Next Article in Journal
Development and Sequential Analysis of a New Multi-Agent, Anti-Acne Formulation Based on Plant-Derived Antimicrobial and Anti-Inflammatory Compounds
Next Article in Special Issue
Methylglyoxal-Glyoxalase 1 Balance: The Root of Vascular Damage
Previous Article in Journal
Hypoxia Inducible Factor 1α Promotes Endogenous Adaptive Response in Rat Model of Chronic Cerebral Hypoperfusion
Previous Article in Special Issue
Bacterial Responses to Glyoxal and Methylglyoxal: Reactive Electrophilic Species
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Glycative Stress and Its Defense Machinery Glyoxalase 1 in Renal Pathogenesis

1
Division of Nephrology and Endocrinology, The University of Tokyo Graduate School of Medicine, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
2
Division of Chronic Kidney Disease (CKD) Pathophysiology, The University of Tokyo Graduate School of Medicine, 7-3-1, Hongo, Bunkyo-ku, Tokyo 113-8655, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2017, 18(1), 174; https://doi.org/10.3390/ijms18010174
Submission received: 9 December 2016 / Revised: 5 January 2017 / Accepted: 10 January 2017 / Published: 17 January 2017
(This article belongs to the Special Issue Glyoxalase System)

Abstract

:
Chronic kidney disease is a major public health problem around the world. Because the kidney plays a role in reducing glycative stress, renal dysfunction results in increased glycative stress. In turn, glycative stress, especially that due to advanced glycated end products (AGEs) and their precursors such as reactive carbonyl compounds, exacerbates chronic kidney disease and is related to premature aging in chronic kidney disease, whether caused by diabetes mellitus or otherwise. Factors which hinder a sufficient reduction in glycative stress include the inhibition of anti-glycation enzymes (e.g., GLO-1), as well as pathogenically activated endoplasmic reticulum (ER) stress and hypoxia in the kidney. Promising strategies aimed at halting the vicious cycle between chronic kidney disease and increases in glycative stress include the suppression of AGE accumulation in the body and the enhancement of GLO-1 to strengthen the host defense machinery against glycative stress.

1. Introduction

Chronic kidney disease (CKD) is defined as “abnormalities of kidney structure or function, present for over three months, with implications for health”. A diagnosis of CKD is often based on persistent proteinuria and a decreased glomerular filtration rate [1]. The causes and exacerbating factors of CKD are wide-ranging, and include inflammation, hypoxia, endoplasmic reticulum (ER) stress, and glycative stress [2,3,4,5]. With regard to glycative stress, the kidney is the major site of clearance of advanced glycated end products (AGEs), a complex group of compounds formed via glycation of proteins and nucleic acids [6]. However, the kidney is also a target for AGE-mediated organ damage, and any increase in the AGE concentration in the presence of kidney damage thus forms a vicious cycle. This characteristic of AGEs in the context of renal pathogenesis and the progression of CKD has now attracted substantial research attention (Figure 1).
The production of AGEs is thought to be related to senescence. This notion originally arose from research into the effects of glyoxalase 1 (GLO-1) on the lifespan of C. elegans. GLO-1 is an enzyme which is part of the glyoxalase system in cytosol, where it prevents the accumulation of AGEs mediated by alpha-oxoaldehydes such as methylglyoxal and glyoxal [7]. GLO-1 over-expression prolongs the lifespan of C. elegans by ameliorating mitochondrial protein glycation [8,9]. Following more recent studies, AGEs are now recognized as inducers of premature senescence in both the whole body [10] and the kidney [11].
Another reason for the substantial research interest in AGEs in diabetic kidney disease is that AGEs can be involved in “metabolic memory”, a term used to describe the long-term benefit provided by initial strict glycemic control for a short time. Two clinical trials have shown that initial intensive glucose control is associated with fewer diabetic complications even years after treatment intervention for hyperglycemia ends [12,13]. Since AGE formation is greater in hyperglycemia and AGEs produce adverse long-term results in many organs, AGEs are likely a reason for the occurrence of “metabolic memory” [14]. The existence of “metabolic memory” has changed the clinical management of diabetes mellitus and its complications such as diabetic kidney disease, and the role of AGEs in the pathophysiology of diabetes and diabetic complications has in turn captured worldwide attention.

2. Glycative Stress in Pathogenesis of Kidney Disease

2.1. Diabetic Kidney Disease

The characteristic increase in glycative stress under high glucose concentrations has made this stress a frequent topic in research into diabetic complications. One convincing proof of the importance of glycative stress in diabetes is that the level of hemoglobin A1c (HbA1c), an Amadori rearrangement compound, is correlated with diabetic complications, and is clinically used as a surrogate marker of an average of blood glucose over approximately one month [15,16,17]. Further evidence is that the level of skin collagen glycation is proven to be correlated to the development of future diabetic complications [18]. Diabetic kidney disease is one of these complications, and its incidence and clinical course are related to the level of HbA1c. Accordingly, the role of AGEs and the glyoxalase system has attracted strong research interest [19].
The enzyme activity of GLO-1 in humans is influenced by at least two polymorphisms in the GLO-1 gene [20]. The question of whether GLO-1 polymorphism is related to the onset of diabetes or diabetic complications therefore warrants investigation. An answer might already have been found in the several studies in rats which showed convincing evidence for the close involvement of AGEs and GLO-1 in the pathogenesis of diabetic kidney disease. Initially, Karachalias et al. reported that AGEs accumulate in the glomeruli of streptozotocin-induced diabetic rats [21]. The next question was whether an alteration of GLO-1 activity affects the phenotype of diabetic kidney disease. An answer came with the finding that systemic GLO-1 over-expression decreases tissue AGEs, reduces urinary protein, and ameliorates mesangial expansion in streptozotocin-induced diabetic mice without changing the fasting plasma glucose [22,23,24]. Interestingly, GLO-1 knockdown rats have a similar phenotype to diabetic kidney disease, even without streptozotocin injection [23].
A second approach to evaluating the possible impact of GLO-1 polymorphism on the onset of diabetes or diabetic complications is via the blocking of AGE accumulation on target organs. Kaida et al. reported that the administration of high-affinity DNA-aptamer raised against AGEs in type 2 diabetic model mice (KKAy/Ta mice) resulted in a reduction in urinary protein, mesangial expansion, and other phenotypes of diabetic kidney disease [25]. These results indicate that AGEs are not only causative agents of diabetic kidney disease but also aggravating factors, at least in mice. Another study emphasized the role of receptor for AGEs (RAGE) in diabetic kidney disease: Reiniger et al. reported that homozygous RAGE knockout ameliorates albuminuria, the reduction in the glomerular filtration rate and other diabetic kidney disease–related renal changes [26].
Several effects of AGEs are known to influence cultured cells originating from nephrons in diabetic models, including podocytes, mesangial cells, and tubular cells. In podocytes and mesangial cells, some histone-modifying proteins are thought to be related to AGE-induced damage. With regard to podocytes, Wang et al. reported that AGEs increase the protein expression of histone deacetylase (HDAC) 4, similarly to high-glucose incubation or the addition of transforming growth factor β (TGF-β) [27]. Wang’s group also showed that in vivo silencing of HDAC4 in the whole kidney attenuated several diabetic changes, including albuminuria, mesangial expansion, an increase in TGF-β, and a reduced nephrin concentration, and that AGEs increase HDAC5 expression in mesangial cells, although the significance of this latter finding is not clear. Another report showed that AGEs decrease the silent information regulator 2-related protein 1 (Sirt1) protein, a histone deacetylase, in mesangial cells, resulting in the promotion of fibronectin 1 and TGF-β1 expression [28]. Taken together, AGE-induced podocytopathy is likely to be related to epigenetics.
With regard to tubular cells, Liu et al. proposed a novel role of AGEs in diabetic kidney disease following their cultured cell experiments. Exposure to AGE-BSA resulted in an increase in autophagosomes, a decrease in autolysosomes, defect in the acidification function in lysosomes, and suppression of the lysosomal degradation function [29]. Given that renal tubular lysosomal dysfunction is detected in type 1 diabetes in both animals and humans [30,31], and that lipid accumulation is seen in diabetic kidney disease [32], this glycative stress–related lysosomal dysfunction appears to be a pathological mechanism in diabetic kidney disease.

2.2. Other Non-Diabetic CKD (Chronic Kidney Disease)

A clinical study reported that RAGE polymorphism is related to non-diabetic CKD progression [33]. This finding implicates RAGE pathophysiologically in general CKD as well as in diabetic kidney disease [34]. The binding of AGEs with RAGE is one of the major pathogenic roles of AGEs [35,36], and this finding is thus sufficiently convincing to show that AGEs are related to CKD progression regardless of the blood glucose level even in humans. Indeed, there is a report showing that AGE accumulation and RAGE upregulation were seen in the peritoneal membrane in uremic patients [37]. In this study, the comorbidity of diabetes mellitus was only seen in approximately 20% of the patients.
As for experimental models, Vlassara et al. reported in 1994 that administration of AGE-albumin at 25 mg/kg/day causes glomerulosclerosis and albuminuria without diabetes [38]. In cultured cell experiments, AGEs have been proven to induce epithelial-myofibroblast transdifferentiation in tubular cells [39], an inflammatory response in mesangial cells [40], and podocyte apoptosis [41]. The AGE concentration is increased in non-diabetic CKD [42], and AGE-induced nephrotoxicity therefore plays an aggravating role even in non-diabetic CKD. In turn, the progression of CKD results in decreased renal function, followed by further AGE accumulation owing to decreased AGE clearance in the kidney. Clinical evidence of the relationship between RAGE polymorphism and CKD progression might be related to this vicious cycle; if the affinity between RAGE and AGEs changes, the effect of AGE accumulation on CKD progression may change.

3. GLO-1 System in Renal and Vascular Senescence

Since AGEs are closely related to senescence and GLO-1 is a strong detoxifier of AGEs, activating GLO-1 is a promising strategy to avoid renal senescence. Kumagai et al. reported the impact of renal GLO-1 activity in renal ischemia-reperfusion injury (IRI) in rats [43]. After IRI, renal GLO-1 activity was decreased, whereas GLO-1 expression was not changed at the mRNA or protein levels. This result suggests that the decreased activity of GLO-1 in the damaged kidney is caused by some post-transcriptional modification or the existence of inhibitors. Interestingly, the renal damage induced by IRI is ameliorated by over-expression of the human GLO-1 gene, and accompanied by lower levels of carboxyethyl lysine (CEL), oxidative stress, and apoptosis. A decrease in GLO-1 activity without any change in the protein levels of GLO-1 was also seen in aged rats compared with young rats [44], and thus this result can be interpreted to indicate the prevention of renal senescence induced by IRI via the over-expression of GLO-1. Ikeda et al. indicated that GLO-1 over-expression attenuates renal senescence in aged rats [44]: while cellular indicators of cell senescence such as senescence-associated β-galactosidase (SA-β-gal), p53, p21, and p16 were increased in the kidneys of aged rats, this age-related damage was ameliorated in GLO-1 transgenic aged rats. This cell-protective effect against cellular senescence is also seen in Human Renal Proximal Tubule Cells (RPTEC) and etoposide-induced cellular senescence models. These results indicate that reduced GLO-1 activity and subsequent renal senescence occur in CKD, and that the activation of GLO-1 can be an encouraging strategy against CKD.
Given that CKD is a high risk factor for cardiovascular disease [45,46], CKD can be interpreted as a systemic vascular disease. Jo-Watanabe et al. identified the role of GLO-1 in age-related endothelial dysfunction [47]. Age-related aortic glycative stress, the subsequent inhibitory phosphorylation of endothelial NO synthase (eNOS) (Thr495), and endothelial dysfunction were alleviated by GLO-1 over-expression. On this basis, the activation of GLO-1 also appears to be a promising way to reduce vascular senescence, which probably exists in CKD patients.

4. Renal Hypoxia as the Final Common Pathway of Chronic Kidney Disease

Renal tubulointerstitial hypoxia occurs in CKD; in turn, tubulointerstitial hypoxia exacerbates CKD [3,48]. Our group recently established a direct oxygen measurement technique in living mice, and proved that the kidney was clearly hypoxic in a CKD model [49]. The response against hypoxia is mainly regulated by hypoxia inducible factor (HIF). HIF is composed of two subunits, HIF-α, an oxygen-sensitive protein, and the HIF-β subunit. Three isoforms of HIF-α have been identified, and all of these subunits are degraded in cytosol if sufficient oxygen molecules are present. Once oxygen tension is lowered, HIF-α is accumulated in the cytosol where it forms a heterodimer with HIF-β, which then induces the activation of many HIF-downstream genes. Activation of HIF is thought to be renoprotective, and thus the regulation of HIF has attracted attention.
Recently, Yamaguchi et al. identified a strong association between inflammation and hypoxia [50]. They found that CCAAT/enhancer-binding protein δ (CEBPD) regulates HIF-1 mRNA expression, and that IL-1β, a well-known inflammatory cytokine, induces CEBPD. These results indicate that inflammation activates the HIF pathway. Both hypoxia and inflammation induce reactive oxygen species (ROS), and subsequent oxidative stress. Methylglyoxal (MG) is an AGE precursor. The production of MG increases in oxidative stress [51], and MG is reported to inhibit the heterodimer formation of HIF-α and HIF-β. The idea that the hypoactivation of HIF in renal ischemia underlies the progression of CKD suggests that glycative stress and CKD progression are linked in a vicious cycle, partly via renal hypoxia. More research is needed to elucidate how renal inflammation stands between these two factors.

5. Cross-Talk between Glycative Stress, ER (Endoplasmic Reticulum) Stress and Hypoxia

The endoplasmic reticulum (ER) is an organelle which maintains protein homeostasis via posttranscriptional modification, such as folding and degrading unfavorable proteins [5]. ER dysfunction, specifically ER stress, is an important factor in the pathogenesis or progression of various diseases, including kidney disease. ER stress is closely linked to glycative stress. Liu et al. demonstrated one example, showing that the expressions of RAGE, ER stress marker glucose-regulated protein 78 (GRP78), and p21, a cell arrest marker, are all independently correlated with renal senescence, as estimated by SA-β-gal activity, in diabetic kidney disease patients [11]. They also showed that AGEs and ER stress inducers have similar effects on renal tubular cells and result in premature senescence. The link between glycative stress and ER stress has also been shown in other diseases or other organs [52]. This is probably because, since glycation is a non-enzymatic posttranslational modification of proteins, pathogenic glycated proteins may cause protein malfolding, and subsequent ER stress.
It has been shown that ER stress impairs erythropoietin (EPO) production by derangement of the enhancer activity of the EPO gene [53]. Insufficient EPO production or response to EPO is a major mechanism of the anemia experienced by CKD patients, which in turn causes intracellular hypoxia, as shown by our group [49]. AGEs are also related to erythropoietin resistance in CKD patients as uremic toxins [54]. In turn, erythropoietin has a protective effect against AGEs in several cells [55,56,57]. These findings reveal the presence of complicated cross-talk between glycative stress, ER stress, and hypoxia, and therapeutic approaches to CKD must consider these three angles.

6. Intervention in Renal Glycative Stress

6.1. Activating GLO-1

Since GLO-1 is a powerful detoxifying enzyme of AGE precursors, such as glyoxal (GO) and MG, activation of GLO-1 is a promising strategy against glycative stress–induced organ dysfunction. One mild intervention to activate GLO-1 is the enforcement of exercise. Exercise training in aged rats has multiple effects on glycation stress, including activating GLO-1, reducing MG and CML (GO-adducts) concentrations, and suppressing RAGE expression in the aorta [58]. Against this, another study in rats indicated that exercise increased the numbers of AGE-positive damaged tubular cells [59]; these authors speculated that an increase in the single nephron glomerular filtration rate might result in higher AGE excretion from the blood to primitive urine, and subsequent AGE reabsorption into tubules. Taken together, these findings may suggest that exercise reduces the systemic amount of AGE in the body, but increases AGE accumulation in tubules. Further research to determine whether exercise indeed protects the kidney from AGE-induced injury is warranted. A pharmacological intervention to activate GLO-1 has attracted major interest, and several drugs with this effect have been reported [60,61,62]. Among them, activating the Nrf2-Keap1-ARE (antioxidant responsible elements) pathway was recently reported to have the potential to activate GLO-1 [62,63]. Nrf2 is a transcriptional factor which is regulated by Keap1, and in the presence of stress, such as oxidative stress, the dissociation of Keap1 from Nrf2 increases Nrf2 nuclear accumulation, resulting in its binding to ARE and the subsequent enhancement of downstream gene expressions. Bardoxolone methyl, a drug which activates the Nrf2-Keap1-ARE pathway [64], has been proven to protect kidney function in diabetic kidney disease [65,66,67]. This drug also triggered cardiovascular side effects such as congestive heart failure, and its phase III trial was stopped; however, its renal protective effect (improvement of the estimated glomerular filtration rate) on diabetic kidney disease is clear, and it is again in a phase II trial in Japan, with particular attention being paid to its adverse effects. Data from interim analysis demonstrated a significant improvement in the glomerular filtration rate, as estimated by insulin clearance, without any safety concerns in the bardoxolone methyl group compared to the placebo group. The mechanism by which this drug improves kidney function is not sufficiently understood, but previous research suggests that it may be due to GLO-1 activation, at least in part [62,63]. Another promising GLO-1 inducer is a binary combination of trans-resveratrol and hesperetin (tRES-HESP), which synergistically increase GLO-1 expression [68]. tRES-HESP are proven to increase GLO-1 activity, decrease plasma methylglyoxal and total body methylglyoxal-protein glycation, and finally improve metabolic and vascular health in overweight and obese patients [69].

6.2. Suppression of AGE Accumulation and Reduction of AGE Production

The suppression of AGE absorption from the intestine is another strategy. The serum AGE concentration is reported to be related to dietary AGE intake [51]. Although one report insists that the influence of the dietary AGE amount is not sufficient to cause vascular endothelial dysfunction in healthy volunteers [70], this report does not clearly mean that the dietary AGE amount does not matter in diabetic or CKD patients. To elucidate the importance of dietary AGE intake in CKD, a high-quality randomized controlled trial is required [71]. To decrease the AGE content, the cooking method is as important as the food being cooked; methods that use lower temperature and higher moisture (i.e., boiling) are desirable because they reduce AGE formation compared with dry heat cooking methods (i.e., broiling) [72].
AGE absorbents can promote the fecal excretion of AGEs. Sevelamer hydrochloride, a frequently used inorganic phosphate binder in dialysis patients, is reportedly able to reduce the plasma pentosidine level in hemodialytic patients [73]. Similarly, this agent also reduces serum levels of MG and (ε)N-carboxymethyl-lysine in non-dialytic diabetic patients [74]. Given also that sevelamer is not absorbed into the blood from the gut and can bind to AGE-BSA, these effects are derived from the absorption and fecal excretion of AGEs by sevelamer. Subsequent research revealed that sevelamer carbonate ameliorates albuminuria and reduces circulating and cellular AGEs in type 2 diabetes patients [75]. These reports indicated that such absorption agents will be beneficial to patients with diabetes, especially those with diabetic kidney disease.
Another method of reducing the plasma and renal AGE levels is suppression of AGE formation in the body. Aminoguanidine is a prototype therapeutic agent which scavenges dicarbonyl glycating agents, such as methylglyoxal and glyoxal [76]. However, its clinical trial against diabetic nephropathy in type 2 diabetes mellitus needs to be terminated according to safety concerns and an apparent lack of efficacy.
Several derivatives of vitamin B1 and B6 have the potential to inhibit AGE formation; the representative drugs are benfotiamine and pyridoxamine, a form of vitamin B6 [77,78]. Benfotiamine activates transketolase and has antioxidative effects in diabetes. This drug is proven to protect tissue from high-glucose–induced cell damage via alleviation of AGE accumulation [79,80,81]. Pyridoxamine inhibits AGE formation in vitro [82], and inhibits albuminuria in streptozotocin-induced diabetic rats [83]. A recent report by Golegaonkar et al. indicated that rifampicin, an old antibiotic, has the potential to suppress age-related glycative stress and elongate the lifespan of C. elegans [84].

7. Future Directions

As described above, AGEs and the kidney are closely related in that AGEs accumulate with renal dysfunction and have a nephropathic role. Although the mechanism of AGE-induced renal damage is quite complicated, the fact that AGEs damage the kidney has been well proven in animal experiments and cells derived from humans. However, clinical research which proves that AGEs are toxic to the kidney or that anti-AGE treatment is effective against CKD progression is not presently sufficient. It is fortunate that nephrologists can easily obtain urine, which is strongly affected by renal conditions, from CKD patients. Together with recent advances in metabolomics analysis, the role of AGEs in clinical settings must be more thoroughly researched.

Acknowledgments

This work was supported by the Japan Society for the Promotion of Science Grants-in-Aid for Scientific Research (25461207, 15KT0088, and 16K15465 to Reiko Inagi), the Yakult Bio-Science Foundation (to Reiko Inagi), and a research grant from Kyowa Hakko Kirin Co., Ltd. (Tokyo, Japan) (to Reiko Inagi).

Author Contributions

Yosuke Hirakawa wrote the draft and Reiko Inagi edited the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. GFR Decreased. Chapter 1: Definition and classification of CKD. Kidney Int. Suppl. 2013, 3, 19–62. [Google Scholar]
  2. Inagi, R. Glycative stress and glyoxalase in kidney disease and aging. Biochem. Soc. Trans. 2014, 42, 457–460. [Google Scholar] [CrossRef] [PubMed]
  3. Nangaku, M. Chronic hypoxia and tubulointerstitial injury: A final common pathway to end-stage renal failure. J. Am. Soc. Nephrol. 2006, 17, 17–25. [Google Scholar] [CrossRef] [PubMed]
  4. Nikolic-Paterson, D.J.; Wang, S.; Lan, H.Y. Macrophages promote renal fibrosis through direct and indirect mechanisms. Kidney Int. Suppl. 2014, 4, 34–38. [Google Scholar] [CrossRef] [PubMed]
  5. Inagi, R.; Ishimoto, Y.; Nangaku, M. Proteostasis in endoplasmic reticulum—New mechanisms in kidney disease. Nat. Rev. Nephrol. 2014, 10, 369–378. [Google Scholar] [CrossRef] [PubMed]
  6. Miyata, T.; Ueda, Y.; Horie, K.; Nangaku, M.; Tanaka, S.; de Strihou, C.V.Y.; Kurokawa, K. Renal catabolism of advanced glycation end products: The fate of pentosidine. Kidney Int. 1998, 53, 416–422. [Google Scholar] [CrossRef] [PubMed]
  7. Thornalley, P.J. Glyoxalase I—Structure, function and a critical role in the enzymatic defence against glycation. Biochem. Soc. Trans. 2003, 31, 1343–1348. [Google Scholar] [CrossRef] [PubMed]
  8. Morcos, M.; Du, X.; Pfisterer, F.; Hutter, H.; Sayed, A.A.; Thornalley, P.; Ahmed, N.; Baynes, J.; Thorpe, S.; Kukudov, G.; et al. Glyoxalase-1 prevents mitochondrial protein modification and enhances lifespan in Caenorhabditis elegans. Aging Cell 2008, 7, 260–269. [Google Scholar] [CrossRef] [PubMed]
  9. Schlotterer, A.; Kukudov, G.; Bozorgmehr, F.; Hutter, H.; Du, X.; Oikonomou, D.; Ibrahim, Y.; Pfisterer, F.; Rabbani, N.; Thornalley, P.; et al. C. Elegans as model for the study of high glucose-mediated life span reduction. Diabetes 2009, 58, 2450–2456. [Google Scholar] [CrossRef] [PubMed]
  10. Semba, R.D.; Nicklett, E.J.; Ferrucci, L. Does accumulation of advanced glycation end products contribute to the aging phenotype? J. Gerontol. A Biol. Sci. Med. Sci. 2010, 65, 963–975. [Google Scholar] [CrossRef] [PubMed]
  11. Liu, J.; Huang, K.; Cai, G.Y.; Chen, X.M.; Yang, J.R.; Lin, L.R.; Yang, J.; Huo, B.G.; Zhan, J.; He, Y.N. Receptor for advanced glycation end-products promotes premature senescence of proximal tubular epithelial cells via activation of endoplasmic reticulum stress-dependent p21 signaling. Cell Signal. 2014, 26, 110–121. [Google Scholar] [CrossRef] [PubMed]
  12. Nathan, D.M.; Cleary, P.A.; Backlund, J.Y.; Genuth, S.M.; Lachin, J.M.; Orchard, T.J.; Raskin, P.; Zinman, B. Diabetes Control and Complications Trial/Epidemiology of Diabetes Interventions and Complications (DCCT/EDIC) Study Research Group. Intensive diabetes treatment and cardiovascular disease in patients with type 1 diabetes. N. Engl. J. Med. 2005, 353, 2643–2653. [Google Scholar] [PubMed]
  13. Chalmers, J.; Cooper, M.E. UKPDS and the legacy effect. N. Engl. J. Med. 2008, 359, 1618–1620. [Google Scholar] [CrossRef] [PubMed]
  14. Ceriello, A. Hypothesis: The “metabolic memory”, the new challenge of diabetes. Diabetes Res. Clin. Pract. 2009, 86, S2–S6. [Google Scholar] [CrossRef]
  15. Stratton, I.M.; Adler, A.I.; Neil, H.A.; Matthews, D.R.; Manley, S.E.; Cull, C.A.; Hadden, D.; Turner, R.C.; Holman, R.R. Association of glycaemia with macrovascular and microvascular complications of type 2 diabetes (UKPDS 35): Prospective observational study. BMJ 2000, 321, 405–412. [Google Scholar] [CrossRef] [PubMed]
  16. UK Prospective Diabetes Study (UKPDS) Group. Intensive blood-glucose control with sulphonylureas or insulin compared with conventional treatment and risk of complications in patients with type 2 diabetes (UKPDS 33). Lancet 1998, 352, 837–853. [Google Scholar]
  17. American Diabetes Association. The absence of a glycemic threshold for the development of long-term complications: The perspective of the diabetes control and complications trial. Diabetes 1996, 45, 1289–1298. [Google Scholar]
  18. Monnier, V.M.; Bautista, O.; Kenny, D.; Sell, D.R.; Fogarty, J.; Dahms, W.; Cleary, P.A.; Lachin, J.; Genuth, S. Skin collagen glycation, glycoxidation, and crosslinking are lower in subjects with long-term intensive versus conventional therapy of type 1 diabetes: Relevance of glycated collagen products versus HbA1c as markers of diabetic complications. DCCT Skin Collagen Ancillary Study Group. Diabetes Control and Complications Trial. Diabetes 1999, 48, 870–880. [Google Scholar] [PubMed]
  19. Rabbani, N.; Thornalley, P.J. The critical role of methylglyoxal and glyoxalase 1 in diabetic nephropathy. Diabetes 2014, 63, 50–52. [Google Scholar] [CrossRef] [PubMed]
  20. Peculis, R.; Konrade, I.; Skapare, E.; Fridmanis, D.; Nikitina-Zake, L.; Lejnieks, A.; Pirags, V.; Dambrova, M.; Klovins, J. Identification of glyoxalase 1 polymorphisms associated with enzyme activity. Gene 2013, 515, 140–143. [Google Scholar] [CrossRef] [PubMed]
  21. Karachalias, N.; Babaei-Jadidi, R.; Ahmed, N.; Thornalley, P.J. Accumulation of fructosyl-lysine and advanced glycation end products in the kidney, retina and peripheral nerve of streptozotocin-induced diabetic rats. Biochem. Soc. Trans. 2003, 31, 1423–1425. [Google Scholar] [CrossRef] [PubMed]
  22. Brouwers, O.; Niessen, P.M.; Ferreira, I.; Miyata, T.; Scheffer, P.G.; Teerlink, T.; Schrauwen, P.; Brownlee, M.; Stehouwer, C.D.; Schalkwijk, C.G. Overexpression of glyoxalase-I reduces hyperglycemia-induced levels of advanced glycation end products and oxidative stress in diabetic rats. J. Biol. Chem. 2011, 286, 1374–1380. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  23. Giacco, F.; Du, X.; D’Agati, V.D.; Milne, R.; Sui, G.; Geoffrion, M.; Brownlee, M. Knockdown of glyoxalase 1 mimics diabetic nephropathy in nondiabetic mice. Diabetes 2014, 63, 291–299. [Google Scholar] [CrossRef] [PubMed]
  24. Brouwers, O.; Niessen, P.M.; Miyata, T.; Østergaard, J.A.; Flyvbjerg, A.; Peutz-Kootstra, C.J.; Sieber, J.; Mundel, P.H.; Brownlee, M.; Janssen, B.J.; et al. Glyoxalase-1 overexpression reduces endothelial dysfunction and attenuates early renal impairment in a rat model of diabetes. Diabetologia 2014, 57, 224–235. [Google Scholar] [CrossRef] [PubMed]
  25. Kaida, Y.; Fukami, K.; Matsui, T.; Higashimoto, Y.; Nishino, Y.; Obara, N.; Nakayama, Y.; Ando, R.; Toyonaga, M.; Ueda, S.; et al. DNA aptamer raised against ages blocks the progression of experimental diabetic nephropathy. Diabetes 2013, 62, 3241–3250. [Google Scholar] [CrossRef] [PubMed]
  26. Reiniger, N.; Lau, K.; McCalla, D.; Eby, B.; Cheng, B.; Lu, Y.; Qu, W.; Quadri, N.; Ananthakrishnan, R.; Furmansky, M.; et al. Deletion of the receptor for advanced glycation end products reduces glomerulosclerosis and preserves renal function in the diabetic OVE26 mouse. Diabetes 2010, 59, 2043–2054. [Google Scholar] [CrossRef] [PubMed]
  27. Wang, X.; Liu, J.; Zhen, J.; Zhang, C.; Wan, Q.; Liu, G.; Wei, X.; Zhang, Y.; Wang, Z.; Han, H.; et al. Histone deacetylase 4 selectively contributes to podocyte injury in diabetic nephropathy. Kidney Int. 2014, 86, 712–725. [Google Scholar] [CrossRef] [PubMed]
  28. Huang, K.P.; Chen, C.; Hao, J.; Huang, J.Y.; Liu, P.Q.; Huang, H.Q. AGEs-RAGE system down-regulates Sirt1 through the ubiquitin-proteasome pathway to promote FN and TGF-β1 expression in male rat glomerular mesangial cells. Endocrinology 2015, 156, 268–279. [Google Scholar] [CrossRef] [PubMed]
  29. Liu, W.J.; Shen, T.T.; Chen, R.H.; Wu, H.L.; Wang, Y.J.; Deng, J.K.; Chen, Q.H.; Pan, Q.; Huang Fu, C.M.; Tao, J.L.; et al. Autophagy-lysosome pathway in renal tubular epithelial cells is disrupted by advanced glycation end products in diabetic nephropathy. J. Biol. Chem. 2015, 290, 20499–20510. [Google Scholar] [CrossRef] [PubMed]
  30. Shechter, P.; Boner, G.; Rabkin, R. Tubular cell protein degradation in early diabetic renal hypertrophy. J. Am. Soc. Nephrol. 1994, 4, 1582–1587. [Google Scholar] [PubMed]
  31. Osicka, T.M.; Houlihan, C.A.; Chan, J.G.; Jerums, G.; Comper, W.D. Albuminuria in patients with type 1 diabetes is directly linked to changes in the lysosome-mediated degradation of albumin during renal passage. Diabetes 2000, 49, 1579–1584. [Google Scholar] [CrossRef] [PubMed]
  32. Stadler, K.; Goldberg, I.J.; Susztak, K. The evolving understanding of the contribution of lipid metabolism to diabetic kidney disease. Curr. Diabetes Rep. 2015, 15, 40. [Google Scholar] [CrossRef] [PubMed]
  33. Baragetti, I.; Norata, G.D.; Sarcina, C.; Baragetti, A.; Rastelli, F.; Buzzi, L.; Grigore, L.; Garlaschelli, K.; Pozzi, C.; Catapano, A.L. -374 T/A RAGE polymorphism is associated with chronic kidney disease progression in subjects affected by nephrocardiovascular disease. PLoS ONE 2013, 8, e60089. [Google Scholar] [CrossRef] [PubMed]
  34. Inagi, R.; Yamamoto, Y.; Nangaku, M.; Usuda, N.; Okamato, H.; Kurokawa, K.; van Ypersele de Strihou, C.V.Y.; Yamamoto, H.; Miyata, T. A severe diabetic nephropathy model with early development of nodule-like lesions induced by megsin overexpression in RAGE/iNOS transgenic mice. Diabetes 2006, 55, 356–366. [Google Scholar] [CrossRef] [PubMed]
  35. Fleming, T.H.; Humpert, P.M.; Nawroth, P.P.; Bierhaus, A. Reactive metabolites and AGE/RAGE-mediated cellular dysfunction affect the aging process: A mini-review. Gerontology 2011, 57, 435–443. [Google Scholar] [CrossRef] [PubMed]
  36. Coughlan, M.T.; Thorburn, D.R.; Penfold, S.A.; Laskowski, A.; Harcourt, B.E.; Sourris, K.C.; Tan, A.L.; Fukami, K.; Thallas-Bonke, V.; Nawroth, P.P.; et al. RAGE-induced cytosolic ROS promote mitochondrial superoxide generation in diabetes. J. Am. Soc. Nephrol. 2009, 20, 742–752. [Google Scholar] [CrossRef] [PubMed]
  37. Kihm, L.P.; Wibisono, D.; Müller-Krebs, S.; Pfisterer, F.; Morath, C.; Gross, M.L.; Morcos, M.; Seregin, Y.; Bierhaus, A.; Nawroth, P.P.; et al. RAGE expression in the human peritoneal membrane. Nephrol. Dial. Transplant. 2008, 23, 3302–3306. [Google Scholar] [CrossRef] [PubMed]
  38. Vlassara, H.; Striker, L.J.; Teichberg, S.; Fuh, H.; Li, Y.M.; Steffes, M. Advanced glycation end products induce glomerular sclerosis and albuminuria in normal rats. Proc. Natl. Acad. Sci. USA 1994, 91, 11704–11708. [Google Scholar] [CrossRef] [PubMed]
  39. Oldfield, M.D.; Bach, L.A.; Forbes, J.M.; Nikolic-Paterson, D.; McRobert, A.; Thallas, V.; Atkins, R.C.; Osicka, T.; Jerums, G.; Cooper, M.E. Advanced glycation end products cause epithelial-myofibroblast transdifferentiation via the receptor for advanced glycation end products (RAGE). J. Clin. Investig. 2001, 108, 1853–1863. [Google Scholar] [CrossRef] [PubMed]
  40. Lu, C.; He, J.C.; Cai, W.; Liu, H.; Zhu, L.; Vlassara, H. Advanced glycation endproduct (AGE) receptor 1 is a negative regulator of the inflammatory response to age in mesangial cells. Proc. Natl. Acad. Sci. USA 2004, 101, 11767–11772. [Google Scholar] [CrossRef] [PubMed]
  41. Chuang, P.Y.; Yu, Q.; Fang, W.; Uribarri, J.; He, J.C. Advanced glycation endproducts induce podocyte apoptosis by activation of the FOXO4 transcription factor. Kidney Int. 2007, 72, 965–976. [Google Scholar] [CrossRef] [PubMed]
  42. Stinghen, A.E.; Massy, Z.A.; Vlassara, H.; Striker, G.E.; Boullier, A. Uremic toxicity of advanced glycation end products in CKD. J. Am. Soc. Nephrol. 2016, 27, 354–370. [Google Scholar] [CrossRef] [PubMed]
  43. Kumagai, T.; Nangaku, M.; Kojima, I.; Nagai, R.; Ingelfinger, J.R.; Miyata, T.; Fujita, T.; Inagi, R. Glyoxalase I overexpression ameliorates renal ischemia-reperfusion injury in rats. Am. J. Physiol. Ren. Physiol. 2009, 296, F912–F921. [Google Scholar] [CrossRef] [PubMed]
  44. Ikeda, Y.; Inagi, R.; Miyata, T.; Nagai, R.; Arai, M.; Miyashita, M.; Itokawa, M.; Fujita, T.; Nangaku, M. Glyoxalase I retards renal senescence. Am. J. Pathol. 2011, 179, 2810–2821. [Google Scholar] [CrossRef] [PubMed]
  45. Levey, A.S.; de Jong, P.E.; Coresh, J.; El Nahas, M.; Astor, B.C.; Matsushita, K.; Gansevoort, R.T.; Kasiske, B.L.; Eckardt, K.U. The definition, classification, and prognosis of chronic kidney disease: A KDIGO Controversies Conference report. Kidney Int. 2011, 80, 17–28. [Google Scholar] [CrossRef] [PubMed]
  46. Foley, R.N.; Murray, A.M.; Li, S.; Herzog, C.A.; McBean, A.M.; Eggers, P.W.; Collins, A.J. Chronic kidney disease and the risk for cardiovascular disease, renal replacement, and death in the united states medicare population, 1998 to 1999. J. Am. Soc. Nephrol. 2005, 16, 489–495. [Google Scholar] [CrossRef] [PubMed]
  47. Jo-Watanabe, A.; Ohse, T.; Nishimatsu, H.; Takahashi, M.; Ikeda, Y.; Wada, T.; Shirakawa, J.; Nagai, R.; Miyata, T.; Nagano, T.; et al. Glyoxalase I reduces glycative and oxidative stress and prevents age-related endothelial dysfunction through modulation of endothelial nitric oxide synthase phosphorylation. Aging Cell 2014, 13, 519–528. [Google Scholar] [CrossRef] [PubMed]
  48. Mimura, I.; Nangaku, M. The suffocating kidney: Tubulointerstitial hypoxia in end-stage renal disease. Nat. Rev. Nephrol. 2010, 6, 667–678. [Google Scholar] [CrossRef] [PubMed]
  49. Hirakawa, Y.; Yoshihara, T.; Kamiya, M.; Mimura, I.; Fujikura, D.; Masuda, T.; Kikuchi, R.; Takahashi, I.; Urano, Y.; Tobita, S.; et al. Quantitating intracellular oxygen tension in vivo by phosphorescence lifetime measurement. Sci. Rep. 2015, 5, 17838. [Google Scholar] [CrossRef] [PubMed]
  50. Yamaguchi, J.; Tanaka, T.; Eto, N.; Nangaku, M. Inflammation and hypoxia linked to renal injury by CCAAT/enhancer-binding protein δ. Kidney Int. 2015, 88, 262–275. [Google Scholar] [CrossRef] [PubMed]
  51. Uribarri, J.; Cai, W.; Peppa, M.; Goodman, S.; Ferrucci, L.; Striker, G.; Vlassara, H. Circulating glycotoxins and dietary advanced glycation endproducts: Two links to inflammatory response, oxidative stress, and aging. J. Gerontol. A Biol. Sci. Med. Sci. 2007, 62, 427–433. [Google Scholar] [CrossRef] [PubMed]
  52. Inagi, R. RAGE and glyoxalase in kidney disease. Glycoconj. J. 2016, 33, 619–626. [Google Scholar] [CrossRef] [PubMed]
  53. Chiang, C.K.; Nangaku, M.; Tanaka, T.; Iwawaki, T.; Inagi, R. Endoplasmic reticulum stress signal impairs erythropoietin production: A role for ATF4. Am. J. Physiol. Cell Physiol. 2013, 304, C342–C353. [Google Scholar] [CrossRef] [PubMed]
  54. Nangaku, M.; Mimura, I.; Yamaguchi, J.; Higashijima, Y.; Wada, T.; Tanaka, T. Role of uremic toxins in erythropoiesis-stimulating agent resistance in chronic kidney disease and dialysis patients. J. Ren. Nutr. 2015, 25, 160–163. [Google Scholar] [CrossRef] [PubMed]
  55. Lee, S.T.; Chu, K.; Park, J.E.; Jung, K.H.; Jeon, D.; Lim, J.Y.; Lee, S.K.; Kim, M.; Roh, J.K. Erythropoietin improves memory function with reducing endothelial dysfunction and amyloid-β burden in Alzheimer’s disease models. J. Neurochem. 2012, 120, 115–124. [Google Scholar] [CrossRef] [PubMed]
  56. Yu, T.; Li, L.; Chen, T.; Liu, Z.; Liu, H.; Li, Z. Erythropoietin attenuates advanced glycation endproducts-induced toxicity of schwann cells in vitro. Neurochem. Res. 2015, 40, 698–712. [Google Scholar] [CrossRef] [PubMed]
  57. Loeffler, I.; Ruster, C.; Franke, S.; Liebisch, M.; Wolf, G. Erythropoietin ameliorates podocyte injury in advanced diabetic nephropathy in the db/db mouse. Am. J. Physiol. Ren. Physiol. 2013, 305, F911–F918. [Google Scholar] [CrossRef] [PubMed]
  58. Gu, Q.; Wang, B.; Zhang, X.F.; Ma, Y.P.; Liu, J.D.; Wang, X.Z. Contribution of receptor for advanced glycation end products to vasculature-protecting effects of exercise training in aged rats. Eur. J. Pharmacol. 2014, 741, 186–194. [Google Scholar] [CrossRef] [PubMed]
  59. Tsutsumi, E.; Murata, Y.; Sakamoto, M.; Horikawa, E. Effects of exercise on the nephron of Goto-Kakizaki rats: Morphological, and advanced glycation end-products and inducible nitric oxide synthase immunohistochemical analyses. J. Diabetes Complicat. 2015, 29, 472–478. [Google Scholar] [CrossRef] [PubMed]
  60. Liu, Y.W.; Zhu, X.; Zhang, L.; Lu, Q.; Wang, J.Y.; Zhang, F.; Guo, H.; Yin, J.L.; Yin, X.X. Up-regulation of glyoxalase 1 by mangiferin prevents diabetic nephropathy progression in streptozotocin-induced diabetic rats. Eur. J. Pharmacol. 2013, 721, 355–364. [Google Scholar] [CrossRef] [PubMed]
  61. Choi, E.M.; Suh, K.S.; Rhee, S.Y.; Kim, Y.S. Sciadopitysin alleviates methylglyoxal-mediated glycation in osteoblastic MC3T3-E1 cells by enhancing glyoxalase system and mitochondrial biogenesis. Free Radic. Res. 2014, 48, 729–739. [Google Scholar] [CrossRef] [PubMed]
  62. Liu, Y.W.; Cheng, Y.Q.; Liu, X.L.; Hao, Y.C.; Li, Y.; Zhu, X.; Zhang, F.; Yin, X.X. Mangiferin upregulates glyoxalase 1 through activation of Nrf2/are signaling in central neurons cultured with high glucose. Mol. Neurobiol. 2016. [Google Scholar] [CrossRef] [PubMed]
  63. Xue, M.; Rabbani, N.; Momiji, H.; Imbasi, P.; Anwar, M.M.; Kitteringham, N.; Park, B.K.; Souma, T.; Moriguchi, T.; Yamamoto, M.; et al. Transcriptional control of glyoxalase 1 by Nrf2 provides a stress-responsive defence against dicarbonyl glycation. Biochem. J. 2012, 443, 213–222. [Google Scholar] [CrossRef] [PubMed]
  64. Yates, M.S.; Tauchi, M.; Katsuoka, F.; Flanders, K.C.; Liby, K.T.; Honda, T.; Gribble, G.W.; Johnson, D.A.; Johnson, J.A.; Burton, N.C.; et al. Pharmacodynamic characterization of chemopreventive triterpenoids as exceptionally potent inducers of Nrf2-regulated genes. Mol. Cancer Ther. 2007, 6, 154–162. [Google Scholar] [CrossRef]
  65. Pergola, P.E.; Krauth, M.; Huff, J.W.; Ferguson, D.A.; Ruiz, S.; Meyer, C.J.; Warnock, D.G. Effect of bardoxolone methyl on kidney function in patients with T2D and stage 3b–4 CKD. Am. J. Nephrol. 2011, 33, 469–476. [Google Scholar] [CrossRef] [PubMed]
  66. Pergola, P.E.; Raskin, P.; Toto, R.D.; Meyer, C.J.; Huff, J.W.; Grossman, E.B.; Krauth, M.; Ruiz, S.; Audhya, P.; Christ-Schmidt, H.; et al. Bardoxolone methyl and kidney function in CKD with type 2 diabetes. N. Engl. J. Med. 2011, 365, 327–336. [Google Scholar] [CrossRef] [PubMed]
  67. De Zeeuw, D.; Akizawa, T.; Audhya, P.; Bakris, G.L.; Chin, M.; Christ-Schmidt, H.; Goldsberry, A.; Houser, M.; Krauth, M.; Lambers Heerspink, H.J.; et al. Bardoxolone methyl in type 2 diabetes and stage 4 chronic kidney disease. N. Engl. J. Med. 2013, 369, 2492–2503. [Google Scholar] [CrossRef] [PubMed]
  68. Rabbani, N.; Xue, M.; Thornalley, P.J. Dicarbonyls and glyoxalase in disease mechanisms and clinical therapeutics. Glycoconj. J. 2016, 33, 513–525. [Google Scholar] [CrossRef] [PubMed]
  69. Xue, M.; Weickert, M.O.; Qureshi, S.; Kandala, N.B.; Anwar, A.; Waldron, M.; Shafie, A.; Messenger, D.; Fowler, M.; Jenkins, G.; et al. Improved Glycemic Control and Vascular Function in Overweight and Obese Subjects by Glyoxalase 1 Inducer Formulation. Diabetes 2016, 65, 2282–2294. [Google Scholar] [CrossRef] [PubMed]
  70. Semba, R.D.; Gebauer, S.K.; Baer, D.J.; Sun, K.; Turner, R.; Silber, H.A.; Talegawkar, S.; Ferrucci, L.; Novotny, J.A. Dietary intake of advanced glycation end products did not affect endothelial function and inflammation in healthy adults in a randomized controlled trial. J. Nutr. 2014, 144, 1037–1042. [Google Scholar] [CrossRef] [PubMed]
  71. Clarke, R.E.; Dordevic, A.L.; Tan, S.M.; Ryan, L.; Coughlan, M.T. Dietary advanced glycation end products and risk factors for chronic disease: A systematic review of randomised controlled trials. Nutrients 2016, 8, 125. [Google Scholar] [CrossRef] [PubMed]
  72. Uribarri, J.; He, J.C. The low age diet: A neglected aspect of clinical nephrology practice? Nephron 2015, 130, 48–53. [Google Scholar] [CrossRef] [PubMed]
  73. Kakuta, T.; Tanaka, R.; Hyodo, T.; Suzuki, H.; Kanai, G.; Nagaoka, M.; Takahashi, H.; Hirawa, N.; Oogushi, Y.; Miyata, T.; et al. Effect of sevelamer and calcium-based phosphate binders on coronary artery calcification and accumulation of circulating advanced glycation end products in hemodialysis patients. Am. J. Kidney Dis. 2011, 57, 422–431. [Google Scholar] [CrossRef] [PubMed]
  74. Vlassara, H.; Uribarri, J.; Cai, W.; Goodman, S.; Pyzik, R.; Post, J.; Grosjean, F.; Woodward, M.; Striker, G.E. Effects of sevelamer on HbA1c, inflammation, and advanced glycation end products in diabetic kidney disease. Clin. J. Am. Soc. Nephrol. 2012, 7, 934–942. [Google Scholar] [CrossRef] [PubMed]
  75. Yubero-Serrano, E.M.; Woodward, M.; Poretsky, L.; Vlassara, H.; Striker, G.E. Effects of sevelamer carbonate on advanced glycation end products and antioxidant/pro-oxidant status in patients with diabetic kidney disease. Clin. J. Am. Soc. Nephrol. 2015, 10, 759–766. [Google Scholar] [CrossRef] [PubMed]
  76. Thornalley, P.J. Use of aminoguanidine (Pimagedine) to prevent the formation of advanced glycation endproducts. Arch. Biochem. Biophys. 2003, 419, 31–40. [Google Scholar] [CrossRef] [PubMed]
  77. Thomas, M.C.; Baynes, J.W.; Thorpe, S.R.; Cooper, M.E. The role of AGEs and AGE inhibitors in diabetic cardiovascular disease. Curr. Drug Targets 2005, 6, 453–474. [Google Scholar] [CrossRef] [PubMed]
  78. Booth, A.A.; Khalifah, R.G.; Hudson, B.G. Thiamine pyrophosphate and pyridoxamine inhibit the formation of antigenic advanced glycation end-products: Comparison with aminoguanidine. Biochem. Biophys. Res. Commun. 1996, 220, 113–119. [Google Scholar] [CrossRef] [PubMed]
  79. Babaei-Jadidi, R.; Karachalias, N.; Ahmed, N.; Battah, S.; Thornalley, P.J. Prevention of incipient diabetic nephropathy by high-dose thiamine and benfotiamine. Diabetes 2003, 52, 2110–2120. [Google Scholar] [CrossRef] [PubMed]
  80. Kihm, L.P.; Müller-Krebs, S.; Klein, J.; Ehrlich, G.; Mertes, L.; Gross, M.L.; Adaikalakoteswari, A.; Thornalley, P.J.; Hammes, H.P.; Nawroth, P.P.; et al. Benfotiamine protects against peritoneal and kidney damage in peritoneal dialysis. J. Am. Soc. Nephrol. 2011, 22, 914–926. [Google Scholar] [CrossRef] [PubMed]
  81. Muller-Krebs, S.; Nissle, K.; Tsobaneli, J.; Zeier, M.; Kihm, L.P.; Kender, Z.; Fleming, T.; Nawroth, P.P.; Reiser, J.; Schwenger, V. Effect of benfotiamine in podocyte damage induced by peritoneal dialysis fluid. Front. Med. 2015, 2, 10. [Google Scholar]
  82. Booth, A.A.; Khalifah, R.G.; Todd, P.; Hudson, B.G. In vitro kinetic studies of formation of antigenic advanced glycation end products (AGEs). Novel inhibition of post-Amadori glycation pathways. J. Biol. Chem. 1997, 272, 5430–5437. [Google Scholar] [CrossRef] [PubMed]
  83. Degenhardt, T.P.; Alderson, N.L.; Arrington, D.D.; Beattie, R.J.; Basgen, J.M.; Steffes, M.W.; Thorpe, S.R.; Baynes, J.W. Pyridoxamine inhibits early renal disease and dyslipidemia in the streptozotocin-diabetic rat. Kidney Int. 2002, 61, 939–950. [Google Scholar] [CrossRef] [PubMed]
  84. Golegaonkar, S.; Tabrez, S.S.; Pandit, A.; Sethurathinam, S.; Jagadeeshaprasad, M.G.; Bansode, S.; Sampathkumar, S.G.; Kulkarni, M.J.; Mukhopadhyay, A. Rifampicin reduces advanced glycation end products and activates DAF-16 to increase lifespan in Caenorhabditis elegans. Aging Cell 2015, 14, 463–473. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Vicious cycle between CKD and glycative stress. In CKD patients, renal AGE clearance is insufficient because of the low glomerular filtration rate and GLO-1 inhibition. This is followed by AGE accumulation. AGEs themselves have been confirmed to have nephrotoxicity. Thus, CKD and AGE accumulation constitutes a vicious cycle. Abbreviations: AGEs, advanced glycation end products; CKD, chronic kidney disease; GLO-1, glyoxalase 1.
Figure 1. Vicious cycle between CKD and glycative stress. In CKD patients, renal AGE clearance is insufficient because of the low glomerular filtration rate and GLO-1 inhibition. This is followed by AGE accumulation. AGEs themselves have been confirmed to have nephrotoxicity. Thus, CKD and AGE accumulation constitutes a vicious cycle. Abbreviations: AGEs, advanced glycation end products; CKD, chronic kidney disease; GLO-1, glyoxalase 1.
Ijms 18 00174 g001

Share and Cite

MDPI and ACS Style

Hirakawa, Y.; Inagi, R. Glycative Stress and Its Defense Machinery Glyoxalase 1 in Renal Pathogenesis. Int. J. Mol. Sci. 2017, 18, 174. https://doi.org/10.3390/ijms18010174

AMA Style

Hirakawa Y, Inagi R. Glycative Stress and Its Defense Machinery Glyoxalase 1 in Renal Pathogenesis. International Journal of Molecular Sciences. 2017; 18(1):174. https://doi.org/10.3390/ijms18010174

Chicago/Turabian Style

Hirakawa, Yosuke, and Reiko Inagi. 2017. "Glycative Stress and Its Defense Machinery Glyoxalase 1 in Renal Pathogenesis" International Journal of Molecular Sciences 18, no. 1: 174. https://doi.org/10.3390/ijms18010174

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop