Skip to main content

Advertisement

Log in

RAGE/galectin-3 yields intraplaque calcification transformation via sortilin

  • Original Article
  • Published:
Acta Diabetologica Aims and scope Submit manuscript

Abstract

Aims

Macrocalcification and microcalcification present different clinical risks, but the regulatory of their formation was unclear. Therefore, this study explored the underlying mechanisms of macrocalcification and microcalcification in diabetes mellitus.

Methods

Anterior tibial arteries of amputated diabetic feet were collected. According to the calcium content, patients were divided into less-calcification group and more-calcification group. And calcification morphology in plaques was observed. For further study, an in vivo mouse diabetic atherosclerosis model and an in vitro primary mouse aortic smooth muscle cell model were established. After the receptors for AGEs (RAGE) or galectin-3 were silenced, calcified nodule sizes and sortilin expression were determined. Scanning electron microscopy (SEM) was performed to detect the aggregation of matrix vesicles with the inhibition or promotion of sortilin.

Results

Both macro- and microcalcification were found in human anterior tibial artery plaques. Macrocalcification formed after the silencing of RAGE, and microcalcification formed after the silencing of galectin-3. In the process of RAGE- or galcetin-3-induced calcification, sortilin played an important role downstream. SEM showed that sortilin promoted the aggregation of MVs in the early stage of calcification and formed larger calcified nodules.

Conclusion

RAGE downregulated sortilin and then transmitted microcalcification signals, whereas galectin-3 upregulated sortilin, which accelerated the aggregation of MVs in the early stage of calcification and mediated the formation of macrocalcifications, These data illustrate the progression of two calcification types and suggest sortilin as a potential target for early intervention of calcification and as an effective biomarker for the assessment of long-term clinical risk and prognosis.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7

Similar content being viewed by others

Data availability

All data and materials are available upon request.

Abbreviations

VSMCs:

Vascular smooth muscle cells

CML:

Nε-carboxymethyl-lysine

AGEs:

Advanced glycation end products

RAGE:

Receptor for advanced glycation end products

GWAS:

Genome-wide association study

SPF:

Specific pathogen free

STZ:

Streptozotocin

AAV:

Adeno-associated viral

OM:

Osteogenic medium

LV:

Lentivirus vector

oxLDL:

Oxidized low-density lipoprotein

CoIP:

Coimmunoprecipitation

SEM:

Scanning electron microscopy

NTA:

Nanoparticle tracking analysis

MVs:

Matrix vesicles

HFD:

High-fat diet

RUNX2:

Runt-related transcription factor 2

PBS:

Phosphate-buffered saline

TNAP:

Tissue nonspecific alkaline phosphatase

EVs:

Extracellular vesicles

CCK-8:

Cell counting kit-8

MOI:

Multiplicity of infection

References

  1. Benjamin EJ, Virani SS, Callaway CW et al (2018) Heart disease and stroke statistics-2018 update: a report from the american heart association. Circulation 137(12):CIR.0000000000000558

    Article  Google Scholar 

  2. Xu S, Ye F, Li L et al (2017) Ghrelin attenuates vascular calcification in diabetic patients with amputation. Biomed Pharmacother 91:1053–1064

    Article  CAS  PubMed  Google Scholar 

  3. Singh DK, Winocour P, Summerhayes B et al (2012) Prevalence and progression of peripheral vascular calcification in type 2 diabetes subjects with preserved kidney function. Diabetes Res Clin Pract 97:158–165

    Article  CAS  PubMed  Google Scholar 

  4. Kelly-Arnold A, Maldonado N, Laudier D, Aikawa E, Cardoso L, Weinbaum S (2013) Revised microcalcification hypothesis for fibrous cap rupture in human coronary arteries. Proc Natl Acad Sci USA 110:10741–10746

    Article  PubMed  Google Scholar 

  5. Hutcheson JD, Goettsch C, Bertazzo S et al (2016) Genesis and growth of extracellular-vesicle-derived microcalcification in atherosclerotic plaques. Nat Mater 15:335–343

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Janssen R (2017) Magnesium to counteract elastin degradation and vascular calcification in chronic obstructive pulmonary disease. Med Hypotheses 107:74–77

    Article  CAS  PubMed  Google Scholar 

  7. Nakahara T, Dweck MR, Narula N, Pisapia D, Narula J, Strauss HW (2017) Coronary artery calcification: from mechanism to molecular imaging. JACC Cardiovasc Imaging 10:582–593

    Article  Google Scholar 

  8. Yao Y, Bennett BJ, Wang X et al (2010) Inhibition of bone morphogenetic proteins protects against atherosclerosis and vascular calcification. Circ Res 107(4):485–494

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Menini S, Iacobini C, Ricci C et al (2013) The galectin-3/RAGE dyad modulates vascular osteogenesis in atherosclerosis. Cardiovasc Res 100(3):472–480

    Article  CAS  PubMed  Google Scholar 

  10. Wang Z, Jiang Y, Liu N et al (2012) Advanced glycation end-product Nε-carboxymethyl-Lysine accelerates progression of atherosclerotic calcification in diabetes. Atherosclerosis 221:387–396

    Article  CAS  PubMed  Google Scholar 

  11. Wang Z, Li L, Du R et al (2016) CML/RAGE signal induces calcification cascade in diabetes. Diabetol Metab Syndr 8:83–94

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Zhou Z, Immel D, Xi CX et al (2006) Regulation of osteoclast function and bone mass by RAGE. J Exp Med 203:1067–1080

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Stock M, Schäfer H, Stricker S, Gross G, Mundlos S, Otto F (2003) Expression of galectin-3 in skeletal tissues is controlled by Runx2. J Biol Chem 278:17360–17367

    Article  CAS  PubMed  Google Scholar 

  14. Strong A, Ding Q, Edmondson AC et al (2012) Hepatic sortilin regulates both apolipoprotein B secretion and LDL catabolism. J Clin Invest 122:2807–2816

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Greenwood SG, Montroull L, Volosin M et al (2018) A novel neuroprotective mechanism for lithium that prevents association of the p75(NTR)-sortilin receptor complex and attenuates proNGF-Induced neuronal death in vitro and in vivo. eNeuro 5:ENEURO.0257-17

    Article  Google Scholar 

  16. O’Donnell CJ, Kavousi M, Smith AV et al (2011) Genome-wide association study for coronary artery calcification with follow-up in myocardial infarction. Circulation 124:2855–2864

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Kaddai V, Jager J, Gonzalez T et al (2009) Involvement of TNF-alpha in abnormal adipocyte and muscle sortilin expression in obese mice and humans. Diabetologia 52:932–940

    Article  CAS  PubMed  Google Scholar 

  18. Kjolby M, Andersen OM, Breiderhoff T et al (2010) Sort1, encoded by the cardiovascular risk locus 1p13.3, is a regulator of hepatic lipoprotein export. Cell Metab 12:213–223

    Article  CAS  PubMed  Google Scholar 

  19. Alves RD, Eijken M, Bezstarosti K, Demmers JA, van Leeuwen JP (2013) Activin A suppresses osteoblast mineralization capacity by altering extracellular matrix (ECM) composition and impairing matrix vesicle (MV) production. Mol Cell Proteomics 12:2890–2900

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Gungor O, Kocyigit I, Yilmaz MI, Sezer S (2018) Role of vascular calcification inhibitors in preventing vascular dysfunction and mortality in hemodialysis patients. Semin Dial 31:72–81

    Article  PubMed  Google Scholar 

  21. Pérez-Hernández N, Aptilon-Duque G, Blachman-Braun R et al (2017) Vascular calcification: current genetics underlying this complex phenomenon. Chin Med J (Engl) 130:1113–1121

    Article  Google Scholar 

  22. Cui L, Houston DA, Farquharson C, MacRae VE (2016) Characterisation of matrix vesicles in skeletal and soft tissue mineralisation. Bone 87:147–158

    Article  CAS  PubMed  Google Scholar 

  23. Zazzeroni L, Faggioli G, Pasquinelli G (2018) Mechanisms of arterial calcification: the role of matrix vesicles. Eur J Vasc Endovasc Surg 55:425–433

    Article  PubMed  Google Scholar 

  24. Moe SM, O’Neill KD, Duan D et al (2002) Medial artery calcification in ESRD patients is associated with deposition of bone matrix proteins. Kidney Int 61:638–647

    Article  PubMed  Google Scholar 

  25. Marcusson EG, Horazdovsky BF, Cereghino JL, Gharakhanian E, Emr SD (1994) The sorting receptor for yeast vacuolar carboxypeptidase Y is encoded by the VPS10 gene. Cell 77:579–586

    Article  CAS  PubMed  Google Scholar 

  26. Linsel-Nitschke P, Heeren J, Aherrahrou Z et al (2010) Genetic variation at chromosome 1p13.3 affects sortilin mRNA expression, cellular LDL-uptake and serum LDL levels which translates to the risk of coronary artery disease. Atherosclerosis 208:183–189

    Article  CAS  PubMed  Google Scholar 

  27. Bi L, Chiang JY, Ding WX, Dunn W, Roberts B, Li T (2013) Saturated fatty acids activate ERK signaling to downregulate hepatic sortilin 1 in obese and diabetic mice. J Lipid Res 54:2754–2762

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Béraud-Dufour S, Devader C, Massa F, Roulot M, Coppola T, Mazella J (2016) Focal adhesion kinase-dependent role of the soluble form of neurotensin receptor-3/sortilin in colorectal cancer cell dissociation. Int J Mol Sci 17:1860

    Article  CAS  PubMed Central  Google Scholar 

  29. Patel KM, Strong A, Tohyama J et al (2015) Macrophage sortilin promotes LDL uptake, foam cell formation, and atherosclerosis. Circ Res 116:789–796

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Saada S, Marget P, Fauchais AL et al (2012) Differential expression of neurotensin and specific receptors, NTSR1 and NTSR2, in normal and malignant human B lymphocytes. J Immunol 189:5293–5303

    Article  CAS  PubMed  Google Scholar 

  31. Gustafsen C, Glerup S, Pallesen LT et al (2013) Sortilin and SorLA display distinct roles in processing and trafficking of amyloid precursor protein. J Neurosci 33:64–71

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Goettsch C, Hutcheson JD, Aikawa M et al (2016) Sortilin mediates vascular calcification via its recruitment into extracellular vesicles. J Clin Invest 126:1323–1336

    Article  PubMed  PubMed Central  Google Scholar 

  33. Patel JJ, Zhu D, Opdebeeck B et al (2018) Inhibition of arterial medial calcification and bone mineralization by extracellular nucleotides: The same functional effect mediated by different cellular mechanisms. J Cell Physiol 233:3230–3243

    Article  CAS  PubMed  Google Scholar 

  34. Chen NX, O’Neill KD, Moe SM (2018) Matrix vesicles induce calcification of recipient vascular smooth muscle cells through multiple signaling pathways. Kidney Int 93:343–354

    Article  CAS  PubMed  Google Scholar 

  35. Shioi A, Ikari Y (2017) Plaque calcification during atherosclerosis progression and regression. J Atheroscler Thromb 25:294–303

    Article  PubMed  Google Scholar 

  36. Bakhshian Nik A, Hutcheson JD, Aikawa E (2017) Extracellular vesicles as mediators of cardiovascular calcification. Front Cardiovasc Med 4:78

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Pugliese G, Iacobini C, Pesce CM, Menini S (2015) Galectin-3: an emerging all-out player in metabolic disorders and their complications. Glycobiology 25:136–150

    Article  CAS  PubMed  Google Scholar 

  38. Savic-Radojevic A, Pljesa-Ercegovac M, Matic M, Simic D, Radovanovic S, Simic T (2017) Novel biomarkers of heart failure. Adv Clin Chem 79:93–152

    Article  CAS  PubMed  Google Scholar 

  39. Suthahar N, Meijers WC, Silljé HHW, Ho JE, Liu FT, de Boer RA (2018) Galectin-3 activation and inhibition in heart failure and cardiovascular disease: an update. Theranostics 8:593–609

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Funding

This work was supported by the foundations as follows: the National Natural Science Foundation of China (Grant Nos. 81770450, 81370408, 81670405), the Foundation of Jiangsu Province (WSN-044, QNRC2016836), the Open Program of Key Laboratory of Nuclear Medicine, Ministry of Health and Jiangsu Key Laboratory of Molecular Nuclear Medicine (KF201504) and Graduate Student Scientific Research Innovation Projects of Jiangsu Province (KYCX17_1801, SJCX18_0754).

Author information

Authors and Affiliations

Authors

Contributions

ZS performed, and analyzed experiments, produced figures, and wrote the manuscript. LL contributed to section preparation and immunofluorescence analysis. JY helped the in vivo and in vitro models establishment. CS contributed to gene silencing of cells and mice. ZB helped with SEM and data analysis. LJ contributed to MVs isolation and NTA. YG helped with clinical data collection and analysis. PQ and LZ provided suggestions for experimental design. ZW designed and supervised experiments and wrote the manuscript. All authors read and approved the final manuscript.

Corresponding author

Correspondence to Zhongqun Wang.

Ethics declarations

Conflict of interest

The authors declare that they have no conflict of interest.

Human and animal rights disclosure

Human studies conform to the principles outlined in the Declaration of Helsinki (1964) and was approved by the Ethical Committee of the Affiliated Hospital of Jiangsu University. All animal experiments were approved by the Animal Health and Utilization Committee of the Affiliated Hospital of Jiangsu University, and carried out in accordance with the guidelines from Directive 2010/63/EU and “Principles of laboratory animal care” (NIH publication No. 86-23, revised 1985).

Informed consent

All patients gave consent prior to inclusion.

Additional information

Managed By Massimo Porta.

Publisher’s Note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary material 1 (DOCX 4718 KB)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sun, Z., Wang, Z., Li, L. et al. RAGE/galectin-3 yields intraplaque calcification transformation via sortilin. Acta Diabetol 56, 457–472 (2019). https://doi.org/10.1007/s00592-018-1273-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00592-018-1273-1

Keywords

Navigation