Abstract
Non-enzymatic post-translational modifications (nPTMs) of proteins have emerged as novel risk factors for the genesis and progression of various diseases. We now have a variety of experimental and established therapeutic strategies to target harmful nPTMs and potentially improve clinical outcomes. Protein carbamylation and glycation are two common and representative nPTMs that have gained considerable attention lately as favorable therapeutic targets with emerging clinical evidence. Protein carbamylation is associated with the occurrence of cardiovascular disease (CVD) and mortality in patients with chronic kidney disease (CKD); and advanced glycation end products (AGEs), a heterogeneous group of molecules produced in a series of glycation reactions, have been linked to various diabetic complications. Therefore, reducing the burden of protein carbamylation and AGEs is an appealing and promising therapeutic approach.
This review chapter summarizes potential anti-nPTM therapy options in CKD, CVD, and diabetes along with clinical implications. Using two prime examples—protein carbamylation and AGEs—we disucss the varied preventative and therapeutic options to mitigate these pathologic nPTMs in detail. We provide in-depth case studies on carbamylation in the setting of kidney disease and AGEs in metabolic disorders, with an emphasis on the relevance to reducing adverse clinical outcomes such as CKD progression, cardiovascular events, and mortality. Overall, whether specific efforts to lower carbamylation and AGE burden will yield definitive clinical improvement in humans remains largely to be seen. However, the scientific rationale for such pursuits is demonstrated herein.
Keywords: Carbamylation, Cardiovascular diseases, Diabetes mellitus, Glycation, Chronic kidney disease, Post-translational modification
1. Introduction
Proteins in the human body, during both health and disease, are exposed to a variety of chemical reactions capable of altering their active properties. Post-translational modifications (PTMs) of proteins are capable of altering protein charge, structure, and function in various physiological and pathophysiological conditions. The diversification of protein structures by PTMs provides an important means of protein regulation in human health. For example, phosphorylation is commonly required to activate the catalytic function of various enzymes (Walsh et al., 2005). Non-enzymatic post-translational modifications (nPTMs) of proteins are typically irreversible chemical reactions between electrophilic small molecules (endogenously or exogenously derived) and nucleophilic amino acid side chains or the N-terminus of proteins (Harmel and Fiedler, 2018). Without classical enzyme catalysis, nPTMs are usually spontaneous and not highly regulated. In recent years, multiple nPTMs have been found to play key roles in molecular aging and dysfunction (Gorisse et al., 2016), and have been implicated in various disease states such as chronic kidney disease (CKD) (Kalim et al., 2014), cardiovascular disease (CVD) (Verbrugge et al., 2015), diabetes mellitus (DM) (Singh et al., 2001), cancer (Sharma et al., 2019) and neurodegenerative disorders (Schaffert and Carter, 2020).
While discussion of the potential prevention and therapy of all known pathologic nPTMs is beyond the scope of any single review, there are clear areas that have been the primary focus of recent literature. Specifically, the nPTMs carbamylation (particularly in the context of kidney diseases) and glycation (particularly in the context of DM) have gained considerable attention. It is understandable that these entities have garnered such focus given the high prevalence of their commonly associated medical conditions in the general population and because therapeutic pathways appear primed for clinical study. Of note, one of the most common causes of CKD across the globe is DM (Alebiosu and Ayodele, 2005), and much of the morbidity and mortality associated with CKD and DM ultimately relates to cardiovascular causes (Sarnak et al., 2003). Thus, CVD, CKD and DM are often interlinked with each other, collectively acknowledged as cardio-renal-metabolic disease (Rangaswami et al., 2020). This review will look to potential avenues of carbamylation and glycation prevention and therapy across this broad axis. There are still many other PTMs, as noted above, with therapeutic potential in the early investigational stages and, therefore, considered beyond the scope of this review.
1.1. Overview of the carbamylation reaction
Carbamylation is defined as the non-enzymatic reaction between cyanate and amino or sulfhydryl groups of amino acids, peptides, or proteins (Delanghe et al., 2017). As shown in Fig. 1, urea or otherwise derived cyanate (which is in equilibrium with its tautomer and reactive form isocyanate), can spontaneously react with select sites on proteins. The net result of these reactions, which we are calling “carbamylation”, is the addition of a “carbamoyl” moiety (−CONH2) to a functional group. It must be noted that several authorities accurately state that the reaction in question is more correctly deemed “carbamoylation” and, in fact, “carbamylation” denotes a different chemical reaction (the reversible interaction of CO2 with α- and ε-amino groups of proteins) (Jelkmann, 2008). However, over the past decades, the biomedical literature has largely been calling the reaction involving a carbamoyl moiety, “carbamylation”, so we will continue in this manner for convention.
Figure 1.
Schematic representation of chemical genesis of protein carbamylation and potential therapeutic targets.
As kidney function declines, circulating levels of urea and cyanate increase in the human body, and thus so does carbamylation load (Pietrement et al., 2013; Vanholder et al., 2018). An alternative source of cyanate is thiocyanate or cyanide which can derive from diet or smoking and be catalyzed by myeloperoxidase (MPO), an enzyme present in the granules of neutrophils, monocytes and macrophages, at inflammatory sites (Delporte et al., 2018; Wang et al., 2007). Yet in the face of declining kidney function, the increase in urea is felt to be the dominant driver of carbamylation (Kalim et al., 2016, 2013).
Carbamylated proteins undergo structural and functional changes, leading to modified biological activity (Jaisson et al., 2011). Accumulation of carbamylated proteins has been described as a hallmark of molecular aging, and such proteins are involved in the pathogenesis of several conditions including atherosclerosis (Jaisson et al., 2015; Speer et al., 2014; Sun et al., 2016), thrombus formation (Binder et al., 2017; Holy et al., 2016) and renal fibrosis (Gross et al., 2011) for a few examples. Protein carbamylation also plays a role in the generation of abnormal tau protein deposition involved in the development of Alzheimer’s disease (Farías et al., 1997).
1.2. Overview of the non-enzymatic glycation reaction
Glycation is defined as the non-enzymatic reaction between reducing sugars or sugar derivatives and amino groups of amino acids, proteins, lipids or nucleic acids (Simm, 2013). Advanced glycation end products (AGEs) are a heterogeneous class of molecules or compounds that are derived from this process (Vlassara, 1997; Vlassara and Palace, 2003). With initial observations made by the French chemist Louis-Camille Maillard, it is common to see reference to the relevant reaction as the Maillard reaction (Maillard, 1912). This complex reaction involves a series of steps in which reducing sugars undergo non-enzymatic reactions that produce reactive carbonyl compounds and the subsequent glycoxidation of proteins, lipids and nucleic acids. AGEs are ultimately yielded when the glycation targets are rearranged to form stable heterogeneous compounds (Singh et al., 2001). The receptor for AGEs (RAGE) is a multi-ligand pattern-recognition receptor expressed in different cell types (Hudson and Lippman, 2018) and interaction of AGEs with RAGE can activate complex intracellular signaling cascades. RAGE independent effects of AGEs include change in molecular conformation and clearance, alteration of enzyme activity, and interference with receptor recognition (Aronson and Rayfield, 2002).
Notably, as DM is the leading cause of CKD, it is not surprising that the two nPTMs most closely related to these disease processes (non-enzymatic glycation and carbamylation) can co-occur. In vitro and animal studies suggest these two reactions have a competitive relationship with each other as they often target the same protein sites (Nicolas et al., 2019, 2018). However, at more intensive carbamylation states, competitive glycation appears less significant (Nicolas et al., 2019, 2018).
2. The relation between nPTMs and adverse clinical outcomes
As reviewed elsewhere in this series and briefly noted here, carbamylation and non-enzymatic glycation have been clearly and independently associated with adverse clinical outcomes in humans. A detailed accounting of all clinical association studies related to these nPTMs is beyond the scope of this review and it is important to note the difficulty in assigning weight to such diverse studies with varying sample sizes, study design, and even nPTM assay methods. Nevertheless, Table 1 highlights select consistent examples that provide compelling evidence that these nPTMs are associated with adverse clinical outcomes.
Table 1.
Patient-oriented studies linking nPTMs to adverse clinical outcomes
PTM | Biomarker (Measurement method) | Study design | Study population | Associated clinical outcomes | References |
---|---|---|---|---|---|
Carbamylation | Protein-bound homocitrulline (HPLC–MS/MS) | Case-control | 550 patients undergoing cardiac catheterization | MACE | (Wang et al., 2007) |
Cohort | 347 patients on HD | Mortality | (Koeth et al., 2013) | ||
Cohort | 111 patients with chronic systolic heart failure | Mortality; Cardiac transplantation | (Tang et al., 2013) | ||
Carbamylated albumin (HPLC–MS/MS) | Cohort | 187 patients on HD | Mortality | (Berg et al., 2013) | |
Cohort | 158 patients on HD | Erythropoietin resistance; mortality | (Kalim et al., 2013) | ||
Case-control | 366 patients on HD | Mortality | (Kalim et al., 2016) | ||
RCT post-hoc analysis | 1,161 patients on HD with diabetes | Cardiovascular mortality; Sudden cardiac death | (Drechsler et al., 2015) | ||
Case-control | 300 patients with CKD | CKD progression; Mortality | (Kalim et al., 2020) | ||
Glycation | Pentosidine (ELISA) | Case-control | 141 patients with heart failure | Cardiovascular mortality; Re-hospitalization | (Koyama et al., 2007) |
Pentosidine (HPLC–MS/MS) | Cohort | 746 patients with CKD | Mortality | (Machowska et al., 2016) | |
CML (ELISA) | Cohort | 81 patients with CAD | CAD progression | (Basta et al., 2008) | |
Cohort | 154 patients on HD | Mortality | (Wagner et al., 2006) | ||
Skin AF (AF reader) | Case-control | 109 patients on HD | CVD Mortality | (Meerwaldt et al., 2005) |
Abbreviations: AF, autofluorescence; CAD, coronary artery disease; CKD, chronic kidney disease; CML, Nε-carboxymethyl-lysine; ELISA, enzyme-linked immunosorbent assay; HD, hemodialysis; HPLC–MS/MS, high-performance liquid chromatography with tandem mass spectrometry; MACE, major adverse cardiac event; PTM, post-translational modification; RCT, randomized controlled trial.
2.1. Protein carbamylation and adverse clinical outcomes
The motivation for clinical outcome studies related to carbamylation has strong mechanistic underpinnings. Classic experiments have shown that when certain proteins are incubated with urea, a modified form of the protein accumulates that is less positively charged, and urea exposure results in lysine within the protein becoming irreversibly modified into N (6)-carbamoyl-L-lysine (what is known as homocitrulline). This carbamylation reaction yields a relatively stable moiety which does not readily dissociate or oxidize and proteins can accumulate these modifications on their N-terminal α–amino groups or the ε-amino groups of lysine side chains throughout their lifespan. A major chemical effect of carbamylation on protein amino groups is the neutralization of a positive charge which in turn alters ionic protein-water interactions on the protein surface (Beswick and Harding, 1987; Jaisson et al., 2018; Wistow et al., 1983). This disturbance can destabilize secondary and tertiary protein structures resulting in subsequent conformational changes. Dozens of studies have now shown results implicating carbamylation in changes of protein charge (Bobb and Hofstee, 1971; Legendre et al., 1998), conformation (Beswick and Harding, 1987; Monhemi and Tabaee, 2020; Nowicki and Santomé, 1981), and stability (Fazili et al., 1993), with consequent alterations in enzyme and hormone activity (De Furia et al., 1972; Oimomi et al., 1987; Shaw et al., 1964; Van Lente et al., 1986; Veronese et al., 1972), binding properties (Dengler et al., 1992; Lee and Gibson, 1981; Weisgraber et al., 1978), receptor-drug interaction (Erill et al., 1980; Smyth, 1967), and cellular expression and responses (Balion et al., 1998; Garnotel et al., 2004; Ha et al., 2010; Jaisson et al., 2007; Lane and Burka, 1976; Maddock and Westenfelder, 1996). These studies are just a few examples which highlight plausible mechanisms by which hyper-carbamylation could contribute to adverse clinical outcomes.
A few examples of specific pathophysiological pathways shown to accelerate due to carbamylation include atherosclerosis, vascular calcification, and fibrosis of kidney parenchyma. Carbamylation of low density lipoprotein (LDL) can enhance its atherogenic properties partly by decreasing its binding to the LDL-receptor (Wang et al., 2007) and by preventing its clearance from circulation, a phenomenon seen in both animal models of chronic renal failure (Hörkkö et al., 1992; Shapiro, 1993), as well as in uremic patients (Apostolov et al., 2010; Hörkkö et al., 1995, 1994; Shah et al., 2008). Carbamylated LDL has also been shown to readily bind macrophage scavenger receptors, facilitating foam cell formation and inflammatory signaling (Apostolov et al., 2009, 2007; Wang et al., 2007). Moreover, carbamylated LDL can induce endothelial cell apoptosis (Apostolov et al., 2007; Ok et al., 2005) and stimulate vascular smooth muscle proliferation (Asci et al., 2008) while carbamylated collagen increases release of matrix metalloproteinases by monocytes, possibly stimulating remodeling of the extracellular matrix (Garnotel et al., 2004; Jaisson et al., 2006).
Carbamylated proteins at concentrations present in uremia have similarly been shown to activate glomerular mesangial cells to a profibrogenic phenotype and stimulate collagen deposition (Shaykh et al., 1999). Cyanate exposure strongly inhibits the collagenase activity of human and rat mesangial cell MMP-2 (Gross et al., 2011). In general, it is suggested that carbamylation of collagen or enzymes involved in extracellular-matrix remodeling can disrupt the balanced remodeling of extracellular proteins and enhance fibrosis (Jaisson et al., 2007; Kraus et al., 2001). Additionally, carbamylated albumin results in renal tubular cell damage and peritubular fibrosis via stimulation of TGF-β, EGF, NF-κB and endothelin-1 in the amphibian kidney (Gross et al., 2011). Carbamylation of high-density lipoprotein (HDL) can impact migration, angiogenesis, and proliferation in endothelial cells (Holzer et al., 2011; Sun et al., 2016), while other studies suggest cyanate itself has direct pathologic effects on endothelial cells (El-Gamal et al., 2012; Xiao et al., 2001). Similar mechanistic links to endothelial dysfunction can be seen through carbamylation’s contribution to oxidative stress. For example, S-carbamylation of free cysteine and glutathione sulfhydryl groups as well as of hydrogen sulfide interferes with their antioxidant functions (e.g. free radical scavenging) (Praschberger et al., 2013; Schreier et al., 2011). Most recently, carbamylation’s mechanistic role in vascular disease and calcification has been elucidated through in vitro and in vivo studies of carbamylated sortilin, elastin, uromodulin, and mitochondrial proteins all resulting in increased vascular pathology (Alesutan et al., 2021; Doué et al., 2021; Jankowski et al., 2021; Mori et al., 2018).
There are a few biomarkers that have been used to assess and quantify carbamylation burden, including homocitrulline (Jaisson et al., 2016, 2012), carbamylated albumin (Berg et al., 2013), carbamylated hemoglobin (Balion et al., 1998) and carbamylated LDL (Apostolov et al., 2005). Most of the biomarkers are quantified using high-performance liquid chromatography (HPLC) with online tandem mass spectrometry (MS/MS). HPLC-MS/MS confers robust measurement with high precision and reproducibility. For example, the assay of carbamylated albumin quantified using this method has excellent linearity with a coefficient of variation of 4.2% (Berg et al., 2013). However, their use in routine clinical practice is currently limited by time-consuming techniques and expensive equipment (Jaisson et al., 2018). If the clinical utility of carbamylation measures eventually becomes established, the development of efficient, reliable, and cost-effective assays will need to follow.
With such mechanistic underpinnings and available biomarkers, several patient-oriented studies (Table 1) have unsurprisingly shown that carbamylation is an independent risk factor for adverse clinical outcomes including CVD, CKD progression, and mortality in patients with CKD as well as normal kidney function (Kalim et al., 2020, 2016; Tang et al., 2013; Wang et al., 2007). Moreover, carbamylation was associated with CVD and mortality in end-stage renal disease (ESRD) as well (Berg et al., 2013; Drechsler et al., 2015; Koeth et al., 2013).
2.2. AGEs and adverse clinical outcomes
Similarly, the accumulation of AGEs alters the structure and function of proteins, contributing to the pathogenesis of metabolic diseases such as diabetes through a variety of mechanisms (Chaudhuri et al., 2018). As an example, after AGEs bind to RAGE, NF-kB and MAPK signaling pathways are activated, leading to inflammation and endothelial dysfunction (Sadik et al., 2012). AGEs are also implicated in the pathogenesis of neurodegenerative disease (Münch et al., 2012); for example, AGE formation has been found at the periphery of Lewy bodies in patients with Parkinson’s disease (Castellani et al., 1996).
Although markers for glucose control such as hemoglobin A1c are well-established and commonly used in clinical practice (Speeckaert et al., 2014), the biomarkers used to indicate and assess the burden of AGEs based on downstream products of glucose metabolism are only emerging (Chaudhuri et al., 2018). Several patient-oriented studies (examples in Table 1) have shown that AGEs measured by various biomarkers are independent risk factors for adverse clinical outcomes including CKD progression, CVD, and mortality in patients with or without diabetes mellitus (Koyama et al., 2007). Tissue AGE accumulation reflected by skin autofluorescence was associated with vascular calcification (Wang et al., 2014) and predicts mortality in CKD (Machowska et al., 2016) and ESRD patients (Meerwaldt et al., 2005; Wagner et al., 2006).
3. Prevention and Therapy
The identification and characterization of these harmful, mechanistically relevant, nPTMs, set them up as promising therapeutic targets in various disease states, especially CVD, CKD, and DM. There is an increasing body of evidence suggesting that protein carbamylation and AGEs may serve as potential preventative and therapeutic targets to reduce the morbidity and mortality from the relevant cardio-renal-metabolic diseases.
3.1. Prevention and therapy targeting carbamylation
Compelling evidence from animal data and epidemiological studies discussed above have suggested carbamylation may play an important role in a plethora of biochemical alterations with clinical manifestations in renal disease. Given carbamylation is established as an independent mechanistically relevant risk factor for adverse clinical outcomes that is readily quantifiable, it is reasonable to hypothesize that targeting carbamylation in CKD and ESRD may improve clinical outcomes. There are a variety of theoretical and practical strategies proposed to reduce the rate and burden of carbamylation (Fig. 1). These pathways include reducing the source of carbamylation’s cyanate—urea—by more intensive dialysis treatment, kidney transplantation, or reducing dietary protein intake; increasing free amino acid scavengers in circulation (which can react with cyanate to prevent the same reaction from occurring on a protein instead); or alternative therapies to block carbamylation’s chemical reaction from occurring. Notably, to date, the common limitations of studies pursuing such strategies include relatively small sample sizes and the lack of hard clinical outcomes (e.g. mortality or CKD progression). In the future, large, randomized control trials to study clinically meaningful outcomes will be needed.
3.1.1. Urea reduction
3.1.1.1. Dialysis intensification and kidney transplantation
Initiation of dialysis in patients with advanced CKD has been shown to reduce protein carbamylation substantially from pre-dialysis advanced CKD levels. This is felt largely due to enhanced urea clearance via dialysis as well likely an improvement in overall uremic symptoms and thus improvement in nutritional status and amino acid balance (Kalim et al., 2018, 2016). However, there is still an excessive carbamylation burden observed in ESRD patients on routine dialysis treatment, raising the question of whether an intensified dialysis treatment prescription could further reduce carbamylation. A prospective study with 53 ESRD patients investigated the impact of in-center nocturnal extended duration dialysis on the burden of carbamylation and cardiac structure and found that compared with standard dialysis (3–4 hours per session, 3 times per week), extended dialysis (7–8 hours per session, 3 times per week) was associated with reduction of carbamylation levels as assayed by carbamylated albumin at 1 year. Such carbamylation reduction correlated with urea reduction and was associated with reduced left ventricular mass measured by cardiac MRI (Perl et al., 2016). Challenges to the approach of intensified dialysis included patient reluctance to undergo additional hours of treatment each week which is often viewed as a significant adverse impact on quality of life. However, with the changing landscape of favoring home dialysis over in-center dialysis under the Advancing American Kidney Health initiative (Himmelfarb et al., 2020), the implementation of extended duration dialysis could become easier.
Looking beyond dialysis, kidney transplantation is the treatment of choice for most patients with ESRD. A successful kidney transplant improves quality of life and reduces the mortality risk for the majority of patients when compared with maintenance dialysis (Wolfe et al., 1999). The impact of kidney transplant on carbamylation has not been robustly studied, but given carbamylation’s close correlation to eGFR, it stands to reason that as eGFR is restored post-transplant, carbamylation load would decrease. This was briefly studied by Han et al, who showed the carbamylated hemoglobin concentration of 6 patients decreased by 19.7% within 2 to 3 weeks after kidney transplantation (Han et al., 1997). However, the true in vivo kinetics were less clear as there was also an average hemoglobin increase of 25% which may have confounded what the direct effects of restored solute clearance had on carbamylation.
3.1.1.2. Dietary modification
Another strategy to reduce urea in pre-dialysis CKD patients is dietary modifications. A Mediterranean diet and a very low protein diet have been shown beneficial in the reduction of urea burden and decreasing carbamylation. In a randomized crossover controlled trial, 60 patients with advanced CKD were assigned to very low protein diet supplemented with keto-analogues or Mediterranean diet or “free diet”; compared to a “free diet”, both the very low protein and Mediterranean diets were associated with decreases in serum homocitrulline levels and such reduction correlated with urea reduction (Di Iorio et al., 2018). Keto-analogues are a very attractive carbamylation therapy as they are amino acid precursors that utilize an amino group from urea to form amino acids (thus reducing urea/ cyanate and increasing carbamylation scavengers simultaneously). However, dietary modification is challenging for many patients to maintain, and strong motivation is required. Moreover, competing dietary demands from comorbid conditions such as diabetes, limited medical practitioner knowledge of specific dietary recommendations, and complicated psycho-socio-economic, behavioral, and cultural factors all can pose challenges to routine dietary interventions. Nevertheless, the evidence suggests that a low-protein diet supplemented with keto-analogues can significantly reduce carbamylation burden. It is not clear what the relative contribution of the low protein diet vs. the keto-analogue supplementation makes to reducing carbamylation and teasing apart these two interventions is an area for future research as is linking such intervention to clinical outcomes. To that end, a contemporary randomized clinical trial in CKD patients recently showed considerable improvements in the outcome of 50% reduction in eGFR or reaching ESRD with a very low protein diet supplemented with keto-analogues (compared to just a low protein diet), yet the underlying mechanisms for this result were not clear and could not be attributed to changes in nutritional parameters alone (Garneata et al., 2016). Perhaps, those who benefited responded to lower urea, restored amino acid concentrations, and subsequently reduced protein carbamylation. Effect sizes might be greater and more consistent if such studies included only subjects with accelerated protein carbamylation were targeted.
3.1.2. Amino acid supplementation
As mentioned above, free amino acids can compete with proteins to react with cyanate and thus reduce protein carbamylation by serving as “scavengers” (Berg et al., 2013). However, patients with advanced CKD and ESRD are often deficient in free amino acids due to various reasons, including poor nutritional status and loss of amino acids in the dialysate (Duranton et al., 2014; Ikizler et al., 1994). There are different means of repleting amino acids, including intravenous, intraperitoneal, and oral/ dietary approaches. Further work is needed to determine the best dose, route, and type of amino acids to reduce carbamylation and maximize the efficacy, feasibility and safety for CKD patients at different stages.
3.1.2.1. Intravenous amino acid supplementation
In a prospective pilot study, 12 hemodialysis patients who received 250cc of intravenous (IV) amino acids which contained 14g of essential amino acids 3 times per week post-dialysis over 8 weeks had significantly reduced carbamylated albumin levels at 4 weeks and 8 weeks, compared to 11 hemodialysis control patients (Kalim et al., 2015). Importantly there were no safety concerns with this approach and notably the dosing did not result in higher blood urea nitrogen levels in the treated individuals (a concern when providing significant amounts of exogenous nitrogen). Such a strategy is appealing as it could potentially also improve proteinenergy malnutrition, another common morbidity in ESRD populations. Another trial to study the effect of IV amino acids in ESRD patients, CarRAAT-2 (Trial of Carbamylation in Renal Disease-Modulation With Amino Acid Therapy; NCT02472834) has been completed with results anticipated soon.
3.1.1.2. Intraperitoneal amino acid supplementation
In a secondary analysis from the IMPENDIA trial (a randomized trial studying the effect of low-glucose peritoneal dialysis solution in DM) (Li et al., 2013), patients maintained on peritoneal dialysis were treated with either icodextrin and intraperitoneal amino acids, or dextrose only peritoneal dialysis solutions. The intraperitoneal amino acid group did not show reduced carbamylated albumin levels (Trottier et al., 2018). However, the study was limited by the small sample size and lack of generalizability due to the inclusion of diabetic patients only. Overall, this study suggested that intraperitoneal amino acid administration might not be the best approach to mitigate carbamylation. An additional finding from this study was that compared to a cohort of similar hemodialysis patients, urea levels on average were higher in the peritoneal dialysis group and this correlated with greater carbamylation levels. It was postulated that perhaps any benefits or advantages of peritoneal dialysis over hemodialysis may be blunted by a greater urea-driven carbamylation load in the peritoneal dialysis group.
3.1.2.3. Oral amino acid supplementation
We can also give oral protein and/or amino acid supplementations (or keto-analogue supplementation as detailed above), both in non-dialysis CKD patients and dialysis patients. For non-dialysis CKD patients, it is not easy to administrate IV amino acids routinely. Therefore, an oral option becomes very appealing. However, the study of oral nutritional and amino acid supplementation has focused mainly on protein and energy homeostasis in dialysis patients (Lacson et al., 2012; Weiner et al., 2014), with no insights into the effects on carbamylation. While such studies have shown mortality benefits in dialysis patients, similar to the benefits seen in low protein diets supplemented with keto-analogues (Garneata et al., 2016), the mechanisms of such benefits remain unclear. As above, effect sizes may be greater and more consistent across studies if therapies targeted carbamylation levels. Additional studies investigating the effect of oral amino acid and/or keto-analogue supplementation on carbamylation burden and clinical outcomes in CKD patients are needed to clarify this.
3.1.3. Investigative paths to attenuate carbamylation reactions
Carbamylation occurs naturally (at low levels in the absence of CKD or other drivers) but can accumulate on proteins with long half-lives. The accumulation rate of carbamylation over time in the skin of different mammalian species with different life expectancies is inversely correlated with longevity (Gorisse et al., 2016). This suggests that some animals may harbor potential protective mechanisms against carbamylation that are yet to be identified. Similar speculation of an ability to guard against carbamylation’s harmful effects has also been pondered in the context of elasmobranch physiology (sharks, rays, and skates). Relative to humans, these animals have extremely high blood urea concentrations (350–500 mM) to maintain osmotic balance with a marine environment yet have several homologous proteins to humans that do not appear to malfunction in such a milieu. How these animals handle high urea levels without enduring adverse effects is not entirely known (Delanghe et al., 2017; Yancey et al., 1982). The underlying mechanism is still speculative and thought to be related to a coexisting protective effect of methylamines including trimethylamine N-oxide (TMAO), betaine, glycerylphosphorylcholine and sarcosine, which can offset the harmful effects of urea on proteins (Ballantyne, 2016; Lee et al., 1991). This is an important area for future research as the understanding of the underlying mechanism carries large clinical implications for any attempts to reduce carbamylation either nutritionally or pharmacologically in CKD patients with high urea levels.
Experimental studies have also shown various agents successful at blocking carbamylation, but these are yet to be assessed in humans. Therapies including ascorbic acid, α-tocopherol, lycopene, and flavonoids all inhibited LDL carbamylation in vitro (Ghaffari and Shanaki, 2010a, 2010b). Eicosapentaenoic acid supplementation in mice decreased mitochondrial protein carbamylation (Johnson et al., 2015), and triclosan similarly inhibited carbamylation (Bright et al., 2018). Aspirin, ibuprofen, and bendazac have all been shown to inhibit carbamylation events that commonly pathologically occur on lens proteins (Crompton et al., 1985; Lewis et al., 1986; Plater et al., 1997; Rao and Cotlier, 1988). Again, these studies are either in vitro or animal studies, and have not been translated to clinical applications and serve as areas for future investigation.
3.2. Prevention and therapy targeting AGE
Chronic hyperglycemia, characteristic of DM, promotes the formation of AGEs and thus, like reducing urea levels to prevent carbamylation, optimal glycemic control is a cornerstone of preventing accelerated AGE formation. Moreover, several medications including antidiabetic medications, blood pressure medications, and statins appear to block Maillard reactions to varying degrees and thereby reduce the development of AGE in addition to their primary respective function (Jud and Sourij, 2019). However, despite the wide availability and variety of traditional diabetes drugs and the emergence of novel antihyperglycemic agents such as SGLT2 inhibitors and GLP-1 agonists (Brown et al., 2021), the morbidity caused by diabetes including retinopathy, nephropathy, and accelerated CVD is still unacceptably high (Centers for Disease Control and Prevention, 2020). Therefore, therapies to more specifically target AGEs and prevent and reduce diabetic-related end-organ damage are crucial. Since AGE formation is complex and involves many steps, AGE therapy is challenging (Dariya and Nagaraju, 2020). Possible pathways include AGE inhibitors, AGE cross-link breakers and RAGE antagonists. There are also dietary factors and phytochemicals that target AGE pathways through various mechanisms. Many of the following therapies are still limited by a lack of proven clinical efficacy or the presence of safety concerns.
3.2.1. AGE inhibitors
AGE inhibitors prevent AGE formation and cross-linking. Aminoguanidine, a guanidine derivative, acts as a dicarbonyl scavenger to prevent the formation of AGEs (Thornalley, 2003). It was studied in the setting of diabetic nephropathy in the ACTION I trial (A Clinical Trial In Overt Nephropathy of Type 1 Diabetics) where subjects in the aminoguanidine group had slower estimated glomerular filtration rate (eGFR) decline, reduced proteinuria, and improved lipid profiles (Bolton et al., 2004). However, the subsequent ACTION II trial (Aminoguanidine Clinical Trial in Overt Type 2 Diabetic Nephropathy) was stopped early due to futility and safety concerns with the intervention including flu-like symptoms, ANCA vasculitis, and elevated liver enzymes (Freedman et al., 1999). Pyridoxamine, an amine of vitamin B6, acts as a chelator of redox metal ions catalyzing part of Maillard reactions (Voziyan et al., 2003). It was also studied in the setting of diabetic nephropathy. A double-blind, randomized, placebo-controlled trial enrolling 317 patients with type 2 diabetic nephropathy failed to show reno-protective effects of pyridoxamine dihydrochloride (Lewis et al., 2012).
3.2.2. AGE cross-link breakers
AGE cross-link breakers target the cross-links formed between AGEs and other extracellular molecules. Alagebrium, a thiazolium derivative, is the most extensively studied AGE cross-link breaker (Toprak and Yigitaslan, 2019). In a multi-center randomized trial conducted in the US, 93 elderly patients with hypertension and vascular stiffening were enrolled, and the alagebrium group was shown to have improved total arterial compliance compared to placebo (Kass et al., 2001). In another single-arm trial, 23 patients with heart failure with preserved ejection fraction treated with alagebrium showed reduced left ventricular mass and a trend towards improvement in left ventricular diastolic function assessed by E/e’ at 16 weeks compared to baseline (Little et al., 2005). In contrast, in another well-designed randomized, double-blind, placebo-controlled study enrolling 120 patients with heart failure with reduced ejection fraction, the alagebrium group did not show improvement in overall exercise tolerance or systolic dysfunction at 36 weeks (Hartog et al., 2011). Overall, alagebrium seems well tolerated in all the studies without major side effects. Unfortunately, further clinical trials have been terminated due to financial difficulties (Toprak and Yigitaslan, 2019). Currently, there are ongoing trials to evaluate the efficacy and safety of other AGE cross-link breakers in patients with diabetes and heart failure, such as TRC 041266 (NCT04507347).
3.2.3. RAGE antagonists
Blocking RAGE signaling has been shown to reduce downstream inflammation and progression of diabetic vascular complications (Hudson and Lippman, 2018). Soluble RAGE (sRAGE), a recombinant form of RAGE comprising the extracellular region, has been demonstrated to reduce the size and complexity of atherosclerotic lesions (Park et al., 1998), diminish albuminuria with improved renal function (Wendt et al., 2003), and enhance wound healing (Goova et al., 2001) in diabetic rats. However, due to a large size, it is difficult and expensive to produce at therapeutic levels, making it less ideal in clinical use (Kim et al., 2021). Small-molecular RAGE inhibitors which are designed to target the extracellular ligand-binding site of RAGE or block the intracellular signaling pathway seem a more feasible choice. One of them, TTP 488, also known as PF-04494700 or azeliragon, has been tested in multiple clinical trials of patients with Alzheimer’s disease. In 2007, a phase 2 trial recruited 399 patients with mild to moderate Alzheimer’s was terminated early due to safety signals in the high dose group and concern for futility in the low dose group (Burstein et al., 2014). However, subsequent analysis on the follow-up data suggested a possible slower cognitive decline in the low dose group (Galasko et al., 2014). Subsequently, a Phase 3 trial of TTP 488, STEADFAST (Evaluation of the Efficacy and Safety of TTP488 in Patients With Mild Alzheimer’s Disease; NCT02080364), failed to meet its co-primary endpoint of cognitive assessment scores.
3.2.4. Diet and phytochemicals to reduce AGEs
Food is an exogenous source of AGEs. Frying or cooking with high temperature makes food more susceptible to Maillard reactions and thus higher AGE concentrations (Santos and Penha-Silva, 2021). In a randomized crossover dietary interventional trial with 62 healthy adults, consuming a diet prepared at high temperature increased insulin resistance and cholesterol and triglyceride levels compared with a diet prepared through steaming (Birlouez-Aragon et al., 2010). In a meta-analysis of 13 randomized clinical trials, a low-AGE diet improved cardiometabolic parameters, including reduction in insulin resistance, total cholesterol, and low-density lipoprotein (Sohouli et al., 2021). There are many medicinal plants and their derived phytochemicals that have been shown to have anti-AGE activities based on in vitro or animal studies (Parveen et al., 2021). Some examples include apigenin (Zhou et al., 2019), curcumin (Lin et al., 2012), and epigallocatechin 3-gallate (Sampath et al., 2017). They pose future opportunities in the multifaceted therapeutic approach to prevent and treat AGE-mediated diseases. Many additional excellent reviews on this specific topic exist (Guilbaud et al., 2016; Tessier and Birlouez-Aragon, 2012).
4. Conclusions
This review discussed the status of multiple preventative and therapeutic strategies targeting at nPTMs in various diseases. Decades of basic science research with mechanistic studies and animal models have contributed a rich literature suggesting the pathophysiological roles of nPTMs in different disease states. In more recent years, epidemiological studies have shown the mechanistically informed adverse associations of nPTMs to clinical outcomes. Now, a variety of therapeutic approaches aimed at reducing nPTMs are being translated into potential clinical strategies to prevent and treat several different diseases processes. Moving forward, we next need more in human clinical trials with clinical endpoints to say for certain whether nPTM therapies can deliver on their promise to improve patient outcomes.
Acknowledgments
This work was supported by National Institutes of Health grant R01 DK124453 to SK.
Footnotes
Publisher's Disclaimer: This is a PDF file of an unedited manuscript that has been accepted for publication. As a service to our customers we are providing this early version of the manuscript. The manuscript will undergo copyediting, typesetting, and review of the resulting proof before it is published in its final form. Please note that during the production process errors may be discovered which could affect the content, and all legal disclaimers that apply to the journal pertain.
References:
- Alebiosu CO, Ayodele OE, 2005. The global burden of chronic kidney disease and the way forward. Ethn Dis 15, 418–423. [PubMed] [Google Scholar]
- Alesutan I, Luong TTD, Schelski N, Masyout J, Hille S, Schneider MP, Graham D, Zickler D, Verheyen N, Estepa M, Pasch A, Maerz W, Tomaschitz A, Pilz S, Frey N, Lang F, Delles C, Müller OJ, Pieske B, Eckardt K-U, Scherberich J, Voelkl J, 2021. Circulating uromodulin inhibits vascular calcification by interfering with pro-inflammatory cytokine signalling. Cardiovasc Res 117, 930–941. 10.1093/cvr/cvaa081 [DOI] [PubMed] [Google Scholar]
- Apostolov EO, Basnakian AG, Yin X, Ok E, Shah SV, 2007. Modified LDLs induce proliferation-mediated death of human vascular endothelial cells through MAPK pathway. Am J Physiol Heart Circ Physiol 292, H1836–1846. 10.1152/ajpheart.01079.2006 [DOI] [PubMed] [Google Scholar]
- Apostolov EO, Ray D, Savenka AV, Shah SV, Basnakian AG, 2010. Chronic Uremia Stimulates LDL Carbamylation and Atherosclerosis. JASN 21, 1852–1857. 10.1681/ASN.2010040365 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Apostolov EO, Shah SV, Ok E, Basnakian AG, 2005. Quantification of Carbamylated LDL in Human Sera by a New Sandwich ELISA. Clinical Chemistry 51, 719–728. 10.1373/clinchem.2004.044032 [DOI] [PubMed] [Google Scholar]
- Apostolov EO, Shah SV, Ray D, Basnakian AG, 2009. Scavenger receptors of endothelial cells mediate the uptake and cellular proatherogenic effects of carbamylated LDL. Arterioscler Thromb Vasc Biol 29, 1622–1630. 10.1161/ATVBAHA.109.189795 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Aronson D, Rayfield EJ, 2002. How hyperglycemia promotes atherosclerosis: molecular mechanisms. Cardiovasc Diabetol 1, 1. 10.1186/1475-2840-1-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Asci G, Basci A, Shah SV, Basnakian A, Toz H, Ozkahya M, Duman S, Ok E, 2008. Carbamylated low-density lipoprotein induces proliferation and increases adhesion molecule expression of human coronary artery smooth muscle cells. Nephrology (Carlton) 13, 480–486. 10.1111/j.1440-1797.2008.00948.x [DOI] [PubMed] [Google Scholar]
- Balion CM, Draisey TF, Thibert RJ, 1998. Carbamylated hemoglobin and carbamylated plasma protein in hemodialyzed patients. Kidney International 53, 488–495. 10.1046/j.1523-1755.1998.00777.x [DOI] [PubMed] [Google Scholar]
- Ballantyne JS, 2016. Some of the most interesting things we know, and don’t know, about the biochemistry and physiology of elasmobranch fishes (sharks, skates and rays). Comparative Biochemistry and Physiology Part B: Biochemistry and Molecular Biology 199, 21–28. 10.1016/j.cbpb.2016.03.005 [DOI] [PubMed] [Google Scholar]
- Basta G, Berti S, Cocci F, Lazzerini G, Parri S, Papa A, Battaglia D, Lubrano V, Del Turco S, Ravani M, Rizza A, De Caterina R, Marraccini P, Mazzone A, 2008. Plasma N-epsilon-(carboxymethyl)lysine levels are associated with the extent of vessel injury after coronary arterial stenting. Coron Artery Dis 19, 299–305. 10.1097/MCA.0b013e3282fec058 [DOI] [PubMed] [Google Scholar]
- Berg AH, Drechsler C, Wenger J, Buccafusca R, Hod T, Kalim S, Ramma W, Parikh SM, Steen H, Friedman DJ, Danziger J, Wanner C, Thadhani R, Karumanchi SA, 2013. Carbamylation of Serum Albumin as a Risk Factor for Mortality in Patients with Kidney Failure. Sci. Transl. Med 5. 10.1126/scitranslmed.3005218 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Beswick HT, Harding JJ, 1987. High-molecular-weight crystallin aggregate formation resulting from non-enzymic carbamylation of lens crystallins: relevance to cataract formation. Exp Eye Res 45, 569–578. 10.1016/s0014-4835(87)80067-2 [DOI] [PubMed] [Google Scholar]
- Binder V, Bergum B, Jaisson S, Gillery P, Scavenius C, Spriet E, Nyhaug A, Roberts H, Chapple I, Hellvard A, Delaleu N, Mydel P, 2017. Impact of fibrinogen carbamylation on fibrin clot formation and stability. Thromb Haemost 117, 899–910. 10.1160/TH16-09-0704 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Birlouez-Aragon I, Saavedra G, Tessier FJ, Galinier A, Ait-Ameur L, Lacoste F, Niamba C-N, Alt N, Somoza V, Lecerf J-M, 2010. A diet based on high-heat-treated foods promotes risk factors for diabetes mellitus and cardiovascular diseases. Am J Clin Nutr 91, 1220–1226. 10.3945/ajcn.2009.28737 [DOI] [PubMed] [Google Scholar]
- Bobb D, Hofstee BH, 1971. Gel isoelectric focusing for following the successive carbamylations of amino groups in chymotrypsinogen A. Anal Biochem 40, 209–217. 10.1016/0003-2697(71)90094-7 [DOI] [PubMed] [Google Scholar]
- Bolton WK, Cattran DC, Williams ME, Adler SG, Appel GB, Cartwright K, Foiles PG, Freedman BI, Raskin P, Ratner RE, Spinowitz BS, Whittier FC, Wuerth J-P, 2004. Randomized Trial of an Inhibitor of Formation of Advanced Glycation End Products in Diabetic Nephropathy. Am J Nephrol 24, 32–40. 10.1159/000075627 [DOI] [PubMed] [Google Scholar]
