Next Article in Journal
Virtual Screening Strategy and In Vitro Tests to Identify New Inhibitors of the Immunoproteasome
Next Article in Special Issue
Lamin A/C Ablation Restricted to Vascular Smooth Muscle Cells, Cardiomyocytes, and Cardiac Fibroblasts Causes Cardiac and Vascular Dysfunction
Previous Article in Journal
Novel SPEA Superantigen Peptide Agonists and Peptide Agonist-TGFαL3 Conjugate. In Vitro Study of Their Growth-Inhibitory Effects for Targeted Cancer Immunotherapy
Previous Article in Special Issue
Target and Cell Therapy for Atherosclerosis and CVD
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Role of Galectin-3 in Predicting Congenital Heart Disease Outcome: A Review of the Literature

by
Amalia Făgărășan
1,†,
Maria Săsăran
2,*,†,
Liliana Gozar
1,
Andrei Crauciuc
3 and
Claudia Bănescu 
4
1
Department of Pediatrics III, Faculty of Medicine, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Târgu Mures, Gheorghe Marinescu Street no 38, 540136 Târgu Mures, Romania
2
Department of Pediatrics III, Faculty of Medicine in English, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Târgu Mures, Gheorghe Marinescu Street no 38, 540136 Târgu Mures, Romania
3
Department of Medical Genetics, George Emil Palade University of Medicine, Pharmacy, Sciences and Technology of Târgu Mures, Gheorghe Marinescu Street no 38, 540136 Târgu Mures, Romania
4
Genetics Department, Center for Advanced Medical and Pharmaceutical Research, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, Gheorghe Marinescu Street no 38, 540136 Târgu Mures, Romania
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Int. J. Mol. Sci. 2023, 24(13), 10511; https://doi.org/10.3390/ijms241310511
Submission received: 18 April 2023 / Revised: 20 June 2023 / Accepted: 21 June 2023 / Published: 22 June 2023
(This article belongs to the Special Issue Cardiovascular Diseases: Molecular Mechanisms and Potential Therapy)

Abstract

:
Galectin-3 (Gal-3) is a novel pro-fibrotic biomarker that can predict both right and left cardiac dysfunction caused by various cardiovascular conditions. Its expression seems to be progressively altered with evolving cardiac remodeling processes, even before the onset of heart failure. Hence, Gal-3 has been found to be an individual predictor of acute and chronic heart failure or to serve as part of an integrated biomarker panel that can foresee adverse cardiac outcomes. In congenital heart disease (CHD), Gal-3 correlates with cardiac mortality and complications in both children and adults and is proposed as a therapeutic target in order to reverse the activation of pro-fibrosis pathways that lead to heart failure. Positive associations between serum Gal-3 levels, post-operatory hospitalization rates, complications and ventricular dysfunction have also been reported within studies conducted on patients with CHD who underwent corrective surgery. Thus, this review tried to address the potential utility of Gal-3 in patients with CHD and particularly in those who undergo corrective surgery. The heterogeneity of the literature data and the lack of validation of the results obtained by the current studies on larger cohorts cannot be neglected, though. Further longitudinal research is required to establish how Gal-3 can relate to long-term outcomes in pediatric CHD.

1. Introduction

Galectin-3 (Gal-3) represents a β-galactosidase-binding lectin that can mediate cell-to-extracellular matrix (ECM) communication through interactions with ligands such as laminin, integrin and collagen [1]. Although mainly found within the cellular cytoplasmatic compartment, Gal-3 has been identified both within the nucleus and the mitochondria and can be easily externalized through specific exosomes by an alternative secretory pathway [2,3,4]. Its proven involvement in cellular pre-messenger RNA (mRNA) splicing within the nucleus implies a direct correlation with cellular proliferation processes, which has been strengthened by the discovery of its antiapoptotic effects exerted through the inhibition of cytochrome c release and mitochondrial protection [3,5]. As Gal-3 has the ability to bind to both the cell surface and the ECM and to interact with cellular components, multiple experimental studies have proposed this galactosidase as a mediator of the pathways involved in cellular apoptosis, angiogenesis, cell adhesion, migration and inflammation [6]. A multifunctional protein with both intracellular and extracellular roles, Gal-3 is expressed by several types of cells, such as epithelial cells, myeloid cells and immune cells (neutrophils, B- and T-cell lymphocytes, macrophages and natural killer cells) [7]. Moreover, Gal-3 has been identified within the conjunctive, epithelial tissue, respiratory tract, digestive tract, urinary tract and hepatic and heart tissue [8]. Given its complex interaction with cytosolic proteins, as well as its ubiquitous tissular expression, Gal-3 has been proven to play a role in the development of a variety of disorders, including renal disease, cardiovascular disorders, viral infection, autoimmune diseases, neurodegenerative disorders and malignancies [9,10,11,12,13,14,15,16].
Gal-3 has been regarded as a key effector in the promotion of fibrosis after the initial disproportionate inflammation-driven processes, which will enhance the transformation of fibroblasts into myofibroblasts and, therefore, the expansion of the ECM, a mandatory stage in fibrotic tissue genesis [6]. This mechanism is mediated by transforming growth factor-beta (TGF-β) and macrophages, which both promote myofibroblast differentiation [17,18]. It has been known for a long time that Gal-3 is overexpressed within macrophages, as proven by early experimental studies [19]. Moreover, Gal-3 stimulates TGF-β activity and has been proposed as a therapeutic target in the mediation of idiopathic pulmonary fibrosis [20]. Excessive macrophage aggregation and inducement of the TGF-β1/α-smooth muscle actin (SMA)/collagen I (Col I) profibrotic pathway represents essential physiopathological processes leading to atrial fibrosis, according to experimental studies conducted on rats [21,22,23]. The involvement of Gal-3 in the pathogenesis of cardiac fibrosis is briefly illustrated in Figure 1.
Cardiac fibrosis represents a hot topic for the in vitro modulation of Gal-3 expression. Its proven involvement in the renin–angiotensin–aldosterone axis led to the proposal of its inhibition as a therapeutic tool for the prevention of myocardial hypertrophy and interstitial fibrosis [24]. For example, spironolactone administration reduces the Gal-3 concentrations in patients with reduced ejection fractions, which shows a potential beneficial therapeutic outcome of the inhibition of the renin–angiotensin–aldosterone axis for the prevention of myocardial fibrosis and remodeling processes [25]. However, there is a dispute surrounding the cardiac specificity of Gal-3 due to its persistent elevation in patients before and after heart transplantation. There is also a lack of correlation between the circulating levels and Gal-3 expression within myocardial tissue and in post-transplant interstitial fibrosis [26].
Starting from evidence based on the active role of Gal-3 in the modulation of cardiac fibrosis, multiple research articles have proposed this molecule as a biomarker for various cardiovascular diseases, including cardiac insufficiency, atrial fibrillation and congenital cardiac malformations, or as a predictor of long-term outcomes after cardiac resynchronization therapy [27,28,29,30,31]. As most chronic heart disease conditions will eventually lead to the development of chronic heart failure (CHF), the role of Gal-3 as a prognostic factor and mortality predictor in patients diagnosed with severe myocardial disfunction has been thoroughly investigated [32,33,34]. Figure 2 provides a synopsis of the associations between Gal-3 and various cardiovascular disorders.
The present review aims to assess the role of Gal-3 in predicting myocardial dysfunction, as well as specific complications related to cardiac remodeling processes in congenital cardiac malformation. Starting from extensive research that asserted Gal-3 as a marker of ventricular maladaptation and its utility in the follow-up of patients with a diagnosis of heart failure of different etiologies, this review will try to highlight the potential utility of this pro-fibrotic marker in predicting the early onset of heart failure signs and post-operatory complications of corrective surgery in patients with congenital heart disease (CHD).

2. Materials and Methods

After an initial screening of the literature data for Gal-3-based physiopathological mechanisms involved in cardiac remodeling and heart failure, which constitute the grounds upon which Gal-3 has emerged as a biomarker in CHD, our focus was directed toward the role of Gal-3 in CHD and its outcome. The PubMed, Web of Science and Scopus databases were searched for articles published in the English language that assessed the role of Gal-3 in CHD, it complications and corrective cardiac surgery outcomes. The search terms were “Gal-3” AND “congenital heart disease” OR “congenital cardiac malformation” OR “congenital heart defect”. We included adult and pediatric population-based studies that adhered to our aforementioned objectives. Experimental, animal-based studies and studies published in a non-English language were excluded.

3. Gal-3 and Cardiac Remodeling

3.1. Gal-3 and Cardiac Fibrosis

Gal-3 was initially thought to be a marker of progressive cardiac remodeling associated with left ventricular dysfunction and positively correlated with the rate of adverse cardiovascular events [35]. However, Gal-3 expression within the myocardium seems to be altered before the onset of heart failure, as suggested by an experimental study conducted on pericardial Gal-3-infused rats, which exhibited significant collagen production triggered by cardiac fibroblasts [36]. The in vitro pharmacological inhibition of Gal-3 prevented the development of heart failure by hampering cardiac fibrosis and left ventricular dysfunction. Still, left ventricular hypertrophy continued to develop as a result of the intervention used to trigger cardiac remodeling, even with Gal-3 inhibition [37]. Thus, Gal-3 seems to mainly interfere with collagen production. An in vivo example is represented by the onset of cardiac remodeling caused by volume overload in the setting of arterial hypertension, which can already lead to a significant elevation in Gal-3 [38].

3.2. Gal-3 and Left Ventricular Remodeling

The relationship between Gal-3 and left ventricular remodeling is mainly reflected through a low LVEF. Elevated Gal-3, together with increased values of N-terminal pro-B-type natriuretic peptide (NT-proBNP) and C-reactive protein (CRP) were predictive of a very diminished left ventricular ejection fraction (LVEF), with values even lower than 35%, in patients with newly diagnosed dilated cardiomyopathy, according to Rieth et al. [39]. This association between Gal-3 and diminished LVEF was further strengthened by changes seen on cardiac magnetic resonance imaging in subjects who suffered from myocardial infarction [40,41]. These changes often became obvious 30 days post-myocardial infarction, in correlation with diastolic dysfunction progression [42]. It is thought worth mentioning that impaired parietal kinetics has also been significantly associated with mid-range ejection fraction in heart failure patients [43]. However, other studies, such as the one conducted by de Boer et al., showed that the prognostic value of Gal-3 stands out only in those patients with HF and preserved left ventricular ejection fraction (LVEF) [44]. One meta-analysis claimed that Gal-3 can be used as a predictor for new-onset HF with preserved LVEF, which also correlates with left ventricular diastolic dysfunction [45]. A recent review article confirmed these findings and proposed Gal-3 as a marker that can guide the therapeutic approach in HF with preserved LVEF [46]. Still, due to the paucity of studies that have assessed the Gal-3 levels in subjects with HF and preserved LVEF, another recent meta-analysis addressed the need for future research in this particular clinical setting [47].

3.3. Gal-3 and Atrial Remodeling

Gal-3 poses pro-fibrotic effects upon the atrial interstitium, promoting arrhythmogenesis through CD98 (its membrane surface receptor) signaling [48]. As fibrosis is the main precipitant factor for the development of atrial fibrillation (AF), the biomarker role of Gal-3 in AF has been proposed [49]. Since AF usually occurs in the setting of cardiac structural remodeling, namely atrial dilation and cardiomyocyte replacement by fibrotic tissue, research testifying a correlation between its higher incidence in the general population and elevation in Gal-3 levels was foreseeable [50,51]. Moreover, the AF severity and persistence were related to higher Gal-3 values by Ho et al. but without taking into account additional risk factors [51]. Thus, Gal-3 has been proposed as a therapeutic guidance adjuvant that can predict the need for renin–angiotensin–aldosterone system inhibitor therapy, as well as a potential therapeutic target, with the help of β-adrenoceptor antagonists. As a result, conversion to a sinus rhythm can be more easily achieved through the pharmacological modulation of Gal-3 [52,53,54]. Gal-3 might also be useful in the prediction of post-operative AF. An elevation in Gal-3 levels can predict AF occurring after coronary artery bypass surgery, according to Erdem et al. In their study, patients who maintained a sinus rhythm post-operatively had significantly lower Gal-3 levels, but these values were not compared with the ones of healthy controls [55]. Other studies have also related the augmentation of Gal-3 with the arousal of AF after cardiac surgery, but a recent review of available evidence concluded that there was insufficient data available due to contradictory results reported and the limited study samples on which the research on the matter was conducted [56,57,58].

3.4. Gal-3 and Cardiac Remodeling in Relation to Impaired Glucose Metabolism

The relationship between Gal-3 and cardiac remodeling in altered glucose tolerance or diabetes mellitus patients is still being debated. Gal-3 might represent a predictor of early cardiac remodeling in patients with diabetes mellitus but only in junction with other cardiac-specific biomarkers [59]. Moreover, elevation of the Gal-3 levels was reported in both pre-diabetic and diabetic patients within a case–control study without any relationship to the cardiac function or adverse events [60]. In spite of existing proof regarding the augmentation of Gal-3 in patients with heart failure and impaired glucose metabolism, the reduction of the left ventricular contractile reserve among patients with heart failure and diabetes mellitus did not independently alter the plasmatic Gal-3 levels.

4. Gal-3, Heart Failure and Its Associated Complications

Gal-3 has been established as a marker in multiple cardiovascular diseases. Its connection to heart failure has been widely investigated and reviewed in multiple publications, which presented Gal-3 as an useful tool for risk stratification and prognosis evaluation in both acute and chronic heart failure [32,61,62]. This section provides a brief presentation of the role of Gal-3 in acute, incidental and chronic heart failure.

4.1. Gal-3 and Acute Heart Failure

Van Kimmenade et al. were the first to evaluate the utility of novel serum markers in the diagnosis of acute heart failure (AHF). The authors concluded that plasma NT-proBNP represents the best diagnostic marker, with a superiority over both apelin and Gal-3, but suggested that the combination between NT-proBNP and Gal-3 might be the best mortality predictor [63]. In a similar fashion, a review performed on pediatric studies that enrolled patients diagnosed with heart failure concluded that none of the currently available serum biomarkers of myocardial fibrosis perform as well as NT-proBNP in predicting ventricular dysfunction [64]. Gal-3 was afterwards regarded by the randomized SHOCK-COOL trial as a stable biomarker that correlates well with a 30-day mortality of different causes in patients with AHF and which is not influenced by age, sex and body mass index [65]. It is therefore unsurprising that Gal-3 has been proposed as a diagnostic biomarker and potential therapeutic target in AHF [66]. Still, Gal-3 values seem to be dependent upon renal function, with one large-scale study suggesting that the serum levels of Gal-3 need to be adjusted according to the glomerular filtration rate [67]. As a matter of fact, recent research in the field recommends the use of a multi-marker panel for evaluation of the AHF prognosis, which includes Gal-3 and creatinine, as well as growth differentiation factor 15 (GDF-15), NT-proBNP, suppression of tumorigenicity 2 (ST2) and troponin I [68,69]. Wang et al. even sustained that a combined biomarker panel consisting of Gal-3, NT-proBNP and ST2 might be indicative of systemic fibrotic processes evolution, not only of those localized within myocardial tissue [69].

4.2. Gal-3 and Incident Heart Failure

Gal-3 concentrations correlate with incident heart failure risk, as claimed by a meta-analysis conducted on nine studies [70]. Gal-3 elevation enhances the risk of incident heart failure and is associated with higher overall mortality rates and increases in the left ventricular mass, according to Ho et al. [71]. The ARIC study also identified a connection between Gal-3 elevation and incident heart failure but emphasized that Gal-3 values tend to be higher in the obese, especially among females and Caucasian populations [72]. A study conducted on a large population-based cohort (FINIRISK 1997 cohort) initially established that Gal-3 counts as a predictor of three major endpoints—namely, all-cause mortality, cardiovascular death and incident heart failure—after adjusting for the parameters of the Framingham risk score. However, when additionally adjusting for NT-proBNP, the Gal-3 value was not related to an increase in heart failure risk anymore [73]. Still, another meta-analysis of 18 studies concluded that high Gal-3 levels are correlated with incidental heart failure, all-cause mortality and cardiovascular mortality in the general population [74].

4.3. Gal-3 and Chronic Heart Failure

In the case of CHF, Gal-3 also acts as an important predicting factor independent of the severity of HF assessed through the NT-proBNP levels [75]. A cut-off level of 11.5 ng/mL has been proven to be predictive of CHF exacerbations requiring hospitalization, according to Baran et al., who also identified a correlation between Gal-3 and vascular stiffness within their study group [76]. Other research proposed a cut-off value of 8 ng/mL as a risk factor for CHF development. This case–control study also reported an increase in the Gal-3 levels with age among the healthy subjects [77]. The symptom severity, morbidity and hospitalization rates in patients with CHF have been associated with the elevation of the Gal-3 levels. Furthermore, the efficacy of Valsartan treatment in preventing the need for hospitalization seems to be dependent upon the Gal-3 values, as suggested by Anand et al. [78]. It is yet unclear whether Gal-3 is influenced or not by other biomarkers, as suggested by the study performed by Miller et al., in which Gal-3 proved to be a predictor of cardiac adverse events in CHF patients only in conjunction with ST2 and not taken individually [79]. Given the extensive evidence available, Gal-3 has been proposed by The American College of Cardiology Foundation (ACCF) and the American Heart Association (AHA) as an integrated part of a biomarker panel that can assess the extent of cardiac fibrosis [80].

5. Gal-3 in Congenital Heart Disease

In healthy children, Gal-3 presents no variations in age and gender, as opposed to other biomarkers that are also altered in the setting of myocardial fibrosis, such as ST2, NT-proBNP and troponin [81,82]. These findings, provided by two pediatric studies that compared different age groups, are in line with the evidence-based theory of Schindler et al., which sustained the stability of Gal-3 over other cardiac biomarkers [83]. As a result, Gal-3 is considered to be a promising, age-independent biomarker in pediatric cardiovascular disorders that can identify early signs of cardiac disfunction. Table 1 overviews the role of Gal-3 in predicting the early onset of myocardial fibrosis, heart failure and a CHD outcome.
In CHD, the experimental data showed that Gal-3 is highly expressed in the setting of pulmonary artery hypertension (PAH), which represents one of the main causes of right ventricular failure and subsequent death. The pathomechanism involved is represented by the T-helper 2 (Th2) cell-induced release of inflammatory cytokines, such as interleukin(IL)-4 and IL-5 [84]. Thus, Gal-3 can prematurely identify the onset of right ventricle remodeling in patients with PAH and is positively associated with the right ventricular systolic and diastolic indices [85]. On the other hand, Gal-3 was not able to differentiate between adaptive and maladaptive RV remodeling in PAH patients within the study of Keranov et al. [86].
An association between Gal-3 and an right ventricle pressure overload was also revealed by a study performed on adult patients with CHD [87]. However, the prediction of the alteration of the cardiac function parameters and associated complications in children diagnosed with CHD, the main cause of CHF at pediatric ages, still remains a challenge. Gal-3 has emerged as a marker of progressive ventricular remodeling in children with CHD, which positively correlates with pulmonary artery pressure and the left atrial and ventricular diameters, according to Mohammed et al. [88]. Therefore, Gal-3 has also been analyzed in survivors of CHDs such as Tetralogy of Fallot (ToF) and has not been asserted as a potential biomarker candidate for long-term outcomes, as opposed to several other plasmatic biomarkers [89]. Kotby et al. identified an increase in the Gal-3 serum levels with chronic heart failure progression, quantified through Ross classification, regardless of ejection fraction preservation. Approximately one-third of their pediatric study group included patients with a CHD diagnosis, and the echocardiographic assessment yielded a negative correlation between Gal-3 and the systolic and diastolic function parameters [90]. The importance of Gal-3 in heart failure staging was also confirmed in a study conducted on children with CHD through a positive correlation between its serum levels and Ross classification [91]. However, while some authors (Saleh et al.) have claimed that Gal-3 might perform even better than Ross classification in the precocious identification of heart failure among children with CHD, exhibiting excellent sensitivity and specificity with a cut-off value of 10.4 ng/dL, some others (Cura et al.) proved in a case–control study that enrolled infants with ventricular septal defects (VSD) that Gal-3 increases independently from left ventricular dilation [92,93]. On the other hand, a study conducted on adults with CHD revealed that Gal-3 correlates with the global longitudinal strain (GLS) and NT-proBNP values but does not constitute a marker of major adverse cardiac events [94]. Another study (Geenen et al.) claimed that Gal-3 performs better than NT-proBNP or an echocardiographic strain analysis in predicting adverse outcome in adults with a systemic right ventricle and transposition of the great arteries (TGA) [95]. Therefore, given the conflicting results, the previously reported positive correlations between Gal-3 and the systolic and diastolic diameters, as well as end diastolic volumes, still require validation in larger cohorts [90,92].
A Gal-3 level increase is suggestive of the development of supraventricular and ventricular tachycardia in adults with CHD [94]. The presence of an arrhythmia brings about an additional enhancement of Gal-3 expression to the one normally found with a pre-existing disorder, as suggested by a study that comparatively assessed patients with CHD, rheumatic heart disease and rheumatic disease complicated by atrial fibrillation [96]. The correlation between Gal-3 and arrhythmia seems to be independent of a pre-existing underlying condition. A small-scale study discovered an increase in Gal-3 in adolescents with ventricular arrhythmias, as opposed to healthy counterparts. Moreover, the highest Gal-3 serum values were reported in patients with complex ventricular arrythmias. This finding was based on one study that revealed that this marker is dependent upon the severity of left ventricular dysfunction, as expressed through a moderate linear correlation between Gal-3 and the left ventricular diastolic diameter [97].
While taking into consideration the potential modulation of Gal-3 for the prevention of myocardial fibrosis, one study and two randomized controlled trials (RCTs) assessed the effect of an aldosterone antagonist agent, eplerenone, and a prostacyclin analogue, iloprost, on its circulating levels [98,99,100], while, in one RCT, eplerenone did not significantly influence the Gal-3 levels in adults with a childhood diagnosis of ToF or complete TGA. In another one, which enrolled adults with a history of TGA correction, a decrease in Gal-3 was found after one year of treatment [98,100]. In another study, iloprost inhalation did significantly lower the Gal-3 levels in patients with CHD and pulmonary artery hypertension. Thus, iloprost might influence myocardial fibrotic processes through Gal-3 modulation [99].
Gal-3 has also been proposed as a marker of unfavorable outcome in heritable cardiomyopathies. In the study of Hu et al., conducted on non-ischemic cardiomyopathy patients, Gal-3 correlated with major adverse cardiovascular events [101]. However, another study revealed a lack of correlation between Gal-3 and adverse outcomes in subjects with stable, dilated cardiomyopathy [102]. In vitro studies support the implications of Gal-3 in the ventricular remodeling implied by inherited cardiomyopathies. Thus, in a mouse model with dilated cardiomyopathy, deletion of the Gal-3 gene resulted in the attenuation of cardiac remodeling and dysfunction [103].

