Acessibilidade / Reportar erro

Galectin-3 and fibrosis intensity in Chronic Chagas Cardiomyopathy: a systematic review

ABSTRACT

Chronic Chagas Cardiomyopathy (CCC) is the most prevalent type of myocarditis and the main clinical form of the Chagas disease, which has peculiarities such as focal inflammation, structural derangement, hypertrophy, dilation, and intense reparative fibrosis. Many cellular compounds contribute to CCC development. Galectin-3 is a partaker in inflammation and contributes to myocardial fibrosis formation. Some studies showed the connection between Galectin-3 and fibrosis in Chagas disease but are still inconclusive on the guidance for the early implementation of pharmacological therapy. This systematic review evaluated Galectin-3 as a biomarker for fibrosis intensity in CCC. Two independent reviewers have searched five databases (PubMed, EMBASE, Cochrane Library, Scopus, and Lilacs), using the following search terms: galectin-3, biomarkers, fibrosis, Chagas cardiomyopathy, and Chagas disease. Overall, seven studies met the inclusion criteria and made up this review. There were four trials conducted through animal model experiments and three trials with humans. Experimental data in mice indicate an association between Galectin-3 expression and fibrosis in CCC (75% of studies). Data from human studies showed no direct connection between myocardial fibrosis and Galectin-3 expression (80% of studies). Thus, human findings do not provide significant evidence indicating that Galectin-3 is related to fibrosis formation in Chagas disease. Based on the analyzed studies, it is suggested that Galectin-3 might not be a good fibrosis marker in CCC.

Galectin-3; Chronic Chagas Cardiomyopathy; Fibrosis

INTRODUCTION

Chagas disease is caused by the protozoa Trypanosoma cruzi, which leads to inflammatory cardiomyopathy and myocardial fibrosis11. Pan American Health Organization. Chagas disease. [cited 2022 May 13]. Available from: https://www.paho.org/en/topics/chagas-disease
https://www.paho.org/en/topics/chagas-di...
and affects millions of people worldwide. The infection is endemic in Mexico, Central, and South America and represents a severe public health problem22. Garcia MN, Aguilar D, Gorchakov R, Rossmann SN, Montgomery SP, Rivera H, et al. Evidence of autochthonous Chagas disease in southeastern Texas. Am J Trop Med Hyg. 2015;92:325-30.. The clinical course of Chagas disease comprises acute and chronic phases33. Bonney KM, Luthringer DJ, Kim SA, Garg NJ, Engman DM. Pathology and pathogenesis of Chagas heart disease. Annu Rev Pathol. 2019;14:421-47.. Chronic Chagas Cardiomyopathy (CCC) is the most relevant clinical manifestation of Chagas disease due to its frequency, severity, morbidity, and mortality44. Tanowitz HB, Machado FS, Spray DC, Friedman JM, Weiss OS, Lora JN, et al. Developments in the management of Chagas cardiomyopathy. Expert Rev Cardiovasc Ther. 2015;13:1393-409..

Most Chagas disease patients present diffuse myocarditis with fibrosis and hypertrophy55. Nonaka CK, Macêdo CT, Cavalcante BR, Alcântara AC, Silva DN, Bezerra MD, et al. Circulating miRNAs as potential biomarkers associated with cardiac remodeling and fibrosis in Chagas disease cardiomyopathy. Int J Mol Sci. 2019;20:4064.. In CCC, fibrosis occurs due to the extensive and constant infiltration of inflammatory cells into the myocardium, mediated by cytokines and growth factors that regulate cell migration, proliferation, differentiation, production, and degradation of different extracellular matrix components, which are particularities that distinguish CCC from other diseases66. Marin-Neto JA, Cunha-Neto E, Maciel BC, Simões MV. Pathogenesis of chronic Chagas heart disease. Circulation. 2007;115:1109-23.

7. Cunha-Neto E, Chevillard C. Chagas disease cardiomyopathy: immunopathology and genetics. Mediators Inflamm. 2014;2014:683230.
-88. Chaves AT, Menezes CA, Costa HS, Nunes MC, Rocha MO. Myocardial fibrosis in Chagas disease and molecules related to fibrosis. Parasite Immunol. 2019;41:e12663..

Galectin-3 is a β-galactoside-binding lectin99. Liu FT, Rabinovich GA. Galectins: regulators of acute and chronic inflammation. Ann N Y Acad Sci. 2010;1183:158-82. found in several physiological and pathological cellular processes, including proliferation, migration, and cardiac fibrosis1010. de Boer RA, Voors AA, Muntendam P, van Gilst WH, van Veldhuisen DJ. Galectin-3: a novel mediator of heart failure development and progression. Eur J Heart Fail. 2009;11:811-7.,1111. Dumic J, Dabelic S, Flögel M. Galectin-3: an open-ended story. Biochim Biophys Acta. 2006;1760:616-35.. In the context of CCC, Galectin-3 participates in the migration of cells recruited into the heart and contributes to the fibrogenesis process; thus, Galectin-3 could also be a factor in maintaining inflammation and contributing to cardiac fibrosis formation88. Chaves AT, Menezes CA, Costa HS, Nunes MC, Rocha MO. Myocardial fibrosis in Chagas disease and molecules related to fibrosis. Parasite Immunol. 2019;41:e12663.. Galectin-3 may be a potential therapeutic target and the control of its expression could benefit patients with CCC.

