CC BY-NC-ND 4.0 · Thromb Haemost 2022; 122(06): 1047-1057
DOI: 10.1055/a-1711-1055
Atherosclerosis and Ischaemic Disease

Anti-Galectin-2 Antibody Treatment Reduces Atherosclerotic Plaque Size and Alters Macrophage Polarity

Jamie Kane*
1   Department of Nephrology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, Amsterdam, The Netherlands
2   Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, Amsterdam, The Netherlands
3   Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, Amsterdam, The Netherlands
,
Matthijs Jansen*
2   Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, Amsterdam, The Netherlands
4   Department of Cardiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, Amsterdam, The Netherlands
,
Sebastian Hendrix
2   Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, Amsterdam, The Netherlands
,
Laura A. Bosmans
2   Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, Amsterdam, The Netherlands
,
Linda Beckers
2   Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, Amsterdam, The Netherlands
,
Claudia van Tiel
2   Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, Amsterdam, The Netherlands
,
Marion Gijbels
2   Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, Amsterdam, The Netherlands
5   Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM), GROW-School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
,
Noam Zelcer
2   Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, Amsterdam, The Netherlands
,
Carlie J. de Vries
2   Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, Amsterdam, The Netherlands
,
6   Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillian's University, Munich, Germany
,
Marc Vervloet
1   Department of Nephrology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, Amsterdam, The Netherlands
,
Ed Eringa
3   Department of Physiology, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, Amsterdam, The Netherlands
,
Anton J. Horrevoets
7   Department of Molecular Cell Biology and Immunology, Amsterdam University Medical Centre, Amsterdam, The Netherlands
,
Niels van Royen*
8   Department of Cardiology, Radboud University Medical Centre, Nijmegen, The Netherlands
,
Esther Lutgens
2   Department of Medical Biochemistry, Amsterdam Cardiovascular Sciences, Amsterdam University Medical Centre, Amsterdam, The Netherlands
6   Institute for Cardiovascular Prevention (IPEK), Ludwig-Maximillian's University, Munich, Germany
9   German Centre for Cardiovascular Research (DZHK), partner site Munich Heart Alliance, Munich, Germany
10   Cardiovascular Medicine, Experimental Cardiovascular Immunology Laboratory, Mayo Clinic, Rochester, Minnesota, United States
› Institutsangaben
Funding This project has received funding from the European Union's Horizon 2020 research and innovation program under the Marie Skłodowska-Curie Actions, grant agreement No. 812699 (to J.K.). We also acknowledge the support from Amsterdam Cardiovascular Sciences for grant support (to M.J.), as well as the Netherlands Cardiovascular Research Initiative: the Dutch Heart Foundation, Dutch Federation of University Medical Centres, the Netherlands Organization for Health Research, and Development and the Royal Netherlands Academy of Sciences for the GENIUS-II project ‘Generating the best evidence-based pharmaceutical targets for atherosclerosis’ (CVON2017–22 to E.L.). This study was also supported by the Deutsche Forschungsgemeinschaft (CRC 1123 to E.L., P.v.H.).

Abstract

Background Galectins have numerous cellular functions in immunity and inflammation. Short-term galectin-2 (Gal-2) blockade in ischemia-induced arteriogenesis shifts macrophages to an anti-inflammatory phenotype and improves perfusion. Gal-2 may also affect other macrophage-related cardiovascular diseases.

Objectives This study aims to elucidate the effects of Gal-2 inhibition in atherosclerosis.

MethodsApoE −/− mice were given a high-cholesterol diet (HCD) for 12 weeks. After 6 weeks of HCD, intermediate atherosclerotic plaques were present. To study the effects of anti-Gal-2 nanobody treatment on the progression of existing atherosclerosis, treatment with two llama-derived anti-Gal-2 nanobodies (clones 2H8 and 2C10), or vehicle was given for the remaining 6 weeks.

Results Gal-2 inhibition reduced the progression of existing atherosclerosis. Atherosclerotic plaque area in the aortic root was decreased, especially so in mice treated with 2C10 nanobodies. This clone showed reduced atherosclerosis severity as reflected by a decrease in fibrous cap atheromas in addition to decreases in plaque size.

The number of plaque resident macrophages was unchanged; however, there was a significant increase in the fraction of CD206+ macrophages. 2C10 treatment also increased plaque α-smooth muscle content, and Gal-2 may have a role in modulating the inflammatory status of smooth muscle cells. Remarkably, both treatments reduced serum cholesterol concentrations including reductions in very low-density lipoprotein, low-density lipoprotein, and high-density lipoprotein while triglyceride concentrations were unchanged.

Conclusion Prolonged and frequent treatment with anti-Gal-2 nanobodies reduced plaque size, slowed plaque progression, and modified the phenotype of plaque macrophages toward an anti-inflammatory profile. These results hold promise for future macrophage modulating therapeutic interventions that promote arteriogenesis and reduce atherosclerosis.

* Shared authorship.


Supplementary Material



Publikationsverlauf

Eingereicht: 05. November 2020

Angenommen: 26. September 2021

Accepted Manuscript online:
01. Dezember 2021

Artikel online veröffentlicht:
08. Februar 2022

© 2021. The Author(s). This is an open access article published by Thieme under the terms of the Creative Commons Attribution-NonDerivative-NonCommercial License, permitting copying and reproduction so long as the original work is given appropriate credit. Contents may not be used for commercial purposes, or adapted, remixed, transformed or built upon. (https://creativecommons.org/licenses/by-nc-nd/4.0/)

Georg Thieme Verlag KG
Rüdigerstraße 14, 70469 Stuttgart, Germany

 
  • References

  • 1 Gisterå A, Hansson GK. The immunology of atherosclerosis. Nat Rev Nephrol 2017; 13 (06) 368-380
  • 2 Than NG, Romero R, Goodman M. et al. A primate subfamily of galectins expressed at the maternal-fetal interface that promote immune cell death. Proc Natl Acad Sci U S A 2009; 106 (24) 9731-9736
  • 3 Vasta GR. Galectins as pattern recognition receptors: structure, function, and evolution. Adv Exp Med Biol 2012; 946: 21-36
  • 4 Loser K, Sturm A, Voskort M. et al. Galectin-2 suppresses contact allergy by inducing apoptosis in activated CD8+ T cells. J Immunol 2009; 182 (09) 5419-5429
  • 5 Rabinovich GA, Toscano MA. Turning ‘sweet’ on immunity: galectin-glycan interactions in immune tolerance and inflammation. Nat Rev Immunol 2009; 9 (05) 338-352
  • 6 Si Y, Feng S, Gao J. et al. Human galectin-2 interacts with carbohydrates and peptides non-classically: new insight from X-ray crystallography and hemagglutination. Acta Biochim Biophys Sin (Shanghai) 2016; 48 (10) 939-947
  • 7 Sturm A, Lensch M, André S. et al. Human galectin-2: novel inducer of T cell apoptosis with distinct profile of caspase activation. J Immunol 2004; 173 (06) 3825-3837
  • 8 Paclik D, Berndt U, Guzy C. et al. Galectin-2 induces apoptosis of lamina propria T lymphocytes and ameliorates acute and chronic experimental colitis in mice. J Mol Med (Berl) 2008; 86 (12) 1395-1406
  • 9 Meier P, Gloekler S, Zbinden R. et al. Beneficial effect of recruitable collaterals: a 10-year follow-up study in patients with stable coronary artery disease undergoing quantitative collateral measurements. Circulation 2007; 116 (09) 975-983
  • 10 Hollander MR, Horrevoets AJG, van Royen N. Cellular and pharmacological targets to induce coronary arteriogenesis. Curr Cardiol Rev 2014; 10 (01) 29-37
  • 11 van der Laan AM, Schirmer SH, de Vries MR. et al. Galectin-2 expression is dependent on the rs7291467 polymorphism and acts as an inhibitor of arteriogenesis. Eur Heart J 2012; 33 (09) 1076-1084
  • 12 Yıldırım C, Vogel DYS, Hollander MR. et al. Galectin-2 induces a proinflammatory, anti-arteriogenic phenotype in monocytes and macrophages. PLoS One 2015; 10 (04) e0124347
  • 13 Hollander MR, Jansen MF, Hopman LHGA. et al. Stimulation of collateral vessel growth by inhibition of galectin 2 in mice using a single-domain llama-derived antibody. J Am Heart Assoc 2019; 8 (20) e012806
  • 14 Ikeda S, Tanaka N, Arai T, Chida K, Muramatsu M, Sawabe M. Polymorphisms of LTA, LGALS2, and PSMA6 genes and coronary atherosclerosis: a pathological study of 1503 consecutive autopsy cases. Atherosclerosis 2012; 221 (02) 458-460
  • 15 Harmsen MM, De Haard HJ. Properties, production, and applications of camelid single-domain antibody fragments. Appl Microbiol Biotechnol 2007; 77 (01) 13-22
  • 16 Virmani R, Kolodgie FD, Burke AP, Farb A, Schwartz SM. Lessons from sudden coronary death: a comprehensive morphological classification scheme for atherosclerotic lesions. Arterioscler Thromb Vasc Biol 2000; 20 (05) 1262-1275
  • 17 Fedoseienko A, Wijers M, Wolters JC. et al. The COMMD family regulates plasma LDL levels and attenuates atherosclerosis through stabilizing the CCC complex in endosomal LDLR trafficking. Circ Res 2018; 122 (12) 1648-1660
  • 18 Goldstein JL, DeBose-Boyd RA, Brown MS. Protein sensors for membrane sterols. Cell 2006; 124 (01) 35-46
  • 19 Lin J, Kakkar V, Lu X. Impact of MCP-1 in atherosclerosis. Curr Pharm Des 2014; 20 (28) 4580-4588
  • 20 Grundmann S, Hoefer I, Ulusans S. et al. Granulocyte-macrophage colony-stimulating factor stimulates arteriogenesis in a pig model of peripheral artery disease using clinically applicable infusion pumps. J Vasc Surg 2006; 43 (06) 1263-1269
  • 21 Subramanian M, Thorp E, Tabas I. Identification of a non-growth factor role for GM-CSF in advanced atherosclerosis: promotion of macrophage apoptosis and plaque necrosis through IL-23 signaling. Circ Res 2015; 116 (02) e13-e24
  • 22 Sparkes A, De Baetselier P, Brys L. et al. Novel half-life extended anti-MIF nanobodies protect against endotoxic shock. FASEB J 2018; 32 (06) 3411-3422
  • 23 Harman JL, Jørgensen HF. The role of smooth muscle cells in plaque stability: therapeutic targeting potential. Br J Pharmacol 2019; 176 (19) 3741-3753
  • 24 Willemsen L, de Winther MPJ. Macrophage subsets in atherosclerosis as defined by single-cell technologies. J Pathol 2020; 250 (05) 705-714
  • 25 Yvan-Charvet L, Wang N, Tall AR. Role of HDL, ABCA1, and ABCG1 transporters in cholesterol efflux and immune responses. Arterioscler Thromb Vasc Biol 2010; 30 (02) 139-143
  • 26 Brunham LR, Singaraja RR, Duong M. et al. Tissue-specific roles of ABCA1 influence susceptibility to atherosclerosis. Arterioscler Thromb Vasc Biol 2009; 29 (04) 548-554
  • 27 Westerterp M, Murphy AJ, Wang M. et al. Deficiency of ATP-binding cassette transporters A1 and G1 in macrophages increases inflammation and accelerates atherosclerosis in mice. Circ Res 2013; 112 (11) 1456-1465
  • 28 Bochem AE, van Wijk DF, Holleboom AG. et al. ABCA1 mutation carriers with low high-density lipoprotein cholesterol are characterized by a larger atherosclerotic burden. Eur Heart J 2013; 34 (04) 286-291