6. Gal-3 as a Predictor of Complications Associated with CHD Corrective Surgery

An integrated biomarker panel, consisting of ST2, Gal-3, glial fibrillary acidic protein (GFAP) and NT-proBNP, can constitute a relevant addition to clinical data in predicting the 30-day and one-year rehospitalization and mortality rates in children undergoing surgery for CHD [104,105,106]. Similar results were reported by Opotowsky et al., who found a positive relationship between the Gal-3 levels and the presence of Fontan circulation in adults, as well as a positive correlation with cardiovascular hospitalization and death [107]. Gal-3 has also been investigated as a potential predictor of post-operative ventricle remodeling in patients with CHD. DiLorenzo et al., who assessed multiple biomarkers of myocardial fibrosis for the detection of right ventricle remodeling after ToF surgical repair, amongst which was also Gal-3, concluded that only matrix metalloproteinase-1 (MMP-1), a marker of several common congenital cardiac and vascular anomalies, can really be taken into consideration [108,109]. In children undergoing surgery for isolated aortic coarctation with/without aortic hypoplasia, there was no significant linear relationship between the pre-operatory Gal-3 levels and post-operatory left ventricular mass index nor relative wall thickness, which were perceived as markers of left ventricular remodeling [110]. The prognostic role of Gal-3 in patients with functional univentricular hearts and Fontan circulation was also put into question by a study that did not include this biomarker amongst the ones related to major adverse events [111]. Table 2 provides details regarding the main outcomes of studies that investigated the role of Gal-3 in predicting myocardial fibrosis, as well as cardiovascular complications, in subjects who underwent CHD corrective surgery.
A decrease in the Gal-3 levels was reported immediately after the percutaneous closure of atrial septal defects in an adult cohort. However, the parameter returned to its baseline levels three months after the aforementioned procedure, irrespective of the cardiac remodeling processes [112]. On the other hand, Dudnyk et al. claimed that, even if children are completely asymptomatic in post-corrective surgery for CHD, their Gal-3 levels are still elevated and are associated with myocardial disfunction, depicted through modified tissue Doppler imaging measurements [113]. Another study conducted on adult patients confirmed that Gal-3 failed to reflect both left and right ventricle dysfunction in adult patients who had undergone surgical procedures for ToF repair at pediatric ages [114]. Furthermore, Gal-3 did not correlate with the hemodynamic parameters such as right atrial pressure or right ventricular end diastolic pressure in children undergoing transcatheter valve replacement for congenital pulmonary valve stenosis or insufficiency, unlike NT-proBNP and ST2 [115].
In children undergoing cardiopulmonary bypass during CHD corrective surgery, Gal-3 has also been studied in relation to acute kidney injury, occurring as a post-operative complication. Parsons et al. concluded that Gal-3 cannot be considered a reliable marker for acute kidney injury (AKI) in this clinical setting [116]. Contradictorily, Greenberg et al. described an independent association between the pre-operatory and first post-operatory Gal-3 levels and AKI in children undergoing cardiac surgical procedures [117].

7. Conclusions

From what is known so far, Gal-3 is a next-generation biomarker with predictive potential for cardiac dysfunction in right and left heart pathology in children and adults. This review provides comprehensive information regarding the potential utility of Gal-3 in predicting the early onset of heart failure signs and post-operatory complications of corrective surgery in patients with CHD. Its focus on the role of Gal-3 in CHD makes it peculiar among the previous literature reviews that have analyzed the same biomarker in cardiovascular diseases. The limitation of the present work is the heterogeneity and the lack of validation of data on large cohorts. A larger number of patients were enrolled in studies that assessed the role of Gal-3 in CHD post-corrective surgery scenarios. Further longitudinal research is required to establish how Gal-3 can relate to long-term outcomes and complications in both pediatric and adult CHD. The robustness of its predictor role depends on its placement in a clinical context and the integration of other parameters derived from an advanced cardiac imaging analysis (echocardiography and cardiac magnetic resonance).

Author Contributions

A.F. and M.S. were equally involved in the conceptualization, literature data research, drafting of the manuscript, its revision and subsequent correction. L.G., A.C. and C.B. reviewed, added new information where necessary and edited the manuscript. C.B. reviewed the final version of the manuscript, made the necessary amendments and reorganized the information when necessary. All authors have read and agreed to the published version of the manuscript.

Funding

This work was funded by the University of Medicine, Pharmacy, Science and Technology “George Emil Palade” of Târgu Mureș Research, grant number 163/1/10.01.2023.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Ochieng, J.; Furtak, V.; Lukyanov, P. Extracellular Functions of Galectin-3. Glycoconj. J. 2002, 19, 527–535. [Google Scholar] [CrossRef] [PubMed]
  2. Davidson, P.J.; Davis, M.J.; Patterson, R.J.; Ripoche, M.-A.; Poirier, F.; Wang, J.L. Shuttling of Galectin-3 between the Nucleus and Cytoplasm. Glycobiology 2002, 12, 329–337. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Yu, F.; Finley, R.L.; Raz, A.; Kim, H.-R.C. Galectin-3 Translocates to the Perinuclear Membranes and Inhibits Cytochrome c Release from the Mitochondria: A Role for Synexin In Galectin-3 Translocation*. J. Biol. Chem. 2002, 277, 15819–15827. [Google Scholar] [CrossRef] [Green Version]
  4. Nickel, W. The Mystery of Nonclassical Protein Secretion. Eur. J. Biochem. 2003, 270, 2109–2119. [Google Scholar] [CrossRef] [PubMed]
  5. Dagher, S.F.; Wang, J.L.; Patterson, R.J. Identification of Galectin-3 as a Factor in Pre-MRNA Splicing. Proc. Natl. Acad. Sci. USA 1995, 92, 1213–1217. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  6. Li, L.; Li, J.; Gao, J. Functions of Galectin-3 and Its Role in Fibrotic Diseases. J. Pharmacol. Exp. Ther. 2014, 351, 336–343. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Newlaczyl, A.U.; Yu, L.-G. Galectin-3—A Jack-of-All-Trades in Cancer. Cancer Lett. 2011, 313, 123–128. [Google Scholar] [CrossRef]
  8. Van den Brûle, F.A.; Fernandez, P.L.; Buicu, C.; Liu, F.T.; Jackers, P.; Lambotte, R.; Castronovo, V. Differential Expression of Galectin-1 and Galectin-3 during First Trimester Human Embryogenesis. Dev. Dyn. 1997, 209, 399–405. [Google Scholar] [CrossRef]
  9. Clementy, N.; Garcia, B.; André, C.; Bisson, A.; Benhenda, N.; Pierre, B.; Bernard, A.; Fauchier, L.; Piver, E.; Babuty, D. Galectin-3 Level Predicts Response to Ablation and Outcomes in Patients with Persistent Atrial Fibrillation and Systolic Heart Failure. PLoS ONE 2018, 13, e0201517. [Google Scholar] [CrossRef]
  10. Cui, Y.; Qi, X.; Huang, A.; Li, J.; Hou, W.; Liu, K. Differential and Predictive Value of Galectin-3 and Soluble Suppression of Tumorigenicity-2 (SST2) in Heart Failure with Preserved Ejection Fraction. Med. Sci. Monit. 2018, 24, 5139–5146. [Google Scholar] [CrossRef]
  11. Chen, S.-C.; Kuo, P.-L. The Role of Galectin-3 in the Kidneys. Int. J. Mol. Sci. 2016, 17, 565. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. Tan, K.C.B.; Cheung, C.-L.; Lee, A.C.H.; Lam, J.K.Y.; Wong, Y.; Shiu, S.W.M. Galectin-3 Is Independently Associated with Progression of Nephropathy in Type 2 Diabetes Mellitus. Diabetologia 2018, 61, 1212–1219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. de Oliveira, F.L.; Gatto, M.; Bassi, N.; Luisetto, R.; Ghirardello, A.; Punzi, L.; Doria, A. Galectin-3 in Autoimmunity and Autoimmune Diseases. Exp. Biol. Med. 2015, 240, 1019–1028. [Google Scholar] [CrossRef] [Green Version]
  14. Boza-Serrano, A.; Ruiz, R.; Sanchez-Varo, R.; García-Revilla, J.; Yang, Y.; Jimenez-Ferrer, I.; Paulus, A.; Wennström, M.; Vilalta, A.; Allendorf, D.; et al. Galectin-3, a Novel Endogenous TREM2 Ligand, Detrimentally Regulates Inflammatory Response in Alzheimer’s Disease. Acta. Neuropathol. 2019, 138, 251–273. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  15. Aureli, A.; Del Cornò, M.; Marziani, B.; Gessani, S.; Conti, L. Highlights on the Role of Galectin-3 in Colorectal Cancer and the Preventive/Therapeutic Potential of Food-Derived Inhibitors. Cancers 2022, 15, 52. [Google Scholar] [CrossRef]
  16. Lima, T.; Perpétuo, L.; Henrique, R.; Fardilha, M.; Leite-Moreira, A.; Bastos, J.; Vitorino, R. Galectin-3 in Prostate Cancer and Heart Diseases: A Biomarker for These Two Frightening Pathologies? Mol. Biol. Rep. 2022, 50, 2763–2778. [Google Scholar] [CrossRef]
  17. Frangogiannis, N. Transforming Growth Factor-β in Tissue Fibrosis. J. Exp. Med. 2020, 217, e20190103. [Google Scholar] [CrossRef]
  18. Meng, X.-M.; Nikolic-Paterson, D.J.; Lan, H.Y. TGF-β: The Master Regulator of Fibrosis. Nat. Rev. Nephrol. 2016, 12, 325–338. [Google Scholar] [CrossRef]
  19. Elliott, M.J.; Strasser, A.; Metcalf, D. Selective Up-Regulation of Macrophage Function in Granulocyte-Macrophage Colony-Stimulating Factor Transgenic Mice. J. Immunol. 1991, 147, 2957–2963. [Google Scholar] [CrossRef]
  20. Parmar, N.; Tatler, A.; MacKinnon, A.; Ford, P.; Jenkins, R. g. Galectin-3 Promotes Transforming Growth Factor b (TGFb) Activity in the Development of Idiopathic Pulmonary Fibrosis. In A68. Molecular Determinants of Remodeling in Lung Fibrosis; American Thoracic Society International Conference Abstracts; American Thoracic Society: New York, NY, USA, 2018; p. A2207. [Google Scholar]
  21. Falkenham, A.; de Antueno, R.; Rosin, N.; Betsch, D.; Lee, T.D.G.; Duncan, R.; Légaré, J.-F. Nonclassical Resident Macrophages Are Important Determinants in the Development of Myocardial Fibrosis. Am. J. Pathol. 2015, 185, 927–942. [Google Scholar] [CrossRef]
  22. Lippi, G.; Cervellin, G.; Sanchis-Gomar, F. Galectin-3 in Atrial Fibrillation: Simple Bystander, Player or Both? Clin. Biochem. 2015, 48, 818–822. [Google Scholar] [CrossRef] [PubMed]
  23. Shen, H.; Wang, J.; Min, J.; Xi, W.; Gao, Y.; Yin, L.; Yu, Y.; Liu, K.; Xiao, J.; Zhang, Y.-F.; et al. Activation of TGF-Β1/α-SMA/Col I Profibrotic Pathway in Fibroblasts by Galectin-3 Contributes to Atrial Fibrosis in Experimental Models and Patients. Cell Physiol. Biochem. 2018, 47, 851–863. [Google Scholar] [CrossRef] [PubMed]
  24. Vergaro, G.; Prud’homme, M.; Fazal, L.; Merval, R.; Passino, C.; Emdin, M.; Samuel, J.-L.; Cohen Solal, A.; Delcayre, C. Inhibition of Galectin-3 Pathway Prevents Isoproterenol-Induced Left Ventricular Dysfunction and Fibrosis in Mice. Hypertension 2016, 67, 606–612. [Google Scholar] [CrossRef] [Green Version]
  25. Tran, S.K.; Ngo, T.H.; Lai, T.T.; Truong, G.K.; Tran, K.D.D.; Vo, P.M.; Nguyen, P.T.; Nguyen, P.H.; Nguyen, T.T.; Nguyen, O.T.K.; et al. Effectiveness of Spironolactone in Terms of Galectin-3 Levels in Patients with Heart Failure with a Reduced Ejection Fraction in the Vietnamese Population. Healthcare 2023, 11, 253. [Google Scholar] [CrossRef]
  26. Grupper, A.; Nativi-Nicolau, J.; Maleszewski, J.J.; Geske, J.R.; Kremers, W.K.; Edwards, B.S.; Kushwaha, S.S.; Pereira, N.L. Circulating Galectin-3 Levels Are Persistently Elevated After Heart Transplantation and Are Associated With Renal Dysfunction. JACC Heart Fail. 2016, 4, 847–856. [Google Scholar] [CrossRef] [PubMed]
  27. Sarhene, M.; Wang, Y.; Wei, J.; Huang, Y.; Li, M.; Li, L.; Acheampong, E.; Zhengcan, Z.; Xiaoyan, Q.; Yunsheng, X.; et al. Biomarkers in Heart Failure: The Past, Current and Future. Heart Fail. Rev. 2019, 24, 867–903. [Google Scholar] [CrossRef] [PubMed]
  28. Zhang, G.; Wu, Y. Circulating Galectin-3 and Atrial Fibrillation Recurrence after Catheter Ablation: A Meta-Analysis. Cardiovasc. Ther. 2019, 2019, 4148129. [Google Scholar] [CrossRef] [Green Version]
  29. Andre, C.; Piver, E.; Perault, R.; Bisson, A.; Pucheux, J.; Vermes, E.; Pierre, B.; Fauchier, L.; Babuty, D.; Clementy, N. Galectin-3 Predicts Response and Outcomes after Cardiac Resynchronization Therapy. J. Transl. Med. 2018, 16, 299. [Google Scholar] [CrossRef] [Green Version]
  30. Baggen, V.J.M.; van den Bosch, A.E.; Eindhoven, J.A.; Menting, M.E.; Witsenburg, M.; Cuypers, J.A.A.E.; Boersma, E.; Roos-Hesselink, J.W. Prognostic Value of Galectin-3 in Adults with Congenital Heart Disease. Heart 2018, 104, 394–400. [Google Scholar] [CrossRef]
  31. Agnello, L.; Bivona, G.; Lo Sasso, B.; Scazzone, C.; Bazan, V.; Bellia, C.; Ciaccio, M. Galectin-3 in Acute Coronary Syndrome. Clin. Biochem. 2017, 50, 797–803. [Google Scholar] [CrossRef]
  32. Gehlken, C.; Suthahar, N.; Meijers, W.C.; de Boer, R.A. Galectin-3 in Heart Failure: An Update of the Last 3 Years. Heart Fail. Clin. 2018, 14, 75–92. [Google Scholar] [CrossRef] [PubMed]
  33. Magnussen, C.; Blankenberg, S. Biomarkers for Heart Failure: Small Molecules with High Clinical Relevance. J. Intern. Med. 2018, 283, 530–543. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Suthahar, N.; Meijers, W.C.; Silljé, H.H.W.; Ho, J.E.; Liu, F.-T.; de Boer, R.A. Galectin-3 Activation and Inhibition in Heart Failure and Cardiovascular Disease: An Update. Theranostics 2018, 8, 593–609. [Google Scholar] [CrossRef] [PubMed]
  35. Motiwala, S.R.; Szymonifka, J.; Belcher, A.; Weiner, R.B.; Baggish, A.L.; Sluss, P.; Gaggin, H.K.; Bhardwaj, A.; Januzzi, J.L. Serial Measurement of Galectin-3 in Patients with Chronic Heart Failure: Results from the ProBNP Outpatient Tailored Chronic Heart Failure Therapy (PROTECT) Study. Eur. J. Heart Fail. 2013, 15, 1157–1163. [Google Scholar] [CrossRef]
  36. Sharma, U.C.; Pokharel, S.; van Brakel, T.J.; van Berlo, J.H.; Cleutjens, J.P.M.; Schroen, B.; André, S.; Crijns, H.J.G.M.; Gabius, H.-J.; Maessen, J.; et al. Galectin-3 Marks Activated Macrophages in Failure-Prone Hypertrophied Hearts and Contributes to Cardiac Dysfunction. Circulation 2004, 110, 3121–3128. [Google Scholar] [CrossRef]
  37. Yu, L.; Ruifrok, W.P.T.; Meissner, M.; Bos, E.M.; van Goor, H.; Sanjabi, B.; van der Harst, P.; Pitt, B.; Goldstein, I.J.; Koerts, J.A.; et al. Genetic and Pharmacological Inhibition of Galectin-3 Prevents Cardiac Remodeling by Interfering with Myocardial Fibrogenesis. Circ. Heart Fail. 2013, 6, 107–117. [Google Scholar] [CrossRef] [Green Version]
  38. Rieth, A.J.; Jung, C.; Gall, H.; Rolf, A.; Mitrovic, V.; Hamm, C.W.; Sperzel, J.; Liebetrau, C. Association of Galectin-3 with Changes in Left Ventricular Function in Recent-Onset Dilated Cardiomyopathy. Biomarkers 2019, 24, 652–658. [Google Scholar] [CrossRef]
  39. Yao, Y.; Shen, D.; Chen, R.; Ying, C.; Wang, C.; Guo, J.; Zhang, G. Galectin-3 Predicts Left Ventricular Remodeling of Hypertension. J. Clin. Hypertens 2016, 18, 506–511. [Google Scholar] [CrossRef] [Green Version]
  40. Perea, R.J.; Morales-Ruiz, M.; Ortiz-Perez, J.T.; Bosch, X.; Andreu, D.; Borras, R.; Acosta, J.; Penela, D.; Prat-González, S.; de Caralt, T.M.; et al. Utility of Galectin-3 in Predicting Post-Infarct Remodeling after Acute Myocardial Infarction Based on Extracellular Volume Fraction Mapping. Int. J. Cardiol. 2016, 223, 458–464. [Google Scholar] [CrossRef]
  41. van der Velde, A.R.; Lexis, C.P.H.; Meijers, W.C.; van der Horst, I.C.; Lipsic, E.; Dokter, M.M.; van Veldhuisen, D.J.; van der Harst, P.; de Boer, R.A. Galectin-3 and SST2 in Prediction of Left Ventricular Ejection Fraction after Myocardial Infarction. Clin. Chim. Acta 2016, 452, 50–57. [Google Scholar] [CrossRef]
  42. Mitić, B.; Jovanović, A.; Nikolić, V.N.; Stokanović, D.; Andrejić, O.M.; Vučić, R.M.; Pavlović, M.; Ignjatović, A.; Momčilović, S. Trend of Galectin-3 Levels in Patients with Non-ST-Elevation and ST-Elevation Myocardial Infarction. Medicina 2022, 58, 286. [Google Scholar] [CrossRef]
  43. Ardelean, C.L.; Pescariu, S.; Lighezan, D.F.; Pleava, R.; Ursoniu, S.; Nadasan, V.; Mihaicuta, S. Particularities of Older Patients with Obstructive Sleep Apnea and Heart Failure with Mid-Range Ejection Fraction. Medicina 2019, 55, 449. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. de Boer, R.A.; Lok, D.J.A.; Jaarsma, T.; van der Meer, P.; Voors, A.A.; Hillege, H.L.; van Veldhuisen, D.J. Predictive Value of Plasma Galectin-3 Levels in Heart Failure with Reduced and Preserved Ejection Fraction. Ann. Med. 2011, 43, 60–68. [Google Scholar] [CrossRef] [PubMed]
  45. Shi, Y.; Dong, G.; Liu, J.; Shuang, X.; Liu, C.; Yang, C.; Qing, W.; Qiao, W. Clinical Implications of Plasma Galectin-3 in Heart Failure With Preserved Ejection Fraction: A Meta-Analysis. Front. Cardiovasc. Med. 2022, 9, 854501. [Google Scholar] [CrossRef] [PubMed]
  46. Baccouche, B.M.; Rhodenhiser, E.G. Galectin-3 and HFpEF: Clarifying an Emerging Relationship. Curr. Cardiol. Rev. 2023, 19. [Google Scholar] [CrossRef]
  47. Chen, H.; Chhor, M.; Rayner, B.S.; McGrath, K.; McClements, L. Evaluation of the Diagnostic Accuracy of Current Biomarkers in Heart Failure with Preserved Ejection Fraction: A Systematic Review and Meta-Analysis. Arch. Cardiovasc. Dis. 2021, 114, 793–804. [Google Scholar] [CrossRef]
  48. Cheng, W.-L.; Chen, Y.-C.; Li, S.-J.; Lee, T.-I.; Lee, T.-W.; Higa, S.; Chung, C.-C.; Kao, Y.-H.; Chen, S.-A.; Chen, Y.-J. Galectin-3 Enhances Atrial Remodelling and Arrhythmogenesis through CD98 Signalling. Acta. Physiol. 2022, 234, e13784. [Google Scholar] [CrossRef]
  49. Hernández-Romero, D.; Vílchez, J.A.; Lahoz, Á.; Romero-Aniorte, A.I.; Jover, E.; García-Alberola, A.; Jara-Rubio, R.; Martínez, C.M.; Valdés, M.; Marín, F. Galectin-3 as a Marker of Interstitial Atrial Remodelling Involved in Atrial Fibrillation. Sci. Rep. 2017, 7, 40378. [Google Scholar] [CrossRef] [Green Version]
  50. Allessie, M.; Ausma, J.; Schotten, U. Electrical, Contractile and Structural Remodeling during Atrial Fibrillation. Cardiovasc. Res. 2002, 54, 230–246. [Google Scholar] [CrossRef]
  51. Ho, J.E.; Yin, X.; Levy, D.; Vasan, R.S.; Magnani, J.W.; Ellinor, P.T.; McManus, D.D.; Lubitz, S.A.; Larson, M.G.; Benjamin, E.J. Galectin 3 and Incident Atrial Fibrillation in the Community. Am. Heart J. 2014, 167, 729–734.e1. [Google Scholar] [CrossRef] [Green Version]
  52. Du, X.-J.; Zhao, W.-B.; Nguyen, M.-N.; Lu, Q.; Kiriazis, H. β-Adrenoceptor Activation Affects Galectin-3 as a Biomarker and Therapeutic Target in Heart Disease. Br. J. Pharmacol. 2019, 176, 2449–2464. [Google Scholar] [CrossRef] [PubMed]
  53. Wachtell, K.; Lehto, M.; Gerdts, E.; Olsen, M.H.; Hornestam, B.; Dahlöf, B.; Ibsen, H.; Julius, S.; Kjeldsen, S.E.; Lindholm, L.H.; et al. Angiotensin II Receptor Blockade Reduces New-Onset Atrial Fibrillation and Subsequent Stroke Compared to Atenolol: The Losartan Intervention for End Point Reduction in Hypertension (LIFE) Study. J. Am. Coll. Cardiol. 2005, 45, 712–719. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Takemoto, Y.; Ramirez, R.J.; Yokokawa, M.; Kaur, K.; Ponce-Balbuena, D.; Sinno, M.C.; Willis, B.C.; Ghanbari, H.; Ennis, S.R.; Guerrero-Serna, G.; et al. Galectin-3 Regulates Atrial Fibrillation Remodeling and Predicts Catheter Ablation Outcomes. JACC Basic. Transl. Sci. 2016, 1, 143–154. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Erdem, K.; Kurtoglu, E.; Oc, M.; Oc, B.; Ilgenli, T.F.; Unlu, A.; Eryavuz Onmaz, D. The Plasma Galectin-3 Level Has High Specificity and Sensitivity for Predicting Postoperative Atrial Fibrillation after Coronary Artery Bypass Surgery. Eur. Rev. Med. Pharmacol. Sci. 2022, 26, 9072–9078. [Google Scholar] [CrossRef]
  56. Bening, C.; Mazalu, E.-A.; Yaqub, J.; Alhussini, K.; Glanowski, M.; Kottmann, T.; Leyh, R. Atrial Contractility and Fibrotic Biomarkers Are Associated with Atrial Fibrillation after Elective Coronary Artery Bypass Grafting. J. Thorac. Cardiovasc. Surg. 2020, 159, 515–523. [Google Scholar] [CrossRef]
  57. Alexandre, J.; Saloux, E.; Chequel, M.; Allouche, S.; Ollitrault, P.; Plane, A.-F.; Legallois, D.; Fischer, M.-O.; Saplacan, V.; Buklas, D.; et al. Preoperative Plasma Aldosterone and the Risk of Atrial Fibrillation after Coronary Artery Bypass Surgery: A Prospective Cohort Study. J. Hypertens 2016, 34, 2449–2457. [Google Scholar] [CrossRef]
  58. King, M.; Stambulic, T.; Kirupaharan, S.; Baranchuk, A.; Rabinovich, G.A.; Payne, D.; El-Diasty, M. Galectin-3 as a Predictor of Post Cardiac Surgery Atrial Fibrillation: A Scoping Review. Curr. Probl. Cardiol. 2022, 47, 101314. [Google Scholar] [CrossRef]
  59. Chhor, M.; Law, W.; Pavlovic, M.; Aksentijevic, D.; McGrath, K.; McClements, L. Diagnostic and Prognostic Biomarkers Reflective of Cardiac Remodelling in Diabetes Mellitus: A Scoping Review. Diabet. Med. 2023, 40, e15064. [Google Scholar] [CrossRef]
  60. Yilmaz, H.; Cakmak, M.; Inan, O.; Darcin, T.; Akcay, A. Increased Levels of Galectin-3 Were Associated with Prediabetes and Diabetes: New Risk Factor? J. Endocrinol. Investig. 2015, 38, 527–533. [Google Scholar] [CrossRef]
  61. Amin, H.Z.; Amin, L.Z.; Wijaya, I.P. Galectin-3: A Novel Biomarker For The Prognosis Of Heart Failure. Med. Pharm. Rep. 2017, 90, 129–132. [Google Scholar] [CrossRef] [Green Version]
  62. Blanda, V.; Bracale, U.M.; Di Taranto, M.D.; Fortunato, G. Galectin-3 in Cardiovascular Diseases. Int. J. Mol. Sci. 2020, 21, 9232. [Google Scholar] [CrossRef] [PubMed]
  63. van Kimmenade, R.R.; Januzzi, J.L.; Ellinor, P.T.; Sharma, U.C.; Bakker, J.A.; Low, A.F.; Martinez, A.; Crijns, H.J.; MacRae, C.A.; Menheere, P.P.; et al. Utility of Amino-Terminal pro-Brain Natriuretic Peptide, Galectin-3, and Apelin for the Evaluation of Patients with Acute Heart Failure. J. Am. Coll. Cardiol. 2006, 48, 1217–1224. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. McGinn, C.; Casey, F.A.; Watson, C.; Morrison, L. Paediatric Heart Failure—Understanding the Pathophysiology and the Current Role of Cardiac Biomarkers in Clinical Practice. Cardiol Young 2023, 33, 1–11. [Google Scholar] [CrossRef]
  65. Cheng, W.; Fuernau, G.; Desch, S.; Freund, A.; Feistritzer, H.-J.; Pöss, J.; Besler, C.; Lurz, P.; Büttner, P.; Thiele, H. Circulating Galectin-3 in Patients with Cardiogenic Shock Complicating Acute Myocardial Infarction Treated with Mild Hypothermia: A Biomarker Sub-Study of the SHOCK-COOL Trial. J. Clin. Med. 2022, 11, 7168. [Google Scholar] [CrossRef] [PubMed]
  66. Pourafkari, L.; Tajlil, A.; Nader, N.D. Biomarkers in Diagnosing and Treatment of Acute Heart Failure. Biomark Med. 2019, 13, 1235–1249. [Google Scholar] [CrossRef] [PubMed]
  67. Caravaca Perez, P.; González-Juanatey, J.R.; Nuche, J.; Matute-Blanco, L.; Serrano, I.; Martínez Selles, M.; Vázquez García, R.; Martínez Dolz, L.; Gómez-Bueno, M.; Pascual Figal, D.; et al. Renal Function Impact in the Prognostic Value of Galectin-3 in Acute Heart Failure. Front. Cardiovasc. Med. 2022, 9, 861651. [Google Scholar] [CrossRef] [PubMed]
  68. Gürgöze, M.T.; van Vark, L.C.; Baart, S.J.; Kardys, I.; Akkerhuis, K.M.; Manintveld, O.C.; Postmus, D.; Hillege, H.L.; Lesman-Leegte, I.; Asselbergs, F.W.; et al. Multimarker Analysis of Serially Measured GDF-15, NT-ProBNP, ST2, GAL-3, CTnI, Creatinine, and Prognosis in Acute Heart Failure. Circ. Heart Fail. 2023, 16, e009526. [Google Scholar] [CrossRef]
  69. Wang, C.-H.; Yang, N.-I.; Liu, M.-H.; Hsu, K.-H.; Kuo, L.-T. Estimating Systemic Fibrosis by Combining Galectin-3 and ST2 Provides Powerful Risk Stratification Value for Patients after Acute Decompensated Heart Failure. Cardiol. J. 2016, 23, 563–572. [Google Scholar] [CrossRef] [Green Version]
  70. Baccouche, B.M.; A Mahmoud, M.; Nief, C.; Patel, K.; Natterson-Horowitz, B. Galectin-3 Is Associated with Heart Failure Incidence: A Meta-Analysis. Curr. Cardiol. Rev. 2022, 19, 50–62. [Google Scholar] [CrossRef]
  71. Ho, J.E.; Liu, C.; Lyass, A.; Courchesne, P.; Pencina, M.J.; Vasan, R.S.; Larson, M.G.; Levy, D. Galectin-3, a Marker of Cardiac Fibrosis, Predicts Incident Heart Failure in the Community. J. Am. Coll. Cardiol. 2012, 60, 1249–1256. [Google Scholar] [CrossRef] [Green Version]
  72. Florido, R.; Kwak, L.; Echouffo-Tcheugui, J.B.; Zhang, S.; Michos, E.D.; Nambi, V.; Goldberg, R.B.; Hoogeveen, R.C.; Lazo, M.; Gerstenblith, G.; et al. Obesity, Galectin-3, and Incident Heart Failure: The ARIC Study. J. Am. Heart Assoc. 2022, 11, e023238. [Google Scholar] [CrossRef] [PubMed]
  73. Jagodzinski, A.; Havulinna, A.S.; Appelbaum, S.; Zeller, T.; Jousilahti, P.; Skytte-Johanssen, S.; Hughes, M.F.; Blankenberg, S.; Salomaa, V. Predictive Value of Galectin-3 for Incident Cardiovascular Disease and Heart Failure in the Population-Based FINRISK 1997 Cohort. Int. J. Cardiol. 2015, 192, 33–39. [Google Scholar] [CrossRef] [PubMed]
  74. Imran, T.F.; Shin, H.J.; Mathenge, N.; Wang, F.; Kim, B.; Joseph, J.; Gaziano, J.M.; Djoussé, L. Meta-Analysis of the Usefulness of Plasma Galectin-3 to Predict the Risk of Mortality in Patients With Heart Failure and in the General Population. Am. J. Cardiol. 2017, 119, 57–64. [Google Scholar] [CrossRef] [PubMed]
  75. Lok, D.J.A.; Van Der Meer, P.; de la Porte, P.W.B.-A.; Lipsic, E.; Van Wijngaarden, J.; Hillege, H.L.; van Veldhuisen, D.J. Prognostic Value of Galectin-3, a Novel Marker of Fibrosis, in Patients with Chronic Heart Failure: Data from the DEAL-HF Study. Clin. Res. Cardiol. 2010, 99, 323–328. [Google Scholar] [CrossRef] [Green Version]
  76. Baran, J.; Niewiara, Ł.; Podolec, J.; Siedliński, M.; Józefczuk, E.; Bernacik, A.; Badacz, R.; Przewłocki, T.; Pieniążek, P.; Żmudka, K.; et al. Serum and Vascular Stiffness Biomarkers Associated with the Severity of Degenerative Aortic Valve Stenosis and Cardiovascular Outcomes. J. Cardiovasc. Dev. Dis. 2022, 9, 193. [Google Scholar] [CrossRef] [PubMed]
  77. Khadeja Bi, A.; Santhosh, V.; Sigamani, K. Levels of Galectin-3 in Chronic Heart Failure: A Case-Control Study. Cureus 2022, 14, e28310. [Google Scholar] [CrossRef]
  78. Anand, I.S.; Rector, T.S.; Kuskowski, M.; Adourian, A.; Muntendam, P.; Cohn, J.N. Baseline and Serial Measurements of Galectin-3 in Patients with Heart Failure: Relationship to Prognosis and Effect of Treatment with Valsartan in the Val-HeFT. Eur. J. Heart Fail. 2013, 15, 511–518. [Google Scholar] [CrossRef]
  79. Miller, W.L.; Saenger, A.K.; Grill, D.E.; Slusser, J.P.; Bayes-Genis, A.; Jaffe, A.S. Prognostic Value of Serial Measurements of Soluble Suppression of Tumorigenicity 2 and Galectin-3 in Ambulatory Patients With Chronic Heart Failure. J. Card. Fail. 2016, 22, 249–255. [Google Scholar] [CrossRef]
  80. Yancy, C.W.; Jessup, M.; Bozkurt, B.; Butler, J.; Casey, D.E.; Drazner, M.H.; Fonarow, G.C.; Geraci, S.A.; Horwich, T.; Januzzi, J.L.; et al. 2013 ACCF/AHA Guideline for the Management of Heart Failure: A Report of the American College of Cardiology Foundation/American Heart Association Task Force on Practice Guidelines. J. Am. Coll. Cardiol. 2013, 62, e147–e239. [Google Scholar] [CrossRef] [Green Version]
  81. Meeusen, J.W.; Johnson, J.N.; Gray, A.; Wendt, P.; Jefferies, J.L.; Jaffe, A.S.; Donato, L.J.; Saenger, A.K. Soluble ST2 and Galectin-3 in Pediatric Patients without Heart Failure. Clin. Biochem. 2015, 48, 1337–1340. [Google Scholar] [CrossRef]
  82. Caselli, C.; Ragusa, R.; Prontera, C.; Cabiati, M.; Cantinotti, M.; Federico, G.; Del Ry, S.; Trivella, M.G.; Clerico, A. Distribution of Circulating Cardiac Biomarkers in Healthy Children: From Birth through Adulthood. Biomark. Med. 2016, 10, 357–365. [Google Scholar] [CrossRef] [PubMed]
  83. Schindler, E.I.; Szymanski, J.J.; Hock, K.G.; Geltman, E.M.; Scott, M.G. Short- and Long-Term Biologic Variability of Galectin-3 and Other Cardiac Biomarkers in Patients with Stable Heart Failure and Healthy Adults. Clin. Chem. 2016, 62, 360–366. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Shen, Q.; Chen, W.; Liu, J.; Liang, Q. Galectin-3 Aggravates Pulmonary Arterial Hypertension via Immunomodulation in Congenital Heart Disease. Life Sci. 2019, 232, 116546. [Google Scholar] [CrossRef] [PubMed]
  85. Fenster, B.E.; Lasalvia, L.; Schroeder, J.D.; Smyser, J.; Silveira, L.J.; Buckner, J.K.; Brown, K.K. Galectin-3 Levels Are Associated with Right Ventricular Functional and Morphologic Changes in Pulmonary Arterial Hypertension. Heart Vessel. 2016, 31, 939–946. [Google Scholar] [CrossRef]
  86. Keranov, S.; Dörr, O.; Jafari, L.; Liebetrau, C.; Keller, T.; Troidl, C.; Riehm, J.; Rutsatz, W.; Bauer, P.; Kriechbaum, S.; et al. Osteopontin and Galectin-3 as Biomarkers of Maladaptive Right Ventricular Remodeling in Pulmonary Hypertension. Biomark. Med. 2021, 15, 1021–1034. [Google Scholar] [CrossRef] [PubMed]
  87. Kowalik, E.; Kuśmierczyk-Droszcz, B.; Klisiewicz, A.; Wróbel, A.; Lutyńska, A.; Gawor, M.; Niewiadomska, J.; Lipczyńska, M.; Biernacka, E.K.; Grzybowski, J.; et al. Galectin-3 Plasma Levels in Adult Congenital Heart Disease and the Pressure Overloaded Right Ventricle: Reason Matters. Biomark Med. 2020, 14, 1197–1205. [Google Scholar] [CrossRef]
  88. Mohammed, L.A.; Gafar, H.S.; Hussien, N.R. Galectin-3 as Early Detector of Heart Failure in Children with Congenital Acyanotic Heart Disease. Clin. Med. Diagn. 2014, 4, 90–98. [Google Scholar]
  89. van den Bosch, E.; van Genuchten, W.J.; Luijnenburg, S.E.; Duppen, N.; Kamphuis, V.P.; Roos-Hesselink, J.W.; Bartelds, B.; Roest, A.A.W.; Breur, J.M.P.J.; Blom, N.A.; et al. Associations between Blood Biomarkers, Cardiac Function and Adverse Outcome in a Young Tetralogy of Fallot Cohort. Int. J. Cardiol. 2022, 361, 31–37. [Google Scholar] [CrossRef]
  90. Kotby, A.A.; Youssef, O.I.; Elmaraghy, M.O.; El Sharkawy, O.S. Galectin-3 in Children with Chronic Heart Failure with Normal and Reduced Ejection Fraction: Relationship to Disease Severity. Pediatr. Cardiol. 2017, 38, 95–102. [Google Scholar] [CrossRef]
  91. Elhewala, A.A.S.; Ibrahem, M.M.; Abdel Hafez, E.S. Galectin-3 as a Biomarker of Heart Failure in Children with Congenital Heart Disease. Egypt. J. Hosp. Med. 2020, 80, 1008–1013. [Google Scholar] [CrossRef]
  92. Saleh, N.; Khattab, A.; Rizk, M.; Salem, S.; Abo-Haded, H. Value of Galectin-3 Assay in Children with Heart Failure Secondary to Congenital Heart Diseases: A Prospective Study. BMC Pediatr. 2020, 20, 537. [Google Scholar] [CrossRef] [PubMed]
  93. Cura, C.; Argun, M.; Koçer, D. Aldosterone, Galectin-3, and NTproBNP Levels and Their Values as Biomarkers in Infants with Ventricular Septal Defect. Turk. Kardiyol. Dern. Ars. 2022, 50, 131–136. [Google Scholar] [CrossRef] [PubMed]
  94. Frogoudaki, A.A.; Pantelakis, I.; Bistola, V.; Kroupis, C.; Birba, D.; Ikonomidis, I.; Alexopoulos, D.; Filippatos, G.; Parissis, J. Global Longitudinal Strain of the Systemic Ventricle Is Correlated with Plasma Galectin-3 and Predicts Major Cardiovascular Events in Adult Patients with Congenital Heart Disease. Medicina 2020, 56, 305. [Google Scholar] [CrossRef]
  95. Geenen, L.W.; van Grootel, R.W.J.; Akman, K.; Baggen, V.J.M.; Menting, M.E.; Eindhoven, J.A.; Cuypers, J.A.A.E.; Boersma, E.; van den Bosch, A.E.; Roos-Hesselink, J.W. Exploring the Prognostic Value of Novel Markers in Adults With a Systemic Right Ventricle. J. Am. Heart Assoc. 2019, 8, e013745. [Google Scholar] [CrossRef]
  96. Xiao, M.; Zhang, M.; Bie, M.; Wang, X.; Guo, J.; Xiao, H. Galectin-3 Induces Atrial Fibrosis by Activating the TGF-Β1/Smad Pathway in Patients with Atrial Fibrillation. Cardiology 2020, 145, 446–455. [Google Scholar] [CrossRef] [PubMed]
  97. Pietrzak, R.; Książczyk, T.M.; Górska, E.; Małek, Ł.A.; Werner, B. Evaluation of Galectin-3 Plasma Concentration in Adolescents with Ventricular Arrhythmia. Int. J. Environ. Res. Public Health 2021, 18, 2410. [Google Scholar] [CrossRef] [PubMed]
  98. Sodhi, S.S.; Zhang, T.J.; McDonald, R.; Al Rashida, V.; Kondapalli, N.; Barger, P.; Ludbrook, P.; Cedars, A.M. Effects of Eplerenone on Markers of Myocardial Fibrosis, 6-Minute Walk Distance, and Quality of Life in Adults with Tetralogy of Fallot and Complete Transposition of the Great Arteries. Proceedings 2018, 31, 12–19. [Google Scholar] [CrossRef] [Green Version]
  99. Li, X.-Y.; Zheng, Y.; Long, Y.; Zhang, X.; Zhang, L.; Tian, D.; Zhou, D.; Lv, Q.-Z. Effect of Iloprost on Biomarkers in Patients with Congenital Heart Disease-Pulmonary Arterial Hypertension. Clin. Exp. Pharmacol. Physiol. 2017, 44, 914–923. [Google Scholar] [CrossRef] [Green Version]
  100. Dos, L.; Pujadas, S.; Estruch, M.; Mas, A.; Ferreira-González, I.; Pijuan, A.; Serra, R.; Ordóñez-Llanos, J.; Subirana, M.; Pons-Lladó, G.; et al. Eplerenone in Systemic Right Ventricle: Double Blind Randomized Clinical Trial. The Evedes Study. Int. J. Cardiol. 2013, 168, 5167–5173. [Google Scholar] [CrossRef]
  101. Hu, D.-J.; Xu, J.; Du, W.; Zhang, J.-X.; Zhong, M.; Zhou, Y.-N. Cardiac Magnetic Resonance and Galectin-3 Level as Predictors of Prognostic Outcomes for Non-Ischemic Cardiomyopathy Patients. Int. J. Cardiovasc. Imaging. 2016, 32, 1725–1733. [Google Scholar] [CrossRef]
  102. Wojciechowska, C.; Romuk, E.; Nowalany-Kozielska, E.; Jacheć, W. Serum Galectin-3 and ST2 as Predictors of Unfavorable Outcome in Stable Dilated Cardiomyopathy Patients. Hellenic. J. Cardiol. 2017, 58, 350–359. [Google Scholar] [CrossRef] [PubMed]
  103. Nguyen, M.-N.; Ziemann, M.; Kiriazis, H.; Su, Y.; Thomas, Z.; Lu, Q.; Donner, D.G.; Zhao, W.-B.; Rafehi, H.; Sadoshima, J.; et al. Galectin-3 Deficiency Ameliorates Fibrosis and Remodeling in Dilated Cardiomyopathy Mice with Enhanced Mst1 Signaling. Am. J. Physiol. Heart Circ. Physiol. 2019, 316, H45–H60. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Parker, D.M.; Everett, A.D.; Stabler, M.E.; Vricella, L.; Jacobs, M.L.; Jacobs, J.P.; Parikh, C.R.; Pasquali, S.K.; Brown, J.R. Novel Biomarkers Improve Prediction of 365-Day Readmission After Pediatric Congenital Heart Surgery. Ann. Thorac. Surg. 2020, 109, 164–170. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Brown, J.R.; Stabler, M.E.; Parker, D.M.; Vricella, L.; Pasquali, S.; Leyenaar, J.K.; Bohm, A.R.; MacKenzie, T.; Parikh, C.; Jacobs, M.L.; et al. Biomarkers Improve Prediction of 30-Day Unplanned Readmission or Mortality after Paediatric Congenital Heart Surgery. Cardiol. Young 2019, 29, 1051–1056. [Google Scholar] [CrossRef]
  106. Parker, D.M.; Everett, A.D.; Stabler, M.E.; Vricella, L.; Jacobs, M.L.; Jacobs, J.P.; Thiessen-Philbrook, H.; Parikh, C.R.; Brown, J.R. Biomarkers Associated with 30-Day Readmission and Mortality after Pediatric Congenital Heart Surgery. J. Card. Surg. 2019, 34, 329–336. [Google Scholar] [CrossRef]
  107. Opotowsky, A.R.; Baraona, F.; Owumi, J.; Loukas, B.; Singh, M.N.; Valente, A.M.; Wu, F.; Cheng, S.; Veldtman, G.; Rimm, E.B.; et al. Galectin-3 Is Elevated and Associated With Adverse Outcomes in Patients With Single-Ventricle Fontan Circulation. J. Am. Heart Assoc. 2016, 5, e002706. [Google Scholar] [CrossRef] [Green Version]
  108. DiLorenzo, M.P.; DeCost, G.; Mai, A.D.; Hughes, N.; Goldmuntz, E.; Jones, A.; Fogel, M.A.; Mercer-Rosa, L. Comparison of Serum Biomarkers of Myocardial Fibrosis with Cardiac Magnetic Resonance in Patients Operated for Tetralogy of Fallot. Int. J. Cardiol. 2022, 358, 27–33. [Google Scholar] [CrossRef]
  109. Făgărășan, A.; Săsăran, M.O. The Predictive Role of Plasma Biomarkers in the Evolution of Aortopathies Associated with Congenital Heart Malformations. Int. J. Mol. Sci. 2022, 23, 4993. [Google Scholar] [CrossRef]
  110. Frank, B.S.; Urban, T.T.; Lewis, K.; Tong, S.; Cassidy, C.; Mitchell, M.B.; Nichols, C.S.; Davidson, J.A. Circulating Biomarkers of Left Ventricular Hypertrophy in Pediatric Coarctation of the Aorta. Congenit. Heart Dis. 2019, 14, 446–453. [Google Scholar] [CrossRef]
  111. van den Bosch, E.; Bossers, S.S.M.; Kamphuis, V.P.; Boersma, E.; Roos-Hesselink, J.W.; Breur, J.M.P.J.; Ten Harkel, A.D.J.; Kapusta, L.; Bartelds, B.; Roest, A.A.W.; et al. Associations Between Blood Biomarkers, Cardiac Function, and Adverse Outcome in a Young Fontan Cohort. J. Am. Heart Assoc. 2021, 10, e015022. [Google Scholar] [CrossRef]
  112. Geenen, L.W.; Uchoa de Assis, L.; Baggen, V.J.M.; Eindhoven, J.A.; Cuypers, J.A.A.E.; Boersma, E.; Roos-Hesselink, J.W.; van den Bosch, A.E. Evolution of Blood Biomarker Levels Following Percutaneous Atrial Septal Defect Closure in Adults. Int. J. Cardiol. Heart Vasc. 2020, 30, 100582. [Google Scholar] [CrossRef] [PubMed]
  113. Dudnyk, V.; Zborovska, O.; Vyzhga, Y.; Khromykh, K.; Zvenigorodska, G.; Popov, V.; Bakhnivskyi, V. Instrumental Assessment of Myocardial Function by Doppler Tissue Imaging in Children with Congenital Heart Defects after Surgical Correction and Its Correlation with Galectin-3 Amount as Biological Marker of Cardiac Fibrosis. Wiad. Lek. 2019, 72, 1281–1287. [Google Scholar] [CrossRef] [PubMed]
  114. Karali, K.; Makedou, K.; Kallifatidis, A.; Didagelos, M.; Giannakoulas, G.; Davos, C.H.; Karamitsos, T.D.; Ziakas, A.; Karvounis, H.; Hadjimiltiades, S. The Interplay between Myocardial Fibrosis, Strain Imaging and Collagen Biomarkers in Adults with Repaired Tetralogy of Fallot. Diagnostics 2021, 11, 2101. [Google Scholar] [CrossRef]
  115. Zegelbone, P.M.; Ringel, R.E.; Coulson, J.D.; Nies, M.K.; Stabler, M.E.; Brown, J.R.; Everett, A.D. Heart Failure Biomarker Levels Correlate with Invasive Haemodynamics in Pulmonary Valve Replacement. Cardiol. Young 2020, 30, 50–54. [Google Scholar] [CrossRef]
  116. Parsons, M.; Greenberg, J.; Parikh, C.; Brown, J.; Parker, D.; Zhu, J.; Vricella, L.; Everett, A.D. Post-Operative Acute Kidney Injury Is Associated with a Biomarker of Acute Brain Injury after Paediatric Cardiac Surgery. Cardiol. Young 2020, 30, 505–510. [Google Scholar] [CrossRef] [PubMed]
  117. Greenberg, J.H.; Parsons, M.; Zappitelli, M.; Jia, Y.; Thiessen-Philbrook, H.R.; Devarajan, P.; Everett, A.D.; Parikh, C.R. Cardiac Biomarkers for Risk Stratification of Acute Kidney Injury After Pediatric Cardiac Surgery. Ann. Thorac. Surg. 2021, 111, 191–198. [Google Scholar] [CrossRef]
Figure 1. Gal-3 involvement in the pathogenesis of cardiac fibrosis (created with BioRender.com). Legend: Gal-3—galectin-3; TGF-β—transforming growth factor-β.
Figure 1. Gal-3 involvement in the pathogenesis of cardiac fibrosis (created with BioRender.com). Legend: Gal-3—galectin-3; TGF-β—transforming growth factor-β.
Ijms 24 10511 g001
Figure 2. Gal-3 and its associations with various cardiovascular disorders. Legend: AHF—acute heart failure; CHF—chronic heart failure; CHD—congenital heart disease.
Figure 2. Gal-3 and its associations with various cardiovascular disorders. Legend: AHF—acute heart failure; CHF—chronic heart failure; CHD—congenital heart disease.
Ijms 24 10511 g002
Table 1. Characteristics of clinical studies that assessed the role of Gal-3 in predicting myocardial disfunction and related complications associated with CHD.
Table 1. Characteristics of clinical studies that assessed the role of Gal-3 in predicting myocardial disfunction and related complications associated with CHD.
Reference (Author, Year)Type of StudyPopulation and Study Group AssignmentMain Outcome
Fenster et al., 2016 [85]Case-control study25 adult patients
15 patients with PAH in study group
10 patients in control group
Gal-3 levels were significantly higher in the study group when compared to the controls
Significant association between Gal-3 and indices of RV systolic and diastolic function
Kowalik et al., 2020 [87]Cross-sectional study124 adult patients
63 patients with congenitally corrected TGA in study group I
41 patients with Eisenmenger syndrome in study group II
20 healthy controls
Increase in gal-3 levels within the study groups
Gal-3 levels correlated with age, NYHA class, NT-proBNP and parameters of RV function only in patients with congenitally corrected TGA
Mohammed et al., 2014 [88]Case-control study90 pediatric patients
60 patients with left to right shunt CHD:
  • 30 patients with manifestations of HF
  • 30 patients without HF
30 healthy controls
Gal-3 levels were significantly higher in children with HF when compared with the ones of children without HF and the control group
Significant association between Gal-3 levels and severity of HF, assessed with the help of Ross classification subclasses
Van den Bosch et al., 2022 [89]Cross-sectional study137 Fallot patients (adolescents and adults)Gal-3 does not corelates with parameters of cardiac function and long-term outcome
Kotby et al., 2017 [90]Cross-sectional study90 pediatric patients
45 patients with CHF (15 patients with CHD):
  • 22 patients with HFNEF (EF > 50%)
  • 23 patients with HFREF (EF ≤ 50%)
45 healthy controls
Significant increase in Gal-3 levels within the study group
No significant difference in serum Gal-3 levels between patients with HFNEF and those with HFREF
Elhewala et al., 2020 [91]Case-control study60 pediatric patients
40 patients with CHD:
  • 20 patients with HF
  • 20 patients without HF
20 healthy controls
Significant increase in Gal-3 levels within the study group
Significantly higher Gal-3 levels were found in the study group in patients with HF as opposed to those without HF
Saleh et al., 2020 [92]Case-control study115 pediatric patients
75 patients with CHD
  • 30 children with HFNEF
  • 45 children with HFREF
40 healthy controls
Gal-3 might present an important predictive value for the early diagnosis of HF at a cut-off level of 10.4 ng/dl, with excellent sensitivity and specificity
Cura et al., 2022 [93]Cross-sectional study44 pediatric patients
22 infants with isolated VSD who had received HF treatment
22 healthy control infants
Significant increase in Gal-3 levels within the study group
Gal-3 might be able to distinguish between individuals with the investigated pathology and controls at a cut-off level of 3.62 ng/mL, with good sensitivity and specificity
Frogoudaki et al., 2020 [94]Cross-sectional study58 adult patients with CHD:
  • Group A—16 patients with no SVT episode
  • Group B—27 patients with 1–2 episode of SVT or SV extrasystoles
  • Group C—15 patients with multiple SVT episode or atrial fibrillation
  • Group A’—15 patients with no VT episode
  • Group B’—32 patients with ventricular extrasystoles or couplets
  • Group C’—11 patients with triplets or non-sustained VT or more than 1000 extrasystoles
Gal-3 differed significantly between patient subgroups divided according to the presence and severity of SVT/VT
Gal-3 did not correlate with major adverse cardiovascular events
Geenen et al., 2019 [95]Prospective cohort study86 adult patients with TGA and sRV:
  • 65 patients with M-TGA
  • 21 patients with ccTGA
Gal-3 level is a better predictor for death arrythmia than NT-proBNP and echocardiographic strain parameters
Xiao et al., 2020 [96]Observational study30 adult patients:
  • 10 patients with CHD and normal sinus rhythm
  • 10 patients with RHD and normal sinus rhythm
  • 10 patients with RHD and AF
Expression levels of Gal-3 were increased in the human right atrial appendage tissue within the AF study group when compared to the other two study groups
Legend: AF—atrial fibrillation; ccTGA—congenitally corrected transposition of the great arteries; CHD—congenital heart disease; CHF—chronic heart failure; Gal-3—galectin 3; HF—heart failure; HFNEF—heart failure with normal ejection fraction; HFREF—heart failure with reduce ejection fraction; M-TGA—Mustard/Senning procedure applied for correction of transposition of the great arteries; NT-proBNP—N-terminal pro-brain natriuretic peptide; NYHA—New York Heart Association; PAH—pulmonary arterial hypertension; RCT—randomized control trials; RHD—rheumatic heart disease; RV—right ventricle; sRV—systemic right ventricle; SV—supraventricular; SVT—supraventricular tachycardia; TGA—transposition of the great arteries; VSD—ventricular septal defect; VT—ventricular tachycardia.
Table 2. Characteristics of clinical studies that assessed the role of Gal-3 in predicting myocardial disfunction and related complications in patients with CHD who underwent corrective surgery.
Table 2. Characteristics of clinical studies that assessed the role of Gal-3 in predicting myocardial disfunction and related complications in patients with CHD who underwent corrective surgery.
Reference (Author, Year)Type of StudyPopulation and Study Group
Assignment
Main Outcome
Parker et al., 2020 [98]Longitudinal cohort study145 pediatric patients with CHD and corrective surgery involving cardiopulmonary by-passAn integrated biomarker panel, consisting of GFAP, NT-proBNP, ST2 and Gal-3 significantly correlated with an increased risk of readmission and mortality within the study cohort
Brown et al., 2019 [99]Longitudinal cohort study162 pediatric patients with CHD and corrective surgeryAn integrated biomarker panel, consisting of GFAP, NT-proBNP, ST2 and Gal-3 significantly correlated with an increased risk of readmission and mortality 30 days after discharge within the study cohort
Parker et al., 2019 [104,105,106]Longitudinal cohort study162 pediatric patients with CHD and corrective surgeryAn integrated biomarker panel, consisting of GFAP, NT-proBNP, ST2 and Gal-3 significantly correlated with an increased risk of readmission and mortality 30 days after discharge within the study cohort
Opotowsky et al., 2016 [101]Longitudinal cohort study70 adult patients who underwent Fontan procedureHigh Gal-3 levels correlated with on elective cardiovascular hospitalization ad death
DiLorenzo et al., 2022 [102]Longitudinal cohort study60 pediatric patients who underwent corrective surgery for ToFGal-3 did not correlate with parameters of ventricular remodeling, depicted with the help of CMR
Frank et al., 2019 [110]Longitudinal cohort study27 pediatric patients who underwent aortic coarctation repairPost-operatory Gal-3 levels did not vary significantly from the pre-operatory ones
van den Bosch et al., 2021 [111]Longitudinal cohort study133 pediatric patients who underwent the Fontan procedureGal-3 levels did not correlate with major adverse cardiovascular events
Geenen et al., 2020 [112]Longitudinal cohort observational study50 adult patients who underwent percutaneous ASD closureGal-3 returned to baseline levels three months after surgery
Dudnyk et al., 2019 [113]Case-control study224 pediatric patients:
  • 184 patients with CHD and corrective surgery
  • 40 healthy controls
Gal-3 values were significantly higher in the CHD cohort
Karali et al., 2021 [114]Observational cohort study35 adult patients with repaired ToFNo correlation found between Gal-3 and right ventricular myocardial fibrosis, quantified with the help of CMR
Gal-3 levels correlated with moderate/severe pulmonary regurgitation
Zegelbone et al., 2020 [115]Observational cohort study16 adolescent and adult patients who underwent transcatheter pulmonary valve replacementGal-3 did not correlate with any atrial and ventricular function parameters, assessed through CMR, unlike NT-proBNP and ST2
Parsons et al., 2020 [116]Observational cohort study162 pediatric patients with CHD undergoing cardiopulmonary by-passGal-3 levels did not differ significantly between children with post-operatory AKI and those without renal complications
Greenberg et al., 2021 [117]Observational cohort study395 pediatric patients with CHD undergoing cardiopulmonary by-pass:
  • 194 children < 2 years of age
  • 201 children ≥ 2 years of age
Gal-3 levels were independently associated with post-operative AKI but only in children ≥2 years of age
Legend: ASD—atrial septal defect; AKI—acute kidney injury; CHD—congenital heart disease; CMR—cardiac magnetic resonance; Gal-3—galectin 3; GFAP—glial fibrillary acidic protein; NT-proBNP—N-terminal pro-brain natriuretic peptide.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Făgărășan, A.; Săsăran, M.; Gozar, L.; Crauciuc, A.; Bănescu , C. The Role of Galectin-3 in Predicting Congenital Heart Disease Outcome: A Review of the Literature. Int. J. Mol. Sci. 2023, 24, 10511. https://doi.org/10.3390/ijms241310511

AMA Style

Făgărășan A, Săsăran M, Gozar L, Crauciuc A, Bănescu  C. The Role of Galectin-3 in Predicting Congenital Heart Disease Outcome: A Review of the Literature. International Journal of Molecular Sciences. 2023; 24(13):10511. https://doi.org/10.3390/ijms241310511

Chicago/Turabian Style

Făgărășan, Amalia, Maria Săsăran, Liliana Gozar, Andrei Crauciuc, and Claudia Bănescu . 2023. "The Role of Galectin-3 in Predicting Congenital Heart Disease Outcome: A Review of the Literature" International Journal of Molecular Sciences 24, no. 13: 10511. https://doi.org/10.3390/ijms241310511

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