The biomarkers predicting the progression of CCC may guide the early implementation of pharmacological therapy in Chagas disease1212. Bocchi EA, Bestetti RB, Scanavacca MI, Cunha Neto E, Issa VS. Chronic Chagas heart disease management: from etiology to cardiomyopathy treatment. J Am Coll Cardiol. 2017;70:1510-24.. Furthermore, it is challenging to identify predicting factors associated with disease progression, morbidity, and mortality, to assist with the decision-making in the follow-up and treatment of this complex disease. The present study examines previous publications to establish whether Galectin-3 is an adequate biomarker for fibrosis intensity evaluation in CCC patients.

MATERIALS AND METHODS

The Preferred Reporting Items for Systematic Reviews and Meta-Analysis (PRISMA)1313. Moher D, Shamseer L, Clarke M, Ghersi D, Liberati A, Petticrew M, et al. Preferred reporting items for systematic review and meta-analysis protocols (PRISMA-P) 2015 statement. Syst Rev. 2015;4:1. was applied in the writing of this systematic review and registered in the PROSPERO database (CRD42019119309). This review aimed to answer the question “What is the real role of Galectin-3 as a biomarker in fibrosis intensity in CCC?”

Search strategy

The literature search has been done in five electronic databases (PubMed, Embase, Cochrane Library, Scopus, and Lilacs) to recognize clinical trials that examined the role of Galectin-3 as a biomarker in fibrosis intensity in CCC, from database inception to December 2020. Furthermore, we searched all reference lists of qualified studies and related reviews to avoid any irrelevant publications. No search limitations concerning languages, time of publication, and article types were applied.

The search strategy included Medical Subject Headings (MeSH), Descriptors in Health Sciences (DeCS), and Emtree, seeking terms such as “Galectin-3”, “fibrosis”, “Chagas disease”, “Chagas cardiomyopathy”, “biomarkers”, using the operators of quotes, parentheses, “AND”, “OR”, “exp” and “mp”. The study search strategy is shown in Figure 1.

Figure 1
PRISMA Flowchart describing the search strategy and selection of studies on the Galectin-3 assessment as a biomarker in fibrosis intensity in Chronic Chagas Cardiomyopathy.

Eligibility and exclusion criteria

For eligibility and inclusion of the scientific studies, two reviewers read the titles and abstracts. In the second stage, the complete text of each study was evaluated by a third reviewer, and any disagreements were resolved by trying to answer the question that originated the study.

All original published papers regarding the topic of interest were eligible for inclusion, such as (1) cross-sectional, case-control, cohorts, clinical trials, or diagnostics studies; (2) studies with male and female humans or animals; (3) studies that evaluated Galectin-3 as a biomarker in CCC.

The exclusion criteria were: duplicates, narrative or systematic revisions and meta-analysis, case studies, book chapters, editorials, letters to the editor, studies on pregnant or breastfeeding women, children, and adolescents, studies involving themes not suitable for the review, and studies evaluating Gal-3 in relation to fibrosis and cardiomyopathy, but not Chagas disease.

Data extraction

Two reviewers extracted all trial data independently; the third reviewer arbitrated the disagreements, when necessary. We withdrew data into separate spreadsheets. A specif dataset was created once the reviewers resolved all inconsistencies. For each eligible study, we extracted the author’s first name, study design, year of publication, publishing means, impact factor, country, the population of studies, sample size, gender, age, tests used to detect fibrosis and Galectin-3, the statistical analysis used, and the main results of Galectin-3 and fibrosis. Seven articles met all inclusion criteria (Figure 1).

RESULTS

Primarily, the database search generated 435 studies, of which, 280 published articles appeared to be relevant. Ten manuscripts, in duplicates, were removed. After that, 273 works were excluded for different reasons leaving seven eligible manuscript articles1414. Cruz GS, Angelo AL, Larocca TF, Macedo CT, Noya-Rabelo M, Correia LC, et al. Assessment of Galectin-3 polymorphism in subjects with chronic Chagas disease. Arq Bras Cardiol. 2015;105:472-8.

15. Noya-Rabelo MM, Larocca TF, Macêdo CT, Torreão JA, Souza BS, Vasconcelos JF, et al. Evaluation of Galectin-3 as a novel biomarker for Chagas cardiomyopathy. Cardiology. 2017;136:33-9.

16. Echeverría LE, Rojas LZ, Calvo LS, Roa ZM, Rueda-Ochoa OL, Morillo CA, et al. Profiles of cardiovascular biomarkers according to severity stages of Chagas cardiomyopathy. Int J Cardiol. 2017;227:577-82.

17. Ferrer MF, Pascuale CA, Gomez RM, Leguizamón MS. DTU I isolates of Trypanosoma cruzi induce upregulation of Galectin-3 in murine myocarditis and fibrosis. Parasitology. 2014;141:849-58.

18. Souza BS, Silva DN, Carvalho RH, Sampaio GL, Paredes BD, França LA, et al. Association of cardiac Galectin-3 expression, myocarditis, and fibrosis in chronic Chagas disease cardiomyopathy. Am J Pathol. 2017;187:1134-46.

19. Souza BS, Silva KN, Silva DN, Rocha VP, Paredes BD, Azevedo CM, et al. Galectin-3 knockdown impairs survival, migration, and immunomodulatory actions of mesenchymal stromal cells in a mouse model of Chagas disease cardiomyopathy. Stem Cells Int. 2017;2017:3282656.
-2020. Pineda MA, Cuervo H, Fresno M, Soto M, Bonay P. Lack of Galectin-3 prevents cardiac fibrosis and effective immune responses in a murine model of Trypanosoma cruzi infection. J Infect Dis. 2015;212:1160-71.. There were four trials conducted through animal model experiments and three trials with humans, as shown in Table 1. The selected articles were published in important academic journals that presented a range of impact factors from 1.35 to 4.73 and were conducted from 2014 to 2017 in four different countries: Brazil (57.1%), Colombia (14.3%), Argentina (14.3%), and Spain (14.3%). The population of the studies included in this systematic review comprised 216 humans, 103 males (47.7%) and 113 females (52.3%), with a mean age of 56 years old (Table 1). The total number of animals used in the experimental studies was 44. Of which, 29 (65.9%) were females and 15 (34.1%) were males, with a mean age of 6–8 weeks and 21 days, respectively.

Table 1
General characteristics of the studies included in the systematic review.

Furthermore, in Table 2, we present the comparative events extracted from the seven reports that correlated Galectin-3 to the Chronic Chagas Cardiomyopathy in human or animal studies.

Table 2
NI = not informed.

- Characteristics of studies that evaluated Galectin-3 and myocardial fibrosis in Chagas chronic cardiopathy humans and mice.


Cruz et al.1414. Cruz GS, Angelo AL, Larocca TF, Macedo CT, Noya-Rabelo M, Correia LC, et al. Assessment of Galectin-3 polymorphism in subjects with chronic Chagas disease. Arq Bras Cardiol. 2015;105:472-8. evaluated the myocardial fibrosis of patients with and without Galectin-3 polymorphism and compared myocardial fibrosis in individuals with AA, AC, and CC genotypes for the genetic variants at two single nucleotide polymorphic (SNP) sites of the Galectin-3 gene (rs4644 and rs4652). The results demonstrated that there was no statistical difference between the AA, AC and CC genotypes (p = 0.508) or between SNP rs4644 and SNP rs4652 (p = 0.903).

Noya-Rabelo et al.1515. Noya-Rabelo MM, Larocca TF, Macêdo CT, Torreão JA, Souza BS, Vasconcelos JF, et al. Evaluation of Galectin-3 as a novel biomarker for Chagas cardiomyopathy. Cardiology. 2017;136:33-9. studied the percentage of myocardial fibrosis by magnetic resonance imaging and the plasma concentration of Galectin-3 by enzyme-linked immunosorbent assay (ELISA) in different groups of patients (indeterminate cardiac form and cardiac form with or without ventricular dysfunction)1515. Noya-Rabelo MM, Larocca TF, Macêdo CT, Torreão JA, Souza BS, Vasconcelos JF, et al. Evaluation of Galectin-3 as a novel biomarker for Chagas cardiomyopathy. Cardiology. 2017;136:33-9.. The results showed that the proportion of myocardial fibrosis was 9.4% (IQI: 2.4–18.4), with a progressive increase as the disease worsened. Myocardial fibrosis was detected in 6 of 17 individuals with the indeterminate form (median, 4.1%, IQI: 2.1–10.7), in 7 of 16 individuals with the cardiac form without ventricular dysfunction (median 2.3%, IQI: 1.0–5.0), and in 22 of 28 individuals with the cardiac form with ventricular dysfunction (median 15.2%, IQI: 7.8–25.0, p=0.001). The median Galectin-3 concentration was 12.1 ng/mL (IQI: 9.4–14.4). There were no statistically significant differences in Galectin-3 concentration between the groups of patients, with a median concentration of 12.1 ng/mL (IQI: 8.8–18.3). The median Galectin-3 concentration in the indeterminate group was 12.1 ng/mL (IQI: 10.1–13.9). In the cardiac group with or without ventricular dysfunction, the median Galectin-3 concentration was 12.0 ng/mL (IQI: 11.0–14.8). No correlation was found between myocardial fibrosis and Galectin-3 concentration (r = 0.098; p = 0.47).

Echeverria et al.1616. Echeverría LE, Rojas LZ, Calvo LS, Roa ZM, Rueda-Ochoa OL, Morillo CA, et al. Profiles of cardiovascular biomarkers according to severity stages of Chagas cardiomyopathy. Int J Cardiol. 2017;227:577-82. determined the concentration of Galectin-3 by fluorescence enzyme-linked immunosorbent assay (ELFA) and correlated the data with left ventricular ejection fraction (LVEF). The study applied three different models: (1) gross probability ratio; (2) crude probability ratio adjusted for body mass index, age, gender, and estimated glomerular filtration rate; and (3) further adjustment for angiotensin-converting enzyme inhibitor or angiotensin receptor blocker, beta-blocker, aldosterone antagonist, and diuretic use. In the first model, there was a positive correlation between LVEF and Galectin-3 concentration (β = 0.421; IQI: -0.618 to -0.224; p<0.001). In the second and third models, no correlation was found between LVEF and Galectin-3 concentration (model 2: β = 0.154, IQI: -0.378 to -0.699, p = 0.175; model 3: β = 0.123, IQI: 0.320–0.073, p = 0.217).

Regarding the four studies involving animal models 17–20, three of them used mice aged 6 to 8 weeks, while in one of the studies, the authors included 15 males with a mean age of 21 days.

Ferrer et al.1717. Ferrer MF, Pascuale CA, Gomez RM, Leguizamón MS. DTU I isolates of Trypanosoma cruzi induce upregulation of Galectin-3 in murine myocarditis and fibrosis. Parasitology. 2014;141:849-58. measured the expression of Galectin-3 associated with cardiac extracellular matrix (ECM) by analyzing the levels of procollagen I mRNA. Cardiomyopathy was induced by discrete typing unit strains I (DTU Tc I), named Ac and Hc, of T. cruzi. In samples from mice infected with Hc and Ac, the expression of procollagen I mRNA was higher in the animals inoculated with Ac (p<0.05). As for the cardiac histology of infected mice, an increase in the collagen matrix was observed in samples from animals infected with both strains. MEC remodeling was less intense in mice inoculated with Hc than Ac. At the same time, Galectin-3 levels were significantly higher in samples from mice infected with Ac than in the simulated controls (p≤0.001). The Galectin-3 antigen was mainly detected in the interstitial cells and found in myocardial samples.

In 2015, Pineda et al.2020. Pineda MA, Cuervo H, Fresno M, Soto M, Bonay P. Lack of Galectin-3 prevents cardiac fibrosis and effective immune responses in a murine model of Trypanosoma cruzi infection. J Infect Dis. 2015;212:1160-71. evaluated the expression of collagen I, III, IV, and laminin in myocardial samples of C57BL/6 mice and those with the knockout of Galectin-3, 28, and 60 days after T. cruzi infection2020. Pineda MA, Cuervo H, Fresno M, Soto M, Bonay P. Lack of Galectin-3 prevents cardiac fibrosis and effective immune responses in a murine model of Trypanosoma cruzi infection. J Infect Dis. 2015;212:1160-71.. The reduced expression of collagen I, III, IV, and laminin in the heart of Galectin-3 knockout animals, as demonstrated, compared to the control group suggests that galectin-3 is strongly involved in Chagas disease, not only in the immune response against T. cruzi but also in mediating myocardial damage.

In 2017, Souza et al.1818. Souza BS, Silva DN, Carvalho RH, Sampaio GL, Paredes BD, França LA, et al. Association of cardiac Galectin-3 expression, myocarditis, and fibrosis in chronic Chagas disease cardiomyopathy. Am J Pathol. 2017;187:1134-46. correlated the Galectin-3 expression area’s percentage with the fibrosis area’s percentage in cardiac sections of mice at different times of infection with trypomastigotes of the Colombian T. cruzi strain (30 dpi, 90 dpi, and 180 dpi). It was observed an increased myocardial Galectin-3 expression in infected animals compared to non-infected controls. When compared to the uninfected mice, the Galectin-3 cardiac expression peaked at one-month post-infection, remaining high during the chronic phase of the infection, but this expression decreased with time. Regarding infection points, Galectin-3 expression in 30 dpi mice was higher than that of 180 dpi mice (p<0.001). The percentage of fibrosis increased with infection time and fibrosis was more severe in 180 dpi mice than in 30 dpi (p<0.001) and 90 dpi (p<0.001) mice.

A second study1919. Souza BS, Silva KN, Silva DN, Rocha VP, Paredes BD, Azevedo CM, et al. Galectin-3 knockdown impairs survival, migration, and immunomodulatory actions of mesenchymal stromal cells in a mouse model of Chagas disease cardiomyopathy. Stem Cells Int. 2017;2017:3282656. evaluated the fibrosis area percentage and the gene expression of type I collagen (Col1a1) in the mice’s T. cruzi-infected heart. T. cruzi-infected mice received control mesenchymal stromal cells (MSC-WT) or mesenchymal Galectin-3 knockdown stromal cells (MSC-Gal3KD). The analysis of cardiac sections in T. cruzi-infected mice showed extensive areas of fibrosis while the fibrosis content in the heart was not significantly different between the groups. the Collagen type I (Col1a1) gene expression was reduced in infected mice that received wild-type MSC cells but not in those who received MSC knockdown Galectin-3. According to studies using animal models, three studies (75%) showed a positive correlation between Galectin-3 and myocardial fibrosis and one study (25%) presented no correlation between these variables.

DISCUSSION

CCC is the most relevant clinical form of Chagas disease due to its high morbidity, mortality, and social/medical impact2121. Rocha MO, Ribeiro AL, Teixeira MM. Clinical management of chronic Chagas cardiomyopathy. Front Biosci. 2003;8:e44-54.

22. Andrade JP, Marin Neto JA, Paola AA, Vilas-Boas F, Oliveira GM, Bacal F, et al. I Latin American Guidelines for the diagnosis and treatment of Chagas’ heart disease: executive summary. Arq Bras Cardiol. 2011;96:434-42.

23. Ribeiro AL, Nunes MP, Teixeira MM, Rocha MO. Diagnosis and management of Chagas disease and cardiomyopathy. Nat Rev Cardiol. 2012;9:576-89.
-2424. Nunes MC, Dones W, Morillo CA, Encina JJ, Ribeiro AL. Chagas disease: an overview of clinical and epidemiological aspects. J Am Coll Cardiol. 2013;62:767-76.. CCC has a characteristically slow and progressive course, although it can progress rapidly. Its clinical manifestations range from subclinical to severe presentations characterized by heart failure, arrhythmias of variable type and severity, conduction blocks of electrical stimulation, thromboembolism, ischemic stroke, and sudden death that may eventually constitute its first manifestation2121. Rocha MO, Ribeiro AL, Teixeira MM. Clinical management of chronic Chagas cardiomyopathy. Front Biosci. 2003;8:e44-54.,2323. Ribeiro AL, Nunes MP, Teixeira MM, Rocha MO. Diagnosis and management of Chagas disease and cardiomyopathy. Nat Rev Cardiol. 2012;9:576-89.

24. Nunes MC, Dones W, Morillo CA, Encina JJ, Ribeiro AL. Chagas disease: an overview of clinical and epidemiological aspects. J Am Coll Cardiol. 2013;62:767-76.

25. Rassi A Jr, Rassi SG, Rassi A. Sudden death in Chagas’ disease. Arq Bras Cardiol. 2001;76:75-96.
-2626. Chagas C, Villela E. Forma cardíaca da Trypanosomiase Americana. Mem Inst Oswaldo Cruz. 1922;14:5-61..

CCC is the most common and fibrous type of myocarditis worldwide2727. Feldman AM, McNamara D. Myocarditis. N Engl J Med. 2000;343:1388-98.,2828. Henderson NC, Sethi T. The regulation of inflammation by galectin-3. Immunol Rev. 2009;230:160-71.. CCC presents peculiarities such as variable intensities of focal inflammation, structural derangement, hypertrophy, dilation, and intense reactive and reparative fibrosis88. Chaves AT, Menezes CA, Costa HS, Nunes MC, Rocha MO. Myocardial fibrosis in Chagas disease and molecules related to fibrosis. Parasite Immunol. 2019;41:e12663.. A recent study concerning this topic showed a correlation between Gal-3 and cardiac dysfunction, and severe human Chagas cardiomyopathy2929. Fernandes F, Moreira CH, Oliveira LC, Souza-Basqueira M, Ianni BM, Lorenzo CD, et al. Galectin-3 associated with severe forms and long-term mortality in patients with Chagas disease. Arq Bras Cardiol. 2021;116:248-56..

Inflammation is a commonly beneficial protective response to tissue injury, promoting healing and repair. Acute inflammation is the natural response of vascularized tissues to injury, irritation, and infection. In contrast, chronic inflammation is a detrimental process due to the inability to resolve acute inflammation or persistent inflammatory stimuli1010. de Boer RA, Voors AA, Muntendam P, van Gilst WH, van Veldhuisen DJ. Galectin-3: a novel mediator of heart failure development and progression. Eur J Heart Fail. 2009;11:811-7.. Chronic inflammation with fibrosis formation and functional impairment of organs is a significant cause of morbidity and mortality of many chronic diseases3030. Lopez-Andrès N, Rossignol P, Iraqi W, Fay R, Nuée J, Ghio S, et al. Association of galectin-3 and fibrosis markers with long-term cardiovascular outcomes in patients with heart failure, left ventricular dysfunction, and dyssynchrony: insights from the CARE-HF (Cardiac Resynchronization in Heart Failure) trial. Eur J Heart Fail. 2012;14:74-81..

Tissue fibrosis is a progressive disorder characterized by the abundant accumulation of extracellular matrix, leading to organ lesions and dysfunction. Several cytokines, chemokines, growth factors, and angiogenic regulate cells’ activation to produce extracellular matrix in fibrous processes. In most cases, it is relevant to identify the mechanisms of fibrosis formation by identifying irreversible lesions and, consequently, delineate possible antifibrotic therapeutic strategies1010. de Boer RA, Voors AA, Muntendam P, van Gilst WH, van Veldhuisen DJ. Galectin-3: a novel mediator of heart failure development and progression. Eur J Heart Fail. 2009;11:811-7.,3131. MacKinnon AC, Farnworth SL, Hodkinson PS, Henderson NC, Atkinson KM, Leffler H, et al. Regulation of alternative macrophage activation by galectin-3. J Immunol. 2008;180:2650-8..

Galectin-3 is a multifunctional protein of the lectin family that binds to the cell surface and glycans of the extracellular matrix. It plays a crucial role in cell proliferation, adhesion, differentiation, apoptosis, and angiogenesis1010. de Boer RA, Voors AA, Muntendam P, van Gilst WH, van Veldhuisen DJ. Galectin-3: a novel mediator of heart failure development and progression. Eur J Heart Fail. 2009;11:811-7.. Galectin-3 exerts a pro-inflammatory effect by activating macrophages and mediating extracellular matrix-producing cells3232. Li LC, Li J, Gao J. Functions of galectin-3 and its role in fibrotic diseases. J Pharmacol Exp Ther. 2014;351:336-43.,3333. Sharma UC, Pokharel S, van Brakel TJ, van Berlo JH, Cleutjens JP, Schroen B, et al. Galectin-3 marks activated macrophages in failure-prone hypertrophied hearts and contributes to cardiac dysfunction. Circulation. 2004;110:3121-8.. Furthermore, Galectin-3 regulates the inflammatory response through cell activation and migration by regulating apoptosis of immune cells. In acute inflammation, Galectin-3 seems to play a pro-inflammatory and protective role. After the transition to a chronic inflammatory state, it plays a different fibrogenic role in repairing the lesions; ultimately, this results in fibrosis and architectural disruption of organs. A more detailed study of the role of Galectin-3 in cardiac remodeling revealed that it was localized in the fibrosis sites themselves, especially in fibroblasts and macrophages, but not in cardiomyocytes3333. Sharma UC, Pokharel S, van Brakel TJ, van Berlo JH, Cleutjens JP, Schroen B, et al. Galectin-3 marks activated macrophages in failure-prone hypertrophied hearts and contributes to cardiac dysfunction. Circulation. 2004;110:3121-8..

Most heart disease biomarkers, such as troponin, C-reactive protein, and BNP are released into circulation due to the pathogenic process, representing the result and not the cause of the damage. Extracellular Galectin-3 has a causal role in the remodeling process, the inhibition of which seems to block or reverse the process. These measurements and analysis may enable the development of disease-modifying therapies by inhibiting remodeling and stopping or delaying the progression of heart failure1010. de Boer RA, Voors AA, Muntendam P, van Gilst WH, van Veldhuisen DJ. Galectin-3: a novel mediator of heart failure development and progression. Eur J Heart Fail. 2009;11:811-7..

It is already known that Galectin-3 is an important factor in the progression of heart failure (HF). HF patients with low levels of Gal-3 present a better outcome than patients with high Gal-3 levels3434. de Boer RA, van der Velde AR, Mueller C, van Veldhuisen DJ, Anker SD, Peacock WF, et al. Galectin-3: a modifiable risk factor in heart failure. Cardiovasc Drugs Ther. 2014;28:237-46.,3535. de Boer RA, van Veldhuisen DJ, Gansevoort RT, Muller Kobold AC, van Gilst WH, Hillege HL, et al. The fibrosis marker galectin-3 and outcome in the general population. J Intern Med. 2012;272:55-64.. Gal-3 can be used to identify patients with HF at low risk for 30-day and 180-day mortality, and HF patients’ hospital returns after an episode of acute HF3636. Meijers WC, de Boer RA, van Veldhuisen DJ, Jaarsma T, Hillege HL, Maisel AS, et al. Biomarkers and low risk in heart failure: data from COACH and TRIUMPH. Eur J Heart Fail. 2015;17:1271-82..

The results of this systematic review show that 75% of animal model studies demonstrated a positive correlation between Galectin-3 and myocardial fibrosis. These findings provide evidence for a pro-inflammatory role of Galectin-3 in Chagas disease, which is indicative of its ability to promote tissue inflammation, cell infiltration, and cardiac damage.

Although experimental data in mice indicate an association between Gal-3 expression and fibrosis, the few studies in humans are controversial. They do not provide robust evidence for this association in Chagas heart disease. There were biases in these studies that may affect the interpretation of the results. Among them are the small sample size of the studies analyzed and the different methods used to measure the concentration of Gal-3 and the percentage of fibrosis in the myocardium. Moreover, in the clinical studies, Galectin-3 was measured in a single moment; thus, it was challenging to provide information about its importance over time. Furthermore, the circulating concentration of Galectin-3 may not accurately reflect its expression in the myocardium. There is experimental evidence that Gal-3 expression is associated with cardiac fibrosis in CCC. However, studies on humans are scarce, using different methods and presenting controversial results, indicating the need for studies on this subject.

Study limitations

These results must be interpreted with caution and several limitations should be borne in mind. There are three major limitations in this study that could influence the conclusions. First, the small number of studies relating Galectin and cardiac fibrosis in human Chagas disease (3 articles) in association with the number of individuals evaluated in the 3 reviewed articles (216 individuals divided in control, indeterminate and cardiac groups). Second, the methodology used to evaluate the relation between Galectin and fibrosis was different in all three refereed articles (MRI, gene polymorphism, FEVE, plasmatic expression). Third, the evaluation of Gal-3 levels, when measured, was done only once and there was no follow-up to assess if any alteration in these biomarkers occurred over time. Based on this consideration, it would be necessary a multicentric study with a higher number of individuals evaluated and a pattern methodology to identify the real role of Galectin-3 in fibrosis production and maintenance in cardiac Chagas disease.

CONCLUSION

Galectin-3 has been pointed out as an indicator of fibrosis in various tissues, but its specific role in detecting myocardial fibrosis is not well established. Through this systematic review, it was not possible to assume that Gal-3 can be useful to characterize fibrosis in CCC patients. Further research is needed to verify the association of Galectin-3 with cardiac connective tissue, remodeling and ascertaining the potential use of this molecule as a biomarker of fibrosis in CCC.

REFERENCES

  • 1
    Pan American Health Organization. Chagas disease. [cited 2022 May 13]. Available from: https://www.paho.org/en/topics/chagas-disease
    » https://www.paho.org/en/topics/chagas-disease
  • 2
    Garcia MN, Aguilar D, Gorchakov R, Rossmann SN, Montgomery SP, Rivera H, et al. Evidence of autochthonous Chagas disease in southeastern Texas. Am J Trop Med Hyg. 2015;92:325-30.
  • 3
    Bonney KM, Luthringer DJ, Kim SA, Garg NJ, Engman DM. Pathology and pathogenesis of Chagas heart disease. Annu Rev Pathol. 2019;14:421-47.
  • 4
    Tanowitz HB, Machado FS, Spray DC, Friedman JM, Weiss OS, Lora JN, et al. Developments in the management of Chagas cardiomyopathy. Expert Rev Cardiovasc Ther. 2015;13:1393-409.
  • 5
    Nonaka CK, Macêdo CT, Cavalcante BR, Alcântara AC, Silva DN, Bezerra MD, et al. Circulating miRNAs as potential biomarkers associated with cardiac remodeling and fibrosis in Chagas disease cardiomyopathy. Int J Mol Sci. 2019;20:4064.
  • 6
    Marin-Neto JA, Cunha-Neto E, Maciel BC, Simões MV. Pathogenesis of chronic Chagas heart disease. Circulation. 2007;115:1109-23.
  • 7
    Cunha-Neto E, Chevillard C. Chagas disease cardiomyopathy: immunopathology and genetics. Mediators Inflamm. 2014;2014:683230.
  • 8
    Chaves AT, Menezes CA, Costa HS, Nunes MC, Rocha MO. Myocardial fibrosis in Chagas disease and molecules related to fibrosis. Parasite Immunol. 2019;41:e12663.
  • 9
    Liu FT, Rabinovich GA. Galectins: regulators of acute and chronic inflammation. Ann N Y Acad Sci. 2010;1183:158-82.
  • 10
    de Boer RA, Voors AA, Muntendam P, van Gilst WH, van Veldhuisen DJ. Galectin-3: a novel mediator of heart failure development and progression. Eur J Heart Fail. 2009;11:811-7.
  • 11
    Dumic J, Dabelic S, Flögel M. Galectin-3: an open-ended story. Biochim Biophys Acta. 2006;1760:616-35.
  • 12
    Bocchi EA, Bestetti RB, Scanavacca MI, Cunha Neto E, Issa VS. Chronic Chagas heart disease management: from etiology to cardiomyopathy treatment. J Am Coll Cardiol. 2017;70:1510-24.
  • 13
    Moher D, Shamseer L, Clarke M, Ghersi D, Liberati A, Petticrew M, et al. Preferred reporting items for systematic review and meta-analysis protocols (PRISMA-P) 2015 statement. Syst Rev. 2015;4:1.
  • 14
    Cruz GS, Angelo AL, Larocca TF, Macedo CT, Noya-Rabelo M, Correia LC, et al. Assessment of Galectin-3 polymorphism in subjects with chronic Chagas disease. Arq Bras Cardiol. 2015;105:472-8.
  • 15
    Noya-Rabelo MM, Larocca TF, Macêdo CT, Torreão JA, Souza BS, Vasconcelos JF, et al. Evaluation of Galectin-3 as a novel biomarker for Chagas cardiomyopathy. Cardiology. 2017;136:33-9.
  • 16
    Echeverría LE, Rojas LZ, Calvo LS, Roa ZM, Rueda-Ochoa OL, Morillo CA, et al. Profiles of cardiovascular biomarkers according to severity stages of Chagas cardiomyopathy. Int J Cardiol. 2017;227:577-82.
