• No results found

University of Groningen Skin autofluorescence in the general population: associations and prediction van Waateringe, Robert Paul

N/A
N/A
Protected

Academic year: 2021

Share "University of Groningen Skin autofluorescence in the general population: associations and prediction van Waateringe, Robert Paul"

Copied!
11
0
0

Bezig met laden.... (Bekijk nu de volledige tekst)

Hele tekst

(1)

Skin autofluorescence in the general population: associations and prediction

van Waateringe, Robert Paul

IMPORTANT NOTE: You are advised to consult the publisher's version (publisher's PDF) if you wish to cite from it. Please check the document version below.

Document Version

Publisher's PDF, also known as Version of record

Publication date: 2019

Link to publication in University of Groningen/UMCG research database

Citation for published version (APA):

van Waateringe, R. P. (2019). Skin autofluorescence in the general population: associations and prediction. Rijksuniversiteit Groningen.

Copyright

Other than for strictly personal use, it is not permitted to download or to forward/distribute the text or part of it without the consent of the author(s) and/or copyright holder(s), unless the work is under an open content license (like Creative Commons).

Take-down policy

If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

Downloaded from the University of Groningen/UMCG research database (Pure): http://www.rug.nl/research/portal. For technical reasons the number of authors shown on this cover page is limited to 10 maximum.

(2)

Introduction and aim of the thesis

1

(3)

Introduction

Advanced glycation end products

Advanced glycation end products (AGEs) are a group of very heterogeneous compounds formed by non-enzymatic glycation of proteins, lipids and nucleic acids under conditions of glycaemic, oxidative or carbonyl stress (1). This mechanism of AGE formation is also called the Maillard or browning reaction (2). In short, the interaction between the carbonyl groups of reducing sugars and free amino groups of proteins leads to the formation of a Schiff base. Rearrangement of the Schiff base results in Amadori products. A well-known example of an Amadadori product is glycated haemoglobin which is commonly used in clinical practice to asses long-term diabetes control. The slow process of oxidation of the Amadori products leads to reactive carbonyl compounds and subsequently to the formation of AGEs (3, 4). Reactive oxygen species (ROS) are also involved in the lipid peroxidation pathway, altering lipids in reactive carbonyl compounds. This reaction results in the formation of advanced lipoxidation end products (ALEs) (5, 6).

Next to endogenous formation of AGEs, there are also exogenous sources of AGEs such as tobacco smoke and certain foods. Tobacco smoke itself contains high levels reactive glycation products (7). In addition, tobacco smoke causes oxidative stress, which in turn can enhance the formation of AGEs. High levels of AGEs were observed in lenses and blood vessels of tobacco smokers (8). A second exogenous source of AGEs is the modern diet. Intake of food containing high levels of AGEs may lead to an increase of AGEs. Uncooked, animal derived food, rich of protein and fad, generally contain high levels of AGEs (9). However, more important is the way how food is processed. Especially the preparation and prolonged heating of food plays an key role in the generation of AGEs, including grilling, roasting and boiling (9, 10). It has been estimated that approximately 10% of AGEs from food are absorbed from the gastro-intestinal tract into the circulation (11).

AGEs are known to accumulate in the body during ageing (12). The lifetime of AGEs depends on the specific protein on which the AGEs are cross-linked. It has been reported that AGEs reflect long-term (~ 15 years) accumulation of glycaemic and oxidative stress, and might therefore be considered as a marker of ‘metabolic memory’ (13). In people with diabetes, both the formation and accumulation of AGEs is increased as a result of chronic hyperglycaemia (14, 15). In individuals with impaired renal function, the formation of AGEs may be a result of oxidative or carbonyl stress (16, 17), while decreased renal clearance of serum AGEs also contributes to increased accumulation of AGEs (16, 18).

