The synergistic effects of saxagliptin and metformin on CD34+ endothelial progenitor cells in early type 2 diabetes patients: a randomized clinical trial

Fiona J Dore, Cleyton C Domingues, Neeki Ahmadi, Nabanita Kundu, Yana Kropotova, Sara Houston, Carol Rouphael, Aytan Mammadova, Linda Witkin, Anamil Khiyami, Richard L Amdur, Sabyasachi Sen, Fiona J Dore, Cleyton C Domingues, Neeki Ahmadi, Nabanita Kundu, Yana Kropotova, Sara Houston, Carol Rouphael, Aytan Mammadova, Linda Witkin, Anamil Khiyami, Richard L Amdur, Sabyasachi Sen

Abstract

Aims: Type 2 diabetes is associated with endothelial dysfunction leading to cardiovascular disease. CD34+ endothelial Progenitor Cells (EPCs) are responsible for endothelial repair and neo-angiogenesis and can be used as a cardiovascular disease risk biomarker. This study investigated whether the addition of saxagliptin, a DPP-IV inhibitor, to metformin, may reduce cardiovascular disease risk in addition to improving glycemic control in Type 2 diabetes patients.

Methods: In 12 week, double-blind, randomized placebo-controlled trial, 42 subjects already taking metformin 1-2 grams/day were randomized to placebo or saxagliptin 5 mg. Subjects aged 40-70 years with diabetes for < 10 years, with no known cardiovascular disease, BMI 25-39.9, HbA1C 6-9% were included. We evaluated EPCs number, function, surface markers and gene expression, in addition to arterial stiffness, blood biochemistries, resting energy expenditure, and body composition parameters. A mixed model regression to examine saxagliptin vs placebo, accounting for within-subject autocorrelation, was done with SAS (p < 0.05).

Results: Although there was no significant increase in CD34+ cell number, CD31+ cells percentage increased. Saxagliptin increased migration (in response to SDF1α) with a trend of higher colony formation count. MNCs cytometry showed higher percentage of CXCR4 double positivity for both CD34 and CD31 positive cells, indicating a functional improvement. Gene expression analysis showed an upregulation in CD34+ cells for antioxidant SOD1 (p < 0.05) and a downregulation in CD34- cells for IL-6 (p < 0.01). For arterial stiffness, both augmentation index and systolic blood pressure measures went down in saxagliptin subjects (p < 0.05).

Conclusion: Saxagliptin, in combination with metformin, can help improve endothelial dysfunction in early diabetes before macrovascular complications appear. Trial registration Trial is registered under clinicaltrials.gov, NCT02024477.

Keywords: Arterial stiffness; DPP-4 inhibitor; Diabetes; Endothelial progenitor cells; Saxagliptin.

Figures

Fig. 1
Fig. 1
CFU-Hill’s colonies as an indicator of vascular health. Experiments were performed in duplicate and values are given as mean ± SD (p = 0.07, for the visit x treatment interaction t-test in a random effects mixed model)
Fig. 2
Fig. 2
Migration of CD34+ cells in response to SDF-1α (100 ng/mL). Results are expressed as fluorescence ratio between cells exposed to the chemotactic factor and cells exposed to chemoattractant-free media (control) followed by lysis in presence of CyQuant GR dye. Experiments were performed in duplicate and results are given as mean ± SD (p 

Fig. 3

Representative image ( a and…

Fig. 3

Representative image ( a and b ) indicating the CD34+ CXCR4+ expression in…

Fig. 3
Representative image (a and b) indicating the CD34+ CXCR4+ expression in MNCs by flow cytometry. CD34+ CXCR4+ expression is higher for the saxagliptin group at visit 3 than visit 1 (7.95 and 4.64%, respectively). c Double positivity for CD34 and CXCR4 along the visits for placebo and saxagliptin groups (p < 0.01)

Fig. 4

CD31+ CXCR4+ expression in MNCs…

Fig. 4

CD31+ CXCR4+ expression in MNCs assessed by flow cytometry. Double positivity of CD31…

Fig. 4
CD31+ CXCR4+ expression in MNCs assessed by flow cytometry. Double positivity of CD31 and CXCR4 is higher for saxagliptin group at visit 2 and visit 3 in comparison to placebo group (p 

Fig. 5

Effect of saxagliptin on gene…

Fig. 5

Effect of saxagliptin on gene expression of CD34+ ( a ) and CD34−…

Fig. 5
Effect of saxagliptin on gene expression of CD34+ (a) and CD34− (b) cells. A low number of CD34+ cells obtained resulted in a reduced amount of mRNA affecting the number of samples suitable for analysis. The genes affected by saxagliptin treatment were: SOD1 (n = 11), GPX1 (n = 11), CASP3 (n = 10), IL6 (n = 13), IGF1 (n = 7). *p < 0.05; **p < 0.01. Results are relative to visit 1 (control)

Fig. 6

a Fat free mass (kg)…

Fig. 6

a Fat free mass (kg) show that the saxagliptin group had a sharp…
Fig. 6
a Fat free mass (kg) show that the saxagliptin group had a sharp decline after visit 1, but then a rise after visit 2, and the placebo group remained relatively stable (0.072). b  % Body Fat across visits 1–3. Saxagliptin patients had a decline from visit 1 to 3, whereas the control group increased at visit 3 (p = 0.079). c % total body water remained relatively stable for the placebo group, but increase in saxagliptin (p = 0.098)

Fig. 7

a Shows adiponectin values from…

Fig. 7

a Shows adiponectin values from visit 1 to visit 3. Saxagliptin showed a…
Fig. 7
a Shows adiponectin values from visit 1 to visit 3. Saxagliptin showed a decrease throughout the study, after visit 2, while the placebo group increased steadily from visit 1 to visit 3 (p = 0.01). b Shows serum creatinine values from visit 1 to visit 3. There is a steep decline from visit 1 to 2 in the placebo group, while levels remain stable in the saxagliptin group (p = 0.12)

Fig. 8

a Shows the arterial stiffness…

Fig. 8

a Shows the arterial stiffness parameter, augmentation index adjusted for a heart rate…
Fig. 8
a Shows the arterial stiffness parameter, augmentation index adjusted for a heart rate of 75. Saxagliptin subjects remained stable across visit 1 through 3, whereas the control subjects had an increase in arterial stiffness (p = 0.04). b Shows radial systolic blood pressure in saxagliptin, compared to Placebo, across the three visits. Saxagliptin blood pressure remains stable across visits 1 and 2, but then drops in visit 3. In the placebo group, blood pressure drops at visit 2, but then rises dramatically by visit 3 (p = 0.009)
All figures (8)
Similar articles
Cited by
References
    1. NIDDK. National diabetes statistics: 2007 and 2011 fact sheet. Bethesda, MD, USA. Department of Health and Human Services, NIH, 2008 and National Center for Chronic Disease Prevention and Health Promotion, 2011.
    1. American Diabetes Association Standards of medical care in diabetes-2014. Diabetes Care. 2014;37:S14–S80. doi: 10.2337/dc14-S014. - DOI - PubMed
    1. Sheetz MJ, King GL. Molecular understanding of hyperglycemia’s adverse effects for diabetic complications. JAMA. 2002;288:2579–2588. doi: 10.1001/jama.288.20.2579. - DOI - PubMed
    1. Rask-Madsen C, King GL. Mechanisms of disease: endothelial dysfunction in insulin resistance and diabetes. Nat Clin Pract Endocrinol Metab. 2007;3:46–56. doi: 10.1038/ncpendmet0366. - DOI - PubMed
    1. Afkarian M, Sachs MC, Kestenbaum B, Hirsch IB, Tuttle KR, Himmelfarb J, de Boer IH. Kidney disease and increased mortality risk in type 2 diabetes. JASN. 2013;24:302–308. doi: 10.1681/ASN.2012070718. - DOI - PMC - PubMed
Show all 60 references
Publication types
MeSH terms
Associated data
[x]
Cite
Copy Download .nbib
Format: AMA APA MLA NLM
Fig. 3
Fig. 3
Representative image (a and b) indicating the CD34+ CXCR4+ expression in MNCs by flow cytometry. CD34+ CXCR4+ expression is higher for the saxagliptin group at visit 3 than visit 1 (7.95 and 4.64%, respectively). c Double positivity for CD34 and CXCR4 along the visits for placebo and saxagliptin groups (p < 0.01)
Fig. 4
Fig. 4
CD31+ CXCR4+ expression in MNCs assessed by flow cytometry. Double positivity of CD31 and CXCR4 is higher for saxagliptin group at visit 2 and visit 3 in comparison to placebo group (p 

Fig. 5

Effect of saxagliptin on gene…

Fig. 5

Effect of saxagliptin on gene expression of CD34+ ( a ) and CD34−…

Fig. 5
Effect of saxagliptin on gene expression of CD34+ (a) and CD34− (b) cells. A low number of CD34+ cells obtained resulted in a reduced amount of mRNA affecting the number of samples suitable for analysis. The genes affected by saxagliptin treatment were: SOD1 (n = 11), GPX1 (n = 11), CASP3 (n = 10), IL6 (n = 13), IGF1 (n = 7). *p < 0.05; **p < 0.01. Results are relative to visit 1 (control)

Fig. 6

a Fat free mass (kg)…

Fig. 6

a Fat free mass (kg) show that the saxagliptin group had a sharp…
Fig. 6
a Fat free mass (kg) show that the saxagliptin group had a sharp decline after visit 1, but then a rise after visit 2, and the placebo group remained relatively stable (0.072). b  % Body Fat across visits 1–3. Saxagliptin patients had a decline from visit 1 to 3, whereas the control group increased at visit 3 (p = 0.079). c % total body water remained relatively stable for the placebo group, but increase in saxagliptin (p = 0.098)

Fig. 7

a Shows adiponectin values from…

Fig. 7

a Shows adiponectin values from visit 1 to visit 3. Saxagliptin showed a…
Fig. 7
a Shows adiponectin values from visit 1 to visit 3. Saxagliptin showed a decrease throughout the study, after visit 2, while the placebo group increased steadily from visit 1 to visit 3 (p = 0.01). b Shows serum creatinine values from visit 1 to visit 3. There is a steep decline from visit 1 to 2 in the placebo group, while levels remain stable in the saxagliptin group (p = 0.12)

Fig. 8

a Shows the arterial stiffness…

Fig. 8

a Shows the arterial stiffness parameter, augmentation index adjusted for a heart rate…
Fig. 8
a Shows the arterial stiffness parameter, augmentation index adjusted for a heart rate of 75. Saxagliptin subjects remained stable across visit 1 through 3, whereas the control subjects had an increase in arterial stiffness (p = 0.04). b Shows radial systolic blood pressure in saxagliptin, compared to Placebo, across the three visits. Saxagliptin blood pressure remains stable across visits 1 and 2, but then drops in visit 3. In the placebo group, blood pressure drops at visit 2, but then rises dramatically by visit 3 (p = 0.009)
All figures (8)
Fig. 5
Fig. 5
Effect of saxagliptin on gene expression of CD34+ (a) and CD34− (b) cells. A low number of CD34+ cells obtained resulted in a reduced amount of mRNA affecting the number of samples suitable for analysis. The genes affected by saxagliptin treatment were: SOD1 (n = 11), GPX1 (n = 11), CASP3 (n = 10), IL6 (n = 13), IGF1 (n = 7). *p < 0.05; **p < 0.01. Results are relative to visit 1 (control)
Fig. 6
Fig. 6
a Fat free mass (kg) show that the saxagliptin group had a sharp decline after visit 1, but then a rise after visit 2, and the placebo group remained relatively stable (0.072). b  % Body Fat across visits 1–3. Saxagliptin patients had a decline from visit 1 to 3, whereas the control group increased at visit 3 (p = 0.079). c % total body water remained relatively stable for the placebo group, but increase in saxagliptin (p = 0.098)
Fig. 7
Fig. 7
a Shows adiponectin values from visit 1 to visit 3. Saxagliptin showed a decrease throughout the study, after visit 2, while the placebo group increased steadily from visit 1 to visit 3 (p = 0.01). b Shows serum creatinine values from visit 1 to visit 3. There is a steep decline from visit 1 to 2 in the placebo group, while levels remain stable in the saxagliptin group (p = 0.12)
Fig. 8
Fig. 8
a Shows the arterial stiffness parameter, augmentation index adjusted for a heart rate of 75. Saxagliptin subjects remained stable across visit 1 through 3, whereas the control subjects had an increase in arterial stiffness (p = 0.04). b Shows radial systolic blood pressure in saxagliptin, compared to Placebo, across the three visits. Saxagliptin blood pressure remains stable across visits 1 and 2, but then drops in visit 3. In the placebo group, blood pressure drops at visit 2, but then rises dramatically by visit 3 (p = 0.009)

References

    1. NIDDK. National diabetes statistics: 2007 and 2011 fact sheet. Bethesda, MD, USA. Department of Health and Human Services, NIH, 2008 and National Center for Chronic Disease Prevention and Health Promotion, 2011.
    1. American Diabetes Association Standards of medical care in diabetes-2014. Diabetes Care. 2014;37:S14–S80. doi: 10.2337/dc14-S014.
    1. Sheetz MJ, King GL. Molecular understanding of hyperglycemia’s adverse effects for diabetic complications. JAMA. 2002;288:2579–2588. doi: 10.1001/jama.288.20.2579.
    1. Rask-Madsen C, King GL. Mechanisms of disease: endothelial dysfunction in insulin resistance and diabetes. Nat Clin Pract Endocrinol Metab. 2007;3:46–56. doi: 10.1038/ncpendmet0366.
    1. Afkarian M, Sachs MC, Kestenbaum B, Hirsch IB, Tuttle KR, Himmelfarb J, de Boer IH. Kidney disease and increased mortality risk in type 2 diabetes. JASN. 2013;24:302–308. doi: 10.1681/ASN.2012070718.
    1. Hill JM, Zalos G, Halcox JPJ, Schenke WH, Waclawiw MA, Quyyumi AA, Finkel T. Circulating endothelial progenitor cells, vascular function, and cardiovascular risk. N Engl J Med. 2003;348:593–600. doi: 10.1056/NEJMoa022287.
    1. Krenning G, Dankers PYW, Drouven JW, Waanders F, Franssen CFM, van Luyn MJA, Harmsen MC, Popa ER. Endothelial progenitor cell dysfunction in patients with progressive chronic kidney disease. Am J Physiol Renal Physiol. 2009;296:F1314–F1322. doi: 10.1152/ajprenal.90755.2008.
    1. Tepper OM, Galiano RD, Capla JM, Kalka C, Gagne PJ, Jacobowitz GR, Levine JP, Gurtner GC. Human endothelial progenitor cells from type II diabetics exhibit impaired proliferation, adhesion, and incorporation into vascular structures. Circulation. 2002;106:2781–2786. doi: 10.1161/01.CIR.0000039526.42991.93.
    1. Werner N, Wassmann S, Ahlers P, Kosiol S, Nickenig G. Circulating CD31+/annexin V+ apoptotic microparticles correlate with coronary endothelial function in patients with coronary artery disease. Arterioscler Thromb Vasc Biol. 2006;26:112–116. doi: 10.1161/01.ATV.0000191634.13057.15.
    1. Kundu N, Domingues CC, Chou C, Ahmadi N, Houston S, Jerry DJ, Sen S. Use of p53-silenced endothelial progenitor cells to treat ischemia in diabetic peripheral vascular disease. J Am Heart Assoc. 2017;6(4):e005146. doi: 10.1161/JAHA.116.005146.
    1. Caballero S, Sengupta N, Afzal A, Chang KH, Li Calzi S, Guberski DL, Kern TS, Grant MB. Ischemic vascular damage can be repaired by healthy, but not diabetic, endothelial progenitor cells. Diabetes. 2007;56:960–967. doi: 10.2337/db06-1254.
    1. Fadini GP, Boscaro E, Albiero M, Menegazzo L, Frison V, de Kreutzenberg S, Agostini C, Tiengo A, Avogaro A. The oral dipeptidyl peptidase-4 inhibitor sitagliptin increases circulating endothelial progenitor cells in patients with type 2 diabetes: possible role of stromal-derived factor-1alpha. Diabetes Care. 2010;33:1607–1609. doi: 10.2337/dc10-0187.
    1. Huang CY, Shih CM, Tsao NW, Lin YW, Huang PH, Wu SC, Lee AW, Kao YT, Chang NC, Nakagami H, Morishita R, Ou KL, Hou WC, Lin CY, Shyu KG, Lin FY. Dipeptidyl peptidase-4 inhibitor improves neovascularization by increasing circulating endothelial progenitor cells. Br J Pharmacol. 2012;167:1506–1519. doi: 10.1111/j.1476-5381.2012.02102.x.
    1. Losordo DW, Schatz RA, White CJ, Udelson JE, Veereshwarayya V, Durgin M, Poh KK, Weinstein R, Kearney M, Chaudhry M, Burg A, Eaton L, Heyd L, Thorne T, Shturman L, Hoffmeister P, Story K, Zak V, Dowling D, Traverse JH, Olson RE, Flanagan J, Sodano D, Murayama T, Kawamoto A, Kusano KF, Wollins J, Welt F, Shah P, Soukas P, Asahara T, Henry TD. Intramyocardial transplantation of autologous CD34+ stem cells for intractable angina: a phase I/IIa double-blind, randomized controlled trial. Circulation. 2007;115:3165–3172. doi: 10.1161/CIRCULATIONAHA.106.687376.
    1. Marrotte EJ, Chen D-D, Hakim JS, Chen AF. Manganese superoxide dismutase expression in endothelial progenitor cells accelerates wound healing in diabetic mice. J Clin Invest. 2010;120:4207–4219. doi: 10.1172/JCI36858.
    1. Dicker D. DPP-4 inhibitors: impact on glycemic control and cardiovascular risk factors. Diabetes Care. 2011;34(2):S276–S278. doi: 10.2337/dc11-s229.
    1. Li F, Chen J, Leng F, Lu Z, Ling Y. Effect of saxagliptin on circulating endothelial progenitor cells and endothelial function in newly diagnosed type 2 diabetic patients. Exp Clin Endocrinol Diabetes. 2017;125:400–407. doi: 10.1055/s-0042-124421.
    1. American Diabetes Association Pharmacologic approaches to glycemic treatment. Diabetes Care. 2017;40(1):S64–S74. doi: 10.2337/dc17-S011.
    1. Kirpichnikov D, McFarlane SI, Sowers JR. Metformin: an update. Ann Intern Med. 2002;137(1):25–33. doi: 10.7326/0003-4819-137-1-200207020-00009.
    1. UK Prospective Diabetes Study (UKPDS) Group Effect of intensive blood-glucose control with metformin on complications in overweight patients with type 2 diabetes (UKPDS 34. Lancet. 1998;352:854–865. doi: 10.1016/S0140-6736(98)07037-8.
    1. Fung CS, Wan EY, Wong CK, Jiao F, Chan AK. Effect of metformin monotherapy on cardiovascular diseases and mortality: a retrospective cohort study on Chinese type 2 diabetes mellitus patients. Cardiovasc Diabetol. 2015;14:137. doi: 10.1186/s12933-015-0304-2.
    1. Ahmed FW, Rider R, Glanville M, Narayanan K, Razvi S, Weaver JU. Metformin improves circulating endothelial cells and endothelial progenitor cells in type 1 diabetes: MERIT study. Cardiovasc Diabetol. 2016;15:116. doi: 10.1186/s12933-016-0413-6.
    1. Herrera C, Morimoto C, Blanco J, Mallol J, Arenzana F, Lluis C, Franco R. Comodulation of CXCR4 and CD26 in human lymphocytes. J Biol Chem. 2001;276:19532–19539. doi: 10.1074/jbc.M004586200.
    1. Zheng H, Fu G, Dai T, Huang H. Migration of endothelial progenitor cells mediated by stromal cell-derived factor-1alpha/CXCR4 via PI3K/Akt/eNOS signal transduction pathway. J Cardiovasc Pharmacol. 2007;50:274–280. doi: 10.1097/FJC.0b013e318093ec8f.
    1. Yin Y, Zhao X, Fang Y, Yu S, Zhao J, Song M, Huang L. SDF-1alpha involved in mobilization and recruitment of endothelial progenitor cells after arterial injury in mice. Cardiovasc Pathol. 2010;19:218–227. doi: 10.1016/j.carpath.2009.04.002.
    1. Moore MA, Hattori K, Heissig B, Shieh JH, Dias S, Crystal RG, Rafii S. Mobilization of endothelial and hematopoietic stem and progenitor cells by adenovector-mediated elevation of serum levels of SDF-1, VEGF, and angiopoietin-1. Ann NY Acad Sci. 2001;938:36–45. doi: 10.1111/j.1749-6632.2001.tb03572.x.
    1. Werner N, Kosiol S, Schiegl T, Ahlers P, Walenta K, Link A, Böhm M, Nickenig G. Circulating endothelial progenitor cells and cardiovascular outcomes. N Engl J Med. 2005;353(10):999–1007. doi: 10.1056/NEJMoa043814.
    1. Fadini GP, Pucci L, Vanacore R, Baesso I, Penno G, Balbarini A, Di Stefano R, Miccoli R, de Kreutzenberg S, Coracina A, Tiengo A, Agostini C, Del Prato S, Avogaro A. Glucose tolerance is negatively associated with circulating progenitor cells levels. Diabetologia. 2007;50:2156–2163. doi: 10.1007/s00125-007-0732-y.
    1. Fadini GP, Albiero M, Vigili de Kreutzenberg S, Boscaro E, Cappellari R, Marescotti M, Poncina N, Agostini C, Avogaro A. Diabetes impairs stem cell and proangiogenic cell mobilization in humans. Diabetes Care. 2013;36:943–949. doi: 10.2337/dc12-1084.
    1. Rigato M, Bittante C, Albiero M, Avogaro A, Fadini GP. Circulating progenitor cell count predicts microvascular outcomes in type 2 diabetic patients. J Clin Endocrinol Metab. 2015;100:2666–2672. doi: 10.1210/jc.2015-1687.
    1. Sen S, Witkowski S, Lagoy A, Islam AM. A 6-week home exercise program improves endothelial function and CD34+ circulating progenitor cells in patients with pre-diabetes. J Endocrinol Metab. 2015;5:163–171. doi: 10.14740/jem273w.
    1. Urbich C, Dimmeler S. Endothelial progenitor cells functional characterization. Trends Cardiovasc Med. 2004;14:318–322. doi: 10.1016/j.tcm.2004.10.001.
    1. Chen LL, Liao YF, Zeng TS, Yu F, Li HQ, Feng Y. Effect of metformin plus gliclazide compared with metformin alone on circulating endothelial progenitor cell in type 2 diabetic patients. Endocrine. 2010;38:266–275. doi: 10.1007/s12020-010-9383-8.
    1. Bakhashab, et al. Metformin improves the angiogenic potential of human CD34+ cells co-incident with downregulating CXCL10 and TIMP1 gene expression and increasing VEGFA under hyperglycemia and hypoxia within a therapeutic window for myocardial infarction. Cardiovasc Diabetol. 2016;15:27. doi: 10.1186/s12933-016-0344-2.
    1. Bakhashab S, et al. Proangiogenic effect of metformin in endothelial cells is via upregulation of VEGFR1/2 and their signaling under hyperglycemia–hypoxia. Int J Mol Sci. 2018
    1. Landers-Ramos RQ, Sapp RM, Jenkins NT, Murphy AE, Cancre L, Chin ER, Spangenburg EE, Hagberg JM. Chronic endurance exercise affects paracrine action of CD31+ and CD34+ cells on endothelial tube formation. Am J Physio Heart Circ Physiol. 2015;309:H407–H420. doi: 10.1152/ajpheart.00123.2015.
    1. Hu X, Zheng H, Yan T, Pan S, Fang J, Jiang R, Ma S. Physical exercise induces expression of CD31 and facilitates neural function recovery in rats with focal cerebral infarction. Neurol Res. 2010;32:397–402. doi: 10.1179/016164110X12670144526309.
    1. Jarajapu YP, Caballero S, Verma A, Nakagawa T, Lo MC, Li Q, Grant MB. Blockade of NADPH oxidase restores vasoreparative function in diabetic CD34+ cells. Invest Ophthalmol Vis Sci. 2011;52:5093–5104. doi: 10.1167/iovs.10-70911.
    1. Sen S, Domingues CC, Rouphael C, Chou C, Kim C, Yadava N. Genetic modification of human mesenchymal stem cells helps to reduce adiposity and improve glucose tolerance in an obese diabetic mouse model. Stem Cell Res Ther. 2015;6:242. doi: 10.1186/s13287-015-0224-9.
    1. Dhillon S, Weber J. Saxagliptin. Drugs. 2009;69:2103–2114. doi: 10.2165/11201170-000000000-00000.
    1. de Boer SA, Heerspink HJL, Juárez Orozco LE, van Roon AM, Kamphuisen PW, Smit AJ, Slart RHJA, Lefrandt JD, Mulder DJ. Effect of linagliptin on pulse wave velocity in early type 2 diabetes: a randomized, double-blind, controlled 26-week trial (RELEASE) Diabetes Obes Metab. 2017;19:1147–1154. doi: 10.1111/dom.12925.
    1. Goldsmith F, Keenan MJ, Raggio AM, Ye X, Hao Z, Durham H, Geaghan J, Jia W, Martin RJ, Ye J. Induction of energy expenditure by sitagliptin is dependent on GLP-1 receptor. PLoS ONE. 2015;10:e0126177. doi: 10.1371/journal.pone.0126177.
    1. Pannacciulli N, Bunt JC, Koska J, Bogardus C, Krakoff J. Higher fasting plasma concentrations of glucagon-like peptide 1 are associated with higher resting energy expenditure and fat oxidation rates in humans. Am J Clin Nutr. 2006;84:556–560. doi: 10.1093/ajcn/84.3.556.
    1. Scirica BM, Braunwald E, Raz I, Cavender MA, Morrow DA, Jarolim P, Udell JA, Mosenzon O, Im K, Umez-Eronini AA, Pollack PS, Hirshberg B, Frederich R, Lewis BS, McGuire DK, Davidson J, Steg PG, Bhatt DL. SAVOR-TIMI 53 Steering committee and investigators. heart failure, saxagliptin, and diabetes mellitus: observations from the SAVOR-TIMI 53 randomized trial. Circulation. 2014;130:1579–1588. doi: 10.1161/CIRCULATIONAHA.114.010389.
    1. Duvnjak L, Slaslov K. Dipeptidyl peptidate-4 inhibitors improve arterial stiffness, blood pressure, lipid profile and inflammation parameters in patients with type 2 diabetes mellitus. Diabetol Metab Syndr. 2016;8:26. doi: 10.1186/s13098-016-0144-6.
    1. Liu X, Men P, Wang Y, Zhai S, Liu G. Impact of dipeptidyl peptidase-4 inhibitors on serum adiponectin: a meta-analysis. Lipids Health Dis. 2016;15:204. doi: 10.1186/s12944-016-0372-7.
    1. Yu SM, Bonventre JV. Acute kidney injury and progression of diabetic kidney disease. Adv Chronic Kidney Dis. 2018;25(2):166–180. doi: 10.1053/j.ackd.2017.12.005.
    1. Daehn IS. glomerular endothelial cells stress and cross-talk with podocytes in the development of diabetic kidney disease. Front Med (Lausanne). 2018;5:76. doi: 10.3389/fmed.2018.00076.
    1. Cecelja M, Chowienczyk P. Role of arterial stiffness in cardiovascular disease. J R Seoc Med Cardiovasc Dis. 2012;1:11.
    1. Quinn U, Tomlinson LA, Cockcroft JR. Arterial stiffness. J R Soc Med Cardiovas Dis. 2012;1:18.
    1. Stoner L, Young JM, Fryer S. Assessments of arterial stiffness and endothelial function using pulse wave analysis. Int J Vasc Med. 2012;2012:903107.
    1. Khiyami AM, Dore FJ, Mammadova A, Amdur R, Sen S. The correlation of arterial stiffness with biophysical parameters and blood biochemistry. Metab Syndr Relat Disord. 2017;15:178–182. doi: 10.1089/met.2016.0136.
    1. Henry RM, Kostense PJ, Spijkerman AM, Dekker JM, Nijpels G, Heine RJ, Kamp O, Westerhof N, Bouter LM, Stehouwer CD, Hoorn Study Arterial stiffness increases with deteriorating glucose tolerance status. Circulation. 2003;107:2089–2095. doi: 10.1161/01.CIR.0000065222.34933.FC.
    1. Sun F, Wu S, Wang J, Guo S, Chai S, Yang Z, Li L, Zhang Y, Ji L, Zhan S. Effect of glucagon-like peptide-1 receptor agonists on lipid proles among type 2 diabetes: a systematic review and network meta-analysis. Clin Ther. 2015;37:225–241. doi: 10.1016/j.clinthera.2014.11.008.
    1. Richter G, Feddersen O, Wagner U, Barth P, Göke R, Göke B. GLP-1 stimulates secretion of macromolecules from airways and relaxes pulmonary artery. Am J Physiol. 1993;265:L374–L381.
    1. Mannucci E, Ognibene A, Cremasco F, Bardini G, Mencucci A, Pierazzuoli E, Ciani S, Messeri G, Rotella CM. Effect of metformin on glucagon-like peptide 1 (GLP-1) and leptin levels in obese nondiabetic subjects. Diabetes Care. 2001;24:489–494. doi: 10.2337/diacare.24.3.489.
    1. Wu T, Thazhath SS, Bound MJ, Jones KL, Horowitz M, Rayner CK. Mechanism of increase in plasma intact GLP-1 by metformin in type 2 diabetes: stimulation of GLP-1 secretion or reduction in plasma DPP-4 activity? Diabetes Res Clin Pract. 2014;106:e3–e6. doi: 10.1016/j.diabres.2014.08.004.
    1. Marso SP, Daniels GH, Brown-Frandsen K, Kristensen P, Mann JF, Nauck MA, Nissen SE, Pocock S, Poulter NR, Ravn LS, Steinberg WM, Stockner M, Zinman B, Bergenstal RM, Buse JB. LEADER steering committee, LEADER trial investigators. liraglutide and cardiovascular outcomes in type 2 diabetes. N Engl J Med. 2016;375:311–322. doi: 10.1056/NEJMoa1603827.
    1. Zinman B, Wanner C, Lachin JM, Fitchett D, Bluhmki E, Hantel S, Mattheus M, Devins T, Johansen OE, Woerle HJ, Broedl UC, Inzucchi SE. EMPA-REG OUTCOME investigators. empagliflozin, cardiovascular outcomes, and mortality in type 2 diabetes. N Engl J Med. 2015;373:2117–2128. doi: 10.1056/NEJMoa1504720.
    1. Marso SP, Bain SC, Consoli A, Eliaschewitz FG, Jódar E, Leiter LA, Lingvay I, Rosenstock J, Seufert J, Warren ML, Woo V, Hansen O, Holst AG, Pettersson J, Vilsbøll T. SUSTAIN-6 investigators. semaglutide and cardiovascular outcomes in patients with type 2 diabetes. N Engl J Med. 2016;375:1834–1844. doi: 10.1056/NEJMoa1607141.

Source: PubMed

3
Abonnieren