTKs and autophosphorylation of their tyrosine residues, which can serve as docking sites for SH2 domains to activate related downstream signaling pathways such as RAS/MAPK and PI3K/AKT pathways, and initiate IR-induced EMT [22,23,35]. For example, many studies have shown that PDGF may play a role in EMT by promoting β-catenin translated into the nucleus in cooperation with TGF-β [36]. The expression of PDGF is up-regulated in the lung tissue of C57BL/6 mice with radiation-induced lung injury [37,38], and blocking PDGF by the inhibitors of RTKs can reduce the development of RIPF [3,38,39], which suggests that the PDGF–PDGFR system is a very promising potential target for the treatment of RIPF. Connective tissue growth factor (CTGF) is reported to be a TGF-β-mediated pro-fibrotic growth factor that promotes collagen synthesis and ECM deposition [40,41], thus is considered a potential molecular target to alleviate RIPF.

3.3. PI3K/AKT Signaling Pathway

PI3K is an intracellular phosphatidylinositol kinase that has three categories. Class I PI3K is a dimer consisting of catalytic subunit p110 and regulatory subunit p85 [42,43,44,45,46,47]. The activation signal for PI3K is mainly a variety of growth factors/signal transduction complexes, such as FGF, VEGF, EGF, HGF, and insulin [48,49]. In response to growth factor stimulation, the p85 regulatory subunit binds to the phosphotyrosine residues of the growth factor receptor, via its SH2 structural domain and the phosphotyrosine motif on the RTKs (Figure 2), recruiting the p110 catalytic subunit to reach the cell membrane and phosphorylate PIP2 [43,48]. Activated PI3K catalyzes PIP2 phosphorylation into PIP3 (a process inhibited by PTEN), which activates the protein kinase B (PKB or AKT) [49,50,51]. When phosphorylated at the Ser473 and Thr308 sites, AKT can control downstream pathways, including mTOR, MAPK, glycolysis, cell cycle, and apoptosis [42,49].
AT2 cells, in several studies, can promote the development of RIPF through PI3K/AKT signaling pathway-induced EMT. Previous studies have indicated that Re-Du-Ning (RDN) therapy may attenuate RILI in mice via the PI3K/AKT signaling pathway to suppress EMT in alveolar epithelial cells [52]. Additionally, TBK1, as a potential therapy target for RIPF, can directly activate the AKT signaling pathway to promote EMT in alveolar epithelial cells. Moreover, the PI3K/AKT signaling pathway can be activated in bleomycin-induced pulmonary fibrosis (the expression of p-PI3K and p-AKT are increasing) [53,54,55,56,57]. In DEC-deficient mice and cells, DEC deficiency improves idiopathic pulmonary fibrosis by inhibiting the PI3K/AKT/GSK-3β/β-Catenin signaling pathway to suppress EMT in AT2 cells [58]. Treatment with low molecular weight fucose (LMWF) relieves pulmonary fibrosis and EMT by inhibiting the PI3K/AKT signaling pathway, in vivo and in vitro [59]. In particular, the PI3K/AKT signaling pathway is a key target for treating COVID-19-related pulmonary fibrosis [57]. It is thus clear that since AKT downstream signaling triggered by PI3K activation induces significant EMT in AT2 cells, selecting PI3K as a new potential target and inhibiting its activation is more comprehensive and efficient for the treatment of RIPF.

3.4. Epigenetic Factors (MicroRNAs)

Epigenetics refers to the production of heritable phenotypic changes without altering the DNA sequence by mechanisms including DNA methylation, histone modifications and non-coding RNAs [60]. Epigenetics has been shown to play a key role in immune, cancer, endocrine and psychiatric disorders, and epigenetic aberrations can be reversed by drugs [61]. Recent studies have pointed out that epigenetics (such as DNA methylation, m6A modification of RNA, histone modification and the binding of non-coding RNAs to mRNA) can regulate the development of RIPF by promoting or inhibiting the expression of downstream target genes to regulate the activation of EMT-related signaling pathways, and promote RIPF [62,63,64]. Among these, non-coding RNAs are very promising biomarkers to regulate RIPF in epigenetics, which mainly include microRNA (miRNA), long noncoding RNA (lncRNA), circular RNA (circRNA) [60]. Additionally, our findings suggest a clear role for MicroRNAs in mouse RIPF.
MicroRNAs are a class of small non-coding RNAs containing 19–25 nucleotides that can play an important role in the post-translational gene regulation and expression, through gene silence and epigenetics [12,16]. As non-coding genes, MicroRNAs can regulate the expression of major transcription factors in EMT such as Snail, ZEB1/2, Slug or target genes (Figure 2), promoting or inhibiting the progression of EMT [12]. MicroRNA dysregulation is closely associated with initiating EMT in AT2 cells. Current studies have reported that abnormal expression of miR-21 exists in multiple aspects of pulmonary fibrosis, including EMT of alveolar epithelial cells and differentiation of fibroblasts into myofibroblasts [65,66]. Therefore, miR-21 is expected to become a new candidate MicroRNA for intervention in radiation-induced pulmonary fibrosis. In addition to miR-21, our laboratory found that the ectopic expression of miR-155-5p in the lungs of C57BL/6 mice, inverted IR-induced EMT in AT2 cells and attenuated RIPF in mice by downregulating GSK-3β [67]. Similarly, IR could significantly induce Snail and Slug expression in AT2 cells by, respectively, inhibiting miR-486-3p and miR-541-5p (Figure 2), triggering IR-induced EMT and inducing the development of RIPF [68,69]. Through the intervention of the mentioned MicroRNAs, it was found that the reversal effect of IR-induced EMT was very obvious, and the pathological results of RIPF in mice were significantly improved; this suggested to us that targeting miRNAs may be an effective strategy to improve the current treatment dilemma of RIPF.

4. Therapy Strategy of RIPF by Targeting EMT

4.1. Current Clinical Situation of Pulmonary Fibrosis

Currently, the drugs approved by the FDA for the treatment of pulmonary fibrosis are Nintedanib and Pirfenidone, both of which target the activation and proliferation of fibroblasts, thereby acting to inhibit the progression of pulmonary fibrosis. However, as research has progressed, numerous in vivo studies have shown that Nintedanib and Pirfenidone can also target the epithelial–mesenchymal transition of AT2 cells and mitigate the progression of lung fibrosis [70,71,72]. In addition, FG-3019, a human-derived CTGF monoclonal antibody, inhibits the conversion of epithelial cells into fibroblasts via EMT to inhibit pulmonary fibrosis [41]. FG-3019 has now completed Phase I and Phase II clinical trials and is currently undergoing Phase III clinical studies. In a phase II randomized placebo-controlled trial, administration of FG-3019 significantly reduced the decline in pulmonary function compared to placebo, and quantitative pulmonary fibrosis HRCT scores showed that FG-3019 slowed progression in patients with pulmonary fibrosis, was safe and well tolerated, and significantly improved patients’ quality of life [73,74]. Moreover, pulmonary fibrosis is not only one of the sequelae of radiotherapy for chest tumors, but also one of the pathological features of COVID-19 patients. The current national medical program-recommended Chinese medical formula for COVID-19 patients is Qimai Feiluoping decoction, which can significantly improve pulmonary function and reduce clinical symptoms in the clinical phase [75]. Its potential mechanisms of action have been identified through network pharmacology predictions, including inhibition of the TGF-β/Smad3 pathway to inhibit TGF-β1-induced AT2 cell proliferation, and inhibition of EMT to alleviate pulmonary fibrosis [75,76].
However, there are currently limited clinically available drugs for the treatment of pulmonary fibrosis, and clinical data on targeted EMT for the treatment of patients with pulmonary fibrosis are also scarce. We therefore note that many of the anti-tumor drugs currently used in clinical patients are targeted to EMT [77,78]. Galunisertib, the first TβR1 inhibitor to be advanced to clinical trials, inhibits TGF-β pathway-induced EMT in tumor cells. Additionally, clinical data showed lower protein expression levels of p-Smad2 in peripheral blood mononuclear cells of patients receiving the clinically beneficial Galunisertib [79,80]. Vactosertib is also a novel TβR1 inhibitor, currently under development in clinical trials, that inhibits the phosphorylation of Smad2 and Smad3 proteins, thereby suppressing EMT in tumor cells, and has been shown to be safe and well tolerated in phase I clinical studies [80,81,82]. In addition, Gefitinib (an EGFR-tyrosine kinase inhibitor) is among the anti-tumor drugs involved in the inhibition of the EMT process in tumor cells, with good results in clinical studies so far [83,84]. Although all of these clinical agents mentioned above are targeting EMT of tumor cells in cancer patients, investigating their effects on pulmonary fibrosis in vivo is an effective way of providing us with the ability to rapidly develop new drug candidates for the treatment of pulmonary fibrosis, due to their reliable safety and tolerability.

4.2. Pre-Clinical Treatment of RIPF

4.2.1. Targeting TGF-β Signaling Pathway

In RIPF, activation of the TGF-β/Smad signaling pathway is a crucial step. Nowadays, therapeutic strategies in clinical studies regard TGF-β as a potential target that can be divided into three types (Table 1): inhibits latent TGF-β synthesis (antisense molecules), inhibits the activation of TGF-β (neutralizing antibody), and inhibits downstream gene transduction (receptor kinase inhibitor) [25].
For example, as a major TGF-β activator in the pulmonary system, direct inhibition or knockdown of integrin αvβ6 is a potential therapeutic target for RIPF [85]. In addition, removal of ROS also can inhibit the activation of the TGF-β signaling pathway to some extent, as ROS activates TGF-β. Additionally, studies show that Amifostine is currently used as a clinical antiradiation drug with an active metabolite as a ROS scavenger (Table 1) and is a potential drug for the treatment of radiation-induced pulmonary fibrosis [86,87]. To prevent ROS, the superoxide dismutase fusion of TAT (SOD-TAT) significantly ameliorated radiation-induced lung injury in mice [88].
As TβR1 inhibitors, LY2109761 [1,2,3,25,89] directly inhibited pro-fibrotic signaling and balanced the TGF-β/BMP signaling pathway in the RIPF, Galunisertib (LY2157299) [3,25,90] downregulated Smad2 phosphorylation in lung cancer, SB203580 and WP631 inhibited downstream signaling by highly specific inhibition of TβR1 [1,2,91], and SM16 [1,2,92] significantly reduced the extent of RIPF in rats. Furthermore, Smad7, a TGF-β pathway inhibitor, inhibits TGF-β signaling transduction by inhibiting blocking of Smad2/3 phosphorylation [26,27,93]. The natural products, Halofuginone and Verbascoside, can inhibit TGF-β pathway activity by inhibiting Smad2/3 phosphorylation and increasing Smad7 mRNA expression [26,94,95], which are potential RIPF protective molecules.
As mentioned above, there are a wide variety of inhibitors targeting the TGF-β pathway, including antitumor drugs that have entered clinical trials and gained FDA approval. The investigation of the pharmacological activities and molecular mechanisms of these drugs can facilitate the development of new clinical applications, and provide a more immediate and effective approach for the clinical treatment of tumor patients with RIPF induced by radiotherapy. Moreover, the TGF-β signaling pathway is a powerful molecular mechanism in IR-induced EMT, that can amplify the regulatory effects on EMT by activating signaling molecules from other pathways or by crosstalk with other signaling pathways. Therefore, the development of inhibitors of the TGF-β pathway is of great potential value.

4.2.2. Targeting Growth Factors

Damaged AT2 cells, macrophages and endothelial cells secrete large amounts of pro-fibrotic growth factors during the EMT in AT2 cells, by regulating pathways such as the PI3K/AKT pathway [96,97,98]. Therefore, the growth factors have been considered as signaling molecules and targets associated with the development of RIPF (Table 1). Flufenidone (a novel anti-fibrotic agent) reduces cardiac and renal fibrosis by inhibiting CTGF expression [99]. Moreover, PDGF promotes idiopathic pulmonary fibrosis (IPF), bleomycin-induced pulmonary fibrosis, and RIPF [37,100]. Additionally, PDGF receptor tyrosine kinase inhibitors (imatinib/Gleevec, SU9518 and SU11657) significantly attenuated the progression of pulmonary fibrosis lesions and remodeled lung structure in animal models [38,39,101]. In addition, PDGF receptor tyrosine kinase inhibitors are currently in clinical trials [23]. It means that these inhibitors may have more obvious efficacy, selectivity and safety in the treatment of RIPF, and are expected to be candidate medicines for the prevention and treatment of RIPF in the future.

4.2.3. Additional Targets

In addition, we found that Syndecan-2 can inhibit fibroblast differentiation and reduce pulmonary fibrosis in irradiated mice by downregulating PI3K/AKT pathway activity [102]. 2-Methoxyestradiol (2-ME) inhibits radiation-induced elevation of HIF-1α levels, reduces vascular collagen deposition, and inhibits the development of RIPF by inhibiting radiation-induced EndMT and EMT [103]. Additionally, the small molecule inhibitor J2 of heat shock protein-27 (HSP-27), a candidate target used in mouse models for the treatment of pulmonary fibrosis (Table 1), inhibits the development of RIPF by inhibiting IkBa-NFkB signaling after cross-linking HSP27 [104]. It is likely that MyD88 (a key intracellular adapter for TLR signaling) prevents pulmonary fibrosis by regulating NF-κB activation to attenuate RILI (Table 1) [105]. The SIK2 (salt-inducible kinase-2) inhibitor ARN-3236 was recently reported to attenuate bleomycin-induced pulmonary fibrosis in mice [106]. This gives us new candidate targets for a more comprehensive search for appropriate therapeutic approaches.
Table 1. Therapeutic strategies and inhibitors for radiation-induced pulmonary fibrosis.
Table 1. Therapeutic strategies and inhibitors for radiation-induced pulmonary fibrosis.
MechanismNameStructural FormulaTypeTargetRef.
ROSAmifostineIjms 23 15014 i001Organic
thiophosphate
Scavenging
free radicals
[86,87]
SOD-TATIjms 23 15014 i002Recombinant
protein
Oxidative
damage
[88]
TGF-β
signaling
pathway
LY2109761Ijms 23 15014 i003Quinoline
derivatives
TβR1
inhibitor
[89]
GalunisertibIjms 23 15014 i004PyrrolopyrazoleTβR1
inhibitor
[90]
SB203580Ijms 23 15014 i005ImidazolesTβR1
inhibitor
[91]
WP631Ijms 23 15014 i006Bisintercalating
anthracycline
TβR1
inhibitor
[91]
SM16Ijms 23 15014 i007AntibodyTβR1
inhibitor
[92]
HalofuginoneIjms 23 15014 i008Quinazolinone
alkaloid
Smad2/3
phosphorylation
[94]
VerbascosideIjms 23 15014 i009GlucosidesSmad2/3
phosphorylation
Increasing
Smad7
[95]
Tyrosine
kinase
pathway
FlufenidoneIjms 23 15014 i010Anti-fibrotic
drug
CTGF[99]
FG-3019Ijms 23 15014 i011Recombinant
antibody
CTGF[41]
ImatinibIjms 23 15014 i012Antineoplastic
agent
PDGF[101]
SU9518Ijms 23 15014 i013Small
molecule
VEGF
PDGF
[38,39]
Addition2-MEIjms 23 15014 i01417β-hydroxy
steroid
PI3K/AKT
HIF-1α
[103]
J2Ijms 23 15014 i015Small
molecule
IkBa-NFkB
HSP-27
[104]
MyD88Ijms 23 15014 i016Recombinant
protein
NF-κB
activation
[105]

5. Conclusions

Early diagnosis and treatment of RILI in patients with thoracic tumors after radiotherapy is now agreed to be a very important treatment to prevent the development of RIPF. However, although precision radiotherapy can reduce RILI, it affects tumor outcomes and quality of survival, and therefore RIPF remains a pressing challenge for medical research to overcome. RIPF is a progressive disease that involves many complex molecular mechanisms, with various molecular pathways crosstalking each other and cooperatively contributing to the pathological process of RIPF. In addition to molecular targets that target the IR-induced EMT signaling pathway, there are also many drugs in development or in clinical trials that target other molecular targets, such as by inhibiting the recruitment of inflammatory cells or targeting the clearance of senescent cells [107,108,109]. Recently, steroids, diuretics, hormones, antioxidants and enzymes are used clinically to treat RIPF, but because these drugs are not symptomatic and lack specificity, their treatment is not very effective [110,111]. Therefore, further investigation of the molecular mechanisms involved in RIPF is essential to identify new clinical targets.
Currently, it is well established that epithelial cells convert into migratory and invasive mesenchymal cells during tumor development, but the precise role of EMT as a source of pulmonary fibroblasts has been challenged by an AT2 cell fate tracking study in a bleomycin-induced mouse model of pulmonary fibrosis [112]. One study found no evidence of transformation of labeled AT2 cells into myofibroblasts via EMT when using surfactant protein C-CreERT2 knock-in alleles to follow the fate of AT2 cells in vivo [112]. Another study also showed that minor alveolar epithelial cells can be transformed into fibrotic lesions via EMT in a TGF-α-induced pulmonary fibrosis model [113]. Therefore, some researchers argued that the genetic lineage tracing methods—used early to provide evidence of EMT involvement in proliferation of pulmonary fibroblasts in IPF, using β-galactosidase as a genetic marker—cannot rule out false-positive results [13]. Thus, the in vivo evidence that EMT can directly convert into fibroblasts is still somehow unclear during PF/IPF [114,115]. Similarly, there is also no in vivo evidence to support the notion that AT2 cells could directly contribute to myofibroblast population via EMT in RIPF, and lineage tracing studies should be performed to uncover the EMT role during RIPF. Although it is controversial as to whether pulmonary epithelial cells could be directly converted to myofibroblasts via EMT in pulmonary fibrosis, some studies suggested EMT could promote myofibroblast differentiation by secreting factors to promote pulmonary fibrosis [116]. Another study revealed that radiation-induced EMT in AT2 cells can alter the microenvironment by secreting cytokines to recruit macrophages, thereby promoting RIPF [6]. Therefore, it is essential to investigate the molecular mechanisms of EMT in RIPF, which will provide us with more potential targets to find effective strategies for RIPF.
In conclusion, we have summarized the targets in the major IR-induced EMT signaling pathways and systematically collated the inhibitors of the molecular targets involved in EMT (Table 1), with a view of finding more effective therapeutic strategies for the treatment of RIPF, and developing drugs with better efficacy and specificity.

Author Contributions

Conceived and drafted the manuscript, P.W.; Edited the manuscript, Z.Y.; Revised and approved the manuscript, P.-K.Z.; Conceived, revised and approved the manuscript, Y.G. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by grants from the National Natural Science Foundation of China (82073488, 82273568, 31870847 and 81773359).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Acknowledgments

We thank Shenghui Zhou for providing some reference in this manuscript and Yuhao Liu for revising parts of this manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Jin, H.; Yoo, Y.; Kim, Y.; Kim, Y.; Cho, J.; Lee, Y.S. Radiation-Induced Lung Fibrosis: Preclinical Animal Models and Therapeutic Strategies. Cancers 2020, 12, 1561. [Google Scholar] [CrossRef]
  2. Ding, N.H.; Li, J.J.; Sun, L.Q. Molecular mechanisms and treatment of radiation-induced lung fibrosis. Curr. Drug Targets 2013, 14, 1347–1356. [Google Scholar] [CrossRef]
  3. Chen, Z.; Wu, Z.; Ning, W. Advances in Molecular Mechanisms and Treatment of Radiation-Induced Pulmonary Fibrosis. Transl. Oncol. 2019, 12, 162–169. [Google Scholar] [CrossRef]
  4. He, Y.; Thummuri, D.; Zheng, G.; Okunieff, P.; Citrin, D.E.; Vujaskovic, Z.; Zhou, D. Cellular senescence and radiation-induced pulmonary fibrosis. Transl. Res. 2019, 209, 14–21. [Google Scholar] [CrossRef]
  5. Jarzebska, N.; Karetnikova, E.S.; Markov, A.G.; Kasper, M.; Rodionov, R.N.; Spieth, P.M. Scarred Lung. An Update on Radiation-Induced Pulmonary Fibrosis. Front. Med. 2020, 7, 585756. [Google Scholar] [CrossRef]
  6. Park, H.R.; Jo, S.K.; Jung, U. Ionizing Radiation Promotes Epithelial-to-Mesenchymal Transition in Lung Epithelial Cells by TGF-beta-producing M2 Macrophages. In Vivo 2019, 33, 1773–1784. [Google Scholar] [CrossRef] [Green Version]
  7. Son, B.; Kwon, T.; Lee, S.; Han, I.; Kim, W.; Youn, H.; Youn, B. CYP2E1 regulates the development of radiation-induced pulmonary fibrosis via ER stress- and ROS-dependent mechanisms. Am. J. Physiol. Lung Cell. Mol. Physiol. 2017, 313, L916–L929. [Google Scholar] [CrossRef]
  8. Yang, Q.; Zhang, P.; Liu, T.; Zhang, X.; Pan, X.; Cen, Y.; Liu, Y.; Zhang, H.; Chen, X. Magnesium isoglycyrrhizinate ameliorates radiation-induced pulmonary fibrosis by inhibiting fibroblast differentiation via the p38MAPK/Akt/Nox4 pathway. Biomed. Pharmacother 2019, 115, 108955. [Google Scholar] [CrossRef]
  9. Nagaraja, S.S.; Nagarajan, D. Radiation-Induced Pulmonary Epithelial-Mesenchymal Transition: A Review on Targeting Molecular Pathways and Mediators. Curr. Drug Targets 2018, 19, 1191–1204. [Google Scholar] [CrossRef]
  10. De Ruysscher, D.; Granton, P.V.; Lieuwes, N.G.; van Hoof, S.; Wollin, L.; Weynand, B.; Dingemans, A.M.; Verhaegen, F.; Dubois, L. Nintedanib reduces radiation-induced microscopic lung fibrosis but this cannot be monitored by CT imaging: A preclinical study with a high precision image-guided irradiator. Radiother. Oncol. 2017, 124, 482–487. [Google Scholar] [CrossRef]
  11. Qin, W.; Liu, B.; Yi, M.; Li, L.; Tang, Y.; Wu, B.; Yuan, X. Antifibrotic Agent Pirfenidone Protects against Development of Radiation-Induced Pulmonary Fibrosis in a Murine Model. Radiat. Res. 2018, 190, 396–403. [Google Scholar] [CrossRef]
  12. Musavi Shenas, M.H.; Eghbal-Fard, S.; Mehrisofiani, V.; Abd Yazdani, N.; Rahbar Farzam, O.; Marofi, F.; Yousefi, M. MicroRNAs and signaling networks involved in epithelial-mesenchymal transition. J. Cell. Physiol. 2019, 234, 5775–5785. [Google Scholar] [CrossRef]
  13. Bartis, D.; Mise, N.; Mahida, R.Y.; Eickelberg, O.; Thickett, D.R. Epithelial-mesenchymal transition in lung development and disease: Does it exist and is it important? Thorax 2014, 69, 760–765. [Google Scholar] [CrossRef] [Green Version]
  14. Brabletz, S.; Schuhwerk, H.; Brabletz, T.; Stemmler, M.P. Dynamic EMT: A multi-tool. for tumor progression. EMBO J. 2021, 40, e108647. [Google Scholar] [CrossRef]
  15. Wilson, M.M.; Weinberg, R.A.; Lees, J.A.; Guen, V.J. Emerging Mechanisms by which EMT Programs Control. Stemness. Trends Cancer 2020, 6, 775–780. [Google Scholar] [CrossRef]
  16. Mohammadinejad, R.; Biagioni, A.; Arunkumar, G.; Shapiro, R.; Chang, K.C.; Sedeeq, M.; Taiyab, A.; Hashemabadi, M.; Pardakhty, A.; Mandegary, A.; et al. EMT signaling: Potential contribution of CRISPR/Cas gene editing. Cell. Mol. Life Sci. 2020, 77, 2701–2722. [Google Scholar] [CrossRef]
  17. Marconi, G.D.; Fonticoli, L.; Rajan, T.S.; Pierdomenico, S.D.; Trubiani, O.; Pizzicannella, J.; Diomede, F. Epithelial-Mesenchymal Transition (EMT): The Type-2 EMT in Wound Healing, Tissue Regeneration and Organ Fibrosis. Cells 2021, 10, 1587. [Google Scholar] [CrossRef]
  18. Usman, S.; Waseem, N.H.; Nguyen, T.K.N.; Mohsin, S.; Jamal, A.; Teh, M.T.; Waseem, A. Vimentin Is at the Heart of Epithelial Mesenchymal Transition (EMT) Mediated Metastasis. Cancers 2021, 13, 4985. [Google Scholar] [CrossRef]
  19. Imran, S.A.M.; Yazid, M.D.; Idrus, R.B.H.; Maarof, M.; Nordin, A.; Razali, R.A.; Lokanathan, Y. Is There an Interconnection between Epithelial-Mesenchymal Transition (EMT) and Telomere Shortening in Aging? Int. J. Mol. Sci. 2021, 22, 3888. [Google Scholar] [CrossRef]
  20. Dongre, A.; Weinberg, R.A. New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer. Nat. Rev. Mol. Cell Biol. 2019, 20, 69–84. [Google Scholar] [CrossRef]
  21. Zhou, S.; Zhang, M.; Zhou, C.; Wang, W.; Yang, H.; Ye, W. The role of epithelial-mesenchymal transition in regulating radioresistance. Crit. Rev. Oncol. Hematol. 2020, 150, 102961. [Google Scholar] [CrossRef]
  22. Lee, S.Y.; Jeong, E.K.; Ju, M.K.; Jeon, H.M.; Kim, M.Y.; Kim, C.H.; Park, H.G.; Han, S.I.; Kang, H.S. Induction of metastasis, cancer stem cell phenotype, and oncogenic metabolism in cancer cells by ionizing radiation. Mol. Cancer 2017, 16, 10. [Google Scholar] [CrossRef] [Green Version]
  23. Cannito, S.; Novo, E.; di Bonzo, L.V.; Busletta, C.; Colombatto, S.; Parola, M. Epithelial-mesenchymal transition: From molecular mechanisms, redox regulation to implications in human health and disease. Antioxid Redox Signal. 2010, 12, 1383–1430. [Google Scholar] [CrossRef]
  24. Shao, L.; Zhang, Y.; Shi, W.; Ma, L.; Xu, T.; Chang, P.; Dong, L. Mesenchymal stromal cells can repair radiation-induced pulmonary fibrosis via a DKK-1-mediated Wnt/β-catenin pathway. Cell Tissue Res. 2021, 384, 87–97. [Google Scholar] [CrossRef]
  25. Chen, Y.; Di, C.; Zhang, X.; Wang, J.; Wang, F.; Yan, J.F.; Xu, C.; Zhang, J.; Zhang, Q.; Li, H.; et al. Transforming growth factor beta signaling pathway: A promising therapeutic target for cancer. J. Cell. Physiol. 2020, 235, 1903–1914. [Google Scholar] [CrossRef]
  26. Mirzaei, S.; Paskeh, M.D.A.; Saghari, Y.; Zarrabi, A.; Hamblin, M.R.; Entezari, M.; Hashemi, M.; Aref, A.R.; Hushmandi, K.; Kumar, A.P.; et al. Transforming growth factor-beta (TGF-beta) in prostate cancer: A dual function mediator? Int. J. Biol. Macromol. 2022, 206, 435–452. [Google Scholar] [CrossRef]
  27. Sisto, M.; Ribatti, D.; Lisi, S. Organ Fibrosis and Autoimmunity: The Role of Inflammation in TGFbeta-Dependent EMT. Biomolecules 2021, 11, 310. [Google Scholar] [CrossRef]
  28. Jobling, M.F.; Mott, J.D.; Finnegan, M.T.; Jurukovski, V.; Erickson, A.C.; Walian, P.J.; Taylor, S.E.; Ledbetter, S.; Lawrence, C.M.; Rifkin, D.B.; et al. Isoform-specific activation of latent transforming growth factor beta (LTGF-beta) by reactive oxygen species. Radiat. Res. 2006, 166, 839–848. [Google Scholar] [CrossRef]
