Skip to main content

Pulmonary fibrosis: from mechanisms to therapies

Abstract

Pulmonary fibrosis (PF) is a chronic, progressive interstitial lung disease characterized by excessive deposition of extracellular matrix (ECM) and abnormal fibroblast proliferation, which is mainly caused by air pollution, smoking, aging, occupational exposure, environmental pollutants exposure, and microbial infections. Although antifibrotic agents such as pirfenidone and nintedanib, approved by the United States (US) Food and Drug Administration (FDA), can slow the decline in lung function and disease progression, their side effects and delivery inefficiency limit the overall prognosis of PF. Therefore, there is an urgent need to develop effective therapeutic targets and delivery approaches for PF in clinical settings. This review provides an overview of the pathogenic mechanisms, therapeutic drug targeting signaling pathways, and promising drug delivery strategies for treating PF.

Introduction

Pulmonary fibrosis (PF) is a progressive, destructive, and fatal interstitial lung disease characterized by the substantial presence of abnormally activated fibroblasts, tissue remodeling, and excessive ECM proteins (like collagens) deposition, with limited therapeutic options, resulting in an average life expectancy of only 3 to 5 years [1, 2]. The reported incidence of PF ranges from 0.9 to 9.3 cases per 100,000 individuals in Europe and North America, and from 3.5 to 13.0 cases per 100,000 people in Asia and South America [3]. Currently, only nintedanib and pirfenidone are approved as targeted therapies for PF [4]. In the United Kingdom (UK), the annual cost of pirfenidone is approximately $77227.66, whereas in the US, the combined annual cost for both pirfenidone and nintedanib exceeds $100,000 [5, 6]. For Asian countries, a recent study found the median direct medical cost for PF patients in China was 9,378.3 Chinese yuan (approximately $1,300) per patient per admission [7]. Moreover, PF predominantly affects older individuals, with most cases occurring between 50 and 70 years of age, and it is more common in males than in females [8]. However, female mice showed higher mortality rates and more severe fibrosis than male mice, as female mice exhibit direct fibrogenic effects on lung fibroblasts, mediated by their endogenous sex hormones [9]. The growing burden of PF poses a significant threat to public health worldwide, substantially affecting health-related quality of life [10]. Despite its clinical severity, currently approved treatments have shown limited efficacy and considerable side effects, rendering them less than satisfactory [11,12,13]. The dynamic evolution of PF further complicates diagnostic efforts and limits opportunities for early intervention [14]. Therefore, a deeper understanding of the pathogenic mechanisms underlying PF and the development of effective treatment strategies are urgently needed. Here, we review the underlying mechanisms (including risk factors and cellular as well as molecular mechanisms) of PF and also summarize the therapeutic intervention strategies against PF. Collectively, our review aims to shed new light on the pathogenic mechanisms and intervention strategies for PF.

Pathogenesis and mechanisms of PF

Despite the limited understanding of the mechanisms underlying PF, the current paradigm of disease pathogenesis encompasses specific cellular processes and molecular pathways. The initial injury to the alveolar epithelium triggers a cascade of events, including inflammation and alveolar epithelial injury, which is crucial for the onset of the disease [15, 16]. Following this, lung fibroblasts become activated and differentiate into myofibroblasts in response to the release of cytokines and growth factors from the damaged lung epithelium, accompanied by pro-fibrotic signaling pathways. This process ultimately leads to the deposition of ECM and the development of progressive fibrotic scars within the lung parenchyma, resulting in lung dysfunction [17, 18].

Risk factors

Multiple risk factors, including intrinsic factors (e.g., genetic predisposition and aging) and extrinsic factors (e.g., air pollution, smoking, occupational/environmental exposures, and microbial infections), can initiate the progression of PF. The primary risk factors involved in PF are further illustrated in Fig. 1.

Fig. 1
figure 1

Schematic representation of PF potential risk factors. Diverse risk factors, including intrinsic factors (including genetic factors and aging), and extrinsic factors (including air pollution, smoking, occupational exposure, environmental pollutants exposure, and microbial infections) contribute to PF. Image Created in https://BioRender.com

Intrinsic factors

Genetic Factors.

PF arises from the interplay between genetic and environmental factors, with genetic susceptibility playing a crucial role in determining the risk of developing the disease. Previous genome-wide association studies (GWAS) have identified common genetic variants associated with PF, particularly in genes linked to immune function, epithelial barrier function, telomeres, and cell cycle regulation [19, 20]. Ongoing GWAS are expected to uncover additional gene polymorphisms related to PF risk, and these specific variants may influence the disease’s natural history and predict clinical outcomes [21]. In patients with familial pulmonary fibrosis (FPF), telomere maintenance-related causal genes are implicated in about 30% of cases [22]. The variant rs35705950, a common gain-of-function promoter variant located in the promoter region of the MUC5B gene, has been identified as the strongest risk factor for PF, correlating with a five-fold increase in disease risk [23, 24]. The association between rs35705950 and PF has been validated across multiple independent cohorts, including studies conducted in France [25] and Mexico [26]. Besides the MUC5B promoter variant, other rare variants (MAF < 1%) related to telomere and surfactant genes may also contribute to FPF and sporadic PF cases [27]. Furthermore, pro-fibrotic signaling variants, such as those in the toll-interacting protein (TOLLIP) gene variants (including rs111521887, rs5743894, and rs574389), have been suggested to associate with PF [28]. Although our understanding of the genetic risk factors for PF is still evolving, these findings provide valuable biological insights into the susceptibility to the disease.

Aging

Aging is a normal biological process that ultimately leads to death, characterized by the gradual weakening and loss of an organism’s structure and function [29, 30]. Age-related diseases, including cardiovascular diseases, cancers, and interstitial lung diseases, identify aging as a significant risk factor for various chronic conditions in adults [31]. Natural lung aging is a complex process that involves molecular and physiological changes resulting in a decline in lung function, reduced regenerative capacity, and eventual chronic alterations in lung interstitial structure [32]. In contrast to normal cells, senescent cells exhibit distinct genetic and morphologic characteristics, including genomic instability, telomere shortening, epigenetic dysregulation, and mitochondrial dysfunction [33]. Recent studies have investigated mechanisms underlying accelerated cellular aging, particularly the loss of epithelial progenitor cell function during PF progression [34]. Primary lung fibroblasts isolated from PF patients demonstrate a more pronounced senescent phenotype, characterized by the overexpression of α smooth muscle actin (α-SMA) and excessive ECM deposition [35, 36]. Overall, the current understanding of cellular senescence indicates that it is a significant risk factor for the development and progression of PF.

Extrinsic factors

Air Pollution

Recent studies have increasingly highlighted the significant role of air pollution, particularly particulate matter (PM, including PM10 and PM2.5), nitrogen oxides (NO), and other pollutants, in the development and exacerbation of PF. Long-term exposure to PM10 and NO, which are prevalent in urban environments due to traffic and industrial emissions, has been strongly linked to heightened risk and increased mortality in PF patients. This is largely attributed to the triggering of inflammatory responses and oxidative stress [37]. Specifically, PM10 exposure has been associated with lung tissue damage and the progression of fibrosis, likely through mechanisms involving epithelial cell death, oxidative damage, and the activation of inflammatory signaling pathways [38]. Nitrogen oxides, particularly NO2, further exacerbate the inflammatory cascade within the lungs, leading to epithelial injury, fibroblast activation, and the deposition of extracellular matrix components, all of which are critical drivers of fibrosis [39]. Furthermore, exposure to both PM2.5 and NO2 has been found to amplify the risk of acute exacerbations in PF patients, likely through mechanisms involving inflammatory mediators such as interleukins [40, 41]. Studies have also demonstrated that exposure to PM2.5 has been shown to trigger PF through mechanisms such as epithelial-mesenchymal transition (EMT) [42, 43]. Additionally, short-term exposure to PM2.5 is associated with significant lung inflammation and injury [44, 45].

Smoking

Cigarette smoke contributes to the senescence and/or injury of various cell types, including type-2 alveolar epithelial (AEC2) cells, thus playing a role in the progression of PF [46]. A meta-analysis indicated that tobacco smoking correlates with an increased risk of developing PF [47]. A prospective cohort study from the UK Biobank demonstrated that smoking intensity exhibits a dose-response relationship with the risk of PF [48]. Furthermore, electronic cigarettes have gained popularity as a smoking alternative for adults in recent years. Nicotine is one of the only chemicals consistently found in traditional cigarettes and electronic cigarettes [49]. Several reports indicate that electronic cigarettes emit toxic substances, including volatile organic compounds, heavy metals, and known carcinogens, and these are becoming more harmful to pulmonary and cardiac systems [50, 51]. For example, Dai et al. found that long-term chronic exposure to electronic cigarettes induced irreversible damage to cardiovascular and respiratory systems in mice, characterized by cardiac fibrosis and PF [52].

Occupational Exposure

Significant associations have been found between occupational exposure and PF. Occupational exposure to airborne hazardous particles, particularly asbestos, silica, and coal dust, represents the primary etiological factor in the development of PF [53, 54]. Experimental evidence demonstrates that intratracheal asbestos exposure induces marked PF, as evidenced by elevated fibrosis in mouse lung tissue [55]. Similarly, silicosis is a progressive and irreversible occupational lung disease resulting from prolonged inhalation of crystalline silica dust, and with millions of new cases reported annually, it continues to pose a significant global public health challenge [56,57,58]. Coal worker’s pneumoconiosis, another common occupational disease in miners, arises from chronic coal dust exposure, resulting in PF and immune dysfunction [59]. Collectively, these findings underscore occupational hazards as critical contributors to PF, necessitating stringent workplace safety measures and early surveillance to mitigate disease progression.

Environmental Pollutants Exposure

Numerous studies indicate that exposure to environmental pollutants may induce the development of PF. For instance, as a common chemical contaminant, arsenic widely exists in air, drinking water, and soil and has been reported to induce PF via mediating H3K18 lactylation [60]. Other environmental pollutants like 1-nitropyrene, which is formed during gas, oil, and coal combustion, could cause various respiratory system diseases, including PF [61]. Benzo[a]pyrene (BaP) is a well-known environmental pollutant, and Li et al. have found that exposure to BaP might induce PF through EMT [62]. In addition, polystyrene nano- and microplastics (PS-NPs/PS-MPs) have emerged as significant contributors to PF, acting through complex and multifaceted pathological mechanisms [63]. Zhang et al. found that PS-MPs triggered PF by promoting cell ferroptosis [64]. Inhalation exposure to PS-NPs can promote EMT in human alveolar epithelial cells, primarily by enhancing reactive oxygen species (ROS) generation and activating NADPH oxidase 4 (NOX4). This process subsequently triggers mitochondrial dysfunction and endoplasmic reticulum stress, critical events implicated in the fibrotic cascade. Notably, the adverse effects of PS-NPs appear highly dependent on particle characteristics, with smaller size and positive surface charges correlating with heightened cytotoxicity [65]. Moreover, exposure to PS-NPs promotes mitochondrial double-stranded DNA release, further activating the cGAS-STING pathway and amplifying pulmonary inflammation and fibrosis [66]. Our study demonstrates that polystyrene nanoplastics induce PF via two parallel mechanisms: (1) direct stimulation of fibroblast proliferation and activation, and (2) GPX4-dependent ferroptosis in alveolar epithelial cells (unpublished data). Together, these findings underscore the potential pathogenic mechanisms in environmental pollutant-induced lung fibrosis, offering new insights into disease mechanisms and promising avenues for intervention.

Microbial Infections

Increasing evidence suggests that microbial infections (especially viral infections) may play a role in the initiation and exacerbation of PF [67, 68]. Infection with severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2), the virus responsible for COVID-19, can lead to a cytokine storm and trigger profibrotic responses, then result in significant fibrotic changes in the lungs [69, 70]. Additionally, previous studies have shown that infection with γ-Herpes virus-68 can exacerbate bleomycin (BLM)-induced PF in mouse models [71]. Moreover, Huang et al. reviewed various viruses, including Human T-cell leukemia virus (HTLV), Cytomegalovirus (CMV), Human immunodeficiency virus (HIV), and Epstein–Barr virus (EBV), highlighting their potential roles as risk factors for the development of PF following infection [72]. Besides, Chlamydia, Mycoplasma, and Aspergillus fumigatus have also been indicated as potential inducers of PF [73, 74]. Spatial transcriptomics also revealed distinct profibrotic and antifibrotic markers expressed by epithelial and non-epithelial cell populations in influenza virus-infected aged murine lungs. This study uncovered spatially resolved molecular networks that drive fibrotic progression in aged tissues, highlighting how viral infection exacerbates dysregulated fibrogenic pathways [75]. Pseudomonas aeruginosa, an opportunistic Gram-negative pathogen, is a major etiological agent of nosocomial infections and a significant contributor to hospital-acquired pneumonia [76]. Furthermore, chronic pseudomonas aeruginosa infection is strongly associated with progressive structural lung damage in chronic respiratory diseases, including cystic fibrosis and PF [77]. These findings may provide new insights into strategies for managing PF by targeting microbial infections as a therapeutic approach.

The risk factors for PF underscore the multifactorial nature of the disease, driven by the interplay between environmental factors and genetic susceptibility. Collectively, these findings emphasize the importance of PF prevention and management, integrating genetic screening, environmental mitigation, and targeted therapies to address the interplay of aging, genetic susceptibility, and cumulative environmental exposures. Future research should aim to elucidate gene-environment interactions, identify biomarkers for early detection, and develop interventions that disrupt shared pathogenic pathways to mitigate the global burden of PF.

The main effector cells and molecular pathways in PF

Cells involved in PF

PF is a complex and dynamic process that involves cell types throughout the progression of pathological fibrogenesis, including epithelial cells, macrophages, fibroblasts, and endothelial cells [78, 79]. In this section, we will illustrate the roles of these cells in the development of PF (Fig. 2).

Fig. 2
figure 2

Summary of the key effector cells in the initiation of PF. Following profibrotic factors stimulus, macrophages respond and produce inflammatory cytokines. Cytokines attract more immune cells, such as monocytes and neutrophils, which in turn produce more cytokines, creating a cycle of inflammation that damages the lung cells. In addition, activated fibroblasts (including those that originated from EMT and EndoMT ) stimulate the production of myofibroblasts as well as excessive ECM deposition which undergoes pathologic remodeling. Image Created in https://BioRender.com

Alveolar Epithelial Cell

Epithelial cell dysfunction is believed to be susceptible to abnormal injury responses and serves as a critical driver of diffuse parenchymal disease, including PF [80, 81]. In the lung, AEC2 cells play a key role in maintaining alveolar epithelium homeostasis, regenerating surfactant-producing cells, and facilitating the differentiation of type-1 alveolar epithelial (AEC1) cells [82, 83]. When AEC1 cells are injured, the integrity of the epithelial barrier is compromised, leading to aberrant proliferation and differentiation of AEC2. If AEC1 injury persists or the ability to restore the epithelial barrier is impaired, this can result in abnormal activation of fibroblast, increased ECM deposition, and destruction of lung architecture [84, 85].

It has been reported that senescence-associated markers, including the tumor suppressor genes p53 and p21, were upregulated in the distal epithelium of lungs affected by PF [86]. Furthermore, single-cell RNA sequencing of AEC2 derived from both PF and healthy control lungs has highlighted the role of p53 in fibrotic AEC2 [87]. Specifically, oxidative stress and ER stress-induced injury in AECs can promote the release of injury-associated molecules, such as damage-associated molecular patterns (DAMPs), which are linked to the initiation of tissue repair and fibrotic response [88]. EMT is a cellular process in which epithelial cells lose their characteristic features, acquire mesenchymal properties, and adopt fibroblast-like phenotypes [89]. EMT is implicated in several pathological conditions, including cancer and fibrosis [90, 91]. Collectively, these studies suggest that dysfunction in alveolar epithelial cells, including senescence, oxidative and ER stress-induced injury, and dysregulated EMT, significantly contribute to the pathogenesis of PF.

Macrophages

Macrophages play a crucial role in host defense, which includes the phagocytic clearance of microorganisms and apoptotic cells [92]. As the most abundant immune cells in the lungs, comprising approximately 70% of the immune cell population, macrophages have significant modulatory roles in the fibrogenic process at various locations or stages of PF [93]. The lungs contain two distinct groups of macrophages: interstitial macrophages (IMs), located within the lung parenchyma, and alveolar macrophages (AMs), found in the airways [94]. Based on their activation status, macrophages can be polarized into two major subsets: classically activated macrophages (M1), which produce pro-inflammatory cytokines and can be activated by lipopolysaccharide (LPS), and activated macrophages (M2), which dampen inflammation, promote wound healing and can be activated by IL-4 [95, 96]. In fibrotic lungs, both M1 and M2 macrophages contribute to disease progression and regulate the production of inflammatory mediators (such as IL-1, inducible nitric oxide synthase (iNOS), and IL-6) and fibrogenic mediators (such as IL-4, arginase 1 (Arg-1), and IL-13) [97].