- Bright R, Marchant C, Bartold PM, 2018. The effect of triclosan on posttranslational modification of proteins through citrullination and carbamylation. Clin Oral Invest 22, 487–493. 10.1007/s00784-017-2137-8 [DOI] [PubMed] [Google Scholar]
- Brown E, Heerspink HJL, Cuthbertson DJ, Wilding JPH, 2021. SGLT2 inhibitors and GLP-1 receptor agonists: established and emerging indications. The Lancet 398, 262–276. 10.1016/S0140-6736(21)00536-5 [DOI] [PubMed] [Google Scholar]
- Burstein AH, Grimes I, Galasko DR, Aisen PS, Sabbagh M, Mjalli AM, 2014. Effect of TTP488 in patients with mild to moderate Alzheimer’s disease. BMC Neurol 14, 12. 10.1186/1471-2377-14-12 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Castellani R, Smith MA, Richey PL, Perry G, 1996. Glycoxidation and oxidative stress in Parkinson disease and diffuse Lewy body disease. Brain Res 737, 195–200. 10.1016/0006-8993(96)00729-9 [DOI] [PubMed] [Google Scholar]
- Centers for Disease Control and Prevention, 2020. National Diabetes Statistics Report 2020. Estimates of diabetes and its burden in the United States. National Diabetes Statistics Report 32. [Google Scholar]
- Chaudhuri J, Bains Y, Guha S, Kahn A, Hall D, Bose N, Gugliucci A, Kapahi P, 2018. The Role of Advanced Glycation End Products in Aging and Metabolic Diseases: Bridging Association and Causality. Cell Metabolism 28, 337–352. 10.1016/j.cmet.2018.08.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Crompton M, Rixon KC, Harding JJ, 1985. Aspirin prevents carbamylation of soluble lens proteins and prevents cyanate-induced phase separation opacities in vitro: a possible mechanism by which aspirin could prevent cataract. Exp Eye Res 40, 297–311. 10.1016/0014-4835(85)90014-4 [DOI] [PubMed] [Google Scholar]
- Dariya B, Nagaraju GP, 2020. Advanced glycation end products in diabetes, cancer and phytochemical therapy. Drug Discovery Today 25, 1614–1623. 10.1016/j.drudis.2020.07.003 [DOI] [PubMed] [Google Scholar]
- De Furia FG, Miller DR, Cerami A, Manning JM, 1972. The effects of cyanate in vitro on red blood cell metabolism and function in sickle cell anemia. J Clin Invest 51, 566–574. 10.1172/JCI106845 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Delanghe S, Delanghe JR, Speeckaert R, Van Biesen W, Speeckaert MM, 2017. Mechanisms and consequences of carbamoylation. Nat Rev Nephrol 13, 580–593. 10.1038/nrneph.2017.103 [DOI] [PubMed] [Google Scholar]
- Delporte C, Zouaoui Boudjeltia K, Furtmüller PG, Maki RA, Dieu M, Noyon C, Soudi M, Dufour D, Coremans C, Nuyens V, Reye F, Rousseau A, Raes M, Moguilevsky N, Vanhaeverbeek M, Ducobu J, Nève J, Robaye B, Vanhamme L, Reynolds WF, Obinger C, Van Antwerpen P, 2018. Myeloperoxidase-catalyzed oxidation of cyanide to cyanate: A potential carbamylation route involved in the formation of atherosclerotic plaques? Journal of Biological Chemistry 293, 6374–6386. 10.1074/jbc.M117.801076 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Dengler TJ, Robertz-Vaupel GM, Dengler HJ, 1992. Albumin binding in uraemia: quantitative assessment of inhibition by endogenous ligands and carbamylation of albumin. Eur J Clin Pharmacol 43, 491–499. 10.1007/BF02285090 [DOI] [PubMed] [Google Scholar]
- Di Iorio BR, Marzocco S, Bellasi A, De Simone E, Dal Piaz F, Rocchetti MT, Cosola C, Di Micco L, Gesualdo L, 2018. Nutritional therapy reduces protein carbamylation through urea lowering in chronic kidney disease. Nephrology Dialysis Transplantation 33, 804–813. 10.1093/ndt/gfx203 [DOI] [PubMed] [Google Scholar]
- Doué M, Okwieka A, Berquand A, Gorisse L, Maurice P, Velard F, Terryn C, Molinari M, Duca L, Piétrement C, Gillery P, Jaisson S, 2021. Carbamylation of elastic fibers is a molecular substratum of aortic stiffness. Scientific Reports 11, 17827. 10.1038/s41598-021-97293-5 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Drechsler C, Kalim S, Wenger JB, Suntharalingam P, Hod T, Thadhani RI, Karumanchi SA, Wanner C, Berg AH, 2015. Protein carbamylation is associated with heart failure and mortality in diabetic patients with end-stage renal disease. Kidney International 87, 1201–1208. 10.1038/ki.2014.429 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Duranton F, Lundin U, Gayrard N, Mischak H, Aparicio M, Mourad G, Daurès J-P, Weinberger KM, Argilés À, 2014. Plasma and Urinary Amino Acid Metabolomic Profiling in Patients with Different Levels of Kidney Function. CJASN 9, 37–45. 10.2215/CJN.06000613 [DOI] [PMC free article] [PubMed] [Google Scholar]
- El-Gamal D, Holzer M, Gauster M, Schicho R, Binder V, Konya V, Wadsack C, Schuligoi R, Heinemann A, Marsche G, 2012. Cyanate Is a Novel Inducer of Endothelial ICAM-1 Expression. Antioxid Redox Signal 16, 129–137. 10.1089/ars.2011.4090 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Erill S, Calvo R, Carlos R, 1980. Plasma protein carbamylation and decreased acidic drug protein binding in uremia. Clin Pharmacol Ther 27, 612–618. 10.1038/clpt.1980.87 [DOI] [PubMed] [Google Scholar]
- Farías G, González-Billault C, Maccioni RB, 1997. Immunological characterization of epitopes on tau of Alzheimer’s type and chemically modified tau. Mol Cell Biochem 168, 59–66. 10.1023/a:1006838626730 [DOI] [PubMed] [Google Scholar]
- Fazili KM, Mir MM, Qasim MA, 1993. Changes in protein stability upon chemical modification of lysine residues of bovine serum albumin by different reagents. Biochem Mol Biol Int 31, 807–816. [PubMed] [Google Scholar]
- Freedman BI, Wuerth J-P, Cartwright K, Bain RP, Dippe S, Hershon K, Mooradian AD, Spinowitz BS, 1999. Design and Baseline Characteristics for the Aminoguanidine Clinical Trial in Overt Type 2 Diabetic Nephropathy (ACTION II). Controlled Clinical Trials 20, 493–510. 10.1016/S0197-2456(99)00024-0 [DOI] [PubMed] [Google Scholar]
- Galasko D, Bell J, Mancuso JY, Kupiec JW, Sabbagh MN, van Dyck C, 2014. Clinical trial of an inhibitor of RAGE-Ab interactions in Alzheimer disease 7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garneata L, Stancu A, Dragomir D, Stefan G, Mircescu G, 2016. Ketoanalogue-Supplemented Vegetarian Very Low–Protein Diet and CKD Progression. JASN 27, 2164–2176. 10.1681/ASN.2015040369 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Garnotel R, Sabbah N, Jaisson S, Gillery P, 2004. Enhanced activation of and increased production of matrix metalloproteinase-9 by human blood monocytes upon adhering to carbamylated collagen. FEBS Lett 563, 13–16. 10.1016/S0014-5793(04)00233-9 [DOI] [PubMed] [Google Scholar]
- Ghaffari MA, Shanaki M, 2010a. In vitro inhibition of low density lipoprotein carbamylation by vitamins, as an ameliorating atherosclerotic risk in uremic patients. Scandinavian Journal of Clinical and Laboratory Investigation 70, 122–127. 10.3109/00365511003624137 [DOI] [PubMed] [Google Scholar]
- Ghaffari MA, Shanaki M, 2010b. Evalution of in vitro effect of flavonoids on human low-density lipoprotein carbamylation. Iran J Pharm Res 9, 67–74. [PMC free article] [PubMed] [Google Scholar]
- Goova MT, Li J, Kislinger T, Qu W, Lu Y, Bucciarelli LG, Nowygrod S, Wolf BM, Caliste X, Yan SF, Stern DM, Schmidt AM, 2001. Blockade of Receptor for Advanced Glycation End-Products Restores Effective Wound Healing in Diabetic Mice. The American Journal of Pathology 159, 513–525. 10.1016/S0002-9440(10)61723-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gorisse L, Pietrement C, Vuiblet V, Schmelzer CEH, Köhler M, Duca L, Debelle L, Fornès P, Jaisson S, Gillery P, 2016. Protein carbamylation is a hallmark of aging. Proc Natl Acad Sci USA 113, 1191–1196. 10.1073/pnas.1517096113 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Gross M-L, Piecha G, Bierhaus A, Hanke W, Henle T, Schirmacher P, Ritz E, 2011. Glycated and carbamylated albumin are more “nephrotoxic” than unmodified albumin in the amphibian kidney. American Journal of Physiology-Renal Physiology 301, F476–F485. 10.1152/ajprenal.00342.2010 [DOI] [PubMed] [Google Scholar]
- Guilbaud A, Niquet-Leridon C, Boulanger E, Tessier FJ, 2016. How Can Diet Affect the Accumulation of Advanced Glycation End-Products in the Human Body? Foods 5, E84. 10.3390/foods5040084 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Ha E, Bang J-H, Son JN, Cho H-C, Mun K-C, 2010. Carbamylated albumin stimulates microRNA-146, which is increased in human renal cell carcinoma. Mol Med Rep 3, 275–279. 10.3892/mmr_00000251 [DOI] [PubMed] [Google Scholar]
- Han JS, Kim YS, Chin HJ, Jeon US, Ahn C, Kim S, Lee JS, Jang I-J, Shin S-G, 1997. Temporal changes and reversibility of carbamylated hemoglobin in renal failure. American Journal of Kidney Diseases 30, 36–40. 10.1016/S0272-6386(97)90562-X [DOI] [PubMed] [Google Scholar]
- Harmel R, Fiedler D, 2018. Features and regulation of non-enzymatic post-translational modifications. Nat Chem Biol 14, 244–252. 10.1038/nchembio.2575 [DOI] [PubMed] [Google Scholar]
- Hartog JWL, Willemsen S, van Veldhuisen DJ, Posma JL, van Wijk LM, Hummel YM, Hillege HL, Voors AA, for the BENEFICIAL investigators, 2011. Effects of alagebrium, an advanced glycation endproduct breaker, on exercise tolerance and cardiac function in patients with chronic heart failure. European Journal of Heart Failure 13, 899–908. 10.1093/eurjhf/hfr067 [DOI] [PubMed] [Google Scholar]
- Himmelfarb J, Vanholder R, Mehrotra R, Tonelli M, 2020. The current and future landscape of dialysis. Nat Rev Nephrol 16, 573–585. 10.1038/s41581-020-0315-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Holy EW, Akhmedov A, Speer T, Camici GG, Zewinger S, Bonetti N, Beer JH, Lüscher TF, Tanner FC, 2016. Carbamylated Low-Density Lipoproteins Induce a Prothrombotic State Via LOX-1. Journal of the American College of Cardiology 68, 1664–1676. 10.1016/j.jacc.2016.07.755 [DOI] [PubMed] [Google Scholar]
- Holzer M, Gauster M, Pfeifer T, Wadsack C, Fauler G, Stiegler P, Koefeler H, Beubler E, Schuligoi R, Heinemann A, Marsche G, 2011. Protein carbamylation renders high-density lipoprotein dysfunctional. Antioxid Redox Signal 14, 2337–2346. 10.1089/ars.2010.3640 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Hörkkö S, Huttunen K, Kervinen K, Kesäniemi Y, 1994. Decreased clearance of uraemic and mildly carbamylated low-density lipoprotein. European journal of clinical investigation. 10.1111/j.1365-2362.1994.tb00974.x [DOI] [PubMed] [Google Scholar]
- Hörkkö S, Huttunen K, Kesäniemi YA, 1995. Decreased clearance of low-density lipoprotein in uremic patients under dialysis treatment. Kidney International 47, 1732–1740. 10.1038/ki.1995.239 [DOI] [PubMed] [Google Scholar]
- Hörkkö S, Savolainen MJ, Kervinen K, Kesäniemi YA, 1992. Carbamylation-induced alterations in low-density lipoprotein metabolism. Kidney Int 41, 1175–1181. 10.1038/ki.1992.179 [DOI] [PubMed] [Google Scholar]
- Hudson BI, Lippman ME, 2018. Targeting RAGE Signaling in Inflammatory Disease. Annu. Rev. Med. 69, 349–364. 10.1146/annurev-med-041316-085215 [DOI] [PubMed] [Google Scholar]
- Ikizler TA, Flakoll PJ, Parker RA, Hakim RM, 1994. Amino acid and albumin losses during hemodialysis. Kidney Int 46, 830–837. 10.1038/ki.1994.339 [DOI] [PubMed] [Google Scholar]
- Jaisson S, Delevallée-Forte C, Touré F, Rieu P, Garnotel R, Gillery P, 2007. Carbamylated albumin is a potent inhibitor of polymorphonuclear neutrophil respiratory burst. FEBS Lett 581, 1509–1513. 10.1016/j.febslet.2007.03.008 [DOI] [PubMed] [Google Scholar]
- Jaisson S, Gorisse L, Pietrement C, Gillery P, 2012. Quantification of plasma homocitrulline using hydrophilic interaction liquid chromatography (HILIC) coupled to tandem mass spectrometry. Anal Bioanal Chem 402, 1635–1641. 10.1007/s00216-011-5619-6 [DOI] [PubMed] [Google Scholar]
- Jaisson S, Kazes I, Desmons A, Fadel F, Oudart J-B, Santos-Weiss ICRD, Millart H, Touré F, Rieu P, Gillery P, 2016. Homocitrulline as marker of protein carbamylation in hemodialyzed patients. Clinica Chimica Acta 460, 5–10. 10.1016/j.cca.2016.06.009 [DOI] [PubMed] [Google Scholar]
- Jaisson S, Kerkeni M, Santos-Weiss ICR, Addad F, Hammami M, Gillery P, 2015. Increased serum homocitrulline concentrations are associated with the severity of coronary artery disease. Clinical Chemistry and Laboratory Medicine (CCLM) 53. 10.1515/cclm-2014-0642 [DOI] [PubMed] [Google Scholar]
- Jaisson S, Lorimier S, Ricard-Blum S, Sockalingum GD, Delevallée-Forte C, Kegelaer G, Manfait M, Garnotel R, Gillery P, 2006. Impact of carbamylation on type I collagen conformational structure and its ability to activate human polymorphonuclear neutrophils. Chem Biol 13, 149–159. 10.1016/j.chembiol.2005.11.005 [DOI] [PubMed] [Google Scholar]
- Jaisson S, Pietrement C, Gillery P, 2018. Protein Carbamylation: Chemistry, Pathophysiological Involvement, and Biomarkers, in: Advances in Clinical Chemistry. Elsevier, pp. 1–38. 10.1016/bs.acc.2017.12.001 [DOI] [PubMed] [Google Scholar]
- Jaisson S, Pietrement C, Gillery P, 2011. Carbamylation-Derived Products: Bioactive Compounds and Potential Biomarkers in Chronic Renal Failure and Atherosclerosis. Clinical Chemistry 57, 1499–1505. 10.1373/clinchem.2011.163188 [DOI] [PubMed] [Google Scholar]
- Jankowski V, Saritas T, Kjolby M, Hermann J, Speer T, Himmelsbach A, Mahr K, Heuschkel MA, Schunk SJ, Thirup S, Winther S, Bottcher M, Nyegaard M, Nykjaer A, Kramann R, Kaesler N, Jankowski J, Floege J, Marx N, Goettsch C, 2021. Carbamylated sortilin associates with cardiovascular calcification in patients with chronic kidney disease. Kidney International. 10.1016/j.kint.2021.10.018 [DOI] [PubMed] [Google Scholar]
- Jelkmann W, 2008. “O”, erythropoietin carbamoylation versus carbamylation. Nephrol Dial Transplant 23, 3033; author reply 3033–3034. 10.1093/ndt/gfn342 [DOI] [PubMed] [Google Scholar]
- Johnson ML, Lalia AZ, Dasari S, Pallauf M, Fitch M, Hellerstein MK, Lanza IR, 2015. Eicosapentaenoic acid but not docosahexaenoic acid restores skeletal muscle mitochondrial oxidative capacity in old mice. Aging Cell 14, 734–743. 10.1111/acel.12352 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Jud P, Sourij H, 2019. Therapeutic options to reduce advanced glycation end products in patients with diabetes mellitus: A review. Diabetes Research and Clinical Practice 148, 54–63. 10.1016/j.diabres.2018.11.016 [DOI] [PubMed] [Google Scholar]
- Kalim S, Berg AH, Karumanchi SA, Thadhani R, Allegretti AS, Nigwekar S, Zhao S, Srivastava A, Raj D, Deo R, Frydrych A, Chen J, Sondheimer J, Shafi T, Weir M, Lash JP, CRIC Study Investigators, 2020. Protein carbamylation and chronic kidney disease progression in the Chronic Renal Insufficiency Cohort Study. Nephrol Dial Transplant gfaa347. 10.1093/ndt/gfaa347 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalim S, Karumanchi SA, Thadhani RI, Berg AH, 2014. Protein Carbamylation in Kidney Disease: Pathogenesis and Clinical Implications. American Journal of Kidney Diseases 64, 793–803. 10.1053/j.ajkd.2014.04.034 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalim S, Ortiz G, Trottier CA, Deferio JJ, Karumanchi SA, Thadhani RI, Berg AH, 2015. The Effects of Parenteral Amino Acid Therapy on Protein Carbamylation in Maintenance Hemodialysis Patients. Journal of Renal Nutrition 25, 388–392. 10.1053/j.jrn.2015.01.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalim S, Tamez H, Wenger J, Ankers E, Trottier CA, Deferio JJ, Berg AH, Karumanchi SA, Thadhani RI, 2013. Carbamylation of Serum Albumin and Erythropoietin Resistance in End Stage Kidney Disease. Clin J Am Soc Nephrol 14. [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalim S, Trottier CA, Wenger JB, Wibecan J, Ahmed R, Ankers E, Karumanchi SA, Thadhani R, Berg AH, 2016. Longitudinal Changes in Protein Carbamylation and Mortality Risk after Initiation of Hemodialysis. CJASN 11, 1809–1816. 10.2215/CJN.02390316 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kalim S, Wald R, Yan AT, Goldstein MB, Kiaii M, Xu D, Berg AH, Clish C, Thadhani R, Rhee EP, Perl J, 2018. Extended Duration Nocturnal Hemodialysis and Changes in Plasma Metabolite Profiles. Clin J Am Soc Nephrol 13, 436–444. 10.2215/CJN.08790817 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Kass DA, Shapiro EP, Kawaguchi M, Capriotti AR, Scuteri A, deGroof RC, Lakatta EG, 2001. Improved Arterial Compliance by a Novel Advanced Glycation End-Product Crosslink Breaker. Circulation 104, 1464–1470. 10.1161/hc3801.097806 [DOI] [PubMed] [Google Scholar]
- Kim HJ, Jeong MS, Jang SB, 2021. Molecular Characteristics of RAGE and Advances in Small-Molecule Inhibitors. IJMS 22, 6904. 10.3390/ijms22136904 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koeth RA, Kalantar-Zadeh K, Wang Z, Fu X, Tang WHW, Hazen SL, 2013. Protein Carbamylation Predicts Mortality in ESRD. JASN 24, 853–861. 10.1681/ASN.2012030254 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Koyama Y, Takeishi Y, Arimoto T, Niizeki T, Shishido T, Takahashi H, Nozaki N, Hirono O, Tsunoda Y, Nitobe J, Watanabe T, Kubota I, 2007. High Serum Level of Pentosidine, an Advanced Glycation End Product (AGE), is a Risk Factor of Patients with Heart Failure. Journal of Cardiac Failure 13, 199–206. 10.1016/j.cardfail.2006.11.009 [DOI] [PubMed] [Google Scholar]
- Kraus LM, Gaber L, Handorf CR, Marti HP, Kraus AP, 2001. Carbamoylation of glomerular and tubular proteins in patients with kidney failure: a potential mechanism of ongoing renal damage. Swiss Med Wkly 131, 139–134. https://doi.org/2001/11/smw-09691 [DOI] [PubMed] [Google Scholar]
- Lacson E, Wang W, Zebrowski B, Wingard R, Hakim RM, 2012. Outcomes Associated With Intradialytic Oral Nutritional Supplements in Patients Undergoing Maintenance Hemodialysis: A Quality Improvement Report. American Journal of Kidney Diseases 60, 591–600. 10.1053/j.ajkd.2012.04.019 [DOI] [PubMed] [Google Scholar]
- Lane TA, Burka ER, 1976. Decreased life span and membrane damage of carbamylated erythrocytes in vitro. Blood 47, 909–917. [PubMed] [Google Scholar]
- Lee JA, Lee HA, Sadler PJ, 1991. Uraemia: is urea more important than we think? Lancet 338, 1438–1440. 10.1016/0140-6736(91)92733-i [DOI] [PubMed] [Google Scholar]
- Lee TC, Gibson QH, 1981. Allosteric properties of carbamylated hemoglobins. J Biol Chem 256, 4570–4577. [PubMed] [Google Scholar]
- Legendre JM, Bergot A, Turzo A, Morin PP, Humphery-Smith I, 1998. [Hemoglobin alpha chain isoelectric point modification under the action of urea, sodium cyanate, succinic anhydride or diethylene triamine pentaacetic acid anhydride]. Pathol Biol (Paris) 46, 605–612. [PubMed] [Google Scholar]
- Lewis BS, Rixon KC, Harding JJ, 1986. Bendazac prevents cyanate binding to soluble lens proteins and cyanate-induced phase-separation opacities in vitro: a possible mechanism by which bendazac could delay cataract. Exp Eye Res 43, 973–9. [DOI] [PubMed] [Google Scholar]
- Lewis EJ, Greene T, Spitalewiz S, Blumenthal S, Berl T, Hunsicker LG, Pohl MA, Rohde RD, Raz I, Yerushalmy Y, Yagil Y, Herskovits T, Atkins RC, Reutens AT, Packham DK, Lewis JB, for the Collaborative Study Group, 2012. Pyridorin in Type 2 Diabetic Nephropathy. JASN 23, 131–136. 10.1681/ASN.2011030272 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Li PKT, Culleton BF, Ariza A, Do J-Y, Johnson DW, Sanabria M, Shockley TR, Story K, Vatazin A, Verrelli M, Yu AW, Bargman JM, 2013. Randomized, Controlled Trial of Glucose-Sparing Peritoneal Dialysis in Diabetic Patients. J Am Soc Nephrol 24, 1889–1900. 10.1681/ASN.2012100987 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Lin J, Tang Y, Kang Q, Feng Y, Chen A, 2012. Curcumin inhibits gene expression of receptor for advanced glycation end-products (RAGE) in hepatic stellate cells in vitro by elevating PPARγ activity and attenuating oxidative stress: Curcumin inhibits gene expression of RAGE in HSCs. British Journal of Pharmacology 166, 2212–2227. 10.1111/j.1476-5381.2012.01910.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- Little WC, Zile MR, Kitzman DW, Hundley WG, O’Brien TX, deGroof RC, 2005. The Effect of Alagebrium Chloride (ALT-711), a Novel Glucose Cross-Link Breaker, in the Treatment of Elderly Patients With Diastolic Heart Failure. Journal of Cardiac Failure 11, 191–195. 10.1016/j.cardfail.2004.09.010 [DOI] [PubMed] [Google Scholar]
- Machowska A, Sun J, Qureshi AR, Isoyama N, Leurs P, Anderstam B, Heimburger O, Barany P, Stenvinkel P, Lindholm B, 2016. Plasma Pentosidine and Its Association with Mortality in Patients with Chronic Kidney Disease. PLoS One 11, e0163826. 10.1371/journal.pone.0163826 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Maddock AL, Westenfelder C, 1996. Urea induces the heat shock response in human neuroblastoma cells. J Am Soc Nephrol 7, 275–282. 10.1681/ASN.V72275 [DOI] [PubMed] [Google Scholar]
- Maillard L-C, 1912. Action des acides amines sur les sucres: formation des mela- noidines par voie methodique. C R Seances Acad Sci III 154, 66–68. [Google Scholar]
- Meerwaldt R, Hartog JWL, Graaff R, Huisman RJ, Links TP, den Hollander NC, Thorpe SR, Baynes JW, Navis G, Gans ROB, Smit AJ, 2005. Skin Autofluorescence, a Measure of Cumulative Metabolic Stress and Advanced Glycation End Products, Predicts Mortality in Hemodialysis Patients. JASN 16, 3687–3693. 10.1681/ASN.2005020144 [DOI] [PubMed] [Google Scholar]
- Monhemi H, Tabaee SS, 2020. The effects of mutation and modification on the structure and stability of human lysozyme: A molecular link between carbamylation and atherosclerosis. J Mol Graph Model 100, 107703. 10.1016/j.jmgm.2020.107703 [DOI] [PubMed] [Google Scholar]
- Mori D, Matsui I, Shimomura A, Hashimoto N, Matsumoto A, Shimada K, Yamaguchi S, Oka T, Kubota K, Yonemoto S, Sakaguchi Y, Takahashi A, Shintani Y, Takashima S, Takabatake Y, Hamano T, Isaka Y, 2018. Protein carbamylation exacerbates vascular calcification. Kidney Int 94, 72–90. 10.1016/j.kint.2018.01.033 [DOI] [PubMed] [Google Scholar]
- Münch G, Westcott B, Menini T, Gugliucci A, 2012. Advanced glycation endproducts and their pathogenic roles in neurological disorders. Amino Acids 42, 1221–1236. 10.1007/s00726-010-0777-y [DOI] [PubMed] [Google Scholar]
- Nicolas C, Jaisson S, Gorisse L, Tessier FJ, Niquet-Léridon C, Jacolot P, Pietrement C, Gillery P, 2019. Carbamylation and glycation compete for collagen molecular aging in vivo. Sci Rep 9, 18291. 10.1038/s41598-019-54817-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Nicolas C, Jaisson S, Gorisse L, Tessier FJ, Niquet-Léridon C, Jacolot P, Pietrement C, Gillery P, 2018. Carbamylation is a competitor of glycation for protein modification in vivo. Diabetes & Metabolism 44, 160–167. 10.1016/j.diabet.2017.05.006 [DOI] [PubMed] [Google Scholar]
- Nowicki C, Santomé JA, 1981. Modification of lysine 69 reactivity in bovine growth hormone by carbamylation of its N-terminal group. Int J Pept Protein Res 18, 52–60. 10.1111/j.1399-3011.1981.tb02039.x [DOI] [PubMed] [Google Scholar]
- Oimomi M, Hatanaka H, Yoshimura Y, Yokono K, Baba S, Taketomi Y, 1987. Carbamylation of insulin and its biological activity. Nephron 46, 63–66. 10.1159/000184303 [DOI] [PubMed] [Google Scholar]
- Ok E, Basnakian AG, Apostolov EO, Barri YM, Shah SV, 2005. Carbamylated low-density lipoprotein induces death of endothelial cells: a link to atherosclerosis in patients with kidney disease. Kidney Int 68, 173–178. 10.1111/j.1523-1755.2005.00391.x [DOI] [PubMed] [Google Scholar]
- Park L, Raman KG, Lee KJ, Lu Y, Ferran LJ, Chow WS, Stern D, Schmidt AM, 1998. Suppression of accelerated diabetic atherosclerosis by the soluble receptor for advanced glycation endproducts. Nat Med 4, 1025–1031. 10.1038/2012 [DOI] [PubMed] [Google Scholar]
- Parveen A, Sultana R, Lee SM, Kim TH, Kim SY, 2021. Phytochemicals against antidiabetic complications: targeting the advanced glycation end product signaling pathway. Arch. Pharm. Res 44, 378–401. 10.1007/s12272-021-01323-9 [DOI] [PubMed] [Google Scholar]
- Perl J, Kalim S, Wald R, Goldstein MB, Yan AT, Noori N, Kiaii M, Wenger J, Chan C, Thadhani RI, Karumanchi SA, Berg AH, 2016. Reduction of carbamylated albumin by extended hemodialysis: Extended hemodialysis reduces carbamylated albumin. Hemodialysis International 20, 510–521. 10.1111/hdi.12435 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Pietrement C, Gorisse L, Jaisson S, Gillery P, 2013. Chronic Increase of Urea Leads to Carbamylated Proteins Accumulation in Tissues in a Mouse Model of CKD. PLoS ONE 8, e82506. 10.1371/journal.pone.0082506 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Plater ML, Goode D, Crabbe MJ, 1997. Ibuprofen protects alpha-crystallin against posttranslational modification by preventing protein cross-linking. Ophthalmic Res 29, 421–428. 10.1159/000268043 [DOI] [PubMed] [Google Scholar]
- Praschberger M, Hermann M, Laggner C, Jirovetz L, Exner M, Kapiotis S, Gmeiner BMK, Laggner H, 2013. Carbamoylation abrogates the antioxidant potential of hydrogen sulfide. Biochimie 95, 2069–2075. 10.1016/j.biochi.2013.07.018 [DOI] [PubMed] [Google Scholar]
- Rangaswami J, Tuttle K, Vaduganathan M, 2020. Cardio-Renal-Metabolic Care Models. Circulation: Cardiovascular Quality and Outcomes 13, e007264. 10.1161/CIRCOUTCOMES.120.007264 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Rao GN, Cotlier E, 1988. Aspirin prevents the nonenzymatic glycosylation and carbamylation of the human eye lens crystallins in vitro. Biochem Biophys Res Commun 151, 991–996. 10.1016/s0006-291x(88)80463-7 [DOI] [PubMed] [Google Scholar]
- Sadik NAH, Mohamed WA, Ahmed MI, 2012. The association of receptor of advanced glycated end products and inflammatory mediators contributes to endothelial dysfunction in a prospective study of acute kidney injury patients with sepsis. Mol Cell Biochem 359, 73–81. 10.1007/s11010-011-1001-4 [DOI] [PubMed] [Google Scholar]
- Sampath C, Rashid MR, Sang S, Ahmedna M, 2017. Green tea epigallocatechin 3-gallate alleviates hyperglycemia and reduces advanced glycation end products via nrf2 pathway in mice with high fat diet-induced obesity. Biomedicine & Pharmacotherapy 87, 73–81. 10.1016/j.biopha.2016.12.082 [DOI] [PubMed] [Google Scholar]
- Santos HO, Penha-Silva N, 2021. Translating the advanced glycation end products (AGEs) knowledge into real-world nutrition strategies. Eur J Clin Nutr. 10.1038/s41430-021-01028-8 [DOI] [PubMed] [Google Scholar]
- Sarnak MJ, Levey AS, Schoolwerth AC, Coresh J, Culleton B, Hamm LL, McCullough PA, Kasiske BL, Kelepouris E, Klag MJ, Parfrey P, Pfeffer M, Raij L, Spinosa DJ, Wilson PW, American Heart Association Councils on Kidney in Cardiovascular Disease, High Blood Pressure Research, Clinical Cardiology, and Epidemiology and Prevention, 2003. Kidney disease as a risk factor for development of cardiovascular disease: a statement from the American Heart Association Councils on Kidney in Cardiovascular Disease, High Blood Pressure Research, Clinical Cardiology, and Epidemiology and Prevention. Circulation 108, 2154–2169. 10.1161/01.CIR.0000095676.90936.80 [DOI] [PubMed] [Google Scholar]
- Schaffert L-N, Carter WG, 2020. Do Post-Translational Modifications Influence Protein Aggregation in Neurodegenerative Diseases: A Systematic Review. Brain Sciences 10, 232. 10.3390/brainsci10040232 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Schreier SM, Steinkellner H, Jirovetz L, Hermann M, Exner M, Gmeiner BMK, Kapiotis S, Laggner H, 2011. S-carbamoylation impairs the oxidant scavenging activity of cysteine: its possible impact on increased LDL modification in uraemia. Biochimie 93, 772–777. 10.1016/j.biochi.2011.01.007 [DOI] [PubMed] [Google Scholar]
- Shah SV, Apostolov EO, Ok E, Basnakian AG, 2008. Novel mechanisms in accelerated atherosclerosis in kidney disease. J Ren Nutr 18, 65–69. 10.1053/j.jrn.2007.10.014 [DOI] [PubMed] [Google Scholar]
- Shapiro RJ, 1993. Catabolism of low-density lipoprotein is altered in experimental chronic renal failure. Metabolism 42, 162–169. 10.1016/0026-0495(93)90030-r [DOI] [PubMed] [Google Scholar]
- Sharma BS, Prabhakaran V, Desai AP, Bajpai J, Verma RJ, Swain PK, 2019. Post-translational Modifications (PTMs), from a Cancer Perspective: An Overview. Oncogen 2. 10.35702/onc.10012 [DOI] [Google Scholar]
- Shaw DC, Stein WH, Moore S, 1964. Inactivation of Chymotrypsin by Cyanate. Journal of Biological Chemistry 239, PC671–PC673. 10.1016/S0021-9258(18)51734-8 [DOI] [PubMed] [Google Scholar]
- Shaykh M, Pegoraro AA, Mo W, Arruda JA, Dunea G, Singh AK, 1999. Carbamylated proteins activate glomerular mesangial cells and stimulate collagen deposition. J Lab Clin Med 133, 302–308. 10.1016/s0022-2143(99)90086-0 [DOI] [PubMed] [Google Scholar]
- Simm A, 2013. Protein glycation during aging and in cardiovascular disease. Journal of Proteomics 92, 248–259. 10.1016/j.jprot.2013.05.012 [DOI] [PubMed] [Google Scholar]
- Singh R, Barden A, Mori T, Beilin L, 2001. Advanced glycation end-products: a review. Diabetologia 44, 129–146. 10.1007/s001250051591 [DOI] [PubMed] [Google Scholar]
- Smyth DG, 1967. Carbamylation of amino and tyrosine hydroxyl groups. Preparation of an inhibitor of oxytocin with no intrinsic activity on the isolated uterus. J Biol Chem 242, 1579–1591. [PubMed] [Google Scholar]
- Sohouli MH, Fatahi S, Sharifi-Zahabi E, Santos HO, Tripathi N, Lari A, Pourrajab B, Kord-Varkaneh H, Găman M-A, Shidfar F, 2021. The Impact of Low Advanced Glycation End Products Diet on Metabolic Risk Factors: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Advances in Nutrition 12, 766–776. 10.1093/advances/nmaa150 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Speeckaert M, Van Biesen W, Delanghe J, Slingerland R, Wiecek A, Heaf J, Drechsler C, Lacatus R, Vanholder R, Nistor I, for the European Renal Best Practice Guideline Development Group on Diabetes in Advanced CKD, Bilo H, Bolignano D, Couchoud C, Covic A, Coentrao L, Sutter JD, Drechsler C, Gnudi L, Goldsmith D, Heaf J, Heimburger O, Jager K, Nacak H, Nistor I, Soler M, Tomson C, Vanhuffel L, Biesen WV, Laecke SV, Weekers L, Wiecek A, 2014. Are there better alternatives than haemoglobin A1c to estimate glycaemic control in the chronic kidney disease population? Nephrology Dialysis Transplantation 29, 2167–2177. 10.1093/ndt/gfu006 [DOI] [PubMed] [Google Scholar]
- Speer T, Owala FO, Holy EW, Zewinger S, Frenzel FL, Stähli BE, Razavi M, Triem S, Cvija H, Rohrer L, Seiler S, Heine GH, Jankowski V, Jankowski J, Camici GG, Akhmedov A, Fliser D, Lüscher TF, Tanner FC, 2014. Carbamylated low-density lipoprotein induces endothelial dysfunction. Eur Heart J 35, 3021–3032. 10.1093/eurheartj/ehu111 [DOI] [PubMed] [Google Scholar]
- Sun JT, Yang K, Lu L, Zhu ZB, Zhu JZ, Ni JW, Han H, Chen N, Zhang RY, 2016. Increased carbamylation level of HDL in end-stage renal disease: carbamylated-HDL attenuated endothelial cell function. American Journal of Physiology-Renal Physiology 310, F511–F517. 10.1152/ajprenal.00508.2015 [DOI] [PubMed] [Google Scholar]
- Tang WHW, Shrestha K, Wang Z, Borowski AG, Troughton RW, Klein AL, Hazen SL, 2013. Protein Carbamylation in Chronic Systolic Heart Failure: Relationship With Renal Impairment and Adverse Long-Term Outcomes. Journal of Cardiac Failure 19, 219–224. 10.1016/j.cardfail.2013.02.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Tessier FJ, Birlouez-Aragon I, 2012. Health effects of dietary Maillard reaction products: the results of ICARE and other studies. Amino Acids 42, 1119–1131. 10.1007/s00726-010-0776-z [DOI] [PubMed] [Google Scholar]
- Thornalley PJ, 2003. Use of aminoguanidine (Pimagedine) to prevent the formation of advanced glycation endproducts. Archives of Biochemistry and Biophysics 419, 31–40. 10.1016/j.abb.2003.08.013 [DOI] [PubMed] [Google Scholar]
- Toprak C, Yigitaslan S, 2019. Alagebrium and Complications of Diabetes Mellitus. Eurasian J Med 51, 285–292. 10.5152/eurasianjmed.2019.18434 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Trottier C, Perl J, Freeman M, Thadhani R, Berg A, Kalim S, 2018. Protein Carbamylation in Peritoneal Dialysis and the Effect of Low Glucose plus Amino Acid Solutions. Perit Dial Int 38, 149–152. 10.3747/pdi.2017.00176 [DOI] [PubMed] [Google Scholar]
- Van Lente F, McHugh A, Pippenger CE, 1986. Carbamylation of apo-aspartate aminotransferase: a possible mechanism for enzyme inactivation in uremic patients. Clinical Chemistry 32, 2107–2108. 10.1093/clinchem/32.11.2107a [DOI] [PubMed] [Google Scholar]
- Vanholder R, Gryp T, Glorieux G, 2018. Urea and chronic kidney disease: the comeback of the century? (in uraemia research). Nephrology Dialysis Transplantation 33, 4–12. 10.1093/ndt/gfx039 [DOI] [PubMed] [Google Scholar]
- Verbrugge FH, Tang WHW, Hazen SL, 2015. Protein carbamylation and cardiovascular disease. Kidney International 88, 474–478. 10.1038/ki.2015.166 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Veronese FM, Piszkiewicz D, Smith EL, 1972. Inactivation of Bovine Glutamate Dehydrogenase by Carbamyl Phosphate and Cyanate. Journal of Biological Chemistry 247, 754–759. 10.1016/S0021-9258(19)45671-8 [DOI] [PubMed] [Google Scholar]
- Vlassara H, 1997. Recent progress in advanced glycation end products and diabetic complications. Diabetes 46 Suppl 2, S19–25. 10.2337/diab.46.2.s19 [DOI] [PubMed] [Google Scholar]
- Vlassara H, Palace MR, 2003. Glycoxidation: the menace of diabetes and aging. Mt Sinai J Med 70, 232–241. [PubMed] [Google Scholar]
- Voziyan PA, Khalifah RG, Thibaudeau C, Yildiz A, Jacob J, Serianni AS, Hudson BG, 2003. Modification of Proteins In Vitro by Physiological Levels of Glucose. Journal of Biological Chemistry 278, 46616–46624. 10.1074/jbc.M307155200 [DOI] [PubMed] [Google Scholar]
- Wagner Z, Molnár M, Molnár GA, Tamaskó M, Laczy B, Wagner L, Csiky B, Heidland A, Nagy J, Wittmann I, 2006. Serum carboxymethyllysine predicts mortality in hemodialysis patients. Am J Kidney Dis 47, 294–300. 10.1053/j.ajkd.2005.10.010 [DOI] [PubMed] [Google Scholar]
- Walsh CT, Garneau-Tsodikova S, Gatto GJ, 2005. Protein Posttranslational Modifications: The Chemistry of Proteome Diversifications. Angew. Chem. Int. Ed 44, 7342–7372. 10.1002/anie.200501023 [DOI] [PubMed] [Google Scholar]
- Wang AY-M, Wong C-K, Yau Y-Y, Wong S, Chan IH-S, Lam CW-K, 2014. Skin Autofluorescence Associates With Vascular Calcification in Chronic Kidney Disease. Arterioscler Thromb Vasc Biol. 34, 1784–1790. 10.1161/ATVBAHA.114.303378 [DOI] [PubMed] [Google Scholar]
- Wang Z, Nicholls SJ, Rodriguez ER, Kummu O, Hörkkö S, Barnard J, Reynolds WF, Topol EJ, DiDonato JA, Hazen SL, 2007. Protein carbamylation links inflammation, smoking, uremia and atherogenesis. Nat Med 13, 1176–1184. 10.1038/nm1637 [DOI] [PubMed] [Google Scholar]
- Weiner DE, Tighiouart H, Ladik V, Meyer KB, Zager PG, Johnson DS, 2014. Oral Intradialytic Nutritional Supplement Use and Mortality in Hemodialysis Patients. American Journal of Kidney Diseases 63, 276–285. 10.1053/j.ajkd.2013.08.007 [DOI] [PubMed] [Google Scholar]
- Weisgraber KH, Innerarity TL, Mahley RW, 1978. Role of lysine residues of plasma lipoproteins in high affinity binding to cell surface receptors on human fibroblasts. J Biol Chem 253, 9053–9062. [PubMed] [Google Scholar]
- Wendt TM, Tanji N, Guo J, Kislinger TR, Qu W, Lu Y, Bucciarelli LG, Rong LL, Moser B, Markowitz GS, Stein G, Bierhaus A, Liliensiek B, Arnold B, Nawroth PP, Stern DM, D’Agati VD, Schmidt AM, 2003. RAGE Drives the Development of Glomerulosclerosis and Implicates Podocyte Activation in the Pathogenesis of Diabetic Nephropathy. The American Journal of Pathology 162, 1123–1137. 10.1016/S0002-9440(10)63909-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Wistow G, Turnell B, Summers L, Slingsby C, Moss D, Miller L, Lindley P, Blundell T, 1983. X-ray analysis of the eye lens protein gamma-II crystallin at 1.9 A resolution. J Mol Biol 170, 175–202. 10.1016/s0022-2836(83)80232-0 [DOI] [PubMed] [Google Scholar]
- Wolfe RA, Ashby VB, Milford EL, Ojo AO, Ettenger RE, Agodoa LY, Held PJ, Port FK, 1999. Comparison of mortality in all patients on dialysis, patients on dialysis awaiting transplantation, and recipients of a first cadaveric transplant. N Engl J Med 341, 1725–1730. 10.1056/NEJM199912023412303 [DOI] [PubMed] [Google Scholar]
- Xiao S, Wagner L, Mahaney J, Baylis C, 2001. Uremic levels of urea inhibit L-arginine transport in cultured endothelial cells. Am J Physiol Renal Physiol 280, F989–995. 10.1152/ajprenal.2001.280.6.F989 [DOI] [PMC free article] [PubMed] [Google Scholar]
- Yancey PH, Clark ME, Hand SC, Bowlus RD, Somero GN, 1982. Living with Water Stress: Evolution of Osmolyte Systems. Science 217, 1214–1222. 10.1126/science.7112124 [DOI] [PubMed] [Google Scholar]
- Zhou Q, Cheng K-W, Gong J, Li ETS, Wang M, 2019. Apigenin and its methylglyoxal-adduct inhibit advanced glycation end products-induced oxidative stress and inflammation in endothelial cells. Biochemical Pharmacology 166, 231–241. 10.1016/j.bcp.2019.05.027 [DOI] [PubMed] [Google Scholar]