  29. Yazaki, K.; Matsuno, Y.; Yoshida, K.; Sherpa, M.; Nakajima, M.; Matsuyama, M.; Kiwamoto, T.; Morishima, Y.; Ishii, Y.; Hizawa, N. ROS-Nrf2 pathway mediates the development of TGF-β1-induced epithelial-mesenchymal transition through the activation of Notch signaling. Eur. J. Cell Biol. 2021, 100, 151181. [Google Scholar] [CrossRef]
  30. Ramundo, V.; Giribaldi, G.; Aldieri, E. Transforming Growth Factor-β and Oxidative Stress in Cancer: A Crosstalk in Driving Tumor Transformation. Cancers 2021, 13, 3093. [Google Scholar] [CrossRef]
  31. Jung, J.W.; Hwang, S.Y.; Hwang, J.S.; Oh, E.S.; Park, S.; Han, I.O. Ionising radiation induces changes associated with epithelial-mesenchymal transdifferentiation and increased cell motility of A549 lung epithelial cells. Eur. J. Cancer 2007, 43, 1214–1224. [Google Scholar] [CrossRef]
  32. Strieter, R.M.; Mehrad, B. New mechanisms of pulmonary fibrosis. Chest 2009, 136, 1364–1370. [Google Scholar] [CrossRef] [Green Version]
  33. Todd, N.W.; Luzina, I.G.; Atamas, S.P. Molecular and cellular mechanisms of pulmonary fibrosis. Fibrogenesis Tissue Repair 2012, 5, 11. [Google Scholar] [CrossRef] [Green Version]
  34. Ingram, J.L.; Bonner, J.C. EGF and PDGF receptor tyrosine kinases as therapeutic targets for chronic lung diseases. Curr. Mol. Med. 2006, 6, 409–421. [Google Scholar] [CrossRef]
  35. Park, J.S.; Choi, H.I.; Kim, D.H.; Kim, C.S.; Bae, E.H.; Ma, S.K.; Kim, S.W. RON Receptor Tyrosine Kinase Regulates Epithelial Mesenchymal Transition and the Expression of Pro-Fibrotic Markers via Src/Smad Signaling in HK-2 and NRK49F Cells. Int. J. Mol. Sci. 2019, 20, 5489. [Google Scholar] [CrossRef] [Green Version]
  36. Patel, P.; West-Mays, J.; Kolb, M.; Rodrigues, J.C.; Hoff, C.M.; Margetts, P.J. Platelet derived growth factor B and epithelial mesenchymal transition of peritoneal mesothelial cells. Matrix Biol. 2010, 29, 97–106. [Google Scholar] [CrossRef]
  37. Tada, H.; Ogushi, F.; Tani, K.; Nishioka, Y.; Miyata, J.Y.; Sato, K.; Asano, T.; Sone, S. Increased binding and chemotactic capacities of PDGF-BB on fibroblasts in radiation pneumonitis. Radiat. Res. 2003, 159, 805–811. [Google Scholar] [CrossRef]
  38. Abdollahi, A.; Li, M.; Ping, G.; Plathow, C.; Domhan, S.; Kiessling, F.; Lee, L.B.; McMahon, G.; Gröne, H.J.; Lipson, K.E.; et al. Inhibition of platelet-derived growth factor signaling attenuates pulmonary fibrosis. J. Exp. Med. 2005, 201, 925–935. [Google Scholar] [CrossRef] [Green Version]
  39. Li, M.; Ping, G.; Plathow, C.; Trinh, T.; Lipson, K.E.; Hauser, K.; Krempien, R.; Debus, J.; Abdollahi, A.; Huber, P.E. Small molecule receptor tyrosine kinase inhibitor of platelet-derived growth factor signaling (SU9518) modifies radiation response in fibroblasts and endothelial cells. BMC Cancer 2006, 6, 79. [Google Scholar] [CrossRef] [Green Version]
  40. Lipson, K.E.; Wong, C.; Teng, Y.; Spong, S. CTGF is a central mediator of tissue remodeling and fibrosis and its inhibition can reverse the process of fibrosis. Fibrogenesis Tissue Repair 2012, 5, S24. [Google Scholar] [CrossRef]
  41. Bickelhaupt, S.; Erbel, C.; Timke, C.; Wirkner, U.; Dadrich, M.; Flechsig, P.; Tietz, A.; Pföhler, J.; Gross, W.; Peschke, P.; et al. Effects of CTGF Blockade on Attenuation and Reversal of Radiation-Induced Pulmonary Fibrosis. J. Natl. Cancer Inst. 2017, 109, djw339. [Google Scholar] [CrossRef] [Green Version]
  42. Jafari, M.; Ghadami, E.; Dadkhah, T.; Akhavan-Niaki, H. PI3k/AKT signaling pathway: Erythropoiesis and beyond. J. Cell. Physiol. 2019, 234, 2373–2385. [Google Scholar] [CrossRef]
  43. Cuevas, B.D.; Lu, Y.; Mao, M.; Zhang, J.; LaPushin, R.; Siminovitch, K.; Mills, G.B. Tyrosine phosphorylation of p85 relieves its inhibitory activity on phosphatidylinositol. 3-kinase. J. Biol. Chem. 2001, 276, 27455–27461. [Google Scholar] [CrossRef] [Green Version]
  44. Alcazar, I.; Cortes, I.; Zaballos, A.; Hernandez, C.; Fruman, D.A.; Barber, D.F.; Carrera, A.C. p85beta phosphoinositide 3-kinase regulates CD28 coreceptor function. Blood 2009, 113, 3198–3208. [Google Scholar] [CrossRef] [Green Version]
  45. Cortes, I.; Sanchez-Ruiz, J.; Zuluaga, S.; Calvanese, V.; Marques, M.; Hernandez, C.; Rivera, T.; Kremer, L.; Gonzalez-Garcia, A.; Carrera, A.C. p85beta phosphoinositide 3-kinase subunit regulates tumor progression. Proc. Natl. Acad. Sci. USA 2012, 109, 11318–11323. [Google Scholar] [CrossRef] [Green Version]
  46. Rao, L.; Mak, V.C.Y.; Zhou, Y.; Zhang, D.; Li, X.; Fung, C.C.Y.; Sharma, R.; Gu, C.; Lu, Y.; Tipoe, G.L.; et al. p85beta regulates autophagic degradation of AXL to activate oncogenic signaling. Nat. Commun. 2020, 11, 2291. [Google Scholar] [CrossRef]
  47. Rojas, J.M.; Spada, R.; Sanz-Ortega, L.; Morillas, L.; Mejias, R.; Mulens-Arias, V.; Perez-Yague, S.; Barber, D.F. PI3K p85 beta regulatory subunit deficiency does not affect NK cell differentiation and increases NKG2D-mediated activation. J. Leukoc. Biol. 2016, 100, 1285–1296. [Google Scholar] [CrossRef]
  48. Kano, Y.; Hiragami, F.; Motoda, H.; Akiyama, J.; Koike, Y.; Gomita, Y.; Inoue, S.; Kawaura, A.; Furuta, T.; Kawamura, K. C-SH2 point mutation converts p85beta regulatory subunit of phosphoinositide 3-kinase to an anti-aging gene. Sci. Rep. 2019, 9, 12683. [Google Scholar] [CrossRef] [Green Version]
  49. Yang, Q.; Jiang, W.; Hou, P. Emerging role of PI3K/AKT in tumor-related epigenetic regulation. Semin. Cancer Biol. 2019, 59, 112–124. [Google Scholar] [CrossRef]
  50. He, J.; de la Monte, S.; Wands, J.R. The p85beta regulatory subunit of PI3K serves as a substrate for PTEN protein phosphatase activity during insulin mediated signaling. Biochem. Biophys. Res. Commun. 2010, 397, 513–519. [Google Scholar] [CrossRef]
  51. Ueki, K.; Yballe, C.M.; Brachmann, S.M.; Vicent, D.; Watt, J.M.; Kahn, C.R.; Cantley, L.C. Increased insulin sensitivity in mice lacking p85beta subunit of phosphoinositide 3-kinase. Proc. Natl. Acad. Sci. USA 2002, 99, 419–424. [Google Scholar] [CrossRef] [Green Version]
  52. Yang, C.; Song, C.; Wang, Y.; Zhou, W.; Zheng, W.; Zhou, H.; Deng, G.; Li, H.; Xiao, W.; Yang, Z.; et al. Re-Du-Ning injection ameliorates radiation-induced pneumonitis and fibrosis by inhibiting AIM2 inflammasome and epithelial-mesenchymal transition. Phytomedicine 2022, 102, 154184. [Google Scholar] [CrossRef]
  53. Qu, H.; Liu, L.; Liu, Z.; Qin, H.; Liao, Z.; Xia, P.; Yang, Y.; Li, B.; Gao, F.; Cai, J. Blocking TBK1 alleviated radiation-induced pulmonary fibrosis and epithelial-mesenchymal transition through Akt-Erk inactivation. Exp. Mol. Med. 2019, 51, 1–17. [Google Scholar] [CrossRef]
  54. Li, S.; Gao, S.; Jiang, Q.; Liang, Q.; Luan, J.; Zhang, R.; Zhang, F.; Ruan, H.; Li, X.; Li, X.; et al. Clevudine attenuates bleomycin-induced early pulmonary fibrosis via regulating M2 macrophage polarization. Int. Immunopharmacol. 2021, 101, 108271. [Google Scholar] [CrossRef]
  55. Wang, Y.; Zhang, L.; Wu, G.R.; Zhou, Q.; Yue, H.; Rao, L.Z.; Yuan, T.; Mo, B.; Wang, F.X.; Chen, L.M.; et al. MBD2 serves as a viable target against pulmonary fibrosis by inhibiting macrophage M2 program. Sci. Adv. 2021, 7, eabb6075. [Google Scholar] [CrossRef]
  56. Xu, Y.; Wang, X.; Han, D.; Wang, J.; Luo, Z.; Jin, T.; Shi, C.; Zhou, X.; Lin, L.; Shan, J. Revealing the mechanism of Jiegeng decoction attenuates bleomycin-induced pulmonary fibrosis via PI3K/Akt signaling pathway based on lipidomics and transcriptomics. Phytomedicine 2022, 102, 154207. [Google Scholar] [CrossRef]
  57. Yang, F.; Chen, R.; Li, W.Y.; Zhu, H.Y.; Chen, X.X.; Hou, Z.F.; Cao, R.S.; Zang, G.; Li, Y.X.; Zhang, W. D-Limonene Is a Potential Monoterpene to Inhibit PI3K/Akt/IKK-alpha/NF-kappaB p65 Signaling Pathway in Coronavirus Disease 2019 Pulmonary Fibrosis. Front. Med. 2021, 8, 591830. [Google Scholar] [CrossRef]
  58. Hu, X.; Zou, M.; Ni, L.; Zhang, M.; Zheng, W.; Liu, B.; Cheng, Z. Dec1 Deficiency Ameliorates Pulmonary Fibrosis Through the PI3K/AKT/GSK-3beta/beta-Catenin Integrated Signaling Pathway. Front. Pharmacol. 2022, 13, 829673. [Google Scholar] [CrossRef]
  59. Wu, N.; Li, Z.; Wang, J.; Geng, L.; Yue, Y.; Deng, Z.; Wang, Q.; Zhang, Q. Low molecular weight fucoidan attenuating pulmonary fibrosis by relieving inflammatory reaction and progression of epithelial-mesenchymal transition. Carbohydr. Polym. 2021, 273, 118567. [Google Scholar] [CrossRef]
  60. Li, J.; Wang, R.; Shi, W.; Chen, X.; Yi, J.; Yang, X.; Jin, S. Epigenetic regulation in radiation-induced pulmonary fibrosis. Int. J. Radiat. Biol. 2022, 1–12. [Google Scholar] [CrossRef]
  61. Zhang, L.; Lu, Q.; Chang, C. Epigenetics in Health and Disease. Adv. Exp. Med. Biol. 2020, 1253, 3–55. [Google Scholar] [CrossRef]
  62. Kalash, R.; Berhane, H.; Au, J.; Rhieu, B.H.; Epperly, M.W.; Goff, J.; Dixon, T.; Wang, H.; Zhang, X.; Franicola, D.; et al. Differences in irradiated lung gene transcription between fibrosis-prone C57BL/6NHsd and fibrosis-resistant C3H/HeNHsd mice. In Vivo 2014, 28, 147–171. [Google Scholar]
  63. Chen, P.; Tian, K.; Tu, W.; Zhang, Q.; Han, L.; Zhou, X. Sirtuin 6 inhibits MWCNTs-induced epithelial-mesenchymal transition in human bronchial epithelial cells via inactivating TGF-β1/Smad2 signaling pathway. Toxicol. Appl. Pharmacol. 2019, 374, 1–10. [Google Scholar] [CrossRef]
  64. Zhao, J.; Han, D.X.; Wang, C.B.; Wang, X.L. Zbtb7b suppresses aseptic inflammation by regulating m(6)A modification of IL6 mRNA. Biochem. Biophys. Res. Commun. 2020, 530, 336–341. [Google Scholar] [CrossRef]
  65. Jiang, C.; Guo, Y.; Yu, H.; Lu, S.; Meng, L. Pleiotropic microRNA-21 in pulmonary remodeling: Novel insights for molecular mechanism and present advancements. Allergy Asthma Clin. Immunol. 2019, 15, 33. [Google Scholar] [CrossRef] [Green Version]
  66. Liu, Z.; Liang, X.; Li, X.; Liu, X.; Zhu, M.; Gu, Y.; Zhou, P. MiRNA-21 functions in ionizing radiation-induced epithelium-to-mesenchymal transition (EMT) by downregulating PTEN. Toxicol. Res. 2019, 8, 328–340. [Google Scholar] [CrossRef]
  67. Wang, D.; Liu, Z.; Yan, Z.; Liang, X.; Liu, X.; Liu, Y.; Wang, P.; Bai, C.; Gu, Y.; Zhou, P.K. MiRNA-155-5p inhibits epithelium-to-mesenchymal transition (EMT) by targeting GSK-3beta during radiation-induced pulmonary fibrosis. Arch. Biochem. Biophys. 2021, 697, 108699. [Google Scholar] [CrossRef]
  68. Liang, X.; Yan, Z.; Wang, P.; Liu, Y.; Ao, X.; Liu, Z.; Wang, D.; Liu, X.; Zhu, M.; Gao, S.; et al. Irradiation Activates MZF1 to Inhibit miR-541-5p Expression and Promote Epithelial-Mesenchymal Transition (EMT) in Radiation-Induced Pulmonary Fibrosis (RIPF) by Upregulating Slug. Int. J. Mol. Sci. 2021, 22, 11309. [Google Scholar] [CrossRef]
  69. Yan, Z.; Ao, X.; Liang, X.; Chen, Z.; Liu, Y.; Wang, P.; Wang, D.; Liu, Z.; Liu, X.; Zhu, J.; et al. Transcriptional inhibition of miR-486-3p by BCL6 upregulates Snail and induces epithelial-mesenchymal transition during radiation-induced pulmonary fibrosis. Respir. Res. 2022, 23, 104. [Google Scholar] [CrossRef]
  70. Guo, J.; Yang, Z.; Jia, Q.; Bo, C.; Shao, H.; Zhang, Z. Pirfenidone inhibits epithelial-mesenchymal transition and pulmonary fibrosis in the rat silicosis model. Toxicol. Lett. 2019, 300, 59–66. [Google Scholar] [CrossRef]
  71. Li, L.F.; Kao, K.C.; Liu, Y.Y.; Lin, C.W.; Chen, N.H.; Lee, C.S.; Wang, C.W.; Yang, C.T. Nintedanib reduces ventilation-augmented bleomycin-induced epithelial-mesenchymal transition and lung fibrosis through suppression of the Src pathway. J. Cell. Mol. Med. 2017, 21, 2937–2949. [Google Scholar] [CrossRef]
  72. Lv, Q.; Wang, J.; Xu, C.; Huang, X.; Ruan, Z.; Dai, Y. Pirfenidone alleviates pulmonary fibrosis in vitro and in vivo through regulating Wnt/GSK-3β/β-catenin and TGF-β1/Smad2/3 signaling pathways. Mol. Med. 2020, 26, 49. [Google Scholar] [CrossRef]
  73. Raghu, G.; Scholand, M.B.; de Andrade, J.; Lancaster, L.; Mageto, Y.; Goldin, J.; Brown, K.K.; Flaherty, K.R.; Wencel, M.; Wanger, J.; et al. FG-3019 anti-connective tissue growth factor monoclonal antibody: Results of an open-label clinical trial in idiopathic pulmonary fibrosis. Eur. Respir. J. 2016, 47, 1481–1491. [Google Scholar] [CrossRef] [Green Version]
  74. Richeldi, L.; Fernández Pérez, E.R.; Costabel, U.; Albera, C.; Lederer, D.J.; Flaherty, K.R.; Ettinger, N.; Perez, R.; Scholand, M.B.; Goldin, J.; et al. Pamrevlumab, an anti-connective tissue growth factor therapy, for idiopathic pulmonary fibrosis (PRAISE): A phase 2, randomised, double-blind, placebo-controlled trial. Lancet Respir. Med. 2020, 8, 25–33. [Google Scholar] [CrossRef]
  75. Yang, Y.; Ding, L.; Bao, T.; Li, Y.; Ma, J.; Li, Q.; Gao, Z.; Song, S.; Wang, J.; Zhao, J.; et al. Network Pharmacology and Experimental Assessment to Explore the Pharmacological Mechanism of Qimai Feiluoping Decoction Against Pulmonary Fibrosis. Front. Pharmacol. 2021, 12, 770197. [Google Scholar] [CrossRef]
  76. Pandolfi, L.; Bozzini, S.; Frangipane, V.; Percivalle, E.; De Luigi, A.; Violatto, M.B.; Lopez, G.; Gabanti, E.; Carsana, L.; D’Amato, M.; et al. Neutrophil Extracellular Traps Induce the Epithelial-Mesenchymal Transition: Implications in Post-COVID-19 Fibrosis. Front. Immunol. 2021, 12, 663303. [Google Scholar] [CrossRef]
  77. Chen, H.T.; Liu, H.; Mao, M.J.; Tan, Y.; Mo, X.Q.; Meng, X.J.; Cao, M.T.; Zhong, C.Y.; Liu, Y.; Shan, H.; et al. Crosstalk between autophagy and epithelial-mesenchymal transition and its application in cancer therapy. Mol. Cancer 2019, 18, 101. [Google Scholar] [CrossRef] [Green Version]
  78. Zhang, N.; Ng, A.S.; Cai, S.; Li, Q.; Yang, L.; Kerr, D. Novel therapeutic strategies: Targeting epithelial-mesenchymal transition in colorectal cancer. Lancet Oncol. 2021, 22, e358–e368. [Google Scholar] [CrossRef]
  79. Rodón, J.; Carducci, M.; Sepulveda-Sánchez, J.M.; Azaro, A.; Calvo, E.; Seoane, J.; Braña, I.; Sicart, E.; Gueorguieva, I.; Cleverly, A.; et al. Pharmacokinetic, pharmacodynamic and biomarker evaluation of transforming growth factor-β receptor I kinase inhibitor, galunisertib, in phase 1 study in patients with advanced cancer. Investig. New Drugs 2015, 33, 357–370. [Google Scholar] [CrossRef] [Green Version]
  80. Teicher, B.A. TGFβ-Directed Therapeutics: 2020. Pharmacol. Ther 2021, 217, 107666. [Google Scholar] [CrossRef]
  81. Choi, J.; Park, J.; Cho, I.; Sheen, Y. Co-treatment with vactosertib, a novel, orally bioavailable activin receptor-like kinase 5 inhibitor, suppresses radiotherapy-induced epithelial-to-mesenchymal transition, cancer cell stemness, and lung metastasis of breast cancer. Radiol. Oncol. 2022, 56, 185–197. [Google Scholar] [CrossRef]
  82. Jung, S.Y.; Yug, J.S.; Clarke, J.M.; Bauer, T.M.; Keedy, V.L.; Hwang, S.; Kim, S.J.; Chung, E.K.; Lee, J.I. Population pharmacokinetics of vactosertib, a new TGF-β receptor type Ι inhibitor, in patients with advanced solid tumors. Cancer Chemother. Pharmacol. 2020, 85, 173–183. [Google Scholar] [CrossRef]
  83. Hosomi, Y.; Morita, S.; Sugawara, S.; Kato, T.; Fukuhara, T.; Gemma, A.; Takahashi, K.; Fujita, Y.; Harada, T.; Minato, K.; et al. Gefitinib Alone Versus Gefitinib Plus Chemotherapy for Non-Small-Cell Lung Cancer With Mutated Epidermal Growth Factor Receptor: NEJ009 Study. J. Clin. Oncol. 2020, 38, 115–123. [Google Scholar] [CrossRef]
  84. Kanemaru, R.; Takahashi, F.; Kato, M.; Mitsuishi, Y.; Tajima, K.; Ihara, H.; Hidayat, M.; Wirawan, A.; Koinuma, Y.; Hayakawa, D.; et al. Dasatinib Suppresses TGFβ-Mediated Epithelial-Mesenchymal Transition in Alveolar Epithelial Cells and Inhibits Pulmonary Fibrosis. Lung 2018, 196, 531–541. [Google Scholar] [CrossRef]
  85. Puthawala, K.; Hadjiangelis, N.; Jacoby, S.C.; Bayongan, E.; Zhao, Z.; Yang, Z.; Devitt, M.L.; Horan, G.S.; Weinreb, P.H.; Lukashev, M.E.; et al. Inhibition of integrin alpha(v)beta6, an activator of latent transforming growth factor-beta, prevents radiation-induced lung fibrosis. Am. J. Respir. Crit. Care Med. 2008, 177, 82–90. [Google Scholar] [CrossRef] [Green Version]
  86. Li, L.; Nie, X.; Yi, M.; Qin, W.; Li, F.; Wu, B.; Yuan, X. Aerosolized Thyroid Hormone Prevents Radiation Induced Lung Fibrosis. Front. Oncol. 2020, 10, 528686. [Google Scholar] [CrossRef]
  87. Arora, A.; Bhuria, V.; Singh, S.; Pathak, U.; Mathur, S.; Hazari, P.P.; Roy, B.G.; Sandhir, R.; Soni, R.; Dwarakanath, B.S.; et al. Amifostine analog, DRDE-30, alleviates radiation induced lung damage by attenuating inflammation and fibrosis. Life Sci. 2022, 298, 120518. [Google Scholar] [CrossRef]
  88. Pan, J.; Su, Y.; Hou, X.; He, H.; Liu, S.; Wu, J.; Rao, P. Protective effect of recombinant protein SOD-TAT on radiation-induced lung injury in mice. Life Sci. 2012, 91, 89–93. [Google Scholar] [CrossRef]
  89. Flechsig, P.; Dadrich, M.; Bickelhaupt, S.; Jenne, J.; Hauser, K.; Timke, C.; Peschke, P.; Hahn, E.W.; Gröne, H.J.; Yingling, J.; et al. LY2109761 attenuates radiation-induced pulmonary murine fibrosis via reversal of TGF-β and BMP-associated proinflammatory and proangiogenic signals. Clin. Cancer Res. 2012, 18, 3616–3627. [Google Scholar] [CrossRef] [Green Version]
  90. Bueno, L.; de Alwis, D.P.; Pitou, C.; Yingling, J.; Lahn, M.; Glatt, S.; Trocóniz, I.F. Semi-mechanistic modelling of the tumour growth inhibitory effects of LY2157299, a new type I receptor TGF-beta kinase antagonist, in mice. Eur. J. Cancer 2008, 44, 142–150. [Google Scholar] [CrossRef]
  91. Li, Y.; Song, L.W.; Peng, R.Y.; Wang, D.W.; Jin, M.H.; Gao, Y.B.; Ma, J.J. [Effects of SB203580 and WP631 on Smad signal transduction pathway in lung fibroblasts after irradiation]. Chin. J. Cancer 2008, 27, 698–702. [Google Scholar]
  92. Anscher, M.S.; Thrasher, B.; Zgonjanin, L.; Rabbani, Z.N.; Corbley, M.J.; Fu, K.; Sun, L.; Lee, W.C.; Ling, L.E.; Vujaskovic, Z. Small molecular inhibitor of transforming growth factor-beta protects against development of radiation-induced lung injury. Int. J. Radiat. Oncol. Biol. Phys. 2008, 71, 829–837. [Google Scholar] [CrossRef]
  93. Dooley, S.; Hamzavi, J.; Ciuclan, L.; Godoy, P.; Ilkavets, I.; Ehnert, S.; Ueberham, E.; Gebhardt, R.; Kanzler, S.; Geier, A.; et al. Hepatocyte-specific Smad7 expression attenuates TGF-beta-mediated fibrogenesis and protects against liver damage. Gastroenterology 2008, 135, 642–659. [Google Scholar] [CrossRef] [Green Version]
  94. Juárez, P.; Fournier, P.G.J.; Mohammad, K.S.; McKenna, R.C.; Davis, H.W.; Peng, X.H.; Niewolna, M.; Mauviel, A.; Chirgwin, J.M.; Guise, T.A. Halofuginone inhibits TGF-β/BMP signaling and in combination with zoledronic acid enhances inhibition of breast cancer bone metastasis. Oncotarget 2017, 8, 86447–86462. [Google Scholar] [CrossRef] [Green Version]
  95. Wu, C.H.; Chen, C.H.; Hsieh, P.F.; Lee, Y.H.; Kuo, W.W.; Wu, R.C.; Hung, C.H.; Yang, Y.L.; Lin, V.C. Verbascoside inhibits the epithelial-mesenchymal transition of prostate cancer cells through high-mobility group box 1/receptor for advanced glycation end-products/TGF-β pathway. Environ. Toxicol. 2021, 36, 1080–1089. [Google Scholar] [CrossRef]
  96. Wynn, T.A. Integrating mechanisms of pulmonary fibrosis. J. Exp. Med. 2011, 208, 1339–1350. [Google Scholar] [CrossRef] [Green Version]
  97. Vallée, A.; Lecarpentier, Y.; Guillevin, R.; Vallée, J.N. Interactions between TGF-β1, canonical WNT/β-catenin pathway and PPAR γ in radiation-induced fibrosis. Oncotarget 2017, 8, 90579–90604. [Google Scholar] [CrossRef] [Green Version]
  98. Rube, C.E.; Uthe, D.; Schmid, K.W.; Richter, K.D.; Wessel, J.; Schuck, A.; Willich, N.; Rube, C. Dose-dependent induction of transforming growth factor beta (TGF-beta) in the lung tissue of fibrosis-prone mice after thoracic irradiation. Int. J. Radiat. Oncol. Biol. Phys. 2000, 47, 1033–1042. [Google Scholar] [CrossRef]
  99. Wang, L.H.; Liu, J.S.; Ning, W.B.; Yuan, Q.J.; Zhang, F.F.; Peng, Z.Z.; Lu, M.M.; Luo, R.N.; Fu, X.; Hu, G.Y.; et al. Fluorofenidone attenuates diabetic nephropathy and kidney fibrosis in db/db mice. Pharmacology 2011, 88, 88–99. [Google Scholar] [CrossRef]
  100. Westbury, C.B.; Yarnold, J.R. Radiation fibrosis--current clinical and therapeutic perspectives. Clin. Oncol. 2012, 24, 657–672. [Google Scholar] [CrossRef]
  101. Li, M.; Abdollahi, A.; Gröne, H.J.; Lipson, K.E.; Belka, C.; Huber, P.E. Late treatment with imatinib mesylate ameliorates radiation-induced lung fibrosis in a mouse model. Radiat. Oncol. 2009, 4, 66. [Google Scholar] [CrossRef] [Green Version]
  102. Tsoyi, K.; Chu, S.G.; Patino-Jaramillo, N.G.; Wilder, J.; Villalba, J.; Doyle-Eisele, M.; McDonald, J.; Liu, X.; El-Chemaly, S.; Perrella, M.A.; et al. Syndecan-2 Attenuates Radiation-induced Pulmonary Fibrosis and Inhibits Fibroblast Activation by Regulating PI3K/Akt/ROCK Pathway via CD148. Am. J. Respir. Cell Mol. Biol. 2018, 58, 208–215. [Google Scholar] [CrossRef]
  103. Choi, S.H.; Hong, Z.Y.; Nam, J.K.; Lee, H.J.; Jang, J.; Yoo, R.J.; Lee, Y.J.; Lee, C.Y.; Kim, K.H.; Park, S.; et al. A Hypoxia-Induced Vascular Endothelial-to-Mesenchymal Transition in Development of Radiation-Induced Pulmonary Fibrosis. Clin. Cancer Res. 2015, 21, 3716–3726. [Google Scholar] [CrossRef] [Green Version]
  104. Kim, J.Y.; Jeon, S.; Yoo, Y.J.; Jin, H.; Won, H.Y.; Yoon, K.; Hwang, E.S.; Lee, Y.J.; Na, Y.; Cho, J.; et al. The Hsp27-Mediated IkBα-NFκB Signaling Axis Promotes Radiation-Induced Lung Fibrosis. Clin. Cancer Res. 2019, 25, 5364–5375. [Google Scholar] [CrossRef] [Green Version]
  105. Brickey, W.J.; Neuringer, I.P.; Walton, W.; Hua, X.; Wang, E.Y.; Jha, S.; Sempowski, G.D.; Yang, X.; Kirby, S.L.; Tilley, S.L.; et al. MyD88 provides a protective role in long-term radiation-induced lung injury. Int. J. Radiat. Biol. 2012, 88, 335–347. [Google Scholar] [CrossRef]
  106. Zou, L.; Hong, D.; Li, K.; Jiang, B. Salt-inducible kinase 2 (SIK2) inhibitor ARN-3236 attenuates bleomycin-induced pulmonary fibrosis in mice. BMC Pulm. Med. 2022, 22, 140. [Google Scholar] [CrossRef]
  107. Schafer, M.J.; White, T.A.; Iijima, K.; Haak, A.J.; Ligresti, G.; Atkinson, E.J.; Oberg, A.L.; Birch, J.; Salmonowicz, H.; Zhu, Y.; et al. Cellular senescence mediates fibrotic pulmonary disease. Nat. Commun. 2017, 8, 14532. [Google Scholar] [CrossRef] [Green Version]
  108. Lehmann, M.; Korfei, M.; Mutze, K.; Klee, S.; Skronska-Wasek, W.; Alsafadi, H.N.; Ota, C.; Costa, R.; Schiller, H.B.; Lindner, M.; et al. Senolytic drugs target alveolar epithelial cell function and attenuate experimental lung fibrosis ex vivo. Eur. Respir. J. 2017, 50, 1602367. [Google Scholar] [CrossRef] [Green Version]
  109. Katzen, J.; Beers, M.F. Contributions of alveolar epithelial cell quality control. to pulmonary fibrosis. J. Clin. Investig. 2020, 130, 5088–5099. [Google Scholar] [CrossRef]
  110. Williams, J.P.; Johnston, C.J.; Finkelstein, J.N. Treatment for radiation-induced pulmonary late effects: Spoiled for choice or looking in the wrong direction? Curr. Drug Targets 2010, 11, 1386–1394. [Google Scholar] [CrossRef]
  111. Medhora, M.; Gao, F.; Jacobs, E.R.; Moulder, J.E. Radiation damage to the lung: Mitigation by angiotensin-converting enzyme (ACE) inhibitors. Respirology 2012, 17, 66–71. [Google Scholar] [CrossRef]
  112. Rock, J.R.; Barkauskas, C.E.; Cronce, M.J.; Xue, Y.; Harris, J.R.; Liang, J.; Noble, P.W.; Hogan, B.L. Multiple stromal populations contribute to pulmonary fibrosis without evidence for epithelial to mesenchymal transition. Proc. Natl. Acad. Sci. USA 2011, 108, E1475–E1483. [Google Scholar] [CrossRef]
  113. Hardie, W.D.; Hagood, J.S.; Dave, V.; Perl, A.K.; Whitsett, J.A.; Korfhagen, T.R.; Glasser, S. Signaling pathways in the epithelial origins of pulmonary fibrosis. Cell Cycle 2010, 9, 2769–2776. [Google Scholar] [CrossRef] [Green Version]
  114. Kage, H.; Borok, Z. EMT and interstitial lung disease: A mysterious relationship. Curr. Opin. Pulm. Med. 2012, 18, 517–523. [Google Scholar] [CrossRef]
  115. Hill, C.; Jones, M.G.; Davies, D.E.; Wang, Y. Epithelial-mesenchymal transition contributes to pulmonary fibrosis via aberrant epithelial/fibroblastic cross-talk. J. Lung Health Dis. 2019, 3, 31–35. [Google Scholar]
  116. Yao, L.; Conforti, F.; Hill, C.; Bell, J.; Drawater, L.; Li, J.; Liu, D.; Xiong, H.; Alzetani, A.; Chee, S.J.; et al. Paracrine signalling during ZEB1-mediated epithelial-mesenchymal transition augments local myofibroblast differentiation in lung fibrosis. Cell Death Differ. 2019, 26, 943–957. [Google Scholar] [CrossRef]
Figure 1. Radiation-induced EMT. Ionizing radiation induces epithelial–mesenchymal transition in AT2 cells, which lose their tight intercellular junctions and gradually lose their epithelial phenotype, eventually allowing them to acquire a mesenchymal phenotype and differentiate into fibroblasts. Thereafter, fibroblasts proliferate and activate in large numbers, further differentiating into myofibroblasts, which secrete large amounts of extracellular matrix components, leading to excessive deposition of extracellular matrix and promoting the development of RIPF. (AT1-alveolar type I epithelial, AT2-alveolar type II epithelial, ECM-extracellular matrix, ZO-1-zonula occludens-1).
Figure 1. Radiation-induced EMT. Ionizing radiation induces epithelial–mesenchymal transition in AT2 cells, which lose their tight intercellular junctions and gradually lose their epithelial phenotype, eventually allowing them to acquire a mesenchymal phenotype and differentiate into fibroblasts. Thereafter, fibroblasts proliferate and activate in large numbers, further differentiating into myofibroblasts, which secrete large amounts of extracellular matrix components, leading to excessive deposition of extracellular matrix and promoting the development of RIPF. (AT1-alveolar type I epithelial, AT2-alveolar type II epithelial, ECM-extracellular matrix, ZO-1-zonula occludens-1).
Ijms 23 15014 g001
Figure 2. Signaling pathways and targets of EMT, and its therapeutic approach in RIPF. ROS, TGF-β signaling pathway, growth factors and MicroRNA are promising targets for therapeutic and developmental applications in RIPF, and their targeted inhibitors have potent inhibitory effects on IR-induced EMT in AT2 cells, respectively, inhibiting further development of RIPF. (TGF-β- Transforming growth factor β, TβR1-TGF-β type I receptor, TβR2-TGF-β type II receptor, ROS- Reactive oxygen species, RTKs- receptor tyrosine kinases, PIP2-phosphatidylinositol-4,5-biphosphate, PIP3-phosphatidylinositol triphosphate, α-SMA-α-smooth muscle actin). All arrows indicate the transduction process and protein changes in the EMT signaling pathway, and the red crosses indicate that the above inhibitors can ultimately suppress the change of EMT-related marker proteins by inhibiting the elevation of transcription factors, ultimately inhibiting IR-induced EMT in AT2.
Figure 2. Signaling pathways and targets of EMT, and its therapeutic approach in RIPF. ROS, TGF-β signaling pathway, growth factors and MicroRNA are promising targets for therapeutic and developmental applications in RIPF, and their targeted inhibitors have potent inhibitory effects on IR-induced EMT in AT2 cells, respectively, inhibiting further development of RIPF. (TGF-β- Transforming growth factor β, TβR1-TGF-β type I receptor, TβR2-TGF-β type II receptor, ROS- Reactive oxygen species, RTKs- receptor tyrosine kinases, PIP2-phosphatidylinositol-4,5-biphosphate, PIP3-phosphatidylinositol triphosphate, α-SMA-α-smooth muscle actin). All arrows indicate the transduction process and protein changes in the EMT signaling pathway, and the red crosses indicate that the above inhibitors can ultimately suppress the change of EMT-related marker proteins by inhibiting the elevation of transcription factors, ultimately inhibiting IR-induced EMT in AT2.
Ijms 23 15014 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Wang, P.; Yan, Z.; Zhou, P.-K.; Gu, Y. The Promising Therapeutic Approaches for Radiation-Induced Pulmonary Fibrosis: Targeting Radiation-Induced Mesenchymal Transition of Alveolar Type II Epithelial Cells. Int. J. Mol. Sci. 2022, 23, 15014. https://doi.org/10.3390/ijms232315014

AMA Style

Wang P, Yan Z, Zhou P-K, Gu Y. The Promising Therapeutic Approaches for Radiation-Induced Pulmonary Fibrosis: Targeting Radiation-Induced Mesenchymal Transition of Alveolar Type II Epithelial Cells. International Journal of Molecular Sciences. 2022; 23(23):15014. https://doi.org/10.3390/ijms232315014

Chicago/Turabian Style

Wang, Ping, Ziyan Yan, Ping-Kun Zhou, and Yongqing Gu. 2022. "The Promising Therapeutic Approaches for Radiation-Induced Pulmonary Fibrosis: Targeting Radiation-Induced Mesenchymal Transition of Alveolar Type II Epithelial Cells" International Journal of Molecular Sciences 23, no. 23: 15014. https://doi.org/10.3390/ijms232315014

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop