The role of circulating galectin-1 in type 2 diabetes and chronic kidney disease: evidence from cross-sectional, longitudinal and Mendelian randomisation analyses

Isabel Drake, Emanuel Fryk, Lena Strindberg, Annika Lundqvist, Anders H Rosengren, Leif Groop, Emma Ahlqvist, Jan Borén, Marju Orho-Melander, Per-Anders Jansson, Isabel Drake, Emanuel Fryk, Lena Strindberg, Annika Lundqvist, Anders H Rosengren, Leif Groop, Emma Ahlqvist, Jan Borén, Marju Orho-Melander, Per-Anders Jansson

Abstract

Aims/hypothesis: Galectin-1 modulates inflammation and angiogenesis, and cross-sectional studies indicate that galectin-1 may be a uniting factor between obesity, type 2 diabetes and kidney function. We examined whether circulating galectin-1 can predict incidence of chronic kidney disease (CKD) and type 2 diabetes in a middle-aged population, and if Mendelian randomisation (MR) can provide evidence for causal direction of effects.

Methods: Participants (n = 4022; 58.6% women) in the Malmö Diet and Cancer Study-Cardiovascular Cohort enrolled between 1991 and 1994 (mean age 57.6 years) were examined. eGFR was calculated at baseline and after a mean follow-up of 16.6 ± 1.5 years. Diabetes status was ascertained through registry linkage (mean follow-up of 18.4 ± 6.1 years). The associations of baseline galectin-1 with incident CKD and type 2 diabetes were assessed with Cox regression, adjusting for established risk factors. In addition, a genome-wide association study on galectin-1 was performed to identify genetic instruments for two-sample MR analyses utilising the genetic associations obtained from the Chronic Kidney Disease Genetics (CKDGen) Consortium (41,395 cases and 439,303 controls) and the DIAbetes Genetics Replication And Meta-analysis (DIAGRAM) consortium (74,124 cases and 824,006 controls). One genome-wide significant locus in the galectin-1 gene region was identified (sentinel SNP rs7285699; p = 2.4 × 10-11). The association between galectin-1 and eGFR was also examined in individuals with newly diagnosed diabetes from the All New Diabetics In Scania (ANDIS) cohort.

Results: Galectin-1 was strongly associated with lower eGFR at baseline (p = 2.3 × 10-89) but not with incident CKD. However, galectin-1 was associated with increased risk of type 2 diabetes (per SD increase, HR 1.12; 95% CI 1.02, 1.24). Two-sample MR analyses could not ascertain a causal effect of galectin-1 on CKD (OR 0.92; 95% CI 0.82, 1.02) or type 2 diabetes (OR 1.05; 95% CI 0.98, 1.14) in a general population. However, in individuals with type 2 diabetes from ANDIS who belonged to the severe insulin-resistant diabetes subgroup and were at high risk of diabetic nephropathy, genetically elevated galectin-1 was significantly associated with higher eGFR (p = 5.7 × 10-3).

Conclusions/interpretation: Galectin-1 is strongly associated with lower kidney function in cross-sectional analyses, and two-sample MR analyses suggest a causal protective effect on kidney function among individuals with type 2 diabetes at high risk of diabetic nephropathy. Future studies are needed to explore the mechanisms by which galectin-1 affects kidney function and whether it could be a useful target among individuals with type 2 diabetes for renal improvement.

Keywords: ANDIS; Chronic kidney disease; Galectin-1; Human; Malmö Diet Cancer; Mendelian randomisation; Population-based; Prospective; Type 2 diabetes.

© 2021. The Author(s).

Figures

Fig. 1
Fig. 1
A flow chart of participants of the MDCS-CC with circulating levels of galectin-1 who were included in the cross-sectional and longitudinal analyses and in GWAS and MR analyses.T2D, type 2 diabetes
Fig. 2
Fig. 2
Genome-wide analyses for fasting serum concentration of galectin-1 in the MDCS-CC. (a) Manhattan plot of SNPs with p values <0.10 based on genome-wide analysis (chromosomes 1–22) in n = 4086 participants from the MDCS-CC. Red line indicates the genome-wide significant p value of 5 × 10−8. (b) Quantile–quantile (QQ) plot. (c) Regional locus zoom plot of associations at/near the galectin-1 gene (LGALS1). The purple diamond indicates the sentinel SNP (rs7285699; p = 2.4 × 10−11), and all identified SNPs within different degrees of perfect linkage disequilibrium are also shown (r2 ≥ 0.80 [red], <0.8–0.6 [orange], <0.6–0.4 [green], <0.4–0.2 [light blue] and ≤0.2 [dark blue]) at this locus. cM, centimorgans; Mb, megabase
Fig. 3
Fig. 3
Two-sample MR analyses for the association of genetically predicted serum galectin-1 levels with (a) CKD and type 2 diabetes and (b) creatinine-based eGFR, overall and stratified by diabetes mellitus status. aWuttke et al (2019) [25]; bPattaro et al (2016) [26]; cANDIS cohort. T2D, type 2 diabetes; WR, Wald ratio

References

    1. Zimmet PZ. Diabetes and its drivers: the largest epidemic in human history? Clin Diabetes Endocrinol. 2017;3:1. doi: 10.1186/s40842-016-0039-3.
    1. Thomas MC, Brownlee M, Susztak K, et al. Diabetic kidney disease. Nat Rev Dis Primers. 2015;1:15018. doi: 10.1038/nrdp.2015.18.
    1. Ahlqvist E, Storm P, Karajamaki A, et al. Novel subgroups of adult-onset diabetes and their association with outcomes: a data-driven cluster analysis of six variables. Lancet Diabetes Endocrinol. 2018;6(5):361–369. doi: 10.1016/S2213-8587(18)30051-2.
    1. Reily C, Stewart TJ, Renfrow MB, Novak J. Glycosylation in health and disease. Nat Rev Nephrol. 2019;15(6):346–366. doi: 10.1038/s41581-019-0129-4.
    1. Brinchmann MF, Patel DM, Iversen MH. The role of galectins as modulators of metabolism and inflammation. Mediat Inflamm. 2018;2018:9186940. doi: 10.1155/2018/9186940.
    1. Johannes L, Jacob R, Leffler H. Galectins at a glance. J Cell Sci. 2018;131(9):jcs208884. doi: 10.1242/jcs.208884.
    1. Al-Obaidi N, Mohan S, Liang S, et al. Galectin-1 is a new fibrosis protein in type 1 and type 2 diabetes. FASEB J. 2018;33(1):373–387. doi: 10.1096/fj.201800555RR.
    1. Liu Y, Long L, Yuan F, et al. High glucose-induced Galectin-1 in human podocytes implicates the involvement of Galectin-1 in diabetic nephropathy. Cell Biol Int. 2015;39(2):217–223. doi: 10.1002/cbin.10363.
    1. Kuo CS, Chou RH, Lu YW, Tsai YL, Huang PH, Lin SJ. Increased circulating galectin-1 levels are associated with the progression of kidney function decline in patients undergoing coronary angiography. Sci Rep. 2020;10(1):1435. doi: 10.1038/s41598-020-58132-1.
    1. Carlos CP, Silva AA, Gil CD, Oliani SM. Pharmacological treatment with galectin-1 protects against renal ischaemia-reperfusion injury. Sci Rep. 2018;8(1):9568. doi: 10.1038/s41598-018-27907-y.
    1. Mukherjee R, Kim SW, Park T, Choi MS, Yun JW. Targeted inhibition of galectin 1 by thiodigalactoside dramatically reduces body weight gain in diet-induced obese rats. Int J Obes. 2015;39(9):1349–1358. doi: 10.1038/ijo.2015.74.
    1. Baum LG, Seilhamer JJ, Pang M, Levine WB, Beynon D, Berliner JA. Synthesis of an endogeneous lectin, galectin-1, by human endothelial cells is up-regulated by endothelial cell activation. Glycoconj J. 1995;12(1):63–68. doi: 10.1007/bf00731870.
    1. Hsieh SH, Ying NW, Wu MH, et al. Galectin-1, a novel ligand of neuropilin-1, activates VEGFR-2 signaling and modulates the migration of vascular endothelial cells. Oncogene. 2008;27(26):3746–3753. doi: 10.1038/sj.onc.1211029.
    1. Lin YT, Chen JS, Wu MH, et al. Galectin-1 accelerates wound healing by regulating the neuropilin-1/Smad3/NOX4 pathway and ROS production in myofibroblasts. J Invest Dermatol. 2015;135(1):258–268. doi: 10.1038/jid.2014.288.
    1. Hagberg CE, Falkevall A, Wang X, et al. Vascular endothelial growth factor B controls endothelial fatty acid uptake. Nature. 2010;464(7290):917–921. doi: 10.1038/nature08945.
    1. Fryk E, Sundelin JP, Strindberg L, et al. Microdialysis and proteomics of subcutaneous interstitial fluid reveals increased galectin-1 in type 2 diabetes patients. Metabolism. 2016;65(7):998–1006. doi: 10.1016/j.metabol.2016.04.003.
    1. Fryk E, Strindberg L, Lundqvist A, et al. Galectin-1 is inversely associated with type 2 diabetes independently of obesity – a SCAPIS pilot study. Metab Open. 2019;4(2019):100017. doi: 10.1016/j.metop.2019.100017.
    1. Berglund G, Elmstahl S, Janzon L, Larsson SA. The Malmo diet and Cancer study. Design and feasibility. J Intern Med. 1993;233(1):45–51. doi: 10.1111/j.1365-2796.1993.tb00647.x.
    1. Hedblad B, Nilsson P, Janzon L, Berglund G. Relation between insulin resistance and carotid intima-media thickness and stenosis in non-diabetic subjects. Results from a cross-sectional study in Malmo, Sweden. Diabet Med. 2000;17(4):299–307. doi: 10.1046/j.1464-5491.2000.00280.x.
    1. Schulz CA, Engstrom G, Christensson A, Nilsson PM, Melander O, Orho-Melander M. Genetic predisposition for renal dysfunction and incidence of CKD in the Malmo diet and Cancer study. Kidney Int Rep. 2019;4:1143–1151. doi: 10.1016/j.ekir.2019.05.003.
    1. Levey AS, Stevens LA, Schmid CH, et al. A new equation to estimate glomerular filtration rate. Ann Intern Med. 2009;150(9):604–612. doi: 10.7326/0003-4819-150-9-200905050-00006.
    1. Rosvall M, Persson M, Ostling G, et al. Risk factors for the progression of carotid intima-media thickness over a 16-year follow-up period: the Malmo diet and Cancer study. Atherosclerosis. 2015;239(2):615–621. doi: 10.1016/j.atherosclerosis.2015.01.030.
    1. Chang CC, Chow CC, Tellier LC, Vattikuti S, Purcell SM, Lee JJ. Second-generation PLINK: rising to the challenge of larger and richer datasets. Gigascience. 2015;4:7. doi: 10.1186/s13742-015-0047-8.
    1. Levy JC, Matthews DR, Hermans MP. Correct homeostasis model assessment (HOMA) evaluation uses the computer program. Diabetes Care. 1998;21:2191–2192. doi: 10.2337/diacare.21.12.2191.
    1. Wuttke M, Li Y, Li M, et al. A catalog of genetic loci associated with kidney function from analyses of a million individuals. Nat Genet. 2019;51(6):957–972. doi: 10.1038/s41588-019-0407-x.
    1. Pattaro C, Teumer A, Gorski M, et al. Genetic associations at 53 loci highlight cell types and biological pathways relevant for kidney function. Nat Commun. 2016;7:10023. doi: 10.1038/ncomms10023.
    1. Mahajan A, Taliun D, Thurner M, et al. Fine-mapping type 2 diabetes loci to single-variant resolution using high-density imputation and islet-specific epigenome maps. Nat Genet. 2018;50(11):1505–1513. doi: 10.1038/s41588-018-0241-6.
    1. Hemani G, Bowden J, Davey Smith G. Evaluating the potential role of pleiotropy in Mendelian randomization studies. Hum Mol Genet. 2018;27(R2):R195–R208. doi: 10.1093/hmg/ddy163.
    1. Okano K, Tsuruta Y, Yamashita T, Takano M, Echida Y, Nitta K. Suppression of renal fibrosis by galectin-1 in high glucose-treated renal epithelial cells. Exp Cell Res. 2010;316(19):3282–3291. doi: 10.1016/j.yexcr.2010.08.015.
    1. Potthoff SA, Sitek B, Stegbauer J, et al. The glomerular proteome in a model of chronic kidney disease. Proteomics Clin Appl. 2008;2(7–8):1127–1139. doi: 10.1002/prca.200800010.
    1. Whaley-Connell A, Sowers JR. Obesity and kidney disease: from population to basic science and the search for new therapeutic targets. Kidney Int. 2017;92(2):313–323. doi: 10.1016/j.kint.2016.12.034.
    1. Hirano T. Abnormal lipoprotein metabolism in diabetic nephropathy. Clin Exp Nephrol. 2014;18(2):206–209. doi: 10.1007/s10157-013-0880-y.
    1. Webster AC, Nagler EV, Morton RL, Masson P. Chronic kidney disease. Lancet. 2017;389(10075):1238–1252. doi: 10.1016/S0140-6736(16)32064-5.
    1. Zhao JV, Schooling CM. Sex-specific associations of insulin resistance with chronic kidney disease and kidney function: a bi-directional Mendelian randomisation study. Diabetologia. 2020;63(8):1554–1563. doi: 10.1007/s00125-020-05163-y.
    1. Acar S, Paketci A, Kume T, et al. Serum galectin-1 levels are positively correlated with body fat and negatively with fasting glucose in obese children. Peptides. 2017;95:51–56. doi: 10.1016/j.peptides.2017.07.009.
    1. Blois SM, Gueuvoghlanian-Silva BY, Tirado-Gonzalez I, et al. Getting too sweet: galectin-1 dysregulation in gestational diabetes mellitus. Mol Hum Reprod. 2014;20(7):644–649. doi: 10.1093/molehr/gau021.
    1. Roumans NJT, Vink RG, Bouwman FG, Fazelzadeh P, van Baak MA, Mariman ECM. Weight loss-induced cellular stress in subcutaneous adipose tissue and the risk for weight regain in overweight and obese adults. Int J Obes. 2017;41(6):894–901. doi: 10.1038/ijo.2016.221.
    1. Kanda A, Dong Y, Noda K, Saito W, Ishida S. Advanced glycation endproducts link inflammatory cues to upregulation of galectin-1 in diabetic retinopathy. Sci Rep. 2017;7(1):16168. doi: 10.1038/s41598-017-16499-8.
    1. Rothman KJ. No adjustments are needed for multiple comparisons. Epidemiology. 1990;1(1):43–46. doi: 10.1097/00001648-199001000-00010.
    1. Muhammad IF, Borne Y, Hedblad B, Nilsson PM, Persson M, Engstrom G. Acute-phase proteins and incidence of diabetes: a population-based cohort study. Acta Diabetol. 2016;53:981–989. doi: 10.1007/s00592-016-0903-8.
    1. Matthews DR, Hosker JP, Rudenski AS, Naylor BA, Treacher DF, Turner RC. Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia. 1985;28:412–419. doi: 10.1007/BF00280883.

Source: PubMed

3
订阅