(4)

Introduction and aim of the thesis 13 In diabetes, accumulation of AGEs is associated with both micro- and macrovascular complications (19). Accumulation of AGEs in the kidney may cause thickening of the capillary basement membrane and sclerosis of the glomeruli (20). Increased accumulation of AGEs in the axoplasm of myelinated and unmyelinated neurons and Schwann cells supports its involvement in the development and progression of diabetic neuropathy (21). Accumulation of AGEs induce the production of vascular endothelial growth factor (VEGF), monocyte chemotactic protein-1 (MCP-1), and transforming growth factor-b (TGF-b), which are key cytokines related to the development of diabetic retinopathy (22, 23). Cross-linking of proteins within the vascular wall will reduce vascular elasticity and increase arterial stiffness (24, 25). In contrast, treatment with AGEs breakers for 1-3 weeks has been shown to effectively reverse arterial stiffness in rats with streptozotocin-induced diabetes (26). AGEs accumulation have also been found in carotid atherosclerotic plaques which underlines that accumulation of AGEs is involved in the development of atherosclerosis (27).

Furthermore, binding of AGEs to its receptor (RAGE) might play a role in the development of diabetes cardiovascular complications. Activation of RAGE causes intracellular signaling which leads to a cascade of pro-inflammatory cytokines including nuclear factor-kappa B, interleukin 6 and tumor necrosis factor alpha (28, 29). Furthermore, it stimulates smooth muscle cell proliferation, increases oxidative stress and causes endothelial dysfunction (28, 30).

Non-invasive measurement of skin AGEs

In the past, several investigators have tried to assess AGE levels in the blood and in tissues obtained by (skin) biopsies. However, this method is invasive, time-consuming and associated with high costs. Measurement of AGEs is nowadays performed using a device that is called the AGE Reader (Diagnoptics). This device is developed in Groningen, The Netherlands. It estimated skin AGE content non-invasively based on its fluorescent characteristics of many AGE compounds (31). Therefore, measuring skin AGEs using its fluorescent capacities is called skin autofluorescence (SAF). The measurement of SAF is easily performed in the office on the volar side of the forearm, 10cm below the elbow. The AGE Reader illuminates a skin surface of approximately 4 cm², guarded against surrounding light, with an excitation light source with a wavelength between 300 and 420nm (peak intensity at ~ 370nm). Emission light and reflected excitation light from the skin are measured with an internal spectrometer in the range 300 to 600nm. SAF is calculated by dividing the average emitted light intensity per nanometer in the range of 420-600 nm by the average excitated light intensity per nanometer in the range

(5)

420 nm and multiplied by 100. SAF levels are expressed in arbitrary units and increase or decrease per arbitrary unit (AU). It has been shown that SAF levels correlate well with tissue AGEs measured in skin biopsies from subjects with and without diabetes (31, 32). Intra-individual variance among diabetic and control subjects was estimated at around 5% when repeated SAF measurement were taken over a single day (31). SAF levels positively correlate with ageing and are higher among subjects with diabetes and impaired renal function (33).

Skin autofluorescence and previous studies

Over the past 10 years, many studies have been performed using SAF as a biomarker for several clinical conditions, including type 2 diabetes, renal failure and several cardiovascular diseases. Among subjects with type 2 diabetes, higher SAF levels were associated with both micro-and macrovascular complications, and with the severity of diabetes-related complications (34, 35). Furthermore, SAF have been demonstrated to be a strong predictor for cardiac mortality among subjects with type 2 diabetes (36). Addition of SAF measurement to the United Kingdom Prospective Diabetes Study (UKPDS) risk score resulted in re-classification of around 25% of the study population from the low-risk to the high-low-risk group (37). The clinical value of SAF has also been studied in subjects with renal failure. A Japanese study reported that high SAF levels are associated with the progression of chronic kidney disease (38). Other studies have shown that SAF predicts both overall and cardiovascular mortality among (pre)hemodialysis patients (39-41). Furthermore, SAF independently predicts chronic graft dysfunction in renal transplant recipients (42). Only a small number of studies have investigated SAF in subjects without diabetes or impaired renal function. It has been shown that SAF levels are elevated in subjects with (sub-clinical) atherosclerosis and acute ST elevated myocardial infarction, whereas a SAF above the median was of predictive value for future cardiovascular events (43, 44). SAF levels are elevated in subjects with stable coronary artery heart disease and associated with carotid artery intima media thickness (45, 46). Furthermore, SAF levels are higher in individuals with carotid artery stenosis and peripheral artery disease and associated with 5-years risk of mortality and cardiovascular events (47, 48).

Outline of thesis

Most previous research on SAF has focused on populations with either type 2 diabetes, peripheral artery disease, or (end-stage) renal failure. In this thesis, we aim to describe the association between skin autofluorescence and cardiometabolic risk factors in the general population. For these studies, we use data collected in people participating in

(6)

Introduction and aim of the thesis 15 the Dutch LifeLines Cohort Study. In addition, we evaluate whether skin autofluorescence is able to predict incident type 2 diabetes, cardiovascular disease and overall mortality within the general population.

The formation and accumulation of AGEs is a complex pathway including either endogenous as well as exogenous pathways. Moreover, when measuring skin autofluorescence, many factors have to be taken into account. Therefore, we describe both clinical and lifestyle determinants of skin autofluorescence among subjects with and without type 2 diabetes in chapter 2. Tobacco smoking is an exogenous source of AGEs accumulation and higher SAF levels are shown in individuals who smoke. However, the effect of secondhand smoking and smoking cessation on AGEs accumulation needs to be determined. The results of this study are presented in chapter 3. The metabolic syndrome is a combination of several cardiometabolic abnormalities associated with a higher risk of both cardiovascular disease and type 2 diabetes. Explorative associations between skin autofluorescence and the individual metabolic syndrome components are shown in chapter 4. In chapter 5 we evaluate the predictive value of skin autofluorescence as a biomarker for incident type 2 diabetes, cardiovascular disease and overall mortality. Next, we assess the association between thyroid hormone levels and incident type 2 diabetes, cardiovascular disease and overall mortality in the general population and whether the association is independent of SAF. The results are shown in chapter 6. Inflammation plays an essential role in the development of insulin resistance, type 2 diabetes and atherosclerosis. Preliminary data from our group showed that SAF levels increase during pregnancy, and do not return to levels found before pregnancy (Groen et al. unpublished data). This may be caused by increased oxidative and glycemic stress during pregnancy. In chapter 7 we examined the relationship between SAF and the number of pregnancies. In chapter 8 we assessed the reproducibility and standardization of several inflammatory biomarkers, including hsCRP, hsIL6 and TNFα, taking into account the influence of assay variability, the reproducibility of a specific measurement between laboratories as well as the influence of sample storage and storage time. In chapter 9 we summarize and discuss the separate chapters and reflect on possible future research on SAF in the general population, and its current application in screening for incident type 2 diabetes, cardiovascular disease and mortality.

(7)

References

1. Singh R, Barden A, Mori T, Beilin L. Advanced glycation end-products: a review. Diabetologia. 2001 Feb;44(2):129-46.

2. Monnier VM. Nonenzymatic glycosylation, the Maillard reaction and the aging process. J Gerontol. 1990 Jul;45(4):B105-11.

3. Goldin A, Beckman JA, Schmidt AM, Creager MA. Advanced glycation end products: sparking the development of diabetic vascular injury. Circulation. 2006 Aug 8;114(6):597-605. 4. Baynes JW, Thorpe SR. Role of oxidative stress in diabetic complications: a new perspective

on an old paradigm. Diabetes. 1999 Jan;48(1):1-9.

5. Negre-Salvayre A, Coatrieux C, Ingueneau C, Salvayre R. Advanced lipid peroxidation end products in oxidative damage to proteins. Potential role in diseases and therapeutic prospects for the inhibitors. Br J Pharmacol. 2008 Jan;153(1):6-20.

6. Vistoli G, De Maddis D, Cipak A, Zarkovic N, Carini M, Aldini G. Advanced glycoxidation and lipoxidation end products (AGEs and ALEs): an overview of their mechanisms of formation. Free Radic Res. 2013 Aug;47 Suppl 1:3-27.

7. Cerami C, Founds H, Nicholl I, Mitsuhashi T, Giordano D, Vanpatten S, et al. Tobacco smoke is a source of toxic reactive glycation products. Proc Natl Acad Sci U S A. 1997 Dec 9;94(25):13915-20.

8. Nicholl ID, Stitt AW, Moore JE, Ritchie AJ, Archer DB, Bucala R. Increased levels of advanced glycation endproducts in the lenses and blood vessels of cigarette smokers. Mol Med. 1998 Sep;4(9):594-601.

9. Uribarri J, Woodruff S, Goodman S, Cai W, Chen X, Pyzik R, et al. Advanced glycation end products in foods and a practical guide to their reduction in the diet. J Am Diet Assoc. 2010 Jun;110(6):911,16.e12.

10. Goldberg T, Cai W, Peppa M, Dardaine V, Baliga BS, Uribarri J, et al. Advanced glycoxidation end products in commonly consumed foods. J Am Diet Assoc. 2004 Aug;104(8):1287-91. 11. Koschinsky T, He CJ, Mitsuhashi T, Bucala R, Liu C, Buenting C, et al. Orally absorbed reactive

glycation products (glycotoxins): an environmental risk factor in diabetic nephropathy. Proc Natl Acad Sci U S A. 1997 Jun 10;94(12):6474-9.

12. Semba RD, Nicklett EJ, Ferrucci L. Does accumulation of advanced glycation end products contribute to the aging phenotype? J Gerontol A Biol Sci Med Sci. 2010 Sep;65(9):963-75. 13. Verzijl N, DeGroot J, Thorpe SR, Bank RA, Shaw JN, Lyons TJ, et al. Effect of collagen turnover on

the accumulation of advanced glycation end products. J Biol Chem. 2000 Dec 15;275(50):39027-31.

14. Baynes JW, Thorpe SR. Glycoxidation and lipoxidation in atherogenesis. Free Radic Biol Med. 2000 Jun 15;28(12):1708-16.

15. Wolffenbuttel BH, Giordano D, Founds HW, Bucala R. Long-term assessment of glucose control by haemoglobin-AGE measurement. Lancet. 1996 Feb 24;347(9000):513-5.

(8)

Introduction and aim of the thesis 17

16. Miyata T, Wada Y, Cai Z, Iida Y, Horie K, Yasuda Y, et al. Implication of an increased oxidative stress in the formation of advanced glycation end products in patients with end-stage renal failure. Kidney Int. 1997 Apr;51(4):1170-81.

17. Lapolla A, Reitano R, Seraglia R, Sartore G, Ragazzi E, Traldi P. Evaluation of advanced glycation end products and carbonyl compounds in patients with different conditions of oxidative stress. Mol Nutr Food Res. 2005 Jul;49(7):685-90.

18. Miyata T, Ueda Y, Yoshida A, Sugiyama S, Iida Y, Jadoul M, et al. Clearance of pentosidine, an advanced glycation end product, by different modalities of renal replacement therapy. Kidney Int. 1997 Mar;51(3):880-7.

19. Goh SY, Cooper ME. Clinical review: The role of advanced glycation end products in progression and complications of diabetes. J Clin Endocrinol Metab. 2008 Apr;93(4):1143-52.

20. Monnier VM, Sell DR, Nagaraj RH, Miyata S, Grandhee S, Odetti P, et al. Maillard reaction-mediated molecular damage to extracellular matrix and other tissue proteins in diabetes, aging, and uremia. Diabetes. 1992 Oct;41 Suppl 2:36-41.

21. Jack M, Wright D. Role of advanced glycation endproducts and glyoxalase I in diabetic peripheral sensory neuropathy. Transl Res. 2012 May;159(5):355-65.

22. Yokoi M, Yamagishi SI, Takeuchi M, Ohgami K, Okamoto T, Saito W, et al. Elevations of AGE and vascular endothelial growth factor with decreased total antioxidant status in the vitreous fluid of diabetic patients with retinopathy. Br J Ophthalmol. 2005 Jun;89(6):673-5.

23. Fokkens BT, Mulder DJ, Schalkwijk CG, Scheijen JL, Smit AJ, Los LI. Vitreous advanced glycation endproducts and alpha-dicarbonyls in retinal detachment patients with type 2 diabetes mellitus and non-diabetic controls. PLoS One. 2017 Mar 6;12(3):e0173379.

24. Aronson D. Cross-linking of glycated collagen in the pathogenesis of arterial and myocardial stiffening of aging and diabetes. J Hypertens. 2003 Jan;21(1):3-12.

25. Willemsen S, Hartog JW, Hummel YM, van Ruijven MH, van der Horst IC, van Veldhuisen DJ, et al. Tissue advanced glycation end products are associated with diastolic function and aerobic exercise capacity in diabetic heart failure patients. Eur J Heart Fail. 2011 Jan;13(1):76-82. 26. Wolffenbuttel BH, Boulanger CM, Crijns FR, Huijberts MS, Poitevin P, Swennen GN, et al.

Breakers of advanced glycation end products restore large artery properties in experimental diabetes. Proc Natl Acad Sci U S A. 1998 Apr 14;95(8):4630-4.

27. Hanssen NM, Wouters K, Huijberts MS, Gijbels MJ, Sluimer JC, Scheijen JL, et al. Higher levels of advanced glycation endproducts in human carotid atherosclerotic plaques are associated with a rupture-prone phenotype. Eur Heart J. 2014 May;35(17):1137-46.

28. Bierhaus A, Humpert PM, Morcos M, Wendt T, Chavakis T, Arnold B, et al. Understanding RAGE, the receptor for advanced glycation end products. J Mol Med (Berl). 2005 Nov;83(11):876-86. 29. Ramasamy R, Yan SF, Schmidt AM. Receptor for AGE (RAGE): signaling mechanisms in the

pathogenesis of diabetes and its complications. Ann N Y Acad Sci. 2011 Dec;1243:88-102. 30. Basta G, Lazzerini G, Massaro M, Simoncini T, Tanganelli P, Fu C, et al. Advanced glycation end

products activate endothelium through signal-transduction receptor RAGE: a mechanism for amplification of inflammatory responses. Circulation. 2002 Feb 19;105(7):816-22.

(9)

31. Meerwaldt R, Graaff R, Oomen PH, Links TP, Jager JJ, Alderson NL, et al. Simple non-invasive assessment of advanced glycation endproduct accumulation. Diabetologia. 2004 Jul;47(7):1324-30.

32. Meerwaldt R, Links T, Graaff R, Thorpe SR, Baynes JW, Hartog J, et al. Simple noninvasive measurement of skin autofluorescence. Ann N Y Acad Sci. 2005 Jun;1043:290-8.

33. Koetsier M, Lutgers HL, de Jonge C, Links TP, Smit AJ, Graaff R. Reference values of skin autofluorescence. Diabetes Technol Ther. 2010 May;12(5):399-403.

34. Lutgers HL, Graaff R, Links TP, Ubink-Veltmaat LJ, Bilo HJ, Gans RO, et al. Skin autofluorescence as a noninvasive marker of vascular damage in patients with type 2 diabetes. Diabetes Care. 2006 Dec;29(12):2654-9.

35. Noordzij MJ, Mulder DJ, Oomen PH, Brouwer T, Jager J, Castro Cabezas M, et al. Skin autofluorescence and risk of micro- and macrovascular complications in patients with Type 2 diabetes mellitus-a multi-centre study. Diabet Med. 2012 Dec;29(12):1556-61.

36. Meerwaldt R, Lutgers HL, Links TP, Graaff R, Baynes JW, Gans RO, et al. Skin autofluorescence is a strong predictor of cardiac mortality in diabetes. Diabetes Care. 2007 Jan;30(1):107-12. 37. Lutgers HL, Gerrits EG, Graaff R, Links TP, Sluiter WJ, Gans RO, et al. Skin autofluorescence

provides additional information to the UK Prospective Diabetes Study (UKPDS) risk score for the estimation of cardiovascular prognosis in type 2 diabetes mellitus. Diabetologia. 2009 May;52(5):789-97.

38. Tanaka K, Nakayama M, Kanno M, Kimura H, Watanabe K, Tani Y, et al. Skin autofluorescence is associated with the progression of chronic kidney disease: a prospective observational study. PLoS One. 2013 Dec 12;8(12):e83799.

39. Meerwaldt R, Hartog JW, Graaff R, Huisman RJ, Links TP, den Hollander NC, et al. Skin autofluorescence, a measure of cumulative metabolic stress and advanced glycation end products, predicts mortality in hemodialysis patients. J Am Soc Nephrol. 2005 Dec;16(12):3687-93.

40. Tanaka K, Tani Y, Asai J, Nemoto F, Kusano Y, Suzuki H, et al. Skin autofluorescence is associated with renal function and cardiovascular diseases in pre-dialysis chronic kidney disease patients. Nephrol Dial Transplant. 2011 Jan;26(1):214-20.

41. Gerrits EG, Lutgers HL, Smeets GH, Groenier KH, Smit AJ, Gans RO, et al. Skin autofluorescence: a pronounced marker of mortality in hemodialysis patients. Nephron Extra. 2012 Jan;2(1):184-91.

42. Hartog JW, de Vries AP, Bakker SJ, Graaff R, van Son WJ, van der Heide JJ, et al. Risk factors for chronic transplant dysfunction and cardiovascular disease are related to accumulation of advanced glycation end-products in renal transplant recipients. Nephrol Dial Transplant. 2006 Aug;21(8):2263-9.

43. Mulder DJ, van Haelst PL, Graaff R, Gans RO, Zijlstra F, Smit AJ. Skin autofluorescence is elevated in acute myocardial infarction and is associated with the one-year incidence of major adverse cardiac events. Neth Heart J. 2009 Apr;17(4):162-8.

(10)

Introduction and aim of the thesis 19

44. den Dekker MA, Zwiers M, van den Heuvel ER, de Vos LC, Smit AJ, Zeebregts CJ, et al. Skin autofluorescence, a non-invasive marker for AGE accumulation, is associated with the degree of atherosclerosis. PLoS One. 2013 Dec 23;8(12):e83084.

45. Mulder DJ, van Haelst PL, Gross S, de Leeuw K, Bijzet J, Graaff R, et al. Skin autofluorescence is elevated in patients with stable coronary artery disease and is associated with serum levels of neopterin and the soluble receptor for advanced glycation end products. Atherosclerosis. 2008 Mar;197(1):217-23.

46. Lutgers HL, Graaff R, de Vries R, Smit AJ, Dullaart RP. Carotid artery intima media thickness associates with skin autofluoresence in non-diabetic subjects without clinically manifest cardiovascular disease. Eur J Clin Invest. 2010 Sep;40(9):812-7.

47. de Vos LC, Mulder DJ, Smit AJ, Dullaart RP, Kleefstra N, Lijfering WM, et al. Skin autofluorescence is associated with 5-year mortality and cardiovascular events in patients with peripheral artery disease. Arterioscler Thromb Vasc Biol. 2014 Apr;34(4):933-8.

48. Noordzij MJ, Lefrandt JD, Loeffen EA, Saleem BR, Meerwaldt R, Lutgers HL, et al. Skin autofluorescence is increased in patients with carotid artery stenosis and peripheral artery disease. Int J Cardiovasc Imaging. 2012 Feb;28(2):431-8.

(11)

Referenties

GERELATEERDE DOCUMENTEN

Multivariate backward regression analysis showed that in the non-diabetic population, SAF was significantly and independently associated with age, BMI, HbA1c, creatinine

Using metabolomics data we have shown that cotinine N-oxide, a biomarker for environmental tobacco smoke exposure, is significantly associated with higher SAF levels in a group

Furthermore, SAF was significantly and independently associated with the presence of MetS and some of its individual components, particularly elevated blood pressure, impaired

In the present study, we examined whether the measurement of skin autofluorescence can predict 4 year risk of incident type 2 diabetes, cardiovascular disease (CVD) and mortality

Therefore, the goal of this study was to assess whether thyroid hormone levels are associated with skin autofluorescence measurements and whether they modulate the association

Skin autofluorescence, a marker of advanced glycation end products and oxidative stress, is increased in recently preeclamptic women. Coffeng SM, Blaauw J, Souwer ET, Rakhorst G,

With the Bender MedSystems ELISA used at location B, two samples could not be measured due to insufficient sample material, while three samples were below the detection limit of

Higher FT3 levels were associated with increased risk of both type 2 diabetes and cardiovascular disease, even after correction for SAF, age and the presence of the