Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Apr 1.
Published in final edited form as: Exp Gerontol. 2020 Jan 17;132:110836. doi: 10.1016/j.exger.2020.110836

Cell Senescence and Fibrotic Lung Diseases

Rui-Ming Liu 1, Gang Liu 1
PMCID: PMC7036279  NIHMSID: NIHMS1553991  PMID: 31958492

Abstract

Idiopathic pulmonary fibrosis (IPF) is a progressive fatal lung disorder with an unknown etiology and very limited therapeutic options. The incidence and severity of IPF increase with advanced age, suggesting that aging is a major risk factor for IPF. The mechanism underlying the aging-related susceptibility to IPF, however, remains unclear. Cellular senescence, a permanent arrest of cell growth, has been increasingly recognized as an important contributor to aging and aging-related diseases, including IPF. Senescent cells have been identified in IPF lungs and in experimental lung fibrosis models. Removal of senescent cells pharmacologically or genetically improves lung function and reverses pulmonary fibrosis induced by different stimuli in experimental fibrosis models. Treatment with senolytic drugs also improves clinical symptoms in IPF patients. These intriguing findings suggest that cellular senescence contributes importantly to the pathogenesis of fibrotic lung diseases and targeting senescent cells may represent a novel approach for the treatment of fibrotic lung disorders. In this mini review, we summarize the recent advance in the field regarding the role of cellular senescence in fibrotic lung diseases, with a focus on IPF.

Keywords: aging, cell senescence, lung fibrosis

Cellular Senescence

Cellular senescence, including replicative senescence (RS) and stress-induced premature senescence (SIPS), is a permanent arrest of cell growth. Cellular senescence, although linked to aging, can occur at any life stage from embryo to adulthood1, 2. Cellular senescence is a highly regulated process and has both beneficial (fetal development and tumor suppression) and detrimental (premature aging-related diseases) effects, when this process is perturbed2. Senescent cells are characterized by macromolecule damage, irreversible cell cycle arrest, altered metabolism and cell shape, and secretion of various pro-inflammatory cytokines, chemokines, growth factors, a feature best known as senescence associated secretory phenotype (SASP)1, 3, 4. It has been well documented that the number of senescent cells increases with age and in aging-related diseases. The mechanisms responsible for cellular senescence under either physiological or pathological conditions remain poorly understood, although various hypotheses, including telomere shortening, oncogene activation, oxidative stress, DNA damage, and mitochondrial dysfunction have been put forward (Fig 1).

Fig 1. Schematic flow chart of cause and consequence of cell senescence.

Fig 1.

SASP, senescence associated secretory phenotype.

It has been long recognized that cellular senescence contributes importantly to the aging-related decline in tissue regeneration capacity and the pathogenesis of aging-related diseases, including IPF38. The mechanism whereby senescent cells contribute to aging and aging-related diseases, however, remains unclear. Progenitor cells or stem cells are particularly sensitive to senescence stresses. Therefore, one of the hypotheses is that cellular senescence leads to exhaustion of progenitor cells or stem cells, which, in turn, causes a decline in tissue regenerative capacity during aging or upon injury. Another potential mechanism whereby senescent cells contribute to aging and aging-related diseases is through secretion of pro-inflammatory cytokines/chemokines/growth factors (SASP). These biologically active molecules can exert profound effects on themselves (autocrine function) or on neighboring cells (paracrine functions). It has been reported that the SASP can spread senescence signals to surrounding cells, contribute to persistent inflammation, and induce profibrotic phenotypic changes of fibroblasts or macrophages912. The third potential mechanism whereby cell senescence contribute to the development of lung fibrosis is protecting myofibroblasts, the major producer of extracellular matrix proteins, from apoptosis (Fig 1). Despite an incomplete understanding of the mechanism by which senescent cells contribute to aging and aging-related diseases, therapeutic approaches targeting senescent cells for the improvement of healthy aging and for the treatment of aging-related diseases have been growing rapidly1318. Further elucidation of the mechanisms underlying cell senescence and the mechanisms whereby senescent cells contribute to the pathogenesis of the diseases may lead to the discovery of novel therapeutics for the treatment of these aging-related diseases.

Idiopathic Pulmonary Fibrosis

Idiopathic pulmonary fibrosis is a progressive fatal lung disorder with unknown etiology. The disease is characterized by an excessive deposition of extracellular matrix proteins in the lung interstitium, leading to the destruction of lung structure and function. The median survival of IPF is 2–3 years after diagnosis and the mortality rate of IPF surpasses many types of cancers. There is a very limited therapeutic option for this devastating disease due to an incomplete understanding of its etiology and pathogenesis. Several risk factors have been postulated, including gene mutations, cigarette smoking, occupational exposure, and infectious agents; but the strongest link with IPF clinical disease expression/progression is aging1923. Therefore, IPF is considered a disease of aging6, 21, 22, 24. Despite intensive studies, the mechanism underlying the aging-related susceptibility to IPF remains unclear. A current disease paradigm is that lung fibrosis develops as a result of unremitting insults plus genetic and aging-related risk factors, leading to alveolar epithelial cell injuries. Injured epithelial cells then secrete an array of growth factors, chemokines/cytokines, and proteases, which promote the activation of myofibroblasts and replacement of injured alveolar epithelium with fibrotic tissue, due to a decreased reparative capacity of the alveolar epithelium (senescence). Therefore, elucidation of the mechanism underlying cell senescence in lung cells, especially epithelial cells, may be a key for our understanding of the disease pathogenesis and for the development of effective treatments for this devastating disease.

Cell Senescence in IPF

Emerging evidence suggests that cellular senescence contributes importantly to the pathophysiology of IPF. Various types of cells, including alveolar epithelial type II cells, fibroblasts, and endothelial cells, have been shown to undergo senescence in IPF lungs14, 2535. Senescent cells have also been found in experimental lung fibrosis models induced by different stimuli12, 3541. Most importantly, studies have shown that elimination of senescent cells pharmacologically or genetically attenuates lung fibrosis and restores the lung function in mice with experimental lung fibrosis13, 14, 30. A pilot clinical study further showed that senotherapy significantly and clinically-meaningfully improved the physical function evaluated as 6-min walk distance, 4-m gait speed, and chair-stands time in IPF patients18. Together, the data suggest that cellular senescence may underlie the pathophysiology of IPF and targeting senescent cells may be effective for the treatment of IPF.

Clinical findings:

Different types of cells, including alveolar epithelial type II cells, fibroblasts, and endothelial cells, have been shown to undergo senescence in IPF lung14, 2535. ATII cells can self-renew and can also differentiate into type I alveolar epithelial cells and therefore are considered as alveolar progenitor cells42, 43. There is overwhelming evidence showing that alveolar epithelial cells in IPF lung undergo senescence 5, 14, 25, 28, 30, 35. Using immunostaining as well as x-gal staining techniques, several investigators have shown that epithelial cells in IPF lung express higher levels of cell cycle repressors p21, p16, and/or p53 and have higher activity of senescence associated beta-galactosidase (SA-β-gal), as compared to ATII cells in control lungs14, 25, 28, 30, 35. Using single cell RNA sequence techniques, Reyfman et al. identified rare cell populations, including airway stem cells and senescent epithelial cells, in IPF lungs31. These senescent epithelial cells secrete various cytokines and chemokines related to SASP33. Importantly, many alveolar epithelial cells in IPF lungs are also positive for TUNEL staining, a classical method to detect apoptotic cells, indicating that senescent epithelial cells may be prone to apoptosis25, 44.

Fibroblasts are the primary producers of extracellular matrix proteins and play a central role in the development of lung fibrosis. Several studies have shown that fibroblasts in IPF lungs exhibit senescent phenotype, having an increased level of DNA damage, expressing higher levels of cell cycle repressors p21, p16, and/or p53, demonstrating positive SA-β-gal staining, and secreting various SASP cytokines26, 27, 29, 45, 46. These senescent fibroblasts also express higher levels of myofibroblast marker alpha smooth muscle actin (α-SMA) and produce more extracellular matrix proteins26, 27, 29. Interestingly, Yanai et al. reported that fibroblasts from IPF lungs exhibit accelerated replicative cellular senescence phenotype but are resistant to oxidative stress-induced cellular senescence27. In contrast to senescent epithelial cells, senescent fibroblasts from IPF lungs are highly resistant to oxidative stress-induced cytotoxicity or apoptosis26, 27, 29.

Besides ATII cells and fibroblasts, other types of cells in IPF lung also show a senescent phenotype. Increased expression of p21 and activity of SA-β-gal are detected in bronchial epithelial cells in IPF lungs 5. It has also been reported that bone-marrow-derived mesenchymal stem cells (B-MSCs) from IPF lungs have an increased expression of p21, enhanced activity of SA-β-gal, and a higher level of DNA damage as well as a decreased differentiation capacity, compared to fibroblasts from age-matched healthy controls11. Incubation of normal-aged fibroblasts with the conditioned media from senescent IPF B-MSCs stimulates the expression of p21 and p53 as well as collagen genes in these normal fibroblasts, suggesting that these senescent fibroblasts have a paracrine pro-senescence capacity11.

Experimental lung fibrosis models:

Senescent cells have also been detected in the lung of experimental fibrosis models12, 3541. Bleomycin is an anti-cancer drug with a side effect of lung fibrosis. Therefore, bleomycin-induced lung fibrosis has been widely used to study the pathogenesis of lung fibrosis and the therapeutic potential of anti-fibrosis candidate drugs. It has been well-documented that intratracheal instillation of bleomycin induces senescence in alveolar epithelial cells and lung fibroblasts in mice, associated with lung fibrosis35, 37, 38, 41, 47, 48. Radiation-induced lung fibrosis is another commonly used experimental lung fibrosis model. It has been reported that the number of ATII cells that express p21 and stained positive for SA-β-gal is significantly increased in mice exposed to thoracic radiation, compared to unexposed mice39. Treatment with NADPH oxidase inhibitor diphenyleneiodonium (DPI) attenuates radiation-induced ATII cell senescence as well as lung fibrosis39, suggesting that ATII cell senescence may contribute to radiation-induced lung fibrosis.

Although it has been well documented that ATII cells undergo senescence in IPF lung and in experimental fibrosis models, how senescent ATII cells contribute to the development of lung fibrosis remains poorly understood. Emerging evidence suggests that senescent cells may affect the functions of nearby cells through secretion of biological active molecules such as cytokines, chemokines, growth factors, and proteases. Consistent with this hypothesis, studies have shown that senescent alveolar epithelial cells secreted IL-6, TNF-α, MMP-2, and MMP-933,38. Treatment of ATII cells with bleomycin in vitro also induced cell senescence and promoted the SASP35. Transforming growth factor beta 1 (TGF-β1) is one of the most ubiquitous and potent profibrogenic cytokines. Our recent studies showed that treatment with TGF-β1 under the conditions that did not cause apoptosis induced ATII cell senescence and stimulated secretion of pro-inflammatory and pro-fibrotic cytokine/growth factors in vitro12. Incubation of primary alveolar macrophages with the conditioned media from the senescent ATII cells promoted a profibrotic phenotype of macrophages12. Similarly, Tian et al. reported that culture of lung fibroblasts with the conditioned media from senescent ATII cells stimulated collagen deposition in fibroblasts35. Together, these observations suggest that senescent ATII cells may promote lung fibrosis partially by secreting biologically active molecules that promote a profibrotic phenotype of adjacent fibroblasts and alveolar macrophages12, 35.

Molecular Mechanisms Underlying Cellular Senescence in IPF Lungs

The mechanisms underlying cell senescence in either physiological or pathological conditions remain unclear. Although telomere shortening is believed to be responsible for replicative senescence in proliferating cells, the molecular mechanism linking telomere shortening to cell senescence is still not well defined. Furthermore, although it has been well documented that various stress conditions, including DNA damage and oxidative stress, induce premature cell senescence, the signaling pathways leading to cell senescence under these different stress conditions remain poorly understood. In this section, we summarize recent findings regarding the potential mechanisms underlying lung cell senescence in IPF (Fig 2).

Fig 2. Potential mechanisms underlying cell senescence in IPF lung.

Fig 2.

TERT, telomerase reverse transcriptase; PARN, polyadenylation-specific ribonuclease deadenylation nuclease; RTEL1, telomere elongation helicase 1; ROS, reactive oxygen species; PAI-1, plasminogen activator inhibitor 1.

Telomere shortening and cell senescence in IPF lungs:

A telomere is a region of tandem repeats of short DNA sequences at the end of a chromosome, which protects the chromosome from erosion. During the cell cycle, an incomplete DNA replication leads to the loss of a part of the telomere and thereby instability of the chromosome. Telomerase, consisting of telomerase reverse transcriptase (TERT) and telomerase RNA (TERC), on the other hand, restores the telomere length. It has been well documented that telomere length is shortened and DNA damage is increased in IPF lungs, compared to controls4953. It has been reported that 15 % of familial IPF patients and 3% of sporadic IPF patients bear a loss of function mutation in TERT, whereas 30% of familial IPF and 25%−50% of sporadic IPF have telomere shortening49, 50, 54, 55. These results suggest that TERT mutations only account partially for the telomere shortening in IPF. A recent study from sequencing exome of familial kindreds with pulmonary fibrosis showed that mutations in polyadenylation-specific ribonuclease deadenylation nuclease (PARN) and regulator of telomere elongation helicase 1 (RTEL1) are associated with telomere shortening and familial pulmonary fibrosis52. Cigarette smoking is one of the risk factors for IPF. Cigarette smoking has been shown to induce telomere shortening5659, although the findings are not always consistent59. It has also been reported that TERT mutation carriers may be more sensitive to cigarette smoke induced pulmonary fibrosis60. Individuals with short telomeres and/or known telomere-related mutations have higher levels of fibrotic tissues and show more rapid disease progression and shorter lung transplant–free survival53, 61, 62, further suggesting a link between telomere shortening and IPF pathophysiology.

Experimentally, several studies have shown that deletion of telomerase or inducing severe telomere dysfunction leads to cell senescence and lung fibrosis in animal models6366. Chen et al. reported that knockout of mouse gene Terc or Tert caused pulmonary alveolar stem cell replicative senescence, epithelial impairment, lung inflammation, myofibroblast differentiation, and lung fibrosis64. Naikawadi et al. showed that deletion of telomeric repeat-binding factor 1 (TERF1 or TRF1) specifically in ATII cells resulted in telomere shortening, epithelial cell senescence, and lung fibrosis65. Deletion of TRF1 in collagen expressing cells, however, cause pulmonary edema, not fibrosis, suggesting that telomere shortening in ATII cells, but not in collagen-expressing cells, is associated with lung fibrosis65. Liu et al. further showed that specific deletion of the Tert gene in ATII cells in mice did not lead to lung fibrosis; however, these mice were more susceptible to bleomycin-induced ATII cell senescence and lung fibrosis66. As telomere shortening is closely related to cell senescence, whereas cell senescence is a hallmark of aging, it is hypothesized that one of the mechanisms underlying lung cell senescence and thus aging-related susceptibility to IPF is telomere shortening49, 65, 6769.

Epigenetic mechanisms of cell senescence in IPF:

Epigenetic modifications are heritable changes in the expression of genes without involving in DNA sequence [28]. The most important epigenetic mechanisms are DNA methylation, histone post-translational modifications, and non-coding RNA-mediated gene silencing70. Accumulated evidence indicates that epigenetic modifications, including both DNA methylation and histone modifications, are involved in cellular senescence in fibrotic lung diseases5, 71, 72. Sanders et al. reported that senescent fibroblasts had global increases in histone (H4K20) trimethylation and decreases in histone (H4K16) acetylation72. These fibroblasts also had marked enrichment of the Bcl-2 gene that was H4K16 acetylated and H4K20 demethylated. These data suggest that these epigenetic modifications led to active transcription of the Bcl-2 gene, which renders senescent fibroblasts an increased resistance to apoptosis71. Sanders et al. also reported that the NADPH oxidase 4 (Nox4) gene, which codes a reactive oxygen species generating enzyme involved in fibrogenesis, is enriched with the activation histone marker, H4K16Ac, but deprive of the repressive histone mark, H4K20Me3, suggesting that epigenetic mechanisms are involved in the regulation of this gene72.

PAI-1, p53, and cell senescence in IPF lungs:

p53, a tumor repressor, is a cell cycle repressor and a master controller of cell senescence and apoptosis. The p53 pathway is among the most dysregulated pathways identified in IPF lungs29, 41, 73. Although it is well known that DNA damage leads to increased stability and activation of p53 protein through a variety of posttranslational modifications in cancer cells, how p53 is activated in lung cells in fibrotic diseases is unclear74, 75. In a recent study, Zhang et al. reported that interleukin-18 (IL-18) promotes fibroblast senescence in fibrotic lung through down-regulation of Kloth expression, which is associated with increased expression of cell cycle repressor p53, suggesting that increased IL-18 may be one of initial signals inducing p53 in senescent fibroblasts in fibrotic lung disease76.

Plasminogen activator inhibitor 1 (PAI-1) is a primary inhibitor of tissue type and urokinase type plasminogen activators (tPA and uPA), which convert plasminogen into plasmin, a serine proteinase that plays an essential role in fibrinolysis. In addition to blocking fibrinolysis through inhibition of tPA and uPA, PAI-1 is also involved in the regulation of many other biological processes, including degradation of extracellular matrix proteins, cell adhesion, migration, proliferation, and apoptosis77. PAI-1 expression increases with age in the plasma of human beings, in klotho (kl/kl) mice, a murine model of aging, and in many aging-related pathological conditions including IPF7884. Studies, including ours, suggest that increased PAI-1 expression plays a key role in the development of lung fibrosis, although the underlying mechanism remains elusive41, 8588.

Notably, increased PAI-1 expression has long been used as a marker of replicative and stress- induced cellular senescence. Emerging evidence, including that from our studies, further suggests that PAI-1 is not merely a marker but also a key mediator of cell senescence both in vitro and in vivo12, 41, 8993. Chung et al. reported that intraperitoneal injection of a truncated PAI-1 protein significantly attenuated irradiation-induced pneumocyte senescence and lung fibrosis in mice91. In a recent study, we showed that bleomycin instillation increased PAI-1 expression and induced ATII cell senescence as well as lung fibrosis in mice41. Specific ablation of the PAI-1 gene in ATII cells in mice significantly attenuated bleomycin-induced p53 and p21 expression and stimulated phosphorylation of retinoblastoma protein (pRb) in these cells in mice41. This is associated with a reduction in lung fibrosis41. Silencing p53, on the other hand, eliminated PAI-1 protein-induced senescence in rat lung epithelial cells in vitro41. Together, our data suggest that increased PAI-1 mediates bleomycin-induced ATII cell senescence through activation of p53-p21-pRb cell cycle repression axis and that ATII cell senescence contributes importantly to bleomycin-induced lung fibrosis. In another study, we showed that TGF-β1 induced ATII cell senescence through induction of p16, not p5312 and that senescent ATII cells secreted numerous pro-inflammatory and pro-fibrotic cytokines/chemokines, which promoted a pro-fibrotic phenotype in alveolar macrophages12. As PAI-1 expression is increased with age and in IPF, it is suggested that increased PAI-1 expression may underlie ATII cell senescence in IPF lungs.

The mechanism underlying increased PAI-1 expression in senescent cells in fibrotic lung diseases and the mechanism whereby PAI-1 promotes p53 expression are not well understood. Previous studies from this lab and from others have shown that reactive oxygen species (ROS) or oxidative stress induces PAI-1 in cultured cells and in vivo. As oxidative stress is increased in fibrotic lung diseases and many profibrotic mediators stimulate ROS production, it is believed that one of the mechanisms underlying increased PAI-1 expression in fibrotic lung is increased oxidative stress. Our previous studies have also shown that PAI-1 mediates bleomycin-induced phosphorylation of p53 at serine 18 in mouse lung tissue (equals serine 20 in human p53)41. As increased phosphorylation of p53 at serine 18 in mouse or at serine 20 in human prevents the binding of p53 to murine double minute 2 (MDM2), a major E3 ubiquitin ligase involved in p53 degradation94, it is hypothesized that PAI-1 increases p53 protein abundance in alveolar epithelial cells probably by stabilizing p53 protein. More studies are needed to prove this hypothesis and to reveal the molecular mechanism whereby PAI-1 modulates p53 phosphorylation.

microRNAs and cell senescence in IPF lung:

MicroRNAs (miRNAs) are small single strain noncoding RNAs with an average length of around 18–22 nucleotides. miRNAs regulate gene expression through inducing degradation of target mRNAs or inhibiting the translation of target mRNAs by binding to their 3’-untranslated regions. Emerging evidence indicates that miRNAs are involved in the regulation of cell senescence in various pathological conditions, including IPF28, 47, 9597. Disayabutr et al. reported that miR-34a, miR-34b, and miR-34c were significantly elevated in alveolar type II cells in IPF lungs, compared to patients with other interstitial lung diseases or normal controls28. This was associated with increased expression/activity of senescence markers p16, p21, p53, and SA-β-gal, and decreased expression of miR-34 target genes, including E2F1, c-Myc, and cyclin E228. Overexpression of miR-34a, miR-34b, or miR-34c in lung epithelial cells also induced cell senescence28. These results suggest that increased expression of miR-34a, miR-34b, and miR-34c may contribute to ATII cell senescence in IPF lungs.

Our studies further showed that, although miR-34a promoted senescence in both fibroblasts and alveolar epithelial cells (AECs) in bleomycin-induced lung fibrosis model, the role of miR-34a in lung fibrosis depended on the age of mice47, 98. miR-34a expression was significantly increased, mainly in myofibroblasts, in the lung of bleomycin challenged young mice98. Deletion of miR-34a augmented the sensitivity of young mice to bleomycin-induced lung fibrosis98. This was associated with diminished senescence and apoptosis in lung fibroblasts. These results suggest that miR-34a protects young mice from bleomycin-induced lung fibrosis at least in part by inducing lung fibroblast senescence and apoptosis98. In another study, we showed, however, that miR-34a was increased in AECs, but not in lung fibroblasts, in old mice and that bleomycin challenge further increased miR-34a and induced senescence and apoptosis in AECs47. Ablation of miR-34a attenuated AEC senescence and lung fibrosis in old mice47. Together, our data suggest that miR-34a expression may be distinctively regulated in different types of lung cells at different ages. In young mice, it is mainly expressed in fibroblasts and promotes fibroblast senescence and apoptosis and therefore protects mice from lung fibrosis. In old mice, however, it is mainly expressed in AECs, which is associated with AEC senescence and apoptosis as well as lung fibrosis. It should be stressed, although miR-34a associated fibroblast senescence seems to be beneficial in young mice in our study, this phenotype has been frequently implicated in the pathogenesis of lung fibrosis in aged animals and in patients. A plausible explanation for such a discrete effect may be attributable to relative contributions of different types of lung cells to this pathology between young and old subjects. Moreover, we want to point out that senescent fibroblasts are generally believed to be more resistant to cell death. However, this is not the case in miR-34a-induced phenotype change in lung fibroblasts. This suggests that other miR-34a induced effects may render these cells more conducive to apoptosis, which worth further investigations.

Senescent Cells as Therapeutic Targets for the Treatment of IPF

Senescent cells not only lose normal functions and regeneration capacity, they also secrete an array of pathogenically active molecules, which are detrimental to the surrounding tissue/cells. Therefore, removal of senescence cells has been proposed for the treatment of various aging-related diseases, including Alzheimer’s disease, diabetic kidney disease, and IPF13, 14, 1618, 30. Pan et al. reported that exposure of C57BL/6 mice to a single dose (17 Gy) of ionizing radiation induces a persistent pulmonary fibrosis 16 weeks after radiation exposure. Treatment of mice with a senolytic drug ABT-263 for 2 cycles (5 days/cycle) 16 weeks after ionizing radiation, on the other hand, significantly reduced senescent cells and reversed lung fibrosis13. Using the Ink-Attac mouse model, in which AP20187 induces apoptosis of p16 expressing cells, Schafer et al. showed that clearance of senescent cells (p16 expressing cells) significantly reduced bleomycin-induced inflammatory responses and lung fibrosis and improved lung function14. Treatment with dasatinib plus quercetin (DQ), a senolytic cocktail, replicated the beneficial effects of genetic depletion of p16 positive cells14. Similar results have been reported by another study from a different group, which confirmed the protective effect of DQ on bleomycin-induced lung fibrosis30. Importantly, first-in-human open-label pilot study showed that administration of DQ to IPF patients, 3 times/week for 3 weeks, was tolerable and side effects were mild-moderate18. Most importantly, administration of DQ significantly improved the clinical measures, such as 6-min walk distance, and performance of IPF patients, although pulmonary function, clinical chemistries, and frailty index were unchanged18. This study provides initial evidence that senolytic drugs may have therapeutic potential for IPF.

In summary, cellular senescence has emerged as an important mechanism underlying the pathogenesis of IPF. More studies are needed to improve our understanding of the molecular mechanisms leading to cellular senescence in IPF lungs and the mechanisms whereby senescent cells contribute to the development of lung fibrosis. Most importantly, the efficacy of senotherapy for the treatment of IPF warrants further investigation.

Highlight.

  1. Cellular senescence is a pathological feature of IPF lung and is evident in experimental lung fibrosis models.

  2. Various types of lung cells, including alveolar epithelial cells, fibroblasts, and endothelial cells undergo senescence in fibrotic lung diseases

  3. Different mechanisms are involved in lung cell senescence, including telomere shortening, epigenetic modifications, increased oxidative stress or inflammation, which leads to increased expression of senescence mediators PAI-1 and p53, as well as increased microRNAs, especially miR-34a.

  4. Targeting senescent cells may be effective for the treatment of fibrotic lung disease.

Acknowledgements

This work is supported by National Heart, Lung, and Blood Institute to Rui-Ming Liu (5R01HL088141; R56HL131054) and to Gang Liu (R35HL135830).

Footnotes

Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.

References

  • [1].Gorgoulis V, Adams PD, Alimonti A, Bennett DC, Bischof O, Bishop C, Campisi J, Collado M, Evangelou K, Ferbeyre G, Gil J, Hara E, Krizhanovsky V, Jurk D, Maier AB, Narita M, Niedernhofer L, Passos JF, Robbins PD, Schmitt CA, Sedivy J, Vougas K, von Zglinicki T, Zhou D, Serrano M, Demaria M: Cellular Senescence: Defining a Path Forward. Cell 2019, 179:813–27. [DOI] [PubMed] [Google Scholar]
  • [2].Rhinn M, Ritschka B, Keyes WM: Cellular senescence in development, regeneration and disease. Development 2019, 146. [DOI] [PubMed] [Google Scholar]
  • [3].Tacutu R, Budovsky A, Yanai H, Fraifeld VE: Molecular links between cellular senescence, longevity and age-related diseases - a systems biology perspective. Aging 2011, 3:1178–91. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [4].Childs BG, Durik M, Baker DJ, van Deursen JM: Cellular senescence in aging and age-related disease: from mechanisms to therapy. Nat Med 2015, 21:1424–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [5].Minagawa S, Araya J, Numata T, Nojiri S, Hara H, Yumino Y, Kawaishi M, Odaka M, Morikawa T, Nishimura SL, Nakayama K, Kuwano K: Accelerated epithelial cell senescence in IPF and the inhibitory role of SIRT6 in TGF-beta-induced senescence of human bronchial epithelial cells. Am J Physiol Lung Cell Mol Physiol 2011, 300:L391–401. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Faner R, Rojas M, Macnee W, Agusti A: Abnormal lung aging in chronic obstructive pulmonary disease and idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2012, 186:306–13. [DOI] [PubMed] [Google Scholar]
  • [7].Kuwano K, Araya J, Hara H, Minagawa S, Takasaka N, Ito S, Kobayashi K, Nakayama K: Cellular senescence and autophagy in the pathogenesis of chronic obstructive pulmonary disease (COPD) and idiopathic pulmonary fibrosis (IPF). Respiratory investigation 2016, 54:397–406. [DOI] [PubMed] [Google Scholar]
  • [8].Barnes PJ, Baker J, Donnelly LE: Cellular Senescence as a Mechanism and Target in Chronic Lung Diseases. Am J Respir Crit Care Med 2019, 200:556–64. [DOI] [PubMed] [Google Scholar]
  • [9].Acosta JC, Banito A, Wuestefeld T, Georgilis A, Janich P, Morton JP, Athineos D, Kang TW, Lasitschka F, Andrulis M, Pascual G, Morris KJ, Khan S, Jin H, Dharmalingam G, Snijders AP, Carroll T, Capper D, Pritchard C, Inman GJ, Longerich T, Sansom OJ, Benitah SA, Zender L, Gil J: A complex secretory program orchestrated by the inflammasome controls paracrine senescence. Nat Cell Biol 2013, 15:978–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [10].Kadota T, Fujita Y, Yoshioka Y, Araya J, Kuwano K, Ochiya T: Emerging role of extracellular vesicles as a senescence-associated secretory phenotype: Insights into the pathophysiology of lung diseases. Mol Aspects Med 2018, 60:92–103. [DOI] [PubMed] [Google Scholar]
  • [11].Cardenes N, Alvarez D, Sellares J, Peng Y, Corey C, Wecht S, Nouraie SM, Shanker S, Sembrat J, Bueno M, Shiva S, Mora AL, Rojas M: Senescence of bone marrow-derived mesenchymal stem cells from patients with idiopathic pulmonary fibrosis. Stem Cell Res Ther 2018, 9:257. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [12].Rana T, Jiang C, Liu G, Miyata T, Antony V, Thannickal VJ, Liu RM: PAI-1 Regulation of TGF-beta1-induced ATII Cell Senescence, SASP Secretion, and SASP-mediated Activation of Alveolar Macrophages. Am J Respir Cell Mol Biol 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [13].Pan J, Li D, Xu Y, Zhang J, Wang Y, Chen M, Lin S, Huang L, Chung EJ, Citrin DE, Wang Y, Hauer-Jensen M, Zhou D, Meng A: Inhibition of Bcl-2/xl With ABT-263 Selectively Kills Senescent Type II Pneumocytes and Reverses Persistent Pulmonary Fibrosis Induced by Ionizing Radiation in Mice. Int J Radiat Oncol Biol Phys 2017, 99:353–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [14].Schafer MJ, White TA, Iijima K, Haak AJ, Ligresti G, Atkinson EJ, Oberg AL, Birch J, Salmonowicz H, Zhu Y, Mazula DL, Brooks RW, Fuhrmann-Stroissnigg H, Pirtskhalava T, Prakash YS, Tchkonia T, Robbins PD, Aubry MC, Passos JF, Kirkland JL, Tschumperlin DJ, Kita H, LeBrasseur NK: Cellular senescence mediates fibrotic pulmonary disease. Nat Commun 2017, 8:14532. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [15].Childs BG, Li H, van Deursen JM: Senescent cells: a therapeutic target for cardiovascular disease. J Clin Invest 2018, 128:1217–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [16].Bussian TJ, Aziz A, Meyer CF, Swenson BL, van Deursen JM, Baker DJ: Clearance of senescent glial cells prevents tau-dependent pathology and cognitive decline. Nature 2018, 562:578–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [17].Zhang P, Kishimoto Y, Grammatikakis I, Gottimukkala K, Cutler RG, Zhang S, Abdelmohsen K, Bohr VA, Misra Sen J, Gorospe M, Mattson MP: Senolytic therapy alleviates Abeta-associated oligodendrocyte progenitor cell senescence and cognitive deficits in an Alzheimer’s disease model. Nat Neurosci 2019, 22:719–28. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [18].Justice JN, Nambiar AM, Tchkonia T, LeBrasseur NK, Pascual R, Hashmi SK, Prata L, Masternak MM, Kritchevsky SB, Musi N, Kirkland JL: Senolytics in idiopathic pulmonary fibrosis: Results from a first-in-human, open-label, pilot study. EBioMedicine 2019. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [19].Fernandez Perez ER, Daniels CE, Schroeder DR, St Sauver J, Hartman TE, Bartholmai BJ, Yi ES, Ryu JH: Incidence, prevalence, and clinical course of idiopathic pulmonary fibrosis: a population-based study. Chest 2010, 137:129–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [20].Fell CD, Martinez FJ, Liu LX, Murray S, Han MK, Kazerooni EA, Gross BH, Myers J, Travis WD, Colby TV, Toews GB, Flaherty KR: Clinical predictors of a diagnosis of idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 2010, 181:832–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [21].Thannickal VJ: Mechanistic links between aging and lung fibrosis. Biogerontology 2013, 14:609–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [22].Leung J, Cho Y, Lockey RF, Kolliputi N: The Role of Aging in Idiopathic Pulmonary Fibrosis. Lung 2015, 193:605–10. [DOI] [PubMed] [Google Scholar]
  • [23].Wolters PJ, Collard HR, Jones KD: Pathogenesis of idiopathic pulmonary fibrosis. Annual review of pathology 2014, 9:157–79. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [24].Thannickal VJ, Murthy M, Balch WE, Chandel NS, Meiners S, Eickelberg O, Selman M, Pardo A, White ES, Levy BD, Busse PJ, Tuder RM, Antony VB, Sznajder JI, Budinger GR: Blue journal conference. Aging and susceptibility to lung disease. Am J Respir Crit Care Med 2015, 191:261–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [25].Kuwano K, Kunitake R, Kawasaki M, Nomoto Y, Hagimoto N, Nakanishi Y, Hara N: P21Waf1/Cip1/Sdi1 and p53 expression in association with DNA strand breaks in idiopathic pulmonary fibrosis. Am J Respir Crit Care Med 1996, 154:477–83. [DOI] [PubMed] [Google Scholar]
  • [26].Hecker L, Logsdon NJ, Kurundkar D, Kurundkar A, Bernard K, Hock T, Meldrum E, Sanders YY, Thannickal VJ: Reversal of persistent fibrosis in aging by targeting nox4-nrf2 redox imbalance. Science translational medicine 2014, 6:231ra47. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Yanai H, Shteinberg A, Porat Z, Budovsky A, Braiman A, Ziesche R, Fraifeld VE: Cellular senescence-like features of lung fibroblasts derived from idiopathic pulmonary fibrosis patients. Aging 2015, 7:664–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [28].Disayabutr S, Kim EK, Cha SI, Green G, Naikawadi RP, Jones KD, Golden JA, Schroeder A, Matthay MA, Kukreja J, Erle DJ, Collard HR, Wolters PJ: miR-34 miRNAs Regulate Cellular Senescence in Type II Alveolar Epithelial Cells of Patients with Idiopathic Pulmonary Fibrosis. PloS one 2016, 11:e0158367. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [29].Alvarez D, Cardenes N, Sellares J, Bueno M, Corey C, Hanumanthu VS, Peng Y, D’Cuhna H, Sembrat J, Nouraie M, Shanker S, Caufield C, Shiva S, Armanios M, Mora AL, Rojas M: IPF lung fibroblasts have a senescent phenotype. Am J Physiol Lung Cell Mol Physiol 2017:ajplung 00220 2017. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [30].Lehmann M, Korfei M, Mutze K, Klee S, Skronska-Wasek W, Alsafadi HN, Ota C, Costa R, Schiller HB, Lindner M, Wagner DE, Gunther A, Konigshoff M: Senolytic drugs target alveolar epithelial cell function and attenuate experimental lung fibrosis ex vivo. Eur Respir J 2017, 50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [31].Reyfman PA, Walter JM, Joshi N, Anekalla KR, McQuattie-Pimentel AC, Chiu S, Fernandez R, Akbarpour M, Chen CI, Ren Z, Verma R, Abdala-Valencia H, Nam K, Chi M, Han S, Gonzalez-Gonzalez FJ, Soberanes S, Watanabe S, Williams KJN, Flozak AS, Nicholson TT, Morgan VK, Winter DR, Hinchcliff M, Hrusch CL, Guzy RD, Bonham CA, Sperling AI, Bag R, Hamanaka RB, Mutlu GM, Yeldandi AV, Marshall SA, Shilatifard A, Amaral LAN, Perlman H, Sznajder JI, Argento AC, Gillespie CT, Dematte J, Jain M, Singer BD, Ridge KM, Lam AP, Bharat A, Bhorade SM, Gottardi CJ, Budinger GRS, Misharin AV: Single-Cell Transcriptomic Analysis of Human Lung Provides Insights into the Pathobiology of Pulmonary Fibrosis. Am J Respir Crit Care Med 2019, 199:1517–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [32].Moore C, Blumhagen RZ, Yang IV, Walts A, Powers J, Walker T, Bishop M, Russell P, Vestal B, Cardwell J, Markin CR, Mathai SK, Schwarz MI, Steele MP, Lee J, Brown KK, Loyd JE, Crapo JD, Silverman EK, Cho MH, James JA, Guthridge JM, Cogan JD, Kropski JA, Swigris JJ, Bair C, Kim DS, Ji W, Kim H, Song JW, Maier LA, Pacheco KA, Hirani N, Poon AS, Li F, Jenkins RG, Braybrooke R, Saini G, Maher TM, Molyneaux PL, Saunders P, Zhang Y, Gibson KF, Kass DJ, Rojas M, Sembrat J, Wolters PJ, Collard HR, Sundy JS, O’Riordan T, Strek ME, Noth I, Ma SF, Porteous MK, Kreider ME, Patel NB, Inoue Y, Hirose M, Arai T, Akagawa S, Eickelberg O, Fernandez IE, Behr J, Mogulkoc N, Corte TJ, Glaspole I, Tomassetti S, Ravaglia C, Poletti V, Crestani B, Borie R, Kannengiesser C, Parfrey H, Fiddler C, Rassl D, Molina-Molina M, Machahua C, Worboys AM, Gudmundsson G, Isaksson HJ, Lederer DJ, Podolanczuk AJ, Montesi SB, Bendstrup E, Danchel V, Selman M, Pardo A, Henry MT, Keane MP, Doran P, Vasakova M, Sterclova M, Ryerson CJ, Wilcox PG, Okamoto T, Furusawa H, Miyazaki Y, Laurent G, Baltic S, Prele C, Moodley Y, Shea BS, Ohta K, Suzukawa M, Narumoto O, Nathan SD, Venuto DC, Woldehanna ML, Kokturk N, de Andrade JA, Luckhardt T, Kulkarni T, Bonella F, Donnelly SC, McElroy A, Armstong ME, Aranda A, Carbone RG, Puppo F, Beckman KB, Nickerson DA, Fingerlin TE, Schwartz DA: Resequencing Study Confirms That Host Defense and Cell Senescence Gene Variants Contribute to the Risk of Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med 2019, 200:199–208. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Okuda R, Aoshiba K, Matsushima H, Ogura T, Okudela K, Ohashi K: Cellular senescence and senescence-associated secretory phenotype: comparison of idiopathic pulmonary fibrosis, connective tissue disease-associated interstitial lung disease, and chronic obstructive pulmonary disease. J Thorac Dis 2019, 11:857–64. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Qiu T, Tian Y, Gao Y, Ma M, Li H, Liu X, Wu H, Zhang Y, Ding H, Cao M, Zhang J, Dai J, Chen J, Cai H: PTEN loss regulates alveolar epithelial cell senescence in pulmonary fibrosis depending on Akt activation. Aging 2019, 11:7492–509. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [35].Tian Y, Li H, Qiu T, Dai J, Zhang Y, Chen J, Cai H: Loss of PTEN induces lung fibrosis via alveolar epithelial cell senescence depending on NF-kappaB activation. Aging Cell 2019, 18:e12858. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Aoshiba K, Tsuji T, Nagai A: Bleomycin induces cellular senescence in alveolar epithelial cells. Eur Respir J 2003, 22:436–43. [DOI] [PubMed] [Google Scholar]
  • [37].Shivshankar P, Brampton C, Miyasato S, Kasper M, Thannickal VJ, Le Saux CJ: Caveolin-1 deficiency protects from pulmonary fibrosis by modulating epithelial cell senescence in mice. Am J Respir Cell Mol Biol 2012, 47:28–36. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [38].Aoshiba K, Tsuji T, Kameyama S, Itoh M, Semba S, Yamaguchi K, Nakamura H: Senescence-associated secretory phenotype in a mouse model of bleomycin-induced lung injury. Experimental and toxicologic pathology : official journal of the Gesellschaft fur Toxikologische Pathologie 2013, 65:1053–62. [DOI] [PubMed] [Google Scholar]
  • [39].Citrin DE, Shankavaram U, Horton JA, Shield W 3rd, Zhao S, Asano H, White A, Sowers A, Thetford A, Chung EJ: Role of type II pneumocyte senescence in radiation-induced lung fibrosis. J Natl Cancer Inst 2013, 105:1474–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [40].Cui H, Ge J, Xie N, Banerjee S, Zhou Y, Liu RM, Thannickal VJ, Liu G: miR-34a promotes fibrosis in aged lungs by inducing alveolar epithelial dysfunctions. Am J Physiol Lung Cell Mol Physiol 2016:ajplung 00335 2016. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [41].Jiang C, Liu G, Luckhardt T, Antony V, Zhou Y, Carter AB, Thannickal VJ, Liu RM: Serpine 1 induces alveolar type II cell senescence through activating p53-p21-Rb pathway in fibrotic lung disease. Aging Cell 2017, 16:1114–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [42].Hogan BL, Barkauskas CE, Chapman HA, Epstein JA, Jain R, Hsia CC, Niklason L, Calle E, Le A, Randell SH, Rock J, Snitow M, Krummel M, Stripp BR, Vu T, White ES, Whitsett JA, Morrisey EE: Repair and regeneration of the respiratory system: complexity, plasticity, and mechanisms of lung stem cell function. Cell stem cell 2014, 15:123–38. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Barkauskas CE, Noble PW: Cellular mechanisms of tissue fibrosis. 7. New insights into the cellular mechanisms of pulmonary fibrosis. Am J Physiol Cell Physiol 2014, 306:C987–96. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [44].Akram KM, Lomas NJ, Forsyth NR, Spiteri MA: Alveolar epithelial cells in idiopathic pulmonary fibrosis display upregulation of TRAIL, DR4 and DR5 expression with simultaneous preferential over-expression of pro-apoptotic marker p53. International journal of clinical and experimental pathology 2014, 7:552–64. [PMC free article] [PubMed] [Google Scholar]
  • [45].Schuliga M, Pechkovsky DV, Read J, Waters DW, Blokland KEC, Reid AT, Hogaboam CM, Khalil N, Burgess JK, Prele CM, Mutsaers SE, Jaffar J, Westall G, Grainge C, Knight DA: Mitochondrial dysfunction contributes to the senescent phenotype of IPF lung fibroblasts. J Cell Mol Med 2018, 22:5847–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [46].Habiel DM, Hohmann MS, Espindola MS, Coelho AL, Jones I, Jones H, Carnibella R, Pinar I, Werdiger F, Hogaboam CM: DNA-PKcs modulates progenitor cell proliferation and fibroblast senescence in idiopathic pulmonary fibrosis. BMC Pulm Med 2019, 19:165. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [47].Cui H, Ge J, Xie N, Banerjee S, Zhou Y, Liu RM, Thannickal VJ, Liu G: miR-34a promotes fibrosis in aged lungs by inducing alveolarepithelial dysfunctions. Am J Physiol Lung Cell Mol Physiol 2017, 312:L415–L24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [48].Feng F, Wang Z, Li R, Wu Q, Gu C, Xu Y, Peng W, Han D, Zhou X, Wu J, He H: Citrus alkaline extracts prevent fibroblast senescence to ameliorate pulmonary fibrosis via activation of COX-2. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie 2019, 112:108669. [DOI] [PubMed] [Google Scholar]
  • [49].Alder JK, Chen JJ, Lancaster L, Danoff S, Su SC, Cogan JD, Vulto I, Xie M, Qi X, Tuder RM, Phillips JA 3rd, Lansdorp PM, Loyd JE, Armanios MY: Short telomeres are a risk factor for idiopathic pulmonary fibrosis. Proc Natl Acad Sci U S A 2008, 105:13051–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [50].Cronkhite JT, Xing C, Raghu G, Chin KM, Torres F, Rosenblatt RL, Garcia CK: Telomere shortening in familial and sporadic pulmonary fibrosis. Am J Respir Crit Care Med 2008, 178:729–37. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [51].Tsang AR, Wyatt HD, Ting NS, Beattie TL: hTERT mutations associated with idiopathic pulmonary fibrosis affect telomerase activity, telomere length, and cell growth by distinct mechanisms. Aging Cell 2012, 11:482–90. [DOI] [PubMed] [Google Scholar]
  • [52].Stuart BD, Choi J, Zaidi S, Xing C, Holohan B, Chen R, Choi M, Dharwadkar P, Torres F, Girod CE, Weissler J, Fitzgerald J, Kershaw C, Klesney-Tait J, Mageto Y, Shay JW, Ji W, Bilguvar K, Mane S, Lifton RP, Garcia CK: Exome sequencing links mutations in PARN and RTEL1 with familial pulmonary fibrosis and telomere shortening. Nat Genet 2015, 47:512–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [53].McDonough JE, Martens DS, Tanabe N, Ahangari F, Verleden SE, Maes K, Verleden GM, Kaminski N, Hogg JC, Nawrot TS, Wuyts WA, Vanaudenaerde BM: A role for telomere length and chromosomal damage in idiopathic pulmonary fibrosis. Respir Res 2018, 19:132. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [54].Tsakiri KD, Cronkhite JT, Kuan PJ, Xing C, Raghu G, Weissler JC, Rosenblatt RL, Shay JW, Garcia CK: Adult-onset pulmonary fibrosis caused by mutations in telomerase. Proc Natl Acad Sci U S A 2007, 104:7552–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [55].Armanios MY, Chen JJ, Cogan JD, Alder JK, Ingersoll RG, Markin C, Lawson WE, Xie M, Vulto I, Phillips JA 3rd, Lansdorp PM, Greider CW, Loyd JE: Telomerase mutations in families with idiopathic pulmonary fibrosis. N Engl J Med 2007, 356:1317–26. [DOI] [PubMed] [Google Scholar]
  • [56].Valdes AM, Andrew T, Gardner JP, Kimura M, Oelsner E, Cherkas LF, Aviv A, Spector TD: Obesity, cigarette smoking, and telomere length in women. Lancet 2005, 366:662–4. [DOI] [PubMed] [Google Scholar]
  • [57].Morla M, Busquets X, Pons J, Sauleda J, MacNee W, Agusti AG: Telomere shortening in smokers with and without COPD. Eur Respir J 2006, 27:525–8. [DOI] [PubMed] [Google Scholar]
  • [58].Zhang C, Lauderdale DS, Pierce BL: Sex-Specific and Time-Varying Associations Between Cigarette Smoking and Telomere Length Among Older Adults. Am J Epidemiol 2016, 184:922–32. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [59].Astuti Y, Wardhana A, Watkins J, Wulaningsih W: Cigarette smoking and telomere length: A systematic review of 84 studies and meta-analysis. Environ Res 2017, 158:480–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [60].Diaz de Leon A, Cronkhite JT, Katzenstein AL, Godwin JD, Raghu G, Glazer CS, Rosenblatt RL, Girod CE, Garrity ER, Xing C, Garcia CK: Telomere lengths, pulmonary fibrosis and telomerase (TERT) mutations. PloS one 2010, 5:e10680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Stuart BD, Lee JS, Kozlitina J, Noth I, Devine MS, Glazer CS, Torres F, Kaza V, Girod CE, Jones KD, Elicker BM, Ma SF, Vij R, Collard HR, Wolters PJ, Garcia CK: Effect of telomere length on survival in patients with idiopathic pulmonary fibrosis: an observational cohort study with independent validation. Lancet Respir Med 2014, 2:557–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [62].Dai J, Cai H, Li H, Zhuang Y, Min H, Wen Y, Yang J, Gao Q, Shi Y, Yi L: Association between telomere length and survival in patients with idiopathic pulmonary fibrosis. Respirology 2015, 20:947–52. [DOI] [PubMed] [Google Scholar]
  • [63].Povedano JM, Martinez P, Flores JM, Mulero F, Blasco MA: Mice with Pulmonary Fibrosis Driven by Telomere Dysfunction. Cell Rep 2015, 12:286–99. [DOI] [PubMed] [Google Scholar]
  • [64].Chen R, Zhang K, Chen H, Zhao X, Wang J, Li L, Cong Y, Ju Z, Xu D, Williams BR, Jia J, Liu JP: Telomerase Deficiency Causes Alveolar Stem Cell Senescence-associated Low-grade Inflammation in Lungs. J Biol Chem 2015, 290:30813–29. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [65].Naikawadi RP, Disayabutr S, Mallavia B, Donne ML, Green G, La JL, Rock JR, Looney MR, Wolters PJ: Telomere dysfunction in alveolar epithelial cells causes lung remodeling and fibrosis. JCI insight 2016, 1:e86704. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [66].Liu T, Gonzalez De Los Santos F, Zhao Y, Wu Z, Rinke AE, Kim KK, Phan SH: Telomerase reverse transcriptase ameliorates lung fibrosis by protecting alveolar epithelial cells against senescence. J Biol Chem 2019, 294:8861–71. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [67].Snetselaar R, van Batenburg AA, van Oosterhout MFM, Kazemier KM, Roothaan SM, Peeters T, van der Vis JJ, Goldschmeding R, Grutters JC, van Moorsel CHM: Short telomere length in IPF lung associates with fibrotic lesions and predicts survival. PloS one 2017, 12:e0189467. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [68].Petrovski S, Todd JL, Durheim MT, Wang Q, Chien JW, Kelly FL, Frankel C, Mebane CM, Ren Z, Bridgers J, Urban TJ, Malone CD, Finlen Copeland A, Brinkley C, Allen AS, O’Riordan T, McHutchison JG, Palmer SM, Goldstein DB: An Exome Sequencing Study to Assess the Role of Rare Genetic Variation in Pulmonary Fibrosis. Am J Respir Crit Care Med 2017, 196:82–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Courtwright AM, El-Chemaly S: Telomeres in Interstitial Lung Disease: The Short and the Long of It. Ann Am Thorac Soc 2019, 16:175–81. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [70].Lopez-Ramirez C, Suarez Valdivia L, Rodriguez Portal JA: Causes of Pulmonary Fibrosis in the Elderly. Med Sci (Basel) 2018, 6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [71].Sanders YY, Liu H, Zhang X, Hecker L, Bernard K, Desai L, Liu G, Thannickal VJ: Histone modifications in senescence-associated resistance to apoptosis by oxidative stress. Redox biology 2013, 1:8–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [72].Sanders YY, Liu H, Liu G, Thannickal VJ: Epigenetic mechanisms regulate NADPH oxidase-4 expression in cellular senescence. Free Radic Biol Med 2015, 79:197–205. [DOI] [PubMed] [Google Scholar]
  • [73].Kusko RL, Brothers JF 2nd, Tedrow J, Pandit K, Huleihel L, Perdomo C, Liu G, Juan-Guardela B, Kass D, Zhang S, Lenburg M, Martinez F, Quackenbush J, Sciurba F, Limper A, Geraci M, Yang I, Schwartz DA, Beane J, Spira A, Kaminski N: Integrated Genomics Reveals Convergent Transcriptomic Networks Underlying Chronic Obstructive Pulmonary Disease and Idiopathic Pulmonary Fibrosis. Am J Respir Crit Care Med 2016, 194:948–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [74].Bhandary YP, Shetty SK, Marudamuthu AS, Ji HL, Neuenschwander PF, Boggaram V, Morris GF, Fu J, Idell S, Shetty S: Regulation of Lung Injury and Fibrosis by p53-Mediated Changes in Urokinase and Plasminogen Activator Inhibitor-1. Am J Pathol 2013, 183:131–43. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [75].Shetty SK, Tiwari N, Marudamuthu AS, Puthusseri B, Bhandary YP, Fu J, Levin J, Idell S, Shetty S: p53 and miR-34a Feedback Promotes Lung Epithelial Injury and Pulmonary Fibrosis. Am J Pathol 2017, 187:1016–34. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [76].Zhang LM, Zhang J, Zhang Y, Fei C, Wang L, Yi ZW, Zhang ZQ: Interleukin-18 promotes fibroblast senescence in pulmonary fibrosis through down-regulating Klotho expression. Biomedicine & pharmacotherapy = Biomedecine & pharmacotherapie 2019, 113:108756. [DOI] [PubMed] [Google Scholar]
  • [77].Lijnen HR: Pleiotropic functions of plasminogen activator inhibitor-1. J Thromb Haemost 2005, 3:35–45. [DOI] [PubMed] [Google Scholar]
  • [78].Abbate R, Prisco D, Rostagno C, Boddi M, Gensini GF: Age-related changes in the hemostatic system. Int J Clin Lab Res 1993, 23:1–3. [DOI] [PubMed] [Google Scholar]
  • [79].Tofler GH, Massaro J, Levy D, Mittleman M, Sutherland P, Lipinska I, Muller JE, D’Agostino RB: Relation of the prothrombotic state to increasing age (from the Framingham Offspring Study). Am J Cardiol 2005, 96:1280–3. [DOI] [PubMed] [Google Scholar]
  • [80].Takeshita K, Yamamoto K, Ito M, Kondo T, Matsushita T, Hirai M, Kojima T, Nishimura M, Nabeshima Y, Loskutoff DJ, Saito H, Murohara T: Increased expression of plasminogen activator inhibitor-1 with fibrin deposition in a murine model of aging, “Klotho” mouse. Semin Thromb Hemost 2002, 28:545–54. [DOI] [PubMed] [Google Scholar]
  • [81].Lijnen HR: Extracellular proteolysis in the development and progression of atherosclerosis. Biochem Soc Trans 2002, 30:163–7. [DOI] [PubMed] [Google Scholar]
  • [82].Brown NJ: Therapeutic potential of plasminogen activator inhibitor-1 inhibitors. Therapeutic advances in cardiovascular disease 2010, 4:315–24. [DOI] [PubMed] [Google Scholar]
  • [83].Serrano R, Barrenetxe J, Orbe J, Rodriguez JA, Gallardo N, Martinez C, Andres A, Paramo JA: Tissue-specific PAI-1 gene expression and glycosylation pattern in insulin-resistant old rats. Am J Physiol Regul Integr Comp Physiol 2009, 297:R1563–9. [DOI] [PubMed] [Google Scholar]
  • [84].Liu RM, van Groen T, Katre A, Cao D, Kadisha I, Ballinger C, Wang L, Carroll SL, Li L: Knockout of plasminogen activator inhibitor 1 gene reduces amyloid beta peptide burden in a mouse model of Alzheimer’s disease. Neurobiol Aging 2011, 32:1079–89. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [85].Chang W, Wei K, Jacobs SS, Upadhyay D, Weill D, Rosen GD: SPARC suppresses apoptosis of idiopathic pulmonary fibrosis fibroblasts through constitutive activation of beta-catenin. J Biol Chem 2010, 285:8196–206. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [86].Zhang YP, Li WB, Wang WL, Liu J, Song SX, Bai LL, Hu YY, Yuan YD, Zhang M: siRNA against plasminogen activator inhibitor-1 ameliorates bleomycin-induced lung fibrosis in rats. Acta Pharmacol Sin 2012, 33:897–908. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [87].Huang WT, Vayalil PK, Miyata T, Hagood J, Liu RM: Therapeutic value of small molecule inhibitor to plasminogen activator inhibitor-1 for lung fibrosis. Am J Respir Cell Mol Biol 2012, 46:87–95. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [88].Huang WT, Akhter H, Jiang C, MacEwen M, Ding Q, Antony V, Thannickal VJ, Liu RM: Plasminogen activator inhibitor 1, fibroblast apoptosis resistance, and aging-related susceptibility to lung fibrosis. Exp Gerontol 2015, 61:62–75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [89].Elzi DJ, Lai Y, Song M, Hakala K, Weintraub ST, Shiio Y: Plasminogen activator inhibitor 1--insulin-like growth factor binding protein 3 cascade regulates stress-induced senescence. Proc Natl Acad Sci U S A 2012, 109:12052–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [90].Eren M, Boe AE, Murphy SB, Place AT, Nagpal V, Morales-Nebreda L, Urich D, Quaggin SE, Budinger GR, Mutlu GM, Miyata T, Vaughan DE: PAI-1-regulated extracellular proteolysis governs senescence and survival in Klotho mice. Proc Natl Acad Sci U S A 2014, 111:7090–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [91].Chung EJ, McKay-Corkum G, Chung S, White A, Scroggins BT, Mitchell JB, Mulligan-Kehoe MJ, Citrin D: Truncated Plasminogen Activator Inhibitor-1 Protein Protects From Pulmonary Fibrosis Mediated by Irradiation in a Murine Model. Int J Radiat Oncol Biol Phys 2016, 94:1163–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [92].Ghosh AK, Rai R, Park KE, Eren M, Miyata T, Wilsbacher LD, Vaughan DE: A small molecule inhibitor of PAI-1 protects against doxorubicin-induced cellular senescence. Oncotarget 2016, 7:72443–57. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [93].Omer A, Patel D, Lian XJ, Sadek J, Di Marco S, Pause A, Gorospe M, Gallouzi IE: Stress granules counteract senescence by sequestration of PAI-1. EMBO Rep 2018, 19. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [94].Maclaine NJ, Hupp TR: The regulation of p53 by phosphorylation: a model for how distinct signals integrate into the p53 pathway. Aging 2009, 1:490–502. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [95].Cui H, Banerjee S, Xie N, Ge J, Liu RM, Matalon S, Thannickal VJ, Liu G: MicroRNA-27a-3p Is a Negative Regulator of Lung Fibrosis by Targeting Myofibroblast Differentiation. Am J Respir Cell Mol Biol 2016, 54:843–52. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [96].Munk R, Panda AC, Grammatikakis I, Gorospe M, Abdelmohsen K: Senescence-Associated MicroRNAs. Int Rev Cell Mol Biol 2017, 334:177–205. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [97].Baker JR, Vuppusetty C, Colley T, Hassibi S, Fenwick PS, Donnelly LE, Ito K, Barnes PJ: MicroRNA-570 is a novel regulator of cellular senescence and inflammaging. Faseb J 2019, 33:1605–16. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].Cui H, Ge J, Xie N, Banerjee S, Zhou Y, Antony VB, Thannickal VJ, Liu G: miR-34a Inhibits Lung Fibrosis by Inducing Lung Fibroblast Senescence. Am J Respir Cell Mol Biol 2017, 56:168–78. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES