SciELO - Scientific Electronic Library Online

 
vol.42 número2Tele-enfermería en pacientes crónicos: revisión sistemáticaOnabotulinumtoxinA en el tratamiento de la odontalgia atípica: descripción de un caso clínico índice de autoresíndice de materiabúsqueda de artículos
Home Pagelista alfabética de revistas  

Servicios Personalizados

Revista

Articulo

Indicadores

Links relacionados

  • En proceso de indezaciónCitado por Google
  • No hay articulos similaresSimilares en SciELO
  • En proceso de indezaciónSimilares en Google

Compartir


Anales del Sistema Sanitario de Navarra

versión impresa ISSN 1137-6627

Anales Sis San Navarra vol.42 no.2 Pamplona may./ago. 2019  Epub 02-Mar-2020

https://dx.doi.org/10.23938/assn.0643 

Reviews

Galectin-3 as a novel biotarget in cardiovascular alterations associated to development of severe aortic stenosis

La galectina-3, una nueva diana terapéutica para las alteraciones cardiovasculares asociadas al desarrollo de la estenosis aórtica severa

V Arrieta1  2  , JR Sádaba1  2  , V Álvarez1  2  , JA Rodríguez3  4  , N López-Andrés1  2 

1Navarrabiomed. Universidad Pública de Navarra.

2Instituto de Investigación Sanitaria de Navarra. Pamplona. Spain.

3Laboratory of Atherothrombosis. Program of Cardiovascular Diseases. Center for Applied Medical Research. University of Navarra. Pamplona. Spain.

4CIBERCV. Madrid. Spain.

Abstract

Aortic stenosis is one of the most common heart valve diseases, as well as one of the most common causes of heart failure in the elderly. Currently, there are no medical therapies to prevent or slow the progression of the disease. When symptoms develop alongside severe aortic stenosis, there is a poor prognosis unless aortic valve replacement is performed. Aortic stenosis is a heterogeneous disease with a complex pathophysiology involving structural and biological changes of the valve, as well as adaptive and maladaptive compensatory changes in the myocardium and vasculature in response to chronic pressure overload. Galectin-3 serves important functions in numerous biological activities including cell growth, apoptosis, differentiation, inflammation and fibrosis. With evidence emerging to support the function of Galectin-3, the current review aims to summarize the latest literature regarding the potential of Galectin-3 as therapeutic target in aortic valve and cardiovascular alterations associated with aortic stenosis.

Keywords: Galectin-3; Aortic stenosis; Myocardial fibrosis; Valve calcification

Resumen

La estenosis aórtica severa degenerativa (EA) es una enfermedad muy prevalente, cuya incidencia se incrementará en los próximos años debido al envejecimiento de la población. Actualmente no existe ningún tratamiento farmacológico que retarde su progresión y, cuando aparecen los síntomas, la cirugía de recambio valvular es la única opción. La EA se caracteriza por la calcificación de la válvula aórtica y por la aparición de fibrosis miocárdica. Sin embargo, no se conocen los mecanismos fisiopatológicos de la EA necesarios para identificar y desarrollar nuevas estrategias terapéuticas adecuadas. La Galectina-3 (Gal-3) regula funciones biológicas como el crecimiento, la diferenciación, la apoptosis, la inflamación o la fibrosis. Esta revisión resume los principales trabajos que describen el potencial de la Gal-3 como diana terapéutica para las alteraciones cardíacas y valvulares asociadas con el desarrollo de EA.

Palabras clave: Galectina-3; Estenosis aórtica; Fibrosis miocárdica; Calcificación valvular

Aortic stenosis

Aortic stenosis (AS) is the most common heart valve disease (43%) and represents a major healthcare burden, since it is the third leading cause of cardiovascular disease1. Risk factors include male gender, smoking, diabetes mellitus, hypertension, high levels of circulating lipids, and metabolic syndrome2. With the increase in the aging population, there is a surge in the prevalence of calcific aortic valve disease. A prediction on the number of elderly (≥70 years) for the next few decades estimated that patients with severe AS will increase 2.4 fold by the year 2040 and more than triple by the year 20603. Patients with AS have an 80% risk of valve replacement, progression to heart failure (HF), or death in the next 5 years after diagnosis4.

The aortic valve is composed of three leaflets attached to the fibrous ring at the outlet of the left ventricle5. The leaflets are composed of a dense extracellular matrix usually delineated into three layers with different matrix composition, populated with valve interstitial cells (VICs) and the entire structure covered by valve endothelial cells: lamina fibrosa is the widest layer and faces the aortic or arterial side of the valve cusp, and it is composed principally by collagen circumferentially oriented to provide tensile strength6; lamina spongiosa is rich in glycosaminoglycans and proteoglycans that are believed to confer flexibility, dampen vibrations from closing, and resist delamination7; lamina ventricularis is a dense sheet of elastic fibres on the inflow side of the valve that is compliant, and provides elasticity and preload to the leaflets8. During embryogenesis the endothelial cells covering the primordial valve cushions migrate inside the underlying matrix and undergo endothelial to mesenchymal transition to become the interstitial cells9.

The pathophysiology underlying calcific aortic valve disease remains incompletely defined and there are currently no effective medical treatments capable of altering its course10. Chronic inflammation, fibrosis and calcification play an important role in the progression of the disease11. The aortic valve leaflets are a highly specialized structure consisting mostly of VICs and complex extracellular matrix structures12-13. An inflammatory and fibrotic process in aortic valve in humans and animal models has been previously reported14-15. Aberrant remodelling of the extracellular matrix is also caused by the deregulated overexpression of matrix metalloproteinases, associated with inflammation16. These events occur during the activation of VICs towards an osteogenic-like phenotype, promoted by the up-regulation of bone morphogenetic proteins pathway17. Therefore, it has been shown that calcific aortic valve disease shares features with vascular calcification and atherosclerosis such as chronic inflammation, increased extracellular matrix remodelling, proliferation and differentiation of VICs and the development of calcific lesions12 18. Of note, although retrospective studies had suggested that statins could delay the hemodynamic progression rate of AS19-20, in contrast, randomized controlled studies reported that a lipid-lowering strategy neither resulted in lower aortic valve-related events nor in a slower progression rate of stenosis21-22.

Moreover, chronic pressure overload in AS induces a structural remodeling of the left ventricle and may promote HF23 In the initial phases, the increased afterload imposed by aortic valve narrowing induces adaptive left ventricular hypertrophy that acts to maintain wall stress and cardiac output. Ultimately, this process decompensates, and patients transition from hypertrophy to HF and the development of symptoms and adverse cardiovascular events18. This transition is predominantly driven by myocardial fibrosis and myocyte cell death24. Thus, the transition from hypertrophy to HF plays a key role in AS. A better knowledge of the underlying mechanisms may highlight novel mediators of cardiac remodeling and decompensation which could identify biotargets for novel pharmacological therapies.

Galectin-3

Galectin-3 (Gal-3) is a 29-35 kDa protein, member of a β-galactoside binding lectin family, localized in nucleus, cytoplasm, cell surface and extracellular space25. It is composed of a highly conserved N-terminal domain and a C-terminal carbohydrate recognition domain, which interacts with glycoproteins26. The damaging effects of Gal-3 have been associated to its capacity to bind matrix proteins such as cell surface receptors (integrins), collagen, elastin or fibronectin27. The expression of this lectin has been reported in many tissues, including heart, vessels and kidney28. Moreover, Gal-3 is expressed in many cell types of the cardiovascular system such as cardiac fibroblasts29, vascular smooth muscle cells30, endothelial cells31, VICs32 and inflammatory cells33. Gal-3 is involved in numerous physiological and pathological processes some of which, inflammation and fibrosis, are pivotal contributing to pathophysiological mechanisms in the development and progression of HF.

The effects of Gal-3 in cells from the cardiovascular system have been largely investigated (Fig.1).

VCMCs: vascular smooth muscle cells; VICs: valve interstitial cells

Figure 1. Involvement of Galectin-3 in cellular pathophysiological processes associated with aortic stenosis 

Indeed, it has been demonstrated in cell culture that Gal-3 turns quiescent fibroblasts into myofibroblasts that produce and secrete matrix proteins, including collagen29 34. Gal-3 exerts its effects during several other stages of fibrogenesis besides collagen production, such as collagen maturation and cross-linking, which underscores the pivotal importance of Gal-3 in cardiovascular fibrosis35-36. Moreover, Gal-3 has emerged as a potential mediator of cardiovascular damage in different pathological situations through its ability to stimulate key pro-inflammatory molecules33. Thus, it has been demonstrated in human cardiac fibroblasts that Gal-3 enhances the production and the secretion of proinflammatory and profibrotic mediators such as interleukin-1β, IL-6, monocyte chemoattractant protein-1, collagen type I, collagen type III, fibronectin as well as the activity of metalloproteinases-1, -2 and -937. At the vascular level, Gal-3 increases the production and secretion of pro-fibrotic and pro-inflammatory markers in vascular smooth muscle cells30, contributing to arterial stiffness. In endothelial cells, Gal-3 increases the expression of inflammatory factors (interleukin-6, interleukin-8, and interleukin-1β), chemokines (monocyte chemoattractant protein-1) and adhesion molecules38. Furthermore, Gal-3 modulates cell surface expression and activation of vascular endothelial growth factor receptor 2 in human endothelial cells contributing to the plasma membrane retention and exerting a pro-angiogenic function39. In VICs from aortic valves, Gal-3 also increases the secretion of pro-inflammatory and pro-fibrotic markers as well as the expression of calcification markers32.

Beneficial effects of Galectin-3 blockade on aortic valve alterations in aortic stenosis

Chronic pressure overload due to AS results in pathological morphological changes in the cardiovascular system. These changes result in an initially compensatory phase, whose persistance could produce an important impact on cardiovascular function40. Pressure overload induces a modification in the aortic valves and the valve cusps become progressively thickened, fibrosed and calcified41. Moreover, a combination of endothelial damage and lipid deposition causes inflammation within the aortic valve that facilitates the infiltration of inflammatory cells which release proinflammatory factors42 43-44. In addition, matrix metalloproteinases secreted by VICs and inflammatory cells have an important and complex role in the restructuring of the aortic valve matrix43. Thus, abnormal remodeling in the aortic valve is also accompanied by the deregulated expression of metalloproteinases and inflammation16. As the stenosis-induced pressure overload progresses, wall shear stress across the aortic valve dramatically increases45, activating transforming growth factor-β146, that can also induce fibrosis and calcification44.

Gal-3 expression has been recently reported in VICs from aortic valves in patients undergoing aortic valve replacement32. Moreover, Gal-3 co-localized with the expression of osteogenic and inflammatory markers in human aortic valves32. Furthermore, in vitro, in human VICs, Gal-3 pharmacological inhibition with modified citrus pectin (MCP) as well as Gal-3 silencing attenuated the pro-inflammatory, pro-fibrotic and pro-osteogenic response32. A recent study described an association of Gal-3 with mortality after balloon aortic valvuloplasty, which is indicative of a contribution of local valvular Gal-3 expression to post-valvuloplasty restenosis47. In pressure overload, there is evidence of aberrant matrix deposition and valve fibrosis, which contributes to the calcification48.

In agreement with these data, AS animals presented increased aortic valve inflammation, fibrosis, metalloproteinase activities and calcification markers. The pharmacological inhibition of Gal-3 was able to decrease the aortic valve inflammation, fibrosis, metalloproteinase activities and calcification in absence of increased blood pressure levels in the pressure overload group, showing the potential therapeutic benefit of Gal-3 inhibition both in the primary (i.e., in early stages of pressure overload) and secondary prevention settings (i.e., when pressure overload is installed) (Fig. 2)49.

(from Ibarrola et al, 2017)56

Figure 2. Beneficial effects of Galectin-3 blockade with modified citrus pectin (MCP) in aortic valve remodeling in an experimental model of aortic stenosis (AS). For collagen quantification (fibrosis), Sirius red staining was performed. Representative immunohistochemistry for cd68 and bone morphogenetic protein 4 are showed as examples of aortic valve inflammation and calcification respectively 56  

Beneficial effects of Galectin-3 blockade on cardiac alterations in aortic stenosis

AS accompanied by chronic pressure overload is a known precursor of left ventricular remodeling, involving cardiac fibrosis and inflammation. Patients display a marked variation in the magnitude of their left ventricular remodeling. This has recently been demonstrated to be of prognostic importance50. As with fibrosis in the valve, an imbalance in metalloproteinases and tissue inhibitor of metalloproteinase activities and inflammation have all been implicated in this process. In human myocardium, Gal-3 is mainly expressed by cardiac fibroblasts and can be found in extracellular matrix51. Moreover, increased Gal-3 expression has been previously shown in myocardium from AS patients with depressed ejection fraction, as compared to myocardium from AS patients with preserved ejection fraction29, suggesting a role for Gal-3 in cardiac dysfunction associated with AS. Besides, cardiac Gal-3 expression has been found to be increased in animal models of pressure overload34 51-52 and paralleled the severity of left ventricular diastolic dysfunction52.

Several findings reported by our group deal with the potential consequences of Gal-3 overexpression in myocardium of AS patients. Firstly, cardiac Gal-3 overexpression is associated with cardiac fibrosis and inflammation51. Secondly, both cardiac and circulating Gal-3 levels positively correlated with cardiac fibrosis in AS patients53. Thirdly, a recent study showed that Gal-3 may serve as a prognostic biomarker after transcatheter aortic valve implantation by reflecting the degree of myocardial fibrosis54. Additionally, cardiac Gal-3 expression is associated with inflammatory markers and metalloproteinase-1 in myocardial biopsies from AS patients18, reinforcing the key role of this lectin in the inflammatory process and in extracellular matrix remodeling that accompanies the development of AS.

Previous studies have demonstrated that Gal-3 pharmacological inhibition prevented cardiac dysfunction, fibrosis and inflammation in several pathophysiological conditions such as hyperaldosteronism,37 55 obesity37 or hypertension37. Similar beneficial effects have been reported on cardiac fibrosis, remodeling and dysfunction in Gal-3 knockout mice subjected to thoracic aortic constriction34. In line with these findings, pharmacological blockade of Gal-3 is able to prevent cardiac fibrosis, inflammation and functional alterations in an animal model of early stages of AS (Fig. 3). Thus, these results show the key role of Gal-3 in the cardiac remodeling associated with AS development and the beneficial effects of Gal-3 pharmacological inhibition on cardiac fibrosis and inflammation, the two key processes underlying the cardiac functional alterations which finally could affect cardiac function and AS progression, leading to HF.

(from Ibarrola et al, 201756 and Arrieta et al, 201751)

Figure 3. Beneficial effects of Galectin-3 blockade with modified citrus pectin (MCP) in cardiac and vascular remodeling in an experimental model of aortic stenosis (AS). For collagen quantification (fibrosis), Sirius red staining was performed in cardiac and aortic sections. In myocardium, cd68 was used as inflammatory marker, whereas in aortic sections monocyte chemoattractant protein-1 was chosen 

Beneficial effects of Galectin-3 blockade on vascular alterations in aortic stenosis

At vascular level, pressure overload induces and increment of the aortic diameter and thickening of aortic wall through the extracellular matrix remodeling, characterized by an increment of fibrosis, inflammation and calcification in vessels and aortic valves45-46. Ascending aortic constriction is the most common surgical model for creating pressure overload-induced cardiovascular alterations. Gal-3 may contribute toward adverse cardiovascular effects in-part through an effect on aortic stiffness, effects which cannot be attributed to generalized inflammation.

In a recent study, it has been demonstrated that pharmacological Gal-3 inhibition by MCP could delay vascular remodeling and inflammation in a rat model of pressure overload (Fig. 3)51 56. Gal-3 inhibition exerts beneficial effects, decreasing aortic tunica media hypertrophy. Moreover, the use of MCP also decreases aortic fibrosis induced by pressure overload. Thus, the expression of collagen type I, fibronectin, α-smooth muscle actin, transforming growth factor-β1 and connective tissue growth factor was decreased in AS rats treated with the Gal-3 pharmacological inhibitor MCP. Complementarily, MCP treatment diminishes the expression of the inflammatory markers interleukin-6, interleukin-1β, tumor necrosis factor-α, monocyte chemoattractant protein-1, osteopontin, cd45 and cd68 in pressure-overloaded aortae (Fig. 3) 51 56. These results suggest that Gal-3 may contribute toward adverse cardiovascular effects in part through an effect on aortic stiffness. In line with these findings, it has been shown that Gal-3 also contributes to ventricular-vascular uncoupling in HF patients57.

Conclusions

Aortic stenosis is a disease of both the valve and the myocardium, characterized by fibrosis and calcification of valve leaflets, progressive left ventricular hypertrophy and cardiovascular fibrosis. In aortic stenosis, Gal-3 expression is increased in aortic valves, myocardium and aorta. Moreover, Gal-3 is colocalized with calcification markers in aortic valves, and with fibroblasts and extracellular matrix markers in myocardium. Gal-3 promotes inflammation, fibrosis and calcification in primary valvular interstitial cells and enhances the expression of fibrotic and inflammatory markers in cardiac fibroblasts and in vascular smooth muscle cells. Importantly, Gal-3 inhibition blocked aortic valve calcification, cardiac and vascular fibrosis and inflammation in vivo in an experimental model of pressure overload. Targeting Gal-3 may be an upstream therapeutic option for the treatment of aortic valve and cardiovascular remodeling that accompanies the progression of aortic stenosis. More in-depth mechanistic studies would be needed to understand the mechanisms by which Gal-3 inhibition blocks cardiovascular damage in aortic stenosis. Further clinical studies are required to establish the potential therapeutic benefit of Gal-3 inhibition in aortic stenosis patients.

Bibliografía

1. Iung B, Baron G, Butchart EG, Delahaye F, Gohlke-Bärwolf C, Levang OW, et al. A prospective survey of patients with valvular heart disease in Europe: The Euro heart survey on valvular heart disease. Eur Heart J 2003; 24: 1231-1243. https://doi.org/10.1016/S0195668X(03)00201-X [ Links ]

2. O'Brien KD. Epidemiology and genetics of calcific aortic valve disease. J Investig Med 2007; 55: 284-291. https://doi.org/10.2310/6650.2007.00010 [ Links ]

3 Danielsen R, Aspelund T, Harris TB, Gudnason V. The prevalence of aortic stenosis in the elderly in Iceland and predictions for the coming decades: the AGES-Reykjavík study. Int J Cardiol 2014; 20: 916-922. https://doi.org/10.1016/j.ijcard.2014.08.053 [ Links ]

4. Otto CM, Burwash IG, Legget ME, Munt BI, Fujioka M, Healy NL, et al. Prospective study of asymptomatic valvular aortic stenosis. Clinical, echocardiographic, and exercise predictors of outcome. Circulation 1997; 6: 2262-2270; https://doi.org/10.1161/01.CIR.95.9.2262 [ Links ]

5. Misfeld M, Sievers H-H. Heart valve macro- and microstructure. Philos Trans R Soc Lond B Biol Sci 2007; 29: 1421-1436. ; https://doi.org/10.1161/01.CIR.95.9.2262 [ Links ]

6. Doehring TC, Kahelin M, Vesely I. Mesostructures of the aortic valve. J Heart Valve Dis 2005; 14: 679-686. [ Links ]

7. Stephens EH, Chu C-K, Grande-Allen KJ. Valve proteoglycan content and glycosaminoglycan fine structure are unique to microstructure, mechanical load and age: Relevance to an age-specific tissue-engineered heart valve. Acta Biomater 2008; 4: 1148-1160. https://doi.org/10.1016/j.actbio.2008.03.014 [ Links ]

8. Helske S, Kupari M, Lindstedt KA, Kovanen PT. Aortic valve stenosis: an active atheroinflammatory process. Curr Opin Lipidol 2007; 18: 483-491. https://doi.org/10.1097/MOL.0b013e3282a66099 [ Links ]

9. Butcher JT, Nerem RM. Valvular endothelial cells and the mechanoregulation of valvular pathology. Philos Trans R Soc B Biol Sci 2007; 29: 1445-1457. https://doi.org/10.1098/rstb.2007.2127 [ Links ]

10. Bossé Y, Miqdad A, Fournier D, Pépin A, Pibarot P, Mathieu P. Refining molecular pathways leading to calcific aortic valve stenosis by studying gene expression profile of normal and calcified stenotic human aortic valves. Circ Cardiovasc Genet 2009; 2: 489-498. https://doi.org/10.1161/CIRCGENETICS.108.820795 [ Links ]

11. Mohler ER, Gannon F, Reynolds C, Zimmerman R, Keane MG, Kaplan FS. Bone formation and inflammation in cardiac valves. Circulation 2001; 20: 1522-1528. https://doi.org/10.1161/01.CIR.103.11.1522 [ Links ]

12. Bäck M, Gasser TC, Michel J-B, Caligiuri G. Biomechanical factors in the biology of aortic wall and aortic valve diseases. Cardiovasc Res 2013; 15: 232-241. https://doi.org/10.1093/cvr/cvt040 [ Links ]

13. Merryman WD, Youn I, Lukoff HD, Krueger PM, Guilak F, Hopkins RA, et al. Correlation between heart valve interstitial cell stiffness and transvalvular pressure: implications for collagen biosynthesis. Am J Physiol Heart Circ Physiol 2006; 290: H224-231. https://doi.org/10.1152/ajpheart.00521.2005 [ Links ]

14. Towler DA. Molecular and cellular aspects of calcific aortic valve disease. Circ Res 2013; 5: 198-208. https://doi.org/10.1161/CIRCRESAHA.113.300155 [ Links ]

15. Miller JD, Weiss RM, Serrano KM, Brooks RM, Berry CJ, Zimmerman K, et al. Lowering plasma cholesterol levels halts progression of aortic valve disease in mice. Circulation 2009; 26: 2693-2701. https://doi.org/10.1161/CIRCULATIONAHA.108.834614 [ Links ]

16. Vanhoutte D, Van Almen GC, Van Aelst LNL, Van Cleemput J, Droogné W, Jin Y, et al. Matricellular proteins and matrix metalloproteinases mark the inflammatory and fibrotic response in human cardiac allograft rejection. Eur Heart J 2013; 1: 1930-1941. https://doi.org/10.1093/eurheartj/ehs375 [ Links ]

17. Poggio P, Sainger R, Branchetti E, Grau JB, Lai EK, Gorman RC, et al. Noggin attenuates the osteogenic activation of human valve interstitial cells in aortic valve sclerosis. Cardiovasc Res 2013; 1: 402-410. https://doi.org/10.1093/cvr/cvt055 [ Links ]

18. Dweck MR, Boon NA, Newby DE. Calcific Aortic Stenosis. J Am Coll Cardiol 2012; 6: 1854-1863. https://doi.org/10.1016/j.jacc.2012.02.093 [ Links ]

19. Rajamannan NM, Subramaniam M, Springett M, Sebo TC, Niekrasz M, McConnell JP, et al. Atorvastatin inhibits hypercholesterolemia-induced cellular proliferation and bone matrix production in the rabbit aortic valve. Circulation 2002; 4: 2660-2665. https://doi.org/10.1161/01.CIR.0000017435.87463.72 [ Links ]

20. Rajamannan NM, Subramaniam M, Stock SR, Stone NJ, Springett M, Ignatiev KI, et al. Atorvastatin inhibits calcification and enhances nitric oxide synthase production in the hypercholesterolaemic aortic valve. Heart 2005; 1: 806-810. https://doi.org/10.1136/hrt.2003.029785 [ Links ]

21. Chan KL, Teo K, Dumesnil JG, Ni A, Tam J, Astronomer Investigators. Effect of lipid lowering with rosuvastatin on progression of aortic stenosis: results of the aortic stenosis progression observation: measuring effects of rosuvastatin (ASTRONOMER) trial. Circulation 2010; 19: 306-314. https://doi.org/10.1161/CIRCULATIONAHA.109.900027 [ Links ]

22. Cowell SJ, Newby DE, Prescott RJ, Bloomfield P, Reid J, Northridge DB, et al. A randomized trial of intensive lipid-lowering therapy in calcific aortic stenosis. N Engl J Med 2005; 352: 2389-2397. https://doi.org/10.1056/NEJMoa043876 [ Links ]

23. Mann DL. Left ventricular size and shape: determinants of mechanical signal transduction pathways. Heart Fail Rev 2005; 10: 95-100. https://doi.org/10.1007/s10741-0054636-y [ Links ]

24. Hein S, Arnon E, Kostin S, Schönburg M, Elsässer A, Polyakova V, et al. Progression from compensated hypertrophy to failure in the pressure-overloaded human heart: structural deterioration and compensatory mechanisms. Circulation 2003; 107: 984-991. https://doi.org/10.1161/01.CIR.0000051865.66123.B7 [ Links ]

25. Dumic J, Dabelic S, Flögel M. Galectin-3: an open-ended story. Biochim biophys acta. 2006; 1760: 616-635. https://doi.org/10.1016/j.bbagen.2005.12.020 [ Links ]

26. Dennis JW, Pawling J, Cheung P, Partridge E, Demetriou M. UDP-N-acetylglucosamine: alpha-6-D-mannoside beta1,6 N-acetylglucosaminyltransferase V (Mgat5) deficient mice. Biochim Biophys Acta 2002; 19: 414-422. https://doi.org/10.1016/S0304-4165(02)00411-7 [ Links ]

27. Ochieng J, Furtak V, Lukyanov P. Extracellular functions of galectin-3. Glycoconj J 2002; 19: 527-535. https://doi.org/10.1023/B:GLYC.0000014082.99675.2f [ Links ]

28. Kim H, Lee J, Hyun JW, Park JW, Joo H, Shin T. Expression and immunohistochemical localization of galectin-3 in various mouse tissues. Cell Biol Int 2007; 31: 655-662. https://doi.org/10.1016/j.cellbi.2006.11.036 [ Links ]

29. Sharma UC, Pokharel S, van Brakel TJ, van Berlo JH, Cleutjens JPM, Schroen B, et al. Galectin-3 marks activated macrophages in failure-prone hypertrophied hearts and contributes to cardiac dysfunction. Circulation 2004; 110: 3121-3128. https://doi.org/10.1161/01.CIR.0000147181.65298.4D [ Links ]

30. Calvier L, Miana M, Reboul P, Cachofeiro V, Martinez-Martinez E, de Boer RA, et al. Galectin-3 mediates aldosterone-induced vascular fibrosis. Arterioscler Thromb Vasc Biol 2013; 33: 67-75. https://doi.org/10.1161/ATVBAHA.112.300569 [ Links ]

31. Wan SY, Zhang TF, Ding Y. Galectin-3 enhances proliferation and angiogenesis of endothelial cells differentiated from bone marrow mesenchymal stem cells. Transplant Proc 2011; 43: 3933-3938. https://doi.org/10.1016/j.transproceed.2011.10.050 [ Links ]

32. Sádaba JR, Martínez-Martínez E, Arrieta V, Álvarez V, Fernández-Celis A, Ibarrola J, et al. Role for galectin-3 in calcific aortic valve stenosis. J Am Heart Assoc 2016; 5. https://doi.org/10.1161/JAHA.116.004360 [ Links ]

33. Papaspyridonos M, McNeill E, de Bono JP, Smith A, Burnand KG, Channon KM, et al. Galectin-3 is an amplifier of inflammation in atherosclerotic plaque progression through macrophage activation and monocyte chemoattraction. Arterioscler Thromb Vasc Biol 2008; 28: 433-440.https://doi.org/10.1161/ATVBAHA.107.159160 [ Links ]

34. Yu L, Ruifrok WPT, Meissner M, Bos EM, van Goor H, Sanjabi B, et al. Genetic and pharmacological inhibition of galectin-3 prevents cardiac remodeling by interfering with myocardial fibrogenesis. Circ Heart Fail 2013; 6: 107-117. https://doi.org/10.1161/CIRCHEARTFAILURE.112.971168 [ Links ]

35. Ho MK, Springer TA. Mac-2, a novel 32,000 Mr mouse macrophage subpopulation-specific antigen defined by monoclonal antibodies. J Immunol 1982; 128: 1221-1228. [ Links ]

36. 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: 237-246. https://doi.org/10.1007/s10557-014-6520-2 [ Links ]

37. Martínez-Martínez E, López-Ándres N, Jurado-López R, Rousseau E, Bartolomé MV, Fernández-Celis A, et al. Galectin-3 participates in cardiovascular remodeling associated with obesity. Hypertension 2015; 66: 961-969. https://doi.org/10.1161/HYPERTENSIONAHA.115.06032 [ Links ]

38. Chen X, Zhang R, Zhang Q, Xu Z, Xu F, Li D, et al. Microtia patients: Auricular chondrocyte ECM is promoted by CGF through IGF-1 activation of the IGF-1R/PI3K/AKT pathway. J Cell Physiol 2018. https://doi.org/10.1002/jcp.27316 [ Links ]

39. Markowska AI, Jefferies KC, Panjwani N. Galectin-3 protein modulates cell surface expression and activation of vascular endothelial growth factor receptor 2 in human endothelial cells. J Biol Chem 2011; 25: 29913-29921. https://doi.org/10.1074/jbc.M111.226423 [ Links ]

40. Gs AK, Raj B, Santhosh KS, Sanjay G, Kartha CC. Ascending aortic constriction in rats for creation of pressure overload cardiac hypertrophy model. J Vis Exp 2014; e50983. https://doi.org/10.3791/50983 [ Links ]

41. Mourino-Alvarez L, Baldan-Martin M, Gonzalez-Calero L, Martinez-Laborde C, Sastre-Oliva T, Moreno-Luna R, et al. Patients with calcific aortic stenosis exhibit systemic molecular evidence of ischemia, enhanced coagulation, oxidative stress and impaired cholesterol transport. Int J Cardiol 2016; 225: 99-106. https://doi.org/10.1016/j.ijcard.2016.09.089 [ Links ]

42. Kaden JJ, Dempfle C-E, Grobholz R, Tran H-T, Kiliç R, Sarikoç A, et al. Interleukin-1 beta promotes matrix metalloproteinase expression and cell proliferation in calcific aortic valve stenosis. Atherosclerosis 2003; 25: 205-211. https://doi.org/10.1016/S0021-9150(03)00284-3 [ Links ]

43. Malek AM, Gibbons GH, Dzau VJ, Izumo S. Fluid shear stress differentially modulates expression of genes encoding basic fibroblast growth factor and platelet-derived growth factor B chain in vascular endothelium. J Clin Invest 1993; 1: 2013-2021. https://doi.org/10.1172/JCI116796 [ Links ]

44. Laurent S, Boutouyrie P, Asmar R, Gautier I, Laloux B, Guize L, et al. Aortic stiffness is an independent predictor of all-cause and cardiovascular mortality in hypertensive patients. Hypertens (Dallas, Tex 1979): 2001; 25: 1236-1241. https://doi.org/10.1161/01.HYP.37.5.1236 [ Links ]

45. Gould ST, Srigunapalan S, Simmons CA, Anseth KS. Hemodynamic and cellular response feedback in calcific aortic valve disease. Circ Res 2013; 5: 186-197. https://doi.org/10.1161/CIRCRESAHA.112.300154 [ Links ]

46. Ahamed J, Burg N, Yoshinaga K, Janczak CA, Rifkin DB, Coller BS. In vitro and in vivo evidence for shear-induced activation of latent transforming growth factor- 1. Blood 2008; 1: 3650-3660. https://doi.org/10.1182/blood-2008-04-151753 [ Links ]

47. Bobrowska B, Wieczorek-Surdacka E, Kruszelnicka O, Chyrchel B, Surdacki A, Dudek D. Clinical correlates and prognostic value of plasma galectin-3 levels in degenerative aortic stenosis: a single-center prospective study of patients referred for invasive treatment. Int J Mol Sci 2017; 29: 947. https://doi.org/10.3390/ijms18050947 [ Links ]

48. Bäck M, Gasser TC, Michel J-B, Caligiuri G. Biomechanical factors in the biology of aortic wall and aortic valve diseases. Cardiovasc Res 2013; 15: 232-241. https://doi.org/10.1093/cvr/cvt040 [ Links ]

49. Sádaba JR, Martínez-Martínez E, Arrieta V, Álvarez V, Fernández-Celis A, Ibarrola J, et al. Role for galectin-3 in calcific aortic valve stenosis. J Am Heart Assoc 2016; 4. https://doi.org/10.1161/JAHA.116.004360 [ Links ]

50. Cioffi G, Faggiano P, Vizzardi E, Tarantini L, Cramariuc D, Gerdts E, et al. Prognostic effect of inappropriately high left ventricular mass in asymptomatic severe aortic stenosis. Heart 2011; 15: 301-330. https://doi.org/10.1136/hrt.2010.192997 [ Links ]

51. Arrieta V, Martinez-Martinez E, Ibarrola J, Alvarez V, Sádaba R, Garcia-Peña A, et al. A role for galectin-3 in the development of early molecular alterations in short-term aortic stenosis. Clin Sci (Lond) 2017; 1: 935-949. https://doi.org/10.1042/CS20170145 [ Links ]

52. Wu CK, Su MY, Lee JK, Chiang FT, Hwang JJ, Lin JL, et al. Galectin-3 level and the severity of cardiac diastolic dysfunction using cellular and animal models and clinical indices. Sci Rep 2015; 5: 17007. https://doi.org/10.1038/srep17007 [ Links ]

53. Zhou K, Zhou Y, Zhao Y, Tan C, Yuan Z, Li J, et al. The relationship between galectin-3 and different patterns of ventricular geometry remodelling in aortic valve stenosis. Heart Lung Circ 2016; 25: 371-377. https://doi.org/10.1016/j.hlc.2015.08.021 [ Links ]

54. Baldenhofer G, Zhang K, Spethmann S, Laule M, Eilers B, Leonhardt F, et al. Galectin-3 predicts short- and long-term outcome in patients undergoing transcatheter aortic valve implantation (TAVI). Int J Cardiol 2014; 20: 912-917. https://doi.org/10.1016/j.ijcard.2014.10.010 [ Links ]

55. Calvier L, Martinez-Martinez E, Miana M, Cachofeiro V, Rousseau E, Sádaba JR, et al. The impact of galectin-3 inhibition on aldosterone-induced cardiac and renal injuries. JACC Heart Fail 2015; 3: 59-67. https://doi.org/10.1016/j.jchf.2014.08.002 [ Links ]

56. Ibarrola J, Martínez-Martínez E, Sádaba JR, Arrieta V, García-Peña A, Álvarez V et al. Beneficial effects of galectin-3 blockade in vascular and aortic valve alterations in an experimental pressure overload model. Int J Mol Sci 2017; 31: 1664. https://doi.org/10.3390/ijms18081664 [ Links ]

57. Lala RI, Darabantiu D, Pilat L, Puschita M. Galectin-3: A Link between myocardial and arterial stiffening in patients with acute decompensated heart failure? Arq Bras Cardiol 2016; 25: 121-129. https://doi.org/10.5935/abc.20150149 [ Links ]

Received: April 30, 2019; Revised: May 31, 2019; Accepted: June 10, 2019

Correspondencia: Natalia López-Andrés Navarrabiomed Irunlarrea 3 31008 Pamplona España. E-mail: nlopezan@navarra.es

Creative Commons License This is an open-access article distributed under the terms of the Creative Commons Attribution License