  • 17
    Ferrer MF, Pascuale CA, Gomez RM, Leguizamón MS. DTU I isolates of Trypanosoma cruzi induce upregulation of Galectin-3 in murine myocarditis and fibrosis. Parasitology. 2014;141:849-58.
  • 18
    Souza BS, Silva DN, Carvalho RH, Sampaio GL, Paredes BD, França LA, et al. Association of cardiac Galectin-3 expression, myocarditis, and fibrosis in chronic Chagas disease cardiomyopathy. Am J Pathol. 2017;187:1134-46.
  • 19
    Souza BS, Silva KN, Silva DN, Rocha VP, Paredes BD, Azevedo CM, et al. Galectin-3 knockdown impairs survival, migration, and immunomodulatory actions of mesenchymal stromal cells in a mouse model of Chagas disease cardiomyopathy. Stem Cells Int. 2017;2017:3282656.
  • 20
    Pineda MA, Cuervo H, Fresno M, Soto M, Bonay P. Lack of Galectin-3 prevents cardiac fibrosis and effective immune responses in a murine model of Trypanosoma cruzi infection. J Infect Dis. 2015;212:1160-71.
  • 21
    Rocha MO, Ribeiro AL, Teixeira MM. Clinical management of chronic Chagas cardiomyopathy. Front Biosci. 2003;8:e44-54.
  • 22
    Andrade JP, Marin Neto JA, Paola AA, Vilas-Boas F, Oliveira GM, Bacal F, et al. I Latin American Guidelines for the diagnosis and treatment of Chagas’ heart disease: executive summary. Arq Bras Cardiol. 2011;96:434-42.
  • 23
    Ribeiro AL, Nunes MP, Teixeira MM, Rocha MO. Diagnosis and management of Chagas disease and cardiomyopathy. Nat Rev Cardiol. 2012;9:576-89.
  • 24
    Nunes MC, Dones W, Morillo CA, Encina JJ, Ribeiro AL. Chagas disease: an overview of clinical and epidemiological aspects. J Am Coll Cardiol. 2013;62:767-76.
  • 25
    Rassi A Jr, Rassi SG, Rassi A. Sudden death in Chagas’ disease. Arq Bras Cardiol. 2001;76:75-96.
  • 26
    Chagas C, Villela E. Forma cardíaca da Trypanosomiase Americana. Mem Inst Oswaldo Cruz. 1922;14:5-61.
  • 27
    Feldman AM, McNamara D. Myocarditis. N Engl J Med. 2000;343:1388-98.
  • 28
    Henderson NC, Sethi T. The regulation of inflammation by galectin-3. Immunol Rev. 2009;230:160-71.
  • 29
    Fernandes F, Moreira CH, Oliveira LC, Souza-Basqueira M, Ianni BM, Lorenzo CD, et al. Galectin-3 associated with severe forms and long-term mortality in patients with Chagas disease. Arq Bras Cardiol. 2021;116:248-56.
  • 30
    Lopez-Andrès N, Rossignol P, Iraqi W, Fay R, Nuée J, Ghio S, et al. Association of galectin-3 and fibrosis markers with long-term cardiovascular outcomes in patients with heart failure, left ventricular dysfunction, and dyssynchrony: insights from the CARE-HF (Cardiac Resynchronization in Heart Failure) trial. Eur J Heart Fail. 2012;14:74-81.
  • 31
    MacKinnon AC, Farnworth SL, Hodkinson PS, Henderson NC, Atkinson KM, Leffler H, et al. Regulation of alternative macrophage activation by galectin-3. J Immunol. 2008;180:2650-8.
  • 32
    Li LC, Li J, Gao J. Functions of galectin-3 and its role in fibrotic diseases. J Pharmacol Exp Ther. 2014;351:336-43.
  • 33
    Sharma UC, Pokharel S, van Brakel TJ, van Berlo JH, Cleutjens JP, Schroen B, et al. Galectin-3 marks activated macrophages in failure-prone hypertrophied hearts and contributes to cardiac dysfunction. Circulation. 2004;110:3121-8.
  • 34
    de Boer RA, van der Velde AR, Mueller C, van Veldhuisen DJ, Anker SD, Peacock WF, et al. Galectin-3: a modifiable risk factor in heart failure. Cardiovasc Drugs Ther. 2014;28:237-46.
  • 35
    de Boer RA, van Veldhuisen DJ, Gansevoort RT, Muller Kobold AC, van Gilst WH, Hillege HL, et al. The fibrosis marker galectin-3 and outcome in the general population. J Intern Med. 2012;272:55-64.
  • 36
    Meijers WC, de Boer RA, van Veldhuisen DJ, Jaarsma T, Hillege HL, Maisel AS, et al. Biomarkers and low risk in heart failure: data from COACH and TRIUMPH. Eur J Heart Fail. 2015;17:1271-82.
  • FUNDING: The study was supported by a grant from Fundação de Amparo à Pesquisa do Estado de Minas Gerais (FAPEMIG). ATC and MOCR are CNPq fellows.

Publication Dates

  • Publication in this collection
    24 June 2022
  • Date of issue
    2022

History

  • Received
    14 Feb 2022
  • Accepted
    15 May 2022
Instituto de Medicina Tropical de São Paulo Av. Dr. Enéas de Carvalho Aguiar, 470, 05403-000 - São Paulo - SP - Brazil, Tel. +55 11 3061-7005 - São Paulo - SP - Brazil
E-mail: revimtsp@usp.br