In the current paradigm of PF pathogenesis, inflammation is recognized as an initiating pathological feature and triggers aberrant wound repair responses throughout the progression of the disease [98]. During the early inflammatory stage, M1 macrophages activate and then produce extracellular matrix metalloproteinases (MMPs), which lead to the degradation of the ECM. Notably, the inhibition of histone methyltransferase enhancer of zeste homolog 2 (EZH2) has been shown to attenuate lung inflammation and fibrosis by modulating macrophage polarization in an LPS-induced acute respiratory distress syndrome (ARDS)-associated PF mouse model [99].

M2 macrophages are thought to regulate the progression of fibrotic lung disease by decreasing inflammatory factors and increasing the production of chemokines, MMPs, TIMPs, and fibronectin [100, 101]. Animal studies have also indicated that M2 macrophages could serve as therapeutic targets for the treatment of PF [102, 103]. However, it is noted that M2 macrophages may exacerbate PF by producing amounts of transforming growth factor-beta 1(TGF-β1) and platelet-derived growth factor (PDGF), which induce fibroblast proliferation and differentiation [104]. In summary, macrophages are implicated in the fibrotic remodeling of organs and play a crucial role in the pathogenesis of PF.

Fibroblasts

Fibroblast-to-myofibroblast transition (FMT) is characterized by excessive ECM deposition in the lung parenchyma and plays a central role in the pathogenesis of PF [105]. TGF-β1 is the primary stimulating factor driving the FMT process. Myofibroblasts are key effector cells of PF and are crucial for the reconstruction of the ECM during the wound healing repair process. After the repair process is completed, the number of myofibroblasts decreases, along with their presence in remodeled areas, which ultimately contributes to the progression of chronic lung diseases [106].

Activated myofibroblasts are characterized by high expression of α-SMA, accompanied by collagen deposition and migrating and the formation of fibrotic foci [107]. In a comprehensive analysis using multicolor clonal cell labeling, Ting Xie et al. reported a significant population of fibroblasts in the lungs of mice with PF, including those expressing α-SMA, Col1α1, vimentin, desmin, (platelet-derived growth factor receptor, alpha polypeptide) Pdgfrα, and (platelet-derived growth factor receptor, beta polypeptide) Pdgfrβ [108]. These fibroblasts develop contractile properties similar to those of smooth muscle cells (SMCs), leading to increased mechanical stress and matrix stiffness, ultimately resulting in a feed-forward mechanism of progressive fibrotic remodeling [109]. Therefore, targeting fibroblasts or FMT progression may represent novel therapeutic targets for the treatment of PF.

Endothelial Cells.

In the process of PF, loss of pulmonary microvascular endothelial cells (PMVECs) and dysregulation of angiogenesis are recognized as significant pathological changes [110, 111]. Additionally, endothelial cells serve as a source of myofibroblasts through the endothelial-to-mesenchymal transition (EndoMT) process [112, 113]. During EndoMT, endothelial cells undergo a phenotypic shift to acquire mesenchymal characteristics, resulting in a myofibroblast-like phenotype marked by high expression of α-SMA and vimentin, alongside low expression of vascular endothelial cadherin (VE-cadherin) [113, 114]. Recent studies by Zhang et al. have demonstrated that the FDA-approved antifibrotic drug Pirfenidone mitigates LPS-induced PF by inhibiting EndoMT through the Hedgehog signaling pathway [115]. Furthermore, a subpopulation of Cxcl12+ endothelial cells expands in the lungs of mice treated with BLM and is closely associated with pro-fibrotic processes, including the regulation of angiogenesis, collagen binding, and chemokine activity [116]. Collectively, these findings suggest that endothelial cells play a crucial role in the pathogenesis of PF.

The pathogenesis of PF hinges on intricate cellular crosstalk, yet critical ambiguities persist. A critical gap remains in understanding how epithelial injury, macrophage plasticity, fibroblast activation, and endothelial dysfunction synergize across disease stages. Future research needs to consider the role of intercellular crosstalk in PF.

Molecular pathways in PF

Numerous molecular signaling pathways, including TGF-β, Wnt, phosphoinositol-3 kinase (PI3K)-Akt, and Hippo, have been extensively studied for their roles in regulating the progression of PF. This section primarily focuses on the contributions of these signaling pathways to the pathophysiology of PF (Fig. 3).

Fig. 3
figure 3

Overview of molecular signaling pathways in PF. In the TGF-β signaling, TGF-β binding to TGF-βRs activates the TGF-β-Smad signalings. In the Wnt signaling, activated DVL inhibits the destruction complex and then allows β-catenin accumulation to enter the nucleus where it can act together with either p300 or CBP as a transcriptional co-activator for TCF/LEF to activate Wnt-related fibrotic genes. In the Wnt signaling, MAP4K and TAOK families of kinases phosphorylate and activate LATS1/2. Meanwhile, YAP activity is regulated by the LATS1/2 kinases, which promote YAP nucleus accumulation and bind to several transcription factors, such as the TEAD family, eventually changing profibrotic gene expression. In the PI3K-Akt signaling, PI3Ks can be activated by various growth factors like VEGF, PDGF, and TGF-β. The common downstream of receptor-mediated PI3K activation is Akt, which regulates the activity of mTOR, HIF-1α, and Fox3 to involve cell proliferation, differentiation, and ECM deposition in PF. Image Created in https://BioRender.com

TGF-β Signaling

TGF-β plays a multifaceted role in regulating inflammation responses, cell proliferation, and wound healing of fibrotic tissues [117, 118]. There are three isoforms of TGF-βs, termed TGF-β1, TGF-β2, and TGF-β3 [119]. TGF-β1 is primarily expressed in endothelial, hematopoietic, and connective tissue cells, while TGF-β2 is predominantly found in epithelial and neuronal cells. TGF-β3 is mainly expressed in mesenchymal cells [120, 121]. Notably, among various pro-fibrotic factors, TGF-β1 stands out as the most critical contributor to the fibrogenic process. TGF-β1 regulates fibrotic diseases through both canonical (Smad-based) and non-canonical (non-Smad-based) signaling pathways, leading to myofibroblast activation and ECM deposition [122].

In the canonical signaling pathway, TGF-β1 binds to its receptors, TβRI and TβRII, resulting in the phosphorylation of Smad2 and Smad3 [123]. Smad7 acts as a negative regulator of the TGF-β1/Smad pathway. Phosphorylated Smad2 and Smad3 then associate with Smad4 and translocate into the nucleus, where they regulate the expression of downstream target genes [124, 125]. Besides, the cellular homolog of the avian Sloan-Kettering virus (SKI) encodes the SKI proto-oncogene protein and is commonly regarded as a negative regulator of the canonical TGF-β signaling pathway [126]. The SKI protein complex plays a critical role in regulating fibroblast activation and EMT by modulating TGF-β1/Smad signaling. The inhibition of SKI significantly suppresses TGF-β1-induced fibroblast proliferation, highlighting its importance in cellular activation processes [127]. Additionally, SKI acts as a key negative regulator of EMT by antagonizing Smad-dependent signaling, as evidenced by its ability to reverse the process of EMT [128]. These findings underscore SKI’s dual function in both fibroblast proliferation and EMT regulation, positioning it as a central player in TGF-β1-mediated cellular responses.

In the non-canonical signaling pathway, TGF-β interacts with ALK1, leading to the activation of Smad1/5, which is implicated in tissue disease [129]. During the process of PF, TGF-β can be released by alveolar epithelial cells, macrophages, or regulatory T cells following injury. It subsequently recruits and activates circulating fibrocytes and fibroblasts, resulting in ECM production and deposition [130, 131]. Furthermore, TGF-β inhibits the production of anti-fibrotic molecules, such as prostaglandin E2, thereby exacerbating the progression of PF [132].

Wnt Signaling

The Wnt signal pathways are critically recognized as important pathways for the development and homeostasis of organs and tissue [133, 134]. Wnt genes encode proteins that are highly conserved throughout evolution. These pathways are involved in various biological functions, including cell proliferation, polarity, and differentiation [135]. Moreover, sustained activation of Wnt signaling is implicated in wound repair and the pathogenesis of fibrosis, including lung, kidney, and liver fibrosis [136, 137].

Wnt signaling pathways are principally characterized by two classes: the canonical Wnt pathway and the noncanonical Wnt/calcium pathway [138]. The canonical Wnt signaling, also referred to as Wnt/β-Catenin signaling, is typically initiated by its central effector, β-Catenin. The transcription activity of β-Catenin is generally regulated by canonical Wnt proteins (Wnt1, Wnt2, Wnt3, Wnt3a, Wnt8b, Wnt10a, and Wnt10b) [139, 140]. In the “WNT ON state” (Wnt ligands exit), the formation of the destruction complex (composed by AXIN, CK1α, GSK3β, and APC, which phosphorylates β-catenin) is inhibited, leading to the nuclear translocation of β-catenin. Conversely, in the “WNT OFF state” (in the absence of Wnt ligands), the destruction complex facilitates the ubiquitin-proteasomal degradation of β-Catenin [141].

Recent literature has demonstrated that aberrant activation of Wnt signaling is observed in PF, along with persistent expression of the β-Catenin-dependent gene. Notably, nuclear accumulation of β-Catenin has been found to increase proliferative epithelial lesions and fibroblastic foci in the lungs of patients with PF [142]. Additionally, the expression of Wnt1, Wnt7b, and Wnt10b was significantly elevated in PF [143]. Furthermore, various studies have shown that Wnt signaling can regulate tissue remodeling by MMPs and vascular endothelial growth factor (VEGF) [144, 145]. Overall, it is evident that Wnt signaling plays a crucial role in the development of PF.

PI3K-Akt Signaling

Recent studies have shown that the PI3K/Akt signaling pathway serves as a master regulator in PF by influencing cell growth, differentiation, apoptosis, and angiogenesis [146, 147]. PI3K functions as a lipid kinase and is classified into three classes based on its molecular structure [148]. Among these, Class I PI3K is the most extensively studied in various diseases [149]. Following the activation of receptor tyrosine kinases, PI3K translocates from the cytoplasm to the plasma membrane. This translocation activates its catalytic subunit (p110α) and promotes the phosphorylation of phosphoinositides, leading to the formation of a complex consisting of PI3,4-bisphosphate and PI3,4,5-trisphosphate that binds to the pleckstrin-homology domain of Akt [150]. Moreover, serine 473 and threonine 308 residues of Akt are phosphorylated by pyruvate dehydrogenase kinase 1 (PDK1) and pyruvate dehydrogenase kinase 2 (PDK2), respectively, resulting in the activation of downstream signaling molecules. Key downstream targets of the PI3K/Akt signaling pathway implicated in fibrotic diseases include mTOR, Hif-1α, GSK-3, Forkhead box proteins O (FoxOs, such as FoxO3), and nitric oxide synthase (NOS) [151].

Evidence suggests that the activation of the PI3K/Akt signaling pathway is linked to fibroblast activation, epithelial cell damage, and macrophage polarization. For instance, the PI3K-Akt-mTOR/PFKFB3 pathway has been shown to regulate fibroblast glycolysis and collagen synthesis in LPS-induced PF [152]. Lu et al. demonstrated that BLM can induce lung fibroblast activation by upregulating collagen expression and enhancing cell proliferation through the PI3K/Akt pathway [153]. Besides, increased levels of PI3K have been observed in silica-induced lung fibrosis tissues and epithelial cells; the knockdown of PI3K has been shown to reduce the EMT process and attenuate PF [154]. Furthermore, enhanced PI3K/Akt signaling may worsen PF progression by promoting the M2 polarization of macrophages [155].In conclusion, PI3K/Akt signaling plays a significant role in the development of PF.

Hippo-YAP Signaling

The Hippo signaling pathway is a critical and evolutionarily conserved regulator of organismal homeostasis, consisting of a three-step cascade involving mammalian sterile 20-like kinases 1 and 2 (MST1/2, also known as STK4/3), Salvador (SAV1), large tumor suppressor 1 and 2 (LATS1/2), MOB kinase activators 1 A and 1B (MOB1a/b), and Yes-associated protein (YAP)/transcriptional co-activator with PDZ-binding motif (TAZ, also known as WWTR1) [156, 157]. It is well established that the Hippo signaling pathway regulates cellular proliferation, differentiation, and survival through its downstream effector YAP [158, 159]. Mechanistically, YAP interacts with transcription factors such as TEAD1–4 to modulate the expression of fibrotic genes [160].

In the context of PF pathogenesis, the Hippo-YAP signaling pathway plays a multifaceted role by targeting various effector cell types. Liu et al. demonstrated that the Hippo pathway is activated in fibrotic lung tissue. Inhibition of YAP and TAZ was shown to attenuate PF by suppressing collagen synthesis and lung fibroblast proliferation [161]. Furthermore, the downregulation of the Hippo signaling pathway inhibited the expression of Collagen I and Fibronectin in fibroblasts induced by TGF-β1 [162]. It is noteworthy that the Hippo/YAP signaling pathway is involved in normal alveolar development and repair processes. Activated YAP in airway basal cells can lead to hyperplasia and impair terminal differentiation [163]. Additionally, nuclear transcription of YAP is essential for airway epithelial cells to respond effectively to TGF-β. The deletion of TAZ in mouse AEC2 cells resulted in a decrease in AEC1 cell differentiation following BLM-induced lung injury [164]. Therefore, selective activation of the Hippo/YAP pathway in AEC2 cells may promote alveolar regeneration and facilitate tissue repair in PF.

TGF-β, Wnt, PI3K-Akt, and Hippo-YAP pathways drive PF through fibroblast activation, ECM remodeling, and epithelial dysfunction, yet key challenges persist. TGF-β1’s dual Smad/non-Smad signaling and SKI’s regulatory duality offer therapeutic potential but face hurdles due to TGF-β’s pleiotropy. Wnt/β-catenin activation in fibrosis is well-documented, yet the conflicting roles of Wnt isoforms (e.g., Wnt7b vs. Wnt10b) and unclear cell-specific effects limit targeted interventions. PI3K-Akt’s broad influence on fibroblast metabolism and macrophage polarization raises off-target risks. Besides, studies often neglect pathway crosstalk (e.g., Wnt-YAP/β-catenin interplay) and spatial dynamics. Finally, using specific inhibitors targeting these signaling pathways may be the potential strategy for the treatment of PF.

Therapeutic approaches

Traditional approaches to managing PF encompass a range of interventions, including oxygen therapy, pulmonary rehabilitation, symptom management with medications, lifestyle modifications, and eventual lung transplantation [165]. However, as shown in Table 1, there is a significant cost disparity between these conventional therapies and newer-generation agents like pirfenidone and nintedanib, particularly in Europe and the US. This economic consideration highlights the need for cost-effective yet efficacious treatment options.

Table 1 Comparison of Annual Costs for PF Medications in Europe vs. US

To better evaluate current therapeutic strategies, Table 2 provides an infographic summarizing the recommended treatments based on guidelines from leading scientific societies (mainly from the ATS/ERS/JRS/ALAT 2011 guideline) [169]. This comparative analysis helps identify evidence-based approaches while underscoring the ongoing demand for novel, affordable anti-fibrotic therapies.

Table 2 Summary of available therapies for PF based on scientific society guidelines

Pharmacological treatment

Current Anti-fibrotic drugs

Current drug therapies for PF include synthetic compounds, natural compounds, and antibody-based treatments. Despite only two drugs having received regulatory approval for PF treatment to date, numerous investigational compounds and antibodies have advanced to clinical trials (Table 3). The following sections will provide a detailed overview of these therapeutic candidates.

Table 3 Emerging Anti-Pulmonary fibrosis drugs in clinical development

Targeting signaling pathways

Current evidence spans the therapeutic agents from preclinical validation to clinical trial evaluation across different signaling pathways (including TGF-β, Wnt, PI3K-Akt, and Hippo-YAP signaling) for PF treatment. There are several effective strategies for inhibiting TGF-β. Preclinical approaches include blocking TGF-β synthesis using antisense oligonucleotides, targeting TGF-β ligands with proteoglycans or soluble TGF-β receptors, inhibiting TGF-β receptors through ALK5 inhibitors, and preventing the activation of latent TGF-β using neutralizing antibodies or small molecule inhibitors [117]. Notably, increased SKI expression effectively blocked BLM-induced TGF-β1 signaling activation and exerted an anti-fibrotic action in PF [187], suggesting that SKI modulation could serve as a novel therapeutic strategy for PF treatment. In line with this finding, apigenin suppresses TGF-β1 signaling by downregulating the Ski protein complex expression, which subsequently decreases both total and phosphorylated Smad2/3 levels (p-Smad2/3) [188]. As a result, this mechanism effectively inhibits fibroblast-to-myofibroblast differentiation and reduces ECM deposition. Additionally, the use of antagonists, small interfering RNA, and transcription inhibitors has proven effective in inhibiting Wnt signaling in fibrotic diseases [136, 189]. For the PI3K signaling pathway, targeted inhibitors such as Omipalisib and Rapamycin have been evaluated in clinical trials for PF treatment [172, 190]. In the context of the Hippo-YAP pathway, G-protein-coupled receptor (GPCR) signaling plays a significant role and serves as a promising therapeutic target [191]. Consequently, drugs directed at GPCRs may be utilized for the pharmacological targeting of Hippo-YAP signaling.

Targeting inflammation

Targeted anti-inflammatory therapies have emerged as a promising adjunctive strategy in the treatment of PF, focusing on modulating specific inflammatory mediators and immune pathways that contribute to disease progression. While the role of inflammation in PF has been debated historically, recent evidence has clarified that inflammation plays a significant role in the disease, particularly in the context of acute exacerbations and certain disease subtypes [192]. Key inflammatory mediators, such as cytokines (e.g., IL-13, IL-6), chemokines (e.g., CCL2), and growth factors like TGF-β, have been identified as critical drivers of fibroblast activation and extracellular matrix deposition, making them promising therapeutic targets [193]. Furthermore, recent studies have shown that blocking members of the tumor necrosis factor (TNF) superfamily, such as LIGHT (TNFSF14) and TL1A (TNFSF15), can significantly reverse fibrosis in preclinical models, further supporting the idea of targeting specific inflammatory drivers in PF therapy [194]. However, attempts to broadly suppress inflammation with corticosteroids and immunosuppressants have often been ineffective or even detrimental, underscoring the need for more precise, mechanism-based approaches [195, 196]. Despite the promising preclinical results, the clinical application of combination therapies that integrate both anti-inflammatory and anti-fibrotic strategies faces challenges related to patient selection, the risk of side effects, and the need for sustained, targeted immune modulation [197]. Nonetheless, the growing body of research and these promising findings provide a solid foundation for the future development of personalized, inflammation-targeted treatments for PF.

Drug delivery strategies

In the past decade, various drug delivery strategies and systems have been explored for the treatment of PF to minimize side effects (Fig. 4). Effective targeted delivery routes and technologies should possess several desirable characteristics, including reduced side effects, appropriate biodegradability, high bioavailability, and optimal drug efficacy [198]. Consequently, there has been a growing interest in the development of therapeutic approaches utilizing pulmonary drug delivery systems.

Fig. 4
figure 4

Drug delivery strategies for PF treatment. Drug delivery strategies of various therapeutic modalities (including nanoparticle delivery system, cell-mediated delivery system, and adeno-associated virus vectors-mediated delivery system) for the treatment of PF. Image Created in https://BioRender.com

Nanoparticle delivery system

Currently, conventional drug delivery approaches, such as oral administration, intravenous injection, nasal delivery, inhalation, and intratracheal administration, face significant limitations. Notably, certain routes (e.g., oral delivery) subject drugs to hepatic first-pass metabolism, reducing systemic bioavailability and impairing their therapeutic potential upon reaching target tissues like the lungs [199, 200]. To address these challenges, nanotechnology-based delivery systems have emerged as a transformative solution. By encapsulating therapeutics within engineered nanomaterials, these platforms significantly enhance drug stability, bioavailability, and targeted accumulation at disease sites [201, 202]. Among the most promising nanocarriers are liposomes, polymer-based nanoparticles, and mesoporous silica nanoparticles, each offering unique advantages for controlled release, tissue-specific targeting, and improved therapeutic outcomes in pulmonary and systemic diseases [203, 204]. The following describes various nanomaterial-based delivery approaches.

Liposomal Nanoparticles Delivery

Lipid nanoparticles are submicron capsules characterized by a homogeneous lipid core that is surrounded and stabilized by lipid layers [205, 206]. In recent years, they have emerged as a promising platform for drug delivery, with various liposomal formulations being utilized in a wide range of pharmaceutical applications [207, 208]. Notably, liposomes exhibit favorable properties, including reasonable biodegradability, excellent biocompatibility, and high encapsulation efficiency [209]. These nanoparticles can serve as effective delivery vehicles for oligonucleotides, peptides, and siRNA in the treatment of various diseases. In 2018, the U.S. FDA approved the first siRNA drug formulated with liposomes. Previous studies have shown that siRNA-loaded liposomes represent a promising strategy for siRNA-based therapy targeting PF [210, 211]. Furthermore, liposomes co-decorated with collagen-binding peptides and collagenase have the potential to specifically target fibrotic lung tissues and help restore normal lung architecture [212]. Thus, lipid nanoparticles have emerged as one of the most prominent nanocarriers, particularly important for the delivery of peptides and siRNAs.

Polymeric Nanoparticles Delivery

Polymeric nanoparticles are colloidal and offer enhanced drug loading efficiency and improved drug solubility [213]. These nanoparticles can either chemically conjugate active compounds or adsorb them onto their surface, functioning as either a reservoir (by dissolving or dispersing the actives) or a matrix (by entrapping the actives) [214]. However, several challenges must be addressed for effective pulmonary delivery of polymeric nanoparticles, including particle characteristics (such as size, shape, and surface charge), mucociliary clearance, and cellular internalization [215, 216]. A recent study conducted by Lee et al. utilized inhaled tacrolimus-loaded chitosan-coated poly nanoparticles in a mouse model of BLM-induced PF [217]. The surface modification of PLGA nanoparticles with chitosan offers several pharmaceutical advantages, including reduced burst effects of drug release and improved tissue retention and permeation. Another study demonstrated that GSE4-loaded PLGA nanoparticles could serve as a potential therapy for BLM-induced fibrosis by enhancing telomerase activity and reducing cell apoptosis and DNA damage [218]. These findings indicate that polymeric nanoparticles represent a promising strategy for the treatment of PF.

Mesoporous Silica nanoparticles Delivery

Mesoporous silica nanoparticles, characterized by their amorphous structure and lack of distinct geometric shape, are primarily composed of silicon dioxide. These nanoparticles are widely utilized in various applications, particularly in drug delivery, due to their exceptional biocompatibility, large surface area, and tunable particle size [219]. The use of mesoporous silica nanoparticles in drug delivery systems offers enhanced stability and presents a promising alternative to traditional delivery mechanisms [220]. Typically, the particle size and surface modifications of mesoporous silica nanoparticles play critical roles in influencing cellular uptake and therapeutic efficacy during targeted drug delivery processes [221, 222]. For instance, a nanosystem composed of Janus Au/mesoporous silica core/shell nanoparticles is designed for effective therapeutic and real-time tracing of pirfenidone in PF therapy [223]. Additionally, the intelligent nanodrug based on dendritic mesoporous silica nanoparticles loaded with the sonosensitizer protoporphyrin IX (PpIX) demonstrated significant anti-pulmonary fibrosis efficacy [224]. These findings underscore the significant potential of mesoporous silica nanoparticles in advancing biomedical sciences, particularly in the development of targeted drug delivery systems for PF treatment.

Cell-mediated delivery system

Recently, cell-mediated drug-delivery targeting systems have emerged as a significant area of research due to their therapeutic specificity in addressing various diseases [225]. These systems leverage the natural circulation of blood and the presence of surface ligands, which enhance therapeutic efficacy while minimizing side effects [226]. In the context of PF treatment, various types of cells can serve as effective drug carriers. By utilizing the unique properties of these cells, researchers aim to improve the precision of drug delivery, ensuring that therapeutic agents are effectively transported to the targeted sites of action. This approach not only holds promise for enhancing treatment outcomes but also for reducing the systemic exposure and associated adverse effects commonly seen with conventional drug delivery methods.

Macrophages-mediated Delivery

Macrophages, which are natural immune cells and antigen-presenting cells within the leukocyte lineage, originate from monocytes and play crucial roles as regulators of the innate immune system [227, 228]. They are integral to various biological processes, including responses to pathogens, the resolution of inflammation, and tissue repair [229]. Due to their long circulation time in the bloodstream and their ability to phagocytose foreign particles [230], macrophages present a promising platform for drug delivery systems aimed at treating diseases. Recent studies have demonstrated the use of macrophages to deliver dexamethasone, enhancing the therapeutic outcomes for PF by modulating the immune microenvironment [231]. In summary, macrophages represent a viable and effective vehicle for drug delivery, offering significant potential for improving therapeutic strategies in various diseases.

Neutrophil-mediated Delivery

Neutrophils are vital effector cells of the immune system and serve as the first line of defense against pathogens during inflammation and immune responses [232, 233]. They play a significant role in various chronic respiratory diseases, including asthma, chronic obstructive pulmonary disease (COPD), and PF. In the progression of these diseases, neutrophils can undergo profound infiltration and activation, triggering a cascade of inflammatory responses and cytokine release that ultimately leads to tissue remodeling and damage [234]. Similar to other immune cells, neutrophils possess the capability to facilitate targeted drug delivery. For instance, therapeutic nanoparticles combined with denatured bovine serum albumin (BSA) can be internalized by activated neutrophils, allowing them to migrate across blood vessels and deliver the therapeutic agents to inflamed tissues [235]. While neutrophil-mediated drug delivery has shown promise in targeting inflammatory diseases, current literature reveals no reported studies specifically investigating this approach for PF treatment. Given neutrophils’ natural tropism for fibrotic lesions and their demonstrated utility as drug carriers in other pathological contexts, this represents an unexplored therapeutic opportunity. Future research should evaluate: (1) neutrophil-hitchhiking strategies for antifibrotic agents (e.g., pirfenidone, nintedanib), (2) biomimetic neutrophil-membrane-coated nanoparticles for lung targeting, and (3) combination approaches leveraging neutrophils’ innate immune functions with precision drug delivery.

Stem Cell-mediated Delivery

Stem cells possess the unique ability to self-renew and differentiate into various specialized cell types [236]. Traditionally, stem cells are classified into two main categories based on their origin and plasticity: embryonic stem cells and somatic stem cells [237]. Recent research indicates that stem cells can produce a range of bioactive factors, including cytokines, chemokines, and antioxidant molecules, as well as angiogenic and antifibrotic agents during tissue repair processes [238,239,240]. Given these capabilities, stem cells represent a promising strategy for targeted drug delivery. For instance, Lata G. Menon et al. demonstrated the use of human bone marrow-derived mesenchymal stromal cells (hMSCs) as cellular vehicles for delivering tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) to glioma tumors [241]. Additionally, the administration of hMSCs engineered to produce interferon-beta (IFN-β) has been shown to inhibit the growth of pulmonary metastases through the systemic release of IFN-β at cancer sites [242]. In conclusion, stem cells may serve as a valuable platform for the targeted delivery of therapeutic agents, offering significant potential for enhancing treatment outcomes in various diseases.

Fibroblast-mediated Delivery

Targeting fibroblast and drug delivery has emerged as a promising therapeutic strategy against PF. For example, Sun et al. developed the clodronate-loaded liposomal and fibroblast-derived exosomal hybrid system as an available drug delivery strategy for PF treatment [243]. Also, researchers have engineered a core-shell microneedle platform for stable fibroblast delivery, featuring a cell-encapsulating gelatin methacryloyl hydrogel core surrounded by a protective polylactic-co-glycolic acid (PLGA) sheath. This minimally invasive design combines cellular viability preservation with structural robustness, providing a clinically adaptable solution for targeted fibroblast delivery [244]. Another promising strategy involves fibroblast-targeting dual siRNA-loaded micelles (including siPTPN13 and siNOX4), which offers a potential strategy with promising prospects in PF therapy [245]. Given the pivotal role of activated fibroblasts in pulmonary fibrogenesis, there is an urgent need to develop more targeted drug delivery strategies to effectively intervene in disease progression.

Adeno-associated Vectors-mediated delivery system

Gene therapy for PF has garnered attention with the advancement of viral vector technologies, particularly adeno-associated viruses (AAVs) and adenoviral vectors (AdVs). AAVs, such as AAV9 and AAV1, are favored due to their ability to target specific lung cells, including alveolar epithelial cells, fibroblasts, and endothelial cells, which are involved in the disease’s pathogenesis. AAVs are known for their low immunogenicity, which enables long-term gene expression and sustained therapeutic effects in chronic diseases like PF. Preclinical studies have shown that AAV9, used to overexpress sphingosine-1-phosphate lyase (SGPL1), reduces lung fibrosis by modulating pro-fibrotic pathways like TGF-β [246]. Additionally, AAV2/9-mediated delivery of LIM domain-only protein 7 (Lmo7) shRNA significantly ameliorated the progression of BLM-induced PF [247]. In contrast, AdVs offer high transduction efficiency and the ability to deliver larger genes but are limited by high immunogenicity, leading to transient expression and inflammation. New-generation adenoviral vectors, such as helper-dependent adenoviral vectors (HDAds), have been engineered to reduce immune responses and improve transgene persistence, showing promise in targeting key pathways like TGF-β signaling [248]. Despite these advancements, challenges such as efficient vector delivery, sustained expression, and overcoming immune responses remain. Moreover, no clinical trials have been published yet for AAVs or new-generation AdVs in PF treatment. Nonetheless, preclinical successes and the application of AAVs in other lung diseases, such as cystic fibrosis, provide a strong foundation for future clinical trials.

Conclusion and future perspective

While the understanding of its pathophysiology and potential therapeutic options have advanced in recent years, there remains no effective intervention to halt or cure PF. A variety of risk factors, including air pollution, smoking, aging, occupational exposure, environmental pollutants exposure, and microbial infections contribute to the progression of PF. Currently, only two drugs, pirfenidone and nintedanib, have been approved by the FDA for the treatment of PF. However, numerous other pharmacological agents targeting different aberrant signaling pathways are under development in clinical trials. Furthermore, the challenges posed by the low solubility and poor specificity of fibrosis-related therapeutic agents necessitate the creation of reliable delivery systems to enhance their efficacy. In summary, the current landscape of PF treatment highlights the urgent need for further research into the underlying pathogenic mechanisms, the identification of novel therapeutic targets, and the development of effective delivery systems for anti-fibrotic drugs.

Data availability

Not applicable.

Abbreviations

ARDS:

Acute respiratory distress syndrome

AAVs:

Adeno-associated viruses

AdVs:

Adenoviral vectors

Ams:

Alveolar macrophages

AST:

American Thoracic Society

Arg-1:

Arginase 1

α-SMA:

αsmooth muscle actin

BaP:

Benzo[a]pyrene

BLM:

Bleomycin

BSA:

Bovine serum albumin

COPD:

Chronic obstructive pulmonary disease

M1 macrophage:

Classically activated macrophages

CMV:

Cytomegalovirus

EndoMT:

Endothelial-to-mesenchymal transition

EMT:

Epithelial-mesenchymal transition

EBV:

Epstein–Barr virus

ERS:

European Respiratory Society

ECM:

Extracellular matrix

FPF:

Familial pulmonary fibrosis

FMT:

Fibroblast-to-myofibroblast transition

FoxOs:

Forkhead box proteins O

FDA:

Food and Drug Administration

GPCR:

G-protein-coupled receptor

GWAS:

Genome-wide association studies

HDAds:

Helper-dependent adenoviral vectors

HIV:

Human immunodeficiency virus

HTLV:

Human T-cell leukemia virus

Ims:

Interstitial macrophages

IFN-β:

Interferon-beta

iNOS:

Inducible nitric oxide synthase

JRS:

Japanese Respiratory Society

ALAT:

Latin American Thoracic Association

Lmo7:

LIM domain-only protein 7

MST1/2:

Mammalian sterile 20-like kinases 1 and 2

MMPs:

Matrix metalloproteinases

MOB1a/b:

MOB kinase activators 1 A and 1B

NOX4:

NADPH oxidase 4

NO:

Nitrogen oxides

PM:

Particularly particulate matter

PI3K:

Phosphoinositol-3 kinase

PDGF:

Platelet-derived growth factor

Pdgfrα:

Platelet-derived growth factor receptor alpha polypeptide

Pdgfrβ:

Platelet-derived growth factor receptor, beta polypeptide

PLGA:

Polylactic-co-glycolic acid

PS-NPs/PS-MPs:

Polystyrene nano- and microplastics

PpIX:

Protoporphyrin IX

PF:

Pulmonary fibrosis

PMVECs:

Pulmonary microvascular endothelial cells

PDK1:

Pyruvate dehydrogenase kinase 1

PDK2:

Pyruvate dehydrogenase kinase 2

ROS:

Reactive oxygen species

SAV1:

Salvador 1

SARS-CoV-2:

Severe acute respiratory syndrome coronavirus-2

SGPL1:

Sphingosine-1-phosphate lyase

SMCs:

Smooth muscle cells

SKI:

Sloan-Kettering virus

TGF-β1:

Transforming growth factor-beta 1

TAZ:

Transcriptional co-activator with PDZ-binding motif

AEC1:

Tumor necrosis factor (TNF), Type-1 alveolar epithelial

AEC2:

Type-2 alveolar epithelial

VE-cadherin:

Vascular endothelial cadherin

VEGF:

Vascular endothelial growth factor

YAP:

Yes-associated protein

EZH2:

Zeste homolog 2

References

  1. Sun Y, Ren Y, Song LY, Wang YY, Li TG, Wu YL, Li L, Yang ZS. Targeting iron-metabolism:a potential therapeutic strategy for pulmonary fibrosis. Biomed Pharmacother. 2024;172:116270.

    Article  CAS  PubMed  Google Scholar 

  2. Fang Y, Chung SSW, Xu L, Xue C, Liu X, Jiang D, Li R, Korogi Y, Yuan K, Saqi A, Hibshoosh H, Huang Y, Lin CS, Tsukui T, Sheppard D, Sun X, J., Que. RUNX2 promotes fibrosis via an alveolar-to-pathological fibroblast transition, Nature, (2025).

  3. Hu Y, Huang Y, Zong L, Lin J, Liu X, Ning S. Emerging roles of ferroptosis in pulmonary fibrosis: current perspectives, opportunities and challenges. Cell Death Discov. 2024;10:301.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Ren F, Aliper A, Chen J, Zhao H, Rao S, Kuppe C, Ozerov IV, Zhang M, Witte K, Kruse C, Aladinskiy V, Ivanenkov Y, Polykovskiy D, Fu Y, Babin E, Qiao J, Liang X, Mou Z, Wang H, Pun FW, Torres-Ayuso P, Veviorskiy A, Song D, Liu S, Zhang B, Naumov V, Ding X, Kukharenko A, Izumchenko E, Zhavoronkov A. A small-molecule TNIK inhibitor targets fibrosis in preclinical and clinical models. Nat Biotechnol. 2025;43:63–75.

    Article  CAS  PubMed  Google Scholar 

  5. Dempsey TM, Thao V, Moriarty JP, Borah BJ, Limper AH. Cost-effectiveness of the anti-fibrotics for the treatment of idiopathic pulmonary fibrosis in the united States. BMC Pulm Med. 2022;22:18.

    Article  PubMed  PubMed Central  Google Scholar 

  6. Rinciog C, Watkins M, Chang S, Maher TM, LeReun C, Esser D, Diamantopoulos A. A Cost-Effectiveness analysis of nintedanib in idiopathic pulmonary fibrosis in the UK. Volume 35. Pharmacoeconomics; 2017. pp. 479–91.

  7. Zheng XF, Xie BB, Liu Y, Zhu M, Zhang S, Ban CJ, Geng J, Jiang DY, Ren YH, Dai HP, Wang C. Direct medical costs of hospitalized patients with idiopathic pulmonary fibrosis in a tertiary hospital in China. Chin Med J (Engl). 2020;133:2498–500.

    Article  PubMed  Google Scholar 

  8. Wijsenbeek M, Suzuki A, Maher TM. Interstitial lung diseases. Lancet. 2022;400:769–86.

    Article  PubMed  Google Scholar 

  9. Gharaee-Kermani M, Hatano K, Nozaki Y, Phan SH. Gender-based differences in bleomycin-induced pulmonary fibrosis. Am J Pathol. 2005;166:1593–606.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Diamantopoulos A, Wright E, Vlahopoulou K, Cornic L, Schoof N, Maher T.M. The burden of illness of idiopathic pulmonary fibrosis: A comprehensive evidence review. PharmacoEconomics. 2018;36:779–807.

    Article  PubMed  PubMed Central  Google Scholar 

  11. Zhang Y, Lu W, Zhang X, Lu J, Xu S, Chen S, Zhong Z, Zhou T, Wang Q, Chen J, Liu P. Cryptotanshinone protects against pulmonary fibrosis through inhibiting Smad and STAT3 signaling pathways. Pharmacol Res. 2019;147:104307.

    Article  CAS  PubMed  Google Scholar 

  12. Lu W, Teoh A, Waters M, Haug G, Shakeel I, Hassan I, Shahzad AM, Callerfelt AL, Piccari L, Sohal SS. Pathology of idiopathic pulmonary fibrosis with particular focus on vascular endothelium and epithelial injury and their therapeutic potential. Pharmacol Ther. 2025;265:108757.

    Article  CAS  PubMed  Google Scholar 

  13. Ghanem M, Archer G, Justet A, Jaillet M, Vasarmidi E, Mordant P, Castier Y, Mal H, Cazes A, Pote N, Crestani B, Mailleux A. FGF21 signaling exerts antifibrotic properties during pulmonary fibrosis. Am J Respir Crit Care Med. 2025;211:486–98.

    Article  CAS  PubMed  Google Scholar 

  14. Wang Y, Jiao L, Qiang C, Chen C, Shen Z, Ding F, Lv L, Zhu T, Lu Y, Cui X. The role of matrix metalloproteinase 9 in fibrosis diseases and its molecular mechanisms. Biomed Pharmacother. 2024;171:116116.

    Article  CAS  PubMed  Google Scholar 

  15. Watanabe S, Markov NS, Lu Z, Piseaux Aillon R, Soberanes S, Runyan CE, Ren Z, Grant RA, Maciel M, Abdala-Valencia H, Politanska Y, Nam K, Sichizya L, Kihshen HG, Joshi N, McQuattie-Pimentel AC, Gruner KA, Jain M, Sznajder JI, Morimoto RI, Reyfman PA, Gottardi CJ, Budinger GRS, Misharin AV. Resetting proteostasis with ISRIB promotes epithelial differentiation to attenuate pulmonary fibrosis. Proc Natl Acad Sci U S A; 2021. p. 118.

  16. Borok Z, Horie M, Flodby P, Wang H, Liu Y, Ganesh S, Firth AL, Minoo P, Li C, Beers MF, Lee AS, Zhou B. Grp78 loss in epithelial progenitors reveals an Age-linked role for Endoplasmic reticulum stress in pulmonary fibrosis. Am J Respir Crit Care Med. 2020;201:198–211.

    Article  PubMed  PubMed Central  Google Scholar 

  17. Tzouvelekis A, Gomatou G, Bouros E, Trigidou R, Tzilas V, Bouros D. Common pathogenic mechanisms between idiopathic pulmonary fibrosis and lung Cancer. Chest. 2019;156:383–91.

    Article  PubMed  Google Scholar 

  18. Borchers AT, Chang C, Keen CL, Gershwin ME. Idiopathic pulmonary fibrosis-an epidemiological and pathological review. Clin Rev Allergy Immunol. 2011;40:117–34.

    Article  PubMed  Google Scholar 

  19. Kropski JA, Blackwell TS. Progress in Understanding and treating idiopathic pulmonary fibrosis. Annu Rev Med. 2019;70:211–24.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Allen RJ, Stockwell A, Oldham JM, Guillen-Guio B, Schwartz DA, Maher TM, Flores C, Noth I, Yaspan BL, Jenkins RG, Wain LV, International IPFGC. Genome-wide association study across five cohorts identifies five novel loci associated with idiopathic pulmonary fibrosis. Thorax. 2022;77:829–33.

    Article  PubMed  Google Scholar 

  21. Meyer KC. Pulmonary fibrosis, part I: epidemiology, pathogenesis, and diagnosis. Expert Rev Respir Med. 2017;11:343–59.

    CAS  PubMed  Google Scholar 

  22. Duckworth A, Gibbons MA, Allen RJ, Almond H, Beaumont RN, Wood AR, Lunnon K, Lindsay MA, Wain LV, Tyrrell J, Scotton CJ. Telomere length and risk of idiopathic pulmonary fibrosis and chronic obstructive pulmonary disease: a Mendelian randomisation study. Lancet Respir Med. 2021;9:285–94.

    Article  CAS  PubMed  Google Scholar 

  23. Nakano Y, Yang IV, Walts AD, Watson AM, Helling BA, Fletcher AA, Lara AR, Schwarz MI, Evans CM, Schwartz DA. MUC5B promoter variant rs35705950 affects MUC5B expression in the distal airways in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 2016;193:464–6.

    Article  PubMed  PubMed Central  Google Scholar 

  24. Kropski JA, Blackwell TS, Loyd JE. The genetic basis of idiopathic pulmonary fibrosis. Eur Respir J. 2015;45:1717–27.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Borie R, Crestani B, Dieude P, Nunes H, Allanore Y, Kannengiesser C, Airo P, Matucci-Cerinic M, Wallaert B, Israel-Biet D, Cadranel J, Cottin V, Gazal S, Peljto AL, Varga J, Schwartz DA, Valeyre D, Grandchamp B. The MUC5B variant is associated with idiopathic pulmonary fibrosis but not with systemic sclerosis interstitial lung disease in the European Caucasian population. PLoS ONE. 2013;8:e70621.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Peljto AL, Zhang Y, Fingerlin TE, Ma SF, Garcia JG, Richards TJ, Silveira LJ, Lindell KO, Steele MP, Loyd JE, Gibson KF, Seibold MA, Brown KK, Talbert JL, Markin C, Kossen K, Seiwert SD, Murphy E, Noth I, Schwarz MI, Kaminski N, Schwartz DA. Association between the MUC5B promoter polymorphism and survival in patients with idiopathic pulmonary fibrosis. JAMA. 2013;309:2232–9.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Petrovski S, Todd JL, Durheim MT, Wang Q, Chien JW, Kelly FL, Frankel C, Mebane CM, Ren Z, Bridgers J, Urban TJ, Malone CD, Finlen Copeland A, Brinkley C, Allen AS, O’Riordan T, McHutchison JG, Palmer SM. Goldstein, an exome sequencing study to assess the role of rare genetic variation in pulmonary fibrosis. Am J Respir Crit Care Med. 2017;196:82–93.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Michalski JE, Schwartz DA. Genetic risk factors for idiopathic pulmonary fibrosis: insights into Immunopathogenesis. J Inflamm Res. 2020;13:1305–18.

    Article  CAS  PubMed  Google Scholar 

  29. Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. Hallmarks of aging: an expanding universe. Cell. 2023;186:243–78.

    Article  CAS  PubMed  Google Scholar 

  30. Han JJ. The ticking of aging clocks. Trends Endocrinol Metab. 2024;35:11–22.

    Article  CAS  PubMed  Google Scholar 

  31. Ge T, Shao Y, Bao X, Xu W, Lu C. Cellular senescence in liver diseases: from mechanisms to therapies. Int Immunopharmacol. 2023;121:110522.

    Article  CAS  PubMed  Google Scholar 

  32. Cho SJ. Stout-Delgado, aging and lung disease. Annu Rev Physiol. 2020;82:433–59.

    Article  CAS  PubMed  Google Scholar 

  33. Lopez-Otin C, Blasco MA, Partridge L, Serrano M, Kroemer G. The hallmarks of aging. Cell. 2013;153:1194–217.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Cui H, Xie N, Banerjee S, Dey T, Liu RM, Antony VB, Sanders YY, Adams TS, Gomez JL, Thannickal VJ, Kaminski N, Liu G. CD38 mediates lung fibrosis by promoting alveolar epithelial cell aging. Am J Respir Crit Care Med. 2022;206:459–75.

    Article  PubMed  PubMed Central  Google Scholar 

  35. Han D, Gong H, Wei Y, Xu Y, Zhou X, Wang Z, Feng F. Hesperidin inhibits lung fibroblast senescence via IL-6/STAT3 signaling pathway to suppress pulmonary fibrosis. Phytomedicine. 2023;112:154680.

    Article  CAS  PubMed  Google Scholar 

  36. Alvarez D, Cardenes N, Sellares J, Bueno M, Corey C, Hanumanthu VS, Peng Y, D’Cunha H, Sembrat J, Nouraie M, Shanker S, Caufield C, Shiva S, Armanios M, Mora AL, Rojas M. IPF lung fibroblasts have a senescent phenotype. Am J Physiol Lung Cell Mol Physiol. 2017;313:L1164–73.

    Article  PubMed  PubMed Central  Google Scholar 

  37. Yoon HY, Kim SY, Kim OJ, Song JW. Nitrogen dioxide increases the risk of mortality in idiopathic pulmonary fibrosis. Eur Respir J, 57 (2021).

  38. Tomos I, Dimakopoulou K, Manali ED, Papiris SA, Karakatsani A. Long-term personal air pollution exposure and risk for acute exacerbation of idiopathic pulmonary fibrosis. Environ Health. 2021;20:99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Liang L, Cai Y, Lyu B, Zhang D, Chu S, Jing H, Rahimi K, Tong Z. Air pollution and hospitalization of patients with idiopathic pulmonary fibrosis in Beijing: a time-series study. Respir Res. 2022;23:81.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Lan D, Fermoyle CC, Troy LK, Knibbs LD, Corte TJ. The impact of air pollution on interstitial lung disease: a systematic review and meta-analysis. Front Med (Lausanne). 2023;10:1321038.

    Article  PubMed  Google Scholar 

  41. Cui F, Sun Y, Xie J, Li D, Wu M, Song L, Hu Y, Tian Y. Air pollutants, genetic susceptibility and risk of incident idiopathic pulmonary fibrosis. Eur Respir J, 61 (2023).

  42. Ning J, Du H, Zhang Y, Liu Q, Jiang T, Pang Y, Tian X, Yan L, Niu Y, Zhang R. N6-Methyladenosine modification of CDH1 mRNA promotes PM2.5-Induced pulmonary fibrosis via mediating epithelial mesenchymal transition. Toxicol Sci. 2022;185:143–57.

    Article  CAS  PubMed  Google Scholar 

  43. Chao CC, Hsiao SY, Kao WC, Chiou PC, Huang CC, Wang MT, Chen PC. Pyrroloquinoline Quinone ameliorates PM2.5-induced pulmonary fibrosis through targeting epithelial-mesenchymal transition. J Cell Mol Med. 2024;28:e18299.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Yue W, Tong L, Liu X, Weng X, Chen X, Wang D, Dudley S.C., Weir E.K., Ding W, Lu Z, Xu Y, Chen Y. Short term Pm2.5 exposure caused a robust lung inflammation, vascular remodeling, and exacerbated transition from left ventricular failure to right ventricular hypertrophy. Redox Biol. 2019;22:101161.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Wang J, He W, Yue H, Zhao P, Li J. Effective-components combination alleviates PM2.5-induced inflammation by evoking macrophage autophagy in COPD. J Ethnopharmacol. 2024;321:117537.

    Article  CAS  PubMed  Google Scholar 

  46. Zhang Y, Huang W, Zheng Z, Wang W, Yuan Y, Hong Q, Lin J, Li X, Meng Y. Cigarette smoke-inactivated SIRT1 promotes autophagy-dependent senescence of alveolar epithelial type 2 cells to induce pulmonary fibrosis. Free Radic Biol Med. 2021;166:116–27.

    Article  CAS  PubMed  Google Scholar 

  47. Pardo A, Selman M. The interplay of the genetic architecture, aging, and environmental factors in the pathogenesis of idiopathic pulmonary fibrosis. Am J Respir Cell Mol Biol. 2021;64:163–72.

    Article  CAS  PubMed  Google Scholar 

  48. Bellou V, Belbasis L, Evangelou E. Tobacco smoking and risk for pulmonary fibrosis: A prospective cohort study from the UK biobank. Chest. 2021;160:983–93.

    Article  PubMed  Google Scholar 

  49. Tsai M, Byun MK, Shin J, Crotty LE, Alexander. Effects of e-cigarettes and vaping devices on cardiac and pulmonary physiology. J Physiol. 2020;598:5039–62.

    Article  CAS  PubMed  Google Scholar 

  50. Zhao D, Aravindakshan A, Hilpert M, Olmedo P, Rule AM, Navas-Acien A, Aherrera A. Metal/Metalloid levels in electronic cigarette liquids, aerosols, and human biosamples: A systematic review. Environ Health Perspect. 2020;128:36001.

    Article  CAS  PubMed  Google Scholar 

  51. Sun L, Wang X, Gu T, Hu B, Luo J, Qin Y, Wan C. Nicotine triggers islet beta cell senescence to facilitate the progression of type 2 diabetes. Toxicology. 2020;441:152502.

    Article  CAS  PubMed  Google Scholar 

  52. Dai Y, Yang W, Song H, He X, Guan R, Wu Z, Jiang X, Li M, Liu P, Chen J. Long-term effects of chronic exposure to electronic cigarette aerosol on the cardiovascular and pulmonary system in mice: A comparative study to cigarette smoke. Environ Int. 2024;185:108521.

    Article  CAS  PubMed  Google Scholar 

  53. Spagnolo P, Ryerson CJ, Guler S, Feary J, Churg A, Fontenot AP, Piciucchi S, Udwadia Z, Corte TJ, Wuyts WA, Johannson KA, Cottin V. Occupational interstitial lung diseases. J Intern Med. 2023;294:798–815.

    Article  CAS  PubMed  Google Scholar 

  54. Murgia N, Akgun M, Blanc PD, Costa JT, Moitra S, Munoz X, Toren K, Ferreira AJ. Issue 3-The occupational burden of respiratory diseases, an update. Pulmonology. 2025;31:2416808.

    Article  CAS  PubMed  Google Scholar 

  55. Sui J, Boatz JC, Shi J, Hu Q, Li X, Zhang Y, Konigshoff M, Kliment CR. Loss of ANT1 increases fibrosis and epithelial cell senescence in idiopathic pulmonary fibrosis. Am J Respir Cell Mol Biol. 2023;69:556–69.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Zhang L, Tian J, Li N, Wang Y, Jin Y, Bian H, Xiong M, Zhang Z, Meng J, Han Z, Duan S. Exosomal MiRNA reprogramming in pyroptotic macrophage drives silica-induced fibroblast-to-myofibroblast transition and pulmonary fibrosis. J Hazard Mater. 2025;483:136629.

    Article  CAS  PubMed  Google Scholar 

  57. Jin C, Tao X, Zhang W, Xu H, Wu Y, Chen Q, Li S, Ning A, Wang W, Wu Q, Chu M. Multi-omics and multi-stages integration identified a novel variant associated with silicosis risk. Arch Toxicol. 2024;98:2907–18.

    Article  CAS  PubMed  Google Scholar 

  58. Li Z, Zhang W, Li S, Tao X, Xu H, Wu Y, Chen Q, Ning A, Tian T, Zhang L, Cui J, Wang W, Chu M. Integration of ApaQTL and eQTL analysis reveals novel SNPs associated with occupational pulmonary fibrosis risk. Arch Toxicol. 2024;98:2117–29.

    Article  CAS  PubMed  Google Scholar 

  59. Li B, Wang J, Zhao Y, Zou Y, Cao H, Jin H, Tao X, Mu M. Vitamin D3 reverses immune tolerance and enhances the cytotoxicity of effector T cells in coal pneumoconiosis. Ecotoxicol Environ Saf. 2024;271:115972.

    Article  CAS  PubMed  Google Scholar 

  60. Wang P, Xie D, Xiao T, Cheng C, Wang D, Sun J, Wu M, Yang Y, Zhang A, Liu Q. H3K18 lactylation promotes the progression of arsenite-related idiopathic pulmonary fibrosis via YTHDF1/m6A/NREP. J Hazard Mater. 2024;461:132582.

    Article  CAS  PubMed  Google Scholar 

  61. Li SR, Kang NN, Wang RR, Li MD, Chen LH, Zhou P, Xu DX, Zhao H, Fu L. ALKBH5 SUMOylation-mediated FBXW7 m6A modification regulates alveolar cells senescence during 1-nitropyrene-induced pulmonary fibrosis. J Hazard Mater. 2024;468:133704.

    Article  CAS  PubMed  Google Scholar 

  62. Li MD, Chen LH, Xiang HX, Jiang YL, Lv BB, Xu DX, Zhao H, Fu L. Benzo[a]pyrene evokes epithelial-mesenchymal transition and pulmonary fibrosis through AhR-mediated Nrf2-p62 signaling. J Hazard Mater. 2024;473:134560.

    Article  CAS  PubMed  Google Scholar 

  63. Sui A, Yao C, Chen Y, Li Y, Yu S, Qu J, Wei H, Tang J, Chen G. Polystyrene nanoplastics inhibit star expression by activating HIF-1alpha via ERK1/2 MAPK and AKT pathways in TM3 Leydig cells and testicular tissues of mice. Food Chem Toxicol. 2023;173:113634.

    Article  CAS  PubMed  Google Scholar 

  64. Zhang J, Du J, Liu D, Zhuo J, Chu L, Li Y, Gao L, Xu M, Chen W, Huang W, Xie L, Chen J, Meng X, Zou F, Cai S, Dong H. Polystyrene microplastics induce pulmonary fibrosis by promoting alveolar epithelial cell ferroptosis through cGAS/STING signaling. Ecotoxicol Environ Saf. 2024;277:116357.

    Article  CAS  PubMed  Google Scholar 

  65. Halimu G, Zhang Q, Liu L, Zhang Z, Wang X, Gu W, Zhang B, Dai Y, Zhang H, Zhang C, Xu M. Toxic effects of nanoplastics with different sizes and surface charges on epithelial-to-mesenchymal transition in A549 cells and the potential toxicological mechanism. J Hazard Mater. 2022;430:128485.

    Article  CAS  PubMed  Google Scholar 

  66. Liu J, Xu F, Guo M, Gao D, Song Y. Nasal instillation of polystyrene nanoplastics induce lung injury via mitochondrial DNA release and activation of the Cyclic GMP-AMP synthase-stimulator of interferon genes-signaling cascade. Sci Total Environ. 2024;948:174674.

    Article  CAS  PubMed  Google Scholar 

  67. Hatabu H, Kaye KM, Christiani DC. Viral infection, pulmonary fibrosis, and long COVID. Am J Respir Crit Care Med. 2023;207:647–9.

    Article  PubMed  Google Scholar 

  68. Matsunaga T, Sano H, Takita K, Morita M, Yamanaka S, Ichikawa T, Numakura T, Ida T, Jung M, Ogata S, Yoon S, Fujino N, Kyogoku Y, Sasaki Y, Koarai A, Tamada T, Toyama A, Nakabayashi T, Kageyama L, Kyuwa S, Inaba K, Watanabe S, Nagy P, Sawa T, Oshiumi H, Ichinose M, Yamada M, Sugiura H, Wei FY, Motohashi H, Akaike T. Supersulphides provide airway protection in viral and chronic lung diseases. Nat Commun. 2023;14:4476.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. George PM, Wells AU, Jenkins RG. Pulmonary fibrosis and COVID-19: the potential role for antifibrotic therapy. Lancet Respir Med. 2020;8:807–15.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. de Rooij L, Becker LM, Teuwen LA, Boeckx B, Jansen S, Feys S, Verleden S, Liesenborghs L, Stalder AK, Libbrecht S, Van Buyten T, Philips G, Subramanian A, Dumas SJ, Meta E, Borri M, Sokol L, Dendooven A, Truong AK, Gunst J, Van Mol P, Haslbauer JD, Rohlenova K, Menter T, Boudewijns R, Geldhof V, Vinckier S, Amersfoort J, Wuyts W, Van Raemdonck D, Jacobs W, Ceulemans LJ, Weynand B, Thienpont B, Lammens M, Kuehnel M, Eelen G, Dewerchin M, Schoonjans L, Jonigk D, van Dorpe J, Tzankov A, Wauters E, Mazzone M, Neyts J, Wauters J, Lambrechts D, Carmeliet P. The pulmonary vasculature in lethal COVID-19 and idiopathic pulmonary fibrosis at single-cell resolution. Cardiovasc Res. 2023;119:520–35.

    Article  PubMed  Google Scholar 

  71. Ashley SL, Jegal Y, Moore TA, van Dyk LF, Laouar Y. Moore, gamma-Herpes virus-68, but not Pseudomonas aeruginosa or influenza A (H1N1), exacerbates established murine lung fibrosis. Am J Physiol Lung Cell Mol Physiol. 2014;307:L219–230.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  72. Huang WJ, Tang XX. Virus infection induced pulmonary fibrosis. J Transl Med. 2021;19:496.

    Article  PubMed  PubMed Central  Google Scholar 

  73. Chen Y, He H, Luo L, Liu K, Jiang M, Li S, Zhang X, Yang X, Liu Q. Studying pulmonary fibrosis due to microbial infection via automated microscopic image analysis. Front Microbiol. 2023;14:1176339.

    Article  PubMed  PubMed Central  Google Scholar 

  74. Chabi ML, Goracci A, Roche N, Paugam A, Lupo A, Revel MP. Pulmonary aspergillosis. Diagn Interv Imaging. 2015;96:435–42.

    Article  CAS  PubMed  Google Scholar 

  75. Kasmani MY, Topchyan P, Brown AK, Brown RJ, Wu X, Chen Y, Khatun A, Alson D, Wu Y, Burns R, Lin CW, Kudek MR, Sun J, Cui W. A Spatial sequencing atlas of age-induced changes in the lung during influenza infection. Nat Commun. 2023;14:6597.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Reynolds D, Kollef M. The epidemiology and pathogenesis and treatment of Pseudomonas aeruginosa infections: an update. Volume 81. Drugs; 2021. pp. 2117–31.

  77. Wan X, Wang W, Zhu J, Xiao Y. Antibacterial peptide Reg4 ameliorates Pseudomonas aeruginosa-induced pulmonary inflammation and fibrosis. Microbiol Spectr. 2024;12:e0390523.

    Article  PubMed  Google Scholar 

  78. Wynn TA. Integrating mechanisms of pulmonary fibrosis. J Exp Med. 2011;208:1339–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Barkauskas CE, Noble PW. Cellular mechanisms of tissue fibrosis. 7. New insights into the cellular mechanisms of pulmonary fibrosis. Am J Physiol Cell Physiol. 2014;306:C987–996.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Katzen J, Beers MF. Contributions of alveolar epithelial cell quality control to pulmonary fibrosis. J Clin Invest. 2020;130:5088–99.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Shao M, Cheng H, Li X, Qiu Y, Zhang Y, Chang Y, Fu J, Shen M, Xu X, Feng D, Han Y, Yue S, Zhou Y, Luo Z. Abnormal mitochondrial iron metabolism damages alveolar type II epithelial cells involved in bleomycin-induced pulmonary fibrosis. Theranostics. 2024;14:2687–705.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Barkauskas CE, Cronce MJ, Rackley CR, Bowie EJ, Keene DR, Stripp BR, Randell SH, Noble PW, Hogan BL. Type 2 alveolar cells are stem cells in adult lung. J Clin Invest. 2013;123:3025–36.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Desai TJ, Brownfield DG, Krasnow MA. Alveolar progenitor and stem cells in lung development, renewal and cancer. Nature. 2014;507:190–4.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Chen H, He A, Li H, Chen H, Xie H, Luo L, Huang Y, Chen J, Guan J, He Q, Ma J, Ou C, Tao A, Yan J. TSSK4 upregulation in alveolar epithelial type-II cells facilitates pulmonary fibrosis through HSP90-AKT signaling restriction and AT-II apoptosis. Cell Death Dis. 2021;12:938.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Kim KK, Dotson MR, Agarwal M, Yang J, Bradley PB, Subbotina N, Osterholzer JJ, Sisson TH. Efferocytosis of apoptotic alveolar epithelial cells is sufficient to initiate lung fibrosis. Cell Death Dis. 2018;9:1056.

    Article  PubMed  PubMed Central  Google Scholar 

  86. Chen X, Xu H, Hou J, Wang H, Zheng Y, Li H, Cai H, Han X, Dai J. Epithelial cell senescence induces pulmonary fibrosis through Nanog-mediated fibroblast activation. Aging. 2019;12:242–59.

    Article  PubMed  PubMed Central  Google Scholar 

  87. Xu Y, Mizuno T, Sridharan A, Du Y, Guo M, Tang J, Wikenheiser-Brokamp KA, Perl AT, Funari VA, Gokey JJ, Stripp BR, Whitsett JA. Single-cell RNA sequencing identifies diverse roles of epithelial cells in idiopathic pulmonary fibrosis. JCI Insight. 2016;1:e90558.

    Article  PubMed  PubMed Central  Google Scholar 

  88. Yang J, Pan X, Wang L, Yu G. Alveolar cells under mechanical stressed niche: critical contributors to pulmonary fibrosis. Mol Med. 2020;26:95.

    Article  PubMed  PubMed Central  Google Scholar 

  89. Manfioletti G, Fedele M. Epithelial-Mesenchymal transition (EMT). Int J Mol Sci, 24 (2023).

  90. Park SJ, Kim TH, Lee K, Kang MA, Jang HJ, Ryu HW, Oh SR, Lee HJ. Kurarinone attenuates BLM-Induced pulmonary fibrosis via inhibiting TGF-beta signaling pathways. Int J Mol Sci, 22 (2021).

  91. Hua R, Yu J, Yan X, Ni Q, Zhi X, Li X, Jiang B, Zhu J. Syndecan-2 in colorectal cancer plays oncogenic role via epithelial-mesenchymal transition and MAPK pathway. Biomed Pharmacother. 2020;121:109630.

    Article  CAS  PubMed  Google Scholar 

  92. Gurski CJ, Dittel BN. Myeloperoxidase as a marker to differentiate mouse monocyte/macrophage subsets. Int J Mol Sci, 23 (2022).

  93. Mou Y, Wu GR, Wang Q, Pan T, Zhang L, Xu Y, Xiong W, Zhou Q, Wang Y. Macrophage-targeted delivery of SiRNA to silence Mecp2 gene expression attenuates pulmonary fibrosis. Bioeng Transl Med. 2022;7:e10280.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Spasov A, Kosolapov V, Babkov D, Klochkov V, Sokolova E, Miroshnikov M, Borisov A, Velikorodnaya Y, Smirnov A, Savateev K, Fedotov V, Kotovskaya S, Rusinov V. Discovery of Nitro-azolo[1,5-a]pyrimidines with Anti-Inflammatory and protective activity against LPS-Induced acute lung injury. Pharmaceuticals (Basel), 15 (2022).

  95. Vasse GF, Nizamoglu M, Heijink IH, Schleputz M, van Rijn P, Thomas MJ, Burgess JK, Melgert BN. Macrophage-stroma interactions in fibrosis: biochemical, biophysical, and cellular perspectives. J Pathol. 2021;254:344–57.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Locati M, Mantovani A, Sica A. Macrophage activation and polarization as an adaptive component of innate immunity. Adv Immunol. 2013;120:163–84.

    Article  CAS  PubMed  Google Scholar 

  97. Ge Z, Chen Y, Ma L, Hu F, Xie L. Macrophage polarization and its impact on idiopathic pulmonary fibrosis. Front Immunol. 2024;15:1444964.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Wang L, Li S, Yao Y, Yin W, Ye T. The role of natural products in the prevention and treatment of pulmonary fibrosis: a review. Food Funct. 2021;12:990–1007.

    Article  PubMed  Google Scholar 

  99. Bao X, Liu X, Liu N, Zhuang S, Yang Q, Ren H, Zhao D, Bai J, Zhou X, Tang L. Inhibition of EZH2 prevents acute respiratory distress syndrome (ARDS)-associated pulmonary fibrosis by regulating the macrophage polarization phenotype. Respir Res. 2021;22:194.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Polverino F, Balestro E, Spagnolo P. Clinical presentations, pathogenesis, and therapy of sarcoidosis: state of the Art. J Clin Med, 9 (2020).

  101. Medbury HJ, James V, Ngo J, Hitos K, Wang Y, Harris DC, Fletcher JP. Differing association of macrophage subsets with atherosclerotic plaque stability. Int Angiol. 2013;32:74–84.

    CAS  PubMed  Google Scholar 

  102. Tao B, Jin W, Xu J, Liang Z, Yao J, Zhang Y, Wang K, Cheng H, Zhang X, Ke Y. Myeloid-specific disruption of tyrosine phosphatase Shp2 promotes alternative activation of macrophages and predisposes mice to pulmonary fibrosis. J Immunol. 2014;193:2801–11.

    Article  CAS  PubMed  Google Scholar 

  103. Murray LA, Rosada R, Moreira AP, Joshi A, Kramer MS, Hesson DP, Argentieri RL, Mathai S, Gulati M, Herzog EL, Hogaboam CM. Serum amyloid P therapeutically attenuates murine bleomycin-induced pulmonary fibrosis via its effects on macrophages. PLoS ONE. 2010;5:e9683.

    Article  PubMed  PubMed Central  Google Scholar 

  104. Luo J, Li P, Dong M, Zhang Y, Lu S, Chen M, Zhou H, Lin N, Jiang H, Wang Y. SLC15A3 plays a crucial role in pulmonary fibrosis by regulating macrophage oxidative stress. Cell Death Differ. 2024;31:417–30.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Liu P, Miao K, Zhang L, Mou Y, Xu Y, Xiong W, Yu J, Wang Y. Curdione ameliorates bleomycin-induced pulmonary fibrosis by repressing TGF-beta-induced fibroblast to myofibroblast differentiation. Respir Res. 2020;21:58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Liu G, Philp AM, Corte T, Travis MA, Schilter H, Hansbro NG, Burns CJ, Eapen MS, Sohal SS, Burgess JK, Hansbro PM. Therapeutic targets in lung tissue remodelling and fibrosis. Pharmacol Ther. 2021;225:107839.

    Article  CAS  PubMed  Google Scholar 

  107. Pan X, Li J, Tu X, Wu C, Liu H, Luo Y, Dong X, Li X, Pan LL, Sun J. Lysine-specific demethylase-1 regulates fibroblast activation in pulmonary fibrosis via TGF-beta1/Smad3 pathway. Pharmacol Res. 2020;152:104592.

    Article  CAS  PubMed  Google Scholar 

  108. Xie T, Liang J, Liu N, Huan C, Zhang Y, Liu W, Kumar M, Xiao R, D’Armiento J, Metzger D, Chambon P, Papaioannou VE, Stripp BR, Jiang D, Noble PW. Transcription factor TBX4 regulates myofibroblast accumulation and lung fibrosis. J Clin Invest. 2016;126:3063–79.

    Article  PubMed  PubMed Central  Google Scholar 

  109. Tschumperlin DJ, Ligresti G, Hilscher MB, Shah VH. Mechanosensing and fibrosis. J Clin Invest. 2018;128:74–84.

    Article  PubMed  PubMed Central  Google Scholar 

  110. Wan YY, Tian GY, Guo HS, Kang YM, Yao ZH, Li XL, Liu QH, Lin DJ. Endostatin, an angiogenesis inhibitor, ameliorates bleomycin-induced pulmonary fibrosis in rats. Respir Res. 2013;14:56.

    Article  PubMed  PubMed Central  Google Scholar 

  111. Ackermann M, Werlein C, Plucinski E, Leypold S, Kuhnel MP, Verleden SE, Khalil HA, Langer F, Welte T, Mentzer SJ, Jonigk DD. The role of vasculature and angiogenesis in respiratory diseases, Angiogenesis, (2024).

  112. Gorelova A, Berman M, Ghouleh IA. Endothelial-to-Mesenchymal transition in pulmonary arterial hypertension. Antioxid Redox Signal. 2021;34:891–914.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Cheng D, Lian W, Jia X, Wang T, Sun W, Liu Y, Ni C. LGALS3 regulates endothelial-to-mesenchymal transition via PI3K/AKT signaling pathway in silica-induced pulmonary fibrosis. Toxicology. 2024;509:153962.

    Article  CAS  PubMed  Google Scholar 

  114. Su Q, Chen X, Ling X, Li D, Ren X, Zhao Y, Yang Y, Liu Y, He A, Zhu X, Yang X, Lu W, Wu H, Qi Y. SUMOylation of Smad2 mediates TGF-beta-regulated endothelial-mesenchymal transition. J Biol Chem. 2023;299:105244.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  115. Zhang R, Tan Y, Yong C, Jiao Y, Tang X, Wang D. Pirfenidone ameliorates early pulmonary fibrosis in LPS-induced acute respiratory distress syndrome by inhibiting endothelial-to-mesenchymal transition via the Hedgehog signaling pathway. Int Immunopharmacol. 2022;109:108805.

    Article  CAS  PubMed  Google Scholar 

  116. Liu X, Qin X, Qin H, Jia C, Yuan Y, Sun T, Chen B, Chen C, Zhang H. Characterization of the heterogeneity of endothelial cells in bleomycin-induced lung fibrosis using single-cell RNA sequencing. Angiogenesis. 2021;24:809–21.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Ong CH, Tham CL, Harith HH, Firdaus N, Israf DA. TGF-beta-induced fibrosis: A review on the underlying mechanism and potential therapeutic strategies. Eur J Pharmacol. 2021;911:174510.

    Article  CAS  PubMed  Google Scholar 

  118. Massague J, Sheppard D. TGF-beta signaling in health and disease. Cell. 2023;186:4007–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  119. Saito A, Horie M, Nagase T. TGF-beta signaling in lung health and disease. Int J Mol Sci, 19 (2018).

  120. Nickel J, Ten Dijke P, Mueller TD. TGF-beta family co-receptor function and signaling. Acta Biochim Biophys Sin (Shanghai). 2018;50:12–36.

    Article  CAS  PubMed  Google Scholar 

  121. Chaikuad A, Bullock AN. Structural basis of intracellular TGF-beta signaling: receptors and Smads. Cold Spring Harb Perspect Biol, 8 (2016).

  122. Meng XM, Nikolic-Paterson DJ, Lan HY. TGF-beta: the master regulator of fibrosis. Nat Rev Nephrol. 2016;12:325–38.

    Article  CAS  PubMed  Google Scholar 

  123. Zhang XL, Zhang XY, Ge XQ, Liu MX. Mangiferin prevents hepatocyte epithelial-mesenchymal transition in liver fibrosis via targeting HSP27-mediated JAK2/STAT3 and TGF-beta1/Smad pathway. Phytother Res. 2022;36:4167–82.

    Article  CAS  PubMed  Google Scholar 

  124. Li J, Ge F, Wuken S, Jiao S, Chen P, Huang M, Gao X, Liu J, Tu P, Chai X, Huang L. Zerumbone, a humulane sesquiterpene from Syringa pinnatifolia, attenuates cardiac fibrosis by inhibiting of the TGF-beta1/Smad signaling pathway after myocardial infarction in mice. Phytomedicine. 2022;100:154078.

    Article  PubMed  Google Scholar 

  125. Gifford CC, Tang J, Costello A, Khakoo NS, Nguyen TQ, Goldschmeding R, Higgins PJ, Samarakoon R. Negative regulators of TGF-beta1 signaling in renal fibrosis; pathological mechanisms and novel therapeutic opportunities. Clin Sci (Lond). 2021;135:275–303.

    Article  CAS  PubMed  Google Scholar 

  126. Schepers D, Doyle AJ, Oswald G, Sparks E, Myers L, Willems PJ, Mansour S, Simpson MA, Frysira H, Maat-Kievit A, Van Minkelen R, Hoogeboom JM, Mortier GR, Titheradge H, Brueton L, Starr L, Stark Z, Ockeloen C, Lourenco CM, Blair E, Hobson E, Hurst J, Maystadt I, Destree A, Girisha KM, Miller M, Dietz HC, Loeys B, Van Laer L. The SMAD-binding domain of SKI: a hotspot for de Novo mutations causing Shprintzen-Goldberg syndrome. Eur J Hum Genet. 2015;23:224–8.

    Article  CAS  PubMed  Google Scholar 

  127. Li P, Wang QS, Zhai Y, Xiong RP, Chen X, Liu P, Peng Y, Zhao Y, Ning YL, Yang N, Zhou YG. Ski mediates TGF-beta1-induced fibrosarcoma cell proliferation and promotes tumor growth. J Cancer. 2020;11:5929–40.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  128. Yang H, Zhan L, Yang T, Wang L, Li C, Zhao J, Lei Z, Li X, Zhang HT. Ski prevents TGF-beta-induced EMT and cell invasion by repressing SMAD-dependent signaling in non-small cell lung cancer. Oncol Rep. 2015;34:87–94.

    Article  CAS  PubMed  Google Scholar 

  129. Finnson KW, Almadani Y, Philip A. Non-canonical (non-SMAD2/3) TGF-beta signaling in fibrosis: mechanisms and targets. Semin Cell Dev Biol. 2020;101:115–22.

    Article  CAS  PubMed  Google Scholar 

  130. Inui N, Sakai S, Kitagawa M. Molecular pathogenesis of pulmonary fibrosis, with focus on pathways related to TGF-beta and the Ubiquitin-Proteasome pathway. Int J Mol Sci, 22 (2021).

  131. Blobe GC, Schiemann WP, Lodish HF. Role of transforming growth factor beta in human disease. N Engl J Med. 2000;342:1350–8.

    Article  CAS  PubMed  Google Scholar 

  132. Bauman KA, Wettlaufer SH, Okunishi K, Vannella KM, Stoolman JS, Huang SK, Courey AJ, White ES, Hogaboam CM, Simon RH, Toews GB, Sisson TH, Moore BB. Peters-Golden, the antifibrotic effects of plasminogen activation occur via prostaglandin E2 synthesis in humans and mice. J Clin Invest. 2010;120:1950–60.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Duchartre Y, Kim YM, Kahn M. The Wnt signaling pathway in cancer. Crit Rev Oncol Hematol. 2016;99:141–9.

    Article  PubMed  Google Scholar 

  134. Qi X, Hu Q, Elghobashi-Meinhardt N, Long T, Chen H, Li X. Molecular basis of Wnt biogenesis, secretion, and Wnt7-specific signaling. Cell. 2023;186:5028–e50405014.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  135. Guo X, Zhu Y, Sun Y, Li X. IL-6 accelerates renal fibrosis after acute kidney injury via DNMT1-dependent FOXO3a methylation and activation of Wnt/beta-catenin pathway. Int Immunopharmacol. 2022;109:108746.

    Article  CAS  PubMed  Google Scholar 

  136. Hu HH, Cao G, Wu XQ, Vaziri ND, Zhao YY. Wnt signaling pathway in aging-related tissue fibrosis and therapies. Ageing Res Rev. 2020;60:101063.

    Article  CAS  PubMed  Google Scholar 

  137. Ming WH, Luan ZL, Yao Y, Liu HC, Hu SY, Du CX, Zhang C, Zhao YH, Huang YZ, Sun XW, Qiao RF, Xu H, Guan YF, Zhang XY. Pregnane X receptor activation alleviates renal fibrosis in mice via interacting with p53 and inhibiting the Wnt7a/beta-catenin signaling. Acta Pharmacol Sin. 2023;44:2075–90.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  138. Duspara K, Bojanic K, Pejic JI, Kuna L, Kolaric TO, Nincevic V, Smolic R, Vcev A, Glasnovic M, Curcic IB, Smolic M. Targeting the Wnt signaling pathway in liver fibrosis for drug options: an update. J Clin Transl Hepatol. 2021;9:960–71.

    PubMed  PubMed Central  Google Scholar 

  139. Staal FJ, Clevers HC. WNT signalling and haematopoiesis: a WNT-WNT situation. Nat Rev Immunol. 2005;5:21–30.

    Article  CAS  PubMed  Google Scholar 

  140. Sidaway P. Prostate cancer: Wnt signalling induces resistance. Nat Rev Urol. 2015;12:597.

    Article  CAS  PubMed  Google Scholar 

  141. Cheng X, Xu X, Chen D, Zhao F, Wang W. Therapeutic potential of targeting the Wnt/beta-catenin signaling pathway in colorectal cancer. Biomed Pharmacother. 2019;110:473–81.

    Article  CAS  PubMed  Google Scholar 

  142. Chilosi M, Poletti V, Zamo A, Lestani M, Montagna L, Piccoli P, Pedron S, Bertaso M, Scarpa A, Murer B, Cancellieri A, Maestro R, Semenzato G, Doglioni C. Aberrant Wnt/beta-catenin pathway activation in idiopathic pulmonary fibrosis. Am J Pathol. 2003;162:1495–502.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Konigshoff M, Eickelberg O. WNT signaling in lung disease: a failure or a regeneration signal? Am J Respir Cell Mol Biol. 2010;42:21–31.

    Article  PubMed  Google Scholar 

  144. Crosby LM, Waters CM. Epithelial repair mechanisms in the lung. Am J Physiol Lung Cell Mol Physiol. 2010;298:L715–731.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Nelson WJ, Nusse R. Convergence of Wnt, beta-catenin, and Cadherin pathways. Science. 2004;303:1483–7.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Wang J, Hu K, Cai X, Yang B, He Q, Wang J, Weng Q. Targeting PI3K/AKT signaling for treatment of idiopathic pulmonary fibrosis. Acta Pharm Sin B. 2022;12:18–32.

    Article  PubMed  Google Scholar 

  147. Liu H, Wang J, Zhang Q, Geng L, Yang Y, Wu N. Protective effect of fucoidan against MPP(+)-Induced SH-SY5Y cells apoptosis by affecting the PI3K/Akt pathway. Mar Drugs, 18 (2020).

  148. Alzahrani AS. PI3K/Akt/mTOR inhibitors in cancer: at the bench and bedside. Semin Cancer Biol. 2019;59:125–32.

    Article  CAS  PubMed  Google Scholar 

  149. Guo H, German P, Bai S, Barnes S, Guo W, Qi X, Lou H, Liang J, Jonasch E, Mills GB, Ding Z. The PI3K/AKT pathway and renal cell carcinoma. J Genet Genomics. 2015;42:343–53.

    Article  PubMed  PubMed Central  Google Scholar 

  150. Song F, Wang CG, Wang TL, Tao YC, Mao JZ, Hu CW, Zhang Y, Tang PJ, Lu CL, Qing HL, Han L, Chen Z. Enhancement of gemcitabine sensitivity in intrahepatic cholangiocarcinoma through Saikosaponin-a mediated modulation of the p-AKT/BCL-6/ABCA1 axis. Phytomedicine. 2024;133:155944.

    Article  CAS  PubMed  Google Scholar 

  151. Qin W, Cao L, Massey IY. Role of PI3K/Akt signaling pathway in cardiac fibrosis. Mol Cell Biochem. 2021;476:4045–59.

    Article  CAS  PubMed  Google Scholar 

  152. Hu X, Xu Q, Wan H, Hu Y, Xing S, Yang H, Gao Y, He Z. PI3K-Akt-mTOR/PFKFB3 pathway mediated lung fibroblast aerobic Glycolysis and collagen synthesis in lipopolysaccharide-induced pulmonary fibrosis. Lab Invest. 2020;100:801–11.

    Article  CAS  PubMed  Google Scholar 

  153. Lu Y, Azad N, Wang L, Iyer AK, Castranova V, Jiang BH, Rojanasakul Y. Phosphatidylinositol-3-kinase/akt regulates bleomycin-induced fibroblast proliferation and collagen production. Am J Respir Cell Mol Biol. 2010;42:432–41.

    Article  CAS  PubMed  Google Scholar 

  154. Yan W, Wu Q, Yao W, Li Y, Liu Y, Yuan J, Han R, Yang J, Ji X, Ni C. MiR-503 modulates epithelial-mesenchymal transition in silica-induced pulmonary fibrosis by targeting PI3K p85 and is sponged by LncRNA MALAT1. Sci Rep. 2017;7:11313.

    Article  PubMed  PubMed Central  Google Scholar 

  155. Wang Y, Zhang L, Wu GR, Zhou Q, Yue H, Rao LZ, Yuan T, Mo B, Wang FX, Chen LM, Sun F, Song J, Xiong F, Zhang S, Yu Q, Yang P, Xu Y, Zhao J, Zhang H, Xiong W, Wang CY. MBD2 serves as a viable target against pulmonary fibrosis by inhibiting macrophage M2 program. Sci Adv, 7 (2021).

  156. Zheng Z, Li C, Shao G, Li J, Xu K, Zhao Z, Zhang Z, Liu J, Wu H. Hippo-YAP/MCP-1 mediated tubular maladaptive repair promote inflammation in renal failed recovery after ischemic AKI. Cell Death Dis. 2021;12:754.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  157. Song J, Wang T, Chi X, Wei X, Xu S, Yu M, He H, Ma J, Li X, Du J, Sun X, Wang Y, Zhan J, Zhang H. Kindlin-2 inhibits the Hippo signaling pathway by promoting degradation of MOB1. Cell Rep. 2019;29:3664–e36773665.

    Article  CAS  PubMed  Google Scholar 

  158. Pan C, Hao X, Deng X, Lu F, Liu J, Hou W, Xu T. The roles of Hippo/YAP signaling pathway in physical therapy. Cell Death Discov. 2024;10:197.

    Article  PubMed  PubMed Central  Google Scholar 

  159. Zhang Z, Wang Y, Xu Q, Zhou X, Ling Y, Zhang J, Mao L. Methyl Canthin-6-one-2-carboxylate restrains the migration/invasion properties of Fibroblast-like synoviocytes by suppressing the Hippo/YAP signaling pathway. Pharmaceuticals (Basel), 16 (2023).

  160. Moya IM, Halder G. Hippo-YAP/TAZ signalling in organ regeneration and regenerative medicine. Nat Rev Mol Cell Biol. 2019;20:211–26.

    Article  CAS  PubMed  Google Scholar 

  161. Liu F, Lagares D, Choi KM, Stopfer L, Marinkovic A, Vrbanac V, Probst CK, Hiemer SE, Sisson TH, Horowitz JC, Rosas IO, Fredenburgh LE, Feghali-Bostwick C, Varelas X, Tager AM, Tschumperlin DJ. Mechanosignaling through YAP and TAZ drives fibroblast activation and fibrosis. Am J Physiol Lung Cell Mol Physiol. 2015;308:L344–357.

    Article  CAS  PubMed  Google Scholar 

  162. Du W, Tang Z, Yang F, Liu X, Dong J. Icariin attenuates bleomycin-induced pulmonary fibrosis by targeting Hippo/YAP pathway. Biomed Pharmacother. 2021;143:112152.

    Article  CAS  PubMed  Google Scholar 

  163. Mahoney JE, Mori M, Szymaniak AD, Varelas X, Cardoso WV. The Hippo pathway effector Yap controls patterning and differentiation of airway epithelial progenitors. Dev Cell. 2014;30:137–50.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. LaCanna R, Liccardo D, Zhang P, Tragesser L, Wang Y, Cao T, Chapman HA, Morrisey EE, Shen H, Koch WJ, Kosmider B, Wolfson MR, Tian Y. Yap/Taz regulate alveolar regeneration and resolution of lung inflammation. J Clin Invest. 2019;129:2107–22.

    Article  PubMed  PubMed Central  Google Scholar 

  165. Cores J, Hensley MT, Kinlaw K, Rikard SM, Dinh PU, Paudel D, Tang J, Vandergriff AC, Allen TA, Li Y, Liu J, Niu B, Chi Y, Caranasos T, Lobo LJ, Cheng K. Safety and efficacy of allogeneic lung spheroid cells in a mismatched rat model of pulmonary fibrosis. Stem Cells Transl Med. 2017;6:1905–16.

    Article  PubMed  PubMed Central  Google Scholar 

  166. Kreuter M, Picker N, Schwarzkopf L, Baumann S, Cerani A, Postema R, Maywald U, Dittmar A, Langley J, Patel H. Epidemiology, healthcare utilization, and related costs among patients with IPF: results from a German claims database analysis. Respir Res. 2022;23:62.

    Article  PubMed  PubMed Central  Google Scholar 

  167. Raimundo K, Chang E, Broder MS, Alexander K, Zazzali J, Swigris JJ. Clinical and economic burden of idiopathic pulmonary fibrosis: a retrospective cohort study. BMC Pulm Med. 2016;16:2.

    Article  PubMed  PubMed Central  Google Scholar 

  168. Rinciog C, Diamantopoulos A, Gentilini A, Bondue B, Dahlqvist C, Froidure A, Wuyts WA, Soulard S. Cost-Effectiveness analysis of nintedanib versus Pirfenidone in idiopathic pulmonary fibrosis in Belgium. Pharmacoecon Open. 2020;4:449–58.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  169. Raghu G, Collard HR, Egan JJ, Martinez FJ, Behr J, Brown KK, Colby TV, Cordier JF, Flaherty KR, Lasky JA, Lynch DA, Ryu JH, Swigris JJ, Wells AU, Ancochea J, Bouros D, Carvalho C, Costabel U, Ebina M, Hansell DM, Johkoh T, Kim DS, King TE Jr., Kondoh Y, Myers J, Muller NL, Nicholson AG, Richeldi L, Selman M, Dudden RF, Griss BS, Protzko SL, Schunemann HJ, Fibrosis P. An official ATS/ERS/JRS/ALAT statement: idiopathic pulmonary fibrosis: evidence-based guidelines for diagnosis and management, Am J Respir Crit Care Med, 183 (2011) 788–824.

  170. King TE Jr., Bradford WZ, Castro-Bernardini S, Fagan EA, Glaspole I, Glassberg MK, Gorina E, Hopkins PM, Kardatzke D, Lancaster L, Lederer DJ, Nathan SD, Pereira CA, Sahn SA, Sussman R, Swigris JJ, Noble PW, Group AS. A phase 3 trial of Pirfenidone in patients with idiopathic pulmonary fibrosis. N Engl J Med. 2014;370:2083–92.

    Article  PubMed  Google Scholar 

  171. Flaherty KR, Wells AU, Cottin V, Devaraj A, Walsh SLF, Inoue Y, Richeldi L, Kolb M, Tetzlaff K, Stowasser S, Coeck C, Clerisme-Beaty E, Rosenstock B, Quaresma M, Haeufel T, Goeldner RG, Schlenker-Herceg R, Brown KK. Investigators, nintedanib in progressive fibrosing interstitial lung diseases. N Engl J Med. 2019;381:1718–27.

    Article  CAS  PubMed  Google Scholar 

  172. Mercer P.F., Woodcock H.V., Eley J.D., Plate M., Sulikowski M.G., Durrenberger P.F., Franklin L., Nanthakumar C.B., Man Y., Genovese F., McAnulty R.J., Yang S., Maher T.M., Nicholson A.G., Blanchard A.D., Marshall R.P., Lukey P.T., Chambers R.C. Exploration of a potent PI3 kinase/mtor inhibitor as a novel anti-fibrotic agent in IPF. Thorax. 2016;71:701–11.

  173. Maher TM, Simpson JK, Porter JC, Wilson FJ, Chan R, Eames R, Cui Y, Siederer S, Parry S, Kenny J, Slack RJ, Sahota J, Paul L, Saunders P, Molyneaux PL, Lukey PT, Rizzo G, Searle GE, Marshall RP, Saleem A, Kang’ombe AR, Fairman D, Fahy WA. Vahdati-Bolouri, A positron emission tomography imaging study to confirm target engagement in the lungs of patients with idiopathic pulmonary fibrosis following a single dose of a novel inhaled alphavbeta6 integrin inhibitor. Respir Res. 2020;21:75.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Desroy N, Housseman C, Bock X, Joncour A, Bienvenu N, Cherel L, Labeguere V, Rondet E, Peixoto C, Grassot JM, Picolet O, Annoot D, Triballeau N, Monjardet A, Wakselman E, Roncoroni V, Le Tallec S, Blanque R, Cottereaux C, Vandervoort N, Christophe T, Mollat P, Lamers M, Auberval M, Hrvacic B, Ralic J, Oste L, van der Aar E, Brys R, Heckmann B. thiazole-5-carbonitrile (GLPG1690), a First-in-Class autotaxin inhibitor undergoing clinical evaluation for the treatment of idiopathic pulmonary fibrosis. J Med Chem. 2017;60:3580–90. Discovery of 2-[[2-Ethyl-6-[4-[2-(3-hydroxyazetidin-1-yl)-2-oxoethyl]piperazin-1-yl]-8-methylimidazo[1,2-a]pyridin-3-yl]methylamino]-4-(4-fluorophenyl).

    Article  CAS  PubMed  Google Scholar 

  175. Khalil N, Manganas H, Ryerson CJ, Shapera S, Cantin AM, Hernandez P, Turcotte EE, Parker JM, Moran JE, Albert GR, Sawtell R, Hagerimana A, Laurin P, Gagnon L, Cesari F, Kolb M. Phase 2 clinical trial of PBI-4050 in patients with idiopathic pulmonary fibrosis. Eur Respir J, 53 (2019).

  176. Hirani N, MacKinnon AC, Nicol L, Ford P, Schambye H, Pedersen A, Nilsson UJ, Leffler H, Sethi T, Tantawi S, Gravelle L, Slack RJ, Mills R, Karmakar U, Humphries D, Zetterberg F, Keeling L, Paul L, Molyneaux PL, Li F, Funston W, Forrest IA, Simpson AJ, Gibbons MA, Maher TM. Target Inhibition of galectin-3 by inhaled TD139 in patients with idiopathic pulmonary fibrosis. Eur Respir J, 57 (2021).

  177. Hanyu F, Zheng H, Jiaqi W, Tairan D, Yiyuanzi Z, Qiwen Y, Ying L, Hongchun Z, Lu L. Protective effects and mechanism of Curcumin in animal models of pulmonary fibrosis: a preclinical systematic review and meta-analysis. Front Pharmacol. 2023;14:1258885.

    Article  PubMed  PubMed Central  Google Scholar 

  178. Singh N, Nagar E, Gautam A, Kapoor H, Arora N. Resveratrol mitigates miR-212-3p mediated progression of diesel exhaust-induced pulmonary fibrosis by regulating SIRT1/FoxO3. Sci Total Environ. 2023;902:166063.

    Article  CAS  PubMed  Google Scholar 

  179. Jiang J, Kao TC, Hu S, Li Y, Feng W, Guo X, Zeng J, Ma X. Protective role of Baicalin in the dynamic progression of lung injury to idiopathic pulmonary fibrosis: A meta-analysis. Phytomedicine. 2023;114:154777.

    Article  CAS  PubMed  Google Scholar 

  180. Nambiar A, Kellogg D 3rd, Justice J, Goros M, Gelfond J, Pascual R, Hashmi S, Masternak M, Prata L, LeBrasseur N, Limper A, Kritchevsky S, Musi N, Tchkonia T, Kirkland J. Senolytics dasatinib and quercetin in idiopathic pulmonary fibrosis: results of a phase I, single-blind, single-center, randomized, placebo-controlled pilot trial on feasibility and tolerability, EBioMedicine, 90 (2023) 104481.

  181. Lu P, Li J, Liu C, Yang J, Peng H, Xue Z, Liu Z. Salvianolic acid B dry powder inhaler for the treatment of idiopathic pulmonary fibrosis. Asian J Pharm Sci. 2022;17:447–61.

    CAS  PubMed  PubMed Central  Google Scholar 

  182. Zhou Y, Li M, Shen T, Yang T, Shi G, Wei Y, Chen C, Wang D, Wang Y, Zhang T. Celastrol targets Cullin-Associated and Neddylation-Dissociated 1 to prevent Fibroblast-Myofibroblast transformation against pulmonary fibrosis. ACS Chem Biol. 2022;17:2734–43.

    Article  CAS  PubMed  Google Scholar 

  183. Khanna D, Lin CJF, Furst DE, Goldin J, Kim G, Kuwana M, Allanore Y, Matucci-Cerinic M, Distler O, Shima Y, van Laar JM, Spotswood H, Wagner B, Siegel J, Jahreis A, Denton CP. i. focu, Tocilizumab in systemic sclerosis: a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet Respir Med. 2020;8:963–74.

    Article  CAS  PubMed  Google Scholar 

  184. Richeldi L, Fernandez Perez ER, Costabel U, Albera C, Lederer DJ, Flaherty KR, Ettinger N, Perez R, Scholand MB, Goldin J, Peony Yu KH, Neff T, Porter S, Zhong M, Gorina E, Kouchakji E, Raghu G. Pamrevlumab, an anti-connective tissue growth factor therapy, for idiopathic pulmonary fibrosis (PRAISE): a phase 2, randomised, double-blind, placebo-controlled trial. Lancet Respir Med. 2020;8:25–33.

    Article  CAS  PubMed  Google Scholar 

  185. Raghu G, Brown KK, Collard HR, Cottin V, Gibson KF, Kaner RJ, Lederer DJ, Martinez FJ, Noble PW, Song JW, Wells AU, Whelan TP, Wuyts W, Moreau E, Patterson SD, Smith V, Bayly S, Chien JW, Gong Q, Zhang JJ. O’Riordan, efficacy of Simtuzumab versus placebo in patients with idiopathic pulmonary fibrosis: a randomised, double-blind, controlled, phase 2 trial. Lancet Respir Med. 2017;5:22–32.

    Article  CAS  PubMed  Google Scholar 

  186. Parker JM, Glaspole IN, Lancaster LH, Haddad TJ, She D, Roseti SL, Fiening JP, Grant EP, Kell CM, Flaherty KR. A phase 2 randomized controlled study of tralokinumab in subjects with idiopathic pulmonary fibrosis. Am J Respir Crit Care Med. 2018;197:94–103.

    Article  CAS  PubMed  Google Scholar 

  187. Bisserier M, Milara J, Abdeldjebbar Y, Gubara S, Jones C, Bueno-Beti C, Chepurko E, Kohlbrenner E, Katz MG, Tarzami S, Cortijo J, Leopold J, Hajjar RJ, Sassi Y, Hadri L. AAV1.SERCA2a gene therapy reverses pulmonary fibrosis by blocking the STAT3/FOXM1 pathway and promoting the SNON/SKI Axis. Mol Ther. 2020;28:394–410.

    Article  CAS  PubMed  Google Scholar 

  188. Wang F, Fan K, Zhao Y, Xie ML. Apigenin attenuates TGF-beta1-stimulated cardiac fibroblast differentiation and extracellular matrix production by targeting miR-155-5p/c-Ski/Smad pathway. J Ethnopharmacol. 2021;265:113195.

    Article  CAS  PubMed  Google Scholar 

  189. Wang X, Dai W, Wang Y, Gu Q, Yang D, Zhang M. Blocking the Wnt/beta-Catenin pathway by Lentivirus-Mediated short hairpin RNA targeting beta-Catenin gene suppresses Silica-Induced lung fibrosis in mice. Int J Environ Res Public Health. 2015;12:10739–54.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  190. Jin X, Dai H, Ding K, Xu X, Pang B, Wang C. Rapamycin attenuates bleomycin-induced pulmonary fibrosis in rats and the expression of metalloproteinase-9 and tissue inhibitors of metalloproteinase-1 in lung tissue. Chin Med J (Engl). 2014;127:1304–9.

    Article  CAS  PubMed  Google Scholar 

  191. Qing J, Ren Y, Zhang Y, Yan M, Zhang H, Wu D, Ma Y, Chen Y, Huang X, Wu Q, Mazhar M, Wang L, Liu J, Ding BS, Cao Z. Dopamine receptor D2 antagonism normalizes profibrotic macrophage-endothelial crosstalk in non-alcoholic steatohepatitis. J Hepatol. 2022;76:394–406.

    Article  CAS  PubMed  Google Scholar 

  192. Bringardner BD, Baran CP, Eubank TD, Marsh CB. The role of inflammation in the pathogenesis of idiopathic pulmonary fibrosis. Antioxid Redox Signal. 2008;10:287–301.

    Article  CAS  PubMed  Google Scholar 

  193. Ma H, Liu S, Li S, Xia Y. Targeting growth factor and cytokine pathways to treat idiopathic pulmonary fibrosis. Front Pharmacol. 2022;13:918771.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  194. Steele H, Willicut A, Dell G, Ghastine A, Nguyen XX, Lembicz P, Doerflein H, Suchoski T, Kato E, Feghali-Bostwick C, Croft M, Herro R. Combination therapy blocking TNF superfamily members 14 and 15 reverses pulmonary fibrosis. J Immunol, (2025).

  195. Jackson C, Stewart ID, Plekhanova T, Cunningham PS, Hazel AL, Al-Sheklly B, Aul R, Bolton CE, Chalder T, Chalmers JD, Chaudhuri N, Docherty AB, Donaldson G, Edwardson CL, Elneima O, Greening NJ, Hanley NA, Harris VC, Harrison EM, Ho LP, Houchen-Wolloff L, Howard LS, Jolley CJ, Jones MG, Leavy OC, Lewis KE, Lone NI, Marks M, McAuley HJC, McNarry MA, Patel BV, Piper-Hanley K, Poinasamy K, Raman B, Richardson M, Rivera-Ortega P, Rowland-Jones SL, Rowlands AV, Saunders RM, Scott JT, Sereno M, Shah AM, Shikotra A, Singapuri A, Stanel SC, Thorpe M, Wootton DG, Yates T, Gisli Jenkins R, Singh SJ, Man WD, Brightling CE, Wain LV, Porter JC, Thompson AAR, Horsley A, Molyneaux PL, Evans RA, Jones SE, Rutter MK, Blaikley JF. P.-C.S.C. Group, effects of sleep disturbance on dyspnoea and impaired lung function following hospital admission due to COVID-19 in the UK: a prospective multicentre cohort study. Lancet Respir Med. 2023;11:673–84.

    Article  PubMed  Google Scholar 

  196. Mora AL, Rojas M, Pardo A, Selman M. Emerging therapies for idiopathic pulmonary fibrosis, a progressive age-related disease. Nat Rev Drug Discov. 2017;16:810.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Saito S, Alkhatib A, Kolls JK, Kondoh Y, Lasky JA. Pharmacotherapy and adjunctive treatment for idiopathic pulmonary fibrosis (IPF). J Thorac Dis. 2019;11:S1740–54.

    Article  PubMed  PubMed Central  Google Scholar 

  198. Li R, Jia Y, Kong X, Nie Y, Deng Y, Liu Y. Novel drug delivery systems and disease models for pulmonary fibrosis. J Control Release. 2022;348:95–114.

    Article  CAS  PubMed  Google Scholar 

  199. Miller MR, Raftis JB, Langrish JP, McLean SG, Samutrtai P, Connell SP, Wilson S, Vesey AT, Fokkens PHB, Boere AJF, Krystek P, Campbell CJ, Hadoke PWF, Donaldson K, Cassee FR, Newby DE, Duffin R, Mills NL. Inhaled Nanoparticles Accumulate at Sites of Vascular Disease, ACS Nano, 11 (2017) 4542–4552.

  200. Miao D, Yu Y, Chen Y, Liu Y, Su G. Facile construction of i-Motif DNA-Conjugated gold nanostars as Near-Infrared and pH Dual-Responsive targeted drug delivery systems for combined Cancer therapy. Mol Pharm. 2020;17:1127–38.

    Article  CAS  PubMed  Google Scholar 

  201. Ghumman M, Dhamecha D, Gonsalves A, Fortier L, Sorkhdini P, Zhou Y, Menon JU. Emerging drug delivery strategies for idiopathic pulmonary fibrosis treatment. Eur J Pharm Biopharm. 2021;164:1–12.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  202. Gu J, Jiang L, Chen Z, Qi J. A simple nanoplatform of thermo-sensitive liposomes and gold nanorods to treat bone metastasis through improved chemotherapy combined with photothermal therapy. Int J Pharm X. 2024;8:100282.

    CAS  PubMed  PubMed Central  Google Scholar 

  203. Liyanage PY, Hettiarachchi SD, Zhou Y, Ouhtit A, Seven ES, Oztan CY, Celik E, Leblanc RM. Nanoparticle-mediated targeted drug delivery for breast cancer treatment. Biochim Biophys Acta Rev Cancer. 2019;1871:419–33.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  204. Pramanik S, Mohanto S, Manne R, Rajendran RR, Deepak A, Edapully SJ, Patil T, Katari O. Nanoparticle-Based drug delivery system: the magic bullet for the treatment of chronic pulmonary diseases. Mol Pharm. 2021;18:3671–718.

    Article  CAS  PubMed  Google Scholar 

  205. Eygeris Y, Gupta M, Kim J, Sahay G. Chemistry of lipid nanoparticles for RNA delivery. Acc Chem Res. 2022;55:2–12.

    Article  CAS  PubMed  Google Scholar 

  206. Song L, Hao Y, Wang C, Han Y, Zhu Y, Feng L, Miao L, Liu Z. Liposomal oxaliplatin prodrugs loaded with Metformin potentiate immunotherapy for colorectal cancer. J Control Release. 2022;350:922–32.

    Article  CAS  PubMed  Google Scholar 

  207. Chen G, Ullah A, Xu G, Xu Z, Wang F, Liu T, Su Y, Zhang T, Wang K. Topically applied liposome-in-hydrogels for systematically targeted tumor photothermal therapy. Drug Deliv. 2021;28:1923–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  208. Lu P, Zhang X, Li F, Xu KF, Li YH, Liu X, Yang J, Zhu B, Wu FG. Cationic liposomes with different lipid ratios: antibacterial activity, antibacterial mechanism, and cytotoxicity evaluations. Pharmaceuticals (Basel), 15 (2022).

  209. Cheng D, Wang Y, Li Z, Xiong H, Sun W, Xi S, Zhou S, Liu Y, Ni C. Liposomal UHRF1 SiRNA shows lung fibrosis treatment potential by regulating fibroblast activation. JCI Insight; 2022.

  210. Pan T, Zhou Q, Miao K, Zhang L, Wu G, Yu J, Xu Y, Xiong W, Li Y, Wang Y. Suppressing Sart1 to modulate macrophage polarization by siRNA-loaded liposomes: a promising therapeutic strategy for pulmonary fibrosis. Theranostics. 2021;11:1192–206.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  211. Wang Q, Liu J, Hu Y, Pan T, Xu Y, Yu J, Xiong W, Zhou Q, Wang Y. Local administration of liposomal-based Srpx2 gene therapy reverses pulmonary fibrosis by blockading fibroblast-to-myofibroblast transition. Theranostics. 2021;11:7110–25.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Yang MY, Lin YJ, Han MM, Bi YY, He XY, Xing L, Jeong JH, Zhou TJ, Jiang HL. Pathological collagen targeting and penetrating liposomes for idiopathic pulmonary fibrosis therapy. J Control Release. 2022;351:623–37.

    Article  CAS  PubMed  Google Scholar 

  213. Bamrungsap S, Zhao Z, Chen T, Wang L, Li C, Fu T, Tan W. Nanotechnology in therapeutics: a focus on nanoparticles as a drug delivery system. Nanomed (Lond). 2012;7:1253–71.

    Article  CAS  Google Scholar 

  214. Prabhu RH, Patravale VB, Joshi MD. Polymeric nanoparticles for targeted treatment in oncology: current insights. Int J Nanomed. 2015;10:1001–18.

    CAS  Google Scholar 

  215. Lee WH, Loo CY, Traini D, Young PM. Nano- and micro-based inhaled drug delivery systems for targeting alveolar macrophages. Expert Opin Drug Deliv. 2015;12:1009–26.

    Article  CAS  PubMed  Google Scholar 

  216. Griffiths G, Nystrom B, Sable SB, Khuller GK. Nanobead-based interventions for the treatment and prevention of tuberculosis. Nat Rev Microbiol. 2010;8:827–34.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  217. Lee C, Seo J, Hwang HS, Thao LQ, Lee S, Lee ES, Lee EH, Choi HG, Youn YS. Treatment of bleomycin-induced pulmonary fibrosis by inhaled tacrolimus-loaded chitosan-coated poly(lactic-co-glycolic acid) nanoparticles. Biomed Pharmacother. 2016;78:226–33.

    Article  CAS  PubMed  Google Scholar 

  218. Pintado-Berninches L, Montes-Worboys A, Manguan-Garcia C, Arias-Salgado EG, Serrano A, Fernandez-Varas B, Guerrero-Lopez R, Iarriccio L, Planas L, Guenechea G, Egusquiaguirre SP, Hernandez RM, Igartua M, Luis Pedraz J, Cortijo J, Sastre L, Molina-Molina M, Perona R. GSE4-loaded nanoparticles a potential therapy for lung fibrosis that enhances pneumocyte growth, reduces apoptosis and DNA damage. FASEB J. 2021;35:e21422.

    Article  CAS  PubMed  Google Scholar 

  219. Huang Y, Li P, Zhao R, Zhao L, Liu J, Peng S, Fu X, Wang X, Luo R, Wang R, Zhang Z. Silica nanoparticles: Biomedical applications and toxicity, Biomed Pharmacother, 151 (2022) 113053.

  220. Florek J, Caillard R, Kleitz F. Evaluation of mesoporous silica nanoparticles for oral drug delivery - current status and perspective of MSNs drug carriers. Nanoscale. 2017;9:15252–77.

    Article  CAS  PubMed  Google Scholar 

  221. Huang X, Li L, Liu T, Hao N, Liu H, Chen D, Tang F. The shape effect of mesoporous silica nanoparticles on biodistribution, clearance, and biocompatibility in vivo. ACS Nano. 2011;5:5390–9.

    Article  CAS  PubMed  Google Scholar 

  222. Cho M, Cho WS, Choi M, Kim SJ, Han BS, Kim SH, Kim HO, Sheen YY, Jeong J. The impact of size on tissue distribution and elimination by single intravenous injection of silica nanoparticles. Toxicol Lett. 2009;189:177–83.

    Article  CAS  PubMed  Google Scholar 

  223. Li X, Li Y, Yu C, Bao H, Cheng S, Huang J, Zhang Z. ROS-Responsive Janus Au/Mesoporous silica core/shell nanoparticles for drug delivery and Long-Term CT imaging tracking of MSCs in pulmonary fibrosis treatment. ACS Nano. 2023;17:6387–99.

    Article  CAS  PubMed  Google Scholar 

  224. Li X, He G, Jin H, Xiang X, Li D, Peng R, Tao J, Li X, Wang K, Luo Y, Liu X. Ultrasound-Activated precise Sono-Immunotherapy for breast Cancer with reduced pulmonary fibrosis. Adv Sci (Weinh). 2025;12:e2407609.

    Article  PubMed  Google Scholar 

  225. Yu H, Yang Z, Li F, Xu L, Sun Y. Cell-mediated targeting drugs delivery systems. Drug Deliv. 2020;27:1425–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  226. Sun Y, Su J, Liu G, Chen J, Zhang X, Zhang R, Jiang M, Qiu M. Advances of blood cell-based drug delivery systems. Eur J Pharm Sci. 2017;96:115–28.

    Article  CAS  PubMed  Google Scholar 

  227. Wynn TA, Chawla A, Pollard JW. Macrophage biology in development, homeostasis and disease. Nature. 2013;496:445–55.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  228. Mitchell MJ, King MR. Leukocytes as carriers for targeted cancer drug delivery. Expert Opin Drug Deliv. 2015;12:375–92.

    Article  CAS  PubMed  Google Scholar 

  229. Zhang X, Li W, Sun J, Yang Z, Guan Q, Wang R, Li X, Li Y, Feng Y, Wang Y. How to use macrophages to realise the treatment of tumour. J Drug Target. 2020;28:1034–45.

    Article  PubMed  Google Scholar 

  230. Murray PJ, Wynn TA. Protective and pathogenic functions of macrophage subsets. Nat Rev Immunol. 2011;11:723–37.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  231. Sang X, Wang Y, Xue Z, Qi D, Fan G, Tian F, Zhu Y, Yang J. Macrophage-Targeted lung delivery of dexamethasone improves pulmonary fibrosis therapy via regulating the immune microenvironment. Front Immunol. 2021;12:613907.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  232. Chu D, Dong X, Shi X, Zhang C, Wang Z. Neutrophil-Based Drug Delivery Syst Adv Mater. 2018;30:e1706245.

    Google Scholar 

  233. Sun D, Du X, Cao X, Wu B, Li S, Zhao Y, Liu T, Xu L, Huang H. Neutrophil-Based bionic delivery system breaks through the capillary barrier of liver sinusoidal endothelial cells and inhibits the activation of hepatic stellate cells. Mol Pharm. 2024;21:2043–57.

    Article  CAS  PubMed  Google Scholar 

  234. Chellappan DK, Yee LW, Xuan KY, Kunalan K, Rou LC, Jean LS, Ying LY, Wie LX, Chellian J, Mehta M, Satija S, Singh SK, Gulati M, Dureja H, Da Silva MW, Tambuwala MM, Gupta G, Paudel KR, Wadhwa R, Hansbro PM, Dua K. Targeting neutrophils using novel drug delivery systems in chronic respiratory diseases. Drug Dev Res. 2020;81:419–36.

    Article  CAS  PubMed  Google Scholar 

  235. Chu D, Gao J, Wang Z. Delivery of therapeutic nanoparticles across blood vessel barrier for treatment of inflammation and infection. ACS Nano. 2015;9:11800–11.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  236. Ramalho-Santos M, Willenbring H. On the origin of the term stem cell. Cell Stem Cell. 2007;1:35–8.

    Article  CAS  PubMed  Google Scholar 

  237. Chu GY, Chen YF, Chen HY, Chan MH, Gau CS, Weng SM. Stem cell therapy on skin: mechanisms, recent advances and drug reviewing issues. J Food Drug Anal. 2018;26:14–20.

    Article  CAS  PubMed  Google Scholar 

  238. Kusuma GD, Carthew J, Lim R, Frith JE. Effect of the microenvironment on mesenchymal stem cell paracrine signaling: opportunities to engineer the therapeutic effect. Stem Cells Dev. 2017;26:617–31.

    Article  CAS  PubMed  Google Scholar 

  239. Liang X, Ding Y, Zhang Y, Tse HF, Lian Q. Paracrine mechanisms of mesenchymal stem cell-based therapy: current status and perspectives. Cell Transpl. 2014;23:1045–59.

    Article  Google Scholar 

  240. Baraniak PR, McDevitt TC. Stem cell paracrine actions and tissue regeneration. Regen Med. 2010;5:121–43.

    Article  PubMed  Google Scholar 

  241. Menon LG, Kelly K, Yang HW, Kim SK, Black PM, Carroll RS. Human bone marrow-derived mesenchymal stromal cells expressing S-TRAIL as a cellular delivery vehicle for human glioma therapy. Stem Cells. 2009;27:2320–30.

    Article  CAS  PubMed  Google Scholar 

  242. Studeny M, Marini FC, Dembinski JL, Zompetta C, Cabreira-Hansen M, Bekele BN, Champlin RE, Andreeff M. Mesenchymal stem cells: potential precursors for tumor stroma and targeted-delivery vehicles for anticancer agents. J Natl Cancer Inst. 2004;96:1593–603.

    Article  CAS  PubMed  Google Scholar 

  243. Sun L, Fan M, Huang D, Li B, Xu R, Gao F, Chen Y. Clodronate-loaded liposomal and fibroblast-derived Exosomal hybrid system for enhanced drug delivery to pulmonary fibrosis. Biomaterials. 2021;271:120761.

    Article  CAS  PubMed  Google Scholar 

  244. Medico F, Kim S, Surwase SS, Liu H, Kim YC. A core-shell microneedle system for stable fibroblast delivery in cell-based therapies. Drug Deliv Transl Res, (2024).

  245. Hou J, Ji Q, Ji J, Ju S, Xu C, Yong X, Xu X, Muddassir M, Chen X, Xie J, Han X. Co-delivery of siPTPN13 and siNOX4 via (myo)fibroblast-targeting polymeric micelles for idiopathic pulmonary fibrosis therapy, Theranostics, 11 (2021) 3244–3261.

  246. Bhattacharyya A, Khan R, Lee JY, Tassew G, Oskouian B, Allende ML, Proia RL, Yin X, Ortega JG, Bhattacharya M, Saba JD. Gene therapy with AAV9-SGPL1 in an animal model of lung fibrosis. J Pathol. 2024;263:22–31.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  247. Sun L, Zhang HB, Jiang HC, Li W, Li MK, Yang XY, Cai YY, Xue KK, Gou YS, Liu XY, Liang Q, Zuo LG, Hu JG, Qian F. LMO7 drives profibrotic fibroblast polarization and pulmonary fibrosis in mice through TGF-beta signalling. Acta Pharmacol Sin, (2025).

  248. Wang J, Lai X, Yao S, Chen H, Cai J, Luo Y, Wang Y, Qiu Y, Huang Y, Wei X, Wang B, Lu Q, Guan Y, Wang T, Li S, Xiang AP. Nestin promotes pulmonary fibrosis via facilitating recycling of TGF-beta receptor I. Eur Respir J, 59 (2022).

Download references

Acknowledgements

Not applicable.

Funding

This work was supported by the National Natural Science Foundation of China (82173554, 82203993, 82360641); The Natural Science Research of Jiangsu Higher Education Institutions (22KJB330003); Jiangxi Provincial Natural Science Foundation (20232BAB206138).

Author information

Authors and Affiliations

Authors

Contributions

Mengna Jiang, Wenxia Bu: writing– original draft (lead); Xuehai Wang: Conceptualization; Jialing Ruan, Weijian Shi: writing– review & editing; Shiqi Yu, Lizhen Huang: writing– review & editing (equal); Peng Xue, Juan Tang: supervision; Xinyuan Zhao, Liling Su, Demin Cheng: conceptualization (lead) and project administration (lead). All authors have read and agreed to the published version of the manuscript.

Corresponding authors

Correspondence to Xinyuan Zhao, Liling Su or Demin Cheng.

Ethics declarations

Competing interests

The authors declare that they have no known competing financial interests.

Additional information

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Rights and permissions

Open Access This article is licensed under a Creative Commons Attribution-NonCommercial-NoDerivatives 4.0 International License, which permits any non-commercial use, sharing, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if you modified the licensed material. You do not have permission under this licence to share adapted material derived from this article or parts of it. The images or other third party material in this article are included in the article’s Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by-nc-nd/4.0/.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Jiang, M., Bu, W., Wang, X. et al. Pulmonary fibrosis: from mechanisms to therapies. J Transl Med 23, 515 (2025). https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-025-06514-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doiorg.publicaciones.saludcastillayleon.es/10.1186/s12967-025-06514-2

Keywords