Next Article in Journal
Warburg-like Metabolic Reprogramming in Endometriosis: From Molecular Mechanisms to Therapeutic Approaches
Previous Article in Journal
Chemoprevention of Colorectal Cancer—With Emphasis on Low-Dose Aspirin and Anticoagulants
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Epithelial Cell Dysfunction in Pulmonary Fibrosis: Mechanisms, Interactions, and Emerging Therapeutic Targets

1
Department of Physiology, School of Medicine, Southeast University, Nanjing 210009, China
2
Jiangsu Provincial Key Laboratory of Critical Care Medicine, Zhongda Hospital, School of Medicine, Southeast University, Nanjing 210009, China
*
Author to whom correspondence should be addressed.
Pharmaceuticals 2025, 18(6), 812; https://doi.org/10.3390/ph18060812
Submission received: 8 April 2025 / Revised: 21 May 2025 / Accepted: 27 May 2025 / Published: 28 May 2025
(This article belongs to the Section Biopharmaceuticals)

Abstract

:
Pulmonary fibrosis (PF) is a progressive and fatal interstitial lung disease characterized by chronic epithelial injury and excessive deposition of extracellular matrix (ECM) driven by dysregulated repair. Increasing evidence has shown that epithelial cell dysfunction plays a key role in PF, involving epithelial–mesenchymal transition (EMT), chronic oxidative stress, disruption of epithelial–immune interactions, and promoting pathological remodeling. Single-cell analyses have identified functionally distinct subpopulations of type 2 alveolar (AT2) cells with pro-fibrotic potential. Epithelial cells exhibit metabolic and epigenetic alterations during PF, which provide new approaches for therapeutic targets. This review summarizes the molecular mechanisms driving epithelial dysfunction in fibrosis progression, with a focus on key regulatory pathways, including transforming growth factor-beta (TGF-β), Wnt, and Notch signaling pathways, as well as miRNA-mediated networks. We also explored emerging epithelial-targeted therapies, ranging from FDA-approved agents (pirfenidone, nintedanib) to experimental inhibitors targeting Galectin-3 and Wnt/β-catenin, providing insights into precision anti-fibrosis strategies for clinical translation.

1. Introduction

Tissue fibrosis is a pathological process characterized by the excessive deposition of extracellular matrix (ECM) components, resulting in structural disruption and functional impairment of affected tissues [1]. This process is commonly observed in various chronic diseases, including liver fibrosis, pulmonary fibrosis (PF), and renal fibrosis. The progression of fibrosis is closely linked to inflammation, ECM remodeling, and fibroblast activation, leading to irreversible tissue scarring and organ dysfunction [2]. In particular, PF, notably idiopathic pulmonary fibrosis (IPF), is marked by progressive dyspnea, non-productive cough, and an irreversible decline in lung function, which often results in respiratory failure [3]. The median survival following diagnosis is typically 3 to 5 years [4]; however, current therapies, such as pirfenidone and nintedanib, significantly slow the annual decline in forced vital capacity (FVC) and reduce the risk of acute exacerbations in IPF patients [5,6]. Still, neither agent can halt or reverse established fibrotic remodeling. Real-world and systematic evaluations have also demonstrated sustained tolerability and survival benefits, but highlighted the persistent absence of therapies capable of regressing existing fibrosis [7]. Recent bibliometric analysis revealed a rapid increase in studies prioritizing lung-function protection over fibrosis reversal, underscoring the urgent need to deepen our understanding of PF pathogenesis and to identify truly disease-modifying therapeutic targets [5].
Epithelial cells, which line the surfaces of organs and tissues, perform essential functions including barrier formation, secretion, absorption, transport, and sensory perception [8]. Under normal physiological conditions, epithelial cells help maintain tissue integrity and contribute to repair processes through complex regulatory mechanisms. However, the dysfunction of epithelial cells plays a key role in the progression of fibrosis. Epithelial cell damage or abnormal repair responses are recognized as early driving factors in the development of PF [9]. In response to physical, chemical, or pathogenic insults, epithelial cells activate regenerative, inflammatory, and repair mechanisms to restore tissue function [10]. However, under pathological conditions, such as chronic inflammation or repeated injury, epithelial cells may undergo abnormal repair responses, leading to epithelial–mesenchymal transition (EMT). Through EMT, epithelial cells differentiate into fibroblast-like cells, directly contributing to the progression of fibrosis [11,12].
Additionally, the interaction between epithelial cells and immune cells during fibrosis progression has garnered increasing attention in recent studies [13]. Damage to epithelial cells, coupled with dysregulation of their repair mechanisms, is considered a pivotal event in the onset of numerous chronic fibrotic diseases. The injury to epithelial cells, their stress responses, and impaired repair processes may be critical steps in the initiation of fibrosis [14,15,16]. These findings suggest epithelial-centered pathways might be promising clinical therapeutic targets for PF. Consequently, elucidating the mechanisms by which functional changes in epithelial cells occur during fibrosis is essential for the development of novel therapeutic strategies for PF. This review will examine the role of epithelial cells in fibrosis, with a focus on the molecular mechanisms underlying epithelial dysfunction in PF, and will explore current and emerging therapeutic strategies targeting epithelial-centered pathways.

2. Epithelial Cell Dysfunction Involved in Pulmonary Fibrosis Process

In PF, epithelial cells, particularly alveolar epithelial cells comprising both type I alveolar epithelial cells (AT1) and type II alveolar epithelial cells (AT2), play multifaceted roles in the fibrotic process. These cells not only serve as key initiators of fibrosis but also contribute significantly to its progression. They promote fibrosis through various mechanisms, including epithelial injury, EMT, pro-fibrotic factors secretion, ECM remodeling, and inflammatory signaling (Figure 1). Damaged epithelial cells lose their ability to repair, leading to destruction of the alveolar structure and triggering fibrotic responses [4]. Through these processes, epithelial cells interact with other cell types, such as immune cells, ultimately promoting fibrosis and leading to the aberrant generation of fibrous tissue.

2.1. Epithelial Cell Injury and Repair Dysregulation

In the early stages of fibrosis, persistent epithelial cell injury drives the fibrotic process. Damaged epithelial cells lose their barrier function, allowing pathogens and proinflammatory factors to penetrate into deeper lung tissues. These infiltrators then trigger an inflammatory response that activates fibroblasts and immune cells, thereby accelerating fibrotic remodeling [17,18]. Additionally, injured cells secrete pro-fibrotic factors like transforming growth factor-beta (TGF-β), promoting fibroblast activation and ECM deposition, which leads to fibrosis through direct or autocrine/paracrine signaling [18,19,20,21]. In a healthy state, epithelial cells can regenerate and repair tissue, while in fibrosis, persistent injury and chronic inflammation impair this repair process, leading to apoptosis or autophagy [22]. These damaged cells worsen inflammation and fibrosis, leading to irreversible tissue damage [23]. Abnormal autophagy can also cause excessive ECM accumulation, preventing regeneration [24]. Ultimately, the loss of epithelial cell regenerative capacity is a key factor in the irreversible loss of tissue function during fibrosis progression.

2.2. Epithelial–Mesenchymal Transition

EMT refers to the transformation of epithelial cells into cells with mesenchymal properties, which is particularly critical in fibrosis and is one of the core mechanisms driving its development. Various cytokines, such as TGF-β, can initiate EMT by inducing epithelial cells to lose their polarity and cell–cell connections, while gaining motility [25,26]. This transition results in the formation of mesenchymal cells, including fibroblasts and myofibroblasts, which possess enhanced migratory and proliferative capabilities and secrete substantial amounts of ECM components, such as collagen. The excessive deposition and stiffening of the ECM not only alter the mechanical properties of the tissue but also further impact cellular activities, thereby promoting the progression of fibrosis [27,28].

2.3. Epithelial Cell Oxidative Stress

Oxidative stress promotes fibrosis by affecting cell signaling, inflammation, and ECM remodeling. In the process of chronic injury, the accumulation of reactive oxygen species (ROS) damages epithelial cells, triggers EMT, destroys tissue structure, and forms a vicious cycle of fibrosis [29]. ROS can activate the TGF-β pathway, increase Smad2/3 phosphorylation, and promote epithelial cell activation and ECM accumulation [30]. ROS not only upregulates ECM components expression but also inhibits the activity of matrix metalloproteinases (MMPs). Studies have shown that oxidative stress significantly reduces the activity of MMP-2 and MMP-9, leading to diminished degradation of the ECM, exacerbating tissue stiffness, and promoting fibrosis [31].
NADPH oxidase 4 (NOX4), located on the mitochondrial inner membrane, plays a critical role in PF by generating excessive ROS [32]. ROS accumulation could activate pro-fibrotic pathways such as TGF-β/Smad pathway. This promotes cell proliferation and collagen synthesis, contributing to fibrosis progression [33,34,35]. The pro-fibrotic responses and ECM deposition in the lungs induced by TGF-β1 were inhibited by siRNA-mediated NOX4 knockdown [36]. Deficiency in NOX4 showed protection in mice subjected to bleomycin (BLM) [32]. NOX4 also links to cellular senescence, accelerating the aging of lung cells and exacerbating fibrosis [37,38].

2.4. Interactions Between Epithelial Cells and Immune Cells

Epithelial cells interact closely with immune cells, such as macrophages and T cells, which play a crucial role in fibrosis. When damaged, epithelial cells release chemokines like C-C motif ligand 2 (CCL2) and C-X-C motif chemokine ligand 10 (CXCL10), attracting immune cells to the injured area and amplifying the inflammatory response [39]. Studies in mouse models show that this leads to increased infiltration of macrophages and T cells, further enhancing the local inflammatory response [39,40]. Activated immune cells release pro-fibrotic factors like Interleukin-13 (IL-13) and TGF-β, promoting EMT and fibrosis, and creating a positive feedback loop that worsens fibrosis over time [41]. Blocking TGF-β signaling in mouse models has been shown to reduce fibrosis [42]. Additionally, in silica-induced lung fibrosis, epithelial cells in the fibrotic regions can independently synthesize and secrete IgA, which may exacerbate fibrosis by activating fibroblasts [43]. Overall, the interaction between epithelial and immune cells plays a critical role in amplifying the fibrotic response.

2.5. Abnormal Accumulation of Extracellular Matrix

During fibrosis, damaged epithelial cells release large amounts of fibronectin, type I collagen, and other ECM components, leading to abnormal ECM accumulation. Normally, ECM degradation and remodeling are balanced by MMPs and their inhibitors, but injury disrupts this balance, causing ECM accumulation and promoting fibrosis [44,45]. Excessive ECM deposition increases tissue stiffness, which activates fibroblasts and worsens fibrosis [46]. Young’s modulus, the ratio of axial stress to axial strain within the material’s linear elastic region, is used to quantify the material’s stiffness under tension or compression. Research shows that fibrotic areas have a significantly higher Young’s modulus, making the tissue stiffer and further activating fibroblasts through mechanosensitive molecules like integrins [47]. The fibrotic ECM not only increases tissue stiffness but also reduces its compliance, thereby restricting alveolar expansion and contraction [48]. This leads to increased respiratory effort, dyspnea, and reduced gas exchange efficiency, causing impaired oxygenation. Additionally, ECM accumulation inhibits tissue regeneration, as seen in cardiac injury, where excessive ECM deposition hampers cardiomyocyte repair [49].

3. Key Mechanisms of Epithelial Cells Involved in Fibrosis

3.1. TGF-β Signaling Pathway

TGF-β is a key regulator in fibrosis, driving EMT and fibroblast activation, which increases ECM production and promotes fibrogenesis. It is upregulated in fibrosis across various organs, including the kidney, liver, and lungs, through both Smad-dependent and Smad-independent pathways [50,51]. TGF-β1 and its receptors (TGFβRI and TGFβRII) play a critical role in fibrosis progression. TGF-β was also increased in the airway and alveolar epithelium of patients with chronic obstructive pulmonary disease (COPD), revealing the complex and important role of TGF-β signaling in lung fibrosis [52]. In addition to the classical Smad-dependent pathway, TGF-β can also accelerate the fibrotic process through other non-classical pathways, such as the activation of PI3K/Akt and MAPK signaling pathways [53,54]. The amount of evidence suggests that targeting TGF-β cascades has the potential to treat patients with fibrosis.

3.2. Wnt/β-Catenin Signaling Pathway

The Wnt/β-catenin signaling pathway plays a pivotal role in regulating epithelial cell injury repair and differentiation. During fibrosis, the Wnt/β-catenin pathway becomes excessively activated. Upon translocation to the nucleus, β-catenin induces the expression of fibrosis-related genes. This upregulation leads to increased protein synthesis and secretion, leading to abnormal ECM accumulation and fibrosis progression [55,56]. In the early stages of fibrosis, the dysregulation of Wnt signaling triggers phenotypic changes in epithelial cells, resulting in EMT and the accumulation of ECM proteins [55,57]. Inhibition of the Wnt/β-catenin pathway significantly reduces the expression of fibrosis-related genes, leading to decreased collagen and fibronectin synthesis, as well as reduced ECM deposition [58]. Moreover, β-catenin inhibition has been shown to suppress EMT and reduce ECM production [59]. These findings highlight the critical role of Wnt/β-catenin signaling in the development of fibrosis.

3.3. Notch Signaling Pathway

The Notch signaling pathway plays a key role in regulating cell fate, including activation, proliferation, and differentiation. Its activation has been shown to promote EMT, thus advancing the fibrotic process [60,61]. Specifically, Notch1 and Notch3 activation are closely associated with abnormalities in epithelial cell injury and repair. In PF, Notch1 is a central regulator of AT2 fate, inducing alveolar epithelial proliferation and loss of Napsin A and surfactant proprotein processing, which contributes to fibrosis [62]. Notch inhibitors significantly reduce EMT-related gene expression [61]. In BLM-induced PF, mice lacking Notch1 show reduced fibrosis and collagen deposition, underscoring the role of Notch1 in fibrosis [63]. The activation of Notch3 promotes fibroblast proliferation and differentiation, driving the expansion of the fibroblast population. In contrast, the loss of Notch3 can alleviate PF by affecting the activation of fibroblasts, suggesting that Notch3 is expected to be a therapeutic target for PF [64,65].

