Next Article in Journal
Ex-Vivo Pharmacological Defatting of the Liver: A Review
Next Article in Special Issue
Molecular Genetic Approach and Evaluation of Cardiovascular Events in Patients with Clinical Familial Hypercholesterolemia Phenotype from Romania
Previous Article in Journal
Impact of Antibiotic Therapy and Metabolic Parameters in Non-Small Cell Lung Cancer Patients Receiving Checkpoint Inhibitors
Previous Article in Special Issue
Symptomatic and Asymptomatic Patients in the Polish Atrial Fibrillation (POL-AF) Registry
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

TRAIL and Cardiovascular Disease—A Risk Factor or Risk Marker: A Systematic Review

by
Katarzyna Kakareko
1,*,
Alicja Rydzewska-Rosołowska
1,
Edyta Zbroch
2 and
Tomasz Hryszko
1
1
2nd Department of Nephrology and Hypertension with Dialysis Unit, Medical University of Białystok, 15-276 Białystok, Poland
2
Department of Internal Medicine and Hypertension, Medical University of Białystok, 15-276 Białystok, Poland
*
Author to whom correspondence should be addressed.
J. Clin. Med. 2021, 10(6), 1252; https://doi.org/10.3390/jcm10061252
Submission received: 28 January 2021 / Revised: 12 March 2021 / Accepted: 15 March 2021 / Published: 18 March 2021
(This article belongs to the Special Issue The Risk Factors of Cardiovascular Diseases)

Abstract

:
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is a pro-apoptotic protein showing broad biological functions. Data from animal studies indicate that TRAIL may possibly contribute to the pathophysiology of cardiomyopathy, atherosclerosis, ischemic stroke and abdominal aortic aneurysm. It has been also suggested that TRAIL might be useful in cardiovascular risk stratification. This systematic review aimed to evaluate whether TRAIL is a risk factor or risk marker in cardiovascular diseases (CVDs) focusing on major adverse cardiovascular events. Two databases (PubMed and Cochrane Library) were searched until December 2020 without a year limit in accordance to the PRISMA guidelines. A total of 63 eligible original studies were identified and included in our systematic review. Studies suggest an important role of TRAIL in disorders such as heart failure, myocardial infarction, atrial fibrillation, ischemic stroke, peripheral artery disease, and pulmonary and gestational hypertension. Most evidence associates reduced TRAIL levels and increased TRAIL-R2 concentration with all-cause mortality in patients with CVDs. It is, however, unclear whether low TRAIL levels should be considered as a risk factor rather than a risk marker of CVDs. Further studies are needed to better define the association of TRAIL with cardiovascular diseases.

1. Introduction

The Greek term “apoptosis” meaning “falling leaves in autumn” was first used for the description of programmed cell death in 1972 by John Kerr [1]. Its origin dates back to Hippocrates [2], and since then, substantial progress has been made in our understanding of this naturally occurring process. Apoptosis can be triggered by various factors (biological, chemical, physical) and it is tightly regulated by a family of proteases called caspases, which are activated during cellular response to a specific trigger. Apoptotic signaling can be internal (intrinsic apoptosis) or external (extrinsic apoptosis). The intrinsic pathway can be induced by cellular stress or DNA damage. The extrinsic pathway is induced by signals activating cell-surface death-receptors. Apart from an important role in physiological processes such as aging, apoptosis (especially when it is inadequate) contributes to the development of many disorders including malignancies [3], and neurological [4] and cardiovascular diseases (CVDs) [5]. In recent years, a large number of studies have tried to clarify how this complex process is actively driven by the cells. One of the proteins involved in inducing apoptosis is TNF-related apoptosis-inducing ligand (TRAIL). TRAIL, which is also known as Apo-2 ligand (Apo-2L) or TNF super family 10 (TNFSN10), is a 281-amino acid type II transmembrane protein [6]. The membrane-bound TRAIL can be cleaved by cysteine proteases, and secreted into circulation, leading to the formation of the soluble form of TRAIL. Activated monocytes and neutrophils are the most important source of soluble TRAIL [7]. Interest in TRAIL rapidly increased when it turned out that TRAIL could selectively kill cancer cells whilst sparing normal cells [8]. This potential anti-tumor effect led to identifying TRAIL as a promising anti-cancer agent [9]. Those varied biological functions result from different receptors being activated by TRAIL.
Five TRAIL receptors have been identified: TRAIL-R1 and TRAIL-R2 (death receptors), TRAIL-R3 and TRAIL-R4 (decoy receptors), and osteoprotegerin (OPG, which is a soluble decoy receptor). Their alternative names are detailed in Figure 1. Decoy receptors act as molecular traps for agonists, binding TRAIL and keeping it from binding with death receptors. TRAIL-R1 and TRAIL-R2 have intracellular death domain motifs while TRAIL-R3 and TRAIL-R4 lack the cytoplasmic domains necessary to transduce apoptotic signal (Figure 1). Thus, TRAIL may exert pleiotropic effects, inducing apoptosis when binding to death receptors, but with TRAIL-induced apoptosis blocked by the decoy receptors. Soluble forms of those receptors are also found in plasma. TRAIL and its receptor system therefore seem to maintain a balance between apoptotic and antiapoptotic actions.
Recently, a lot of attention has been focused on the components of OPG/TRAIL axis as proteins implicated in the development of CVDs. The TRAIL receptors are expressed in the cardiovascular (CV) system in vascular smooth muscle cells [10] and cardiomyocytes [11]. Data from animal studies shows that TRAIL may possibly contribute to the pathophysiology of cardiomyopathy [12,13], atherosclerosis [14,15], ischemic stroke [16], pulmonary hypertension [17] and abdominal aortic aneurysm [18]. It has also been suggested that TRAIL might be potentially useful in CV risk stratification [19].
The association between TRAIL and CVDs is not well understood. We aimed to answer the question of whether TRAIL is a risk factor or a risk marker in CVDs, particularly taking into consideration major adverse CV events defined as a composite endpoint: nonfatal myocardial infarction, nonfatal stroke and CV death. We analyzed clinical and experimental data from human studies aiming to gather full knowledge on the subject in question.

2. Materials and Methods

The report of the methods used for this systematic review was in accordance with the Preferred Reporting Items for Systematic Reviews and Meta-Analyses (PRISMA) consensus statement. We conducted a comprehensive literature search in the electronic databases including PubMed and Cochrane Library up to December 29, 2020. Search strategies included keywords and Medical Subject Heading (MeSH) terms with all subheadings included. The MeSH terms were “cardiovascular diseases”, “cerebrovascular disorders” and “pulmonary embolus” (searched together using the Boolean operator “OR”). The keyword used were: “TRAIL” and “cardiovascular risk”. The terms were combined using the Boolean operator “AND”. We also searched the combined mode of MeSH or keywords. The results were merged with duplicates discarded. Further studies were sought by manually searching reference lists of the relevant articles. Relevant articles were selected based on their title, abstract or full text. Articles were excluded if they were clearly related to another subject matter or were not published in English, French, or German.

3. Results

The detailed process of literature searching is illustrated by flowchart (Figure 2). Initially, 1559 articles were identified from electronic databases, 1279 were screened, and 107 were retrieved and assessed for eligibility. Finally, 63 original studies were included in our systematic review. We have identified 48 clinical and 15 basic studies. All of them are detailed in Table 1, Table 2 and Table 3. Fifteen of those papers reported cardiovascular event endpoints. Due to their heterogeneity (different endpoints and measured proteins), the risk of bias was not assessed. We decided to summarize them together since they all included patient-related outcome measures (Table 3). From those 15 studies, nine were regarding TRAIL and six regarding TRAIL-R2. TRAIL was inversely correlated with all-cause death in patients with CVD in seven out of nine studies (one found an association that was not statistically significant); cardiovascular death was inversely correlated with TRAIL only in two out of three studies. In four out of six studies, higher TRAIL-R2 levels predicted CV death or a CV event.

4. Discussion

The intention of this review was to summarize current knowledge about the involvement of the TRAIL pathway in the development and prognosis of CVDs. Moreover, we tried to evaluate published studies focusing on cardiovascular outcomes. Unfortunately, we were unable to assess the quality of data on TRAIL and major adverse CV events because of the heterogenicity of the groups. Conducted studies had various endpoints and the number of studies with a similar endpoint assessing CVD risk was insufficient for our objective. We decided, therefore, to include all studies since we think the information gathered from them is very interesting and shows new clinical data in terms of TRAIL receptors and their prognostic utility.
The most important finding is that prospective studies demonstrated that lower TRAIL concentration predicted a poor prognosis in patients with CVD, e.g., Volpato et al. [69], Osmancik et al. [70], Richter et al. [72]. In the study by Volpato et al., a large cohort of patients with pre-existing CVD was evaluated over a period of six years. The authors demonstrated that baseline TRAIL concentration was inversely and independently related to all-cause and CV mortality. Participants with the lowest levels of TRAIL (the first and second quartiles) had a three-fold increased risk of CV and all-cause death compared to those with the highest TRAIL levels [69]. Similar observations were made previously in smaller cohorts of patients with heart failure [66] and acute myocardial infarction [67]. In the aforementioned study by Volpato et al., those conclusions were extended to patients with a wider range of CV conditions. The authors, based on their analysis, even suggested the usefulness of TRAIL for predicting outcomes in patients with subclinical CV conditions.
The theories behind the protective effect of TRAIL on mortality risk in CVD are various depending on the etiology of the CV condition, which are included in the discussion below. The mechanism of TRAIL’s beneficial effect might be exerted either by a direct impact on cells or indirectly by modulating systemic immune functions. In vivo (rodent models) studies showed that recombinant TRAIL injections contributed to a slower progression of atherosclerosis and plaque stabilization mainly by the induction of the apoptosis of infiltrating macrophages [81]. TRAIL recruits activated leukocytes to particular tissue and initiates apoptosis to terminate the immune response. On the other hand, it is speculated that TRAIL exerts its protective role not by stimulating the extrinsic apoptotic pathway but by activation of the survival/proliferation pathways [82], as is the case in HF patients [83].
At the same time, when low levels of TRAIL are determining the survival rate, TRAIL-R2 concentration is reported also as having predictive value but with an inverse relationship [74]. The soluble form of TRAIL death receptor was identified as a prognostic marker in CVD by proteomic studies [74,79,80]. During three-years of follow-up, a high concentration of TRAIL-R2 independently predicted future CV events in patients with advanced atherosclerosis, even after adjusting for traditional CV risk factors [52]. A cross-sectional study in patients with lupus erythematosus showed that patients with a range of CVDs had markedly higher TRAIL-R2 concentration than those without [51]. Mattison et al. performed a genome-wide association study to determine the role of death receptor-activated apoptosis in CVD and investigated whether genetic variants of death receptors were linked to CVD risk [75]. Identification of several single-nucleotide polymorphisms (SNPs) associated with the level of plasma soluble form of death receptors remained significant even when the impact of other metabolic factors was considered. Genetic factors play a role in determining the concentrations of soluble death receptors in circulation. A weak association was identified between SNP in the gene region for TRAIL-R2 and coronary artery disease, but as the authors concluded, it is unlikely that soluble death receptors alone play a functional role in CVD [75]. It is probably the whole process of apoptosis that increases the risk.
Interestingly, in two studies, TRAIL, apart from showing a correlation with all-cause mortality, was correlated also with death from infectious causes [68,71]. That observation might be a result of the possible role of TRAIL and its death receptors in defending organisms from viral and bacterial infections. In experimental models, strong up-regulation of the TRAIL apoptotic pathway was observed in response to infection. As a consequence, virus- or bacteria-infected cells were eliminated efficiently by TRAIL-induced apoptosis [84,85]. These results from animal data correspond with reports showing an inverse TRAIL relation with mortality in patients with sepsis [86].
A lot of data come from the population of patients with chronic kidney disease (CKD) and its most pronounced manifestation—end-stage kidney disease. These patients have a high mortality rate and the main cause of death in this group is CVD. Apart from accelerated atherosclerosis, the CKD population is characterized by an inadequate inflammatory response. The investigations on the potential involvement of TRAIL, since it modulates both processes, are therefore highly interesting. Liabeuf at al. found that TRAIL was inversely associated with mortality risk in CKD patients [68]. However, further investigations focused on hemodialysis patients have yielded contradicting results [71,73]. The association of TRAIL with CV mortality in those patients was reported as non-significant [68,71,73], but interestingly, data from a proteomic study identified TRAIL-R2 as a protein associated with CV mortality in hemodialysis patients [79]. One thing that we must keep in mind when analyzing these studies is that patients with kidney failure have a unique risk factor profile and do not reflect the standard population.
Apart from predicting the outcome and therefore serving as a risk marker, TRAIL is considered to participate in the pathogenesis of various CVDs, including heart failure, atherosclerosis, coronary artery disease, atrial fibrillation (AF), and stroke. The discussion is ongoing about whether TRAIL and its death receptors act also as risk factors. Below, we summarize the knowledge on selected topics.

4.1. Heart Failure

Higher levels of TRAIL have been associated with better prognosis in HF patients both with reduced ejection fraction [66] and preserved ejection fraction [74]. Low levels of TRAIL in patients with HF were found to be an independent predictor of the risk of rehospitalization and death (in a 16-month follow-up study) [66]. The risk of death was confirmed in the same population in an extended follow-up of five years [72]. Data from animal models show that mechanical stretch (from mechanical overload) activated death receptor-mediated apoptotic signaling in cardiomyocytes [87]. These results suggest that activation of the TRAIL pathway has been implicated in the development and progression of heart failure, but the mechanism of cardioprotection exerted by TRAIL has not been yet fully elucidated. One suggestion is that higher levels of TRAIL possibly reflects the need for TRAIL-induced apoptosis to resolve inflammation [74], but data from an animal model actually suggests the opposite. The injection of recombinant TRAIL significantly reduced cardiac fibrosis and apoptosis and, as consequence, prevented more relevant cardiac structural changes in a mouse model of cardiomyopathy [13]. In this study, it was suggested that, contrary to its pro-apoptotic effects, it may be the result of triggering non-apoptotic signals in normal cells (promoting survival, migration and proliferation of primary vascular smooth muscles cells) [10,83]. However, in patients with Chagas cardiomyopathy serum, TRAIL levels were enhanced, correlated with decreased left ventricular ejection fraction and left ventricular diastolic dimension [23]. Similar results come from a study of men with nonischemic dilated cardiomyopathy, where the TRAIL concentration was also higher compared to the control group [22]. The prognostic value of these results and their agreement with data from HF patients is unknown.

4.2. Coronary Artery Disease

Emerging evidence has also demonstrated that TRAIL is involved in atherosclerosis development. However, the role of TRAIL in this process is equivocal. One study showed that TRAIL induces apoptosis and has pro-inflammatory effects in human endothelial cells [88]. On the other hand, several studies demonstrated possible anti-atherogenic and anti-inflammatory activity both in vitro and in vivo [15,81]. Cross-sectional and prospective human studies are also in agreement with this hypothesis [29,33,34]. Although we must remember that a cause-and-effect relationship cannot be concluded from observational studies, taking into consideration results from in vitro studies, there seems to be enough evidence to support the vasoprotective role of TRAIL. Potentially, since both endothelial cells and vascular smooth muscle cells express TRAIL receptors, TRAIL may be a molecule that can promote cell survival by activating intracellular signaling pathways, such as ERK/MAPK, Akt and NF-κB, which are known to promote survival and proliferation [10,82].
Few studies have focused on diabetic patients. Metabolic factors are involved in the vascular injury that accelerates the development of atherosclerosis [89]; therefore, there is a higher incidence of CVDs in diabetic patients. A hypothesis has been formed that the higher incidence of CVDs in diabetic patients is related to increased death-receptor activated apoptosis in the CV system in response to metabolic stress [75]. Kawano et al. tried to examine the association of TRAIL with atherosclerosis (the surrogate marker measured was intima-media thickness) in patients with type 2 diabetes but without any symptoms of coronary artery disease and HF [29]. The study showed the correlation between TRAIL and intima-media thickness only in a subset of diabetic patients with macrovascular disease where TRAIL was significantly and inversely correlated with carotid intima-media thickness. The authors concluded that TRAIL might not be helpful in the detection of early-stage atherosclerotic lesions.
Several studies showed that the levels of TRAIL are decreased in patients with coronary artery disease but with different significance. A cross-sectional study in patients undergoing angiography reported a borderline reduction of TRAIL levels compared to healthy controls [32]. Yet, another study found that circulating TRAIL was substantially lower in patients with acute coronary syndrome than those with stable angina and healthy subjects [56]. These differences in the impact of TRAIL might be explained by another study, which found that TRAIL can be a potential marker of the severity of coronary artery disease. Serum TRAIL levels were not only significantly lower in patients with coronary artery disease but also inversely associated with the number of diseased vessels independent of other coronary risk factors [34]. Secchiero et al. reported that in the acute phase of myocardial infarction serum concentration of TRAIL was acutely decreased and tended to increase in following days [67]. After 6 to 12 months of follow-up, its level was not significantly different from those of healthy control subjects. TRAIL was also inversely correlated with biochemical markers of myocardial injury (creatinine kinase and creatinine kinase-MB fraction) and cardiac dysfunction (B-type natriuretic peptide). Moreover, TRAIL predicted not only in-hospital and long-term mortality (follow-up of 12 months) but also the incidence of heart failure [67]. Thus, low TRAIL levels at discharge represent a possible predictor of future CV events following acute myocardial infarction.
Although myocardial infarction is a known cause of the release of apoptotic biomarkers, explanations for low levels of TRAIL in acute coronary syndrome are inconsistent. The most commonly proposed reason is its consumption of the atherosclerotic plaques. Other reasons may include the simultaneous increase in OPG, which is a decoy receptor and may bind TRAIL in the acute phase of myocardial infarction and increase in metalloproteinase-2, which is also released following acute myocardial infarction and is responsible for inducing TRAIL cleavage [62]. Additionally, transcription factor Egr-1, upregulated during atherosclerosis, is known to downregulate TRAIL expression in endothelial cells [90]. Thus, the hypotheses about TRAIL consumption into the plaques or reduction in its production need to be further pursued and clarified.

4.3. Atrial Fibrillation

AF promotes tissue fibrosis [91], which is an important contributor of AF recurrences, resistance to therapy, and complications [92]. Cardiac remodeling is considered a reparative process of replacing degenerative or dead myocytes. Atrial fibrosis involves processes of necrosis and apoptosis, hence its pathophysiological link to TRAIL. In agreement with this are reports that the restoration of sinus rhythm in patients with AF is associated with a decrease in the serum concentration of TRAIL [24,27]. On the other hand, published studies did not confirm the prognostic utility of TRAIL in the prediction of AF occurrence in patients with sinus rhythm [26] and the restoration of sinus rhythm in patients with AF treated pharmacologically [27] or with electrical cardioversion [25]. Deftereos et al. reported that TRAIL concentration shows transcardiac gradient (coronary sinus concentration minus aortic root concentration) in patients with AF after electrical cardioversion and in their half-year follow-up transcardiac gradient, but not TRAIL concentration alone, was inversely correlated with recurrence of AF [25]. Based on these results, the hypothesis of cardiac TRAIL production was made. Proteomic studies have also identified TRAIL-R2 as a risk factor for AF [28].

4.4. Cerebral Ischemia

Since the involvement of TRAIL in atherosclerosis was proven, further studies on its relevance in cerebrovascular diseases were conducted. The abundance of studies on the involvement of TRAIL in atherosclerosis has led to further research on its relevance in cerebrovascular disorders. TRAIL detection in cases of brain ischemia can be explained not only by atherosclerosis; TRAIL in the brain can be released by glia [93], injured neurons, and also circulating leukocytes [94]. Moreover, data from animal models showed TRAIL expression in ischemic areas of the brain, suggesting its potency in inducing death in neurons [16]. Kang et al. analyzed the serum levels of TRAIL in patients with acute ischemic stroke. In these patients, low levels of TRAIL were associated with ischemic stroke severity and stroke volume assessed by imaging [45]. This is in agreement with another study where TRAIL concentration in patients with large-artery atherosclerotic stroke was significantly lower compared to healthy controls. In a three-month follow-up, levels of TRAIL were found to be negatively correlated with prognosis measured by the modified Rankin Scale [46]. It is suggested from in vivo studies that blocking the TRAIL apoptosis pathway may be effective in preventing neuronal death in patients with stroke [95].

5. Conclusions

Considering all of these data together, the TRAIL pathway system undoubtedly has gathered attention as a player in increased CV risk. Its role has been investigated in various types of CVDs. The evidence from most of them associate reduced TRAIL and increased TRAIL-R2 concentration with many forms of heart disorders. It is, however, unclear whether low TRAIL levels should be considered as a risk factor rather than a risk marker of CVD. Understanding the association between CVD and TRAIL has important implications for disease management such as targeted therapies or prognosis. Since human-recombinant TRAIL is being tested in cancer studies, it is possible to apply this therapy to another field. To do so, further work is needed to better define the role of TRAIL in CVDs.

Author Contributions

Conceptualizations, K.K., A.R.-R. and T.H.; methodology, A.R.-R.; software, T.H.; validation, K.K., A.R.-R. and E.Z.; formal analysis, K.K., A.R.-R. and T.H.; investigation, K.K.; resources, K.K. and A.R.-R.; data curation, T.H.; writing—original draft preparation, K.K. writing—review and editing, A.R.-R., E.Z. and T.H.; visualization, K.K. and A.R.-R.; supervision, T.H.; project administration, A.R.-R. and T.H.; funding acquisition, T.H. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

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

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Kerr, J.F.R.; Wyllie, A.H.; Currie, A.R. Apoptosis: A Basic Biological Phenomenon with Wideranging Implications in Tissue Kinetics. Br. J. Cancer 1972, 26, 239–257. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  2. Diamantis, A.; Magiorkinis, E.; Sakorafas, G.H.; Androutsos, G. A Brief History of Apoptosis: From Ancient to Modern Times. Oncol. Res. Treat. 2008, 31, 702–706. [Google Scholar] [CrossRef] [PubMed]
  3. Cotter, T.G. Apoptosis and cancer: The genesis of a research field. Nat. Rev. Cancer 2009, 9, 501–507. [Google Scholar] [CrossRef] [PubMed]
  4. Sawa, A.; Wiegand, G.W.; Cooper, J.K.; Margolis, R.L.; Sharp, A.H.; Lawler, J.F.; Greenamyre, J.T.; Snyder, S.H.; Ross, C.A. Increased apoptosis of Huntington disease lymphoblasts associated with repeat length-dependent mitochondrial depolarization. Nat. Med. 1999, 5, 1194–1198. [Google Scholar] [CrossRef]
  5. Saraste, A.; Pulkki, K.; Kallajoki, M.; Henriksen, K.; Parvinen, M.; Voipio-Pulkki, L.-M. Apoptosis in Human Acute Myocardial Infarction. Circulation 1997, 95, 320–323. [Google Scholar] [CrossRef]
  6. Wiley, S.R.; Schooley, K.; Smolak, P.J.; Din, W.S.; Huang, C.-P.; Nicholl, J.K.; Sutherland, G.R.; Smith, T.D.; Rauch, C.; Smith, C.A.; et al. Identification and characterization of a new member of the TNF family that induces apoptosis. Immunity 1995, 3, 673–682. [Google Scholar] [CrossRef] [Green Version]
  7. Tecchio, C.; Huber, V.; Scapini, P.; Calzetti, F.; Margotto, D.; Todeschini, G.; Pilla, L.; Martinelli, G.; Pizzolo, G.; Rivoltini, L.; et al. IFNα-stimulated neutrophils and monocytes release a soluble form of TNF-related apoptosis-inducing ligand (TRAIL/Apo-2 ligand) displaying apoptotic activity on leukemic cells. Blood 2004, 103, 3837–3844. [Google Scholar] [CrossRef] [Green Version]
  8. Kimberley, F.C.; Screaton, G.R. Following a TRAIL: Update on a ligand and its five receptors. Cell Res. 2004, 14, 359–372. [Google Scholar] [CrossRef]
  9. Stuckey, D.W.; Shah, K. TRAIL on trial: Preclinical advances in cancer therapy. Trends Mol. Med. 2013, 19, 685–694. [Google Scholar] [CrossRef] [Green Version]
  10. Secchiero, P.; Zerbinati, C.; Rimondi, E.; Corallini, F.; Milani, D.; Grill, V.; Forti, G.; Capitani, S.; Zauli, G. TRAIL promotes the survival, migration and proliferation of vascular smooth muscle cells. Cell. Mol. Life Sci. 2004, 61, 1965–1974. [Google Scholar] [CrossRef]
  11. Spierings, D.C.; De Vries, E.G.; Vellenga, E.; Heuvel, F.A.V.D.; Koornstra, J.J.; Wesseling, J.; Hollema, H.; De Jong, S. Tissue distribution of the death ligand TRAIL and its receptors. J. Histochem. Cytochem. 2004, 52, 821–831. [Google Scholar] [CrossRef] [Green Version]
  12. Tanner, M.A.; Thomas, T.P.; Grisanti, L.A. Death receptor 5 contributes to cardiomyocyte hypertrophy through epidermal growth factor receptor transactivation. J. Mol. Cell. Cardiol. 2019, 136, 1–14. [Google Scholar] [CrossRef] [PubMed]
  13. Toffoli, B.; Bernardi, S.; Candido, R.; Zacchigna, S.; Fabris, B.; Secchiero, P. TRAIL shows potential cardioprotective activity. Investig. New Drugs 2011, 30, 1257–1260. [Google Scholar] [CrossRef]
  14. Di Bartolo, B.A.; Cartland, S.P.; Harith, H.H.; Bobryshev, Y.V.; Schoppet, M.; Kavurma, M.M. TRAIL-Deficiency Accelerates Vascular Calcification in Atherosclerosis via Modulation of RANKL. PLoS ONE 2013, 8, e74211. [Google Scholar] [CrossRef]
  15. Watt, V.; Chamberlain, J.; Steiner, T.; Francis, S.; Crossman, D. TRAIL attenuates the development of atherosclerosis in apolipoprotein E deficient mice. Atherosclerosis 2011, 215, 348–354. [Google Scholar] [CrossRef] [PubMed]
  16. Martin-Villalba, A.; Herr, I.; Jeremias, I.; Hahne, M.; Brandt, R.; Vogel, J.; Schenkel, J.; Herdegen, T.; Debatin, K.-M. CD95 Ligand (Fas-L/APO-1L) and Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand Mediate Ischemia-Induced Apoptosis in Neurons. J. Neurosci. 1999, 19, 3809–3817. [Google Scholar] [CrossRef] [Green Version]
  17. Hameed, A.G.; Arnold, N.D.; Chamberlain, J.; Pickworth, J.A.; Paiva, C.; Dawson, S.; Cross, S.; Long, L.; Zhao, L.; Morrell, N.W.; et al. Inhibition of tumor necrosis factor–related apoptosis-inducing ligand (TRAIL) reverses experimental pulmonary hypertension. J. Exp. Med. 2012, 209, 1919–1935. [Google Scholar] [CrossRef] [PubMed]
  18. Bumdelger, B.; Kokubo, H.; Kamata, R.; Fujii, M.; Yoshimura, K.; Aoki, H.; Orita, Y.; Ishida, T.; Ohtaki, M.; Nagao, M.; et al. Osteoprotegerin Prevents Development of Abdominal Aortic Aneurysms. PLoS ONE 2016, 11, e0147088. [Google Scholar] [CrossRef] [PubMed]
  19. Venuraju, S.M.; Yerramasu, A.; Corder, R.; Lahiri, A. Osteoprotegerin as a Predictor of Coronary Artery Disease and Cardiovascular Mortality and Morbidity. J. Am. Coll. Cardiol. 2010, 55, 2049–2061. [Google Scholar] [CrossRef] [Green Version]
  20. Osmančík, P.; Heřman, D.; Stros, P.; Línková, H.; Vondrak, K.; Paskova, E. Changes and Prognostic Impact of Apoptotic and Inflammatory Cytokines in Patients Treated with Cardiac Resynchronization Therapy. Cardiology 2013, 124, 190–198. [Google Scholar] [CrossRef]
  21. Stenemo, M.; Nowak, C.; Byberg, L.; Sundström, J.; Giedraitis, V.; Lind, L.; Ingelsson, E.; Fall, T.; Ärnlöv, J. Circulating proteins as predictors of incident heart failure in the elderly. Eur. J. Heart Fail. 2017, 20, 55–62. [Google Scholar] [CrossRef] [PubMed]
  22. Schoppet, M.; Ruppert, V.; Hofbauer, L.C.; Henser, S.; Al-Fakhri, N.; Christ, M.; Pankuweit, S.; Maisch, B. TNF-related apoptosis-inducing ligand and its decoy receptor osteoprotegerin in nonischemic dilated cardiomyopathy. Biochem. Biophys. Res. Commun. 2005, 338, 1745–1750. [Google Scholar] [CrossRef] [PubMed]
  23. Lula, J.F.; Rocha, M.O.D.C.; Nunes, M.D.C.P.; Ribeiro, A.L.P.; Teixeira, M.M.; Bahia, M.T.; Talvani, A. Plasma concentrations of tumour necrosis factor-alpha, tumour necrosis factor-related apoptosis-inducing ligand, and FasLigand/CD95L in patients with Chagas cardiomyopathy correlate with left ventricular dysfunction. Eur. J. Heart Fail. 2009, 11, 825–831. [Google Scholar] [CrossRef] [PubMed]
  24. Osmančík, P.; Peroutka, Z.; Budera, P.; Heřman, D.; Stros, P.; Straka, Z.; Vondrak, K. Decreased Apoptosis following Successful Ablation of Atrial Fibrillation. Cardiology 2010, 116, 302–307. [Google Scholar] [CrossRef]
  25. Deftereos, S.; Giannopoulos, G.; Kossyvakis, C.; Raisakis, K.; Angelidis, C.; Efremidis, M.; Panagopoulou, V.; Kaoukis, A.; Theodorakis, A.; Toli, K.; et al. Association of post-cardioversion transcardiac concentration gradient of soluble tumor necrosis factor-related apoptosis-inducing ligand (sTRAIL) and inflammatory biomarkers to atrial fibrillation recurrence. Clin. Biochem. 2013, 46, 1020–1025. [Google Scholar] [CrossRef]
  26. Müller, P.; Deneke, T.; Schiedat, F.; Bösche, L.; Strauch, J.; Dietrich, J.W.; Vogt, M.; Tannapfel, A.; Stiegler, H.; Mügge, A.; et al. Increased Preoperative Serum Apoptosis Marker Fas Ligand Correlates with Histopathology and New-Onset of Atrial Fibrillation in Patients After Cardiac Surgery. J. Cardiovasc. Electrophysiol. 2013, 24, 1110–1115. [Google Scholar] [CrossRef]
  27. Rewiuk, K.; Grodzicki, T. Osteoprotegerin and TRAIL in Acute Onset of Atrial Fibrillation. BioMed. Res. Int. 2015, 2015, 1–9. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Chua, W.; Purmah, Y.; Cardoso, V.R.; Gkoutos, G.V.; Tull, S.P.; Neculau, G.; Thomas, M.R.; Kotecha, D.; Lip, G.Y.H.; Kirchhof, P.; et al. Data-driven discovery and validation of circulating blood-based biomarkers associated with prevalent atrial fibrillation. Eur. Heart J. 2019, 40, 1268–1276. [Google Scholar] [CrossRef] [Green Version]
  29. Kawano, N.; Mori, K.; Emoto, M.; Lee, E.; Kobayashi, I.; Yamazaki, Y.; Urata, H.; Morioka, T.; Koyama, H.; Shoji, T.; et al. Association of serum TRAIL levels with atherosclerosis in patients with type 2 diabetes mellitus. Diabetes Res. Clin. Pr. 2011, 91, 316–320. [Google Scholar] [CrossRef]
  30. Deftereos, S.; Giannopoulos, G.; Kossyvakis, C.; Kaoukis, A.; Raisakis, K.; Panagopoulou, V.; Miliou, A.; Theodorakis, A.; Driva, M.; Pyrgakis, V.; et al. Association of soluble tumour necrosis factor-related apoptosis-inducing ligand levels with coronary plaque burden and composition. Heart 2011, 98, 214–218. [Google Scholar] [CrossRef]
  31. Arcidiacono, M.V.; Rimondi, E.; Maietti, E.; Melloni, E.; Tisato, V.; Gallo, S.; Valdivielso, J.M.; Fernández, E.; Betriu, À.; Voltan, R.; et al. Relationship between low levels of circulating TRAIL and atheromatosis progression in patients with chronic kidney disease. PLoS ONE 2018, 13, e0203716. [Google Scholar] [CrossRef]
  32. Schoppet, M.; Sattler, A.M.; Schaefer, J.R.; Hofbauer, L.C. Osteoprotegerin (OPG) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) levels in atherosclerosis. Atherosclerosis 2006, 184, 446–447. [Google Scholar] [CrossRef]
  33. Satoh, D.; Inami, N.; Shimazu, T.; Kajiura, T.; Yamada, K.; Iwasaka, T.; Nomura, S. Soluble TRAIL prevents RANTES-dependent restenosis after percutaneous coronary intervention in patients with coronary artery disease. J. Thromb. Thrombolysis 2009, 29, 471–476. [Google Scholar] [CrossRef]
  34. Mori, K.; Ikari, Y.; Jono, S.; Shioi, A.; Ishimura, E.; Emoto, M.; Inaba, M.; Hara, K.; Nishizawa, Y. Association of serum TRAIL level with coronary artery disease. Thromb. Res. 2010, 125, 322–325. [Google Scholar] [CrossRef]
  35. Secchiero, P.; Corallini, F.; Beltrami, A.P.; Ceconi, C.; Bonasia, V.; Di Chiara, A.; Ferrari, R.; Zauli, G. An imbalanced OPG/TRAIL ratio is associated to severe acute myocardial infarction. Atherosclerosis 2010, 210, 274–277. [Google Scholar] [CrossRef] [PubMed]
  36. Shaker, O.G.; El-Shehaby, A.; Nabih, M. Possible Role of Osteoprotegerin and Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand as Markers of Plaque Instability in Coronary Artery Disease. Angiology 2010, 61, 756–762. [Google Scholar] [CrossRef] [PubMed]
  37. Deftereos, S.; Giannopoulos, G.; Panagopoulou, V.; Raisakis, K.; Kossyvakis, C.; Kaoukis, A.; Tzalamouras, V.; Mavri, M.; Pyrgakis, V.; Cleman, M.W.; et al. Inverse Association of Coronary Soluble Tumor Necrosis Factor-Related Apoptosis Inducing Ligand (sTRAIL) Levels to In-Stent Neointimal Hyperplasia. Cardiology 2012, 123, 97–102. [Google Scholar] [CrossRef] [PubMed]
  38. Song, S.; Choi, K.; Kwon, K.; Choi, C. Soluble tumor necrosis factor-related apoptosis-inducing ligand after percutaneous coronary intervention: A potential biomarker for vascular remodeling. J. Cardiovasc. Med. 2012, 13, 292–293. [Google Scholar] [CrossRef]
  39. Luz, A.; Santos, M.; Magalhães, R.; Oliveira, J.C.; Pacheco, A.; Silveira, J.; Cabral, S.; Torres, S.; Leite-Moreira, A.F.; Carvalho, H. Soluble TNF-related apoptosis induced ligand (sTRAIL) is augmented by Post-Conditioning and correlates to infarct size and left ventricle dysfunction in STEMI patients: A substudy from a randomized clinical trial. Heart Vessel. 2016, 32, 117–125. [Google Scholar] [CrossRef] [PubMed]
  40. Cholan, P.M.; Cartland, S.P.; Dang, L.; Rayner, B.S.; Patel, S.; Thomas, S.R.; Kavurma, M.M. TRAIL protects against endothelial dysfunction in vivo and inhibits angiotensin-II-induced oxidative stress in vascular endothelial cells in vitro. Free. Radic. Biol. Med. 2018, 126, 341–349. [Google Scholar] [CrossRef]
  41. Teringova, E.; Kozel, M.; Knot, J.; Kocka, V.; Benesova, K.; Tousek, P. Relationship between TRAIL and Left Ventricular Ejection Fraction in Patients with ST-Elevation Myocardial Infarction Treated with Primary Percutaneous Coronary Intervention. BioMed Res. Int. 2018, 2018, 1–8. [Google Scholar] [CrossRef] [Green Version]
  42. Chasseraud, M.; Liabeuf, S.; Mozar, A.; Mentaveri, R.; Brazier, M.; Massy, Z.A.; Kamel, S. Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand and Vascular Calcification. Ther. Apher. Dial. 2011, 15, 140–146. [Google Scholar] [CrossRef] [PubMed]
  43. Moon, A.R.; Park, Y.; Chang, J.H.; Lee, S.S. Inverse regulation of serum osteoprotegerin and tumor necrosis factor-related apoptosis-inducing ligand levels in patients with leg lesional vascular calcification. Medicine 2019, 98, e14489. [Google Scholar] [CrossRef] [PubMed]
  44. Sarchielli, P.; Nardi, K.; Chiasserini, D.; Eusebi, P.; Tantucci, M.; Di Piero, V.; Altieri, M.; Marini, C.; Russo, T.; Silvestrini, M.; et al. Immunological Profile of Silent Brain Infarction and Lacunar Stroke. PLoS ONE 2013, 8, e68428. [Google Scholar] [CrossRef]
  45. Kang, Y.H.; Park, M.-G.; Noh, K.-H.; Park, H.R.; Lee, H.W.; Son, S.M.; Park, K.-P. Low serum TNF-related apoptosis-inducing ligand (TRAIL) levels are associated with acute ischemic stroke severity. Atherosclerosis 2015, 240, 228–233. [Google Scholar] [CrossRef]
  46. Pan, X.; Pang, M.; Ma, A.; Wang, K.; Zhang, Z.; Zhong, Q.; Yang, S. Association of TRAIL and Its Receptors with Large-Artery Atherosclerotic Stroke. PLoS ONE 2015, 10, e0136414. [Google Scholar] [CrossRef] [Green Version]
  47. O’Sullivan, E.P.; Ashley, D.T.; Davenport, C.; Kelly, J.; Devlin, N.; Crowley, R.; Leahy, A.L.; Kelly, C.J.; Agha, A.; Thompson, C.J.; et al. Osteoprotegerin is higher in peripheral arterial disease regardless of glycaemic status. Thromb. Res. 2010, 126, e423–e427. [Google Scholar] [CrossRef]
  48. Karatolios, K.; Pankuweit, S.; Goettsch, C.; Hofbauer, L.C.; Timmesfeld, N.; Al-Fakhri, N.; Maisch, B.; Schoppet, M. Osteoprotegerin (OPG) and TNF-related apoptosis-inducing ligand (TRAIL) levels in malignant and benign pericardial effusions. Clin. Biochem. 2012, 45, 237–242. [Google Scholar] [CrossRef] [PubMed]
  49. Zhou, C.; Long, Y.; Yang, H.; Zhu, C.; Ma, Q.; Zhang, Y. TRAIL Is Decreased Before 20 Weeks Gestation in Women with Hypertensive Disorders of Pregnancy. PLoS ONE 2015, 10, e0128425. [Google Scholar] [CrossRef] [Green Version]
  50. Liu, H.; Yang, E.; Lu, X.; Zuo, C.; He, Y.; Jia, D.; Zhu, Q.; Yu, Y.; Lv, A. Serum levels of tumor necrosis factor-related apoptosis-inducing ligand correlate with the severity of pulmonary hypertension. Pulm. Pharmacol. Ther. 2015, 33, 39–46. [Google Scholar] [CrossRef]
  51. Wigren, M.; Svenungsson, E.; Mattisson, I.Y.; Gustafsson, J.T.; Gunnarsson, I.; Zickert, A.; Elvin, K.; Jensen-Urstad, K.; Bengtsson, A.; Gullstrand, B.; et al. Cardiovascular disease in systemic lupus erythematosus is associated with increased levels of biomarkers reflecting receptor-activated apoptosis. Atherosclerosis 2018, 270, 1–7. [Google Scholar] [CrossRef] [PubMed]
  52. Gonçalves, I.; Singh, P.; Tengryd, C.; Cavalera, M.; Mattisson, I.Y.; Nitulescu, M.; Persson, A.F.; Volkov, P.; Engström, G.; Orho-Melander, M.; et al. sTRAIL-R2 (Soluble TNF [Tumor Necrosis Factor]-Related Apoptosis-Inducing Ligand Receptor 2) a Marker of Plaque Cell Apoptosis and Cardiovascular Events. Stroke 2019, 50, 1989–1996. [Google Scholar] [CrossRef] [PubMed]
  53. Yndestad, A.; Damås, J.K.; Eiken, H.G.; Holm, T.; Haug, T.; Simonsen, S.; Frøland, S.S.; Gullestad, L.; Aukrust, P. Increased gene expression of tumor necrosis factor superfamily ligands in peripheral blood mononuclear cells during chronic heart failure. Cardiovasc. Res. 2002, 54, 175–182. [Google Scholar] [CrossRef] [Green Version]
  54. Cao, H.; Wang, J.; Xi, L.; Røe, O.D.; Chen, Y.; Wang, N. Dysregulated atrial gene expression of osteoprotegerin/receptor activator of nuclear factor-?B (RANK)/RANK ligand axis in the development and progression of atrial fibrillation. Circ. J. 2011, 75, 2781–2788. [Google Scholar] [CrossRef] [Green Version]
  55. Schoppet, M.; Al-Fakhri, N.; Franke, F.E.; Katz, N.; Barth, P.J.; Maisch, B.; Preissner, K.T.; Hofbauer, L.C. Localization of Osteoprotegerin, Tumor Necrosis Factor-Related Apoptosis-Inducing Ligand, and Receptor Activator of Nuclear Factor-κB Ligand in Mönckeberg’s Sclerosis and Atherosclerosis. J. Clin. Endocrinol. Metab. 2004, 89, 4104–4112. [Google Scholar] [CrossRef] [Green Version]
  56. Michowitz, Y.; Goldstein, E.; Roth, A.; Afek, A.; Abashidze, A.; Ben Gal, Y.; Keren, G.; George, J. The involvement of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) in atherosclerosis. J. Am. Coll. Cardiol. 2005, 45, 1018–1024. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Niessner, A.; Sato, K.; Chaikof, E.L.; Colmegna, I.; Goronzy, J.J.; Weyand, C.M. Pathogen-Sensing Plasmacytoid Dendritic Cells Stimulate Cytotoxic T-Cell Function in the Atherosclerotic Plaque Through Interferon-α. Circulation 2006, 114, 2482–2489. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Nakajima, H.; Yanase, N.; Oshima, K.; Sasame, A.; Hara, T.; Fukazawa, S.; Takata, R.; Hata, K.; Mukai, K.; Yamashina, A.; et al. Enhanced Expression of the Apoptosis Inducing Ligand TRAIL in Mononuclear Cells After Myocardial Infarction. Jpn. Heart J. 2003, 44, 833–844. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Sato, K.; Niessner, A.; Kopecky, S.L.; Frye, R.L.; Goronzy, J.J.; Weyand, C.M. TRAIL-expressing T cells induce apoptosis of vascular smooth muscle cells in the atherosclerotic plaque. J. Exp. Med. 2006, 203, 239–250. [Google Scholar] [CrossRef]
  60. Corallini, F.; Secchiero, P.; Beltrami, A.P.; Cesselli, D.; Puppato, E.; Ferrari, R.; Beltrami, C.A.; Zauli, G. TNF-α modulates the migratory response of mesenchymal stem cells to TRAIL. Cell. Mol. Life Sci. 2010, 67, 1307–1314. [Google Scholar] [CrossRef] [PubMed]
  61. Sato, K.; Nuki, T.; Gomita, K.; Weyand, C.M.; Hagiwara, N. Statins reduce endothelial cell apoptosis via inhibition of TRAIL expression on activated CD4 T cells in acute coronary syndrome. Atherosclerosis 2010, 213, 33–39. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Secchiero, P.; Gonelli, A.; Corallini, F.; Ceconi, C.; Ferrari, R.; Zauli, G. Metalloproteinase 2 cleaves in vitro recombinant TRAIL: Potential implications for the decreased serum levels of TRAIL after acute myocardial infarction. Atherosclerosis 2010, 211, 333–336. [Google Scholar] [CrossRef] [PubMed]
  63. Liu, X.; Winrow, V.R.; Horrocks, M.; Stevens, C.R. Differential expression of TRAIL and its receptors relative to calcification in AAA. Biochem. Biophys. Res. Commun. 2007, 358, 18–23. [Google Scholar] [CrossRef] [PubMed]
  64. Galeone, A.; Brunetti, G.; Oranger, A.; Greco, G.; Di Benedetto, A.; Mori, G.; Colucci, S.; Zallone, A.; Paparella, D.; Grano, M. Aortic valvular interstitial cells apoptosis and calcification are mediated by TNF-related apoptosis-inducing ligand. Int. J. Cardiol. 2013, 169, 296–304. [Google Scholar] [CrossRef]
  65. Tisato, V.; Secchiero, P.; Rimondi, E.; Gianesini, S.; Menegatti, E.; Casciano, F.; Zamboni, P.; Zauli, G. GM-CSF Exhibits Anti-Inflammatory Activity on Endothelial Cells Derived from Chronic Venous Disease Patients. Mediat. Inflamm. 2013, 2013, 1–9. [Google Scholar] [CrossRef]
  66. Niessner, A.; Hohensinner, P.J.; Rychli, K.; Neuhold, S.; Zorn, G.; Richter, B.; Hülsmann, M.; Berger, R.; Mörtl, D.; Huber, K.; et al. Prognostic value of apoptosis markers in advanced heart failure patients. Eur. Heart J. 2008, 30, 789–796. [Google Scholar] [CrossRef] [Green Version]
  67. Secchiero, P.; Corallini, F.; Ceconi, C.; Parrinello, G.; Volpato, S.; Ferrari, R.; Zauli, G. Potential Prognostic Significance of Decreased Serum Levels of TRAIL after Acute Myocardial Infarction. PLoS ONE 2009, 4, e4442. [Google Scholar] [CrossRef]
  68. Liabeuf, S.; Barreto, D.V.; Barreto, F.C.; Chasseraud, M.; Brazier, M.; Choukroun, G.; Kamel, S.; Massy, Z.A. The circulating soluble TRAIL is a negative marker for inflammation inversely associated with the mortality risk in chronic kidney disease patients. Nephrol. Dial. Transpl. 2010, 25, 2596–2602. [Google Scholar] [CrossRef] [Green Version]
  69. Volpato, S.; Ferrucci, L.; Secchiero, P.; Corallini, F.; Zuliani, G.; Fellin, R.; Guralnik, J.M.; Bandinelli, S.; Zauli, G. Association of tumor necrosis factor-related apoptosis-inducing ligand with total and cardiovascular mortality in older adults. Atherosclerosis 2011, 215, 452–458. [Google Scholar] [CrossRef] [Green Version]
  70. Osmancik, P.; Teringova, E.; Toušek, P.; Paulu, P.; Widimsky, P. Prognostic Value of TNF-Related Apoptosis Inducing Ligand (TRAIL) in Acute Coronary Syndrome Patients. PLoS ONE 2013, 8, e53860. [Google Scholar] [CrossRef] [Green Version]
  71. Mori, K.; Okuno, S.; Shoji, T.; Emoto, M.; Kakutani, Y.; Yamakawa, K.; Imanishi, Y.; Ishimura, E.; Yamakawa, T.; Shoji, S.; et al. Tumor necrosis factor-related apoptosis-inducing ligand as an independent predictor of mortality in hemodialysis patents. Cytokine 2013, 61, 912–916. [Google Scholar] [CrossRef]
  72. Richter, B.; Koller, L.; Hohensinner, P.J.; Zorn, G.; Brekalo, M.; Berger, R.; Mörtl, D.; Maurer, G.; Pacher, R.; Huber, K.; et al. A multi-biomarker risk score improves prediction of long-term mortality in patients with advanced heart failure. Int. J. Cardiol. 2013, 168, 1251–1257. [Google Scholar] [CrossRef] [PubMed]
  73. Kuzniewski, M.; Fedak, D.; Dumnicka, P.; Stępień, E.; Kusnierz-Cabala, B.; Cwynar, M.; Sułowicz, W. Osteoprotegerin and osteoprotegerin/TRAIL ratio are associated with cardiovascular dysfunction and mortality among patients with renal failure. Adv. Med. Sci. 2016, 61, 269–275. [Google Scholar] [CrossRef] [PubMed]
  74. Hage, C.; Michaëlsson, E.; Linde, C.; Donal, E.; Daubert, J.-C.; Gan, L.-M.; Lund, L.H. Inflammatory Biomarkers Predict Heart Failure Severity and Prognosis in Patients with Heart Failure with Preserved Ejection Fraction. Circ. Cardiovasc. Genet. 2017, 10, e001633. [Google Scholar] [CrossRef] [Green Version]
  75. Mattisson, I.Y.; Björkbacka, H.; Wigren, M.; Edsfeldt, A.; Melander, O.; Fredrikson, G.N.; Bengtsson, E.; Gonçalves, I.; Orho-Melander, M.; Engström, G.; et al. Elevated Markers of Death Receptor-Activated Apoptosis are Associated with Increased Risk for Development of Diabetes and Cardiovascular Disease. EBioMedicine 2017, 26, 187–197. [Google Scholar] [CrossRef] [Green Version]
  76. Skau, E.; Henriksen, E.; Wagner, P.; Hedberg, P.; Siegbahn, A.; Leppert, J. GDF-15 and TRAIL-R2 are powerful predictors of long-term mortality in patients with acute myocardial infarction. Eur. J. Prev. Cardiol. 2017, 24, 1576–1583. [Google Scholar] [CrossRef]
  77. Ajala, O.; Zhang, Y.; Gupta, A.; Bon, J.; Sciurba, F.; Chandra, D. Decreased serum TRAIL is associated with increased mortality in smokers with comorbid emphysema and coronary artery disease. Respir. Med. 2018, 145, 21–27. [Google Scholar] [CrossRef]
  78. Nowak, C.; Carlsson, A.C.; Östgren, C.J.; Nyström, F.H.; Alam, M.; Feldreich, T.; Sundström, J.; Carrero, J.-J.; Leppert, J.; Hedberg, P.; et al. Multiplex proteomics for prediction of major cardiovascular events in type 2 diabetes. Diabetologia 2018, 61, 1748–1757. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Feldreich, T.; Nowak, C.; Fall, T.; Carlsson, A.C.; Carrero, J.-J.; Ripsweden, J.; Qureshi, A.R.; Heimbürger, O.; Barany, P.; Stenvinkel, P.; et al. Circulating proteins as predictors of cardiovascular mortality in end-stage renal disease. J. Nephrol. 2019, 32, 111–119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Ferreira, J.P.; Sharma, A.; Mehta, C.; Bakris, G.; Rossignol, P.; White, W.B.; Zannad, F. Multi-proteomic approach to predict specific cardiovascular events in patients with diabetes and myocardial infarction: Findings from the EXAMINE trial. Clin. Res. Cardiol. 2020, 1–14. [Google Scholar] [CrossRef] [PubMed]
  81. Secchiero, P.; Candido, R.; Corallini, F.; Zacchigna, S.; Toffoli, B.; Rimondi, E.; Fabris, B.; Giacca, M.; Zauli, G. Systemic Tumor Necrosis Factor–Related Apoptosis-Inducing Ligand Delivery Shows Antiatherosclerotic Activity in Apolipoprotein E–Null Diabetic Mice. Circulation 2006, 114, 1522–1530. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Bernardi, S.; Bossi, F.; Toffoli, B.; Fabris, B. Roles and Clinical Applications of OPG and TRAIL as Biomarkers in Cardiovascular Disease. BioMed. Res. Int. 2016, 2016, 1–12. [Google Scholar] [CrossRef] [PubMed]
  83. Di Pietro, R.; Zauli, G. Emerging non-apoptotic functions of tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)/Apo2L. J. Cell. Physiol. 2004, 201, 331–340. [Google Scholar] [CrossRef] [PubMed]
  84. Falschlehner, C.; Schaefer, U.; Walczak, H. Following TRAIL’s path in the immune system. Immunology 2009, 127, 145–154. [Google Scholar] [CrossRef] [PubMed]
  85. Cziupka, K.; Busemann, A.; Partecke, L.I.; Pötschke, C.; Rath, M.; Traeger, T.; Koerner, P.; Von Bernstorff, W.; Kessler, W.; Diedrich, S.; et al. Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) improves the innate immune response and enhances survival in murine polymicrobial sepsis. Crit. Care Med. 2010, 38, 2169–2174. [Google Scholar] [CrossRef] [PubMed]
  86. Tian, Y.; Tao, T.; Zhu, J.; Zou, Y.; Wang, J.; Li, J.; Bo, L.; Deng, X. Soluble Tumor Necrosis Factor Related Apoptosis Inducing Ligand Level as a Predictor of Severity of Sepsis and the Risk of Mortality in Septic Patients. PLoS ONE 2013, 8, e82204. [Google Scholar] [CrossRef] [Green Version]
  87. Liao, X.; Wang, X.; Gu, Y.; Chen, Q.; Chen, L.-Y. Involvement of death receptor signaling in mechanical stretch-induced cardiomyocyte apoptosis. Life Sci. 2005, 77, 160–174. [Google Scholar] [CrossRef] [PubMed]
  88. Li, J.H.; Kirkiles-Smith, N.C.; McNiff, J.M.; Pober, J.S. TRAIL induces apoptosis and inflammatory gene expression in human endothelial cells. J. Immunol. 2003, 171, 1526–1533. [Google Scholar] [CrossRef] [Green Version]
  89. Hansson, G.K. Inflammation, Atherosclerosis, and Coronary Artery Disease. N. Engl. J. Med. 2005, 352, 1685–1695. [Google Scholar] [CrossRef] [Green Version]
  90. Oever, I.A.M.V.D.; Raterman, H.G.; Nurmohamed, M.T.; Simsek, S. Endothelial Dysfunction, Inflammation, and Apoptosis in Diabetes Mellitus. Mediat. Inflamm. 2010, 2010, 1–15. [Google Scholar] [CrossRef] [Green Version]
  91. Boldt, A.; Wetzel, U.; Lauschke, J.; Weigl, J.; Gummert, J.; Hindricks, G.; Kottkamp, H.; Dhein, S. Fibrosis in left atrial tissue of patients with atrial fibrillation with and without underlying mitral valve disease. Heart 2004, 90, 400–405. [Google Scholar] [CrossRef] [Green Version]
  92. Nattel, S. Molecular and Cellular Mechanisms of Atrial Fibrosis in Atrial Fibrillation. JACC Clin. Electrophysiol. 2017, 3, 425–435. [Google Scholar] [CrossRef] [PubMed]
  93. Cantarella, G.; Risuglia, N.; Lombardo, G.; Lempereur, L.; Nicoletti, F.; Memo, M.; Bernardini, R. Protective effects of estradiol on TRAIL-induced apoptosis in a human oligodendrocytic cell line: Evidence for multiple sites of interactions. Cell Death Differ. 2004, 11, 503–511. [Google Scholar] [CrossRef] [PubMed]
  94. Cantarella, G.; Uberti, D.; Carsana, T.; Lombardo, G.; Bernardini, R.; Memo, M. Neutralization of TRAIL death pathway protects human neuronal cell line from β-amyloid toxicity. Cell Death Differ. 2003, 10, 134–141. [Google Scholar] [CrossRef] [PubMed]
  95. Cui, M.; Wang, L.; Liang, X.; Ma, X.; Liu, Y.; Yang, M.; Liu, K.; Wei, X.; Zhou, Z.; Chen, Y.H.; et al. Blocking TRAIL-DR5 signaling with soluble DR5 reduces delayed neuronal damage after transient global cerebral ischemia. Neurobiol. Dis. 2010, 39, 138–147. [Google Scholar] [CrossRef]
Figure 1. TRAIL receptors and their alternative names. TRAIL-R1 is also known as DR4 (death receptor 4) and TNFRSF10A (tumor necrosis factor receptor superfamily member 10A); TRAIL-R2 is also known as DR5 (death receptor 5) and TNFRSF10B (tumor necrosis factor receptor superfamily member 10B); TRAIL-R3 is also known as DcR1 (decoy receptor 1), TRAIL-R4 is also known as DcR2 (decoy receptor 2); OPG—osteoprotegerin (soluble decoy receptor).
Figure 1. TRAIL receptors and their alternative names. TRAIL-R1 is also known as DR4 (death receptor 4) and TNFRSF10A (tumor necrosis factor receptor superfamily member 10A); TRAIL-R2 is also known as DR5 (death receptor 5) and TNFRSF10B (tumor necrosis factor receptor superfamily member 10B); TRAIL-R3 is also known as DcR1 (decoy receptor 1), TRAIL-R4 is also known as DcR2 (decoy receptor 2); OPG—osteoprotegerin (soluble decoy receptor).
Jcm 10 01252 g001
Figure 2. Search strategy and results.
Figure 2. Search strategy and results.
Jcm 10 01252 g002
Table 1. Original Studies Assessing the Relationship between TRAIL and TRAIL-R2 and Cardiovascular Risk.
Table 1. Original Studies Assessing the Relationship between TRAIL and TRAIL-R2 and Cardiovascular Risk.
AuthorYearDesign of the StudyPopulationNumber of PatientsParameters AssessedResults and Key Observations
Heart Failure
Osmancik et al. [20]2013Prospective observational studyPatients with heart failure treated with CRT81Six-month evaluation and two-year follow-up after implantationTRAIL did not differ in responders and nonresponders, TRAIL did not predict mortality
Stenemo et al. [21]2017Prospective observational studyElderly patients without heart failure at baseline1586Eight-year and 10-year follow-up/proteomics profillingTRAIL-R2 associated with incident heart failure at follow-up and worsened LV systolic function at baseline
Cardiomyopathy
Schoppet et al. [22]2005Case-control studyMan with nonischemic dilated cardiomyopathy105 and 86 controlsComparison to control groupTRAIL elevated comparing to controls, correlating with LV end-diastolic diameter
Lula et al. [23]2009Case-control studyPatients with Chagas cardiomyopathy31 and 15 controlsTRAIL concentration compared between groups and hemodynamic parametersTRAIL concentration enhanced and correlate with LV ejection fraction and LV diastolic diameter
Atrial Fibrillation (AF)
Osmancik et al. [24]2010Prospective observational studyPatients with AF25Intervention-ablationTRAIL concentration decreased in patients successfully ablated
Deftereos et al. [25]2012Prospective observational studyPatients with persisted AF, successfully cardioverted to sinus rhythm45Six-month follow-up, measured transcardiac gradients (coronary sinus concentration minus aortic root concentration)TRAIL concentration without differences in group with and without AF recurrence, TRAIL transcardiac gradient was negative predictor of AF recurrence
Muller et al. [26]2013Prospective observational studyPatients with sinus rhythm, without history of AF, undergoing cardiac surgery33TRAIL concentration measured pre- and postoperativelyTRAIL did not predict postoperative AF
Rewiuk et al. [27]2015Prospective observational studyPatients with acute onset of AF 60TRAIL concentration measured initially and 7 to 10 days after pharmacological cardioversionTRAIL did not predict restoration of sinus rhythm, increase in TRAIL concentration in sinus rhythm maintenance group
Chua et al. [28] 2019Prospective observational studyPatients with known AF638Logistic regression with machine learning algorithms to determine AF risk factorsTRAIL-R2 associated with AF
Atherosclerosis
Kawano et al. [29]2011Cross-sectional studyPatients with type 2 diabetes without symptoms of CAD416TRAIL concentration measured in correlation with atherosclerotic lesion (IMT)TRAIL did not correlate with IMT and not differ in group with calcified plaque and without
Deftereos et al. [30]2011Cross-sectional studyPatients with stable angina or positive ischemia noninvasive test56TRAIL concentration measured during left cardiac catherization; IVUS plaque assessmentTRAIL associated with histologic prototype of plaque
Arcidiacono et al. [31]2018Prospective observational studyPatients with chronic kidney disease without previous CV events378TRAIL concentration compared with appearance of plaque baseline and after 24-month of follow-upTRAIL associated negatively with plaque at baseline and with new atheromatous plaques after 24 months
Coronary Artery Disease
Schoppet et al. [32]2005Cross-sectional studyMen undergoing coronary angiography for suspected CAD363TRAIL concentration compared between group with and without CADTRAIL and TRAIL/OPG ratio not correlated with presence or severity of CAD
Satoh et al. [33]2009Prospective observational studyPatients with CAD treated with percutaneous coronary intervention85 and 50 controls0.5-year follow-upTRAIL concentration higher in CAD patients and non-restenosis group
Mori et al. [34] 2010Cross-sectional studyPatients undergoing coronary angiography285TRAIL concentration assessed with CAD severityTRAIL concentration was inversely correlated with severity of CAD
Secchiero et al. [35]2010Case-control studyPatients with AMI113 and 21 with unstable angina and 120 controlsTRAIL concentration compared between groupsTRAIL concentration decreased in AMI patients, CAD patients characterized by an increased OPG/TRAIL ratio
Shaker et al. [36]2010Case-control studyMale patients with AMI and CAD28 with AMI, 30 with CAD and 20 controlsTRAIL concentration compared between groups TRAIL concentration lover in AMI patients
Deftereos et al. [37]2012Prospective observational studyPatients undergoing percutaneous coronary intervention with drug-eluting stent6712-months follow-upTRAIL negatively correlated indices of neointimal hyperplasia and positively correlated in-stent minimum lumen area
Song et al. [38]2012Case-control studyPatients with CAD undergoing stent implantation42 and 17 controls0.5-year follow-upTRAIL concentration was increased 1 month after angioplasty
Luz et al. [39]2016Prospective observational studyPatients with STEMI66TRAIL concentration measured between group treated with postconditioning and without, nine-month follow-upTRAIL was augmented by postconditioning and correlated to infarct size and LVEF
Manuneedhi Cholan et al. [40]2018Case-control studyPatients with stable CAD9 and 7 controlsTRAIL and F2-isoprostanes concentration compared between groupsTRAIL concentration was reduced in CAD patients and correlated with marker of oxidative stress
Teringova et al. [41]2018Prospective observational studyPatients with STEMI treated with primary percutaneous coronary intervention101TRAIL concentration measured at baseline and one month after, two-year follow-upTRAIL reaches its lowest serum concentration after reperfusion, low TRAIL concentration is associated with worse LVEF
Vascular Calcification
Chasseraud et al. [42]2011Cross-sectional studyHemodialysis patients38TRAIL concentration measured in hemodialysis patients and compared with calcification levelsTRAIL decreased in serum of hemodialysis patients but not correlate with calcification
Moon et al. [43]2019Cross-sectional studyPatients with diabetes and without diabetes71Diagnosis of PAD based on ABI results; calcification determined by computed tomography scanTRAIL was downregulated in patient with PAD and vascular calcification
Cerebrovascular Disease
Sarchielli et al. [44]2013Case-control studyPatients with silent brain infarction and lacunar infarct49 and 31 controlsAssessment of pathophysiology of silent brain infarctionTRAIL concentration was higher in patients with silent brain infarction
Kang et al. [45]2015Cross-sectional studyPatients with ischemic acute stroke293Assessment of TRAIL correlation with stroke volumeLow concentration of TRAIL correlated with stroke severity
Pan et al. [46]2015Prospective observational studyPatients with large artery atherosclerosis stroke132 and 60 controlsThree-month follow-upTRAIL concentration lower in patients with large artery atherosclerosis stroke; TRAIL negatively correlated with prognosis
Other
O’Sullivan et al. [47]2010Case-control studyPatients with PAD83 and 21 controlsPAD assessed using ABI, TRAIL concentration measured between groups with PAD and diabetesTRAIL concentration was higher in patients with PAD
Karatolios et al. [48]2011Cross-sectional studyPatients with pericardial effusion83Assessment TRAIL concentration in pericardial effusion associated with malignancy, CAD and non-malignantTRAIL concentration was higher in pericardial effusion associated with malignancy and CAD
Zhou et al. [49]2014Validation studyPregnant women812Serum samples taken from 8 to 20 week gestation; than assessed those who developed hypertensionTRAIL concentration lower in patients who developed pregnancy hypertension than uncomplicated pregnancies
Liu et al. [50]2015Prospective observational studyPatients with pulmonary hypertension78 and 80 controlsComparison of TRAIL concentration between groups, 2-years follow-upTRAIL concentration higher in patients with pulmonary hypertension; elevated TRAIL associated with eventual complications
General Cardiovascular Risk
Wigren et al. [51]2018Cross-sectional studyPatients with systemic lupus erythematosus484 and 253 controlsComparison of TRAIL-R2 concentration between groups14% of patients had CVD (CAD, cerebrovascular disease, PAD); patients with CVD had higher concentration of TRAIL-R2 than those without
Goncalves et al. [52]2019Prospective observational studyCPIP cohort (patients with atherosclerosis)55837-month follow-up Higher TRAIL-R2 associated with higher CV risk in future
Abbreviations: ABI—ankle-brachial index, AF—atrial fibrillation, AMI—acute myocardial infarction, CAD—coronary artery disease, CRT—cardiac resynchronization therapy, CV—cardiovascular, IMT—intima-media thickness, IVUS—intravascular ultrasound, LV—left ventricle, LVEF—left ventricular ejection fraction, OPG—osteoprotegerin, PAD—peripheral artery disease, STEMI—ST elevation myocardial infarction TRAIL—TNF-related apoptosis-inducing ligand, TRAIL-R2—TNF-related apoptosis-inducing ligand receptor 2.
Table 2. In Vitro Studies on Human Samples Assessing Role of TRAIL and TRAIL Receptors in Cardiovascular Diseases.
Table 2. In Vitro Studies on Human Samples Assessing Role of TRAIL and TRAIL Receptors in Cardiovascular Diseases.
AuthorYearInvestigated DiseasePopulationNumber of PatientsParameters AssessedResults and Key Observations
Yndestad et al. [53]2002Heart failurePatients with heart failure8 and 8 controlsAnalysis of gene expression in peripheral blood mononuclear cellsTRAIL gene downregulated in chronic heart failure patients
Cao et al. [54]2011Atrial fibrillationPatients with AF48 and 48 controlsTissue obtained during mitral valve surgeryTRAIL gene expression upregulated
Schoppet et al. [55]2004AtherosclerosisPatients’ samples of vascular tissue8 and 4 normal vesselsAnalysis of samples of Mönckeberg’s sclerosis and atherosclerotic arteriesTRAIL detected in calcified regions of Mönckeberg’s sclerosis and atherosclerotic arteries
Michowitz et al. [56]2005AtherosclerosisPatient’s samples of atherosclerotic plaques24TRAIL expression assessed on peripheral mononuclear cells when stimulated by oxLDLTRAIL expression enhanced by oxLDL in atherosclerotic lesions; TRAIL serum concentration reduced in patients with unstable CAD
Niessner et al. [57]2006AtherosclerosisPatients with ACS31TRAIL expression assessed on CD4 T-cells when stimulated by IFNα produced by activated plasmacytoid dendritic cellsTRAIL expression is enhanced by IFNα in atherosclerotic lesions
Goncalvez et al.2019AtherosclerosisPatients’ samples of carotid plaques202TRAIL expression analyzed in atherosclerotic lesionTRAIL-R2 and TRAIL expression were increased in symptomatic carotid plaques
Nakajima et al. [58]2003Coronary artery diseasePatients with AMI26 and 16 controlsCompared expression of TRAIL on peripheral blood mononuclear cells between groupsTRAIL expression was enhanced in AMI patients
Sato et al. [59]2006Coronary artery diseasePatients with ACS50 and 33 controlsExamined whether the TRAIL pathway was involved in CD4 T cell-mediated apoptosisTRAIL expression enhanced on CD4 T-cells in patients with ACS
Corallini et al. [60]2009Coronary artery diseasePatients with AMI80 and 40 controlsAnalysed the relationship of TRAIL with mesenchymal stem cells (role in regeneration after AMI)TNFα enhanced the migration of mesenchymal stem cells in response to TRAIL
Sato et al. [61]2010Coronary artery diseasePatients with ACS55 and 34 controlsIntervention-administration of statins and TRAIL-specific antibodiesTRAIL-R2 upregulated on endothelial cells, T cell mediated endothelial death was dependent on the TRAIL pathway
Secchiero et al. [62]2010Coronary artery diseasePatients with AMI30Evaluated the potential role of metalloproteinase 2 in promoting the cleavage of TRAIL after AMITRAIL concentration showed inverse correlation with MMP2/TIMP2 ratio
Liu et al. [63]2007Vascular calcificationPatients’ aortic samples taken during abdominal aortic aneurysm33 and 8 controlscalcification levels were determined by computed tomography scanTRAIL and TRAIL-R1 expression were higher in aneurysm samples and in calcified samples
Galeone et al. [64]2013Vascular calcificationPatients’ samples of severe calcific aortic stenosis taken during valve replacement10 and 10 controlsImmunohistochemistry and confocal microscopy used for TRAIL immunostaininghigher TRAIL concentration detected in calcific aortic valves and serum
Hameed et al. [17]2012Pulmonary hypertensionSpecimens of human pulmonary artery smooth muscle cells-Assessment of TRAIL expression in smooth muscle cells TRAIL upregulated in pulmonary artery smooth muscle cells in patients with pulmonary hypertension
Tisato et al. [65]2013Chronic venous diseaseSpecimens of venous endothelial cells from patients with chronic venous disease20Assessment of TRAIL expression in correlation with hemodynamic parameters and after GM-CSF exposureTRAIL expression correlated positively with resistance index and GM-CSF
Abbreviations: AF—atrial fibrillation, AMI—acute myocardial infarction, ACS—acute coronary syndrome, CAD—coronary artery disease, GM-CSF—granulocyte-macrophage colony-stimulating factor, IFNα—interferon α, MMP2/TIMP2—metalloproteinase-2/tissue inhibitor molecule 2, oxLDL—oxidized low-density lipoprotein, TNFα—tumor necrosis factor α, TRAIL—TNF-related apoptosis-inducing ligand, TRAIL-R1—TNF-related apoptosis-inducing ligand receptor 1, TRAIL-R2—TNF-related apoptosis-inducing ligand receptor 2.
Table 3. Summary of the Studies Assessing the Association between TRAIL or TRAIL-R2 with Cardiovascular Patient-Related Outcome Measures.
Table 3. Summary of the Studies Assessing the Association between TRAIL or TRAIL-R2 with Cardiovascular Patient-Related Outcome Measures.
AuthorYearPopulationMeasured ProteinNumber of PatientsFollow-UpDeath/CV DeathsAssessed OutcomeRelation TRAIL to All-Cause MortalityRelation TRAIL to CV MortalityComments
MIStrokeCV DeathAll-Cause DeathRe-Hospitalization
Niessner et al. [66]2009Patients with HF *TRAIL35116 months93/ND---yesyesinversely
p < 0.001
n/aTRAIL predicted outcome (all-cause mortality and rehospitalization)
Secchiero et al. [67]2009Patients with AMITRAIL60 and 60 control group12 months10/9--yes yes-NDinversely
p = 0.001
TRAIL concentration decreased at baseline, low levels of TRAIL at discharge was prognostic factor of cardiac death and heart failure at 12 months
Liabeuf et al. [68]2010Chronic kidney disease patientsTRAIL1302 years36/19--yesyes-inversely
p = 0.010
NSSlowest TRAIL was associated with infectious but not CV mortality
Volpato et al. [69]2011inCHIANTI study (older people with CVD)TRAIL12826 years259/112--yesyes-inversely
p = 0.008
inversely
ND
an association was found between prevalent CV disease and TRAIL
Osmancik et al. [70]2013Patients with ACSTRAIL2950.5 year12/NDyesyes (but not evaluated)-yesyesinversely
p = 0.001
n/aTRAIL predicted all-cause mortality, re-MI and combined end point (death and hospitalization for HF)
Mori et al. [71]2013Male hemodialysis patientsTRAIL14936 months33/11--yesyes-inversely
p = 0.011
NSSTRAIL associated with infectious and all-cause mortality but not CV mortality
Richter et al. [72]2013Patients with HF *TRAIL3495 years195/145---yes-inversely
p < 0.001
n/aTRAIL predicted all-cause mortality
Kuzniewski et al. [73]2016Hemodialysis patientsTRAIL69 and 35 controls7 years39/31--yesyes-NSSNSSTRAIL did not predict CV mortality; OPG/TRAIL ratio positively predicted all-cause and CV mortality
Hage et al. [74]2017Patients with HF with preserved ejection fraction/proteomic studyTRAIL and TRAIL-R2861.5 years11/ND---yesyesND (composite outcome)n/aTRAIL and TRAIL-R2 predicting outcome (all-cause mortality or re-hospitalization
Mattisson et al. [75]2017MDC study (general population)TRAIL-R2474219 yearsND/278yesyesyes--n/an/ahigher TRAIL-R2 was associated with increased risk of CV events (myocardial infarction and stroke)
Skau et al. [76]2017Patients with AMITRAIL-R28477 years207/ND---yes-n/an/aTRAIL-R2 predicted all-cause mortality
Ajala et al. [77]2018SmokersTRAIL4748 years83/ND---yes-inversely
p = 0.004
n/aTRAIL concentration reduced in smokers with comorbid emphysema and CAD; related to reduced survival/ CAD assessed by quantifying coronary artery calcium
Nowak et al. [78]2018Patients with diabetesTRAIL-R212116 yearsNDyesyes-yes-n/an/aTRAIL-R2 increased concentration associated with incident major adverse CV events
Feldreich et al. [79]2019MIMICK study
(hemodialysis patients)/proteomic study
TRAIL-R218343 monthsND/45--yes--n/an/aTRAIL-R2 associated with CV mortality
Ferreira et al. [80]2020Patients with diabetes after MI/proteomic studyTRAIL-R251311.5 year302/226yesyesyesyesyesn/an/aTRAIL-R2 prognose all-cause mortality and CV death or HF hospitalization
* the same population. Abbreviations: ACS—acute coronary syndrome, AMI—acute myocardial infarction, CV—cardiovascular, CVD—cardiovascular disease, HF—heart failure, MI—myocardial infarction, ND—no data, NSS—not statistically significant.
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kakareko, K.; Rydzewska-Rosołowska, A.; Zbroch, E.; Hryszko, T. TRAIL and Cardiovascular Disease—A Risk Factor or Risk Marker: A Systematic Review. J. Clin. Med. 2021, 10, 1252. https://doi.org/10.3390/jcm10061252

AMA Style

Kakareko K, Rydzewska-Rosołowska A, Zbroch E, Hryszko T. TRAIL and Cardiovascular Disease—A Risk Factor or Risk Marker: A Systematic Review. Journal of Clinical Medicine. 2021; 10(6):1252. https://doi.org/10.3390/jcm10061252

Chicago/Turabian Style

Kakareko, Katarzyna, Alicja Rydzewska-Rosołowska, Edyta Zbroch, and Tomasz Hryszko. 2021. "TRAIL and Cardiovascular Disease—A Risk Factor or Risk Marker: A Systematic Review" Journal of Clinical Medicine 10, no. 6: 1252. https://doi.org/10.3390/jcm10061252

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