Next Article in Journal
Removal of Copper(II) from Aqueous Environment Using Silk Sericin-Derived Carbon
Next Article in Special Issue
Tracking of Extracellular Vesicles’ Biodistribution: New Methods and Approaches
Previous Article in Journal
Tryptamine, a Microbial Metabolite in Fermented Rice Bran Suppressed Lipopolysaccharide-Induced Inflammation in a Murine Macrophage Model
Previous Article in Special Issue
Methods of the Large-Scale Production of Extracellular Vesicles
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Role of Mesenchymal Stem Cells and Extracellular Vesicles in Idiopathic Pulmonary Fibrosis

by
Sevindzh Kletukhina
1,
Guzel Mutallapova
1,
Angelina Titova
2 and
Marina Gomzikova
1,*
1
Laboratory of Intercellular Communication, Kazan Federal University, 420008 Kazan, Russia
2
Morphology and General Pathology Department, Kazan Federal University, 420008 Kazan, Russia
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(19), 11212; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms231911212
Submission received: 31 July 2022 / Revised: 20 September 2022 / Accepted: 20 September 2022 / Published: 23 September 2022
(This article belongs to the Special Issue Recent Advances in Immuno-Oncology and Intercellular Communication)

Abstract

:
Idiopathic pulmonary fibrosis (IPF) is a progressive interstitial fibrotic disease that leads to disability and death within 5 years of diagnosis. Pulmonary fibrosis is a disease with a multifactorial etiology. The concept of aberrant regeneration of the pulmonary epithelium reveals the pathogenesis of IPF, according to which repeated damage and death of alveolar epithelial cells is the main mechanism leading to the development of progressive IPF. Cell death provokes the migration, proliferation and activation of fibroblasts, which overproduce extracellular matrix, resulting in fibrotic deformity of the lung tissue. Mesenchymal stem cells (MSCs) and extracellular vesicles (EVs) are promising therapies for pulmonary fibrosis. MSCs, and EVs derived from MSCs, modulate the activity of immune cells, inhibit the expression of profibrotic genes, reduce collagen deposition and promote the repair of damaged lung tissue. This review considers the molecular mechanisms of the development of IPF and the multifaceted role of MSCs in the therapy of IPF. Currently, EVs-MSCs are regarded as a promising cell-free therapy tool, so in this review we discuss the results available to date of the use of EVs-MSCs for lung tissue repair.

1. Introduction

Idiopathic pulmonary fibrosis (IPF) is a specific form of idiopathic interstitial pneumonia. It has been shown to have a high mortality rate and an average of 2 to 3 years lifespan after diagnosis [1]. The disease has been shown to occur in middle or older age people. Primary symptoms of IPF include dyspnoea, hypoxaemia, radiographically marked pulmonary infiltrates and accumulation of fibroblasts in the tissue. These symptoms are as a result of the initial scarring of the lung tissues caused by IPF [2].
IPF has been shown to be caused by damages to alveolar epithelial cells due to genetic, environmental and immunological factors, which leads to metabolic dysfunction, aberrant activation of damaged epithelial cells and pathological epithelial repair. Molecular changes in the alveolar epithelial cells during EMT-induced damage causes the cells to actively express profibrotic factors leading to migration, proliferation, differentiation of fibroblasts and myofibroblasts and subsequent accumulation of extracellular matrix, thereby leading to pulmonary fibrosis [3,4,5]. However, the etiology of this progressive parenchymal disease is still unknown.
There are several factors that contribute to the development of IPF. These include and may not be limited to autoimmune/connective tissue diseases (e.g., rheumatoid arthritis or scleroderma) [6,7,8]; antiarrhythmic drugs administration (amiodarone) [9,10]; chemotherapeutic agents (bleomycin) [11]; antimicrobial agents (nitrofurantoin) [12]; smoking [13]; infectious agents such as mycoplasma [14], legionella [15], rickettsia [16]; viruses (e.g., SARS-CoV-2) [17]; radiation [18,19]; occupational exposures (mining; production of ceramic, acid- and fire-resistant materials; work with sandblasters) [20,21,22]; toxic fumes and gases [23]; genetic predisposition [24,25,26]; diabetes [27]; and gastroesophageal reflux disease [28,29]. The therapeutic modality is determined by the etiology of the fibrosis, e.g., discontinuing the drug that caused the fibrosis, preventing allergic or occupational diseases, or treating the underlying autoimmune disease with immunosuppressive drugs [30].
Current therapies for IPF include lung transplantation, pharmacotherapy and cell therapy. Transplantation has the potential of increasing the quality and life span of patients with lung diseases. However, shortage of donor material, risk of acute graft versus host disease and infection, limit the application of lung transplantation as a therapy measure [31,32]. Pharmacotherapy measures for IPF are predominantly limited with the use of antifibrotic drugs such as pirfenidone and nintedanib. These drugs slow disease progression but do not repair damaged lung tissue and have a number of side effects [33,34,35,36]. Cell therapy for pulmonary fibrosis (PF) involves the use of mesenchymal stem cells (MSCs). MSCs are polypotent cells, capable of differentiating into various cell types and providing immunomodulatory, antiproliferative and anti-inflammatory effects [37,38,39,40]. MSCs secrete growth factors, anti-inflammatory cytokines, chemokines and basic proteins that reduce the deposition of extracellular matrix (ECM) and collagen, as well as promote alveolar repair [41]. However, with MSCs therapy, there is a risk of their differentiation into fibroblasts which produce collagen. Undesirable differentiation and accumulation of mutations during cultivation limit the use of MSCs in clinical practice [42,43,44].
Extracellular vesicles naturally released from MSCs (nEVs-MSCs) are an alternative to cell therapy. It has been shown that the positive effects observed from the use of nEVs- MSCs in IPF therapy has to do with their ability to suppress the activity of growth factors, chemokines and cytokines that stimulate fibrosis development, similarly observed to parental cells [45,46]. It is worth mentioning that injection of nEVs-MSCs, in contrast to MSCs transplantation, does not cause arrhythmia, tumour formation and calcification in the tissues [47,48]. To increase the yield of EVs, protocols are being developed to obtain induced vesicles. These induced vesicles exhibit immunosuppressive properties, similarly observed in natural EVs and parental cells. These immunosuppressive properties may have a positive effect on the therapy of IPF [49,50]. Thus, the use of mesenchymal stem cells extracellular vesicles has reason to be of high interest in the treatment of IPF.

2. Fibroblast Heterogeneity in Pulmonary Fibrosis

IPF is the chronic damage of the alveolar epithelium resulting in its inflammation, synthesis and deposition of collagen in the lung interstitium. Chronic inflammation, which is often associated with the formation of a patchy interstitial infiltrate of lymphocytes and plasma cells, has long been thought to be the key mechanism underlying IPF [51,52]. Pulmonary fibrosis was previously thought to be a purely inflammatory process, but, according to current understanding of the pathogenesis of the disease, IPF is primarily characterized by a process of pulmonary fibrosis. Epithelial cell damage and apoptosis represent the initial step in the development of pulmonary fibrosis [53,54]. Various agents, such as allergens, radiation, chemicals, viruses, cigarette smoke, AOFs (active oxygen forms) cause damage to alveolar epithelial cells, which contributes to an inflammatory response [55,56,57,58]. Damaged lung epithelial cells release proinflammatory mediators, namely chemokines, which induce recruitment of circulating blood monocytes, fibrocytes and bone marrow progenitor fibroblast cells [59] to sites of damage [60,61]. Inflammatory cells actively secrete profibrotic activated cytokines and fibrocytes, which subsequently, infiltrate tissue, damage sites of infiltration and promote pulmonary fibrosis by differentiating into active collagen-producing fibroblasts and myofibroblasts [62,63].
Fibrogenesis results from the repeated microdamage of alveolar epithelial cells, followed by the production of factors responsible for activation of a fibrotic program leading to an abbreviated repair of the epithelial cells. This process known as tissue damage-induced epithelial–mesenchymal transition (EMT) on the one hand promotes wound healing and repair of damaged tissue, but on the other hand promotes fibrosis [64,65]. In pathological processes such as fibrosis and in wound healing, the change of epithelial cells to the mesenchymal phenotype is believed to be a necessary process. Epithelial and endothelial cells divide, migrate and participate in the regeneration of damaged tissue [66]. The molecular changes that occur to alveolar epithelial cells during EMT allow them to acquire the mesenchymal cell phenotype. They begin to actively express growth factors (TGF-β, TNF-α, HGF, interleukin-4), secrete numerous mediators (matrix metalloproteinases, angiogenesis inhibitors, and procoagulation factors) leading to their further migration, proliferation, differentiation of fibroblasts into myofibroblasts, and accumulation of extracellular matrix components [67,68,69]. The deposition of excessive amounts of extracellular matrix components leads to an abnormal increase in tissue stiffness. Increased stiffness of lung tissue causes PGE2 (an autocrine inhibitor of fibrogenesis) synthesis suppression, induces further activation of fibroblasts through mechanotransduction pathways and their differentiation into myofibroblasts, thereby contributing to IPF progression [70,71,72,73]. The formed myofibroblasts release numerous profibrotic mediators and synthesize components of the extracellular matrix such as collagen and fibronectin [74,75]. Myofibroblasts, due to their contractile properties, are able to manipulate fibers of the extracellular matrix to heal open wounds, and influence inflammation. All these processes induced by myofibroblasts, in turn, contribute to fibrosis and PF progression.
Myofibroblasts can arise from different cell types such as epithelial cells, mesenchymal stem cells, pericytes, circulating bone marrow fibrocytes, preadipocytes and adipocytes. These are often collectively referred to as fibroblasts (Figure 1). Fibroblasts is the general term often used to describe these heterogeneous cell populations. Stimulation of TGF-β1 is a known factor to activate fibroblasts and transform them into myofibroblasts, which express α-smooth muscle actin (α-SMA) and secrete cytoskeletal and contractile extracellular matrix proteins (Figure 1). TGF-β binds to type I and type II receptor complexes localized on the cell membrane and stimulates receptor activation leading to phosphorylation of SMAD2/3 in the cytosol. Activated SMAD2/3 additionally bind to SMAD4 and travel to the nucleus where they control the expression of target extracellular matrix proteins by binding to the SMAD binding element in the promoter. The expression of these proteins, as previously mentioned, is a characteristic feature of myofibroblasts [76,77]. Myofibroblasts contribute to lung tissue remodeling through collagen production, profibrotic factors and proteins. In this regard, 25-hydroxycholesterol has been shown to be involved in tissue remodeling in fibrosis. 25-hydroxycholesterol induces NF-κβ p65 transport into the nucleus. Activation of NF-κβ signaling promotes enhanced TGF-β1 release, α-SMA expression, collagen I production and matrix metalloproteinase release [78].
Fibroblast differentiation into the myofibroblast phenotype can also be induced by hypoxia and expression of insulin-like growth factor type II (IGF-II). Hypoxia leads to hypermethylation of the Thy-1 promoter in normal human lung fibroblasts and increases the levels of the profibrotic markers: α-SMA, collagen-1, collagen-3, characteristic of the myofibroblastic phenotype [103]. IGF-II expression is activated during IGF. IGF-II transmits signals through IGF1R/IR, PI3K/AKT/GSK3β and JNK/c-Jun receptors, resulting in the increased activity of ACTA2, TIMP1/4, collagen, fibronectin, TGF-β2/3 and SMAD2/3. Due to the expression of these genes, IGF-II induces myofibroblast formation [104].
According to recent studies, lipofibroblasts play an important role in myofibroblast formation. Lipofibroblasts are resident lung fibroblasts that are located in the thickness of the interalveolar septa near type II alveolocytes and can differentiate into myofibroblasts. This makes them able to contribute to pulmonary fibrosis.
Exposure to nicotine, hyperoxia and Wnt/β-catenin signaling induce differentiation of lipofibroblasts into activated myofibroblasts during IPF formation (Figure 1).
TGF-β expression also promotes myofibroblast formation from lipofibroblasts by upregulating SMAD2, SMAD3, Sp1 and c-Myc (Figure 1). Conversely, thiazolidinediones, metformin—an anti-diabetic drug, and PPARγ signaling are able to convert myofibroblasts back to lipofibroblasts. The use of PPARγ and its agonists to preserve the lipogenic phenotype and prevent differentiation into the myogenic phenotype in IPF can offer an alternative to existing ineffective drugs (pirfenidone and nintedanib) [76,105]. It has also been found that increased FGF-10 expression in lung tissue during IPF promotes fibrosis resolution through its ability to suppress the activation of profibrotic cytokines, especially TGF-β1, and presumably contributes to the differentiation of activated myofibroblasts into lipofibroblasts [76].
After wound healing, myofibroblasts can follow different pathways: they may die due to apoptosis induced by soluble proapoptotic factors (IL-1β, FGF1 and prostaglandin E2 (PGE2)) [106,107,108]; they may be deactivated and turn into fibroblasts; and they may be aged by the CCN family proteins [109]. This does not occur in IPF, as myofibroblasts in fibrous tissues show high resistance to apoptosis, thus ensuring the preservation of highly active cells at the sites of damage. Resistance to apoptosis is most likely due to the persistent activity of TGF-β1 and myofibroblast-induced deposition of extracellular matrix components, leading to prolonged survival and activity of myofibroblasts during fibrosis development [110,111,112].

3. Multifaceted Role of MSCs in Pulmonary Fibrosis

MSCs located in the stromal and perivascular compartments in close proximity to fibroblasts are believed to modulate myofibroblast activity.
There is evidence of the effectiveness of MSCs in the therapy of bleomycin-induced fibrotic lung lesions in animals. Stem cells modulate the activity of B-cells by inhibiting their maturation and recruitment to the sites of lung damage during IPF. Modulation of B-cell activity leads to inhibition of the chronic inflammatory process with subsequent formation of fibrotic lesions [113]. On a model of bleomycin-induced lung damage it was shown that MSCs reduce costimulatory protein expression in dendritic cells and monocyte-derived macrophages, reduce their ability to induce antigen-specific T-cell immune responses, and promote generation of immune cells (Treg) and cytokines (IL-10) with immune regulatory functions [114].
Various deleterious factors induce damage and apoptosis of alveolar epithelial cells, which is commonly accompanied by mitochondrial dysfunction. One of the consequences of mitochondrial dysfunction is an increase in AFC in alveolar epithelial cells (AECs), which contribute to further damage, while there is a persistence of apoptosis-resistant myofibroblasts and excessive deposition of extracellular matrix components [115].
The efficacy of PF treatment with MSCs has been shown to be linked to their ability to deliver mitochondria to the damaged epithelial cells [116], reduce collagen deposition [117], promote tissue repair through the secretion of anti-inflammatory and the anti-fibrotic factors they possess [118]. Mitochondrial delivery reverses the effects of mitochondrial dysfunction and maintains normal mitochondrial homeostasis by delivering healthy mitochondria into damaged epithelial cells [116,119]. Gene expression analysis has shown that MSCs powerfully suppress profibrotic genes induced by bleomycin and inhibit pro-inflammatory transcripts [120]. Human MSCs reduce the levels of TGF-β1 and TNF-α by secreting PGE2 and hepatocyte growth factor (HGF). And TGF-β is known to be a key factor influencing the process of extracellular matrix deposition and myofibroblast differentiation. Other favorable factors secreted by MSCs include PGE2 which has the ability to inhibit TGF-β1-induced proliferation of fibroblasts and collagen production by inducing myofibroblast apoptosis through the increased activity of PTEN (phosphatase and tensin homolog); HGF which reduces the deposition of extracellular matrix in alveolar type II (ATII) cells and induces myofibroblast apoptosis through MMP activation; MMP-9 which promotes myofibroblast death by degrading the FN-CCB domain, and MMP-1 which function as a collagenase to prevent lung tissue thickening [118,121]. FGF-2, secreted by adipose tissue stromal cells, can block further cell differentiation. This has been evidenced by observed decreased gene and protein expression of mesenchymal markers and gene expression of extracellular matrix components. FGF-2 treatment increased the matrix metalloproteinase gene expression and decreased the expression of the metalloproteinase gene TIMP-2 inhibitors in tissues [122,123]. The expression of FGF-10 by lung mesenchymal cells is believed to be involved in the suppression of pulmonary fibrosis. It was shown that FGF-10 expression in lung MSCs inversely correlates with fibrosis progression and TGF-β1 expression in activated myofibroblasts. FGF-10 expression was significantly reduced in stromal cells isolated from bronchoalveolar lavage of patients with IPF, in contrast to MSCs of healthy lungs. This confirms that FGF-10 deficiency can be the cause of disease progression and induction of this factor expression can serve as an effective therapy for IPF [124,125,126].
However, there is alternative evidence for the involvement of MSCs in IPF. In addition to having regenerative activity, MSCs have been shown to contribute to the development and progression of pulmonary fibrosis. Overexpression of α-SMA, COL1α1, fibronectin, TGF-β1, IL-6 and TNF-α in MSCs microenvironment induces MSCs differentiation into myofibroblasts, which in turn promotes collagen deposition and accumulation of extracellular matrix components [24,25,26,27]. Thus, in radiation-induced lung fibrosis, increased expression of cytokines TNF-α, TGF-β1 and adhesion molecules ICAM-1 and VCAM-1 in lung epithelial cells and in damaged lung areas induces bone marrow MSCs migration through the circulatory system to the sites of lung damage and their differentiation into myofibroblasts [127]. A similar effect of microenvironment on the regenerative properties of MSCs was investigated on tissue-resistant lung MSCs, human umbilical cord MSCs, and VEGF-positive MSCs actively used for therapeutic purposes [128,129,130,131]. Moreover, it was shown that bleomycin-induced pulmonary fibrosis induces Wnt10a expression and activation of Shh/Gli signaling cascade resulting in myofibroblast differentiation of lung MSCs. The activation of Shh/Glioblastoma system through Wnt/β-catenin signaling control regulates MSCs transition into the myofibroblast phenotype and enhances pulmonary fibrosis [132].
Excessive activation of PDGFRα signaling leads to fibrosis and vascular calcification. Santini et al. showed that in stem cells actively expressing PDGFRα, there is an upregulation of genes involved in fibrosis, namely, growth factor TGF-β1 and fibroblast activation protein (FAP). Subsequently, MSCs differentiate into myofibroblast-like profibrotic cells, thus contributing to the development of fibrosis [84].
In the manifestation of the profibrotic effects of the MSCs, the stage of fibrosis in which the infusion of stem cells was carried out also plays a role. Yan et al. indicate that infusion of MSCs at a later stage of lung damage enhances IPF. MSCs that were infused at an early stage of fibrosis (after 4 h of fibrosis induction time) were safely engrafted into the lung tissues, whereas MSCs injected at a later stage (at days 60 and 120) were detected as myofibroblasts in the lung interstitial space. This is most likely due to the overexpression of profibrotic cytokines, including TGF-β1, which affect MSC differentiation. These data indicate that MSCs infusion in the late stages of the disease enhances scarring in the damaged tissue rather than having a regenerative effect [131].
Despite the controversial properties, MSCs are currently promising candidates for therapeutic use. For the safe application of MSCs as a therapeutic agent, factors such as optimal dosing, age of the cell donor and the phase of fibrosis at which the stem cells are injected should be considered. Stem cells have maximum beneficial effects when stem cells are administered in the early inflammatory phase of pulmonary fibrosis, whereas a negative effect is observed when they are administered in the late phase of fibrosis. The beneficial effect in the early phase is explained by the fact that MSCs express TGF-β1, which is involved in modulation of immune reactions [39]. At the same time, MSCs serve as a target for TGF-β1, contributing to disease progression at later stages. MSCs of old mice, unlike the stem cells of young mice, divide more slowly in vitro and do not exhibit protective properties [133], hence it is recommended to use MSCs of younger donors in therapy.

4. Immunologic Regulation of Pulmonary Fibrosis

Studies on the mechanisms of pulmonary fibrosis have demonstrated the importance of cytokines in pulmonary fibrogenesis in animals and humans. Epithelial and mesenchymal cells, T- and B-lymphocytes, macrophages, neutrophils, eosinophils and platelets are potential sources of cytokines in the lung.
Neutrophils are known to play a key role in the progression of pulmonary fibrosis. They are recruited in the initial phases to exert pro-inflammatory effects. An increased percentage of neutrophils in bronchoalveolar lavage (BAL) fluid has been shown to predict early mortality in individuals with IPF [134]. Infiltration and migration of neutrophils into damaged lung tissue is induced in response to overexpression of CXCL2 on resident alveolar macrophages [135], as well as increased expression of CXCL1 and CXCL2/3, IL-8/CXCL8 [136,137]. In addition, neutrophils produce neutrophil elastase (NE), a neutrophil-specific serine protease, which in turn promotes fibroblast proliferation, myofibroblast differentiation and TGFβ1 activation [138].
Hyperexpression of B-cell antibody genes and focal aggregations of these lymphocytes have been observed in the lungs in IPF [139,140,141,142]. In patients with IPF, B-cell-produced antigen–antibody complexes are present in the lung parenchyma, bloodstream and BAL, which trigger cytotoxic and proinflammatory cascades [143,144,145]. The delivery of B-cells to the foci of inflammation is ensured by the expression of a specific mediator-CXCL13, the sources of which are CD68+ macrophage line cells [146].
Macrophages play a recognized role in wound healing and fibrogenesis through the production and release of chemokines that can engage inflammatory cells and lead to proliferation and activation of myofibroblasts [147,148,149]. In a mouse model of bleomycin-induced pulmonary fibrosis, Baran et al. demonstrated that levels of macrophage colony-stimulating factor, which they showed controls mononuclear phagocyte recruitment and CCL2 production, are elevated in BAL in patients with IPF [150]. Repetitive lung injury in a bleomycin-induced pulmonary fibrosis model showed activation of pulmonary capillary endothelial cells (PCEC) and perivascular macrophages, which in turn inhibits alveolar repair and promotes fibrosis. Suppression of the chemokine receptor CXCR7 expressed on PCEC leads to recruitment of perivascular macrophages expressing VEGFR1 and subsequent stimulation of Wnt/β-catenin-dependent activation of Notch Jagged1 ligand with profibrotic consequences [151]. It has been shown that in addition to initiating the immune response, alveolar macrophages do generate reactive oxygen species (ROS), especially mitochondrial H2O2, which contributes to the development of fibrosis by increasing the expression of TGF-β1 [152,153]. The data obtained by Larson-Casey et al. demonstrate that alveolar macrophages are a major source of TGF-β1. Similarly, their studies suggest that Akt1-mediated mitophagy promotes alveolar macrophage resistance to apoptosis and is essential for the development of pulmonary fibrosis [154].
A study by Scott et al. showed that increased monocyte counts are associated with an increased risk of adverse outcomes in patients with IPF [155]. Classic monocytes have been shown to infiltrate into the fibrotic lung in the early stages of fibrosis and subsequently mature to profibrotic inflammatory alveolar macrophages [156].
Enhanced monocyte migration into the lungs of patients with IPF occurs in response to increased production of CCL2 (or monocyte chemoattractant protein-1, MCP1) by endothelial cells and increased serum CCL2 concentrations [157,158].
The accumulation of mature dendritic cells (DCs) in large numbers with local maturation potential in ectopic lymphoid follicles has been shown to be induced by resident cells that express chemokines such as CCL19, CXCL12 and CCL2, which ultimately contributes to chronic inflammation in IPF [141,159]. TGF-β is able to modulate the accumulation and phenotypic maturation of CD11c+ pulmonary DCs in a mouse model of pulmonary fibrosis. Thus, the absence of TGF-β leads to a decrease in the percentage of CD11c+CD11b+ cells in the group treated with bleomycin [160].
According to the literature, an increased number of mast cells (MCs) are found in the lungs of patients with IPF [161,162]. In addition, increased levels of tryptase (a trypsin-like enzyme found in mast cell granules and basophils) are found in IPF lung tissue samples. In vitro experiments showed that co-culture of human lung MCs with human lung fibroblasts induced MCs activation and stimulated human lung fibroblasts proliferation, which in turn showed a significantly higher growth rate compared to controls [163]. In addition, it has been shown that lung fibrotic ECM regulates MCs function and that degranulating MCs activate TGF-β1 [164]. Overed-Sayer et al. showed a positive correlation between the number of MCs and foci of fibroblasts in patients with IPF, confirming the link between MCs density and mortality from IPF [165].
The T-cell response is known to contribute to the pathogenesis of idiopathic pulmonary fibrosis [166]. In addition, decreased expression of CD28 on circulating T cells is associated with adverse outcomes in patients with IPF [167]. The regulation of IPF by T cells is carried out through the interaction of Fas–Fas ligand (FasL), cAMP chloride channels or T-cells depletion [168,169,170]. T cells such as Th1/Th2, Th17, Th9 and regulatory T cells are thought to contribute most to the progression of IPF. For example, Th1 secrete antifibrotic factors IFN-γ and IL-12, which inhibit fibroblast proliferation and fibrous tissue formation, while the IL-4 and IL-13 released by Th2 stimulate fibroblast proliferation, collagen production and fibroblast differentiation into myofibroblasts, as well as incline macrophages towards the pro-fibrotic phenotype [171,172,173]. Thus, a Th1/Th2 imbalance has been shown to play an important role in the pathogenesis of pulmonary fibrosis. Th17 cells and IL-17 are found in and around inflammatory infiltrates in patients with IPF, confirming their role in fibrosis development and inflammatory progression [142]. In vitro experiments have shown that IL-17 promotes proliferation of pulmonary fibroblasts, which in turn leads to increased synthesis of type I collagen, TGF-β and IL-6 expression [174]. A key cytokine secreted by Th9 cells is IL-9, which has both antifibrotic and profibrotic effects [175]. However, recent studies have shown that Th9 cells do contribute to pulmonary fibrosis by increasing IL-4-mediated Th2 cell differentiation, and IL-9 neutralisation effectively reduces the degree of pulmonary fibrosis [176]. Regulatory T cells can also have both anti- [177] and profibrotic effects [178,179]. Boveda-Ruiz et al. showed that depletion of CD4+CD25+ regulatory T-cells subpopulation at early stages of BLM-induced lung damage is associated with fibrosis reduction, but this depletion later leads to increased lung fibrosis, meaning that Tregs can be harmful at early stages, but protective at later stages of pulmonary fibrosis in mice [180].
Thus, inflammation may act as an important component in the etiology of IPF, and disease progression is accompanied by innate and adaptive immune responses. Therefore, the immunosuppressive properties of MSCs help to suppress inflammation and promote epithelial tissue repair, which makes them a promising tool for the therapy of IPF.

5. Effect of MSCs-Derived EVs on Pulmonary Fibrosis

The therapeutic efficacy of EVs-MSCs has been investigated in preclinical animal models and in many diseases and injuries. Currently, there are more than 300 published reports on the therapeutic properties of exosomes/microvesicles/extracellular vesicles derived from MSCs listed in the PubMed database, covering many categories of diseases.
For example, human EVs-MSCs have been shown to be therapeutically effective in an E. coli endotoxin-induced acute lung injury model in mice [181,182]. Intravenous administration of EVs derived from MSCs prevents and treats bleomycin-induced pulmonary fibrosis, with subsequent improvement in both lung morphology and architecture and reduction in collagen deposition. In addition, PF modulates lung macrophage phenotypes by shifting the proportions of pro-inflammatory/classical and non-classical lung monocytes and alveolar macrophages towards the monocyte/macrophage profiles of control mice, thereby reducing inflammation [183]. Xu et al. showed that human umbilical cord EVs-MSCs can inhibit silica gel-induced pulmonary fibrosis, as well as regulate pulmonary function [184]. Lei et al. found that by transferring miR-214-3p in EVs derived from human placental MSCs attenuation of radiation-induced endothelial damage, vascular dysfunction and inflammation as well as pulmonary fibrosis through suppression of the ATM/P53/P21 pathway occurs. They also found that the expression of collagen type 1α-1 (COL1α1), TGF-β, α-SMA, fibronectin and MMP9 genes involved in fibrosis development was significantly reduced after treatment with EVs-MSCs (Figure 2). Moreover, EVs-MSCs increased the expression of several antifibrotic genes including the TIMP-1, TIMP-2 and bone morphogenetic protein-7 (BMP-7) [185]. miR-186, delivered by EVs from bone marrow MSCs (BM-MSCs), reduces fibroblast activation by downregulating the transcription factors SOX4 and DKK1, while slowing the progression of IPF in mice (Figure 2) [186]. Li et al. demonstrated that EVs-MSCs treatment can reduce pulmonary edema, pulmonary dysfunction and inflammation, which were caused by ischemic/reperfusion lung injury in mice [187]. The results of another group of researchers showed that treatment with EVs-MSCs prevents activation of hypoxia signaling, which underlies lung inflammation and the development of pulmonary hypertension in a mouse model. They also showed that administration of EVs-MSCs suppressed hypoxic induction of both MCP-1 and HIMF/FIZZ1 in the lungs, which are strongly activated during lung hypoxia and are potent pro-inflammatory mediators (Figure 2) [188]. EVs BM-MSCs, with miR-29b-3p overexpression, effectively inhibit fibroblast proliferation, invasion migration and differentiation by inhibiting FZD6 expression, contributing to the antifibrotic effects of pulmonary fibroblasts [189]. Treatment of lipopolysaccharide-induced acute lung injury in mice using EVs derived from immature MSCs reduced inflammatory cell accumulation and alveolar septum thickness 48 h after injury compared to the control. In addition, young EVs-MSCs significantly reduced the protein, total cell and neutrophil counts as well as the level of pro-inflammatory cytokine IL-1β and increased the level of anti-inflammatory IL-10 in BAL after 48 h [190]. The results of Sun et al. showed that therapy with EVs from menstrual blood MSCs helped to reduce symptoms of pulmonary fibrosis, reduced collagen deposition and reduced the severity of pulmonary edema. After treatment of bleomycin-induced fibrosis with EVs-MSCs, levels of hydroxyproline (a marker of collagen deposition) and malondialdehyde (a biomarker of oxidative stress in the lungs) were reduced, and levels of glutathione peroxidase (which has an important protective function in the lungs [191]) were increased compared with the untreated group (Figure 2) [192].
Injection of EVs from human umbilical cord MSCs cultured under normal conditions (Nor-EVs) and EVs from human umbilical cord MSCs cultured under hypoxic conditions (Hypo-EVs) significantly reduced total cell count in BAL fluid of eosinophils and proinflammatory mediators (IL-4 and IL-13) in mice with simulated asthma. Compared to Nor-EVs, Hypo-EVs additionally prevented chronic allergic airway remodeling in mice, which was accompanied by a decrease in the expression of the profibrogenic markers α-smooth muscle actin (α-SMA), collagen-1 and TGF-β1-p-smad2/3 signaling pathway [193]. EVs derived from human umbilical cord blood MSCs (hUC-MSCs) therapy reduced inflammation in the lungs and the number of goblet cells, and inhibited lung destruction in a rat model of chronic obstructive pulmonary disease. Additionally, EVs hUC-MSCs partially reduced inflammation through the expression of PRKCZ and p65 and p50 NF-κβ subunits [194]. Treatment of mice with lipopolysaccharide-induced lung injury by intravenous and intratracheal administration of EVs-MSCs significantly reduced inflammatory cell infiltration, septal thickening, lung permeability, total cell and neutrophil counts in BAL. Here, a significant reduction of pro-inflammatory cytokines including interleukin-1β, interleukin-6 and TNF-α in BAL was also shown [195]. Treatment of EVs-MSCs carrying miR-23a-3p and miR-182-5p reversed the progression of lipopolysaccharide-induced lung damage and fibrosis by inhibiting NF-κB and Hedgehog pathways by downregulating Ikbkb and destabilizing IKKβ (Figure 2) [196].

6. Clinical Trials of IPF Treatment with MSCs and EVs

A number of clinical trials have been conducted to assess the safety and efficacy of IPF therapy with mesenchymal stem cells. For example, in one study, patients with mild to moderately severe IPF received a single intravenous injection of bone marrow MSCs (20, 100 and 200 × 106/mL). No serious adverse effects were observed, but by 60 weeks after the infusion, there was an average 3.0% decrease in FVC and a 5.4% decrease in carbon monoxide diffusing capacity of the lungs [197]. In another study, patients received intravenous umbilical cord MSCs (10 mL) with a cell density of 5 × 106–1 × 107/mL. There was a slight improvement in lung carbon monoxide diffusion capacity (from 46.3 to 63.1%) and fibrosis score (from 4.6 to 9.8%). Dyspnea scores decreased, with dyspnea at rest decreasing from 3.0 to 1.5; there was a 32.2% increase in maximal inspiratory pressure. The results of the study showed a trend towards an increase in systolic pulmonary artery pressure at 6 and 12 months after administration [198]. In addition, a clinical trial was conducted to investigate the safety of fat-derived MSCs in three endobronchial infusions (0.5 million cells per kg body weight per infusion) in patients with mild to moderate IPF. There were no serious or clinically significant adverse events and no deterioration in functional parameters or quality of life indicators [199].
Another study showed that endobronchial injection of bone marrow MSCs did not cause immediate serious adverse events in patients with IPF, but a certain proportion of patients suffered clinical progression of the disease. Genetic instability of MSCs during cultivation was also observed, which may serve as a limitation for the use of autologous MSCs for IPF therapy. [200]. When intravenous administration of bone marrow MSCs in concentrations of 2 × 107 MSCs/kg (group 1) and 1 × 108 MSCs/kg (group 2), a slower progression of pulmonary fibrosis and a smaller decrease in pulmonary diffusion capacity index were observed in the subjects from group 2 than in the subjects from group 1 [201].
The therapeutic properties of MSCs derived from the placenta were also evaluated. Patients with mild-to-moderate IPF were intravenously injected with MSCs at concentrations of 1 × 106 MSC/kg (n = 4) or 2 × 106 MSC/kg (n = 4). Further, forced vital capacity (FVC) and pulmonary diffusion capacity were assessed, and a 6 min walk test and chest computed tomography (CT) scan were performed. As a result, there was an increase in FVC (52.5–74.5%) and pulmonary diffusion capacity (29.5–40%); the 6 min walk test and CT scan were unchanged from the baseline [202].
In a small cohort study (n = 2) it was demonstrated that nebulisation or intravenous injection of telomerase-positive stem cells increased first-second forced expiratory volume by 14% to 27% and 25% to 70%, respectively [203].
Mesenchymal stem cells have received particular attention because of their ability to modulate the immune system and inhibit inflammation and fibrosis induced by SARS-CoV-2. A study was conducted to assess the safety and efficacy of intravenous administration of Wharton’s jelly-derived MSCs (150 × 106 cells per injection) to patients with COVID-19. The results showed that IL-10 and SDF-1 levels increased after cell therapy, while VEGF, TGF-β, IFN-γ, IL-6 and TNF-α levels decreased. At the same time, the zonal lesion score in both lungs was improved [204].
In 2020, Sengupta et al. conducted the first clinical trial on the safety and efficacy of EVs from allogeneic BM-MSCs (ExoFlo) for therapy of severe COVID-19. Intravenous administration of ExoFlo proved to be safe and was not associated with the occurrence of side effects at any time point. The survival rate was 83%. A single intravenous injection of ExoFlo showed an improvement in oxygenation, a 32% reduction in neutrophil counts and lymphopenia with an increase in mean CD3+, CD4+ and CD8+ lymphocyte counts of approximately 45%. Overall, this study demonstrates that therapy with EVs-MSCs contributed to reversing hypoxia, restoring immunity and suppressing the cytokine storm without treatment-related side effects [205].

7. Conclusions

The mechanism of idiopathic pulmonary fibrosis involves a complex interaction of different cell types, factors and signaling pathways. To date, there is a problem with the effectiveness of the applied methods of IPF therapy. Mesenchymal stem cells are the most attractive candidates because they are effective in repairing lung tissue, reducing collagen deposition, suppressing profibrotic genes, inhibiting pro-inflammatory transcripts, and inducing myofibroblast apoptosis. However, the limitations of their use have forced the search for alternative cell-free therapies. EVs-MSCs exhibit the properties of parental cells, promote lung tissue repair and have been confirmed to be safe in clinical trials.

Author Contributions

Conceptualization, M.G. and S.K.; writing—original draft preparation, S.K. and G.M.; writing—review and editing, M.G. and A.T.; visualization, S.K. and G.M.; supervision, M.G. All authors have read and agreed to the published version of the manuscript.

Funding

The reported study was funded by RSF according to the research project No. 21-75-10035.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All data generated or analyzed during this study are included in this published article. The data that support the findings of this study are available from the corresponding author upon request.

Acknowledgments

This paper has been supported by the Kazan Federal University Strategic Academic Leadership Program (PRIORITY-2030).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ley, B.; Collard, H.R.; King, T.E., Jr. Clinical Course and Prediction of Survival in Idiopathic Pulmonary Fibrosis. Am. J. Respir. Crit. Care Med. 2011, 183, 431–440. [Google Scholar] [CrossRef] [PubMed]
  2. Gross, T.J.; Hunninghake, G.W. Idiopathic Pulmonary Fibrosis. N. Engl. J. Med. 2001, 345, 517–525. [Google Scholar] [CrossRef] [PubMed]
  3. Yamashita, M.; Yamauchi, K.; Chiba, R.; Iwama, N.; Date, F.; Shibata, N.; Kumagai, H.; Risteli, J.; Sato, S.; Takahashi, T.; et al. The definition of fibrogenic processes in fibroblastic foci of idiopathic pulmonary fibrosis based on morphometric quantification of extracellular matrices. Hum. Pathol. 2009, 40, 1278–1287. [Google Scholar] [CrossRef] [PubMed]
  4. Shochet, G.E.; Brook, E.; Israeli-Shani, L.; Edelstein, E.; Shitrit, D. Fibroblast paracrine TNF-α signaling elevates integrin A5 expression in idiopathic pulmonary fibrosis (IPF). Respir. Res. 2017, 18, 122. [Google Scholar] [CrossRef]
  5. Pan, L.-H.; Yamauchi, K.; Uzuki, M.; Nakanishi, T.; Takigawa, M.; Inoue, H.; Sawai, T. Type II alveolar epithelial cells and interstitial fibroblasts express connective tissue growth factor in IPF. Eur. Respir. J. 2001, 17, 1220–1227. [Google Scholar] [CrossRef] [PubMed]
  6. Renaud, L.; da Silveira, W.A.; Takamura, N.; Hardiman, G.; Feghali-Bostwick, C. Prominence of IL6, IGF, TLR, and Bioenergetics Pathway Perturbation in Lung Tissues of Scleroderma Patients with Pulmonary Fibrosis. Front. Immunol. 2020, 11, 383. [Google Scholar] [CrossRef] [PubMed]
  7. Juge, P.-A.; Borie, R.; Kannengiesser, C.; Gazal, S.; Revy, P.; Wemeau-Stervinou, L.; Debray, M.-P.; Ottaviani, S.; Marchand-Adam, S.; Nathan, N.; et al. Shared genetic predisposition in rheumatoid arthritis-interstitial lung disease and familial pulmonary fibrosis. Eur. Respir. J. 2017, 49, 1602314. [Google Scholar] [CrossRef]
  8. Redente, E.F.; Aguilar, M.A.; Black, B.P.; Edelman, B.L.; Bahadur, A.N.; Humphries, S.M.; Lynch, D.A.; Wollin, L.; Riches, D.W.H. Nintedanib reduces pulmonary fibrosis in a model of rheumatoid arthritis-associated interstitial lung disease. Am. J. Physiol. Cell. Mol. Physiol. 2018, 314, L998–L1009. [Google Scholar] [CrossRef]
  9. Weng, J.; Tu, M.; Wang, P.; Zhou, X.; Wang, C.; Wan, X.; Zhou, Z.; Wang, L.; Zheng, X.; Li, J.; et al. Amiodarone induces cell proliferation and myofibroblast differentiation via ERK1/2 and p38 MAPK signaling in fibroblasts. Biomed. Pharmacother. 2019, 115, 108889. [Google Scholar] [CrossRef]
  10. Weng, J.; Chen, H.; Wu, H.; Tu, M.; Wang, Z.; Chen, D.; Wang, Z.; Chen, C. Amiodarone induces epithelial-mesenchymal transition in A549 cells via activation of TGF-β1. Drug Chem. Toxicol. 2018, 43, 415–422. [Google Scholar] [CrossRef]
  11. Zhang, H.; Wang, H.; Xia, Y.; Qi, N. Dual Effects of Hypoxia-Inducible Factors-1 Alpha in Bleomycin-Induced Pulmonary Fibrosis Treated by Human Umbilical Cord Mesenchymal Stem Cells. Stem Cells Int. 2021, 2021, 6658855. [Google Scholar] [CrossRef] [PubMed]
  12. Mikkelsen, L.F.; Rubak, S. Reversible lung fibrosis in a 6-year-old girl after long term nitrofurantoin treatment. BMC Pulm. Med. 2020, 20, 313. [Google Scholar] [CrossRef] [PubMed]
  13. 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, 116–127. [Google Scholar] [CrossRef] [PubMed]
  14. Lin, Y.; Tan, D.; Kan, Q.; Xiao, Z.; Jiang, Z. The Protective Effect of Naringenin on Airway Remodeling after Mycoplasma Pneumoniae Infection by Inhibiting Autophagy-Mediated Lung Inflammation and Fibrosis. Mediat. Inflamm. 2018, 2018, 8753894. [Google Scholar] [CrossRef]
  15. Camastra, G.; Ciolina, F.; Arcari, L.; Danti, M.; Musarò, S.; Cacciotti, L.; Sbarbati, S.; Ansalone, G. Heart and Lung Involvement Detected by Cardiac Magnetic Resonance Imaging in a Patient with Legionella Pneumophila Infection: Case Report. SN Compr. Clin. Med. 2021, 3, 1955–1959. [Google Scholar] [CrossRef]
  16. Svendsen, C.B.; Milman, N.; Nielsen, H.W.; Krogfelt, K.A.; Larsen, K.R. A prospective study evaluating the presence of Rickettsia in Danish patients with sarcoidosis. Scand. J. Infect. Dis. 2009, 41, 745–752. [Google Scholar] [CrossRef]
  17. Venkataraman, T.; Coleman, C.M.; Frieman, M.B. Overactive Epidermal Growth Factor Receptor Signaling Leads to Increased Fibrosis after Severe Acute Respiratory Syndrome Coronavirus Infection. J. Virol. 2017, 91, e00182-17. [Google Scholar] [CrossRef]
  18. Su, L.; Dong, Y.; Wang, Y.; Wang, Y.; Guan, B.; Lu, Y.; Wu, J.; Wang, X.; Li, D.; Meng, A.; et al. Potential role of senescent macrophages in radiation-induced pulmonary fibrosis. Cell Death Dis. 2021, 12, 527. [Google Scholar] [CrossRef]
  19. Kim, H.; Park, S.-H.; Han, S.Y.; Lee, Y.-S.; Cho, J.; Kim, J.-M. LXA4-FPR2 signaling regulates radiation-induced pulmonary fibrosis via crosstalk with TGF-β/Smad signaling. Cell Death Dis. 2020, 11, 653. [Google Scholar] [CrossRef]
  20. Albuquerque, D.A.; Seidl, V.R.; Santos, V.C.; Oliveira-Neto, J.A.; Capelozzi, V.L.; Rocco, P.R.; Zin, W.A. The Effect of Experimental Pleurodesis Caused by Aluminum Hydroxide on Lung and Chest Wall Mechanics. Lung 2001, 179, 293–303. [Google Scholar] [CrossRef]
  21. Blum, J.L.; Rosenblum, L.K.; Grunig, G.; Beasley, M.B.; Xiong, J.Q.; Zelikoff, J.T. Short-term inhalation of cadmium oxide nanoparticles alters pulmonary dynamics associated with lung injury, inflammation, and repair in a mouse model. Inhal. Toxicol. 2014, 26, 48–58. [Google Scholar] [CrossRef] [PubMed]
  22. Skoczyńska, A.; Gruszczyński, L.; Wojakowska, A.; Ścieszka, M.; Turczyn, B.; Schmidt, E. Association between the Type of Workplace and Lung Function in Copper Miners. BioMed Res. Int. 2016, 2016, 5928572. [Google Scholar] [CrossRef]
  23. Song, L.C.; Chen, X.X.; Meng, J.G.; Hu, M.; Huan, J.B.; Wu, J.; Xiao, K.; Han, Z.H.; Xie, L.X. Effects of different corticosteroid doses and durations on smoke inhalation-induced acute lung injury and pulmonary fibrosis in the rat. Int. Immunopharmacol. 2019, 71, 392–403. [Google Scholar] [CrossRef]
  24. Steele, M.P.; Speer, M.C.; Loyd, J.E.; Brown, K.K.; Herron, A.; Slifer, S.H.; Burch, L.H.; Wahidi, M.M.; Phillips, J.A.; Sporn, T.A.; et al. Clinical and Pathologic Features of Familial Interstitial Pneumonia. Am. J. Respir. Crit. Care Med. 2005, 172, 1146–1152. [Google Scholar] [CrossRef] [PubMed]
  25. Tsakiri, K.D.; Cronkhite, J.T.; Kuan, P.J.; Xing, C.; Raghu, G.; Weissler, J.C.; Rosenblatt, R.L.; Shay, J.W.; Garcia, C.K. Adult-onset pulmonary fibrosis caused by mutations in telomerase. Proc. Natl. Acad. Sci. USA 2007, 104, 7552–7557. [Google Scholar] [CrossRef]
  26. Nogee, L.M.; Dunbar, A.E.; Wert, S.E.; Askin, F.; Hamvas, A.; Whitsett, J.A. A Mutation in the Surfactant Protein C Gene Associated with Familial Interstitial Lung Disease. N. Engl. J. Med. 2001, 344, 573–579. [Google Scholar] [CrossRef] [PubMed]
  27. Hu, Y.; Ma, Z.; Guo, Z.; Zhao, F.; Wang, Y.; Cai, L.; Yang, J. Type 1 Diabetes Mellitus is an Independent Risk Factor for Pulmonary Fibrosis. Cell Biophys. 2014, 70, 1385–1391. [Google Scholar] [CrossRef]
  28. Gao, F.; Hobson, A.R.; Shang, Z.M.; Pei, Y.X.; Gao, Y.; Wang, J.X.; Huang, W.N. The prevalence of gastro-esophageal reflux disease and esophageal dysmotility in Chinese patients with idiopathic pulmonary fibrosis. BMC Gastroenterol. 2015, 15, 26. [Google Scholar] [CrossRef]
  29. Perng, D.-W.; Chang, K.-T.; Su, K.-C.; Wu, Y.-C.; Wu, M.-T.; Hsu, W.-H.; Tsai, C.-M.; Lee, Y.-C. Exposure of Airway Epithelium to Bile Acids Associated with Gastroesophageal Reflux Symptoms. Chest 2007, 132, 1548–1556. [Google Scholar] [CrossRef]
  30. Tang, Y.-W.; Johnson, J.E.; Browning, P.J.; Cruz-Gervis, R.A.; Davis, A.; Graham, B.S.; Brigham, K.L.; Oates, J.A., Jr.; Loyd, J.E.; Stecenko, A.A. Herpesvirus DNA Is Consistently Detected in Lungs of Patients with Idiopathic Pulmonary Fibrosis. J. Clin. Microbiol. 2003, 41, 2633–2640. [Google Scholar] [CrossRef] [Green Version]
  31. Vicente, R.; Morales, P.; Ramos, F.; Sole, A.; Mayo, M.; Villalain, C. Perioperative Complications of Lung Transplantation in Patients with Emphysema and Fibrosis: Experience From 1992–2002. Transplant. Proc. 2006, 38, 2560–2562. [Google Scholar] [CrossRef] [PubMed]
  32. Wei, D.; Gao, F.; Wu, B.; Zhou, M.; Zhang, J.; Yang, H.; Liu, N.; Fan, L.; Chen, J. Single versus bilateral lung transplantation for idiopathic pulmonary fibrosis. Clin. Respir. J. 2019, 13, 376–383. [Google Scholar] [CrossRef] [PubMed]
  33. Barratt, S.L.; Mulholland, S.; Al Jbour, K.; Steer, H.; Gutsche, M.; Foley, N.; Srivastava, R.; Sharp, C.; Adamali, H.I. South–West of England’s Experience of the Safety and Tolerability Pirfenidone and Nintedanib for the Treatment of Idiopathic Pulmonary Fibrosis (IPF). Front. Pharmacol. 2018, 9, 1480. [Google Scholar] [CrossRef]
  34. Oltmanns, U.; Kahn, N.; Palmowski, K.; Träger, A.; Wenz, H.; Heussel, C.P.; Schnabel, P.A.; Puderbach, M.; Wiebel, M.; Ehlers-Tenenbaum, S.; et al. Pirfenidone in Idiopathic Pulmonary Fibrosis: Real-Life Experience from a German Tertiary Referral Center for Interstitial Lung Diseases. Respiration 2014, 88, 199–207. [Google Scholar] [CrossRef] [PubMed]
  35. Kato, M.; Sasaki, S.; Nakamura, T.; Kurokawa, K.; Yamada, T.; Ochi, Y.; Ihara, H.; Takahashi, F.; Takahashi, K. Gastrointestinal adverse effects of nintedanib and the associated risk factors in patients with idiopathic pulmonary fibrosis. Sci. Rep. 2019, 9, 12062. [Google Scholar] [CrossRef]
  36. Abe, M.; Tsushima, K.; Sakayori, M.; Suzuki, K.; Ikari, J.; Terada, J.; Tatsumi, K. Utility of nintedanib for severe idiopathic pulmonary fibrosis: A single-center retrospective study. Drug Des. Dev. Ther. 2018, 12, 3369–3375. [Google Scholar] [CrossRef]
  37. da Silva Meirelles, L.; Chagastelles, P.C.; Nardi, N.B. Mesenchymal stem cells reside in virtually all post-natal organs and tissues. J. Cell Sci. 2006, 119, 2204–2213. [Google Scholar] [CrossRef]
  38. Yu, J.; Zheng, C.; Ren, X.; Li, J.; Liu, M.; Zhang, L.; Liang, L.; Du, W.; Han, Z.C. Intravenous Administration of Bone Marrow Mesenchymal Stem Cells Benefits Experimental Autoimmune Myasthenia Gravis Mice Through an Immunomodulatory Action. Scand. J. Immunol. 2010, 72, 242–249. [Google Scholar] [CrossRef]
  39. Di Nicola, M.; Carlo-Stella, C.; Magni, M.; Milanesi, M.; Longoni, P.D.; Matteucci, P.; Grisanti, S.; Gianni, A.M. Human bone marrow stromal cells suppress T-lymphocyte proliferation induced by cellular or nonspecific mitogenic stimuli. Blood 2002, 99, 3838–3843. [Google Scholar] [CrossRef]
  40. Moodley, Y.; Vaghjiani, V.; Chan, J.; Baltic, S.; Ryan, M.; Tchongue, J.; Samuel, C.S.; Murthi, P.; Parolini, O.; Manuelpillai, U. Anti-Inflammatory Effects of Adult Stem Cells in Sustained Lung Injury: A Comparative Study. PLoS ONE 2013, 8, e69299. [Google Scholar] [CrossRef] [Green Version]
  41. Jiang, H.; Zhang, J.; Zhang, Z.; Ren, S.; Zhang, C. Effect of transplanted adipose-derived stem cells in mice exhibiting idiopathic pulmonary fibrosis. Mol. Med. Rep. 2015, 12, 5933–5938. [Google Scholar] [CrossRef] [PubMed]
  42. Aguilar, S.; Nye, E.; Chan, J.; Loebinger, M.; Spencer-Dene, B.; Fisk, N.; Stamp, G.; Bonnet, D.; Janes, S.M. Murine but Not Human Mesenchymal Stem Cells Generate Osteosarcoma-Like Lesions in the Lung. Stem Cells 2007, 25, 1586–1594. [Google Scholar] [CrossRef] [PubMed]
  43. Baertschiger, R.M.; Serre-Beinier, V.; Morel, P.; Bosco, D.; Peyrou, M.; Clément, S.; Sgroi, A.; Kaelin, A.; Buhler, L.H.; Gonelle-Gispert, C. Fibrogenic Potential of Human Multipotent Mesenchymal Stromal Cells in Injured Liver. PLoS ONE 2009, 4, e6657. [Google Scholar] [CrossRef]
  44. Kuriyan, A.E.; Albini, T.A.; Townsend, J.H.; Rodriguez, M.; Pandya, H.K.; Leonard, R.E.; Parrott, M.B.; Rosenfeld, P.J.; Flynn, H.W.; Goldberg, J.L. Vision Loss after Intravitreal Injection of Autologous “Stem Cells” for AMD. N. Engl. J. Med. 2017, 376, 1047–1053. [Google Scholar] [CrossRef]
  45. Chen, W.; Huang, Y.; Han, J.; Yu, L.; Li, Y.; Lu, Z.; Li, H.; Liu, Z.; Shi, C.; Duan, F.; et al. Immunomodulatory effects of mesenchymal stromal cells-derived exosome. Immunol. Res. 2016, 64, 831–840. [Google Scholar] [CrossRef]
  46. Raghav, A.; Khan, Z.; Upadhayay, V.; Tripathi, P.; Gautam, K.; Mishra, B.; Ahmad, J.; Jeong, G.-B. Mesenchymal Stem Cell-Derived Exosomes Exhibit Promising Potential for Treating SARS-CoV-2-Infected Patients. Cells 2021, 10, 587. [Google Scholar] [CrossRef]
  47. Breitbach, M.; Bostani, T.; Roell, W.; Xia, Y.; Dewald, O.; Nygren, J.M.; Fries, J.W.U.; Tiemann, K.; Bohlen, H.; Hescheler, J.; et al. Potential risks of bone marrow cell transplantation into infarcted hearts. Blood 2007, 110, 1362–1369. [Google Scholar] [CrossRef]
  48. Chang, M.G.; Tung, L.; Sekar, R.B.; Chang, C.Y.; Cysyk, J.; Dong, P.; Marbán, E.; Abraham, M.R. Proarrhythmic Potential of Mesenchymal Stem Cell Transplantation Revealed in an In Vitro Coculture Model. Circulation 2006, 113, 1832–1841. [Google Scholar] [CrossRef]
  49. Gomzikova, M.O.; Zhuravleva, M.N.; Miftakhova, R.R.; Arkhipova, S.S.; Evtugin, V.G.; Khaiboullina, S.; Kiyasov, A.P.; Persson, J.L.; Mongan, N.P.; Pestell, R.G.; et al. Cytochalasin B-induced membrane vesicles convey angiogenic activity of parental cells. Oncotarget 2017, 8, 70496–70507. [Google Scholar] [CrossRef]
  50. Gomzikova, M.O.; Aimaletdinov, A.M.; Bondar, O.V.; Starostina, I.G.; Gorshkova, N.V.; Neustroeva, O.A.; Kletukhina, S.K.; Kurbangaleeva, S.V.; Vorobev, V.V.; Garanina, E.E.; et al. Immunosuppressive properties of cytochalasin B-induced membrane vesicles of mesenchymal stem cells: Comparing with extracellular vesicles derived from mesenchymal stem cells. Sci. Rep. 2020, 10, 10740. [Google Scholar] [CrossRef]
  51. Katzenstein, A.-L.A.; Myers, J.L. Idiopathic Pulmonary Fibrosis. Am. J. Respir. Crit. Care Med. 1998, 157, 1301–1315. [Google Scholar] [CrossRef]
  52. Ryu, J.H.; Colby, T.V.; Hartman, T.E. Idiopathic Pulmonary Fibrosis: Current Concepts. Mayo Clin. Proc. 1998, 73, 1085–1101. [Google Scholar] [CrossRef] [PubMed]
  53. Kim, K.K.; Dotson, M.R.; Agarwal, M.; Yang, J.; Bradley, P.B.; Subbotina, N.; Osterholzer, J.J.; Sisson, T.H. Efferocytosis of apoptotic alveolar epithelial cells is sufficient to initiate lung fibrosis. Cell Death Dis. 2018, 9, 1056. [Google Scholar] [CrossRef] [PubMed]
  54. Lee, J.-M.; Yoshida, M.; Kim, M.-S.; Lee, J.-H.; Baek, A.-R.; Jang, A.S.; Kim, D.J.; Minagawa, S.; Chin, S.S.; Park, C.-S.; et al. Involvement of Alveolar Epithelial Cell Necroptosis in Idiopathic Pulmonary Fibrosis Pathogenesis. Am. J. Respir. Cell Mol. Biol. 2018, 59, 215–224. [Google Scholar] [CrossRef] [PubMed]
  55. Yuan, X.; Wang, E.; Xiao, X.; Wang, J.; Yang, X.; Yang, P.; Li, G.; Liu, Z. The role of IL-25 in the reduction of oxidative stress and the apoptosis of airway epithelial cells with specific immunotherapy in an asthma mouse model. Am. J. Transl. Res. 2017, 9, 4137–4148. [Google Scholar]
  56. Mendes, J.A.; Ribeiro, M.C.; Filho, G.J.R.; Rocha, T.; Muscará, M.N.; Costa, S.K.; Ferreira, H.H. Hydrogen sulfide inhibits apoptosis and protects the bronchial epithelium in an allergic inflammation mice model. Int. Immunopharmacol. 2019, 73, 435–441. [Google Scholar] [CrossRef]
  57. Carcaterra, M.; Caruso, C. Alveolar epithelial cell type II as main target of SARS-CoV-2 virus and COVID-19 development via NF-Kb pathway deregulation: A physio-pathological theory. Med. Hypotheses 2020, 146, 110412. [Google Scholar] [CrossRef]
  58. Tsuji, T.; Aoshiba, K.; Nagai, A. Cigarette Smoke Induces Senescence in Alveolar Epithelial Cells. Am. J. Respir. Cell Mol. Biol. 2004, 31, 643–649. [Google Scholar] [CrossRef]
  59. Wilson, M.S.; Wynn, T.A. Pulmonary fibrosis: Pathogenesis, etiology and regulation. Mucosal Immunol. 2009, 2, 103–121. [Google Scholar] [CrossRef]
  60. Witherden, I.R.; Bon, E.J.V.; Goldstraw, P.; Ratcliffe, C.; Pastorino, U.; Tetley, T.D. Primary Human Alveolar Type II Epithelial Cell Chemokine Release. Am. J. Respir. Cell Mol. Biol. 2004, 30, 500–509. [Google Scholar] [CrossRef]
  61. Xiang, G.-A.; Zhang, Y.-D.; Su, C.-C.; Ma, Y.-Q.; Li, Y.-M.; Zhou, X.; Wei, L.-Q.; Ji, W.-J. Dynamic changes of mononuclear phagocytes in circulating, pulmonary alveolar and interstitial compartments in a mouse model of experimental silicosis. Inhal. Toxicol. 2016, 28, 393–402. [Google Scholar] [CrossRef] [PubMed]
  62. Li, H.-P.; Li, X.; He, G.-J.; Yi, X.-H.; Kaplan, A.P. The influence of dexamethasone on the proliferation and apoptosis of pulmonary inflammatory cells in bleomycin-induced pulmonary fibrosis in rats. Respirology 2004, 9, 25–32. [Google Scholar] [CrossRef] [PubMed]
  63. Weng, C.-M.; Chen, B.-C.; Wang, C.-H.; Feng, P.-H.; Lee, M.-J.; Huang, C.-D.; Kuo, H.-P.; Lin, C.-H. The Endothelin A Receptor Mediates Fibrocyte Differentiation in Chronic Obstructive Asthma. The Involvement of Connective Tissue Growth Factor. Am. J. Respir. Crit. Care Med. 2013, 188, 298–308. [Google Scholar] [CrossRef] [PubMed]
  64. Hill, C.; Jones, M.; Davies, D.; Wang, Y. Epithelial-Mesenchymal Transition Contributes to Pulmonary Fibrosis via Aberrant Epithelial/Fibroblastic Cross-Talk. J. Lung Health Dis. 2019, 3, 31–35. [Google Scholar] [CrossRef]
  65. Strieter, R.M. Pathogenesis and Natural History of Usual Interstitial Pneumonia. Chest 2005, 128, 526S–532S. [Google Scholar] [CrossRef]
  66. Zolak, J.S.; de Andrade, J.A. Idiopathic Pulmonary Fibrosis. Immunol. Allergy Clin. N. Am. 2012, 32, 473–485. [Google Scholar] [CrossRef]
  67. Li, X.; Bi, Z.; Liu, S.; Gao, S.; Cui, Y.; Huang, K.; Huang, M.; Mao, J.; Li, L.; Gao, J.; et al. Antifibrotic Mechanism of Cinobufagin in Bleomycin-Induced Pulmonary Fibrosis in Mice. Front. Pharmacol. 2019, 10, 1021. [Google Scholar] [CrossRef]
  68. Balestro, E.; Calabrese, F.; Turato, G.; Lunardi, F.; Bazzan, E.; Marulli, G.; Biondini, D.; Rossi, E.; Sanduzzi, A.; Rea, F.; et al. Immune Inflammation and Disease Progression in Idiopathic Pulmonary Fibrosis. PLoS ONE 2016, 11, e0154516. [Google Scholar] [CrossRef]
  69. Selman, M.; Pardo, A. The leading role of epithelial cells in the pathogenesis of idiopathic pulmonary fibrosis. Cell. Signal. 2019, 66, 109482. [Google Scholar] [CrossRef]
  70. Liu, F.; Mih, J.D.; Shea, B.S.; Kho, A.T.; Sharif, A.S.; Tager, A.M.; Tschumperlin, D.J. Feedback amplification of fibrosis through matrix stiffening and COX-2 suppression. J. Cell Biol. 2010, 190, 693–706. [Google Scholar] [CrossRef]
  71. Greenberg, R.S.; Bernstein, A.M.; Benezra, M.; Gelman, I.H.; Taliana, L.; Masur, S.K. FAK-dependent regulation of myofibroblast differentiation. FASEB J. 2006, 20, 1006–1008. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Akhmetshina, A.; Dees, C.; Pileckyte, M.; Szucs, G.; Spriewald, B.M.; Zwerina, J.; Distler, O.; Schett, G.; Distler, J.H.W. Rho-associated kinases are crucial for myofibroblast differentiation and production of extracellular matrix in scleroderma fibroblasts. Arthritis Care Res. 2008, 58, 2553–2564. [Google Scholar] [CrossRef] [PubMed]
  73. Georges, P.; Hui, J.-J.; Gombos, Z.; McCormick, M.E.; Wang, A.Y.; Uemura, M.; Mick, R.; Janmey, P.A.; Furth, E.E.; Wells, R.G. Increased stiffness of the rat liver precedes matrix deposition: Implications for fibrosis. Am. J. Physiol. Liver Physiol. 2007, 293, G1147–G1154. [Google Scholar] [CrossRef]
  74. Herrera, J.; Forster, C.; Pengo, T.; Montero, A.; Swift, J.; Schwartz, M.A.; Henke, C.A.; Bitterman, P.B. Registration of the extracellular matrix components constituting the fibroblastic focus in idiopathic pulmonary fibrosis. JCI Insight 2019, 4, e125185. [Google Scholar] [CrossRef]
  75. Xia, H.; Gilbertsen, A.; Herrera, J.; Racila, E.; Smith, K.; Peterson, M.; Griffin, T.; Benyumov, A.; Yang, L.; Bitterman, P.; et al. Calcium-binding protein S100A4 confers mesenchymal progenitor cell fibrogenicity in idiopathic pulmonary fibrosis. J. Clin. Investig. 2017, 127, 2586–2597. [Google Scholar] [CrossRef] [PubMed]
  76. El Agha, E.; Moiseenko, A.; Kheirollahi, V.; De Langhe, S.; Crnkovic, S.; Kwapiszewska, G.; Szibor, M.; Kosanovic, D.; Schwind, F.; Schermuly, R.T.; et al. Two-Way Conversion between Lipogenic and Myogenic Fibroblastic Phenotypes Marks the Progression and Resolution of Lung Fibrosis. Cell Stem Cell 2017, 20, 261–273.e3. [Google Scholar] [CrossRef] [PubMed]
  77. Walton, K.L.; Johnson, K.E.; Harrison, C.A. Targeting TGF-β Mediated SMAD Signaling for the Prevention of Fibrosis. Front. Pharmacol. 2017, 8, 461. [Google Scholar] [CrossRef] [PubMed]
  78. Ichikawa, T.; Sugiura, H.; Koarai, A.; Kikuchi, T.; Hiramatsu, M.; Kawabata, H.; Akamatsu, K.; Hirano, T.; Nakanishi, M.; Matsunaga, K.; et al. 25-Hydroxycholesterol promotes fibroblast-mediated tissue remodeling through NF-κB dependent pathway. Exp. Cell Res. 2013, 319, 1176–1186. [Google Scholar] [CrossRef] [PubMed]
  79. Marangoni, R.G.; Korman, B.; Varga, J. Adipocytic Progenitor Cells Give Rise to Pathogenic Myofibroblasts: Adipocyte-to-Mesenchymal Transition and Its Emerging Role in Fibrosis in Multiple Organs. Curr. Rheumatol. Rep. 2020, 22, 79. [Google Scholar] [CrossRef]
  80. Rehan, V.K.; Wang, Y.; Sugano, S.; Romero, S.; Chen, X.; Santos, J.; Khazanchi, A.; Torday, J.S. Mechanism of nicotine-induced pulmonary fibroblast transdifferentiation. Am. J. Physiol. Cell. Mol. Physiol. 2005, 289, L667–L676. [Google Scholar] [CrossRef]
  81. Rehan, V.K.; Torday, J.S. Hyperoxia augments pulmonary lipofibroblast-to-myofibroblast transdifferentiation. Cell Biophys. 2003, 38, 239–250. [Google Scholar] [CrossRef]
  82. Waghray, M.; Cui, Z.; Horowitz, J.; Subramanian, I.M.; Martinez, F.J.; Toews, G.B.; Thannickal, V.J. Hydrogen peroxide is a diffusible paracrine signal for the induction of epithelial cell death by activated myofibroblasts. FASEB J. 2005, 19, 854–856. [Google Scholar] [CrossRef] [PubMed]
  83. Kim, K.K.; Wei, Y.; Szekeres, C.; Kugler, M.C.; Wolters, P.J.; Hill, M.L.; Frank, J.A.; Brumwell, A.N.; Wheeler, S.E.; Kreidberg, J.A.; et al. Epithelial cell α3β1 integrin links β-catenin and Smad signaling to promote myofibroblast formation and pulmonary fibrosis. J. Clin. Investig. 2008, 119, 213–224. [Google Scholar] [CrossRef] [PubMed]
  84. Santini, M.P.; Malide, D.; Hoffman, G.; Pandey, G.; D’Escamard, V.; Nomura-Kitabayashi, A.; Rovira, I.; Kataoka, H.; Ochando, J.; Harvey, R.; et al. Tissue-Resident PDGFRα+ Progenitor Cells Contribute to Fibrosis versus Healing in a Context- and Spatiotemporally Dependent Manner. Cell Rep. 2020, 30, 555–570.e7. [Google Scholar] [CrossRef] [PubMed]
  85. Sava, P.; Ramanathan, A.; Dobronyi, A.; Peng, X.; Sun, H.; Ledesma-Mendoza, A.; Herzog, E.L.; Gonzalez, A.L. Human pericytes adopt myofibroblast properties in the microenvironment of the IPF lung. JCI Insight 2017, 2, e96352. [Google Scholar] [CrossRef]
  86. Moore, B.B.; Murray, L.; Das, A.; Wilke, C.A.; Herrygers, A.B.; Toews, G.B. The Role of CCL12 in the Recruitment of Fibrocytes and Lung Fibrosis. Am. J. Respir. Cell Mol. Biol. 2006, 35, 175–181. [Google Scholar] [CrossRef]
  87. Phillips, R.J.; Burdick, M.D.; Hong, K.; Lutz, M.A.; Murray, L.A.; Xue, Y.Y.; Belperio, J.A.; Keane, M.P.; Strieter, R.M. Circulating fibrocytes traffic to the lungs in response to CXCL12 and mediate fibrosis. J. Clin. Investig. 2004, 114, 438–446. [Google Scholar] [CrossRef]
  88. Shao, D.D.; Suresh, R.; Vakil, V.; Gomer, R.H.; Pilling, D. Pivotal Advance: Th-1 cytokines inhibit, and Th-2 cytokines promote fibrocyte differentiation. J. Leukoc. Biol. 2008, 83, 1323–1333. [Google Scholar] [CrossRef]
  89. Wei, J.; Fang, F.; Lam, A.P.; Sargent, J.L.; Hamburg, E.; Hinchcliff, M.E.; Gottardi, C.J.; Atit, R.; Whitfield, M.L.; Varga, J. Wnt/β-catenin signaling is hyperactivated in systemic sclerosis and induces Smad-dependent fibrotic responses in mesenchymal cells. Arthritis Care Res. 2012, 64, 2734–2745. [Google Scholar] [CrossRef]
  90. Jiang, Z.; Liu, G.; Meng, F.; Wang, W.; Hao, P.; Xiang, Y.; Wang, Y.; Han, R.; Li, F.; Wang, L.; et al. Paracrine effects of mesenchymal stem cells on the activation of keratocytes. Br. J. Ophthalmol. 2017, 101, 1583–1590. [Google Scholar] [CrossRef]
  91. Hinz, B. Masters and servants of the force: The role of matrix adhesions in myofibroblast force perception and transmission. Eur. J. Cell Biol. 2006, 85, 175–181. [Google Scholar] [CrossRef] [PubMed]
  92. Hill, C.; Li, J.; Liu, D.; Conforti, F.; Brereton, C.J.; Yao, L.; Zhou, Y.; Alzetani, A.; Chee, S.J.; Marshall, B.G.; et al. Autophagy inhibition-mediated epithelial–mesenchymal transition augments local myofibroblast differentiation in pulmonary fibrosis. Cell Death Dis. 2019, 10, 591. [Google Scholar] [CrossRef] [PubMed]
  93. Wang, Y.-C.; Chen, Q.; Luo, J.-M.; Nie, J.; Meng, Q.-H.; Shuai, W.; Xie, H.; Xia, J.-M.; Wang, H. Notch1 promotes the pericyte-myofibroblast transition in idiopathic pulmonary fibrosis through the PDGFR/ROCK1 signal pathway. Exp. Mol. Med. 2019, 51, 1–11. [Google Scholar] [CrossRef] [PubMed]
  94. Atanassoff, A.P.; Wolfmeier, H.; Schoenauer, R.; Hostettler, A.; Ring, A.; Draeger, A.; Babiychuk, E.B. Microvesicle Shedding and Lysosomal Repair Fulfill Divergent Cellular Needs during the Repair of Streptolysin O-Induced Plasmalemmal Damage. PLoS ONE 2014, 9, e89743. [Google Scholar] [CrossRef] [PubMed]
  95. Andersson-Sjöland, A.; Karlsson, J.C.; Rydell-Törmänen, K. ROS-induced endothelial stress contributes to pulmonary fibrosis through pericytes and Wnt signaling. Lab. Investig. 2015, 96, 206–217. [Google Scholar] [CrossRef]
  96. Strieter, R.M.; Keeley, E.C.; Hughes, M.A.; Burdick, M.D.; Mehrad, B. The role of circulating mesenchymal progenitor cells (fibrocytes) in the pathogenesis of pulmonary fibrosis. J. Leukoc. Biol. 2009, 86, 1111–1118. [Google Scholar] [CrossRef]
  97. Quan, T.E.; Cowper, S.E.; Bucala, R. The role of circulating fibrocytes in fibrosis. Curr. Rheumatol. Rep. 2006, 8, 145–150. [Google Scholar] [CrossRef]
  98. Liu, S.-S.; Liu, C.; Lv, X.-X.; Cui, B.; Yan, J.; Li, Y.-X.; Li, K.; Hua, F.; Zhang, X.-W.; Yu, J.-J.; et al. The chemokine CCL1 triggers an AMFR-SPRY1 pathway that promotes differentiation of lung fibroblasts into myofibroblasts and drives pulmonary fibrosis. Immunity 2021, 54, 2042–2056.e8. [Google Scholar] [CrossRef]
  99. Schneider, D.J.; Wu, M.; Le, T.T.; Cho, S.; Brenner, M.B.; Blackburn, M.R.; Agarwal, S.K. Cadherin-11 contributes to pulmonary fibrosis: Potential role in TGF-β production and epithelial to mesenchymal transition. FASEB J. 2011, 26, 503–512. [Google Scholar] [CrossRef]
  100. Wei, J.; Melichian, D.; Komura, K.; Hinchcliff, M.; Lam, A.P.; Lafyatis, R.; Gottardi, C.; MacDougald, O.; Varga, J. Canonical Wnt signaling induces skin fibrosis and subcutaneous lipoatrophy: A novel mouse model for scleroderma? Arthritis Care Res. 2011, 63, 1707–1717. [Google Scholar] [CrossRef]
  101. Kruglikov, I.L.; Scherer, P.E. The Role of Adipocytes and Adipocyte-Like Cells in the Severity of COVID-19 Infections. Obesity 2020, 28, 1187–1190. [Google Scholar] [CrossRef] [PubMed]
  102. Lagares, D.; Busnadiego, O.; García-Fernández, R.A.; Kapoor, M.; Liu, S.; Carter, D.E.; Abraham, D.; Shi-Wen, X.; Carreira, P.; Fontaine, B.A.; et al. Inhibition of focal adhesion kinase prevents experimental lung fibrosis and myofibroblast formation. Arthritis Care Res. 2011, 64, 1653–1664. [Google Scholar] [CrossRef] [PubMed]
  103. Robinson, C.M.; Neary, R.; Levendale, A.; Watson, C.J.; Baugh, J.A. Hypoxia-induced DNA hypermethylation in human pulmonary fibroblasts is associated with Thy-1 promoter methylation and the development of a pro-fibrotic phenotype. Respir. Res. 2012, 13, 74. [Google Scholar] [CrossRef] [PubMed]
  104. Garrett, S.M.; Hsu, E.; Thomas, J.M.; Pilewski, J.M.; Feghali-Bostwick, C. Insulin-like growth factor (IGF)-II- mediated fibrosis in pathogenic lung conditions. PLoS ONE 2019, 14, e0225422. [Google Scholar] [CrossRef]
  105. Varisco, B.M.; Ambalavanan, N.; Whitsett, J.A.; Hagood, J.S. Thy-1 Signals through PPARγ to Promote Lipofibroblast Differentiation in the Developing Lung. Am. J. Respir. Cell Mol. Biol. 2012, 46, 765–772. [Google Scholar] [CrossRef]
  106. Zhang, H.-Y.; Phan, S.H. Inhibition of Myofibroblast Apoptosis by Transforming Growth Factor β1. Am. J. Respir. Cell Mol. Biol. 1999, 21, 658–665. [Google Scholar] [CrossRef]
  107. MacKenzie, B.; Korfei, M.; Henneke, I.; Sibinska, Z.; Tian, X.; Hezel, S.; Dilai, S.; Wasnick, R.; Schneider, B.; Wilhelm, J.; et al. Increased FGF1-FGFRc expression in idiopathic pulmonary fibrosis. Respir. Res. 2015, 16, 83. [Google Scholar] [CrossRef]
  108. Huang, S.K.; White, E.S.; Wettlaufer, S.H.; Grifka, H.; Hogaboam, C.M.; Thannickal, V.J.; Horowitz, J.C.; Peters-Golden, M. Prostaglandin E2 induces fibroblast apoptosis by modulating multiple survival pathways. FASEB J. 2009, 23, 4317–4326. [Google Scholar] [CrossRef]
  109. Kim, K.-H.; Chen, C.-C.; Monzon, R.I.; Lau, L.F. Matricellular Protein CCN1 Promotes Regression of Liver Fibrosis through Induction of Cellular Senescence in Hepatic Myofibroblasts. Mol. Cell. Biol. 2013, 33, 2078–2090. [Google Scholar] [CrossRef]
  110. Maher, T.M.; Evans, I.C.; Bottoms, S.E.; Mercer, P.F.; Thorley, A.J.; Nicholson, A.G.; Laurent, G.J.; Tetley, T.D.; Chambers, R.C.; McAnulty, R.J. Diminished Prostaglandin E2 Contributes to the Apoptosis Paradox in Idiopathic Pulmonary Fibrosis. Am. J. Respir. Crit. Care Med. 2010, 182, 73–82. [Google Scholar] [CrossRef]
  111. Nho, R.S.; Peterson, M.; Hergert, P.; Henke, C.A. FoxO3a (Forkhead Box O3a) Deficiency Protects Idiopathic Pulmonary Fibrosis (IPF) Fibroblasts from Type I Polymerized Collagen Matrix-Induced Apoptosis via Caveolin-1 (cav-1) and Fas. PLoS ONE 2013, 8, e61017. [Google Scholar] [CrossRef] [PubMed]
  112. Moodley, Y.P.; Caterina, P.; Scaffidi, A.K.; Misso, N.L.; Papadimitriou, J.M.; McAnulty, R.J.; Laurent, G.J.; Thompson, P.J.; Knight, D.A. Comparison of the morphological and biochemical changes in normal human lung fibroblasts and fibroblasts derived from lungs of patients with idiopathic pulmonary fibrosis during FasL-induced apoptosis. J. Pathol. 2004, 202, 486–495. [Google Scholar] [CrossRef] [PubMed]
  113. Komura, K.; Yanaba, K.; Horikawa, M.; Ogawa, F.; Fujimoto, M.; Tedder, T.F.; Sato, S. CD19 regulates the development of bleomycin-induced pulmonary fibrosis in a mouse model. Arthritis Care Res. 2008, 58, 3574–3584. [Google Scholar] [CrossRef] [PubMed]
  114. Cargnoni, A.; Romele, P.; Signoroni, P.B.; Farigu, S.; Magatti, M.; Vertua, E.; Toschi, I.; Cesari, V.; Silini, A.R.; Stefani, F.R.; et al. Amniotic MSCs reduce pulmonary fibrosis by hampering lung B-cell recruitment, retention, and maturation. Stem Cells Transl. Med. 2020, 9, 1023–1035. [Google Scholar] [CrossRef] [PubMed]
  115. Mora, A.L.; Bueno, M.; Rojas, M. Mitochondria in the spotlight of aging and idiopathic pulmonary fibrosis. J. Clin. Investig. 2017, 127, 405–414. [Google Scholar] [CrossRef] [PubMed]
  116. Yao, Y.; Fan, X.-L.; Jiang, D.; Zhang, Y.; Li, X.; Xu, Z.-B.; Fang, S.-B.; Chiu, S.; Tse, H.-F.; Lian, Q.; et al. Connexin 43-Mediated Mitochondrial Transfer of iPSC-MSCs Alleviates Asthma Inflammation. Stem Cell Rep. 2018, 11, 1120–1135. [Google Scholar] [CrossRef]
  117. Shi, L.; Han, Q.; Hong, Y.; Li, W.; Gong, G.; Cui, J.; Mao, M.; Liang, X.; Hu, B.; Li, X.; et al. Inhibition of miR-199a-5p rejuvenates aged mesenchymal stem cells derived from patients with idiopathic pulmonary fibrosis and improves their therapeutic efficacy in experimental pulmonary fibrosis. Stem Cell Res. Ther. 2021, 12, 147. [Google Scholar] [CrossRef]
  118. Zanoni, M.; Cortesi, M.; Zamagni, A.; Tesei, A. The Role of Mesenchymal Stem Cells in Radiation-Induced Lung Fibrosis. Int. J. Mol. Sci. 2019, 20, 3876. [Google Scholar] [CrossRef]
  119. Huang, T.; Zhang, T.; Jiang, X.; Li, A.; Su, Y.; Bian, Q.; Wu, H.; Lin, R.; Li, N.; Cao, H.; et al. Iron oxide nanoparticles augment the intercellular mitochondrial transfer–mediated therapy. Sci. Adv. 2021, 7, eabj0534. [Google Scholar] [CrossRef]
  120. Reddy, M.; Fonseca, L.; Gowda, S.; Chougule, B.; Hari, A.; Totey, S. Human Adipose-derived Mesenchymal Stem Cells Attenuate Early Stage of Bleomycin Induced Pulmonary Fibrosis: Comparison with Pirfenidone. Int. J. Stem Cells 2016, 9, 192–206. [Google Scholar] [CrossRef]
  121. Mizuno, S.; Matsumoto, K.; Li, M.-Y.; Nakamura, T. HGF reduces advancing lung fibrosis in mice: A potential role for MMP-dependent myofibroblast apoptosis. FASEB J. 2005, 19, 1–18. [Google Scholar] [CrossRef] [Green Version]
  122. Liguori, T.T.A.; Liguori, G.R.; Moreira, L.F.P.; Harmsen, M.C. Fibroblast growth factor-2, but not the adipose tissue-derived stromal cells secretome, inhibits TGF-β1-induced differentiation of human cardiac fibroblasts into myofibroblasts. Sci. Rep. 2018, 8, 16633. [Google Scholar] [CrossRef] [PubMed]
  123. Ramos, C.; Montaño, M.; Becerril, C.; Cisneros-Lira, J.; Barrera, L.; Ruíz, V.; Pardo, A.; Selman, M. Acidic fibroblast growth factor decreases α-smooth muscle actin expression and induces apoptosis in human normal lung fibroblasts. Am. J. Physiol. Cell. Mol. Physiol. 2006, 291, L871–L879. [Google Scholar] [CrossRef] [PubMed]
  124. Ramasamy, S.K.; Mailleux, A.A.; Gupte, V.V.; Mata, F.; Sala, F.G.; Veltmaat, J.M.; Del Moral, P.M.; De Langhe, S.; Parsa, S.; Kelly, L.K.; et al. Fgf10 dosage is critical for the amplification of epithelial cell progenitors and for the formation of multiple mesenchymal lineages during lung development. Dev. Biol. 2007, 307, 237–247. [Google Scholar] [CrossRef] [PubMed]
  125. Wu, J.; Chu, X.; Chen, C.; Bellusci, S. Role of Fibroblast Growth Factor 10 in Mesenchymal Cell Differentiation during Lung Development and Disease. Front. Genet. 2018, 9, 545. [Google Scholar] [CrossRef]
  126. Chanda, D.; Kurundkar, A.; Rangarajan, S.; Locy, M.; Bernard, K.; Sharma, N.S.; Logsdon, N.J.; Liu, H.; Crossman, D.K.; Horowitz, J.C.; et al. Developmental Reprogramming in Mesenchymal Stromal Cells of Human Subjects with Idiopathic Pulmonary Fibrosis. Sci. Rep. 2016, 6, 37445. [Google Scholar] [CrossRef]
  127. Epperly, M.W.; Guo, H.; Gretton, J.E.; Greenberger, J.S. Bone Marrow Origin of Myofibroblasts in Irradiation Pulmonary Fibrosis. Am. J. Respir. Cell Mol. Biol. 2003, 29, 213–224. [Google Scholar] [CrossRef]
  128. Kramann, R.; Schneider, R.K.; DiRocco, D.P.; Machado, F.; Fleig, S.; Bondzie, P.A.; Henderson, J.M.; Ebert, B.L.; Humphreys, B.D. Perivascular Gli1+ Progenitors Are Key Contributors to Injury-Induced Organ Fibrosis. Cell Stem Cell 2014, 16, 51–66. [Google Scholar] [CrossRef]
  129. Penke, L.R.; Peters-Golden, M. Molecular determinants of mesenchymal cell activation in fibroproliferative diseases. Experientia 2019, 76, 4179–4201. [Google Scholar] [CrossRef]
  130. Zhao, X.; Wu, X.; Qian, M.; Song, Y.; Wu, D.; Zhang, W. Knockdown of TGF-β1 expression in human umbilical cord mesenchymal stem cells reverts their exosome-mediated EMT promoting effect on lung cancer cells. Cancer Lett. 2018, 428, 34–44. [Google Scholar] [CrossRef]
  131. Yan, X.; Liu, Y.; Han, Q.; Jia, M.; Liao, L.; Qi, M.; Zhao, R.C. Injured microenvironment directly guides the differentiation of engrafted Flk-1+ mesenchymal stem cell in lung. Exp. Hematol. 2007, 35, 1466–1475. [Google Scholar] [CrossRef] [PubMed]
  132. Cao, H.; Chen, X.; Hou, J.; Wang, C.; Xiang, Z.; Shen, Y.; Han, X. The Shh/Gli signaling cascade regulates myofibroblastic activation of lung-resident mesenchymal stem cells via the modulation of Wnt10a expression during pulmonary fibrogenesis. Lab. Investig. 2019, 100, 363–377. [Google Scholar] [CrossRef] [PubMed]
  133. Bustos, M.L.; Huleihel, L.; Kapetanaki, M.G.; Cardenas, C.L.; Mroz, L.; Ellis, B.M.; McVerry, B.J.; Richards, T.J.; Kaminski, N.; Cerdenes, N.; et al. Aging Mesenchymal Stem Cells Fail to Protect Because of Impaired Migration and Antiinflammatory Response. Am. J. Respir. Crit. Care Med. 2014, 189, 787–798. [Google Scholar] [CrossRef]
  134. Kinder, B.W.; Brown, K.K.; Schwarz, M.I.; Ix, J.H.; Kervitsky, A.; King, T.E. Baseline BAL Neutrophilia Predicts Early Mortality in Idiopathic Pulmonary Fibrosis. Chest 2008, 133, 226–232. [Google Scholar] [CrossRef]
  135. Zhang, H.-W.; Wang, Q.; Mei, H.-X.; Zheng, S.-X.; Ali, A.M.; Wu, Q.-X.; Ye, Y.; Xu, H.-R.; Xiang, S.-Y.; Jin, S.-W. RvD1 ameliorates LPS-induced acute lung injury via the suppression of neutrophil infiltration by reducing CXCL2 expression and release from resident alveolar macrophages. Int. Immunopharmacol. 2019, 76, 105877. [Google Scholar] [CrossRef] [PubMed]
  136. Sue, R.D.; Belperio, J.A.; Burdick, M.D.; Murray, L.A.; Xue, Y.Y.; Dy, M.C.; Kwon, J.J.; Keane, M.P.; Strieter, R.M. CXCR2 Is Critical to Hyperoxia-Induced Lung Injury. J. Immunol. 2004, 172, 3860–3868. [Google Scholar] [CrossRef] [PubMed]
  137. Ziegenhagen, M.W.; Zabel, P.; Zissel, G.; Schlaak, M.; Müller-Quernheim, J. Serum Level of Interleukin 8 Is Elevated in Idiopathic Pulmonary Fibrosis and Indicates Disease Activity. Am. J. Respir. Crit. Care Med. 1998, 157, 762–768. [Google Scholar] [CrossRef]
  138. Gregory, A.D.; Kliment, C.R.; Metz, H.E.; Kim, K.-H.; Kargl, J.; Agostini, B.A.; Crum, L.T.; Oczypok, E.A.; Oury, T.A.; Houghton, A.M. Neutrophil elastase promotes myofibroblast differentiation in lung fibrosis. J. Leukoc. Biol. 2015, 98, 143–152. [Google Scholar] [CrossRef]
  139. Zuo, F.; Kaminski, N.; Eugui, E.; Allard, J.; Yakhini, Z.; Ben-Dor, A.; Lollini, L.; Morris, D.; Kim, Y.; DeLustro, B.; et al. Gene expression analysis reveals matrilysin as a key regulator of pulmonary fibrosis in mice and humans. Proc. Natl. Acad. Sci. USA 2002, 99, 6292–6297. [Google Scholar] [CrossRef]
  140. Nuovo, G.J.; Hagood, J.S.; Magro, C.M.; Chin, N.; Kapil, R.; Davis, L.; Marsh, C.B.; Folcik, V.A. The distribution of immunomodulatory cells in the lungs of patients with idiopathic pulmonary fibrosis. Mod. Pathol. 2011, 25, 416–433. [Google Scholar] [CrossRef]
  141. Marchal-Sommé, J.; Uzunhan, Y.; Marchand-Adam, S.; Valeyre, D.; Soumelis, V.; Crestani, B.; Soler, P. Cutting Edge: Nonproliferating Mature Immune Cells Form a Novel Type of Organized Lymphoid Structure in Idiopathic Pulmonary Fibrosis. J. Immunol. 2006, 176, 5735–5739. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  142. Xue, J.; Kass, D.J.; Bon, J.; Vuga, L.; Tan, J.; Csizmadia, E.; Otterbein, L.; Soejima, M.; Levesque, M.C.; Gibson, K.F.; et al. Plasma B Lymphocyte Stimulator and B Cell Differentiation in Idiopathic Pulmonary Fibrosis Patients. J. Immunol. 2013, 191, 2089–2095. [Google Scholar] [CrossRef] [PubMed]
  143. Mayadas, T.N.; Tsokos, G.C.; Tsuboi, N. Mechanisms of Immune Complex–Mediated Neutrophil Recruitment and Tissue Injury. Circulation 2009, 120, 2012–2024. [Google Scholar] [CrossRef] [PubMed]
  144. Dobashi, N.; Fujita, J.; Murota, M.; Ohtsuki, Y.; Yamadori, I.; Yoshinouchi, T.; Ueda, R.; Bandoh, S.; Kamei, T.; Nishioka, M.; et al. Elevation of Anti-Cytokeratin 18 Antibody and Circulating Cytokeratin 18: Anti-Cytokeratin 18 Antibody Immune Complexes in Sera of Patients with Idiopathic Pulmonary Fibrosis. Lung 2000, 178, 171–179. [Google Scholar] [CrossRef] [PubMed]
  145. Dall’Aglio, P.; Pesci, A.; Bertorelli, G.; Brianti, E.; Scarpa, S. Study of Immune Complexes in Bronchoalveolar Lavage Fluids. Respiration 1988, 54, 36–41. [Google Scholar] [CrossRef]
  146. Vuga, L.J.; Tedrow, J.R.; Pandit, K.V.; Tan, J.; Kass, D.J.; Xue, J.; Chandra, D.; Leader, J.K.; Gibson, K.F.; Kaminski, N.; et al. C-X-C Motif Chemokine 13 (CXCL13) Is a Prognostic Biomarker of Idiopathic Pulmonary Fibrosis. Am. J. Respir. Crit. Care Med. 2014, 189, 966–974. [Google Scholar] [CrossRef]
  147. Schupp, J.; Binder, H.; Jäger, B.; Cillis, G.; Zissel, G.; Müller-Quernheim, J.; Prasse, A. Macrophage Activation in Acute Exacerbation of Idiopathic Pulmonary Fibrosis. PLoS ONE 2015, 10, e0116775. [Google Scholar] [CrossRef]
  148. Butler, M.W.; Hackett, N.R.; Salit, J.; Strulovici-Barel, Y.; Omberg, L.; Mezey, J.; Crystal, R.G. Glutathione S-transferase copy number variation alters lung gene expression. Eur. Respir. J. 2011, 38, 15–28. [Google Scholar] [CrossRef]
  149. Nie, Y.; Sun, L.; Wu, Y.; Yang, Y.; Wang, J.; He, H.; Hu, Y.; Chang, Y.; Liang, Q.; Zhu, J.; et al. AKT2 Regulates Pulmonary Inflammation and Fibrosis via Modulating Macrophage Activation. J. Immunol. 2017, 198, 4470–4480. [Google Scholar] [CrossRef]
  150. Baran, C.P.; Opalek, J.M.; McMaken, S.; Newland, C.A.; O’Brien, J.M.; Hunter, M.G.; Bringardner, B.D.; Monick, M.M.; Brigstock, D.R.; Stromberg, P.C.; et al. Important Roles for Macrophage Colony-stimulating Factor, CC Chemokine Ligand 2, and Mononuclear Phagocytes in the Pathogenesis of Pulmonary Fibrosis. Am. J. Respir. Crit. Care Med. 2007, 176, 78–89. [Google Scholar] [CrossRef]
  151. Cao, Z.; Lis, R.; Ginsberg, M.; Chavez, D.; Shido, K.; Rabbany, S.Y.; Fong, G.-H.; Sakmar, T.; Rafii, S.; Ding, B.-S. Targeting of the pulmonary capillary vascular niche promotes lung alveolar repair and ameliorates fibrosis. Nat. Med. 2016, 22, 154–162. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. He, C.; Murthy, S.; McCormick, M.L.; Spitz, D.; Ryan, A.J.; Carter, A.B. Mitochondrial Cu, Zn-Superoxide Dismutase Mediates Pulmonary Fibrosis by Augmenting H2O2 Generation. J. Biol. Chem. 2011, 286, 15597–15607. [Google Scholar] [CrossRef] [PubMed]
  153. Jain, M.; Rivera, S.; Monclus, E.A.; Synenki, L.; Zirk, A.; Eisenbart, J.; Feghali-Bostwick, C.; Mutlu, G.M.; Budinger, G.R.S.; Chandel, N.S. Mitochondrial Reactive Oxygen Species Regulate Transforming Growth Factor-β Signaling. J. Biol. Chem. 2013, 288, 770–777. [Google Scholar] [CrossRef] [PubMed]
  154. Larson-Casey, J.L.; Deshane, J.S.; Ryan, A.J.; Thannickal, V.J.; Carter, A.B. Macrophage Akt1 Kinase-Mediated Mitophagy Modulates Apoptosis Resistance and Pulmonary Fibrosis. Immunity 2016, 44, 582–596. [Google Scholar] [CrossRef]
  155. Scott, M.K.D.; Quinn, K.; Li, Q.; Carroll, R.; Warsinske, H.; Vallania, F.; Chen, S.; Carns, M.A.; Aren, K.; Sun, J.; et al. Increased monocyte count as a cellular biomarker for poor outcomes in fibrotic diseases: A retrospective, multicentre cohort study. Lancet Respir. Med. 2019, 7, 497–508. [Google Scholar] [CrossRef]
  156. Misharin, A.V.; Morales-Nebreda, L.; Reyfman, P.A.; Cuda, C.M.; Walter, J.M.; McQuattie-Pimentel, A.C.; Chen, C.-I.; Anekalla, K.R.; Joshi, N.; Williams, K.J.N.; et al. Monocyte-derived alveolar macrophages drive lung fibrosis and persist in the lung over the life span. J. Exp. Med. 2017, 214, 2387–2404. [Google Scholar] [CrossRef]
  157. Iyonaga, K.; Takeya, M.; Saita, N.; Sakamoto, O.; Yoshimura, T.; Ando, M.; Takahashi, K. Monocyte chemoattractant protein-1 in idiopathic pulmonary fibrosis and other interstitial lung diseases. Hum. Pathol. 1994, 25, 455–463. [Google Scholar] [CrossRef]
  158. Suga, M.; Iyonaga, K.; Ichiyasu, H.; Saita, N.; Yamasaki, H.; Ando, M. Clinical significance of MCP-1 levels in BALF and serum in patients with interstitial lung diseases. Eur. Respir. J. 1999, 14, 370–375. [Google Scholar] [CrossRef]
  159. Marchal-Sommé, J.; Uzunhan, Y.; Marchand-Adam, S.; Kambouchner, M.; Valeyre, M.; Crestani, B.; Soler, P. Dendritic Cells Accumulate in Human Fibrotic Interstitial Lung Disease. Am. J. Respir. Crit. Care Med. 2007, 176, 1007–1014. [Google Scholar] [CrossRef]
  160. Chakraborty, K.; Chatterjee, S.; Bhattacharyya, A. Modulation of CD11c+ lung dendritic cells in respect to TGF-β in experimental pulmonary fibrosis. Cell Biol. Int. 2017, 41, 991–1000. [Google Scholar] [CrossRef]
  161. Cha, S.-I.; Chang, C.S.; Kim, E.K.; Lee, J.W.; Matthay, M.A.; Golden, J.A.; Elicker, B.M.; Jones, K.; Collard, H.R.; Wolters, P.J. Lung mast cell density defines a subpopulation of patients with idiopathic pulmonary fibrosis. Histopathology 2012, 61, 98–106. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  162. Andersson, C.K.; Andersson-Sjöland, A.; Mori, M.; Hallgren, O.; Pardo, A.; Eriksson, L.; Bjermer, L.; Löfdahl, C.-G.; Selman, M.; Westergren-Thorsson, G.; et al. Activated MCTC mast cells infiltrate diseased lung areas in cystic fibrosis and idiopathic pulmonary fibrosis. Respir. Res. 2011, 12, 139. [Google Scholar] [CrossRef] [PubMed]
  163. Wygrecka, M.; Dahal, B.K.; Kosanovic, D.; Petersen, F.; Taborski, B.; von Gerlach, S.; Didiasova, M.; Zakrzewicz, D.; Preissner, K.T.; Schermuly, R.; et al. Mast Cells and Fibroblasts Work in Concert to Aggravate Pulmonary Fibrosis. Am. J. Pathol. 2013, 182, 2094–2108. [Google Scholar] [CrossRef] [PubMed]
  164. Shimbori, C.; Upagupta, C.; Bellaye, P.-S.; Ayaub, E.A.; Sato, S.; Yanagihara, T.; Zhou, Q.; Ognjanovic, A.; Ask, K.; Gauldie, J.; et al. Mechanical stress-induced mast cell degranulation activates TGF-β1 signalling pathway in pulmonary fibrosis. Thorax 2019, 74, 455–465. [Google Scholar] [CrossRef]
  165. Overed-Sayer, C.; Miranda, E.; Dunmore, R.; Marin, E.L.; Beloki, L.; Rassl, D.; Parfrey, H.; Carruthers, A.; Chahboub, A.; Koch, S.; et al. Inhibition of mast cells: A novel mechanism by which nintedanib may elicit anti-fibrotic effects. Thorax 2020, 75, 754–763. [Google Scholar] [CrossRef]
  166. Fireman, E.; Vardinon, N.; Burke, M.; Spizer, S.; Levin, S.; Endler, A.; Stav, D.; Topilsky, M.; Mann, A.; Schwarz, Y.; et al. Predictive value of response to treatment of T-lymphocyte subpopulations in idiopathic pulmonary fibrosis. Eur. Respir. J. 1998, 11, 706–711. [Google Scholar]
  167. Gilani, S.R.; Vuga, L.J.; Lindell, K.O.; Gibson, K.F.; Xue, J.; Kaminski, N.; Valentine, V.G.; Lindsay, E.; George, M.P.; Steele, C.; et al. CD28 Down-Regulation on Circulating CD4 T-Cells Is Associated with Poor Prognoses of Patients with Idiopathic Pulmonary Fibrosis. PLoS ONE 2010, 5, e8959. [Google Scholar] [CrossRef]
  168. Chen, J.H.; Schulman, H.; Gardner, P. A cAMP-Regulated Chloride Channel in Lymphocytes That Is Affected in Cystic Fibrosis. Science 1989, 243, 657–660. [Google Scholar] [CrossRef]
  169. Hao, Z.; Hampel, B.; Yagita, H.; Rajewsky, K. T Cell–specific Ablation of Fas Leads to Fas Ligand–mediated Lymphocyte Depletion and Inflammatory Pulmonary Fibrosis. J. Exp. Med. 2004, 199, 1355–1365. [Google Scholar] [CrossRef]
  170. McKinney, E.F.; Lee, J.C.; Jayne, D.R.W.; Lyons, P.A.; Smith, K.G. T-cell exhaustion, co-stimulation and clinical outcome in autoimmunity and infection. Nature 2015, 523, 612–616. [Google Scholar] [CrossRef]
  171. Prior, C.; Haslam, P.L. In Vivo levels and in vitro production of interferon-gamma in fibrosing interstitial lung diseases. Clin. Exp. Immunol. 1992, 88, 280–287. [Google Scholar] [CrossRef] [PubMed]
  172. Saito, A.; Okazaki, H.; Sugawara, I.; Yamamoto, K.; Takizawa, H. Potential Action of IL-4 and IL-13 as Fibrogenic Factors on Lung Fibroblasts in vitro. Int. Arch. Allergy Immunol. 2003, 132, 168–176. [Google Scholar] [CrossRef] [PubMed]
  173. Smolen, J.S.; Kay, J.; Doyle, M.K.; Landewé, R.; Matteson, E.L.; Wollenhaupt, J.; Gaylis, N.; Murphy, F.T.; Neal, J.S.; Zhou, Y.; et al. Golimumab in patients with active rheumatoid arthritis after treatment with tumour necrosis factor α inhibitors (GO-AFTER study): A multicentre, randomised, double-blind, placebo-controlled, phase III trial. Lancet 2009, 374, 210–221. [Google Scholar] [CrossRef]
  174. Lei, L.; Zhao, C.; Qin, F.; He, Z.-Y.; Wang, X.; Zhong, X.-N. Th17 cells and IL-17 promote the skin and lung inflammation and fibrosis process in a bleomycin-induced murine model of systemic sclerosis. Clin. Exp. Rheumatol. 2016, 34 (Suppl. 100), 14–22. [Google Scholar]
  175. Kaplan, M.H.; Hufford, M.M.; Olson, M.R. The development and in vivo function of T helper 9 cells. Nat. Rev. Immunol. 2015, 15, 295–307. [Google Scholar] [CrossRef]
  176. Deng, K.M.; Yang, X.S.; Luo, Q.; She, Y.X.; Yu, Q.Y.; Tang, X.X. Deleterious Role of Th9 Cells in Pulmonary Fibrosis. Cells 2021, 10, 3209. [Google Scholar] [CrossRef]
  177. Fu, X.-Y.; Zhang, D.-W.; Li, Y.-D.; Jian-Zhao, N.; Tang, Y.-Q.; Niu, J.-Z.; Li, Y. Curcumin Treatment Suppresses CCR7 Expression and the Differentiation and Migration of Human Circulating Fibrocytes. Cell. Physiol. Biochem. 2015, 35, 489–498. [Google Scholar] [CrossRef]
  178. Re, S.L.; Lecocq, M.; Uwambayinema, F.; Yakoub, Y.; Delos, M.; Demoulin, J.-B.; Lucas, S.; Sparwasser, T.; Renauld, J.-C.; Lison, D.; et al. Platelet-Derived Growth Factor–Producing CD4+ Foxp3+ Regulatory T Lymphocytes Promote Lung Fibrosis. Am. J. Respir. Crit. Care Med. 2011, 184, 1270–1281. [Google Scholar] [CrossRef]
  179. Kotsianidis, I.; Nakou, E.; Bouchliou, I.; Tzouvelekis, A.; Spanoudakis, E.; Steiropoulos, P.; Sotiriou, I.; Aidinis, V.; Margaritis, D.; Tsatalas, C.; et al. Global Impairment of CD4+CD25+FOXP3+ Regulatory T Cells in Idiopathic Pulmonary Fibrosis. Am. J. Respir. Crit. Care Med. 2009, 179, 1121–1130. [Google Scholar] [CrossRef]
  180. Boveda-Ruiz, D.; D’Alessandro-Gabazza, C.N.; Toda, M.; Takagi, T.; Naito, M.; Matsushima, Y.; Matsumoto, T.; Kobayashi, T.; Gil-Bernabe, P.; Chelakkot-Govindalayathil, A.-L.; et al. Differential role of regulatory T cells in early and late stages of pulmonary fibrosis. Immunobiology 2012, 218, 245–254. [Google Scholar] [CrossRef]
  181. Zhu, Y.-G.; Feng, X.-M.; Abbott, J.; Fang, X.-H.; Hao, Q.; Monsel, A.; Qu, J.-M.; Matthay, M.A.; Lee, J.W. Human Mesenchymal Stem Cell Microvesicles for Treatment of Escherichia coli Endotoxin-Induced Acute Lung Injury in Mice. Stem Cells 2013, 32, 116–125. [Google Scholar] [CrossRef] [PubMed]
  182. Monsel, A.; Zhu, Y.-G.; Gennai, S.; Hao, Q.; Hu, S.; Rouby, J.-J.; Rosenzwajg, M.; Matthay, M.A.; Lee, J.W. Therapeutic Effects of Human Mesenchymal Stem Cell–derived Microvesicles in Severe Pneumonia in Mice. Am. J. Respir. Crit. Care Med. 2015, 192, 324–336. [Google Scholar] [CrossRef] [PubMed]
  183. Mansouri, N.; Willis, G.R.; Fernandez-Gonzalez, A.; Reis, M.; Nassiri, S.; Mitsialis, S.A.; Kourembanas, S. Mesenchymal stromal cell exosomes prevent and revert experimental pulmonary fibrosis through modulation of monocyte phenotypes. JCI Insight 2019, 4, e128060. [Google Scholar] [CrossRef] [Green Version]
  184. Xu, C.; Zhao, J.; Li, Q.; Hou, L.; Wang, Y.; Li, S.; Jiang, F.; Zhu, Z.; Tian, L. Exosomes derived from three-dimensional cultured human umbilical cord mesenchymal stem cells ameliorate pulmonary fibrosis in a mouse silicosis model. Stem Cell Res. Ther. 2020, 11, 503. [Google Scholar] [CrossRef] [PubMed]
  185. Lei, M.X.; He, M.N.; Zhu, L.; Zhou, M.M.; Zhang, M.K.; Wang, M.C.; Huang, M.H.; Chen, S.; Li, Y.; Liu, Q.; et al. Mesenchymal Stem Cell-Derived Extracellular Vesicles Attenuate Radiation-Induced Lung Injury via miRNA-214-3p. Antioxid. Redox Signal. 2021, 35, 849–862. [Google Scholar] [CrossRef]
  186. Zhou, J.; Lin, Y.; Kang, X.; Liu, Z.; Zhang, W.; Xu, F. microRNA-186 in extracellular vesicles from bone marrow mesenchymal stem cells alleviates idiopathic pulmonary fibrosis via interaction with SOX4 and DKK1. Stem Cell Res. Ther. 2021, 12, 96. [Google Scholar] [CrossRef] [PubMed]
  187. Li, J.W.; Wei, L.; Han, Z.; Chen, Z. Mesenchymal stromal cells-derived exosomes alleviate ischemia/reperfusion injury in mouse lung by transporting anti-apoptotic miR-21-5p. Eur. J. Pharmacol. 2019, 852, 68–76. [Google Scholar] [CrossRef]
  188. Lee, C.; Mitsialis, S.A.; Aslam, M.; Vitali, S.H.; Vergadi, E.; Konstantinou, G.; Sdrimas, K.; Fernandez-Gonzalez, A.; Kourembanas, S. Exosomes Mediate the Cytoprotective Action of Mesenchymal Stromal Cells on Hypoxia-Induced Pulmonary Hypertension. Circulation 2012, 126, 2601–2611. [Google Scholar] [CrossRef]
  189. Wan, X.; Chen, S.; Fang, Y.; Zuo, W.; Cui, J.; Xie, S. Mesenchymal stem cell-derived extracellular vesicles suppress the fibroblast proliferation by downregulating FZD6 expression in fibroblasts via micrRNA-29b-3p in idiopathic pulmonary fibrosis. J. Cell. Physiol. 2020, 235, 8613–8625. [Google Scholar] [CrossRef]
  190. Huang, R.; Qin, C.; Wang, J.; Hu, Y.; Zheng, G.; Qiu, G.; Ge, M.; Tao, H.; Shu, Q.; Xu, J. Differential effects of extracellular vesicles from aging and young mesenchymal stem cells in acute lung injury. Aging 2019, 11, 7996–8014. [Google Scholar] [CrossRef]
  191. Schamberger, A.C.; Schiller, H.B.; Fernandez, I.E.; Sterclova, M.; Heinzelmann, K.; Hennen, E.; Hatz, R.; Behr, J.; Vašáková, M.; Mann, M.; et al. Glutathione peroxidase 3 localizes to the epithelial lining fluid and the extracellular matrix in interstitial lung disease. Sci. Rep. 2016, 6, 29952. [Google Scholar] [CrossRef] [PubMed]
  192. Sun, L.; Zhu, M.; Feng, W.; Lin, Y.; Yin, J.; Jin, J.; Wang, Y. Exosomal miRNA Let-7 from Menstrual Blood-Derived Endometrial Stem Cells Alleviates Pulmonary Fibrosis through Regulating Mitochondrial DNA Damage. Oxidative Med. Cell. Longev. 2019, 2019, 4506303. [Google Scholar] [CrossRef] [PubMed]
  193. Dong, L.; Wang, Y.; Zheng, T.; Pu, Y.; Ma, Y.; Qi, X.; Zhang, W.; Xue, F.; Shan, Z.; Liu, J.; et al. Hypoxic hUCMSC-derived extracellular vesicles attenuate allergic airway inflammation and airway remodeling in chronic asthma mice. Stem Cell Res. Ther. 2021, 12, 4. [Google Scholar] [CrossRef] [PubMed]
  194. Ridzuan, N.; Zakaria, N.; Widera, D.; Sheard, J.; Morimoto, M.; Kiyokawa, H.; Isa, S.A.M.; Singh, G.K.C.; Then, K.-Y.; Ooi, G.-C.; et al. Human umbilical cord mesenchymal stem cell-derived extracellular vesicles ameliorate airway inflammation in a rat model of chronic obstructive pulmonary disease (COPD). Stem Cell Res. Ther. 2021, 12, 54. [Google Scholar] [CrossRef] [PubMed]
  195. Wang, J.; Huang, R.; Xu, Q.; Zheng, G.; Qiu, G.; Ge, M.; Shu, Q.; Xu, J. Mesenchymal Stem Cell–Derived Extracellular Vesicles Alleviate Acute Lung Injury via Transfer of miR-27a-3p. Crit. Care Med. 2020. [Google Scholar] [CrossRef]
  196. Xiao, K.; He, W.; Guan, W.; Hou, F.; Yan, P.; Xu, J.; Zhou, T.; Liu, Y.; Xie, L. Mesenchymal stem cells reverse EMT process through blocking the activation of NF-κB and Hedgehog pathways in LPS-induced acute lung injury. Cell Death Dis. 2020, 11, 863. [Google Scholar] [CrossRef]
  197. Glassberg, M.K.; Minkiewicz, J.; Toonkel, R.L.; Simonet, E.S.; Rubio, G.A.; DiFede, D.; Shafazand, S.; Khan, A.; Pujol, M.V.; LaRussa, V.F.; et al. Allogeneic Human Mesenchymal Stem Cells in Patients With Idiopathic Pulmonary Fibrosis via Intravenous Delivery (AETHER)—A Phase I Safety Clinical Trial. Chest 2017, 151, 971–981. [Google Scholar] [CrossRef]
  198. Zhang, C.; Yin, X.; Zhang, J.; Ao, Q.; Gu, Y.; Liu, Y. Clinical observation of umbilical cord mesenchymal stem cell treatment of severe idiopathic pulmonary fibrosis: A case report. Exp. Ther. Med. 2017, 13, 1922–1926. [Google Scholar] [CrossRef]
  199. Tzouvelekis, A.; Paspaliaris, V.; Koliakos, G.; Ntolios, P.; Bouros, E.; Oikonomou, A.; Zissimopoulos, A.; Boussios, N.; Dardzinski, B.; Gritzalis, D.; et al. A prospective, non-randomized, no placebo-controlled, phase Ib clinical trial to study the safety of the adipose derived stromal cells-stromal vascular fraction in idiopathic pulmonary fibrosis. J. Transl. Med. 2013, 11, 171. [Google Scholar] [CrossRef]
  200. Campo, A.; González-Ruiz, J.M.; Andreu, E.; Alcaide, A.B.; Ocón, M.M.; De-Torres, J.; Pueyo, J.; Cordovilla, R.; Villaron, E.; Sanchez-Guijo, F.; et al. Endobronchial autologous bone marrow-mesenchymal stromal cells in idiopathic pulmonary fibrosis (phase I). ERJ Open Res. 2021, 7, 00773–02020. [Google Scholar] [CrossRef]
  201. Fishman, J.E.; Kim, G.-H.J.; Kyeong, N.-Y.; Goldin, J.G.; Glassberg, M.K. Intravenous stem cell dose and changes in quantitative lung fibrosis and DLCO in the AETHER trial: A pilot study. Eur. Rev. Med. Pharmacol. Sci. 2019, 23, 7568–7572. [Google Scholar] [PubMed]
  202. Chambers, D.C.; Enever, D.; Ilic, N.; Sparks, L.; Whitelaw, K.; Ayres, J.; Yerkovich, S.; Khalil, D.; Atkinson, K.M.; Hopkins, P.M. A phase 1b study of placenta-derived mesenchymal stromal cells in patients with idiopathic pulmonary fibrosis. Respirology 2014, 19, 1013–1018. [Google Scholar] [CrossRef]
  203. Young, H.E.; Speight, M.O. Telomerase-Positive Stem Cells as a Potential Treatment for Idiopathic Pulmonary Fibrosis. Stem Cells Regen. Med. 2020, 4, 1–11. [Google Scholar]
  204. Saleh, M.; Vaezi, A.A.; Aliannejad, R.; Sohrabpour, A.A.; Kiaei, S.Z.F.; Shadnoush, M.; Siavashi, V.; Aghaghazvini, L.; Khoundabi, B.; Abdoli, S.; et al. Cell therapy in patients with COVID-19 using Wharton’s jelly mesenchymal stem cells: A phase 1 clinical trial. Stem Cell Res. Ther. 2021, 12, 410. [Google Scholar] [CrossRef]
  205. Sengupta, V.; Sengupta, S.; Lazo, A.; Woods, P.; Nolan, A.; Bremer, N. Exosomes Derived from Bone Marrow Mesenchymal Stem Cells as Treatment for Severe COVID-19. Stem Cells Dev. 2020, 29, 747–754. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Pathways of myofibroblast formation during IPF. The figure shows the different cell types that respond to stimulation by profibrotic growth factors (TGF-β, CTGF), cytokines (IL-1, IL-13, IL-4, IL-6, TNF-α), signaling pathways (Wnt/β-catenin, Notch1/PDGFRβ/ROCK1), chemokines (CXCL12, SLC, CCL1), specific EMT transcription factors (Snail2, ZEB1, TWIST, p62/SQSTM1), adhesion molecules (ICAM-1,VCAM-1, FAK and CDH-11), hyperoxia, exposure to endothelin-1 and nicotine, overexpression of α-SMA, COL1α1 and fibronectin, upregulation of SMAD2, SMAD3, Sp1 and c-Myc differentiated into myofibroblasts [65,70,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102] (Created with BioRender.com, accessed on 4 July 2022).
Figure 1. Pathways of myofibroblast formation during IPF. The figure shows the different cell types that respond to stimulation by profibrotic growth factors (TGF-β, CTGF), cytokines (IL-1, IL-13, IL-4, IL-6, TNF-α), signaling pathways (Wnt/β-catenin, Notch1/PDGFRβ/ROCK1), chemokines (CXCL12, SLC, CCL1), specific EMT transcription factors (Snail2, ZEB1, TWIST, p62/SQSTM1), adhesion molecules (ICAM-1,VCAM-1, FAK and CDH-11), hyperoxia, exposure to endothelin-1 and nicotine, overexpression of α-SMA, COL1α1 and fibronectin, upregulation of SMAD2, SMAD3, Sp1 and c-Myc differentiated into myofibroblasts [65,70,76,77,78,79,80,81,82,83,84,85,86,87,88,89,90,91,92,93,94,95,96,97,98,99,100,101,102] (Created with BioRender.com, accessed on 4 July 2022).
Ijms 23 11212 g001
Figure 2. Schematic representation of the lung tissue damage therapy mechanism in IPF by means of mesenchymal stem cell-derived extracellular vesicles. The footnotes in the figure indicate: the composition of EVs-MSCs—proteins (PRKCZ, TIMP-1, TIMP-2, BMP-7), transcription factors (NF-κβ p50, NF-κβ p65), miRNAs (miR-182-5p, miR-214-3p, miR-29b-3p, miR-23a-3p) and therapeutic effect of EVs-MSCs (Created with BioRender.com, accessed on 22 September 2022).
Figure 2. Schematic representation of the lung tissue damage therapy mechanism in IPF by means of mesenchymal stem cell-derived extracellular vesicles. The footnotes in the figure indicate: the composition of EVs-MSCs—proteins (PRKCZ, TIMP-1, TIMP-2, BMP-7), transcription factors (NF-κβ p50, NF-κβ p65), miRNAs (miR-182-5p, miR-214-3p, miR-29b-3p, miR-23a-3p) and therapeutic effect of EVs-MSCs (Created with BioRender.com, accessed on 22 September 2022).
Ijms 23 11212 g002
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kletukhina, S.; Mutallapova, G.; Titova, A.; Gomzikova, M. Role of Mesenchymal Stem Cells and Extracellular Vesicles in Idiopathic Pulmonary Fibrosis. Int. J. Mol. Sci. 2022, 23, 11212. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms231911212

AMA Style

Kletukhina S, Mutallapova G, Titova A, Gomzikova M. Role of Mesenchymal Stem Cells and Extracellular Vesicles in Idiopathic Pulmonary Fibrosis. International Journal of Molecular Sciences. 2022; 23(19):11212. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms231911212

Chicago/Turabian Style

Kletukhina, Sevindzh, Guzel Mutallapova, Angelina Titova, and Marina Gomzikova. 2022. "Role of Mesenchymal Stem Cells and Extracellular Vesicles in Idiopathic Pulmonary Fibrosis" International Journal of Molecular Sciences 23, no. 19: 11212. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms231911212

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

Article Metrics

Back to TopTop