Next Article in Journal
Permissive Modulation of Sphingosine-1-Phosphate-Enhanced Intracellular Calcium on BKCa Channel of Chromaffin Cells
Next Article in Special Issue
The History and Mystery of Alveolar Epithelial Type II Cells: Focus on Their Physiologic and Pathologic Role in Lung
Previous Article in Journal
Disabling VEGF-Response of Purkinje Cells by Downregulation of KDR via miRNA-204-5p
Previous Article in Special Issue
Extended Exhaled Nitric Oxide Analysis in Interstitial Lung Diseases: A Systematic Review
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

sFasL—The Key to a Riddle: Immune Responses in Aging Lung and Disease

by
Shulamit B. Wallach-Dayan
1,*,†,
Dmytro Petukhov
1,†,
Ronit Ahdut-HaCohen
2,3,
Mark Richter-Dayan
4 and
Raphael Breuer
1
1
Lung Cellular and Molecular Biology Laboratory, Institute of Pulmonary Medicine, Hadassah Medical Center, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
2
Department of Medical Neurobiology, Institute of Medical Research, Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
3
Department of Science, The David Yellin Academic College of Education, Jerusalem 9103501, Israel
4
Department of Emergency Medicine, Hadassah Medical School, The Hebrew University of Jerusalem, Jerusalem 91120, Israel
*
Author to whom correspondence should be addressed.
These authors contributed equally.
Int. J. Mol. Sci. 2021, 22(4), 2177; https://doi.org/10.3390/ijms22042177
Submission received: 6 December 2020 / Revised: 14 February 2021 / Accepted: 18 February 2021 / Published: 22 February 2021
(This article belongs to the Special Issue Molecular Pathology of Idiopathic Pulmonary Fibrosis)

Abstract

:
By dint of the aging population and further deepened with the Covid-19 pandemic, lung disease has turned out to be a major cause of worldwide morbidity and mortality. The condition is exacerbated when the immune system further attacks the healthy, rather than the diseased, tissue within the lung. Governed by unremittingly proliferating mesenchymal cells and increased collagen deposition, if inflammation persists, as frequently occurs in aging lungs, the tissue develops tumors and/or turns into scars (fibrosis), with limited regenerative capacity and organ failure. Fas ligand (FasL, a ligand of the Fas cell death receptor) is a key factor in the regulation of these processes. FasL is primarily found in two forms: full length (membrane, or mFasL) and cleaved (soluble, or sFasL). We and others found that T-cells expressing the mFasL retain autoimmune surveillance that controls mesenchymal, as well as tumor cell accumulation following an inflammatory response. However, mesenchymal cells from fibrotic lungs, tumor cells, or cells from immune-privileged sites, resist FasL+ T-cell-induced cell death. The mechanisms involved are a counterattack of immune cells by FasL, by releasing a soluble form of FasL that competes with the membrane version, and inhibits their cell death, promoting cell survival. This review focuses on understanding the previously unrecognized role of FasL, and in particular its soluble form, sFasL, in the serum of aged subjects, and its association with the evolution of lung disease, paving the way to new methods of diagnosis and treatment.

1. Introduction

Fas ligand (FasL, CD95L, CD178), a 40 kDa cell surface transmembrane protein, is known to be primarily expressed by activated T-cells [1] and their released extracellular vesicles, from prenatal to somatic stages [2,3,4]. It forms multimere complexes [5] to elicit cell death by induction of a signaling complex (DISC), which binds to Fas (CD95), a TNF-family cell death receptor [4]. The Fas death receptor is commonly expressed on so called “target” cells, such as lung epithelial cells, fibroblasts, T-lymphocytes, virally infected cells and tumor cells [1,3,6]. In some conditions, these same cells (tumors, fibroblasts from fibrotic sites) may also express the Fas ligand, and transform from a defensive situation to an offensive one by counterattacking cells of the immune system and escaping “immune surveillance” just as do cells in immune-privileged sites (e.g., testis Sertoli cells, eye corneal epithelium and neurons) [1,7]. Membrane FasL also enabled myofibroblasts to attain a cytotoxic phenotype and eliminate alveolar epithelial cells and T-cells [7,8].
A soluble form of FasL exists (sFasL), and is known to compete with the full, membrane-anchored protein (mFasL) on the binding site of the Fas death receptor [9], that not only joins this modulation of immune responses [1,2,5], but also prompts cell motility [2,10,11,12].
In this review, we will discuss new perceptions of mechanisms of sFasL increments, mostly in blood serum, and its role in lung health and disease during aging.
This knowledge is important for understanding the role sFasL plays in various conditions of the aging and diseased lung, with emphasis on immunological disorders, including viral infection, and may pave the way for appropriate molecular diagnostics and therapies.

2. Serum Levels of Soluble FasL Increase with Age

Aging is a physiological process characterized by an “inflammaging” response, i.e., a low-grade inflammation comprising the senescence-associated secretory profile (SASP) [13] of pro-inflammatory and cell death effectors affecting tissue repair, regeneration and promoting premalignant responses [13]. Senescence is associated with a variety of lung diseases, including those mediated by viral infection [14,15]. An important SASP-associated cytokine is the Fas ligand. Membrane FasL expression, for example, in T-cells, was shown to decrease with age in the blood serum of healthy individuals [16], whereas the soluble form increased [17,18,19] (see Table 1). A similar increase in sFasL levels was detected in the serum of Werner syndrome patients, a typical accelerated aging syndrome, which was associated with chronic inflammation [17]. Although a few studies reported a mild decrease in the age-related increments of sFasL in serum in normal subjects [20,21], and they were even lower in athletes [22] (Table 1), its increments with aging were further detected in animal studies [5]. In particular, the percentage of activated rat T-lymphocytes expressing FasL increased from about 5% in cells from young animals to more than 50% in old counterparts [23]. This phenomenon and its mechanisms are further detailed.

3. Molecular Signaling and Mechanism of Soluble FasL Increment with Age

The mechanism behind soluble FasL increments in the serum of aging subjects has not been determined with certainty.
Oxidative stress is a known contributing aspect in physiological aging [26], which results from accumulation of oxidants generated during life in normal metabolism, in physiological inflammation, and also in various pathological states [27,28,29,30]. T-lymphocytes, microglial cells, endothelial cells and intestinal and lung epithelial cells are known to increase oxidative stress and induce FasL expression [28,31]. There is clear evidence that oxidative stress, in particular during aging, increases the number of extracellular vesicles that carry and release FasL protein [32]. Among them, we may find exosomes (small microvesicles 10–100 nm in diameter) [33], which are released from the multivesicular bodies through membrane invaginations [34] (see Figure 1) into the circulation and tissues [35]. Phosphorylation and ubiquitination of the FasL molecule are important for its transportation to secretory lysosomes [36]. Microvesicles containing FasL, on their surrounding membrane, were shown to negate T-cell immunity by induction of T-cell death (apoptosis) [37], and FasL, on exosomes, induced antigen-specific apoptosis in autologous CD4+ T-cells by a lymphoblastoid cell line [38]. This may be one of the reasons that exosomes have been indicated as potential biomarkers for aging-related diseases [39].
The literature reflects the involvement of reactive oxygen species (ROS) in FasL gene expression, both at the DNA [40,41] and the RNA [42,43] levels. In particular, the FasL promoter (FASLG), which is responsible for FasL DNA transcription, contains binding sites to transcription factors (e.g., NF-κB), known to be affected by a cell’s redox status [40,41]. Concomitantly, oxidative stress affects FasL mRNA [42,43] and microRNA-mediated FasL gene expression [44,45,46]. A short form (8 kDa) of FasL, which lacks a cytosolic transmembrane tail and a part of the extracellular domain, but retains two of the glycosylation sites of the extracellular domain [47], may result from alternative splicing of FasL mRNA.
Oxidative stress not only enhances FasL DNA and RNA with a subsequent increase in its protein levels; oxidative stress further affects the FasL protein itself. In particular, it promotes FasL cleavage via activation of metalloproteinases (MMPs) [48,49,50,51], which increase with age [26]. Many studies have reported increased cleavage and shedding of membrane FasL to its soluble form by MMPs directly from the cell’s membrane, and indirectly from secretory lysosomes which are further released from cells as extracellular vesicles [5,52,53,54] (see Figure 1). The details of the regulation of FasL cleavage and the generation of its soluble form are as yet incomplete. However, it is known that molecules such as 17β-estradiol upregulate both FasL transcription (increase RNA), and subsequent FasL cleavage by MMP3 (stromelysin-1) through estrogen receptor α (ERα) in osteoblasts and osteoclasts, and that MMP3 blockers inhibited sFasL production [55,56,57]. Similarly, TH2 cytokines, and IL-13 in particular, have been shown to increase shedding of FasL by increasing MMP7 activity in bronchial epithelial cells of patients with severe asthma [58]. This increase in shedding is accompanied by decreased FasL synthesis, and is presumed to be modulated through dephosphorylation of the proteins of the forkhead (FKHR) family, which facilitates their nuclear translocation and binding to the FKHR-responsive element of the FasL promoter [58], which may indicate regulatory feedback inhibition.
In fact, the sFasL-mediated inhibition of apoptosis signaling vs. induction depends on the particular metalloproteinase that cleaves membrane FasL [53]. We have recently reported that MMP-7 knockout mice had decreased sFasL levels in their bloodstream, and further shown that these mice had attenuated lung fibrosis [59]. Particularly, fibroblasts with decreased expression of MMP-7, a shedder of sFasL, have been shown to increase sensitivity to apoptosis [59,60]. Concomitantly, high MMP7 decreases their sensitivity to apoptosis [54,61]. In a mirror situation, sFasL has been demonstrated to increase the production of ROS in neutrophils, with activation of inflammatory pathways [62]. In this context, oxidative stress is also a major factor, causing or exacerbating pulmonary diseases, including fibrosis [28,63,64,65].

4. Soluble FasL Increase in Serum of Patients with Pulmonary Disease

The pulmonary diseases associated with increased levels of the soluble form of FasL include (see Table 2): interstitial lung diseases (ILDs) such as hypersensitivity pneumonitis [66], idiopathic pulmonary fibrosis (IPF) [67], asbestosis and interstitial pneumonia [4,6,66,68]. Increased sFasL levels were also detected in patients diagnosed with lung cancer following chemotherapy, pulmonary sarcoidosis, pulmonary infections of varying etiology [69,70,71,72], acute lung injury (ALI) [73] and acute respiratory distress syndrome (ARDS) [54,73,74,75,76,77]. Similar changes were observed in fibrotic lung diseases in bronchoalveolar lavage fluid (BAL) [4,66]. In patients with chronic obstructive pulmonary disease (COPD), the levels of sFasL are directly related to the disease symptoms [78,79,80]. Patients with cachexic COPD show significantly increased serum sFasL levels compared to non-cachexic patients [78,79,80]. The sFasL content of serum is known to increase in patients diagnosed with asthma and is treated with omalizumab, and in allergic children during the symptomatic period [81,82]. Nevertheless, there is no general direction of change in sFasL serum levels that would be relevant for all asthma patients universally [83]; specifically, there is a significant decrease in plasma sFasL levels of patients with uncontrolled allergic asthma [83]. Of note, although increments in sFasL serum levels have been observed in numerous pulmonary pathologies, it was shown to decrease in pneumonic effusion, acute respiratory distress syndrome and lung cancer [4,6,54,68].
The specific roles of sFasL release in pulmonary diseases may vary per case. For instance, we [6] and others [68,86] have detected an anti-apoptotic role of sFasL (see Figure 2) as a mechanism of IPF by promoting immune suppression, immune privilege and immune escape [6,68,86,87,88,89]. We demonstrated that sFasL inhibits T-cell-induced apoptosis in IPF lung fibroblasts [6], in contrast to the role of mFasL in the cytotoxicity of CD4+ T-cells, which ensures their escape from immune surveillance [7], survival and proliferation in vitro, and in vivo in the experimental lung fibrosis and air pouch models [6]. Concomitantly, in other systems sFasL was demonstrated to promote cell survival as opposed to apoptosis by initiation of signaling pathways via JNK, Akt or ERK activation [9] (see Figure 2). For instance, sFasL binding to the Fas receptor of cell line GM6112 was shown to augment ERK1/2 activation, but not p38 [90]. sFasL induces proliferation in fibroblast-like synuviocytes from rheumatoid arthritis patients by activation of PI3K and caspase-8 signaling pathways, in addition to ERK signaling [91]. Although in the minority, it should be mentioned that a proapoptotic role has also been attributed to sFasL in lung pathologies such as ALI, ARDS and pulmonary adenocarcinoma [73,85,92]. This is akin to the role of membrane-bound Fas ligand [2,3,93].
Interaction of the soluble form of FasL with Fas receptor may also initiate cell migration in target cells (see Figure 2). This FasL-mediated cell migration is postulated to be involved in both the malignant transformation and the fibrotic processes [94]. All this allows not only cell survival, immune regulation and escape from immune surveillance, but also locomotion (e.g., cancer, fibrosis) with consequent disease progression.

5. Immune Disorders and Viral Infection Increase the Levels of Soluble FasL in Blood

Both soluble and membrane forms of FasL are known regulators of the immune system [9]. Soluble FasL is known to be involved in inflammation [2], including chronic inflammation [95] (see Table 3). Its serum concentration is elevated in patients with autoimmune lymphoproliferative syndrome [96]. Mice, exclusively expressing the soluble form of FasL, develop lymphadenopathy due to lack of selective immune cell-induced apoptosis usually performed by the membrane form of Fas ligand [97].
Soluble FasL levels have been consistently demonstrated to reflect the immune mobilization during viral and bacterial infection [75,86,101]. For instance, the concentrations of sFasL in serum and bronchoalveolar lavage (BAL) of asymptomatic carriers of human T-lymphotropic virus type-1 (HTLV-1) were associated with the percentage of CD4+ lymphocytes in BAL [86].
Serum sFasL supplementation may help prevent damage to glandular organs in Sjögren’s syndrome, which was demonstrated to be associated with decreased levels of sFasL in blood [99]. On the other hand, ex vivo depletion of sFasL was efficient in depleting alloreacting human donor anti-host T-cells in graft-versus-host disease [100].
Fas/mFasL signaling has only minimal involvement in both the mechanism of canine coronavirus type II-induced apoptosis [102] and SARS coronavirus-infected human dendritic cells [103], paving the way for the involvement of sFasL. Similarly, cell death induced in human dendritic cells by human coronavirus 229E (responsible for the common cold) was shown to be independent of FasL regulation [104].

6. Discussion

The distinction between the elevation of sFasL levels in the lung caused by disease, versus that caused by aging or even by immune disorders, including viral infection, is a challenging issue. It may be that that they are interconnected by nature. Interestingly, a linear correlation between age and the cubic root of serum sFasL concentration in humans was determined [18]. Sexual dimorphism adds additional complexity to this issue, considering that some pathologies may be overrepresented in one sex. Studies on women undergoing hormone therapy have detected increased serum sFasL levels as well [46].
Oxidative stress, and ROS in particular, is a known factor accompanying normal aging, as well as aging-associated diseases [63]. ROS contribute to cell senescence in the normal process of ontogenesis [63]. SASP is an important indication of cellular senescence, which is associated with lung pathologies [13]. As the role of FasL in the induction of pulmonary fibrosis has also been demonstrated in animals [6], the perspective of interconnection between soluble FasL and SASP presents itself as an interesting topic for further research. Fas ligand, and specifically its soluble form, is gaining recognition as an important factor involved in the progression of various pathological conditions [105].
The knowledge on sFasL involvement in viral diseases is still incomplete. Generally, sFasL release in viral infection is considered a part of the inflammatory immune response, specifically, as a potent chemo-attractant of human polymorphonuclear neutrophils [106]. In relation to lung diseases, serum or BAL sFasL was associated with fibrotic and inflammatory diseases [4,6,68,78] and cancer, including lung cancer [69,77,101].
sFasL has been suggested as a biomarker of disease progression in IPF [4,66], pneumonia [66,107], bronchiolitis obliterans [67], oxidative stress [63], cancer and aging [21]. There is a substantial translational potential for our understanding of the roles and regulation of various forms of released Fas ligand. This includes development of novel approaches to treatment with, for example, FasL-fused humanized antibodies to sensitize target cells to cell death [108,109,110] as suggested in glaucoma treatment [111,112], novel forms of RNA therapy [113], prognosis of long-term allergic outcomes at birth [114] or even for schizophrenia treatment [115]. However, there remain a number of outstanding problems that need to be addressed with regard to soluble Fas ligand as a therapeutic target. The switch between induction of apoptosis and other FasL-dependent signaling cascades, and the origin of the different FasL-associated signaling types through the Fas receptor, stand out as particularly important unanswered questions. Nevertheless, there is little doubt that FasL regulation will remain an important field of investigation in future biochemical and clinical studies.

Funding

This work was supported by a personal grant from Arthur Gutterman.

Conflicts of Interest

The authors have no conflict of interest.

References

  1. Lettau, M.; Paulsen, M.; Schmidt, H.; Janssen, O. Insights into the molecular regulation of FasL (CD178) biology. Eur. J. Cell Biol. 2011, 90, 456–466. [Google Scholar] [CrossRef]
  2. Guégan, J.-P.; Legembre, P. Nonapoptotic functions of Fas/CD95 in the immune response. FEBS J. 2018, 285, 809–827. [Google Scholar] [CrossRef] [PubMed]
  3. Schulte, M.; Reiss, K.; Lettau, M.; Maretzky, T.; Ludwig, A.; Hartmann, D.; de Strooper, B.; Janssen, O.; Saftig, P. ADAM10 regulates FasL cell surface expression and modulates FasL-induced cytotoxicity and activation-induced cell death. Cell Death Differ. 2007, 14, 1040–1049. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Kuwano, K.; Maeyama, T.; Inoshima, I.; Ninomiya, K.; Hagimoto, N.; Yoshimi, M.; Fujita, M.; Nakamura, N.; Shirakawa, K.; Hara, N. Increased circulating levels of soluble Fas ligand are correlated with disease activity in patients with fibrosing lung diseases. Respirology 2002, 7, 15–21. [Google Scholar] [CrossRef] [PubMed]
  5. Gregory, M.S.; Hackett, C.G.; Abernathy, E.F.; Lee, K.S.; Saff, R.R.; Hohlbaum, A.M.; Moody, K.-S.L.; Hobson, M.W.; Jones, A.; Kolovou, P.; et al. Opposing Roles for Membrane Bound and Soluble Fas Ligand in Glaucoma-Associated Retinal Ganglion Cell Death. PLoS ONE 2011, 6, e17659. [Google Scholar] [CrossRef] [PubMed]
  6. Wallach-Dayan, S.B.; Elkayam, L.; Golan-Gerstl, R.; Konikov, J.; Zisman, P.; Dayan, M.R.; Arish, N.; Breuer, R. Cutting edge: FasL(+) immune cells promote resolution of fibrosis. J. Autoimmun. 2015, 59, 67–76. [Google Scholar] [CrossRef] [PubMed]
  7. Wallach-Dayan, S.B.; Golan-Gerstl, R.; Breuer, R. Evasion of myofibroblasts from immune surveillance: A mechanism for tissue fibrosis. Proc. Natl. Acad. Sci. USA 2007, 104, 20460–20465. [Google Scholar] [CrossRef] [Green Version]
  8. Golan-Gerstl, R.; Wallach-Dayan, S.B.; Amir, G.; Breuer, R. Epithelial Cell Apoptosis by Fas Ligand–Positive Myofibroblasts in Lung Fibrosis. Am. J. Respir. Cell Mol. Biol. 2007, 36, 270–275. [Google Scholar] [CrossRef] [Green Version]
  9. Yamada, A.; Arakaki, R.; Saito, M.; Kudo, Y.; Ishimaru, N. Dual Role of Fas/FasL-Mediated Signal in Peripheral Immune Tolerance. Front. Immunol. 2017, 8, 403. [Google Scholar] [CrossRef] [Green Version]
  10. Kleber, S.; Sancho-Martinez, I.; Wiestler, B.; Beisel, A.; Gieffers, C.; Hill, O.; Thiemann, M.; Mueller, W.; Sykora, J.; Kuhn, A.; et al. Yes and PI3K Bind CD95 to Signal Invasion of Glioblastoma. Cancer Cell 2008, 13, 235–248. [Google Scholar] [CrossRef] [Green Version]
  11. Guégan, J.P.; Ginestier, C.; Charafe-Jauffret, E.; Ducret, T.; Quignard, J.-F.; Vacher, P.; Legembre, P. CD95/Fas and metastatic disease: What does not kill you makes you stronger. Semin. Cancer Biol. 2019. [Google Scholar] [CrossRef]
  12. Stock, C.; Schwab, A. Role of the Na+/H+ exchanger NHE1 in cell migration. Acta Physiol. 2006, 187, 149–157. [Google Scholar] [CrossRef]
  13. 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, 857–864. [Google Scholar] [CrossRef]
  14. Wallach-Dayan, S.B.; Rojas, M. Senescence, the Janus of Lung Injury and Repair. Am. J. Respir. Cell Mol. Biol. 2020, 62, 548–549. [Google Scholar] [CrossRef] [PubMed]
  15. Baz-Martínez, M.; Da Silva-Álvarez, S.; Rodríguez, E.; Guerra, J.; El Motiam, A.; Vidal, A.; García-Caballero, T.; González-Barcia, M.; Sánchez, L.; Muñoz-Fontela, C.; et al. Cell senescence is an antiviral defense mechanism. Sci. Rep. 2016, 6, 37007. [Google Scholar] [CrossRef]
  16. Pinti, M. Development of real time PCR assays for the quantification of Fas and FasL mRNA levels in lymphocytes: Studies on centenarians. Mech. Ageing Dev. 2003, 124, 511–516. [Google Scholar] [CrossRef]
  17. Goto, M. Elevation of soluble Fas (APO-1, CD95) ligand in natural aging and Werner syndrome. Biosci. Trends 2008, 2, 124–127. [Google Scholar]
  18. Jiang, S.; Moriarty-Craige, S.E.; Li, C.; Lynn, M.J.; Cai, J.; Jones, D.P.; Sternberg, P. Associations of plasma-soluble fas ligand with aging and age-related macular degeneration. Investig. Ophthalmol. Vis. Sci. 2008, 49, 1345–1349. [Google Scholar] [CrossRef] [PubMed]
  19. Aggarwal, S.; Gupta, S. Increased Apoptosis of T Cell Subsets in Aging Humans: Altered Expression of Fas (CD95), Fas Ligand, Bcl-2, and Bax. J. Immunol. 1998, 160, 1627–1637. [Google Scholar] [PubMed]
  20. Ichikura, T.; Majima, T.; Uchida, T.; Okura, E.; Ogawa, T.; Mochizuki, H. Plasma soluble Fas ligand concentration: Decrease in elderly men and increase in patients with gastric carcinoma. Oncol. Rep. 2001, 8, 311–314. [Google Scholar] [CrossRef]
  21. Kavathia, N.; Jain, A.; Walston, J.; Beamer, B.A.; Fedarko, N.S. Serum markers of apoptosis decrease with age and cancer stage. Aging 2009, 1, 652–663. [Google Scholar] [CrossRef] [Green Version]
  22. Kangas, R.; Törmäkangas, T.; Heinonen, A.; Alen, M.; Suominen, H.; Kovanen, V.; Laakkonen, E.K.; Korhonen, M.T. Declining Physical Performance Associates with Serum FasL, miR-21, and miR-146a in Aging Sprinters. Biomed Res. Int. 2017, 2017, 8468469. [Google Scholar] [CrossRef] [PubMed]
  23. Pahlavani, M.A.; Vargas, D.A. Aging but not dietary restriction alters the activation-induced apoptosis in rat T cells. FEBS Lett. 2001, 491, 114–118. [Google Scholar] [CrossRef] [Green Version]
  24. Duan, P.; Hu, C.; Butler, H.J.; Quan, C.; Chen, W.; Huang, W.; Tang, S.; Zhou, W.; Yuan, M.; Shi, Y.; et al. 4-Nonylphenol induces disruption of spermatogenesis associated with oxidative stress-related apoptosis by targeting p53-Bcl-2/Bax-Fas/FasL signaling. Environ. Toxicol. 2017, 32, 739–753. [Google Scholar] [CrossRef] [Green Version]
  25. Soni, H.; Kaminski, D.; Gangaraju, R.; Adebiyi, A. Cisplatin-induced oxidative stress stimulates renal Fas ligand shedding. Ren. Fail. 2018, 40, 314–322. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  26. Freitas-Rodríguez, S.; Folgueras, A.R.; López-Otín, C. The role of matrix metalloproteinases in aging: Tissue remodeling and beyond. Biochim. Biophys. Acta (BBA) Mol. Cell Res. 2017, 1864, 2015–2025. [Google Scholar] [CrossRef]
  27. Zhou, T.; Prather, E.R.; Garrison, D.E.; Zuo, L. Interplay between ROS and Antioxidants during Ischemia-Reperfusion Injuries in Cardiac and Skeletal Muscle. Int. J. Mol. Sci. 2018, 19, 417. [Google Scholar] [CrossRef] [Green Version]
  28. Wallach-Dayan, S.B.; Izbicki, G.; Cohen, P.Y.; Gerstl-Golan, R.; Fine, A.; Breuer, R. Bleomycin initiates apoptosis of lung epithelial cells by ROS but not by Fas/FasL pathway. Am. J. Physiol. Lung Cell. Mol. Physiol. 2006, 290, L790–L796. [Google Scholar] [CrossRef]
  29. Sonveaux, P. ROS and radiotherapy: More we care. Oncotarget 2017, 8, 35482. [Google Scholar] [CrossRef]
  30. Helmerhorst, H.J.F.; Schultz, M.J.; Van Der Voort, P.H.J.; De Jonge, E.; Van Westerloo, D.J. Bench-to-bedside review: The effects of hyperoxia during critical illness. Crit. Care 2015, 19, 284. [Google Scholar] [CrossRef] [Green Version]
  31. Suzuki, M.; Aoshiba, K.; Nagai, A. Oxidative stress increases Fas ligand expression in endothelial cells. J. Inflamm. (Lond. Engl.) 2006, 3, 11. [Google Scholar] [CrossRef] [Green Version]
  32. Martínez-Lorenzo, M.J.; Anel, A.; Gamen, S.; Monle n, I.; Lasierra, P.; Larrad, L.; Piñeiro, A.; Alava, M.A.; Naval, J. Activated human T cells release bioactive Fas ligand and APO2 ligand in microvesicles. J. Immunol. 1999, 163, 1274–1281. [Google Scholar] [PubMed]
  33. Picca, A.; Guerra, F.; Calvani, R.; Bucci, C.; Lo Monaco, M.; Bentivoglio, A.; Coelho-Júnior, H.; Landi, F.; Bernabei, R.; Marzetti, E. Mitochondrial Dysfunction and Aging: Insights from the Analysis of Extracellular Vesicles. Int. J. Mol. Sci. 2019, 20, 805. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Ståhl, A.-L.; Johansson, K.; Mossberg, M.; Kahn, R.; Karpman, D. Exosomes and microvesicles in normal physiology, pathophysiology, and renal diseases. Pediatric Nephrol. (Berl. Ger.) 2019, 34, 11–30. [Google Scholar] [CrossRef] [Green Version]
  35. Salminen, A.; Kaarniranta, K.; Kauppinen, A. Exosomal vesicles enhance immunosuppression in chronic inflammation: Impact in cellular senescence and the aging process. Cell. Signal. 2020, 75, 109771. [Google Scholar] [CrossRef] [PubMed]
  36. Zuccato, E.; Blott, E.J.; Holt, O.; Sigismund, S.; Shaw, M.; Bossi, G.; Griffiths, G.M. Sorting of Fas ligand to secretory lysosomes is regulated by mono-ubiquitylation and phosphorylation. J. Cell Sci 2007, 120, 191–199. [Google Scholar] [CrossRef] [Green Version]
  37. Kim, J.W.; Wieckowski, E.; Taylor, D.D.; Reichert, T.E.; Watkins, S.; Whiteside, T.L. Fas ligand-positive membranous vesicles isolated from sera of patients with oral cancer induce apoptosis of activated T lymphocytes. Clin. Cancer Res. Off. J. Am. Assoc. Cancer Res. 2005, 11, 1010–1020. [Google Scholar]
  38. Klinker, M.W.; Lizzio, V.; Reed, T.J.; Fox, D.A.; Lundy, S.K. Human B Cell-Derived Lymphoblastoid Cell Lines Constitutively Produce Fas Ligand and Secrete MHCII(+)FasL(+) Killer Exosomes. Front. Immunol. 2014, 5, 144. [Google Scholar] [CrossRef] [Green Version]
  39. Saheera, S.; Potnuri, A.G.; Krishnamurthy, P. Nano-Vesicle (Mis)Communication in Senescence-Related Pathologies. Cells 2020, 9, 1974. [Google Scholar] [CrossRef]
  40. Volpe, E.; Sambucci, M.; Battistini, L.; Borsellino, G. Fas–Fas Ligand: Checkpoint of T Cell Functions in Multiple Sclerosis. Front. Immunol. 2016, 7, 382. [Google Scholar] [CrossRef] [Green Version]
  41. Morgan, M.J.; Liu, Z.-G. Crosstalk of reactive oxygen species and NF-κB signaling. Cell Res. 2011, 21, 103–115. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  42. Nagata, S.; Golstein, P. The Fas death factor. Science 1995, 267, 1449–1456. [Google Scholar] [CrossRef] [PubMed]
  43. Hug, H.; Strand, S.; Grambihler, A.; Galle, J.; Hack, V.; Stremmel, W.; Krammer, P.H.; Galle, P.R. Reactive Oxygen Intermediates Are Involved in the Induction of CD95 Ligand mRNA Expression by Cytostatic Drugs in Hepatoma Cells. J. Biol. Chem. 1997, 272, 28191–28193. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Chen, S.; Yang, C.; Sun, C.; Sun, Y.; Yang, Z.; Cheng, S.; Zhuge, B. miR-21-5p Suppressed the Sensitivity of Hepatocellular Carcinoma Cells to Cisplatin by Targeting FASLG. DNA Cell Biol. 2019, 38, 865–873. [Google Scholar] [CrossRef]
  45. Zhang, B.Y.; Zhao, Z.; Jin, Z. Expression of miR-98 in myocarditis and its influence on transcription of the FAS/FASL gene pair. Genet. Mol. Res. GMR 2016, 15, 1–9. [Google Scholar] [CrossRef] [PubMed]
  46. Kangas, R.; Pöllänen, E.; Rippo, M.R.; Lanzarini, C.; Prattichizzo, F.; Niskala, P.; Jylhävä, J.; Sipilä, S.; Kaprio, J.; Procopio, A.D.; et al. Circulating miR-21, miR-146a and Fas ligand respond to postmenopausal estrogen-based hormone replacement therapy—A study with monozygotic twin pairs. Mech. Ageing Dev. 2014, 143–144, 1–8. [Google Scholar] [CrossRef] [Green Version]
  47. Ayroldi, E.; D’Adamio, F.; Zollo, O.; Agostini, M.; Moraca, R.; Cannarile, L.; Migliorati, G.; Delfino, D.V.; Riccardi, C. Cloning and expression of a short Fas ligand: A new alternatively spliced product of the mouse Fas ligand gene. Blood 1999, 94, 3456–3467. [Google Scholar] [CrossRef]
  48. Ho, B.-Y.; Wu, Y.-M.; Chang, K.-J.; Pan, T.-M. Dimerumic Acid Inhibits SW620 Cell Invasion by Attenuating H2O2-Mediated MMP-7 Expression via JNK/C-Jun and ERK/C-Fos Activation in an AP-1-Dependent Manner. Int. J. Biol. Sci. 2011, 7, 869–880. [Google Scholar] [CrossRef] [Green Version]
  49. Kelly, P.J.; Morrow, J.D.; Ning, M.; Koroshetz, W.; Lo, E.H.; Terry, E.; Milne, G.L.; Hubbard, J.; Lee, H.; Stevenson, E.; et al. Oxidative stress and matrix metalloproteinase-9 in acute ischemic stroke: The Biomarker Evaluation for Antioxidant Therapies in Stroke (BEAT-Stroke) study. Stroke 2008, 39, 100–104. [Google Scholar] [CrossRef] [Green Version]
  50. Haorah, J.; Ramirez, S.H.; Schall, K.; Smith, D.; Pandya, R.; Persidsky, Y. Oxidative stress activates protein tyrosine kinase and matrix metalloproteinases leading to blood-brain barrier dysfunction. J. Neurochem. 2007, 101, 566–576. [Google Scholar] [CrossRef]
  51. Demeule, M.; Brossard, M.; Pagé, M.; Gingras, D.; Béliveau, R. Matrix metalloproteinase inhibition by green tea catechins. Biochim. Biophys. Acta 2000, 1478, 51–60. [Google Scholar] [CrossRef]
  52. Weinlich, R.; Brunner, T.; Amarante-Mendes, G.P. Control of death receptor ligand activity by posttranslational modifications. Cell. Mol. Life Sci. CMLS 2010, 67, 1631–1642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Voss, M.; Lettau, M.; Paulsen, M.; Janssen, O. Posttranslational regulation of Fas ligand function. Cell Commun. Signal. CCS 2008, 6, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Herrero, R.; Kajikawa, O.; Matute-Bello, G.; Wang, Y.; Hagimoto, N.; Mongovin, S.; Wong, V.; Park, D.R.; Brot, N.; Heinecke, J.W.; et al. The biological activity of FasL in human and mouse lungs is determined by the structure of its stalk region. J. Clin. Investig. 2011, 121, 1174–1190. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Garcia, A.J.; Tom, C.; Guemes, M.; Polanco, G.; Mayorga, M.E.; Wend, K.; Miranda-Carboni, G.A.; Krum, S.A. ERα signaling regulates MMP3 expression to induce FasL cleavage and osteoclast apoptosis. J. Bone Miner. Res. 2013, 28, 283–290. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Krum, S.A.; Miranda-Carboni, G.A.; Hauschka, P.V.; Carroll, J.S.; Lane, T.F.; Freedman, L.P.; Brown, M. Estrogen protects bone by inducing Fas ligand in osteoblasts to regulate osteoclast survival. EMBO J. 2008, 27, 535–545. [Google Scholar] [CrossRef] [Green Version]
  57. Nakamura, T.; Imai, Y.; Matsumoto, T.; Sato, S.; Takeuchi, K.; Igarashi, K.; Harada, Y.; Azuma, Y.; Krust, A.; Yamamoto, Y.; et al. Estrogen Prevents Bone Loss via Estrogen Receptor α and Induction of Fas Ligand in Osteoclasts. Cell 2007, 130, 811–823. [Google Scholar] [CrossRef]
  58. Wadsworth, S.J.; Atsuta, R.; McIntyre, J.O.; Hackett, T.-L.; Singhera, G.K.; Dorscheid, D.R. IL-13 and TH2 cytokine exposure triggers matrix metalloproteinase 7–mediated Fas ligand cleavage from bronchial epithelial cells. J. Allergy Clin. Immunol. 2010, 126, 366–374.e368. [Google Scholar] [CrossRef]
  59. Nareznoi, D.; Konikov-Rozenman, J.; Petukhov, D.; Breuer, R.; Wallach-Dayan, S.B. Matrix Metalloproteinases Retain Soluble FasL-mediated Resistance to Cell Death in Fibrotic-Lung Myofibroblasts. Cells 2020, 9, 411. [Google Scholar] [CrossRef] [Green Version]
  60. Fingleton, B.; Vargo-Gogola, T.; Crawford, H.C.; Matrisian, L.M. Matrilysin [MMP-7] Expression Selects for Cells with Reduced Sensitivity to Apoptosis. Neoplasia 2001, 3, 459–468. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Mitsiades, N.; Yu, W.H.; Poulaki, V.; Tsokos, M.; Stamenkovic, I. Matrix metalloproteinase-7-mediated cleavage of Fas ligand protects tumor cells from chemotherapeutic drug cytotoxicity. Cancer Res. 2001, 61, 577–581. [Google Scholar]
  62. Margaryan, S.; Witkowicz, A.; Arakelyan, A.; Partyka, A.; Karabon, L.; Manukyan, G. sFasL-mediated induction of neutrophil activation in patients with type 2 diabetes mellitus. PLoS ONE 2018, 13, e0201087. [Google Scholar] [CrossRef]
  63. Fois, A.G.; Paliogiannis, P.; Sotgia, S.; Mangoni, A.A.; Zinellu, E.; Pirina, P.; Carru, C.; Zinellu, A. Evaluation of oxidative stress biomarkers in idiopathic pulmonary fibrosis and therapeutic applications: A systematic review. Respir. Res. 2018, 19, 1–13. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Kliment, C.R.; Oury, T.D. Oxidative stress, extracellular matrix targets, and idiopathic pulmonary fibrosis. Free Radic. Biol. Med. 2010, 49, 707–717. [Google Scholar] [CrossRef]
  65. Han, M.K.; Murray, S.; Fell, C.D.; Flaherty, K.R.; Toews, G.B.; Myers, J.; Colby, T.V.; Travis, W.D.; Kazerooni, E.A.; Gross, B.H.; et al. Sex differences in physiological progression of idiopathic pulmonary fibrosis. Eur. Respir. J. 2008, 31, 1183–1188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Kuwano, K.; Hagimoto, N.; Kawasaki, M.; Nakamura, N.; Shirakawa, K.; Maeyama, T.; Hara, N. Expression of FasL and Fas protein and their soluble form in patients with hypersensitivity pneumonitis. Int. Arch. Allergy Immunol. 2000, 122, 209–215. [Google Scholar] [CrossRef] [PubMed]
  67. Kuwano, K.; Kawasaki, M.; Maeyama, T.; Hagimoto, N.; Nakamura, N.; Shirakawa, K.; Hara, N. Soluble form of fas and fas ligand in BAL fluid from patients with pulmonary fibrosis and bronchiolitis obliterans organizing pneumonia. Chest 2000, 118, 451–458. [Google Scholar] [CrossRef] [Green Version]
  68. Kopiński, P.; Balicka-Ślusarczyk, B.; Dyczek, A.; Szpechciński, A.; Przybylski, G.; Jarzemska, A.; Wandtke, T.; Jankowski, M.; Iwaniec, T.; Chorostowska-Wynimko, J. Enhanced expression of Fas Ligand (FasL) in the lower airways of patients with fibrotic interstitial lung diseases (ILDs). Folia Histochem. Cytobiol. 2011, 49, 636–645. [Google Scholar] [CrossRef] [Green Version]
  69. Erdoğan, B.; Uzaslan, E.; Budak, F.; Karadağ, M.; Ediger, D.; Oral, B.; Göral, G.; Ege, E.; Gözü, O. The evaluation of soluble Fas and soluble Fas ligand levels of bronchoalveolar lavage fluid in lung cancer patients. Tuberk. Toraks 2005, 53, 127–131. [Google Scholar]
  70. Shimizu, M.; Kondo, M.; Ito, Y.; Kume, H.; Suzuki, R.; Yamaki, K. Soluble Fas and Fas ligand provide new information on metastasis and response to chemotherapy in SCLC patients. Cancer Detect. Prev. 2005, 29, 175–180. [Google Scholar] [CrossRef]
  71. Yoshimura, C.; Nomura, S.; Kanazawa, S.; Kuwana, M.; Muramatsu, M.; Yamaguchi, K.; Fukuhara, S. Analysis of cytotoxic T lymphocytes and Fas/FasL in Japanese patients with non-small cell lung cancer associated with HLA-A2. J. Cancer Res. Clin. Oncol. 2002, 128, 581–588. [Google Scholar] [CrossRef]
  72. Shikuwa, C.; Kadota, J.-I.; Mukae, H.; Iwashita, T.; Kaida, H.; Ishii, H.; Ishimatsu, Y.; Kohno, S. High concentrations of soluble Fas ligand in bronchoalveolar lavage fluid of patients with pulmonary sarcoidosis. Respir. Int. Rev. Thorac. Dis. 2002, 69, 242–246. [Google Scholar] [CrossRef]
  73. Albertine, K.H.; Soulier, M.F.; Wang, Z.; Ishizaka, A.; Hashimoto, S.; Zimmerman, G.A.; Matthay, M.A.; Ware, L.B. Fas and Fas Ligand Are Up-Regulated in Pulmonary Edema Fluid and Lung Tissue of Patients with Acute Lung Injury and the Acute Respiratory Distress Syndrome. Am. J. Pathol. 2002, 161, 1783–1796. [Google Scholar] [CrossRef] [Green Version]
  74. Wang, P.S.; Chen, Y.M.; Hsieh, Y.L.; Yu, C.F.; Tsai, C.M.; Perng, R.P. Pleural effusion and serum soluble fas-ligand levels are elevated in different clinical conditions. Lung 2002, 180, 25–32. [Google Scholar] [CrossRef]
  75. Budak, F.; Uzaslan, E.K.; Cangür, S.; Göral, G.; Oral, H.B. Increased pleural soluble fas ligand (sFasL) levels in tuberculosis pleurisy and its relation with T-helper type 1 cytokines. Lung 2008, 186, 337–343. [Google Scholar] [CrossRef] [PubMed]
  76. Wu, S.H.; Chu, J.J.; Chiang, C.D. Increased soluble Fas ligand concentration in tuberculous pleural effusion. J. Formos. Med Assoc. Taiwan Yi Zhi 2001, 100, 32–34. [Google Scholar] [PubMed]
  77. Naumnik, W.; Izycki, T.; Ossolinska, M.; Chyczewska, E. Serum levels of sFas and sFasL during chemotherapy of lung cancer. Exp. Oncol. 2007, 29, 132–136. [Google Scholar]
  78. Kosacka, M.; Porębska, I.; Korzeniewska, A.; Rubinsztajn, R.; Grabicki, M.; Jankowska, R.; Batura-Gabryel, H.; Chazan, R. Serum levels of apoptosis-related markers (sFasL, TNF-a, p53 and bcl-2) in COPD patients. Pneumonol. I Alergol. Pol. 2016, 84, 11–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Takabatake, N.; Nakamura, H.; Inoue, S.; Terashita, K.; Yuki, H.; Kato, S.; Yasumura, S.; Tomoike, H. Circulating levels of soluble Fas ligand and soluble Fas in patients with chronic obstructive pulmonary disease. Respir. Med. 2000, 94, 1215–1220. [Google Scholar] [CrossRef] [Green Version]
  80. Yasuda, N.; Gotoh, K.; Minatoguchi, S.; Asano, K.; Nishigaki, K.; Nomura, M.; Ohno, A.; Watanabe, M.; Sano, H.; Kumada, H.; et al. An increase of soluble Fas, an inhibitor of apoptosis, associated with progression of COPD. Respir. Med. 1998, 92, 993–999. [Google Scholar] [CrossRef] [Green Version]
  81. Kao, S.-L.; Yu, H.-R.; Kuo, H.-C.; Tsui, K.-Y.; Wu, C.-C.; Chang, L.-S.; Liang, C.-D.; Chung, Y.-H.; Yang, K.D. Higher levels of soluble Fas ligand and transforming growth factor-β after omalizumab treatment: A case report. J. Microbiol. Immunol. Infect. Wei Mian Yu Gan Ran Za Zhi 2012, 45, 69–71. [Google Scholar] [CrossRef] [Green Version]
  82. Mezei, G.; Lévay, M.; Sepler, Z.; Héninger, E.; Kozma, G.T.; Cserháti, E. Seasonal changes of proapoptotic soluble Fas ligand level in allergic rhinitis combined with asthma. Pediatric Allergy Immunol. 2006, 17, 444–449. [Google Scholar] [CrossRef] [PubMed]
  83. Daneshmandi, S.; Pourfath Elah, A.A.; Pourpak, Z.; Heydarnezhad, H. Soluble form of FasL (sFasL) in adult asthma. Iran. Red Crescent Med. J. (IRCMJ) 2009, 11, 301–305. [Google Scholar]
  84. Kuwano, K.; Hagimoto, N.; Kawasaki, M.; Yatomi, T.; Nakamura, N.; Nagata, S.; Suda, T.; Kunitake, R.; Maeyama, T.; Miyazaki, H.; et al. Essential roles of the Fas-Fas ligand pathway in the development of pulmonary fibrosis. J. Clin. Investig. 1999, 104, 13–19. [Google Scholar] [CrossRef] [Green Version]
  85. Matute-Bello, G.; Liles, W.C.; Steinberg, K.P.; Kiener, P.A.; Mongovin, S.; Chi, E.Y.; Jonas, M.; Martin, T.R. Soluble Fas Ligand Induces Epithelial Cell Apoptosis in Humans with Acute Lung Injury (ARDS). J. Immunol. 1999, 163, 2217–2225. [Google Scholar]
  86. Sakamoto, N.; Mukae, H.; Fujii, T.; Kakugawa, T.; Kaida, H.; Kadota, J.-I.; Kohno, S. Soluble form of Fas and Fas ligand in serum and bronchoalveolar lavage fluid of individuals infected with human T-lymphotropic virus type 1. Respir. Med. 2004, 98, 213–219. [Google Scholar] [CrossRef] [Green Version]
  87. Janssen, O.; Qian, J.; Linkermann, A.; Kabelitz, D. CD95 ligand—Death factor and costimulatory molecule? Cell Death Differ. 2003, 10, 1215–1225. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Bohana-Kashtan, O.; Civin, C.I. Fas ligand as a tool for immunosuppression and generation of immune tolerance. Stem Cells 2004, 22, 908–924. [Google Scholar] [CrossRef] [PubMed]
  89. Muschen, M.; Moers, C.; Warskulat, U.; Even, J.; Niederacher, D.; Beckmann, M.W. CD95 ligand expression as a mechanism of immune escape in breast cancer. Immunology 2000, 99, 69–77. [Google Scholar] [CrossRef]
  90. Calmon-Hamaty, F.; Audo, R.; Combe, B.; Morel, J.; Hahne, M. Targeting the Fas/FasL system in Rheumatoid Arthritis therapy: Promising or risky? Cytokine 2015, 75, 228–233. [Google Scholar] [CrossRef]
  91. Audo, R.; Calmon-Hamaty, F.; Papon, L.; Combe, B.; Morel, J.; Hahne, M. Distinct Effects of Soluble and Membrane-Bound Fas Ligand on Fibroblast-like Synoviocytes From Rheumatoid Arthritis Patients. Arthritis Rheumatol. 2014, 66, 3289–3299. [Google Scholar] [CrossRef] [PubMed]
  92. Serrao, K.L.; Fortenberry, J.D.; Owens, M.L.; Harris, F.L.; Brown, L.A. Neutrophils induce apoptosis of lung epithelial cells via release of soluble Fas ligand. Am. J. Physiol. Lung Cell. Mol. Physiol. 2001, 280, L298–L305. [Google Scholar] [CrossRef] [Green Version]
  93. Sharma, K.; Wang, R.X.; Zhang, L.Y.; Yin, D.L.; Luo, X.Y.; Solomon, J.C.; Jiang, R.F.; Markos, K.; Davidson, W.; Scott, D.W.; et al. Death the Fas way: Regulation and pathophysiology of CD95 and its ligand. Pharmacol. Ther. 2000, 88, 333–347. [Google Scholar] [CrossRef]
  94. Salton, F.; Volpe, M.C.; Confalonieri, M. Epithelial–Mesenchymal Transition in the Pathogenesis of Idiopathic Pulmonary Fibrosis. Medicina 2019, 55, 83. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Lotti, R.; Shu, E.; Petrachi, T.; Marconi, A.; Palazzo, E.; Quadri, M.; Lin, A.; O’Reilly, L.A.; Pincelli, C. Soluble Fas Ligand Is Essential for Blister Formation in Pemphigus. Front. Immunol. 2018, 9, 370. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Rieux-Laucat, F.; Magerus-Chatinet, A.; Neven, B. The Autoimmune Lymphoproliferative Syndrome with Defective FAS or FAS-Ligand Functions. J. Clin. Immunol. 2018, 38, 558–568. [Google Scholar] [CrossRef] [PubMed]
  97. O’Reilly, L.A.; Hughes, P.; Lin, A.; Waring, P.; Siebenlist, U.; Jain, R.; Gray, D.H.D.; Gerondakis, S.; Strasser, A. Loss of c-REL but not NF-κB2 prevents autoimmune disease driven by FasL mutation. Cell Death Differ. 2015, 22, 767–778. [Google Scholar] [CrossRef] [Green Version]
  98. Lau, C.-Y.; Mihalek, A.D.; Wang, J.; Dodd, L.E.; Perkins, K.; Price, S.; Webster, S.; Pittaluga, S.; Folio, L.R.; Rao, V.K.; et al. Pulmonary Manifestations of the Autoimmune Lymphoproliferative Syndrome. A Retrospective Study of a Unique Patient Cohort. Ann. Am. Thorac. Soc. 2016, 13, 1279–1288. [Google Scholar] [CrossRef]
  99. Luo, J.; Wang, Y.; Yu, B.; Qian, H.; He, Y.; Shi, G. A Potential of sFasL in Preventing Gland Injury in Sjogren’s Syndrome. Biomed. Res. Int. 2017, 5981432. [Google Scholar] [CrossRef]
  100. Bohana-Kashtan, O.; Morisot, S.; Hildreth, R.; Brayton, C.; Levitsky, H.I.; Civin, C.I. Selective Reduction of Graft-versus-Host Disease-Mediating Human T Cells by Ex Vivo Treatment with Soluble Fas Ligand. J. Immunol. (Baltim. Md. 1950) 2009, 183, 696–705,. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Linkermann, A.; Qian, J.; Lettau, M.; Kabelitz, D.; Janssen, O. Considering Fas ligand as a target for therapy. Expert Opin. Ther. Targets 2005, 9, 119–134. [Google Scholar] [CrossRef]
  102. Marfè, G.; Tafani, M.; Fiorito, F.; Pagnini, U.; Iovane, G.; De Martino, L. Involvement of FOXO Transcription Factors, TRAIL-FasL/Fas, and Sirtuin Proteins Family in Canine Coronavirus Type II-Induced Apoptosis. PLoS ONE 2011, 6, e27313. [Google Scholar] [CrossRef] [Green Version]
  103. Law, H.K.; Cheung, C.; Sia, S.; Chan, Y.; Peiris, J.M.; Lau, Y. Toll-like receptors, chemokine receptors and death receptor ligands responses in SARS coronavirus infected human monocyte derived dendritic cells. BMC Immunol. 2009, 10, 35. [Google Scholar] [CrossRef] [Green Version]
  104. Mesel-Lemoine, M.; Millet, J.; Vidalain, P.O.; Law, H.; Vabret, A.; Lorin, V.; Escriou, N.; Albert, M.L.; Nal, B.; Tangy, F. A Human Coronavirus Responsible for the Common Cold Massively Kills Dendritic Cells but Not Monocytes. J. Virol. 2012, 86, 7577–7587. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Le Gallo, M.; Poissonnier, A.; Blanco, P.; Legembre, P. CD95/Fas, Non-Apoptotic Signaling Pathways, and Kinases. Front. Immunol. 2017, 8, 1216. [Google Scholar] [CrossRef] [Green Version]
  106. O’Connell, J. Immune privilege or inflammation? The paradoxical effects of Fas ligand. Arch. Immunol Exp. (Warsz.) 2000, 48, 73–79. [Google Scholar] [PubMed]
  107. Kuwano, K.; Hagimoto, N.; Maeyama, T.; Fujita, M.; Yoshimi, M.; Inoshima, I.; Nakashima, N.; Hamada, N.; Watanabe, K.; Hara, N. Mitochondria-mediated apoptosis of lung epithelial cells in idiopathic interstitial pneumonias. Lab. Investig. 2002, 82, 1695–1706. [Google Scholar] [CrossRef] [Green Version]
  108. Strasser, A.; Jost, P.J.; Nagata, S. The Many Roles of FAS Receptor Signaling in the Immune System. Immunity 2009, 30, 180–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Chien, M.-H.; Chang, W.-M.; Lee, W.-J.; Chang, Y.-C.; Lai, T.-C.; Chan, D.V.; Sharma, R.; Lin, Y.-F.; Hsiao, M. A Fas Ligand (FasL)-Fused Humanized Antibody Against Tumor-Associated Glycoprotein 72 Selectively Exhibits the Cytotoxic Effect Against Oral Cancer Cells with a Low FasL/Fas Ratio. Mol. Cancer Ther. 2017, 16, 1102–1113. [Google Scholar] [CrossRef] [Green Version]
  110. Muraki, M. Sensitization to cell death induced by soluble Fas ligand and agonistic antibodies with exogenous agents: A review. AIMS Med Sci. 2020, 7, 122–203. [Google Scholar] [CrossRef]
  111. Krishnan, A.; Fei, F.; Jones, A.; Ksander, B.R.; Rothstein, A.M.; Gregory-Ksander, M.S. Gene therapy treatment with AAV-soluble Fas ligand protects retinal ganglion cells during development of Glaucoma. Investig. Ophthalmol. Vis. Sci. 2015, 56, 2594. [Google Scholar]
  112. Krishnan, A.; Fei, F.; Jones, A.; Busto, P.; Marshak-Rothstein, A.; Ksander, B.R.; Gregory-Ksander, M. Overexpression of Soluble Fas Ligand following Adeno-Associated Virus Gene Therapy Prevents Retinal Ganglion Cell Death in Chronic and Acute Murine Models of Glaucoma. J. Immunol. 2016, 197, 4626–4638. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  113. Putzbach, W.; Haluck-Kangas, A.; Gao, Q.Q.; Sarshad, A.A.; Bartom, E.T.; Stults, A.; Qadir, A.S.; Hafner, M.; Peter, M.E. CD95/Fas ligand mRNA is toxic to cells. eLife 2018, 7, e38621. [Google Scholar] [CrossRef] [PubMed]
  114. Su, K.-W.; Yeh, K.-W.; Huang, J.L. Cord Blood Soluble Fas Ligand Predicts Allergic Rhinitis, Obstructive-type Lung Function, and Mite Sensitization at Seven Year-old Children: The PATCH study. J. Allergy Clin. Immunol. 2018, 141, AB203. [Google Scholar] [CrossRef]
  115. Szymona, K.; Karakula-Juchnowicz, H.; Zdzisinska, B.; Flis, M.; Kaławaj, K.; Rosa, W.; Kandefer-Szerszeń, M. Soluble Fas ligand (sFasL) as a predictor of reduction of general psychopathology in schizophrenia after antipsychotic treatment. Eur. Psychiatry 2016, 33, S108. [Google Scholar] [CrossRef]
Figure 1. Mechanisms of FasL secretion by cells. Full-length FasL, termed membrane FasL (mFasL), is found in the cytosol, membrane bodies such as secretory lysosomes and multivesicular bodies, as well as on the cell surface. It may be released in full-length form embedded in the membrane of extracellular bodies as exosomes and microvesicles or, in a shorter version, cleaved by metalloproteinases and shed, termed soluble FasL (sFasL).
Figure 1. Mechanisms of FasL secretion by cells. Full-length FasL, termed membrane FasL (mFasL), is found in the cytosol, membrane bodies such as secretory lysosomes and multivesicular bodies, as well as on the cell surface. It may be released in full-length form embedded in the membrane of extracellular bodies as exosomes and microvesicles or, in a shorter version, cleaved by metalloproteinases and shed, termed soluble FasL (sFasL).
Ijms 22 02177 g001
Figure 2. Mechanisms of FasL action on “target” cells. (A) Interaction of multiple trimers of membrane FasL with Fas receptors of recipient cells (cancer cells, cells with viral infection, etc.) initiates apoptosis. (B) Interaction of soluble form of FasL with Fas receptor, initiating cell migration in target cells. Cell motility is induced by PI3/Akt cascade, anchoring of cytoskeleton to NHE1, through interaction between C-terminal domain of the Na+/H+ exchanger and actin-binding proteins. (C) The motility-inducing signaling complex (MISC), Ca2+ influx, recruitment of NADPH oxidase-3 (NOX3) in MISC and c-Yes kinase activation. (D) sFasL competitive binding to Fas receptor produces anti-apoptotic and anti-cell death effects in nuclei of target cells via activation of pro-survival signaling cascades.
Figure 2. Mechanisms of FasL action on “target” cells. (A) Interaction of multiple trimers of membrane FasL with Fas receptors of recipient cells (cancer cells, cells with viral infection, etc.) initiates apoptosis. (B) Interaction of soluble form of FasL with Fas receptor, initiating cell migration in target cells. Cell motility is induced by PI3/Akt cascade, anchoring of cytoskeleton to NHE1, through interaction between C-terminal domain of the Na+/H+ exchanger and actin-binding proteins. (C) The motility-inducing signaling complex (MISC), Ca2+ influx, recruitment of NADPH oxidase-3 (NOX3) in MISC and c-Yes kinase activation. (D) sFasL competitive binding to Fas receptor produces anti-apoptotic and anti-cell death effects in nuclei of target cells via activation of pro-survival signaling cascades.
Ijms 22 02177 g002
Table 1. Changes in soluble Fas ligand (sFasL) levels during aging, and aged-related conditions and/or diseases.
Table 1. Changes in soluble Fas ligand (sFasL) levels during aging, and aged-related conditions and/or diseases.
ConditionDirection of ChangeNotesReferences
Aging
Normal agingSerum[17,18]
Aging-associated chronic inflammationSerum[20,21]
Werner syndromeSerum[17]
Age-related macular degenerationPlasma[18]
Aging-associated T-cell population decreaseSerum[19]
Aging (athletes)Serum[22]
GlaucomaSerum. Decreased ratio of sFasL to membrane FasL (mFasL) leads to increased susceptibility to fibrosis[5]
Oxidative Stress
ConditionDirection of ChangeNotesReferences
Disruption of spermatogenesisTesticular tissue[24]
Renal degenerationPlasma, urine[25]
Table 2. Changes in sFasL levels in serum and bronchoalveolar lavage fluid (BAL) of patients with lung disease.
Table 2. Changes in sFasL levels in serum and bronchoalveolar lavage fluid (BAL) of patients with lung disease.
ConditionDirection of ChangeNotesReferences
Idiopathic pulmonary fibrosis (IPF) Serum and BAL[67,84]
Hypersensitivity pneumonitisSerum and BAL[66]
Interstitial pneumoniaSerum and BAL[67]
Chronic obstructive pulmonary disease (COPD)Serum[78]
AsthmaSerum[83]
Asthma (uncontrolled allergic patients)Serum[83]
Asthma (omalizumab treatment)Serum[81]
Asthma (allergic children)Serum, during symptomatic period[82]
Acute lung injury [73]
Acute respiratory distress syndromeBAL of patients at risk of death[85]
Lung cancer chemotherapySerum[77]
Lung cancer BAL[69]
Small cell lung cancerSerum[70]
Non-small cell lung cancerSerum[71]
Pulmonary sarcoidosisBAL[72]
Table 3. Changes in sFasL levels in immune-associated disorders.
Table 3. Changes in sFasL levels in immune-associated disorders.
ConditionDirection of ChangeNotesReferences
InflammationSerum[2]
Autoimmune lymphoproliferative syndromeSerum[96,98]
Sjögren’s syndromeSerum[99]
Chronic inflammation (pemphigus)Serum[95]
Graft-versus-host diseaseSerum [100]
Tuberculosis pleurisyBAL[75]
Asymptomatic carriers of human T-lymphotropic virus type-1 (HTLV-1)Serum and BAL[86]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Wallach-Dayan, S.B.; Petukhov, D.; Ahdut-HaCohen, R.; Richter-Dayan, M.; Breuer, R. sFasL—The Key to a Riddle: Immune Responses in Aging Lung and Disease. Int. J. Mol. Sci. 2021, 22, 2177. https://doi.org/10.3390/ijms22042177

AMA Style

Wallach-Dayan SB, Petukhov D, Ahdut-HaCohen R, Richter-Dayan M, Breuer R. sFasL—The Key to a Riddle: Immune Responses in Aging Lung and Disease. International Journal of Molecular Sciences. 2021; 22(4):2177. https://doi.org/10.3390/ijms22042177

Chicago/Turabian Style

Wallach-Dayan, Shulamit B., Dmytro Petukhov, Ronit Ahdut-HaCohen, Mark Richter-Dayan, and Raphael Breuer. 2021. "sFasL—The Key to a Riddle: Immune Responses in Aging Lung and Disease" International Journal of Molecular Sciences 22, no. 4: 2177. https://doi.org/10.3390/ijms22042177

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