3.4. Crosstalk Between Epithelial Signaling Pathways in Pulmonary Fibrosis

In the pathogenesis of PF, multiple signaling pathways work together to drive disease progression through complex crosstalk and synergistic regulatory networks. These pathways interact at multiple levels, including receptor binding, intracellular signaling nodes, and epigenetic modifications, forming an interconnected network rather than acting alone. Through this dynamic network regulation, pro-fibrotic signals are continuously amplified, driving irreversible pathological remodeling (Figure 2). The cascade amplification effect of this signal network suggests that the therapeutic strategy for a single target may have limitations, and it is necessary to analyze the key network nodes from the perspective of systems biology for combined intervention.
TGF-β and Wnt signaling pathways interact closely, particularly in fibrosis development. TGF-β induces Wnt ligand expression (e.g., Wnt5a, Wnt7b) through Smad3 phosphorylation, increasing their protein levels [66]. TGF-β1 has been shown to synergize with Wnt/β-catenin signaling pathway to induce EMT [67]. Wnt/β-catenin activation inhibits GSK3β, stabilizing Smad3 and forming a Smad3/β-catenin complex [68]. These pathways promote the expression of EMT core transcription factors (Snail and Slug) while inhibiting the expression of E-cadherin [69]. This cooperation between TGF-β and Wnt signaling provides insights for future therapeutic approaches.
Notch signaling exacerbates epithelial damage and EMT, working with TGF-β pathways to drive fibrosis [70]. It promotes the expression of key fibrotic factors, enhancing EMT and ECM accumulation. Notch induces myofibroblast differentiation of alveolar epithelial cells via the TGF-β-Smad3 pathway [71]. Studies show that Notch inhibitors can reduce fibrosis induced by TGF-β, highlighting the role of Notch in TGF-β-mediated fibrotic responses [72]. Additionally, Notch signaling influences fibroblast and myofibroblast activation, leading to ECM remodeling [73]. The crosstalk between Notch and TGF-β pathways represents a critical regulatory axis in fibrosis, suggesting that targeting both pathways may provide a more effective therapeutic strategy.
In fibrosis development, a positive feedback loop between oxidative stress and TGF-β signaling plays a critical role across various organs. TGF-β promotes mitochondrial ROS production by upregulating NOX4 and exacerbates oxidative stress, which in turn activates intracellular pathways driving fibrosis [74]. ROS also plays a bidirectional role in TGF-β signaling. Oxidative stress inhibits Smad7, a negative regulator of TGF-β signaling, leading to persistent phosphorylation of Smad2/3. Under normal conditions, Smad7 inhibits excessive TGF-β signaling. However, ROS accumulation reduces Smad7 levels, diminishing its inhibitory effect, which in turn prolongs Smad2/3 activation and then enhances fibroblast proliferation, migration, and ECM deposition, further promoting fibrosis [51,75,76,77]. This positive feedback loop between ROS and TGF-β signaling reveals the critical role of ROS in the progression of fibrosis. Targeting oxidative stress or disrupting this feedback loop may offer novel therapeutic strategies for fibrosis-related diseases.
Oxidative stress and Notch signaling interact through complex molecular mechanisms, in which ROS plays a crucial role in activating the Notch pathway. Mitochondrial-derived ROS activate the JNK signaling pathway, which subsequently induces the expression of the Notch ligand Jagged1, a ligand that activates Notch receptors [72,78]. Additionally, the intracellular domain of Notch competes with Nrf2 for binding to the coactivator CBP, thereby suppressing the expression of antioxidant genes, such as Superoxide Dismutase 2 (SOD2) [70,79]. Functionally, the ROS–Notch axis drives the emergence of a pro-fibrotic phenotype in epithelial cells, which further promotes fibrosis [80,81]. This interaction between oxidative stress and Notch signaling highlights a critical mechanism in fibrotic diseases and suggests potential targets for therapeutic intervention.
The Hippo-YAP pathway interacts with the TGF-β and Wnt signaling pathways through complex molecular mechanisms, regulating processes such as cell proliferation, differentiation, and fibrosis. Mechanical stress and ECM stiffness inhibit the Hippo pathway via integrin-FAK signaling, leading to the nuclear translocation of YAP/TAZ. Once in the nucleus, YAP/TAZ form a complex with Smad2/3, enhancing the efficiency of TGF-β signaling. Additionally, YAP and β-catenin cooperate to bind to TEAD/TCF elements, activating EMT-related genes such as VIM and ZEB1 [82,83,84]. This cross-talk between Hippo-YAP, TGF-β, and Wnt pathways plays a crucial role in regulating fibrosis and EMT, providing insights for potential therapeutic strategies.

3.5. Metabolic and Epigenetic Regulation: A Synergistic Axis in Pulmonary Fibrosis

Emerging evidence highlights the critical interplay between metabolic reprogramming and epigenetic modifications in the pathogenesis of PF. In this process, dysfunctional epithelial cells undergo profound molecular rewiring to adopt pro-fibrotic phenotypes (Figure 3). Mitochondrial dysfunction generates excessive ROS, which oxidize DNA methyltransferases (DNMTs) and histone demethylases (KDMs), leading to locus-specific hypermethylation and chromatin remodeling at pro-fibrotic genes, such as CTGF [85,86,87]. Concurrently, the accumulation of succinate stabilizes HIF-1α, promoting glycolytic flux and increased lactate production, which are key metabolic adaptations in fibrotic cells [88,89].
Additionally, metabolic intermediates like α-ketoglutarate (α-KG) and S-adenosylmethionine (SAM) regulate DNA methylation. AT2 cells with DNMT3A increased exhibit hypermethylation at regeneration-related genes, impairing regenerative potential and promoting fibrotic processes [90,91]. Therapeutic strategies targeting this axis, such as DNMT inhibitors and succinate receptor antagonists, have shown promise in preclinical models by reducing collagen synthesis and normalizing histone lactase, respectively [92,93,94]. Future research should focus on elucidating the spatiotemporal regulation of metabolic-epigenetic crosstalk and evaluating the potential of combination therapies to improve therapeutic outcomes in fibrosis treatment.

4. New Technologies Reveal the Heterogeneity and Functional Diversity of Epithelial Cells

The application of cutting-edge technologies such as single-cell sequencing, spatial transcriptomics, and metabolomics has deepened our understanding of epithelial cell behavior in PF and laid a theoretical foundation for better clinical intervention.

4.1. Heterogeneity Map of Pulmonary Epithelial Cells

4.1.1. Regenerative Function Partitioning of AT1/AT2 Cells

Alveolar epithelial regeneration under normal physiological conditions is primarily driven by AT2 cells, which act as stem cells and repair the lung by differentiating into AT1 cells. However, during PF, the regenerative function of AT2 cells becomes spatially and state-dependent, leading to impaired repair and altered cellular behavior. In the homeostasis zone, distant from the fibrosis lesions, resting AT2 cells maintain low-level proliferation through Notch signaling, primarily contributing to surfactant synthesis [95]. In the injury response zone, at the periphery of the lesion, AT2 cells are activated into a transitional KRT8+ state, showing both proliferation and TGF-β1 secretion. Under mechanical stress induced by increased ECM stiffness, some cells become locked in a pro-fibrotic phenotype (SFTPC+/SCGB3A2+), losing their ability to differentiate into AT1 cells. In the terminal disruption zone, at the core of the lesion, AT1 cells (AGER+/HOPX+) undergo apoptosis or senescence due to mitochondrial dysfunction caused by abnormal shear forces (ROS increase), leading to alveolar collapse [96,97,98]. In this process, residual AT1 cells exacerbate M2 macrophage polarization by secreting IL-33, thereby promoting the progression of fibrosis [99]. Single-cell multi-omics analysis showed that the Wnt/β-catenin pathway was specifically activated (increased nuclear translocation rate of β-catenin) in transitional AT2 cells, which synergistically drove EMT [100].

4.1.2. Double-Edged Basal Cells in Pulmonary Fibrosis

In normal repair, basal cells differentiate into ciliated cells or goblet cells through the Notch signaling pathway, maintaining airway epithelial homeostasis. Upon injury, basal cells activate the Wnt/β-catenin pathway, rapidly proliferate, and form KRT8+ transitional cells, which differentiate into AT1/AT2-like cells to participate in alveolar repair [101,102]. In the pathological process of IPF, basal cells show high heterogeneity in different phenotypes [103]. In the context of fibrosis, under chronic injury, basal cells respond to ECM stiffness through the YAP/TAZ signaling pathway, converting into a KRT17+ pro-fibrotic subpopulation, inducing fibroblast proliferation and collagen production [104,105]. A recent single-cell RNA sequencing revealed an aberrant basaloid cell type with high expression of pro-fibrotic markers and EMT evidence, localizing to the epithelial layer surrounding the fibroblastic foci [100,106]. WNT7A derived from basal cell or basal-like cells can be captured by neighboring fibroblasts and AT2 cells, promoting fibrogenesis at the fibrotic niche in IPF [107].
In addition, IL-13 drives reprogramming through STAT6 to induce the expression of chemokines, which in turn recruit inflammatory cells such as Th2 cells and eosinophils to form a chronic inflammatory microenvironment [108,109,110]. Epigenetic studies have shown that, in the process of fibrosis, the promoter region of EMT-related genes in pro-fibrotic basal cells is demethylated, which enhances their transcriptional activity and promotes the EMT process, thereby aggravating fibrosis [4,111,112]. Thus, reversing the pro-fibrotic phenotype of basal cells can be a therapeutic target in the progression of PF. DNA methyltransferase inhibitors, such as 5-azacytidine, can reverse the abnormal methylation of pro-fibrotic genes, inhibit the activation of fibroblasts and collagen deposition, and thus slow down the progression of PF [113,114]. However, its role in the regulation of basal cell phenotype still needs to be further verified.

4.2. Spatial Microenvironment-Driven Functional Polarization

The spatially heterogeneous microenvironment in PF remodels epithelial cell behavior through mechanical stress, metabolic gradients, and immune–epithelial interactions. Increased ECM stiffness mechanotransduces via YAP/TAZ activation in AT2 cells, promoting their EMT through enhanced nuclear localization of YAP and upregulation of EMT genes [115,116,117].
Succinate, a tricarboxylic acid cycle intermediate, accumulates in IPF and engages its receptor GPR91 to activate ERK and stabilize HIF-1α, driving fibroblast activation [118,119]. Pharmacological inhibition of succinate dehydrogenase attenuates TGF-β1-induced succinate elevation and alleviates fibrosis, whereas exogenous succinate exacerbates it [119,120]. Lactate produced during inflammation induces histone lactylation in macrophages, upregulating pro-fibrotic gene expression and reinforcing fibrogenic signaling [121,122,123,124].
Multiplexed CODEX imaging reveals enrichment of M2-polarized macrophages at fibrotic lesions, where they suppress AT2 cell regeneration via the PD-1/PD-L1 axis and secrete IL-13 to activate fibroblasts and promote collagen deposition [2,125,126]. These findings underscore the complex interplay of mechanical, metabolic, and immune factors driving fibrosis.

4.3. MicroRNAs Regulation

MicroRNAs (miRNAs) are crucial regulators of gene expression in fibrosis. Notably, miR-21 is significantly upregulated in mouse models of PF, enhancing TGF-β signaling by inhibiting the antifibrotic factor Smad7 [127]. Treatment with miR-21 inhibitors leads to a significant decrease in collagen deposition and a rebound in Smad7 expression levels, further confirming the pro-fibrotic role of miR-21 [128]. On the other hand, members of the miR-200 family protect against fibrosis by inhibiting EMT-related factors like ZEB1 and ZEB2. Overexpression of miR-200b in mice decreases collagen I levels and increases E-cadherin expression, suggesting that miR-200b mitigates fibrosis by inhibiting EMT [129]. Conversely, miR-200 knockout models show increased EMT factor expression and collagen deposition, highlighting the protective role of miR-200 in fibrosis suppression [130].

4.4. Clinical Translation Potential of the Heterogeneity Map

The epithelial cell heterogeneity atlas in PF has significant clinical value in diagnosis, targeted therapy, and prognostic monitoring. Single-cell RNA sequencing and spatial transcriptomics have identified disease-specific epithelial subpopulations, such as SCGB3A2+ pro-fibrotic AT2 cells, leading to novel biomarkers like serum MMP7 and sputum KRT17 mRNA. Combined with AI-based radiomics models, diagnostic accuracy has reached 92%. Targeted therapies, including Galectin-3 inhibitors (GB0139) and YAP/TAZ inhibitors (VP-001), are in clinical trials. Dynamic profiling, such as liquid biopsy, further advances precision medicine. However, challenges remain, including high sequencing costs, complex microenvironment dynamics, drug delivery issues, and lack of biomarker standardization. Innovations in technology and optimized clinical strategies offer promise for accelerating the clinical application of heterogeneity maps and providing more precise treatments for PF patients.

5. Anti-Pulmonary Fibrosis Drugs Targeting Epithelial Cells

As mentioned above, alterations in epithelial cell function play an important role in PF. Regulating the injury response of epithelial cells, reducing ECM deposition, and affecting the fibrosis process can effectively promote anti-fibrosis outcomes. In this section, we provide a review of current antifibrotic agents that target the regulation of epithelial cell function in the context of PF. Many new drugs are currently in clinical trials and are expected to provide more treatment options for PF.

5.1. FDA-Approved Drugs for Clinical Use

5.1.1. Pirfenidone

Pirfenidone exhibits antifibrotic, anti-inflammatory, and antioxidant properties, making it a broad-spectrum agent for fibrosis. Although its exact mechanism is not fully understood, it influences EMT by inhibiting pro-fibrotic pathways such as TGF-β and fibroblast growth factors, alleviating fibrosis caused by epithelial injury. Pirfenidone inhibits TGF-β1-induced fibroblast activation and collagen synthesis, confirming its role in suppressing EMT [131]. It also reduces inflammatory factors like TNF-α, IL-6, and IL-1β in animal models, and decreases lung tissue inflammation, demonstrating its anti-inflammatory effects [132]. Furthermore, pirfenidone reduces oxidative stress markers (ROS and MDA) and increases antioxidant enzyme activity (SOD and CAT), boosting cellular antioxidant capacity [133,134].
In the CAPACITY and ASCEND clinical trials, pirfenidone significantly slowed the decline in lung function in patients with IPF. Patients receiving pirfenidone treatment exhibited a significantly lower rate of decline in FVC compared to the placebo group, indicating its effectiveness in clinically delaying disease progression [135].

5.1.2. Nintedanib

Nintedanib is a multi-targeted tyrosine kinase inhibitor that blocks pro-fibrotic pathways such as VEGF, PDGF, and FGF, reducing epithelial cell damage, apoptosis, and fibrosis. Its primary action is to inhibit fibroblast proliferation and migration, but it also regulates epithelial cell function to alleviate PF [136,137]. Research shows that nintedanib reduces apoptosis in alveolar epithelial cells and increases E-cadherin expression, supporting epithelial integrity [137]. In preclinical IPF mouse models, it significantly reduced PF, collagen content, and improved lung function and gas exchange [138]. Additionally, nintedanib has anti-inflammatory effects, inhibiting TNF-α and IL-1β secretion, and reducing inflammatory cell infiltration by blocking the PI3K/Akt/mTOR pathway [139]. Approved for IPF treatment, nintedanib effectively slows disease progression and improves lung function.

5.2. Drugs Developed Based on Molecular Mechanisms

5.2.1. TGF-β Signaling Pathway Inhibitors

TGF-β is a key factor in PF, promoting fibroblast activation and EMT. Inhibiting the TGF-β signaling pathway can reduce the transition of alveolar epithelial cells into fibroblasts, alleviating fibrosis. Drugs targeting TGF-β, such as antisense oligonucleotides or monoclonal antibodies, block its binding to receptors or inhibit downstream signaling (e.g., Smad phosphorylation), effectively reducing fibrosis caused by epithelial cell damage [140]. For example, tranilast suppresses the TGF-β/Smad2 pathway, and (S)-ibuprofen-pirfenidone conjugate 5b reduces lung histopathological changes and collagen deposition in fibrosis models [141]. While TGF-β inhibitors are still in clinical trials, they have not yet been approved for treating PF.

5.2.2. Antioxidants

Oxidative stress is a significant cause of damage and apoptosis in alveolar epithelial cells. Therefore, inhibiting oxidative stress can effectively block the progression of fibrosis. Antioxidants, such as N-acetylcysteine (NAC), can reduce oxidative damage by scavenging free radicals, thereby protecting alveolar epithelial cells and slowing the progression of fibrosis [142,143]. NAC has shown some efficacy in certain patients with PF, but further research is needed.

5.2.3. Wnt/β-Catenin Signaling Pathway Inhibitors

The Wnt/β-catenin pathway is essential for epithelial repair, but excessive activation is linked to fibrosis. Inhibiting Wnt signaling can reduce EMT and slow fibrosis progression. Currently, Wnt inhibitors are in preclinical or early clinical stages, with no approved drugs. IWP-2 is a small molecule that inhibits β-catenin by interfering with Wnt signaling, showing potential antifibrotic effects in PF models [144]. C59 also inhibits the Wnt pathway by blocking β-catenin translocation and gene expression, suppressing fibrosis in liver and lung models [145]. Additionally, low-molecular-weight Fucoidan from sea cucumber Acaudina Molpadioides exhibits anti-fibrotic activity by modulating Wnt/β-catenin signaling [146]. However, recent studies suggest selective activation of Wnt may also benefit IPF [147]. These findings underscore the potential of Wnt/β-catenin as an antifibrotic target, with future studies needed to confirm their efficacy in specific diseases.

5.2.4. FGF Receptor Inhibitors

The FGF family plays a significant role in PF, particularly in promoting the repair and regeneration of epithelial cells. However, excessive FGF signaling can lead to fibrosis [148]. FGF receptor inhibitors work by inhibiting the overactive signaling pathways, thereby reducing the progression of fibrosis. Fibroblast growth factor-10 (FGF-10) is crucial for the regeneration of alveolar epithelial cells. FGF-10 analogs can promote the repair and regeneration of damaged epithelial cells, thus preventing the progression of fibrosis [149,150]. Currently, FGF-10 analogs have shown promise in animal models, but studies in humans are still ongoing.

5.3. New Targeted Drugs Developed by New Technology Applications

5.3.1. Galectin-3 Inhibitors

Galectin-3 is a glycoprotein that plays a crucial role in PF, particularly in mediating interactions between epithelial cells and fibroblasts. Its involvement in PF has attracted significant attention in recent years. Inhibiting Galectin-3 has been shown to disrupt the transmission of fibrotic signals and mitigate the fibrotic response following epithelial cell injury [151]. Currently, several inhibitors targeting Galectin-3 are in clinical trials, offering promising potential for therapeutic intervention.
GB-0139 is an oral Galectin-3 inhibitor developed by Galecto Biotech and is currently undergoing clinical trials to evaluate its efficacy and safety in patients with IPF. Preliminary results indicate that GB-0139 can significantly lower levels of Galectin-3 and may positively impact the progression of PF [152,153]. GR-MD-02, developed by Galectin Therapeutics, is primarily aimed at treating liver fibrosis and other fibrosis-related diseases. This recombinant Galectin-3 inhibitor has shown potential in clinical trials for reducing fibrotic lesions, particularly in patients with chronic liver disease [154,155]. TD139 is another Galectin-3 inhibitor currently under clinical investigation to assess its effects on COPD and PF. Early studies indicate that TD139 has good tolerability and can significantly reduce biomarkers associated with fibrosis [156]. The research on these inhibitors highlights the potential of targeting Galectin-3 as a therapeutic approach for fibrosis-related diseases. However, further clinical trial data are necessary to confirm their long-term efficacy and safety.

5.3.2. MicroRNAs Regulators

Certain miRNAs, such as miR-21 and the miR-200 family, have been identified as key regulators in EMT and fibrosis [157,158,159,160]. These miRNAs play crucial roles in the initiation and progression of fibrosis by modulating various cellular processes, including cell proliferation, migration, and ECM deposition. Interestingly, manipulating miRNA expression, either through inhibitors or enhancers, has emerged as a promising strategy to modify the behavior of epithelial cells and potentially prevent or reverse the development of fibrosis. For instance, miR-29 mimics, such as MRG-201 and MRG-229, have shown significant potential in reducing collagen secretion and maintaining the integrity of lung alveolar architecture, offering a potential therapeutic approach for fibrotic diseases [161,162]. While miRNA-based therapies are still under active investigation, early data suggest that these agents could serve as effective tools for treating a range of fibrosis-related conditions. However, further research and clinical trials are required to fully assess their safety, efficacy, and long-term impact on fibrotic diseases.

5.3.3. Therapies Targeting Epithelial Cell-Derived Exosomes

In response to injury, epithelial cells release exosomes (small extracellular vesicles rich in pro-fibrotic mediators, miRNAs, and proteins) that facilitate intercellular communication and contribute to PF progression. These exosomes promote EMT, activate fibroblasts, and enhance ECM deposition [163]. Notably, miR-21 in exosomes was shown to modulate the TGF-β pathway to further amplify fibrotic signaling [164,165]. The inhibition of exosome secretion with drugs such as GW4869, a neutral sphingomyelinase inhibitor, has been effective in reducing PF in animal models by interfering with pro-fibrotic signaling between epithelial cells and other cells [166,167,168,169]. Although still in the early stages of development, exosome-targeted therapy represents a promising therapeutic avenue for PF and warrants further clinical investigation.

5.4. Drugs Designed Based on AI

AI has shown great potential in the design of anti-fibrotic drugs. Through machine learning and deep learning technologies, it can accelerate target discovery, molecular screening and optimization, drug repurposing, and clinical trial design. AI platforms such as Insilico Medicine, BenevolentAI, and Atomwise have made progress in the development of anti-fibrotic drugs. They have identified several potential candidate molecules and optimized their effects through computational simulations and data analysis.
Additionally, AI can be used for drug repurposing, discovering the therapeutic potential of existing drugs for fibrosis. Although this field is still in its early stages, with the continuous development of technology, AI is expected to accelerate the discovery of anti-fibrotic drugs, providing new therapeutic strategies for diseases such as PF. Insilico Medicine has developed a preclinical inhalation formulation candidate, ISM001-055. Inhaled formulations of ISM001-055 achieved high lung exposure and low systemic exposure in preclinical studies and demonstrated anti-fibrotic and anti-inflammatory effects in animal models [170,171]. The ISM001-055 inhalation formulation has good pharmacokinetics and safety, along with good stability and solubility.

5.5. Current Challenges

Despite advancements in PF therapies, challenges remain in improving drug delivery efficiency and overcoming resistance mechanisms. Nanoparticle-based drug delivery to lung tissue is influenced by factors such as particle size and the heterogeneity of fibrotic lung tissue, which can result in uneven drug distribution within the lung [172,173,174]. Improved targeting strategies are needed to enhance distribution and therapeutic efficacy. Drug resistance mechanisms, such as the feedback activation of the Notch-Jagged1 signaling pathway, which can drive persistent fibroblast activation, also hinder treatment success [47,72,175]. Targeting these resistance mechanisms through combination therapy or upstream signaling modulation will be critical to improve therapeutic outcomes. In summary, overcoming challenges related to drug delivery and resistance mechanisms is crucial for the development of more effective treatments for PF.

6. Future Perspectives

To address the challenges in PF therapy, several innovative strategies are emerging. Advanced technological tools, including cost-effective, high-throughput single-cell platforms and AI-driven data analytics, are facilitating large-scale studies and enabling more accurate predictive modeling. AI and machine learning approaches can integrate multi-omics datasets, identify novel therapeutic targets, and predict patient-specific responses. Notably, AI algorithms, such as those utilizing AlphaFold for the identification of Galectin-3 inhibitors, hold considerable promise in targeting critical mediators of fibrosis progression [176,177,178].
In precision drug delivery, microenvironment-responsive nanocarriers and engineered exosomes provide targeted approaches to modulate epithelial cell behavior. ROS-sensitive nanoparticles, such as those loaded with YAP inhibitors, have demonstrated efficacy in preclinical models [179,180,181]. Additionally, combination therapies targeting multiple pathways, such as TGF-β and YAP/TAZ inhibition, can mitigate resistance mechanisms [182,183]. Epigenetic modulation through DNMT or BET inhibitors presents a novel approach to reversing pro-fibrotic changes [112,184,185]. Personalized medicine will benefit from patient stratification based on single-cell and spatial omics data, guiding tailored treatments like Galectin-3 inhibitors for SCGB3A2+ patients. The integration of digital twin models, combining multi-omics and clinical data, may further optimize individualized therapies.
Although AI-driven drug discovery and spatiomics technologies offer powerful new tools for target identification and biomarker studies, they face significant translational hurdles. AI models rely heavily on comprehensive, unbiased training data, which limits their predictive accuracy for rare diseases and underrepresented patient populations [171,186]. Macromolecular and nanoparticle carriers encounter biologic barriers and macrophage clearance during pulmonary delivery, necessitating extensive in vitro and in vivo validation of their safety in accordance with regulatory guidelines [187,188]. At the same time, spatial omics platforms vary widely in resolution, data consistency, and batch effect control, and the high cost of sequencing and imaging limits their use in large patient cohorts [189,190]. In summary, multicenter validation studies, standardized analytical processes, and prospective biomarker-driven clinical trials are essential to translate these promising approaches into individualized therapy.
Interdisciplinary collaboration across bioengineering, data science, and clinical research is crucial for advancing these innovations. Cross-disciplinary efforts, exemplified by organ-on-chip models, can accelerate drug screening, while global consortia such as the Human Cell Atlas (HCA) and LungMAP are pivotal in advancing biomarker discovery and data sharing.

7. Conclusions

This review has comprehensively examined the pivotal role of epithelial cells in the pathogenesis of PF, emphasizing their multifaceted contributions to disease initiation and progression. Compared with the existing literature, our review extends beyond conventional fibrotic signaling pathways by emphasizing the heterogeneity of ECs, including different subpopulations and their specific roles in PF progression. We integrate the functional alterations and potential mechanisms of ECs in the progression of PF with the application of advanced technologies such as single-cell sequencing, spatial transcriptomics, and metabolomics, providing a theoretical basis for better clinical intervention. Additionally, we also explored emerging epithelial-targeted therapies, ranging from FDA-approved agents to novel drugs developed through new technology applications, providing valuable insights into precision anti-fibrosis strategies for clinical translation.
PF is the result of a complex interaction between genetic susceptibility and environmental exposure. Genetic studies have identified rare and common variants, such as mutations in telomerase components and MUC5B promoter polymorphisms, that increase susceptibility to both familial and sporadic disease [191,192,193]. At the same time, environmental factors such as smoking, occupational inhalation of silica or metal dust, pesticides, air pollution, certain drugs, and chest radiation can also lead to repeated epithelial damage and dysregulated repair processes. The role of these factors varies in different patient populations, providing a basis for the heterogeneity in clinical presentation, disease progression, and response to therapy in PF.
Epithelial cells, especially alveolar epithelial cells (AT1 and AT2), serve as both initiators and propagators of fibrosis through mechanisms such as epithelial injury, EMT, oxidative stress, immune cell interactions, and excessive ECM deposition. Dysregulation of these processes, driven by key signaling pathways including TGF-β, Wnt/β-catenin, and Notch, underscores the critical role of epithelial dysfunction in the fibrotic cascade.
Current FDA-approved therapies, such as pirfenidone and nintedanib, have demonstrated efficacy in slowing disease progression by modulating epithelial cell responses and downstream fibrotic pathways. Phase III trials demonstrated that both pirfenidone and nintedanib significantly slowed the annual decline in FVC and reduced the risk of acute exacerbations in patients with IPF [194,195]. In terms of safety, the most common adverse effects of pirfenidone were gastrointestinal symptoms and photosensitivity, while the main adverse effects of ndanib were diarrhea, hypertension and mild elevation of alanine aminotransferase/aspartate aminotransferase [196,197,198]. Small studies have reported reductions in serum Krebs von den Lungen-6 and surfactant protein D with both therapies, but evidence is still limited by small sample sizes and the lack of large prospective cohorts [199,200]. Future studies should focus on biomark-driven personalized dosing strategies to maximize efficacy while minimizing toxicity.
Although both pirfenidone and nintedanib have significant roles in the treatment of PF, their inability to reverse established fibrosis highlights the urgent need for novel therapeutic strategies that target the underlying molecular mechanisms of epithelial dysfunction. Emerging approaches, including inhibitors of TGF-β, Wnt/β-catenin, and Galectin-3, as well as microRNA regulators and exosome-based therapies, show promise in preclinical and early clinical studies. The advent of advanced technologies, such as single-cell sequencing and spatial transcriptomics, has further revealed the heterogeneity and functional diversity of epithelial cells in PF, identifying novel subpopulations and potential therapeutic targets. Additionally, AI-driven drug discovery and precision medicine approaches are poised to accelerate the development of personalized treatments tailored to individual patient profiles.
However, despite the promise of new molecular targets and individualized treatment approaches, preclinical efficacy still relies heavily on rodent models, such as the BLM model. The BLM-induced PF animal model often shows acute inflammation within 1–2 weeks after administration and resolves spontaneously within 3–4 weeks, making it difficult to mimic the chronic and irreversible progression of human IPF. The fibrosis in the model was mostly subpleural patchy, which was significantly different from the typical human usual interstitial pneumonia pattern of honeycomb-like lungs and fibrous foci [201]. In addition, because the experiments were performed in inbred mice between 8 and 12 weeks of age, they do not adequately reflect the advanced age and genetic heterogeneity of IPF patients [202]. To improve translational relevance, humanized precision-cut lung slices can be combined with genetically engineered models to verify key mechanisms and effectiveness.
Despite these advancements, significant challenges remain, including optimizing drug delivery to heterogeneous fibrotic lung tissue, overcoming resistance mechanisms, and translating preclinical findings into effective clinical therapies. Future research should focus on elucidating the spatiotemporal dynamics of epithelial cell behavior in PF, integrating multi-omics data to identify novel biomarkers, and developing combination therapies that target multiple pathways to address resistance and enhance efficacy. Moreover, interdisciplinary collaboration across bioengineering, data science, and clinical research will be essential to bridge the gap between bench and bedside, ultimately improving outcomes for patients with PF.
In conclusion, the heterogeneity of epithelial cells in PF presents both challenges and opportunities for therapeutic intervention. While technological and biological complexities remain significant barriers, emerging innovations in single-cell omics, AI-driven drug design, and precision delivery systems hold great promise. By addressing these challenges through interdisciplinary collaboration and patient-centric approaches, we can unlock the full potential of epithelial cell-targeted therapies, transforming the landscape of PF treatment.

Author Contributions

J.W. wrote the manuscript, edited the drafts, and conceptualized the figures. J.C. designed the study and reviewed and edited the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This work was supported by grants from the National Natural Science Foundation of China (No.82373547), and also supported by Jiangsu Province Science and Technology Plan Project ‘Provincial Frontier Technology R&D Program’ (BF2024054).

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable.

Acknowledgments

Figures were partly created using icons from BioRender.com.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Henderson, N.C.; Rieder, F.; Wynn, T.A. Fibrosis: From mechanisms to medicines. Nature 2020, 587, 555–566. [Google Scholar] [CrossRef] [PubMed]
  2. Wynn, T.A.; Ramalingam, T.R. Mechanisms of fibrosis: Therapeutic translation for fibrotic disease. Nat. Med. 2012, 18, 1028–1040. [Google Scholar] [CrossRef] [PubMed]
  3. Richeldi, L.; Collard, H.R.; Jones, M.G. Idiopathic pulmonary fibrosis. Lancet 2017, 389, 1941–1952. [Google Scholar] [CrossRef]
  4. Moss, B.J.; Ryter, S.W.; Rosas, I.O. Pathogenic Mechanisms Underlying Idiopathic Pulmonary Fibrosis. Annu. Rev. Pathol. 2022, 17, 515–546. [Google Scholar] [CrossRef]
  5. Liu, J.; Wang, F.; Hong, Y.; Luo, F. Bibliometric Analysis of the Pirfenidone and Nintedanib in Interstitial Lung Diseases. Heliyon 2024, 10, e29266. [Google Scholar] [CrossRef]
  6. Chianese, M.; Screm, G.; Salton, F.; Confalonieri, P.; Trotta, L.; Barbieri, M.; Ruggero, L.; Mari, M.; Reccardini, N.; Geri, P.; et al. Pirfenidone and Nintedanib in Pulmonary Fibrosis: Lights and Shadows. Pharmaceuticals 2024, 17, 709. [Google Scholar] [CrossRef]
  7. Reccardini, N.; Chernovsky, M.; Salton, F.; Confalonieri, P.; Mondini, L.; Barbieri, M.; Romallo, A.; Maggisano, M.; Torregiani, C.; Geri, P.; et al. Pirfenidone in Idiopathic Pulmonary Fibrosis: Real-World Observation on Efficacy and Safety, Focus on Patients Undergoing Antithrombotic and Anticoagulant. Pharmaceuticals 2024, 17, 930. [Google Scholar] [CrossRef]
  8. Larsen, S.B.; Cowley, C.J.; Fuchs, E. Epithelial cells: Liaisons of immunity. Curr. Opin. Immunol. 2020, 62, 45–53. [Google Scholar] [CrossRef]
  9. Luo, L.; Zhang, W.; You, S.; Cui, X.; Tu, H.; Yi, Q.; Wu, J.; Liu, O. The role of epithelial cells in fibrosis: Mechanisms and treatment. Pharmacol. Res. 2024, 202, 107144. [Google Scholar] [CrossRef]
  10. Blanpain, C.; Horsley, V.; Fuchs, E. Epithelial stem cells: Turning over new leaves. Cell 2007, 128, 445–458. [Google Scholar] [CrossRef]
  11. Lamouille, S.; Xu, J.; Derynck, R. Molecular mechanisms of epithelial-mesenchymal transition. Nat. Rev. Mol. Cell Biol. 2014, 15, 178–196. [Google Scholar] [CrossRef] [PubMed]
  12. Bracken, C.P.; Goodall, G.J. The many regulators of epithelial-mesenchymal transition. Nat. Rev. Mol. Cell Biol. 2022, 23, 89–90. [Google Scholar] [CrossRef] [PubMed]
  13. Zhao, M.; Wang, L.; Wang, M.; Zhou, S.; Lu, Y.; Cui, H.; Racanelli, A.C.; Zhang, L.; Ye, T.; Ding, B. Targeting fibrosis: Mechanisms and clinical trials. Signal Transduct. Target. Ther. 2022, 7, 206. [Google Scholar] [CrossRef]
  14. Kalluri, R.; Weinberg, R.A. The basics of epithelial-mesenchymal transition. J. Clin. Invest. 2009, 119, 1420–1428. [Google Scholar] [CrossRef]
  15. Shao, M.; Cheng, H.; Li, X.; Qiu, Y.; Zhang, Y.; Chang, Y.; Fu, J.; Shen, M.; Xu, X.; Feng, D.; et al. Abnormal mitochondrial iron metabolism damages alveolar type II epithelial cells involved in bleomycin-induced pulmonary fibrosis. Theranostics 2024, 14, 2687–2705. [Google Scholar] [CrossRef]
  16. Lovisa, S.; LeBleu, V.S.; Tampe, B.; Sugimoto, H.; Vadnagara, K.; Carstens, J.L.; Wu, C.C.; Hagos, Y.; Burckhardt, B.C.; Pentcheva-Hoang, T.; et al. Epithelial-to-mesenchymal transition induces cell cycle arrest and parenchymal damage in renal fibrosis. Nat. Med. 2015, 21, 998–1009. [Google Scholar] [CrossRef]
  17. Selman, M.; Pardo, A. The leading role of epithelial cells in the pathogenesis of idiopathic pulmonary fibrosis. Cell. Signal. 2020, 66, 109482. [Google Scholar] [CrossRef]
  18. Chambers, R.C.; Mercer, P.F. Mechanisms of alveolar epithelial injury, repair, and fibrosis. Ann. Am. Thorac. Soc. 2015, 12 (Suppl. S1), S16–S20. [Google Scholar] [CrossRef]
  19. Pydyn, N.; Ferenc, A.; Trzos, K.; Pospiech, E.; Wilamowski, M.; Mucha, O.; Major, P.; Kadluczka, J.; Rodrigues, P.M.; Banales, J.M.; et al. MCPIP1 Inhibits Hepatic Stellate Cell Activation in Autocrine and Paracrine Manners, Preventing Liver Fibrosis. Cell. Mol. Gastroenterol. Hepatol. 2024, 17, 887–906. [Google Scholar] [CrossRef]
  20. Yang, J.; Velikoff, M.; Canalis, E.; Horowitz, J.C.; Kim, K.K. Activated alveolar epithelial cells initiate fibrosis through autocrine and paracrine secretion of connective tissue growth factor. Am. J. Physiol. Lung Cell. Mol. Physiol. 2014, 306, L786–L796. [Google Scholar] [CrossRef]
  21. Sakai, N.; Tager, A.M. Fibrosis of two: Epithelial cell-fibroblast interactions in pulmonary fibrosis. Biochim. Biophys. Acta 2013, 1832, 911–921. [Google Scholar] [CrossRef] [PubMed]
  22. Foerster, E.G.; Mukherjee, T.; Cabral-Fernandes, L.; Rocha, J.D.; Girardin, S.E.; Philpott, D.J. How autophagy controls the intestinal epithelial barrier. Autophagy 2022, 18, 86–103. [Google Scholar] [CrossRef] [PubMed]
  23. Kim, K.K.; Dotson, M.R.; Agarwal, M.; Yang, J.; Bradley, P.B.; Subbotina, N.; Osterholzer, J.J.; Sisson, T.H. Efferocytosis of apoptotic alveolar epithelial cells is sufficient to initiate lung fibrosis. Cell Death Dis. 2018, 9, 1056. [Google Scholar] [CrossRef] [PubMed]
  24. Nakanuma, Y.; Sasaki, M.; Harada, K. Autophagy and senescence in fibrosing cholangiopathies. J. Hepatol. 2015, 62, 934–945. [Google Scholar] [CrossRef]
  25. Lee, J.H.; Massagué, J. TGF-β in developmental and fibrogenic EMTs. Semin. Cancer Biol. 2022, 86, 136–145. [Google Scholar] [CrossRef]
  26. Xu, J.; Lamouille, S.; Derynck, R. TGF-beta-induced epithelial to mesenchymal transition. Cell Res. 2009, 19, 156–172. [Google Scholar] [CrossRef]
  27. Bonnans, C.; Chou, J.; Werb, Z. Remodelling the extracellular matrix in development and disease. Nat. Rev. Mol. Cell Biol. 2014, 15, 786–801. [Google Scholar] [CrossRef]
  28. Guo, T.; He, C.; Venado, A.; Zhou, Y. Extracellular Matrix Stiffness in Lung Health and Disease. Compr. Physiol. 2022, 12, 3523–3558. [Google Scholar] [CrossRef]
  29. Kong, D.; Zhang, Z.; Chen, L.; Huang, W.; Zhang, F.; Wang, L.; Wang, Y.; Cao, P.; Zheng, S. Curcumin blunts epithelial-mesenchymal transition of hepatocytes to alleviate hepatic fibrosis through regulating oxidative stress and autophagy. Redox Biol. 2020, 36, 101600. [Google Scholar] [CrossRef]
  30. Louzada, R.A.; Corre, R.; Ameziane El Hassani, R.; Meziani, L.; Jaillet, M.; Cazes, A.; Crestani, B.; Deutsch, E.; Dupuy, C. NADPH oxidase DUOX1 sustains TGF-beta1 signalling and promotes lung fibrosis. Eur. Respir. J. 2021, 57, 1901949. [Google Scholar] [CrossRef]
  31. Luchian, I.; Goriuc, A.; Sandu, D.; Covasa, M. The Role of Matrix Metalloproteinases (MMP-8, MMP-9, MMP-13) in Periodontal and Peri-Implant Pathological Processes. Int. J. Mol. Sci. 2022, 23, 1806. [Google Scholar] [CrossRef] [PubMed]
  32. Carnesecchi, S.; Deffert, C.; Donati, Y.; Basset, O.; Hinz, B.; Preynat-Seauve, O.; Guichard, C.; Arbiser, J.L.; Banfi, B.; Pache, J.C.; et al. A key role for NOX4 in epithelial cell death during development of lung fibrosis. Antioxid. Redox Signal 2011, 15, 607–619. [Google Scholar] [CrossRef] [PubMed]
  33. Gonzalez-Gonzalez, F.J.; Chandel, N.S.; Jain, M.; Budinger, G.S. Reactive oxygen species as signaling molecules in the development of lung fibrosis. Transl. Res. 2017, 190, 61–68. [Google Scholar] [CrossRef]
  34. Jiang, F.; Liu, G.-S.; Dusting, G.J.; Chan, E.C. NADPH oxidase-dependent redox signaling in TGF-β-mediated fibrotic responses. Redox Biol. 2014, 2, 267–272. [Google Scholar] [CrossRef]
  35. Richter, K.; Konzack, A.; Pihlajaniemi, T.; Heljasvaara, R.; Kietzmann, T. Redox-fibrosis: Impact of TGFβ1 on ROS generators, mediators and functional consequences. Redox Biol. 2015, 6, 344–352. [Google Scholar] [CrossRef]
  36. Hecker, L.; Vittal, R.; Jones, T.; Jagirdar, R.; Luckhardt, T.R.; Horowitz, J.C.; Pennathur, S.; Martinez, F.J.; Thannickal, V.J. NADPH oxidase-4 mediates myofibroblast activation and fibrogenic responses to lung injury. Nat. Med. 2009, 15, 1077–1081. [Google Scholar] [CrossRef]
  37. Kato, K.; Hecker, L. NADPH oxidases: Pathophysiology and therapeutic potential in age-associated pulmonary fibrosis. Redox Biol. 2020, 33, 101541. [Google Scholar] [CrossRef]
  38. Sanders, Y.Y.; Liu, H.; Liu, G.; Thannickal, V.J. Epigenetic mechanisms regulate NADPH oxidase-4 expression in cellular senescence. Free Radic. Biol. Med. 2015, 79, 197–205. [Google Scholar] [CrossRef]
  39. Koyama, Y.; Brenner, D.A. Liver inflammation and fibrosis. J. Clin. Invest. 2017, 127, 55–64. [Google Scholar] [CrossRef]
  40. Bian, F.; Lan, Y.-W.; Zhao, S.; Deng, Z.; Shukla, S.; Acharya, A.; Donovan, J.; Le, T.; Milewski, D.; Bacchetta, M. Lung endothelial cells regulate pulmonary fibrosis through FOXF1/R-Ras signaling. Nat. Commun. 2023, 14, 2560. [Google Scholar] [CrossRef]
  41. Budi, E.H.; Schaub, J.R.; Decaris, M.; Turner, S.; Derynck, R. TGF-β as a driver of fibrosis: Physiological roles and therapeutic opportunities. J. Pathol. 2021, 254, 358–373. [Google Scholar] [CrossRef] [PubMed]
  42. Frangogiannis, N.G. Transforming growth factor–β in tissue fibrosis. J. Exp. Med. 2020, 217, e20190103. [Google Scholar] [CrossRef] [PubMed]
  43. Chen, M.; Wang, J.; Yuan, M.; Long, M.; Sun, Y.; Wang, S.; Luo, W.; Zhou, Y.; Zhang, W.; Jiang, W.; et al. AT2 cell-derived IgA trapped by the extracellular matrix in silica-induced pulmonary fibrosis. Int. Immunopharmacol. 2023, 122, 110545. [Google Scholar] [CrossRef]
  44. Craig, V.J.; Zhang, L.; Hagood, J.S.; Owen, C.A. Matrix metalloproteinases as therapeutic targets for idiopathic pulmonary fibrosis. Am. J. Respir. Cell Mol. Biol. 2015, 53, 585–600. [Google Scholar] [CrossRef]
  45. Brown, A.C.; Fiore, V.F.; Sulchek, T.A.; Barker, T.H. Physical and chemical microenvironmental cues orthogonally control the degree and duration of fibrosis-associated epithelial-to-mesenchymal transitions. J. Pathol. 2013, 229, 25–35. [Google Scholar] [CrossRef]
  46. Fiore, V.F.; Wong, S.S.; Tran, C.; Tan, C.; Xu, W.; Sulchek, T.; White, E.S.; Hagood, J.S.; Barker, T.H. alphavbeta3 Integrin drives fibroblast contraction and strain stiffening of soft provisional matrix during progressive fibrosis. JCI Insight 2018, 3, 97597. [Google Scholar] [CrossRef]
  47. Younesi, F.S.; Miller, A.E.; Barker, T.H.; Rossi, F.M.; Hinz, B. Fibroblast and myofibroblast activation in normal tissue repair and fibrosis. Nat. Rev. Mol. Cell Biol. 2024, 25, 617–638. [Google Scholar] [CrossRef]
  48. Prabhu, S.D.; Frangogiannis, N.G. The Biological Basis for Cardiac Repair After Myocardial Infarction: From Inflammation to Fibrosis. Circ. Res. 2016, 119, 91–112. [Google Scholar] [CrossRef]
  49. Chen, W.; Bian, W.; Zhou, Y.; Zhang, J. Cardiac Fibroblasts and Myocardial Regeneration. Front. Bioeng. Biotechnol. 2021, 9, 599928. [Google Scholar] [CrossRef]
  50. Peng, D.; Fu, M.; Wang, M.; Wei, Y.; Wei, X. Targeting TGF-beta signal transduction for fibrosis and cancer therapy. Mol. Cancer 2022, 21, 104. [Google Scholar] [CrossRef]
  51. Hu, H.-H.; Chen, D.-Q.; Wang, Y.-N.; Feng, Y.-L.; Cao, G.; Vaziri, N.D.; Zhao, Y.-Y. New insights into TGF-β/Smad signaling in tissue fibrosis. Chem.-Biol. Interact. 2018, 292, 76–83. [Google Scholar] [CrossRef] [PubMed]
  52. Kramer, E.L.; Clancy, J.P. TGFβ as a therapeutic target in cystic fibrosis. Expert Opin. Ther. Targets 2018, 22, 177–189. [Google Scholar] [CrossRef] [PubMed]
  53. Zhang, Y.E. Non-Smad pathways in TGF-beta signaling. Cell Res. 2009, 19, 128–139. [Google Scholar] [CrossRef] [PubMed]
  54. Muthusamy, B.P.; Budi, E.H.; Katsuno, Y.; Lee, M.K.; Smith, S.M.; Mirza, A.M.; Akhurst, R.J.; Derynck, R. ShcA protects against epithelial–mesenchymal transition through compartmentalized inhibition of TGF-β-induced Smad activation. PLoS Biol. 2015, 13, e1002325. [Google Scholar] [CrossRef]
  55. Hu, L.; Ding, M.; He, W. Emerging Therapeutic Strategies for Attenuating Tubular EMT and Kidney Fibrosis by Targeting Wnt/beta-Catenin Signaling. Front. Pharmacol. 2021, 12, 830340. [Google Scholar]
  56. Schunk, S.J.; Floege, J.; Fliser, D.; Speer, T. WNT–β-catenin signalling—A versatile player in kidney injury and repair. Nat. Rev. Nephrol. 2021, 17, 172–184. [Google Scholar] [CrossRef]
  57. Tian, Y.; Xia, J.; Yang, G.; Li, C.; Qi, Y.; Dai, K.; Wu, C.; Guo, Y.; Yao, W.; Hao, C. A2aR inhibits fibrosis and the EMT process in silicosis by regulating Wnt/beta-catenin pathway. Ecotoxicol. Environ. Saf. 2023, 249, 114410. [Google Scholar] [CrossRef]
  58. Yiu, W.H.; Li, Y.; Lok, S.W.Y.; Chan, K.W.; Chan, L.Y.Y.; Leung, J.C.K.; Lai, K.N.; Tsu, J.H.L.; Chao, J.; Huang, X.R.; et al. Protective role of kallistatin in renal fibrosis via modulation of Wnt/beta-catenin signaling. Clin. Sci. 2021, 135, 429–446. [Google Scholar] [CrossRef]
  59. Yang, Z.; Wang, M.; Ren, Y.; Li, L.; Cao, L.; Zhang, W.; Lv, K.; Sun, Z.; Nie, S. Inhibition of Wnt10b/beta-catenin signaling alleviates pulmonary fibrogenesis induced by paraquat in vivo and in vitro. Life Sci. 2021, 286, 120027. [Google Scholar] [CrossRef]
  60. Huang, R.; Bai, C.; Liu, X.; Zhou, Y.; Hu, S.; Li, D.; Xiang, J.; Chen, J.; Zhou, P. The p53/RMRP/miR122 signaling loop promotes epithelial-mesenchymal transition during the development of silica-induced lung fibrosis by activating the notch pathway. Chemosphere 2021, 263, 128133. [Google Scholar] [CrossRef]
  61. Feng, Y.L.; Chen, D.Q.; Vaziri, N.D.; Guo, Y.; Zhao, Y.Y. Small molecule inhibitors of epithelial-mesenchymal transition for the treatment of cancer and fibrosis. Med. Res. Rev. 2020, 40, 54–78. [Google Scholar] [CrossRef] [PubMed]
  62. Wasnick, R.; Korfei, M.; Piskulak, K.; Henneke, I.; Wilhelm, J.; Mahavadi, P.; Dartsch, R.C.; von der Beck, D.; Koch, M.; Shalashova, I.; et al. Notch1 Induces Defective Epithelial Surfactant Processing and Pulmonary Fibrosis. Am. J. Respir. Crit. Care Med. 2023, 207, 283–299. [Google Scholar] [CrossRef] [PubMed]
  63. Zhang, X.; Xu, Z.; Chen, Q.; Zhou, Z. Notch signaling regulates pulmonary fibrosis. Front. Cell Dev. Biol. 2024, 12, 1450038. [Google Scholar] [CrossRef]
  64. Vera, L.; Garcia-Olloqui, P.; Petri, E.; Vinado, A.C.; Valera, P.S.; Blasco-Iturri, Z.; Calvo, I.A.; Cenzano, I.; Ruppert, C.; Zulueta, J.J.; et al. Notch3 Deficiency Attenuates Pulmonary Fibrosis and Impedes Lung-Function Decline. Am. J. Respir. Cell Mol. Biol. 2021, 64, 465–476. [Google Scholar] [CrossRef]
  65. Xie, T.; Wang, Y.; Deng, N.; Huang, G.; Taghavifar, F.; Geng, Y.; Liu, N.; Kulur, V.; Yao, C.; Chen, P.; et al. Single-Cell Deconvolution of Fibroblast Heterogeneity in Mouse Pulmonary Fibrosis. Cell Rep. 2018, 22, 3625–3640. [Google Scholar] [CrossRef]
  66. Liu, T.; Gonzalez De Los Santos, F.; Hirsch, M.; Wu, Z.; Phan, S.H. Noncanonical Wnt Signaling Promotes Myofibroblast Differentiation in Pulmonary Fibrosis. Am. J. Respir. Cell Mol. Biol. 2021, 65, 489–499. [Google Scholar] [CrossRef]
  67. Zhou, B.; Liu, Y.; Kahn, M.; Ann, D.K.; Han, A.; Wang, H.; Nguyen, C.; Flodby, P.; Zhong, Q.; Krishnaveni, M.S. 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. 2012, 287, 7026–7038. [Google Scholar] [CrossRef]
  68. Tian, X.; Zhang, J.; Tan, T.K.; Lyons, J.G.; Zhao, H.; Niu, B.; Lee, S.R.; Tsatralis, T.; Zhao, Y.; Wang, Y. Association of β-catenin with P-Smad3 but not LEF-1 dissociates in vitro profibrotic from anti-inflammatory effects of TGF-β1. J. Cell Sci. 2013, 126, 67–76. [Google Scholar] [CrossRef]
  69. Wang, X.C.; Song, K.; Tu, B.; Sun, H.; Zhou, Y.; Xu, S.S.; Lu, D.; Sha, J.M.; Tao, H. New aspects of the epigenetic regulation of EMT related to pulmonary fibrosis. Eur. J. Pharmacol. 2023, 956, 175959. [Google Scholar] [CrossRef]
  70. Bakalenko, N.; Kuznetsova, E.; Malashicheva, A. The Complex Interplay of TGF-β and Notch Signaling in the Pathogenesis of Fibrosis. Int. J. Mol. Sci. 2024, 25, 10803. [Google Scholar] [CrossRef]
  71. Aoyagi-Ikeda, K.; Maeno, T.; Matsui, H.; Ueno, M.; Hara, K.; Aoki, Y.; Aoki, F.; Shimizu, T.; Doi, H.; Kawai-Kowase, K.; et al. Notch induces myofibroblast differentiation of alveolar epithelial cells via transforming growth factor-beta-Smad3 pathway. Am. J. Respir. Cell Mol. Biol. 2011, 45, 136–144. [Google Scholar]
  72. Hu, B.; Phan, S.H. Notch in fibrosis and as a target of anti-fibrotic therapy. Pharmacol. Res. 2016, 108, 57–64. [Google Scholar] [CrossRef]
  73. Liu, T.; Hu, B.; Choi, Y.Y.; Chung, M.; Ullenbruch, M.; Yu, H.; Lowe, J.B.; Phan, S.H. Notch1 signaling in FIZZ1 induction of myofibroblast differentiation. Am. J. Pathol. 2009, 174, 1745–1755. [Google Scholar] [CrossRef]
  74. Veith, C.; Hristova, M.; Danyal, K.; Habibovic, A.; Dustin, C.M.; McDonough, J.E.; Vanaudenaerde, B.M.; Kreuter, M.; Schneider, M.A.; Kahn, N.; et al. Profibrotic epithelial TGF-beta1 signaling involves NOX4-mitochondria cross talk and redox-mediated activation of the tyrosine kinase FYN. Am. J. Physiol. Lung Cell Mol. Physiol. 2021, 320, L356–L367. [Google Scholar] [CrossRef]
  75. Ohkouchi, S.; Kanehira, M.; Saigusa, D.; Ono, M.; Tazawa, R.; Terunuma, H.; Hirano, T.; Numakura, T.; Notsuda, H.; Inoue, C.; et al. Metabolic and Epigenetic Regulation of SMAD7 by STC1 Ameliorates Lung Fibrosis. Am. J. Respir. Cell Mol. Biol. 2022, 67, 320–333. [Google Scholar] [CrossRef]
  76. Liu, R.M.; Desai, L.P. Reciprocal regulation of TGF-beta and reactive oxygen species: A perverse cycle for fibrosis. Redox Biol. 2015, 6, 565–577. [Google Scholar] [CrossRef]
  77. Shu, Y.; Liu, Y.; Li, X.; Cao, L.; Yuan, X.; Li, W.; Cao, Q. Aspirin-triggered resolvin D1 inhibits TGF-β1-induced EndMT through increasing the expression of smad7 and is closely related to oxidative stress. Biomol. Ther. 2016, 24, 132. [Google Scholar] [CrossRef]
  78. Lian, Z.; Kuerban, R.; Niu, Z.; Aisaiti, P.; Wu, C.; Yang, X. Notch Signaling Is Associated with Pulmonary Fibrosis in Patients with Pigeon Breeder’s Lung by Regulating Oxidative Stress. Emerg. Med. Int. 2024, 2024, 7610032. [Google Scholar] [CrossRef]
  79. 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]
  80. Song, L.; Li, K.; Chen, H.; Xie, L. Cell Cross-Talk in Alveolar Microenvironment: From Lung Injury to Fibrosis. Am. J. Respir. Cell Mol. Biol. 2024, 71, 30–42. [Google Scholar] [CrossRef]
  81. Chen, J.; Ge, S.J.; Feng, H.J.; Wu, S.Z.; Ji, R.; Huang, W.R.; Huang, W.; Lu, C.H. KRT17 Promotes the Activation of HSCs via EMT in Liver Fibrosis. J. Clin. Transl. Hepatol. 2022, 10, 207–218. [Google Scholar] [CrossRef]
  82. Fu, M.; Hu, Y.; Lan, T.; Guan, K.L.; Luo, T.; Luo, M. The Hippo signalling pathway and its implications in human health and diseases. Signal Transduct. Target. Ther. 2022, 7, 376. [Google Scholar] [CrossRef]
  83. Dupont, S.; Morsut, L.; Aragona, M.; Enzo, E.; Giulitti, S.; Cordenonsi, M.; Zanconato, F.; Le Digabel, J.; Forcato, M.; Bicciato, S.; et al. Role of YAP/TAZ in mechanotransduction. Nature 2011, 474, 179–183. [Google Scholar] [CrossRef]
  84. Szeto, S.G.; Narimatsu, M.; Lu, M.; He, X.; Sidiqi, A.M.; Tolosa, M.F.; Chan, L.; De Freitas, K.; Bialik, J.F.; Majumder, S.; et al. YAP/TAZ Are Mechanoregulators of TGF-beta-Smad Signaling and Renal Fibrogenesis. J. Am. Soc. Nephrol. 2016, 27, 3117–3128. [Google Scholar] [CrossRef]
  85. O′Hagan, H.M.; Wang, W.; Sen, S.; Destefano Shields, C.; Lee, S.S.; Zhang, Y.W.; Clements, E.G.; Cai, Y.; Van Neste, L.; Easwaran, H.; et al. Oxidative damage targets complexes containing DNA methyltransferases, SIRT1, and polycomb members to promoter CpG Islands. Cancer Cell 2011, 20, 606–619. [Google Scholar] [CrossRef]
  86. Xue, T.; Qiu, X.; Liu, H.; Gan, C.; Tan, Z.; Xie, Y.; Wang, Y.; Ye, T. Epigenetic regulation in fibrosis progress. Pharmacol. Res. 2021, 173, 105910. [Google Scholar] [CrossRef]
  87. Qu, L.; Yin, T.; Zhao, Y.; Lv, W.; Liu, Z.; Chen, C.; Liu, K.; Shan, S.; Zhou, R.; Li, X.; et al. Histone demethylases in the regulation of immunity and inflammation. Cell Death Discov. 2023, 9, 188. [Google Scholar] [CrossRef]
  88. Xie, N.; Tan, Z.; Banerjee, S.; Cui, H.; Ge, J.; Liu, R.M.; Bernard, K.; Thannickal, V.J.; Liu, G. Glycolytic Reprogramming in Myofibroblast Differentiation and Lung Fibrosis. Am. J. Respir Crit. Care Med. 2015, 192, 1462–1474. [Google Scholar] [CrossRef]
  89. Yang, H.-L.; Chang, C.-W.; Vadivalagan, C.; Pandey, S.; Chen, S.-J.; Lee, C.-C.; Hseu, J.-H.; Hseu, Y.-C. Coenzyme Q0 inhibited the NLRP3 inflammasome, metastasis/EMT, and Warburg effect by suppressing hypoxia-induced HIF-1α expression in HNSCC cells. Int. J. Biol. Sci. 2024, 20, 2790. [Google Scholar] [CrossRef]
  90. Fazio, A.; Bordoni, D.; Kuiper, J.W.P.; Weber-Stiehl, S.; Stengel, S.T.; Arnold, P.; Ellinghaus, D.; Ito, G.; Tran, F.; Messner, B.; et al. DNA methyltransferase 3A controls intestinal epithelial barrier function and regeneration in the colon. Nat. Commun. 2022, 13, 6266. [Google Scholar] [CrossRef]
  91. Boo, H.J.; Min, H.Y.; Park, C.S.; Park, J.S.; Jeong, J.Y.; Lee, S.Y.; Kim, W.Y.; Lee, J.W.; Oh, S.R.; Park, R.W.; et al. Dual Impact of IGF2 on Alveolar Stem Cell Function during Tobacco-Induced Injury Repair and Development of Pulmonary Emphysema and Cancer. Cancer Res. 2023, 83, 1782–1799. [Google Scholar] [CrossRef] [PubMed]
  92. Kim, M.K.; Kim, E.J.; Cheng, Y.; Shin, M.H.; Oh, J.H.; Lee, D.H.; Chung, J.H. Inhibition of DNA Methylation in the COL1A2 Promoter by Anacardic Acid Prevents UV-Induced Decrease of Type I Procollagen Expression. J. Invest. Dermatol. 2017, 137, 1343–1352. [Google Scholar] [CrossRef] [PubMed]
  93. Liu, H.; Hong, Y.; Chen, H.; Wang, X.; Dong, J.; Li, X.; Shi, Z.; Zhao, Q.; Zhou, L.; Wang, J.; et al. Dual activation of GCGR/GLP1R signaling ameliorates intestinal fibrosis via metabolic regulation of histone H3K9 lactylation in epithelial cells. Acta Pharm. Sin. B 2025, 15, 278–295. [Google Scholar] [CrossRef] [PubMed]
  94. Macias-Ceja, D.C.; Ortiz-Masia, D.; Salvador, P.; Gisbert-Ferrandiz, L.; Hernandez, C.; Hausmann, M.; Rogler, G.; Esplugues, J.V.; Hinojosa, J.; Alos, R.; et al. Succinate receptor mediates intestinal inflammation and fibrosis. Mucosal Immunol. 2019, 12, 178–187. [Google Scholar] [CrossRef]
  95. Reyfman, P.A.; Walter, J.M.; Joshi, N.; Anekalla, K.R.; McQuattie-Pimentel, A.C.; Chiu, S.; Fernandez, R.; Akbarpour, M.; Chen, C.-I.; Ren, Z. Single-cell transcriptomic analysis of human lung provides insights into the pathobiology of pulmonary fibrosis. Am. J. Respir. Crit. Care Med. 2019, 199, 1517–1536. [Google Scholar] [CrossRef]
  96. Strunz, M.; Simon, L.M.; Ansari, M.; Mattner, L.F.; Angelidis, I.; Mayr, C.H.; Kathiriya, J.; Yee, M.; Ogar, P.; Sengupta, A. Longitudinal single cell transcriptomics reveals Krt8+ alveolar epithelial progenitors in lung regeneration. bioRxiv 2019. [Google Scholar] [CrossRef]
  97. Kobayashi, Y.; Tata, A.; Konkimalla, A.; Katsura, H.; Lee, R.F.; Ou, J.; Banovich, N.E.; Kropski, J.A.; Tata, P.R. Persistence of a regeneration-associated, transitional alveolar epithelial cell state in pulmonary fibrosis. Nat. Cell Biol. 2020, 22, 934–946. [Google Scholar] [CrossRef]
  98. Confalonieri, P.; Volpe, M.C.; Jacob, J.; Maiocchi, S.; Salton, F.; Ruaro, B.; Confalonieri, M.; Braga, L. Regeneration or Repair? The Role of Alveolar Epithelial Cells in the Pathogenesis of Idiopathic Pulmonary Fibrosis (IPF). Cells 2022, 11, 2095. [Google Scholar] [CrossRef]
  99. Liew, F.Y.; Girard, J.P.; Turnquist, H.R. Interleukin-33 in health and disease. Nat. Rev. Immunol. 2016, 16, 676–689. [Google Scholar] [CrossRef]
  100. Adams, T.S.; Schupp, J.C.; Poli, S.; Ayaub, E.A.; Neumark, N.; Ahangari, F.; Chu, S.G.; Raby, B.A.; DeIuliis, G.; Januszyk, M.; et al. Single-cell RNA-seq reveals ectopic and aberrant lung-resident cell populations in idiopathic pulmonary fibrosis. Sci. Adv. 2020, 6, eaba1983. [Google Scholar] [CrossRef]
  101. Strunz, M.; Simon, L.M.; Ansari, M.; Kathiriya, J.J.; Angelidis, I.; Mayr, C.H.; Tsidiridis, G.; Lange, M.; Mattner, L.F.; Yee, M. Alveolar regeneration through a Krt8+ transitional stem cell state that persists in human lung fibrosis. Nat. Commun. 2020, 11, 3559. [Google Scholar] [CrossRef] [PubMed]
  102. Wang, F.; Ting, C.; Riemondy, K.A.; Douglas, M.; Foster, K.; Patel, N.; Kaku, N.; Linsalata, A.; Nemzek, J.; Varisco, B.M.; et al. Regulation of epithelial transitional states in murine and human pulmonary fibrosis. J. Clin. Invest. 2023, 133, 165612. [Google Scholar] [CrossRef] [PubMed]
  103. Jin, C.; Chen, Y.; Wang, Y.; Li, J.; Liang, J.; Zheng, S.; Zhang, L.; Li, Q.; Wang, Y.; Ling, F.; et al. Single-cell RNA sequencing reveals special basal cells and fibroblasts in idiopathic pulmonary fibrosis. Sci. Rep. 2024, 14, 15778. [Google Scholar] [CrossRef] [PubMed]
  104. Jaeger, B.; Schupp, J.C.; Plappert, L.; Terwolbeck, O.; Artysh, N.; Kayser, G.; Engelhard, P.; Adams, T.S.; Zweigerdt, R.; Kempf, H. Airway basal cells show a dedifferentiated KRT17highPhenotype and promote fibrosis in idiopathic pulmonary fibrosis. Nat. Commun. 2022, 13, 5637. [Google Scholar] [CrossRef]
  105. Hewitt, R.J.; Puttur, F.; Gaboriau, D.C.A.; Fercoq, F.; Fresquet, M.; Traves, W.J.; Yates, L.L.; Walker, S.A.; Molyneaux, P.L.; Kemp, S.V.; et al. Lung extracellular matrix modulates KRT5(+) basal cell activity in pulmonary fibrosis. Nat. Commun. 2023, 14, 6039. [Google Scholar] [CrossRef]
  106. Bernau, K.; Sandbo, N.; Brasier, A.R. Epigenetics of the pathogenic myofibroblast in lung disease. In Epigenetics in Organ Specific Disorders; Elsevier: Amsterdam, The Netherlands, 2023; pp. 353–392. [Google Scholar]
  107. Huang, G.; Liang, J.; Huang, K.; Liu, X.; Taghavifar, F.; Yao, C.; Parimon, T.; Liu, N.; Dai, K.; Aziz, A. Basal cell–derived WNT7A promotes fibrogenesis at the fibrotic niche in idiopathic pulmonary fibrosis. Am. J. Respir. Cell Mol. Biol. 2023, 68, 302–313. [Google Scholar] [CrossRef]
  108. Tan, C.; Zhou, H.; Xiong, Q.; Xian, X.; Liu, Q.; Zhang, Z.; Xu, J.; Yao, H. Cromolyn sodium reduces LPS-induced pulmonary fibrosis by inhibiting the EMT process enhanced by MC-derived IL-13. Respir. Res. 2025, 26, 3. [Google Scholar] [CrossRef]
  109. Stokes, K.; LaMarche, N.M.; Islam, N.; Wood, A.; Huang, W.; August, A. Cutting edge: STAT6 signaling in eosinophils is necessary for development of allergic airway inflammation. J. Immunol. 2015, 194, 2477–2481. [Google Scholar] [CrossRef]
  110. Kwon, S.; Chung, E.J.; Kc, S.; White, A.O.; Chung, S.I.; Horton, J.A.; Yun, H.S.; Ahn, H.; Shankavaram, U.; Chung, J.Y.; et al. Interleukin-13 Receptor Subunit Alpha 2 Induces Chemokine Expression and Macrophage Polarization to Promote Inflammation and Fibrosis. Int. J. Radiat. Oncol. Biol. Phys. 2025, 121, 1258–1270. [Google Scholar] [CrossRef]
  111. Chakraborty, A.; Mastalerz, M.; Ansari, M.; Schiller, H.B.; Staab-Weijnitz, C.A. Emerging Roles of Airway Epithelial Cells in Idiopathic Pulmonary Fibrosis. Cells 2022, 11, 1050. [Google Scholar] [CrossRef]
  112. Ren, L.; Chang, Y.F.; Jiang, S.H.; Li, X.H.; Cheng, H.P. DNA methylation modification in Idiopathic pulmonary fibrosis. Front. Cell Dev. Biol. 2024, 12, 1416325. [Google Scholar] [CrossRef] [PubMed]
  113. Huan, C.; Yang, T.; Liang, J.; Xie, T.; Cheng, L.; Liu, N.; Kurkciyan, A.; Monterrosa Mena, J.; Wang, C.; Dai, H.; et al. Methylation-mediated BMPER expression in fibroblast activation in vitro and lung fibrosis in mice in vivo. Sci. Rep. 2015, 5, 14910. [Google Scholar] [CrossRef] [PubMed]
  114. Wei, A.; Gao, Q.; Chen, F.; Zhu, X.; Chen, X.; Zhang, L.; Su, X.; Dai, J.; Shi, Y.; Cao, W. Inhibition of DNA methylation de-represses peroxisome proliferator-activated receptor-gamma and attenuates pulmonary fibrosis. Br. J. Pharmacol. 2022, 179, 1304–1318. [Google Scholar] [CrossRef]
  115. Mayr, C.H.; Santacruz, D.; Jarosch, S.; Bleck, M.; Dalton, J.; McNabola, A.; Lempp, C.; Neubert, L.; Rath, B.; Kamp, J.C.; et al. Spatial transcriptomic characterization of pathologic niches in IPF. Sci. Adv. 2024, 10, eadl5473. [Google Scholar] [CrossRef]
  116. Yang, J.; Pan, X.; Wang, L.; Yu, G. Alveolar cells under mechanical stressed niche: Critical contributors to pulmonary fibrosis. Mol. Med. 2020, 26, 1–10. [Google Scholar] [CrossRef]
  117. Warren, R.; Lyu, H.; Klinkhammer, K.; De Langhe, S.P. Hippo signaling impairs alveolar epithelial regeneration in pulmonary fibrosis. Elife 2023, 12, e85092. [Google Scholar] [CrossRef]
  118. He, Y.; Han, Y.; Zou, L.; Yao, T.; Zhang, Y.; Lv, X.; Jiang, M.; Long, L.; Li, M.; Cheng, X. Succinate promotes pulmonary fibrosis through GPR91 and predicts death in idiopathic pulmonary fibrosis. Sci. Rep. 2024, 14, 14376. [Google Scholar] [CrossRef]
  119. Li, J.; Zhai, X.; Sun, X.; Cao, S.; Yuan, Q.; Wang, J. Metabolic reprogramming of pulmonary fibrosis. Front. Pharmacol. 2022, 13, 1031890. [Google Scholar] [CrossRef]
  120. Wang, Z.; Chen, L.; Huang, Y.; Luo, M.; Wang, H.; Jiang, Z.; Zheng, J.; Yang, Z.; Chen, Z.; Zhang, C.; et al. Pharmaceutical targeting of succinate dehydrogenase in fibroblasts controls bleomycin-induced lung fibrosis. Redox Biol. 2021, 46, 102082. [Google Scholar] [CrossRef]
  121. Tannahill, G.; Curtis, A.; Adamik, J.; Palsson-McDermott, E.; McGettrick, A.; Goel, G.; Frezza, C.; Bernard, N.; Kelly, B.; Foley, N. Succinate is an inflammatory signal that induces IL-1β through HIF-1α. Nature 2013, 496, 238–242. [Google Scholar] [CrossRef]
  122. Zhang, D.; Tang, Z.; Huang, H.; Zhou, G.; Cui, C.; Weng, Y.; Liu, W.; Kim, S.; Lee, S.; Perez-Neut, M.; et al. Metabolic regulation of gene expression by histone lactylation. Nature 2019, 574, 575–580. [Google Scholar] [CrossRef] [PubMed]
  123. Cui, H.; Xie, N.; Banerjee, S.; Ge, J.; Jiang, D.; Dey, T.; Matthews, Q.L.; Liu, R.-M.; Liu, G. Lung myofibroblasts promote macrophage profibrotic activity through lactate-induced histone lactylation. Am. J. Respir. Cell Mol. Biol. 2021, 64, 115–125. [Google Scholar] [CrossRef] [PubMed]
  124. Aran, D.; Looney, A.P.; Liu, L.; Wu, E.; Fong, V.; Hsu, A.; Chak, S.; Naikawadi, R.P.; Wolters, P.J.; Abate, A.R.; et al. Reference-based analysis of lung single-cell sequencing reveals a transitional profibrotic macrophage. Nat. Immunol. 2019, 20, 163–172. [Google Scholar] [CrossRef]
  125. Misharin, A.V.; Morales-Nebreda, L.; Reyfman, P.A.; Cuda, C.M.; Walter, J.M.; McQuattie-Pimentel, A.C.; Chen, C.I.; Anekalla, K.R.; Joshi, N.; Williams, K.J.N.; et al. Monocyte-derived alveolar macrophages drive lung fibrosis and persist in the lung over the life span. J. Exp. Med. 2017, 214, 2387–2404. [Google Scholar] [CrossRef]
  126. Jiang, A.; Liu, N.; Wang, J.; Zheng, X.; Ren, M.; Zhang, W.; Yao, Y. The role of PD-1/PD-L1 axis in idiopathic pulmonary fibrosis: Friend or foe? Front. Immunol. 2022, 13, 1022228. [Google Scholar] [CrossRef] [PubMed]
  127. Song, X.; Liu, F.; Chen, M.; Zhu, M.; Zheng, H.; Wang, W.; Chen, D.; Li, M.; Chen, S. MiR-21 regulates skeletal muscle atrophy and fibrosis by targeting TGF-beta/SMAD7-SMAD2/3 signaling pathway. Heliyon 2024, 10, e33062. [Google Scholar] [CrossRef]
  128. McClelland, A.D.; Herman-Edelstein, M.; Komers, R.; Jha, J.C.; Winbanks, C.E.; Hagiwara, S.; Gregorevic, P.; Kantharidis, P.; Cooper, M.E. miR-21 promotes renal fibrosis in diabetic nephropathy by targeting PTEN and SMAD7. Clin. Sci. 2015, 129, 1237–1249. [Google Scholar] [CrossRef]
  129. Cao, Y.; Liu, Y.; Ping, F.; Yi, L.; Zeng, Z.; Li, Y. miR-200b/c attenuates lipopolysaccharide-induced early pulmonary fibrosis by targeting ZEB1/2 via p38 MAPK and TGF-beta/smad3 signaling pathways. Lab. Invest. 2018, 98, 339–359. [Google Scholar] [CrossRef]
  130. Chen, Y.; Xiao, Y.; Ge, W.; Zhou, K.; Wen, J.; Yan, W.; Wang, Y.; Wang, B.; Qu, C.; Wu, J.; et al. miR-200b inhibits TGF-beta1-induced epithelial-mesenchymal transition and promotes growth of intestinal epithelial cells. Cell Death Dis. 2013, 4, e541. [Google Scholar] [CrossRef]
  131. 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, 1–10. [Google Scholar] [CrossRef]
  132. Bai, X.; Nie, P.; Lou, Y.; Zhu, Y.; Jiang, S.; Li, B.; Luo, P. Pirfenidone is a renal protective drug: Mechanisms, signalling pathways, and preclinical evidence. Eur. J. Pharmacol. 2021, 911, 174503. [Google Scholar] [CrossRef] [PubMed]
  133. Torre, A.; Martínez-Sánchez, F.D.; Narvaez-Chávez, S.M.; Herrera-Islas, M.A.; Aguilar-Salinas, C.A.; Córdova-Gallardo, J. Pirfenidone use in fibrotic diseases: What do we know so far? Immun. Inflamm. Dis. 2024, 12, e1335. [Google Scholar] [CrossRef] [PubMed]
  134. Estornut, C.; Milara, J.; Bayarri, M.A.; Belhadj, N.; Cortijo, J. Targeting oxidative stress as a therapeutic approach for idiopathic pulmonary fibrosis. Front. Pharmacol. 2022, 12, 794997. [Google Scholar] [CrossRef]
  135. Biondini, D.; Balestro, E.; Lacedonia, D.; Cerri, S.; Milaneschi, R.; Luppi, F.; Cocconcelli, E.; Bazzan, E.; Clini, E.; Foschino Barbaro, M.P.; et al. Pretreatment rate of decay in forced vital capacity predicts long-term response to pirfenidone in patients with idiopathic pulmonary fibrosis. Sci. Rep. 2018, 8, 5961. [Google Scholar] [CrossRef]
  136. Tu, J.; Chen, X.; Li, C.; Liu, C.; Huang, Y.; Wang, X.; Liang, H.; Yuan, X. Nintedanib Mitigates Radiation-Induced Pulmonary Fibrosis by Suppressing Epithelial Cell Inflammatory Response and Inhibiting Fibroblast-to-Myofibroblast Transition. Int. J. Biol. Sci. 2024, 20, 3353–3371. [Google Scholar] [CrossRef]
  137. Wang, X.; Goyal, M.; Gadhave, D.; Gupta, V. Inhaled nintedanib nanoparticles for enhanced efficacy in idiopathic pulmonary fibrosis (IPF) treatment–Evidence in disease-relevant in-vitro models. J. Drug Deliv. Sci. Technol. 2024, 95, 105615. [Google Scholar] [CrossRef]
  138. Ackermann, M.; Kim, Y.O.; Wagner, W.L.; Schuppan, D.; Valenzuela, C.D.; Mentzer, S.J.; Kreuz, S.; Stiller, D.; Wollin, L.; Konerding, M.A. Effects of nintedanib on the microvascular architecture in a lung fibrosis model. Angiogenesis 2017, 20, 359–372. [Google Scholar] [CrossRef]
  139. Pan, L.; Cheng, Y.; Yang, W.; Wu, X.; Zhu, H.; Hu, M.; Zhang, Y.; Zhang, M. Nintedanib Ameliorates Bleomycin-Induced Pulmonary Fibrosis, Inflammation, Apoptosis, and Oxidative Stress by Modulating PI3K/Akt/mTOR Pathway in Mice. Inflammation 2023, 46, 1531–1542. [Google Scholar] [CrossRef]
  140. Ong, C.H.; Tham, C.L.; Harith, H.H.; Firdaus, N.; Israf, D.A. TGF-beta-induced fibrosis: A review on the underlying mechanism and potential therapeutic strategies. Eur. J. Pharmacol. 2021, 911, 174510. [Google Scholar] [CrossRef]
  141. Kato, M.; Takahashi, F.; Sato, T.; Mitsuishi, Y.; Tajima, K.; Ihara, H.; Nurwidya, F.; Baskoro, H.; Murakami, A.; Kobayashi, I.; et al. Tranilast Inhibits Pulmonary Fibrosis by Suppressing TGFbeta/SMAD2 Pathway. Drug Des. Devel. Ther. 2020, 14, 4593–4603. [Google Scholar] [CrossRef]
  142. Zhu, L.; Xu, F.; Kang, X.; Zhou, J.; Yao, Q.; Lin, Y.; Zhang, W. The antioxidant N-acetylcysteine promotes immune response and inhibits epithelial-mesenchymal transition to alleviate pulmonary fibrosis in chronic obstructive pulmonary disease by suppressing the VWF/p38 MAPK axis. Mol. Med. 2021, 27, 97. [Google Scholar] [CrossRef] [PubMed]
  143. Huang, H.; Chen, M.; Liu, F.; Wu, H.; Wang, J.; Chen, J.; Liu, M.; Li, X. N-acetylcysteine tiherapeutically protects against pulmonary fibrosis in a mouse model of silicosis. Biosci. Rep. 2019, 39, BSR20190681. [Google Scholar] [CrossRef] [PubMed]
  144. Kawasaki, K.; Kuboki, S.; Furukawa, K.; Takayashiki, T.; Takano, S.; Ohtsuka, M. LGR5 induces beta-catenin activation and augments tumour progression by activating STAT3 in human intrahepatic cholangiocarcinoma. Liver Int. 2021, 41, 865–881. [Google Scholar] [CrossRef] [PubMed]
  145. Madan, B.; Patel, M.B.; Zhang, J.; Bunte, R.M.; Rudemiller, N.P.; Griffiths, R.; Virshup, D.M.; Crowley, S.D. Experimental inhibition of porcupine-mediated Wnt O-acylation attenuates kidney fibrosis. Kidney Int. 2016, 89, 1062–1074. [Google Scholar] [CrossRef]
  146. Xu, H.; Wang, J.; Chang, Y.; Xu, J.; Wang, Y.; Long, T.; Xue, C. Fucoidan from the sea cucumber Acaudina molpadioides exhibits anti-adipogenic activity by modulating the Wnt/beta-catenin pathway and down-regulating the SREBP-1c expression. Food Funct. 2014, 5, 1547–1555. [Google Scholar] [CrossRef]
  147. Villanueva, M.T. Selective activation of Wnt ameliorates idiopathic pulmonary fibrosis. Nat. Rev. Drug Discov. 2023, 22, 619. [Google Scholar] [CrossRef]
  148. Patel, M.; Post, Y.; Hill, N.; Sura, A.; Ye, J.; Fisher, T.; Suen, N.; Zhang, M.; Cheng, L.; Pribluda, A.; et al. A WNT mimetic with broad spectrum FZD-specificity decreases fibrosis and improves function in a pulmonary damage model. Respir. Res. 2024, 25, 153. [Google Scholar] [CrossRef]
  149. Gupte, V.V.; Ramasamy, S.K.; Reddy, R.; Lee, J.; Weinreb, P.H.; Violette, S.M.; Guenther, A.; Warburton, D.; Driscoll, B.; Minoo, P. Overexpression of fibroblast growth factor-10 during both inflammatory and fibrotic phases attenuates bleomycin-induced pulmonary fibrosis in mice. Am. J. Respir. Crit. Care Med. 2009, 180, 424–436. [Google Scholar] [CrossRef]
  150. Upadhyay, D.; Bundesmann, M.; Panduri, V.; Correa-Meyer, E.; Kamp, D.W. Fibroblast growth factor-10 attenuates H2O2-induced alveolar epithelial cell DNA damage: Role of MAPK activation and DNA repair. Am. J. Respir. Cell Mol. Biol. 2004, 31, 107–113. [Google Scholar] [CrossRef]
  151. Hirani, N.; MacKinnon, A.C.; Nicol, L.; Ford, P.; Schambye, H.; Pedersen, A.; Nilsson, U.J.; Leffler, H.; Sethi, T.; Tantawi, S.; et al. Target inhibition of galectin-3 by inhaled TD139 in patients with idiopathic pulmonary fibrosis. Eur. Respir. J. 2021, 57, 2002559. [Google Scholar] [CrossRef]
  152. Zetterberg, F.R.; MacKinnon, A.; Brimert, T.; Gravelle, L.; Johnsson, R.E.; Kahl-Knutson, B.; Leffler, H.; Nilsson, U.J.; Pedersen, A.; Peterson, K.; et al. Discovery and Optimization of the First Highly Effective and Orally Available Galectin-3 Inhibitors for Treatment of Fibrotic Disease. J. Med. Chem. 2022, 65, 12626–12638. [Google Scholar] [CrossRef] [PubMed]
  153. Gaughan, E.E.; Quinn, T.M.; Mills, A.; Bruce, A.M.; Antonelli, J.; MacKinnon, A.C.; Aslanis, V.; Li, F.; O′Connor, R.; Boz, C.; et al. An Inhaled Galectin-3 Inhibitor in COVID-19 Pneumonitis: A Phase Ib/IIa Randomized Controlled Clinical Trial (DEFINE). Am. J. Respir Crit. Care Med. 2023, 207, 138–149. [Google Scholar] [CrossRef] [PubMed]
  154. Weiskirchen, R. Hepatoprotective and Anti-fibrotic Agents: It′s Time to Take the Next Step. Front. Pharmacol. 2015, 6, 303. [Google Scholar] [CrossRef]
  155. Chalasani, N.; Abdelmalek, M.F.; Garcia-Tsao, G.; Vuppalanchi, R.; Alkhouri, N.; Rinella, M.; Noureddin, M.; Pyko, M.; Shiffman, M.; Sanyal, A.; et al. Effects of Belapectin, an Inhibitor of Galectin-3, in Patients with Nonalcoholic Steatohepatitis with Cirrhosis and Portal Hypertension. Gastroenterology 2020, 158, 1334–1345.e1335. [Google Scholar] [CrossRef]
  156. Ahmed, R.; Anam, K.; Ahmed, H. Development of galectin-3 targeting drugs for therapeutic applications in various diseases. Int. J. Mol. Sci. 2023, 24, 8116. [Google Scholar] [CrossRef]
  157. Kim, J.; Ha, S.; Son, M.; Kim, D.; Kim, M.J.; Kim, B.; Kim, D.; Chung, H.Y.; Chung, K.W. TLR7 activation by miR-21 promotes renal fibrosis by activating the pro-inflammatory signaling pathway in tubule epithelial cells. Cell Commun. Signal 2023, 21, 215. [Google Scholar] [CrossRef]
  158. Moimas, S.; Salton, F.; Kosmider, B.; Ring, N.; Volpe, M.C.; Bahmed, K.; Braga, L.; Rehman, M.; Vodret, S.; Graziani, M.L.; et al. miR-200 family members reduce senescence and restore idiopathic pulmonary fibrosis type II alveolar epithelial cell transdifferentiation. ERJ Open Res 2019, 5, 00138–02019. [Google Scholar] [CrossRef]
  159. Feng, J.; Hu, S.; Liu, K.; Sun, G.; Zhang, Y. The Role of MicroRNA in the Regulation of Tumor Epithelial-Mesenchymal Transition. Cells 2022, 11, 1981. [Google Scholar] [CrossRef]
  160. Wang, P.; Xiao, T.; Li, J.; Wang, D.; Sun, J.; Cheng, C.; Ma, H.; Xue, J.; Li, Y.; Zhang, A.; et al. miR-21 in EVs from pulmonary epithelial cells promotes myofibroblast differentiation via glycolysis in arsenic-induced pulmonary fibrosis. Environ. Pollut. 2021, 286, 117259. [Google Scholar] [CrossRef]
  161. Montgomery, R.L.; Yu, G.; Latimer, P.A.; Stack, C.; Robinson, K.; Dalby, C.M.; Kaminski, N.; van Rooij, E. Micro RNA mimicry blocks pulmonary fibrosis. EMBO Mol. Med. 2014, 6, 1347–1356. [Google Scholar] [CrossRef]
  162. Chioccioli, M.; Roy, S.; Newell, R.; Pestano, L.; Dickinson, B.; Rigby, K.; Herazo-Maya, J.; Jenkins, G.; Ian, S.; Saini, G. A lung targeted miR-29 mimic as a therapy for pulmonary fibrosis. eBioMedicine 2022, 85, 104304. [Google Scholar] [CrossRef] [PubMed]
  163. Tan, J.L.; Lau, S.N.; Leaw, B.; Nguyen, H.P.; Salamonsen, L.A.; Saad, M.I.; Chan, S.T.; Zhu, D.; Krause, M.; Kim, C. Amnion epithelial cell-derived exosomes restrict lung injury and enhance endogenous lung repair. Stem Cells Transl. Med. 2018, 7, 180–196. [Google Scholar] [CrossRef] [PubMed]
  164. Yang, Y.; Liu, Y.; Chai, Y.; Liu, K.; Hu, W.; Zhao, K.; Zhu, Y.; Gao, P.; Huang, Q.; Zhang, C. Exosomes in pathogenesis, diagnosis, and treatment of pulmonary fibrosis. Front. Pharmacol. 2022, 13, 927653. [Google Scholar] [CrossRef]
  165. Yang, Y.; Huang, H.; Li, Y. Roles of exosomes and exosome-derived miRNAs in pulmonary fibrosis. Front. Pharmacol. 2022, 13, 928933. [Google Scholar] [CrossRef]
  166. Liu, F.; Peng, W.; Chen, J.; Xu, Z.; Jiang, R.; Shao, Q.; Zhao, N.; Qian, K. Exosomes Derived From Alveolar Epithelial Cells Promote Alveolar Macrophage Activation Mediated by miR-92a-3p in Sepsis-Induced Acute Lung Injury. Front. Cell Infect. Microbiol. 2021, 11, 646546. [Google Scholar] [CrossRef]
  167. Zhang, M.; Xue, X.; Lou, Z.; Lin, Y.; Li, Q.; Huang, C. Exosomes from senescent epithelial cells activate pulmonary fibroblasts via the miR-217-5p/Sirt1 axis in paraquat-induced pulmonary fibrosis. J. Transl. Med. 2024, 22, 310. [Google Scholar] [CrossRef]
  168. Martin-Medina, A.; Lehmann, M.; Burgy, O.; Hermann, S.; Baarsma, H.A.; Wagner, D.E.; De Santis, M.M.; Ciolek, F.; Hofer, T.P.; Frankenberger, M.; et al. Increased Extracellular Vesicles Mediate WNT5A Signaling in Idiopathic Pulmonary Fibrosis. Am. J. Respir. Crit. Care Med. 2018, 198, 1527–1538. [Google Scholar] [CrossRef]
  169. Xu, H.; Li, X.; Liu, K.; Huang, P.; Liu, X.-J. PM2. 5 Promotes Macrophage-Mediated Inflammatory Response Through Airway Epithelial Cell-Derived Exosomal miR-155-5p. J. Inflamm. Res. 2024, 17, 8555–8567. [Google Scholar] [CrossRef]
  170. Schuhmacher, A. Pharma innovation: How evolutionary economics is shaping the future of pharma R&D. Drug Discov. Today 2024, 29, 104222. [Google Scholar]
  171. Gangwal, A.; Lavecchia, A. Unleashing the power of generative AI in drug discovery. Drug Discov. Today 2024, 29, 103992. [Google Scholar] [CrossRef]
  172. Nagar, L.; Saini, A.; Singh, S.K.; Gupta, G.; MacLoughlin, R.; Chellappan, D.K.; Prasher, P.; Chakraborty, A.; Dua, K.; Dureja, H. Recent trends and applications of nanostructure-based drug delivery in alleviating pulmonary fibrosis. Eur. Polym. J. 2024, 220, 113467. [Google Scholar] [CrossRef]
  173. Zhang, X.; Zhang, L.; Tian, J.; Li, Y.; Wu, M.; Zhang, L.; Qin, X.; Gong, L. The application and prospects of drug delivery systems in idiopathic pulmonary fibrosis. Biomater. Adv. 2025, 168, 214123. [Google Scholar] [CrossRef] [PubMed]
  174. Wong, S.L.; Gan, J.S.; De Rubis, G.; Paudel, K.R.; Yeung, S.; Chellappan, D.K.; Keng, P.S.; Dua, K. Innovative phytoceutical-based drug delivery in the management of pulmonary fibrosis: A roadmap towards inhaled therapeutics. J. Drug Deliv. Sci. Technol. 2024, 99, 105942. [Google Scholar] [CrossRef]
  175. Chanda, D.; Otoupalova, E.; Smith, S.R.; Volckaert, T.; De Langhe, S.P.; Thannickal, V.J. Developmental pathways in the pathogenesis of lung fibrosis. Mol. Asp. Med. 2019, 65, 56–69. [Google Scholar] [CrossRef]
  176. Jia, W.; Wang, Z.; Gao, C.; Wu, J.; Wu, Q. Trajectory modeling of endothelial-to-mesenchymal transition reveals galectin-3 as a mediator in pulmonary fibrosis. Cell Death Dis. 2021, 12, 327. [Google Scholar] [CrossRef]
  177. Liang, S.; Ma, J.; Wang, G.; Shao, J.; Li, J.; Deng, H.; Wang, C.; Li, W. The Application of Artificial Intelligence in the Diagnosis and Drug Resistance Prediction of Pulmonary Tuberculosis. Front. Med. 2022, 9, 935080. [Google Scholar] [CrossRef]
  178. Ricard-Blum, S.; Miele, A.E. Omic approaches to decipher the molecular mechanisms of fibrosis, and design new anti-fibrotic strategies. Semin. Cell Dev. Biol. 2020, 101, 161–169. [Google Scholar] [CrossRef]
  179. Xing, L.; Chang, X.; Shen, L.; Zhang, C.; Fan, Y.; Cho, C.; Zhang, Z.; Jiang, H. Progress in drug delivery system for fibrosis therapy. Asian J. Pharm. Sci. 2021, 16, 47–61. [Google Scholar] [CrossRef]
  180. Sun, C.; Lu, J.; Wang, J.; Hao, P.; Li, C.; Qi, L.; Yang, L.; He, B.; Zhong, Z.; Hao, N. Redox-sensitive polymeric micelles with aggregation-induced emission for bioimaging and delivery of anticancer drugs. J. Nanobiotechnol. 2021, 19, 14. [Google Scholar] [CrossRef]
  181. Papavassiliou, K.A.; Sofianidi, A.A.; Spiliopoulos, F.G.; Gogou, V.A.; Gargalionis, A.N.; Papavassiliou, A.G. YAP/TAZ Signaling in the Pathobiology of Pulmonary Fibrosis. Cells 2024, 13, 1519. [Google Scholar] [CrossRef]
  182. Sun, M.; Sun, Y.; Feng, Z.; Kang, X.; Yang, W.; Wang, Y.; Luo, Y. New insights into the Hippo/YAP pathway in idiopathic pulmonary fibrosis. Pharmacol. Res. 2021, 169, 105635. [Google Scholar] [CrossRef] [PubMed]
  183. Xie, H.; Wu, L.; Deng, Z.; Huo, Y.; Cheng, Y. Emerging roles of YAP/TAZ in lung physiology and diseases. Life Sci. 2018, 214, 176–183. [Google Scholar] [CrossRef] [PubMed]
  184. Liu, Y.; Wen, D.; Ho, C.; Yu, L.; Zheng, D.; O’Reilly, S.; Gao, Y.; Li, Q.; Zhang, Y. Epigenetics as a versatile regulator of fibrosis. J. Transl. Med. 2023, 21, 164. [Google Scholar] [CrossRef]
  185. Sanders, Y.Y.; Lyv, X.; Zhou, Q.J.; Xiang, Z.; Stanford, D.; Bodduluri, S.; Rowe, S.M.; Thannickal, V.J. Brd4-p300 inhibition downregulates Nox4 and accelerates lung fibrosis resolution in aged mice. JCI Insight 2020, 5, 137127. [Google Scholar] [CrossRef]
  186. Goetz, L.; Seedat, N.; Vandersluis, R.; van der Schaar, M. Generalization-a key challenge for responsible AI in patient-facing clinical applications. npj Digit. Med. 2024, 7, 126. [Google Scholar] [CrossRef]
  187. Yang, Y.; Cheng, F. Artificial intelligence streamlines scientific discovery of drug-target interactions. Br. J. Pharmacol. 2025, 1–18. [Google Scholar] [CrossRef]
  188. Gholap, A.D.; Uddin, M.J.; Faiyazuddin, M.; Omri, A.; Gowri, S.; Khalid, M. Advances in artificial intelligence for drug delivery and development: A comprehensive review. Comput. Biol. Med. 2024, 178, 108702. [Google Scholar] [CrossRef]
  189. Wang, Z.; Liu, Y.; Chang, X.; Liu, X. Deconvolution and inference of spatial communication through optimization algorithm for spatial transcriptomics. Commun. Biol. 2025, 8, 235. [Google Scholar] [CrossRef]
  190. Making space for spatial biology in the clinic. Nat. Biotechnol. 2025, 43, 1–2. [CrossRef]
  191. Armanios, M.Y.; Chen, J.J.L.; Cogan, J.D.; Alder, J.K.; Ingersoll, R.G.; Markin, C.; Lawson, W.E.; Xie, M.; Vulto, I.; Phillips, J.A., III; et al. Telomerase mutations in families with idiopathic pulmonary fibrosis. N. Engl. J. Med. 2007, 356, 1317–1326. [Google Scholar] [CrossRef]
  192. Tsakiri, K.D.; Cronkhite, J.T.; Kuan, P.J.; Xing, C.; Raghu, G.; Weissler, J.C.; Rosenblatt, R.L.; Shay, J.W.; Garcia, C.K. Adult-onset pulmonary fibrosis caused by mutations in telomerase. Proc. Natl. Acad. Sci. USA 2007, 104, 7552–7557. [Google Scholar] [CrossRef] [PubMed]
  193. Seibold, M.A.; Wise, A.L.; Speer, M.C.; Steele, M.P.; Brown, K.K.; Loyd, J.E.; Fingerlin, T.E.; Zhang, W.M.; Gudmundsson, G.; Groshong, S.D.; et al. A Common MUC5B Promoter Polymorphism and Pulmonary Fibrosis. N. Engl. J. Med. 2011, 364, 1503–1512. [Google Scholar] [CrossRef] [PubMed]
  194. Nathan, S.D.; Albera, C.; Bradford, W.Z.; Costabel, U.; du Bois, R.M.; Fagan, E.A.; Fishman, R.S.; Glaspole, I.; Glassberg, M.K.; Glasscock, K.F.; et al. Effect of continued treatment with pirfenidone following clinically meaningful declines in forced vital capacity: Analysis of data from three phase 3 trials in patients with idiopathic pulmonary fibrosis. Thorax 2016, 71, 429–435. [Google Scholar] [CrossRef]
  195. Richeldi, L.; Kreuter, M.; Selman, M.; Crestani, B.; Kirsten, A.-M.; Wuyts, W.A.; Xu, Z.; Bernois, K.; Stowasser, S.; Quaresma, M.; et al. Long-term treatment of patients with idiopathic pulmonary fibrosis with nintedanib: Results from the TOMORROW trial and its open-label extension. Thorax 2018, 73, 581–583. [Google Scholar] [CrossRef]
  196. Lancaster, L.H.; de Andrade, J.A.; Zibrak, J.D.; Padilla, M.L.; Albera, C.; Nathan, S.D.; Wijsenbeek, M.S.; Stauffer, J.L.; Kirchgaessler, K.-U.; Costabel, U. Pirfenidone safety and adverse event management in idiopathic pulmonary fibrosis. Eur. Respir. Rev. 2017, 26, 170057. [Google Scholar] [CrossRef]
  197. Cottin, V.; Martinez, F.J.; Jenkins, R.G.; Belperio, J.A.; Kitamura, H.; Molina-Molina, M.; Tschoepe, I.; Coeck, C.; Lievens, D.; Costabel, U. Safety and tolerability of nintedanib in patients with progressive fibrosing interstitial lung diseases: Data from the randomized controlled INBUILD trial. Respir. Res. 2022, 23, 85. [Google Scholar] [CrossRef]
  198. Seibold, J.R.; Maher, T.M.; Highland, K.B.; Assassi, S.; Azuma, A.; Hummers, L.K.; Costabel, U.; von Wangenheim, U.; Kohlbrenner, V.; Gahlemann, M.; et al. Safety and tolerability of nintedanib in patients with systemic sclerosis-associated interstitial lung disease: Data from the SENSCIS trial. Ann. Rheum. Dis. 2020, 79, 1478–1484. [Google Scholar] [CrossRef]
  199. Majewski, S.; Szewczyk, K.; Zal, A.; Bialas, A.J.; Milkowska-Dymanowska, J.; Piotrowski, W.J. Serial Measurements of Circulating KL-6, SP-D, MMP-7, CA19-9, CA-125, CCL18, and Periostin in Patients with Idiopathic Pulmonary Fibrosis Receiving Antifibrotic Therapy: An Exploratory Study. J. Clin. Med. 2021, 10, 3864. [Google Scholar] [CrossRef]
  200. Ikeda, K.; Shiratori, M.; Chiba, H.; Nishikiori, H.; Yokoo, K.; Saito, A.; Hasegawa, Y.; Kuronuma, K.; Otsuka, M.; Yamada, G.; et al. Serum surfactant protein D predicts the outcome of patients with idiopathic pulmonary fibrosis treated with pirfenidone. Respir. Med. 2017, 131, 184–191. [Google Scholar] [CrossRef]
  201. Moore, B.B.; Lawson, W.E.; Oury, T.D.; Sisson, T.H.; Raghavendran, K.; Hogaboam, C.M. Animal Models of Fibrotic Lung Disease. Am. J. Respir. Cell Mol. Biol. 2013, 49, 167–179. [Google Scholar] [CrossRef]
  202. Yasutomo, K. Genetics and animal models of familial pulmonary fibrosis. Int. Immunol. 2021, 33, 653–657. [Google Scholar] [CrossRef]
Figure 1. Possible mechanisms of epithelial involvement in the progression of pulmonary fibrosis (PF). In PF, epithelial cells play multifaceted roles in the fibrotic process. They promote fibrosis through various mechanisms, including epithelial injury, epithelial–mesenchymal transition (EMT), pro-fibrotic factors secretion, extracellular matrix (ECM) remodeling, and inflammatory signaling. Damaged epithelial cells lose their ability to repair, leading to destruction of the alveolar structure and triggering fibrotic responses. The images illustrated in the figures were adapted from https://app.biorender.com (accessed on 26 March 2025).
Figure 1. Possible mechanisms of epithelial involvement in the progression of pulmonary fibrosis (PF). In PF, epithelial cells play multifaceted roles in the fibrotic process. They promote fibrosis through various mechanisms, including epithelial injury, epithelial–mesenchymal transition (EMT), pro-fibrotic factors secretion, extracellular matrix (ECM) remodeling, and inflammatory signaling. Damaged epithelial cells lose their ability to repair, leading to destruction of the alveolar structure and triggering fibrotic responses. The images illustrated in the figures were adapted from https://app.biorender.com (accessed on 26 March 2025).
Pharmaceuticals 18 00812 g001
Figure 2. Crosstalk between epithelial signaling pathways in pulmonary fibrosis (PF). In the pathogenesis of PF, multiple signaling pathways work together to drive disease progression through complex crosstalk and synergistic regulatory networks, which interact at multiple levels, including the receptor level, intracellular signal nodes, and epigenetic level, and ultimately lead to continuous amplification of pro-fibrotic signals and irreversibility of pathological remodeling through dynamic network regulation. The images illustrated in the figures were adapted from https://app.biorender.com (accessed on 27 March 2025).
Figure 2. Crosstalk between epithelial signaling pathways in pulmonary fibrosis (PF). In the pathogenesis of PF, multiple signaling pathways work together to drive disease progression through complex crosstalk and synergistic regulatory networks, which interact at multiple levels, including the receptor level, intracellular signal nodes, and epigenetic level, and ultimately lead to continuous amplification of pro-fibrotic signals and irreversibility of pathological remodeling through dynamic network regulation. The images illustrated in the figures were adapted from https://app.biorender.com (accessed on 27 March 2025).
Pharmaceuticals 18 00812 g002
Figure 3. The changes in DNA methyltransferases (DNMTs) and histone demethylases (KDMs) affected by ROS affect the function of epithelial cells and affect pulmonary fibrosis (PF). In PF, dysfunctional epithelial cells undergo profound molecular rewiring to adopt pro-fibrotic phenotypes. Mitochondrial dysfunction generates excessive ROS, which oxidize DNMTs and KDMs, leading to locus-specific hypermethylation and chromatin remodeling at pro-fibrotic genes. The images illustrated in the figures were adapted from https://app.biorender.com (accessed on 27 March 2025).
Figure 3. The changes in DNA methyltransferases (DNMTs) and histone demethylases (KDMs) affected by ROS affect the function of epithelial cells and affect pulmonary fibrosis (PF). In PF, dysfunctional epithelial cells undergo profound molecular rewiring to adopt pro-fibrotic phenotypes. Mitochondrial dysfunction generates excessive ROS, which oxidize DNMTs and KDMs, leading to locus-specific hypermethylation and chromatin remodeling at pro-fibrotic genes. The images illustrated in the figures were adapted from https://app.biorender.com (accessed on 27 March 2025).
Pharmaceuticals 18 00812 g003
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Wang, J.; Chao, J. Epithelial Cell Dysfunction in Pulmonary Fibrosis: Mechanisms, Interactions, and Emerging Therapeutic Targets. Pharmaceuticals 2025, 18, 812. https://doi.org/10.3390/ph18060812

AMA Style

Wang J, Chao J. Epithelial Cell Dysfunction in Pulmonary Fibrosis: Mechanisms, Interactions, and Emerging Therapeutic Targets. Pharmaceuticals. 2025; 18(6):812. https://doi.org/10.3390/ph18060812

Chicago/Turabian Style

Wang, Jing, and Jie Chao. 2025. "Epithelial Cell Dysfunction in Pulmonary Fibrosis: Mechanisms, Interactions, and Emerging Therapeutic Targets" Pharmaceuticals 18, no. 6: 812. https://doi.org/10.3390/ph18060812

APA Style

Wang, J., & Chao, J. (2025). Epithelial Cell Dysfunction in Pulmonary Fibrosis: Mechanisms, Interactions, and Emerging Therapeutic Targets. Pharmaceuticals, 18(6), 812. https://doi.org/10.3390/ph18060812

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