Role of Canagliflozin on function of CD34+ve endothelial progenitor cells (EPC) in patients with type 2 diabetes

Seshagiri Rao Nandula, Nabanita Kundu, Hassan B Awal, Beda Brichacek, Mona Fakhri, Nikhila Aimalla, Adrian Elzarki, Richard L Amdur, Sabyasachi Sen, Seshagiri Rao Nandula, Nabanita Kundu, Hassan B Awal, Beda Brichacek, Mona Fakhri, Nikhila Aimalla, Adrian Elzarki, Richard L Amdur, Sabyasachi Sen

Abstract

Background: Endothelial progenitor cells (EPCs) has been shown to be dysfunctional in both type 2 diabetes mellitus (T2DM) and chronic kidney disease (CKD) leading to poor regeneration of endothelium and renal perfusion. EPCs have been shown to be a robust cardiovascular disease (CVD) risk indicator. Effect of sodium glucose channel inhibitors (SGLT2i) such as Canagliflozin (CG) on a cellular biomarker such as CD34+ve progenitor cells, which may help predict CVD risk, in patients with T2DM with established CKD has not been explored.

Methods: This is a pilot study where 29 subjects taking metformin and/or Insulin were enrolled in a 16 week, double blind, randomized placebo matched trial, with a low dose 100 mg CG as the intervention group compared to matched placebo. Type 2 diabetes subjects (30-70 years old), with hemoglobin A1c (HbA1c) of 7-10%, were enrolled. CD34+ve cell number, migratory function, gene expression along with vascular parameters such as arterial stiffness, serum biochemistry pertaining to cardio-metabolic health, resting energy expenditure and body composition were measured. Data were collected at week 0, 8 and 16. A mixed model regression analysis was done and p value less than 0.05 was considered statistically significant.

Results: A significant expression of CXCR4 receptor with a concomittant increase in migratory function of CD34+ve cells was observed in CG treated group as compared to placebo group. Gene expression analysis of CD34+ve cells showed an increase in expression of antioxidants (superoxide dismutase 2 or SOD2, Catalase and Glutathione Peroxidase or GPX) and notable endothelial markers (PECAM1, VEGF-A, and NOS3). A significant reduction in glucose and HbA1c levels were observed along with improved systolic and diastolic blood pressure in the CG group. A significant increase in adiponectin (p = 0.006) was also noted in treatment group. Urinary exosomal protein leak in urine, examining podocyte health (podocalyxin, Wilm's tumor and nephrin) showed reduction with CG CONCLUSION: Low dose Canagliflozin has a beneficial effect on CD34+ cell function, serum biochemistry and urinary podocyte specific exosomes in type 2 diabetes.

Keywords: Canagliflozin; Endothelial function; Progenitor cells; Type 2 diabetes; Urinary exosomes.

Conflict of interest statement

The authors declare that they have no competing interests.

Figures

Fig. 1
Fig. 1
A significant difference in mean systolic blood pressure (a) and diastolic blood pressure (b) is observed between Canagliflozin and placebo groups. In Canagliflozin group both decreased from visit 1 to 2 and increased by visit 3. Whereas in placebo group both increased from visit 1 to 2 and decreased by visit 3. c HbA1c went down substantially in Canagliflozin group whereas it increased in Placebo group. The sharp decrease in HbA1c is seen in visit 1to visit 2 (p = 0.0919) and is stabilized from visit 2 to visit 3 in Canagliflozin group. d Glucose levels also followed the same trend as HbA1c in Canagliflozin group by sharp decrease from visit 1 to 2 and then stabilized till visit 3. Whereas, in placebo group glucose levels increased from visit 1 to 3. e A significant rise in adiponectin levels is noted in Canagliflozin group where as a sharp decrease is observed in placebo group from visit 2 to 3. f Inflammatory marker serum IL6 levels decreased. The difference was significant from visit 2 to 3 (p = 0.0259) in Canagliflozin group as compared to placebo group
Fig. 2
Fig. 2
Migration Assay in response to SDF1alpha at 10 ng/ml concentration and gene expression of SDF1a receptor, CXCR-4 on CD34+ve cells. a Mean migration of CD34+ve cells towards SDF1-α (at 10 ng/ml) increased from visit 1 to 3 in Canagliflozin group as compared to placebo group (for visit 1 to 3 p = 0.03 and for visit 2–3 p = 0.05). b CXCR4 gene expression on CD34 positive cells is increased (p = 0.06) in Canagliflozin group as compared to placebo group at visit 3
Fig. 3
Fig. 3
The effect of Canagliflozin on CD34+ve cell antioxidants gene expression. Fold change in gene expression of antioxidants SOD2 (a) in CD34+ve cells are increased in Canagliflozin group from visit 1 to 3. However, the same for the placebo group is decreased. Whereas the antioxidants, b Catalase (CAT) and c GPX3 gene expression increased in CD34+ve cells in Canagliflozin group from visit 2 to 3. However, the same for the placebo group is decreased. Overall, all the antioxidant genes showed increased expression going from visit 2 to visit 3, whereas the placebo there was a downward trend with catalase expression reaching statistical difference, overall
Fig. 4
Fig. 4
The effect of Canagliflozin on CD34+ve cell Endothelial markers gene expression. Fold change in gene expression of Endothelial markers a VEGF-A, b KDR and c PECAM and endothelial function marker d NOS3 on CD34+ve cells increased significantly in Canagliflozin group from visit 1 to 3. However, the same for the placebo group is decreased. Gene expression is normalized to 18S
Fig. 5
Fig. 5
Urinary exosome markers a Wilm’s tumor-1 (WT-1), b Nephrin and c Podocalyxin (PODXL) were identified by Western blot and followed by quantification. A trend in reduced mean band intensities for Urinary exosome markers are observed in Canagliflozin group from visit 1 to visit 3 as compared to placebo group

References

    1. IDF Diabetes Atlas. 9th edition; 2019. . Accessed 09 Nov 2020.
    1. US Renal Data System Annual Data Report; 2019. . Accessed 09 Nov 2020.
    1. Amorim RG, et al. Kidney disease in diabetes mellitus: cross-linking between hyperglycemia, redox imbalance and inflammation. Arq Bras Cardiol. 2019;112:5.
    1. Giacco F, Brownlee M. Oxidative stress and diabetic complications. Circ Res. 2010;107:9. doi: 10.1161/CIRCRESAHA.110.223545.
    1. Meza CA, et al. Endothelial dysfunction: is there a hyperglycemia-induced imbalance of NOX and NOS? Int J Mol Sci. 2019;20:15. doi: 10.3390/ijms20153775.
    1. Perlman RL, Fredri F, Liu L, et al. Quality of life in chronic kidney disease (CKD): a cross-sectional analysis in the Renal Research Institute-CKD study. Am J Kidney Dis. 2005;45:4. doi: 10.1053/j.ajkd.2004.12.021.
    1. Rosenthal N, et al. Canagliflozin: a sodium glucose co-transporter 2 inhibitor for the treatment of type 2 diabetes mellitus. Ann N Y Acad Sci. 2015;1358:28–43. doi: 10.1111/nyas.12852.
    1. van Bommel EJ, et al. SGLT2 inhibition in the diabetic kidney-from mechanisms to clinical outcome. Clin J Am Soc Nephrol. 2017;12:4.
    1. Huri HZ, Lim LP, Lim SK. Glycemic control and antidiabetic drugs in type 2 diabetes mellitus patients with renal complications. Drug Des Devel Ther. 2015;9:4355. doi: 10.2147/DDDT.S85676.
    1. Perkovic V, et al. Renal effects of canagliflozin in type 2 diabetes mellitus. Curr Med Res Opin. 2015;31:12. doi: 10.1185/03007995.2015.1092128.
    1. Abdul-Ghani MA, DeFronzo RA. Inhibition of renal glucose reabsorption: a novel strategy for achieving glucose control in type 2 diabetes mellitus. Endocr Pract. 2008;14:6. doi: 10.4158/EP.14.6.782.
    1. Lo MC, Lansang MC. Recent and emerging therapeutic medications in type 2 diabetes mellitus: incretin-based, pramlintide, colesevelam, SGLT2 inhibitors, tagatose, succinobucol. Am J Ther. 2013;20:6. doi: 10.1097/MJT.0b013e3181ec9eb2.
    1. Naznin F, et al. Canagliflozin, a sodium glucose cotransporter 2 inhibitor, attenuates obesity-induced inflammation in the nodose ganglion, hypothalamus, and skeletal muscle of mice. Eur J Pharmacol. 2017;794:37–44. doi: 10.1016/j.ejphar.2016.11.028.
    1. Rossing P, de Zeeuw D. Need for better diabetes treatment for improved renal outcome. Kidney Int Suppl. 2011;120:S28–S32. doi: 10.1038/ki.2010.513.
    1. Vallon V, Sharma K. Sodium–glucose transport: role in diabetes mellitus and potential clinical implications. Curr Opin Nephrol Hypertens. 2010;19:5. doi: 10.1097/MNH.0b013e32833bec06.
    1. Ryan PB, et al. Comparative effectiveness of canagliflozin, SGLT2 inhibitors and non-SGLT2 inhibitors on the risk of hospitalization for heart failure and amputation in patients with type 2 diabetes mellitus: a real-world meta-analysis of 4 observational databases (OBSERVE-4D) Diabetes Obes Metab. 2018;20:1. doi: 10.1111/dom.13424.
    1. Ali BH, et al. Effects of the SGLT-2 inhibitor canagliflozin on adenine-induced chronic kidney disease in rats. Cell Physiol Biochem. 2019;52:1. doi: 10.33594/000000001.
    1. Mahaffey KW, et al. Canagliflozin for primary and secondary prevention of cardiovascular events: results from the CANVAS program (canagliflozin cardiovascular assessment study) Circulation. 2018;137:4. doi: 10.1161/CIRCULATIONAHA.117.032038.
    1. Neal B, et al. Canagliflozin and cardiovascular and renal events in type 2 diabetes. N Engl J Med. 2017;377:7. doi: 10.1056/NEJMoa1611925.
    1. Neuen B, et al. Cardiovascular and renal outcomes with canagliflozin according to baseline kidney function. Circulation. 2018;138:15. doi: 10.1161/CIRCULATIONAHA.118.035901.
    1. Patorno E, et al. Cardiovascular outcomes associated with canagliflozin versus other non-gliflozin antidiabetic drugs: population based cohort study. BMJ. 2018;360:k119. doi: 10.1136/bmj.k119.
    1. Rådholm K, et al. Canagliflozin and heart failure in type 2 diabetes mellitus: results from the CANVAS program. Circulation. 2018;138:5. doi: 10.1161/CIRCULATIONAHA.118.034222.
    1. Skelley JW, Carter BS, Roberts MZ. Clinical potential of canagliflozin in cardiovascular risk reduction in patients with type 2 diabetes. Vasc Health Risk Manag. 2018;14:419. doi: 10.2147/VHRM.S168472.
    1. Mancini SJ, et al. Canagliflozin inhibits interleukin-1β-stimulated cytokine and chemokine secretion in vascular endothelial cells by AMP-activated protein kinase-dependent and -independent mechanisms. Sci Rep. 2018;8:1. doi: 10.1038/s41598-017-17765-5.
    1. Sayour AA, et al. Acute canagliflozin treatment protects against in vivo myocardial ischemia-reperfusion injury in non-diabetic male rats and enhances endothelium-dependent vasorelaxation. J Transl Med. 2019;17:1. doi: 10.1186/s12967-019-1881-8.
    1. Sen S. Adult stem cells: beyond regenerative tool, more as a bio-marker in obesity and diabetes. Diabetes Metab J. 2019;43:6. doi: 10.4093/dmj.2019.0175.
    1. Gendron N, Smadja DM. Circulating endothelial cells: a new biomarker of endothelial dysfunction in hematological diseases. Ann Biol Clin. 2016;74:4.
    1. Jain R, Awal H, Sen S. Using adult stem cells to monitor endothelial dysfunction in diabetes mellitus. J Diabetes Complicat. 2020;34(7):107588. doi: 10.1016/j.jdiacomp.2020.107588.
    1. Sen S, Witkowski S, Lagoy A, Islam A. A six-week home exercise program improves endothelial function and CD34+ circulating progenitor cells in patients with pre-diabetes. J Endocrinol Metab. 2015;5:1–2. doi: 10.14740/jem273w.
    1. Rigato M, Fadini GP. Circulating stem/progenitor cells as prognostic biomarkers in macro- and microvascular disease: a narrative review of prospective observational studies. Curr Med Chem. 2018;25:35. doi: 10.2174/0929867324666170920154020.
    1. Rohban R, Prietl B, Pieber TR. Crosstalk between stem and progenitor cellular mediators with special emphasis on vasculogenesis. Transfus Med Hemother. 2017;44:3. doi: 10.1159/000477677.
    1. Prokoph S, et al. Sustained delivery of SDF-1α from heparin-based hydrogels to attract circulating pro-angiogenic cells. Biomaterials. 2012;33:19. doi: 10.1016/j.biomaterials.2012.03.039.
    1. Shen L, et al. A novel mechanism for endothelial progenitor cells homing: the SDF-1/CXCR4-Rac pathway may regulate endothelial progenitor cells homing through cellular polarization. Med Hypotheses. 2011;76:2.
    1. Kutikhin AG, et al. Shear stress: an essential driver of endothelial progenitor cells. J Mol Cell Cardiol. 2018;118:46–69. doi: 10.1016/j.yjmcc.2018.03.007.
    1. Medina RJ, et al. Endothelial progenitors: a consensus statement on nomenclature. Stem Cells Transl Med. 2017;6:5. doi: 10.1002/sctm.16-0360.
    1. Bonora BM, et al. Effects of SGLT2 inhibitors on circulating stem and progenitor cells in patients with type 2 diabetes. J Clin Endocrinol Metab. 2018;103:10. doi: 10.1210/jc.2018-00824.
    1. Mudaliar S, Alloju S, Henry RR. Can a shift in fuel energetics explain the beneficial cardiorenal outcomes in the EMPA-REG OUTCOME study? A unifying hypothesis. Diabetes Care. 2016;39:7. doi: 10.2337/dc16-0542.
    1. Park SH, et al. Empagliflozin improved systolic blood pressure, endothelial dysfunction and heart remodeling in the metabolic syndrome ZSF1 rat. Cardiovasc Diabetol. 2020;19:1. doi: 10.1186/s12933-019-0977-z.
    1. Steven S, et al. The SGLT2 inhibitor empagliflozin improves the primary diabetic complications in ZDF rats. Redox Biol. 2017;13:370–385. doi: 10.1016/j.redox.2017.06.009.
    1. Uthman L, et al. Empagliflozin and dapagliflozin reduce ROS generation and restore NO bioavailability in tumor necrosis factor α-stimulated human coronary arterial endothelial cells. Cell Physiol Biochem. 2019;53:5.
    1. Stoner L, Young JM, Frye S. Assessments of arterial stiffness and endothelial function using pulse wave analysis. Int J Vasc Med. 2012;2012:903107. doi: 10.1155/2012/903107.
    1. Khiyami AM, Dore FJ, Mammadova A, Amdur RL, Sen S. The correlation of arterial stiffness with biophysical parameters and blood biochemistry. Metab Syndr Relat Disord. 2017;15(4):178–182. doi: 10.1089/met.2016.0136.
    1. Dore FJ, Domingues CC, Ahmadi N, Kundu N, Kropotova Y, Houston S, Rouphael C, Mammadova A, Witkin L, Khiyami A, Amdur RL, Sen S. The synergistic effects of saxagliptin and metformin on CD34+ endothelial progenitor cells in early type 2 diabetes patients: a randomized clinical trial. Cardiovasc Diabetol. 2018;17(1):65. doi: 10.1186/s12933-018-0709-9.
    1. Awal HB, Nandula SR, Domingues CC, Dore FJ, Kundu N, Brichacek B, Fakhri M, Elzarki A, Ahmadi N, Safai S, Fosso M, Amdur RL, Sen S. Linagliptin, when compared to placebo, improves CD34+ve endothelial progenitor cells in type 2 diabetes subjects with chronic kidney disease taking metformin and/or insulin: a randomized controlled trial. Cardiovasc Diabetol. 2020;19:1. doi: 10.1186/s12933-020-01046-z.
    1. Fadini GP, Avogar A. SGLT2 inhibitors and amputations in the US FDA adverse event reporting system. Lancet Diabetes Endocrinol. 2017;5:9. doi: 10.1016/S2213-8587(17)30257-7.
    1. Fadini GP, et al. Optimized glycaemic control achieved with add-on basal insulin therapy improves indexes of endothelial damage and regeneration in type 2 diabetic patients with macroangiopathy: a randomized crossover trial comparing detemir versus glargine. Diabetes Obes Metab. 2011;13:8. doi: 10.1111/j.1463-1326.2011.01396.x.
    1. Thiruvenkatarajan V, Meyer EJ, Nanjappa N, Van Wijk RM, Jesudason D. Perioperative diabetic ketoacidosis associated with sodium-glucose co-transporter-2 inhibitors: a systematic review. Br J Anaesth. 2019;123:1. doi: 10.1016/j.bja.2019.03.028.
    1. Yang X, Liu Q, Li Y, Tang Q, Wu T, Chen L, Pu S, Zhao Y, Zhang G, Huang C, Zhang J, Zhang Z, Huang Y, Zou M, Shi X, Jiang W, Wang R, He J. The diabetes medication canagliflozin promotes mitochondrial remodelling of adipocyte via the AMPK-Sirt1-Pgc-1α signalling pathway. Adipocyte. 2020;9:1. doi: 10.1080/21623945.2020.1807850.

Source: PubMed

3
订阅