Abstract
Carbamylation describes a non-enzymatic, posttranslational protein modification mediated by cyanate, a dissociation product of urea. When kidney function declines and urea accumulates, the burden of carbamylation naturally rises. Free amino acids may protect proteins from carbamylation, and protein carbamylation has been shown to increase in uremic patients with amino acid deficiencies. Carbamylation reactions are capable of altering the structure and functional properties of certain proteins, and have been directly implicated in the underlying mechanisms of various disease conditions. A broad range of studies has demonstrated how the irreversible binding of urea-derived cyanate to proteins in the human body causes inappropriate cellular responses leading to adverse outcomes such as accelerated atherosclerosis and inflammation. Given carbamylation’s relationship to urea and the evidence that it contributes to disease pathogenesis, measurements of carbamylated proteins may serve as useful quantitative biomarkers of time-averaged urea concentrations while also offering risk assessment in patients with kidney disease. Moreover, the link between carbamylated proteins and disease pathophysiology creates an enticing therapeutic target for reducing the rate of carbamylation. This article reviews the biochemistry of the carbamylation reaction, its role in specific diseases, and the potential diagnostic and therapeutic implications of these findings based on recent advances.
Keywords: Carbamylation, uremia, posttranslational protein modification (PTM), pathophysiology, chronic kidney disease (CKD), end-stage renal disease (ESRD), cyanate, urea, inflammation, carbamoylation
Background
Proteins in the human body, in both health and disease, are exposed to chemical reactions capable of altering their structural and functional properties. Spontaneous post-translational protein modifications are caused by the non-enzymatic attachment of reactive molecules to protein functional groups, as seen, for example, in glycation reactions. Because post-translational modifications are capable of changing protein structure and function, they can create a mechanistic chemical link to the adverse pathophysiology underlying certain metabolic diseases.
Carbamylation is a protein modification that results from constant exposure to urea and its byproduct, cyanate, which both increase as kidney function declines. Urea-driven carbamylation reactions occur not only on proteins but also on free amino acids, and these targets may compete with each other for binding such that amino acid deficiency can exacerbate protein carbamylation. Furthermore, protein carbamylation may not be solely related to urea; recent work shows that cyanate may also be generated by myeloperoxidase (MPO) and peroxide-catalyzed oxidation of thiocyanate (derived from diet and smoking) at sites of inflammation.
Just as glycation is known to contribute to pathologic sequelae in conditions such as diabetes mellitus, carbamylation has been shown to change the properties of various enzymes, hormones, and other proteins, ultimately contributing to the deleterious effects of reduced kidney function. Similar to measurement of glycated hemoglobin for glucose monitoring in diabetes mellitus, measurement of protein carbamylation offers a time-averaged record of urea concentration and amino acid deficiency, thus characterizing duration and severity of kidney disease, as well as assessing the adequacy of kidney replacement therapies. This review describes the pathogenesis and clinical implications of protein carbamylation in kidney disease, paying special attention to recent advances made in the study of this process.
Case Vignette
A 58-year-old Hispanic man with end-stage renal disease (ESRD) resulting from chronic hypertensive nephrosclerosis was evaluated for worsening dyspnea on exertion. He had been adherent to his thrice weekly hemodialysis treatment regimen over the past 4 years and his latest Kt/V was 1.3. He had been dialyzed to his estimated dry weight of 72 kg at his last treatment and his recent pre-dialysis blood pressures averaged 145/85 mmHg. The patient did not have any prior cardiac history, and a nuclear stress test performed 3 years earlier was within normal limits. There was neither a history of smoking nor of dyslipidemia. On exam, he was without pulmonary or lower extremity edema. Electrocardiogram was significant for left ventricular hypertrophy. His cardiac troponin T level was elevated at 0.31 ng/ml (reference range, <0.03 ng/ml) but failed to rise on serial remeasurements. A myocardial perfusion scan was positive for ischemia. Cardiac catheterization revealed severe left main, left anterior descending, and left circumflex coronary artery disease. The patient was referred for coronary artery bypass graft surgery. A blood sample drawn with the patient’s informed consent for investigative purposes was tested for carbamylated albumin and shown to have 12 mmol carbamylated albumin per mole of total albumin (1.2% serum carbamylated albumin). This amount is well above previously reported average values seen in hemodialysis patients (median serum carbamylated albumin, 0.77%; interquartile range, 0.58%-0.93%).1,2
While well appreciated, the marked excess in cardiovascular burden observed in chronic kidney disease (CKD) patients remains poorly understood and is not entirely explained by traditional risk factors. Novel disease biomarkers may provide insights into the mechanisms of uremic cardiac morbidity and mortality in addition to offering new therapeutic targets. Protein carbamylation has recently been implicated in several disease pathways of uremia, including atherosclerosis and cardiovascular disease. As the mechanisms of protein carbamylation are becoming better understood, approaches to treating increased carbamylation burden and its sequelae are emerging, carrying the potential for meaningful clinical applications into the future.
Pathogenesis
Biochemistry of carbamylation and its link to kidney disease
In 1828 Friedrich Wöhler discovered that urea could be synthesized by reacting cyanate with ammonia, and in 1895 it was found that, under physiologic conditions, urea slowly dissociates into cyanate and its tautomer, isocyanate.3,4 Isocyanate is a highly reactive electrophile that quickly reacts with nucleophilic groups such as primary amines and free sulfhydryls, and by 1949 F. Schutz suggested that urea-derived cyanate could react with the amine and sulfhydryl groups on proteins and free amino acids.5 This claim was shown to be true in 1960 when George Stark et al observed that when ribonuclease is incubated with concentrated urea, a modified form accumulates that is less positively charged. Subsequent experiments found that lysines in the protein had been irreversibly modified to N(6)-carbamoyl-L-lysine (homocitrulline).6 Stark and others demonstrated that cyanate can produce irreversible modifications of primary amines and reversible modifications of thiols, hydroxyls, phenols, and imidazole groups.6-10 The net result of these reactions, referred to as carbamylation, is the addition of a “carbamoyl” moiety (−CONH2) to a functional group. Note that some authorities prefer the term carbamoylation to describe this reaction, but for this review we will use the term more commonly used throughout the biomedical literature, carbamylation.11,12
Because carbamylations of amines are stable, proteins may accumulate these on their amino-terminal α-amino groups or the ε-amino groups of lysine side chains throughout their lifespan. Free amino acids may also be carbamylated on their α-amino group or on nucleophilic groups on their side chains.8,13 Theoretically, carbamylation can occur on any nucleophilic amino acid side chain moiety, depending on its solvent accessibility, nucleophilicity, and the pKa of the group (carbamylation can only occur on amines in their uncharged state).14 As a result, carbamylations have been detected on multiple lysines within proteins, however, due to varying susceptibilities, the relative degree of carbamylation at different sites varies widely.2,15 Furthermore, because of differences in pKa, α-amino groups of free amino acids react about 100 times faster than lysine side chain ε-amino groups on proteins at physiologic pH.8,16
The spontaneous dissociation of urea into cyanate and ammonium depends upon pH and temperature. Under physiologic conditions, the equilibrium between urea and cyanate favors urea, with the cyanate-urea ratio averaging less than 1:100.12,17,18 Nevertheless, because urea levels in the body are relatively high compared to many other biomolecules, significant amounts of cyanate can be generated. When urea concentrations increase with declining kidney function, so does the generation of cyanate, creating a pathologic state that promotes protein carbamylation. The plasma concentration of isocyanate in healthy individuals is about 45 nmol/L, and in uremic patients it reaches 140 nmol/L.19 While this concentration may seem low compared to other biomolecules, recall that urea dissociation is constantly generating more cyanate, which rapidly binds to nearby proteins and amino acids, and thus the rate of protein and amino acid carbamylation may become quite significant. Kraus et al demonstrated that serum concentrations of many carbamylated free amino acids in patients with ESRD actually exceed the concentrations of their unmodified precursors.20
Biochemical rationale for the pathologic effects of protein carbamylation
A number of studies published over the past several decades have demonstrated that carbamylation causes changes in the physical properties of proteins, thus suggesting its involvement in molecular and cellular dysfunction.6,21 A major chemical effect of carbamylation is neutralization of positively charged lysines, which changes protein-water interactions and alters ionic interactions on the protein surface.12 Such changes can alter secondary and tertiary structures, leading to functional conformational changes. In the 1960s, the first enzyme shown to be inactivated by carbamylation was glutamate dehydrogenase.22 At the same time, several other groups were able to demonstrate the wide ranging effects of protein carbamylation.16 For example, investigators demonstrated that carbamylation of pepsinogen transforms the positively charged ε-NH2 groups on its lysine side chains into uncharged homocitrulline side chains .23 This alteration results in changes in electrophoretic mobility and altered enzymatic activity.23 More recent studies demonstrated that carbamylation of type I collagen induces conformational changes within its triple helix structure, altering collagen interactions with inflammatory cells and sensitivity to collagenases.24,25 Others have demonstrated that carbamylated erythropoietin loses its erythropoietic and angiogenic effects, yet maintains several of its tissue-protective properties.26-28 Further research has capitalized on this finding, testing whether carbamylated erythropoietin may be used as a non-hematopoietic, tissue-protective agent in ischemic injuries.29 Dozens of studies through the years have shown similar results implicating carbamylation in changes in protein charge,30 conformation,31,32 stability,33 enzyme and hormone activity,34-38 binding properties,39-41 receptor-drug interaction10,42, and cellular expression and responses.43-48 Lastly, tissue culture and animal model studies of the effects of carbamylated albumin have suggested that carbamylation gives albumin nephrotoxic and neutrophil-inactivating properties, providing further evidence that carbamylation of even a minor fraction of a normal protein may still confer significant pathogenic properties to the protein. 43,44,49
The prevalence of protein carbamylation throughout the body was recently illustrated in a study by Pietrement et al.50 In a chronic kidney disease (CKD) animal model, the authors were able to quantitatively demonstrate using liquid chromatography–tandem mass spectrometry that carbamylation occurrs in a diverse set of tissues including aorta, kidney, bone, skin, liver, and heart, with greater accumulations in the long-lived extracellular matrix proteins. While there appeared to be a basal level of carbamylation occurring in control mice without kidney disease, there was a 2-fold increase in carbamylation burden in 75% nephrectomized mice at 20 weeks in all the aforementioned tissues. Given the ubiquity of urea in all tissue compartments and the potential resulting effects of carbamylation on all of the body’s organs, the disease implications of carbamylation do indeed seem far reaching.
In addition to the effects of carbamylation on proteins, this chemical modification may also have consequences on the bioavailability and pharmacologic properties of free amino acids. When the α-amino group of a free amino acid is irreversibly carbamylated, it cannot participate in protein synthesis or be catabolized as an energy source.18,20 In patients with kidney disease, the proportion of carbamylated plasma free amino acids that accumulate may be significant,18,20 thus raising the possibility that functional amino acid depletion via carbamylation exacerbates the protein energy wasting associated with kidney disease and long-term kidney replacement therapies.51-53 Lastly, S-carbamylation of free sulfhydryl groups on cysteine and glutathione has been shown to interfere with their antioxidant functions, raising the possibility that carbamylation contributes to the thiol oxidative stress associated with uremic states. 54-60
Mechanistic role of carbamylation in specific disease processes
Heart disease
Recent studies from tissue culture and animal models give compelling evidence that protein carbamylation is mechanistically involved in the excessive cardiovascular disease burden associated with kidney failure. Multiple studies have suggested that carbamylation can contribute to atherosclerosis and cardiovascular risk via its effects on lipoproteins, collagen, fibrin, proteoglycans, and fibronectin (see Figure 1 and Table 1).61
Fig. 1.
The chemical pathophysiology of carbamylation in uremia, and the hypothesized pathogenesis of carbamylation-associated disease complications.
Table 1.
Carbamylation of proteins promoting atherosclerosis
| Carbamylated Substrate |
Effect | References |
|---|---|---|
| LDL | Diminishes LDL-receptor recognition/clearance of LDL | 62,65 |
| Triggers macrophage scavenger recognition | 62,69 | |
| Promotes foam cell formation | 62 | |
| Promotes endothelial/ monocyte adhesion | 72 | |
| Induces endothelial apoptosis/ endothelial progenitor cell senescence |
62,71,72,128 | |
| Stimulates vascular smooth muscle proliferation | 62,71,73 | |
| Increases oxidative stress | 128 | |
| Promotes auto-antibodies | 96 | |
| HDL | Promotes foam cell formation | 74 |
| Collagen | Decreases ability to polymerize into normal fibrils | 24 |
| Increases release of matrix metalloproteinase 9 by monocytes | 48 | |
| Increases adhesion of monocytes | 48 | |
| Fibrin | Increases basement membrane thickening | 61 |
| Proteoglycans, fibronectin |
Alters ECM via altered collagen and basement membrane affinity |
61 |
Abbreviations: ECM, extracellular matrix; LDL, low-density lipoprotein; HDL, high-density lipoprotein
Carbamylation of low-density lipoprotein (LDL) may enhance its atherogenic properties partly by decreasing its binding to the LDL-receptor62 and by preventing its clearance from circulation (a phenomenon seen in both animal models of chronic kidney failure63,64 as well as in uremic patients).65-68 Carbamylated LDL has also been shown to readily bind macrophage scavenger receptors, facilitating foam cell formation and inflammatory signaling.62,69,70 Moreover, carbamylated LDL can induce endothelial cell apoptosis71,72 and stimulate vascular smooth muscle proliferation73 while carbamylated collagen increases release of matrix metalloproteinases by monocytes, possibly stimulating remodeling of the extracellular matrix.24,48 Carbamylation of high-density lipoprotein (HDL) similarly appears to confer atherogenic properties,74 and there are studies suggesting that cyanate itself has direct pathologic effects on endothelial cells.75,76 Most recently, epidemiologic studies have further established protein carbamylation as an independent risk factor for all-cause mortality and death from cardiac causes in both uremic and non-uremic populations (see Figure 2 and Recent Advances).2,62,77
Fig. 2.

Kaplan-Meier curve estimates of the incidence of all-cause mortality over 12 months in incident hemodialysis patients in the ArMORR (Accelerated Mortality on Kidney Replacement) cohort. Participants (n=187) were categorized into upper, middle, and lower tertiles according to percent serum carbamylated albumin (%C-Alb) values measured at the outset of the study. Adapted from Berg et al.2
From sickle cell disease to cataracts
The first demonstration of the deleterious effects of carbamylation in vivo came in the 1970s when it was proposed that sickle cell crisis could be prevented by treating sickle cell disease patients with urea or cyanate. The rationale for this novel therapy was based upon the fact that carbamylated hemoglobin S had a higher affinity for oxygen and did not sickle as readily as unmodified hemoglobin S.78-83 Unfortunately, patients being treated with cyanate were observed to later develop cataracts, and carbamylation was soon implicated.84 Subsequent human and animal studies confirmed the finding and demonstrated that cyanate treatment of lens proteins (crystallins) caused them to form inter-chain disulfide bonds and protein aggregates, thus explaining how carbamylation modifications made these proteins cataractogenic.32,85-92
Arthritis
While much of the literature has focused on carbamylation and its effects on the cardiorenal disease axis, an additional consequence of protein carbamylation appears to be the induction of autoantibody responses. Protein carbamylation creates potential neoepitopes that induce multiple autoantibody responses.93-96 Rheumatoid arthritis (RA) is a disease associated with multiple types of autoantibodies, and when investigators examined if anti-carbamylated protein antibody levels are elevated in RA patients, they found that anti-homocitrulline antibodies are present and at a higher level in individuals with RA and arthralgias compared to healthy controls. They also showed positive associations with joint damage and could predict disease development in “pre-RA” patients independent of established biomarkers.95,97 Similar results have been seen in juvenile idiopathic arthritis patients.98
Other Diseases
Because the carbamylation reaction can theoretically occur on any protein, it is not surprising that the reaction has been implicated in many additional disease processes. For example, carbamylated proteins at concentrations present in uremia have been shown to activate glomerular mesangial cells to a profibrogenic phenotype and stimulate collagen deposition.99 Thus, accelerated carbamylation due to kidney failure could conceivably further accelerate the progression of kidney disease, leading to a detrimental positive feedback loop. Examples exist in other diseases as well: carbamylation of the neurologically important τ protein has been reported to result in significantly diminished ability to induce tubulin assembly into microtubules, promoting abnormal polymers that are found in Alzheimer disease.100 When carbamylated albumin was added to mesangial cells, expression of microRNAs associated with human kidney cell carcinoma was found to markedly increase.43 Carbamylation of ceruloplasmin impairs ferroxidase and thus its antioxidant activity, with additional cardiovascular implications as a possible consequence.101
Lastly, urea dissociation is not the only source of cyanate in humans: cyanate may also be derived from the consumption and metabolism of cassava root. Cassava contains cyanogens that are metabolized to cyanate, and it has recently been shown that cassava consumption is associated with increased protein carbamylation and neurologic disease (ie, the paralytic disease Konzo).102-105 The connection between cassava, carbamylation, and neurologic disease is intriguing because it raises the possibility that carbamylation could be a contributor to neurologic complications associated with chronic uremia.
Carbamylated hemoglobin as a biomarker of uremia
Analogous to hemoglobin A1c in diabetes mellitus, when urea-derived cyanate stably attaches to a protein, its half-life is defined by that of the protein. In this sense, carbamylated proteins can give not only a sense of the total body “carbamylation burden” but also a measure of urea concentrations averaged over the lifespan of the protein. Given the possibility that carbamylation contributes directly to the pathogenesis of cardiovascular and other diseases associated with uremia, measurements of carbamylated proteins should behave as ideal biomarkers because of their direct, quantitative, and mechanistic relationships to urea concentrations and disease pathophysiology. A number of studies published over several decades have in fact shown this to be true, beginning with studies of carbamylated hemoglobin in patients with kidney disease.
In 1981 Flückiger et al observed that a sub-fraction of hemoglobin A1 was carbamylated, and that the proportion of carbamylated hemoglobin was related to patient’s blood urea concentrations.106 Several subsequent studies have confirmed this.107-110 It was later shown in a small cohort of dialysis subjects (n=55) that carbamylated hemoglobin is higher in individuals with Kt/V < 1.1 than in those with a Kt/V greater than this; also, as predicted, carbamylation level correlates negatively with both Kt/V and urea reduction ratio.111 Others have similarly demonstrated carbamylated hemoglobin could be a more accurate indicator of average urea between hemodialysis sessions than the standard indices of dialysis adequacy, Kt/V and URR.110,112 In contrast, in a study limited by sample size (n=7), Balion et al found significant variability over time in individuals’ carbamylated hemoglobin levels, with little association with markers of dialysis adequacy.46
A drawback to past studies of carbamylated hemoglobin and dialysis adequacy is that none of the studies went so far as to show carbamylated hemoglobin values were associated with disease outcomes. Without such outcomes studies, it is hard to discern the significance of carbamylated hemoglobin as a clinical biomarker. Furthermore, measurement of carbamylated hemoglobin in ESRD patients may be compromised by the significant variability in the age of erythrocytes in this patient population, in the same way that erythrocyte aging influences hemoglobin A1c levels.113,114 The age of erythrocytes in patients with ESRD depends upon their degree of ESRD-related anemia, dialysis-related red blood cell loss, erythropoietin prescription, and whether they have been recently transfused.113-116 Thus in ESRD, higher carbamylated hemoglobin levels might represent urea load or simply might indicate an older population of circulating red blood cells.
In addition to the application of carbamylated protein measurements as indicators of the adequacy of kidney replacement therapy, these time-averaged biomarkers may also be useful in discriminating between acute and longer-term increases in urea levels.117-119 When looking at individuals with acute, acute-on-chronic, or stable CKD, matched for degree of decrease in kidney function, carbamylated hemoglobin could sufficiently discern between groups, offering utility in identifying individuals with acute and possibly reversible kidney injury from those with chronic and likely stable kidney disease.120,121 Interestingly, in kidney transplant recipients, carbamylated hemoglobin concentration has been reported to decrease by 19.7% within 2 to 3 weeks posttransplantation, but this occurs with an average hemoglobin increase of 25%, making the true in vivo kinetics less clear.121
Recent Advances
Recent years have produced a number of important developments in the field of kidney disease and carbamylation. A first important technical advance was the application of mass spectrometry and proteomics to quantitatively measure global protein carbamylation and to screen for individual carbamylation modifications on proteins in tissue and in blood. Wang and colleagues demonstrated the use of amino acid analysis to allow quantification of protein-bound homocitrulline after protein hydrolysis.62 In this manner, tissues and plasma can be analyzed for their proportional amounts of lysine and its carbamylated form (homocitrulline), thus measuring global accumulated protein carbamylation in a given sample. This analytic method has allowed investigators to confirm the increases in tissue protein carbamylation in uremic states and to correlate them with clinical outcomes. More advanced proteomic techniques have also made it possible to study the significance of carbamylation modifications at specific sites on specific protein targets.15,50 Recently our group devised an assay using high-performance liquid chromatography and tandem mass spectrometry to measure the most abundant site of carbamylation on human serum albumin (the lysine at amino acid 549).2 Development of these new methods for the quantitative analysis of carbamylated proteins has made possible several important clinical studies showing the association between carbamylation of plasma proteins and clinical outcomes in patients with kidney disease.
While the correlation between plasma protein carbamylation and time-averaged urea concentrations provides intriguing evidence of the potential clinical utility of carbamylation measures in people with kidney disease, a more noteworthy association recently elucidated is that carbamylated protein levels are strongly predictive of mortality risk in ESRD. Using measurements of carbamylated albumin or serum protein homocitrulline-lysine ratio as markers of protein carbamylation burden, recent studies of both incident and prevalent hemodialysis patients have shown associations between elevated protein carbamylation and all-cause and cardiovascular mortality in 3 distinct hemodialysis cohorts.2,77 Direct comparisons of carbamylated albumin and homocitrulline as assays of total carbamylation burden are lacking but may be instructive as the field moves forward.
New clinical correlation studies in patients with varying degrees of kidney disease may provide clues as to the pathologic mechanisms underlying the link between protein carbamylation and mortality. In one study, serum protein carbamylation levels in patients with chronic systolic heart failure were shown to be significantly correlated with concentrations of brain natriuretic peptide, possibly invoking an association between carbamylation and progressive ventricular dysfunction.122 In a separate study, serum carbamylated albumin levels were furthermore strongly associated with erythropoietin resistance and mortality in incident dialysis patients after controlling for traditional risk factors such as markers of inflammation and iron indices.1 Together, these observations suggest multiple mechanistic pathways that may be contributing to cardiac death in patients with ESRD.
Another recently proposed concept regarding the pathophysiology of uremic carbamylation is the hypothesis that free amino acids compete with proteins for reaction with cyanate, in essence protecting proteins from the carbamylation reaction. Cyanate’s affinity for the α–amino groups on free amino acids is far greater than that of lysine side chains on proteins, and thus free amino acids compete with proteins for binding to cyanate, and can act as natural ambient scavengers for carbamylation. A corollary to this hypothesis is that amino acid deficiencies will make uremic patients susceptible to even greater degrees of carbamylation, which is noteworthy as free amino acids can become depleted in uremic patients due to protein-energy wasting and hemodialysis.51,52,123 Recent observational work has found that protein carbamylation in dialysis patients is in fact associated with free amino acid deficiencies.2 Furthermore, it was confirmed using in vitro and in vivo mouse model experiments that urea-induced carbamylation is intensified by amino acid deficiencies.2 Thus, it has been hypothesized that amino acid therapy, in select individuals, may reduce the risk of uremic complications by reducing protein carbamylation. This hypothesis is currently under investigation in a first-in-human clinical trial of amino acid therapy for the reduction of carbamylation in hemodialysis patients (ClinicalTrials.gov identifier NCT01612429).
While the aforementioned studies reported that protein carbamylation remains tightly linked with average serum urea nitrogen (SUN), perhaps more significant is the fact that carbamylation is a stronger predictor of mortality than pre-dialysis urea, Kt/V, or urea reduction ratio (URR) values,2 suggesting it is potentially a more sensitive marker to urea load and clearance than simply measuring pre- and post-dialysis urea measures (a practice fraught with limitations).124,125 Although these various measures of dialysis adequacy are all interrelated, there are several reasons why carbamylated albumin measurements may be more tightly associated with outcomes. Rather than depending on single measurements of pre- and post-dialysis urea levels (Kt/V or URR), carbamylated albumin values represent a measure of average urea level over an extended period, thus better accounting for all the variations that occur through time. Furthermore, given the observed association between carbamylated albumin and amino acid levels and erythropoietin resistance, carbamylation measurements may integrate the effects of multiple pathophysiologic pathways: time-averaged uremia as well as protein energy wasting, anemia, and chronic inflammation. These advantages raise the possibility that measurements of circulating carbamylated proteins may represent a better indicator of the adequacy of dialysis than Kt/V or URR. If urea-induced carbamylation is a significant contributor to the pathologic sequelae of kidney disease, as recent studies have suggested, then patients receiving standard regimens of intermittent hemodialysis who are still suffering from excessive carbamylation may benefit from more intensive kidney replacement therapies such as nocturnal hemodialysis.
Although the studies showing associations between carbamylated protein levels and mortality have focused on patients receiving hemodialysis, the same pathophysiologic connections between chronic uremia, protein carbamylation, and resulting end-organ damage are presumably also present in patients on peritoneal dialysis and those with earlier stages of chronic kidney disease. Additional studies are needed to determine the relationship between carbamylated protein levels and clinical outcomes in different patient populations, and if the applications for these tests in therapeutic monitoring remain robust.
Finally, in addition to studies of uremia-associated protein carbamylation, new insights into alternative mechanisms that underlie carbamylation have also been made. Wang and others have demonstrated that beyond the effects of decreased kidney function and hyper-uremia, inflammation can create a unique source of cyanate and carbamylation via myeloperoxidase (MPO).62,126 MPO, which is primarily stored in granules of neutrophils, monocytes, and certain tissue macrophages, can react with hydrogen peroxide and environmentally derived thiocyanate to generate cyanate and thus promote the carbamylation reaction to occur. Thiocyanate derived from foods, smoke exposure, or even air pollution,127 can be oxidized through the MPO-catalyzed reaction with hydrogen peroxide yielding hypo-thiocyanate and cyanate. Given the marked increased levels of MPO at sites of inflammation, it has been demonstrated MPO can further promote carbamylation independent of urea load at sites of inflammation and atherosclerotic plaque. Although the study by Wang, et. al. suggested that tobacco smoking may contribute to MPO-catalyzed carbamylation,62 more recent studies have suggested that MPO may not be as significant a source in patients with kidney disease. In a study of incident hemodialysis patients, no correlation between serum carbamylated albumin levels and MPO enzyme activity could be found.2 Furthermore, in a study of patients with heart failure and variably preserved kidney function, it was shown that carbamylation, as measured by protein bound homocitrulline, appears to relate primarily to kidney indices (even minor deviations), with no significant association between protein-bound homocitrulline and systemic MPO measurement.122
Summary
A rich literature reflecting decades of investigation demonstrates the mechanistic and pathophysiologic involvement of protein carbamylation in the adverse outcomes of kidney disease, which remains a major public health burden. There is evidence that select carbamylated proteins could serve as useful biomarkers of disease and therapeutic response in kidney failure. For example, by integrating information on urea clearance, amino acid balance, and inflammation, elevated carbamylation measures potentially could identify dialysis patients at the greatest risk for uremic complications. This subset of patients may stand to benefit from interventions precisely targeted at reducing carbamylation through modified dialysis prescriptions, nutritional support, or anti-inflammatory therapies. Identifying ‘at risk’ patients like the one in the opening clinical vignette of this report and treating them with dialytic or nutritional interventions to attenuate carbamylation could work to counter the all too common complications individuals with kidney disease must currently endure. Improvements in analytics and a new growth of well-executed clinical studies have shown that carbamylation is both an important marker and effector of uremia and one that is worthy of additional investigation.
Acknowledgements
Support: S.K. has received support from the National Kidney Foundation Young Investigator award; R.T. receives support from NIH award K24 DK094872.; S.A.K. receives support from the Howard Hughes Medical Institute; A.H.B. received support from the American Diabetes Association Junior Faculty Award.
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 citable 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.
Financial Disclosure: Provisional applications for U.S. and International patents related to measurement of carbamylated albumin and treatment of carbamylation with amino acid scavengers have been filed by A.H.B., S.A.K., R.T., and their affiliated institutions. R.T. has served as a consultant to Fresenius Medical Care. Dr Kalim declares that he has no other relevant financial interests.
References
- 1.Kalim S, Tamez H, Wenger J, et al. Carbamylation of serum albumin and erythropoietin resistance in end stage kidney disease. Clin J Am Soc Nephrol. 2013;8:1927–34. doi: 10.2215/CJN.04310413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Berg AH, Drechsler C, Wenger J, et al. Carbamylation of serum albumin as a risk factor for mortality in patients with kidney failure. Sci Transl Med. 2013;5:175ra29. doi: 10.1126/scitranslmed.3005218. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Wöhler F. Ueber künstliche Bildung des Harnstoffs. Ann Phys. 1828;87:253–6. [Google Scholar]
- 4.Walker J, Hambly FJ. Transformation of ammonium cyanate into urea. J Chem Soc. 1895;67:746–67. [Google Scholar]
- 5.Shutz F. Cyanate. Experientia. 1949;5:133–72. doi: 10.1007/BF02174423. [DOI] [PubMed] [Google Scholar]
- 6.Stark GR, Stein WH, Moore S. Reaction of the cyanate present in aqueous urea with amino acids and proteins. J Biol Chem. 1960;235:3177–81. [Google Scholar]
- 7.Stark GR. On the reversible reaction of cyanate with sulfhydryl groups and the determination of NH2-terminal cysteine and cystine in proteins. J Biol Chem. 1964;239:1411–4. [PubMed] [Google Scholar]
- 8.Stark GR. Reactions of cyanate with functional groups of proteins. 3. Reactions with amino and carboxyl groups. Biochemistry. 1965;4:1030–6. doi: 10.1021/bi00882a008. [DOI] [PubMed] [Google Scholar]
- 9.Nyc JF, Mitchell HK. Synthesis of orotic acid from aspartic acid. J Am Chem Soc. 1947;69:1382–4. doi: 10.1021/ja01198a042. [DOI] [PubMed] [Google Scholar]
- 10.Smyth DG. Carbamylation of amino and tyrosine hydroxyl groups. Preparation of an inhibitor of oxytocin with no intrinsic activity on the isolated uterus. J Biol Chem. 1967;242:1579–91. [PubMed] [Google Scholar]
- 11.Jelkmann W. 'O', erythropoietin carbamoylation versus carbamylation. Nephrol Dial Transplant. 2008;23:3033. doi: 10.1093/ndt/gfn342. author reply -4. [DOI] [PubMed] [Google Scholar]
- 12.Jaisson S, Pietrement C, Gillery P. Carbamylation-derived products: bioactive compounds and potential biomarkers in chronic kidney failure and atherosclerosis. Clin Chem. 2011;57:1499–505. doi: 10.1373/clinchem.2011.163188. [DOI] [PubMed] [Google Scholar]
- 13.Stark GR. Modification of proteins with cyanate. Methods Enzymol. 1972;25:579–84. doi: 10.1016/S0076-6879(72)25057-1. [DOI] [PubMed] [Google Scholar]
- 14.Brotzel F, Chu YC, Mayr H. Nucleophilicities of primary and secondary amines in water. J Org Chem. 2007;72:3679–88. doi: 10.1021/jo062586z. [DOI] [PubMed] [Google Scholar]
- 15.Claxton JS, Sandoval PC, Liu G, et al. Endogenous Carbamylation of Kidney Medullary Proteins. PLoS One. 2014;8:e82655. doi: 10.1371/journal.pone.0082655. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Carreras J, Chabas A, Diederich D. Physiological and clinical implications of protein carbamylation. In: Grisolia S, Baguena R, Major F, editors. The Urea Cycle. John Wiley & Sons, Inc.; New York: 1976. pp. 501–48. [Google Scholar]
- 17.Dirnhuber P, Schutz F. The isomeric transformation of urea into ammonium cyanate in aqueous solutions. Biochem J. 1948;42:628–32. [PubMed] [Google Scholar]
- 18.Kraus LM, Kraus AP., Jr. Carbamoylation of amino acids and proteins in uremia. Kidney Int Suppl. 2001;78:S102–7. doi: 10.1046/j.1523-1755.2001.59780102.x. [DOI] [PubMed] [Google Scholar]
- 19.Nilsson L, Lundquist P, Kagedal B, Larsson R. Plasma cyanate concentrations in chronic kidney failure. Clin Chem. 1996;42:482–3. [PubMed] [Google Scholar]
- 20.Kraus LM, Jones MR, Kraus AP., Jr. Essential carbamoyl-amino acids formed in vivo in patients with end-stage kidney disease managed by continuous ambulatory peritoneal dialysis: isolation, identification, and quantitation. J Lab Clin Med. 1998;131:425–31. doi: 10.1016/s0022-2143(98)90143-3. [DOI] [PubMed] [Google Scholar]
- 21.Bobb D, Hofstee BH. Gel isoelectric focusing for following the successive carbamylations of amino groups in chymotrypsinogen A. Anal Biochem. 1971;40:209–17. doi: 10.1016/0003-2697(71)90094-7. [DOI] [PubMed] [Google Scholar]
- 22.Grisolia S. Enzyme regulation by substrate; rapid inactivation of glutamate dehydrogenase by carbamyl phosphate. Biochem Biophys Res Commun. 1968;32:56–9. doi: 10.1016/0006-291x(68)90425-7. [DOI] [PubMed] [Google Scholar]
- 23.Rimon S, Perlmann GE. Carbamylation of pepsinogen and pepsin. J Biol Chem. 1968;243:3566–72. [PubMed] [Google Scholar]
- 24.Jaisson S, Lorimier S, Ricard-Blum S, et al. Impact of carbamylation on type I collagen conformational structure and its ability to activate human polymorphonuclear neutrophils. Chem Biol. 2006;13:149–59. doi: 10.1016/j.chembiol.2005.11.005. [DOI] [PubMed] [Google Scholar]
- 25.Jaisson S, Larreta-Garde V, Bellon G, et al. Carbamylation differentially alters type I collagen sensitivity to various collagenases. Matrix Biol. 2007;26:190–6. doi: 10.1016/j.matbio.2006.10.008. [DOI] [PubMed] [Google Scholar]
- 26.Mun KC, Golper TA. Impaired biological activity of erythropoietin by cyanate carbamylation. Blood Purif. 2000;18:13–7. doi: 10.1159/000014403. [DOI] [PubMed] [Google Scholar]
- 27.Leist M, Ghezzi P, Grasso G, et al. Derivatives of erythropoietin that are tissue protective but not erythropoietic. Science. 2004;305:239–42. doi: 10.1126/science.1098313. [DOI] [PubMed] [Google Scholar]
- 28.Park KD, Mun KC, Chang EJ, Park SB, Kim HC. Inhibition of erythropoietin activity by cyanate. Scand J Urol Nephrol. 2004;38:69–72. doi: 10.1080/00365590310006291. [DOI] [PubMed] [Google Scholar]
- 29.Brines M, Cerami A. Erythropoietin-mediated tissue protection: reducing collateral damage from the primary injury response. J Intern Med. 2008;264:405–32. doi: 10.1111/j.1365-2796.2008.02024.x. [DOI] [PubMed] [Google Scholar]
- 30.Legendre JM, Bergot A, Turzo A, Morin PP, Humphery-Smith I. Modifications du point isoelectrique de la chaine-alpha de l'hemoglobine sous l'action de l'uree, du cyanate de sodium, de l'anhydride succinique ou de l'anhydride diethylene triamine pentaacetique. [Hemoglobin alpha chain isoelectric point modification under the action of urea, sodium cyanate, succinic anhydride or diethylene triamine pentaacetic acid anhydride] Pathol Biol (Paris) 1998;46:605–12. in French. [PubMed] [Google Scholar]
- 31.Nowicki C, Santome JA. Modification of lysine 69 reactivity in bovine growth hormone by carbamylation of its N-terminal group. Int J Pept Protein Res. 1981;18:52–60. doi: 10.1111/j.1399-3011.1981.tb02039.x. [DOI] [PubMed] [Google Scholar]
- 32.Beswick HT, Harding JJ. High-molecular-weight crystallin aggregate formation resulting from non-enzymic carbamylation of lens crystallins: relevance to cataract formation. Exp Eye Res. 1987;45:569–78. doi: 10.1016/s0014-4835(87)80067-2. [DOI] [PubMed] [Google Scholar]
- 33.Fazili KM, Mir MM, Qasim MA. Changes in protein stability upon chemical modification of lysine residues of bovine serum albumin by different reagents. Biochem Mol Biol Int. 1993;31:807–16. [PubMed] [Google Scholar]
- 34.Shaw DC, Stein WH, Moore S. Inactivation of chymotrypsin by cyanate. J Biol Chem. 1964;239:671–3. [PubMed] [Google Scholar]
- 35.De Furia FG, Miller DR, Cerami A, Manning JM. The effects of cyanate in vitro on red blood cell metabolism and function in sickle cell anemia. J Clin Invest. 1972;51:566–74. doi: 10.1172/JCI106845. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Van Lente F, McHugh A, Pippenger CE. Carbamylation of apo-aspartate aminotransferase: a possible mechanism for enzyme inactivation in uremic patients. Clin Chem. 1986;32:2107–8. [PubMed] [Google Scholar]
- 37.Veronese FM, Piszkiewicz D, Smith EL. Inactivation of bovine glutamate dehydrogenase by carbamyl phosphate and cyanate. J Biol Chem. 1972;247:754–9. [PubMed] [Google Scholar]
- 38.Oimomi M, Hatanaka H, Yoshimura Y, et al. Carbamylation of insulin and its biological activity. Nephron. 1987;46:63–6. doi: 10.1159/000184303. [DOI] [PubMed] [Google Scholar]
- 39.Weisgraber KH, Innerarity TL, Mahley RW. Role of lysine residues of plasma lipoproteins in high affinity binding to cell surface receptors on human fibroblasts. J Biol Chem. 1978;253:9053–62. [PubMed] [Google Scholar]
- 40.Lee TC, Gibson QH. Allosteric properties of carbamylated hemoglobins. J Biol Chem. 1981;256:4570–7. [PubMed] [Google Scholar]
- 41.Dengler TJ, Robertz-Vaupel GM, Dengler HJ. Albumin binding in uraemia: quantitative assessment of inhibition by endogenous ligands and carbamylation of albumin. Eur J Clin Pharmacol. 1992;43:491–9. doi: 10.1007/BF02285090. [DOI] [PubMed] [Google Scholar]
- 42.Erill S, Calvo R, Carlos R. Plasma protein carbamylation and decreased acidic drug protein binding in uremia. Clin Pharmacol Ther. 1980;27:612–8. doi: 10.1038/clpt.1980.87. [DOI] [PubMed] [Google Scholar]
- 43.Ha E, Bang JH, Son JN, Cho HC, Mun KC. Carbamylated albumin stimulates microRNA-146, which is increased in human kidney cell carcinoma. Mol Med Rep. 2010;3:275–9. doi: 10.3892/mmr_00000251. [DOI] [PubMed] [Google Scholar]
- 44.Jaisson S, Delevallee-Forte C, Toure F, et al. Carbamylated albumin is a potent inhibitor of polymorphonuclear neutrophil respiratory burst. FEBS Lett. 2007;581:1509–13. doi: 10.1016/j.febslet.2007.03.008. [DOI] [PubMed] [Google Scholar]
- 45.Maddock AL, Westenfelder C. Urea induces the heat shock response in human neuroblastoma cells. J AM Soc Nephrol. 1996;7:275–82. doi: 10.1681/ASN.V72275. [DOI] [PubMed] [Google Scholar]
- 46.Balion CM, Draisey TF, Thibert RJ. Carbamylated hemoglobin and carbamylated plasma protein in hemodialyzed patients. Kidney Int. 1998;53:488–95. doi: 10.1046/j.1523-1755.1998.00777.x. [DOI] [PubMed] [Google Scholar]
- 47.Lane TA, Burka ER. Decreased life span and membrane damage of carbamylated erythrocytes in vitro. Blood. 1976;47:909–17. [PubMed] [Google Scholar]
- 48.Garnotel R, Sabbah N, Jaisson S, Gillery P. Enhanced activation of and increased production of matrix metalloproteinase-9 by human blood monocytes upon adhering to carbamylated collagen. FEBS Lett. 2004;563:13–6. doi: 10.1016/S0014-5793(04)00233-9. [DOI] [PubMed] [Google Scholar]
- 49.Gross ML, Piecha G, Bierhaus A, et al. Glycated and carbamylated albumin is more "nephrotoxic" than unmodified albumin in the amphibian kidney. Am J Physiol Kidney Physiol. 2011;301:F476–85. doi: 10.1152/ajprenal.00342.2010. [DOI] [PubMed] [Google Scholar]
- 50.Pietrement C, Gorisse L, Jaisson S, Gillery P. Chronic Increase of Urea Leads to Carbamylated Proteins Accumulation in Tissues in a Mouse Model of CKD. PloS one. 2013;8:e82506. doi: 10.1371/journal.pone.0082506. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Raj DS, Oladipo A, Lim VS. Amino Acid and protein kinetics in kidney failure: an integrated approach. Semin Nephrol. 2006;26:158–66. doi: 10.1016/j.semnephrol.2005.09.006. [DOI] [PubMed] [Google Scholar]
- 52.Carrero JJ, Stenvinkel P, Cuppari L, et al. Etiology of the protein-energy wasting syndrome in chronic kidney disease: a consensus statement from the International Society of Kidney Nutrition and Metabolism (ISRNM) J Ren Nutr. 2013;23:77–90. doi: 10.1053/j.jrn.2013.01.001. [DOI] [PubMed] [Google Scholar]
- 53.Kalantar-Zadeh K, Cano NJ, Budde K, et al. Diets and enteral supplements for improving outcomes in chronic kidney disease. Nat Rev Nephrol. 2011;7:369–84. doi: 10.1038/nrneph.2011.60. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Schreier SM, Steinkellner H, Jirovetz L, et al. S-carbamoylation impairs the oxidant scavenging activity of cysteine: its possible impact on increased LDL modification in uraemia. Biochimie. 2011;93:772–7. doi: 10.1016/j.biochi.2011.01.007. [DOI] [PubMed] [Google Scholar]
- 55.Iciek M, Bilska A, Lorenc-Koci E, Wlodek LB, Sokolowska MM. The effect of uremic toxin cyanate (OCN−) on anaerobic sulfur metabolism and prooxidative processes in the rat kidney: a protective role of lipoate. Hum Exp Toxicol. 2010;30:1601–8. doi: 10.1177/0960327110394225. [DOI] [PubMed] [Google Scholar]
- 56.Sokolowska M, Niedzielska E, Iciek M, et al. The effect of the uremic toxin cyanate (CNO(−)) on anaerobic cysteine metabolism and oxidative processes in the rat liver: a protective effect of lipoate. Toxicol Mech Methods. 2011;21:473–8. doi: 10.3109/15376516.2011.556155. [DOI] [PubMed] [Google Scholar]
- 57.Go YM, Jones DP. Cysteine/cystine redox signaling in cardiovascular disease. Free Radic Biol Med. 2012;50:495–509. doi: 10.1016/j.freeradbiomed.2010.11.029. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 58.Przemyslaw W, Piotr K, Grazyna C, et al. Total, free, and protein-bound thiols in plasma of peritoneal dialysis and predialysis patients. Int Urol Nephrol. 2011;43:1201–9. doi: 10.1007/s11255-011-9905-1. [DOI] [PubMed] [Google Scholar]
- 59.Prakash M, Upadhya S, Prabhu R. Protein thiol oxidation and lipid peroxidation in patients with uraemia. Scand J Clin Lab Invest. 2004;64:599–604. doi: 10.1080/00365510410002869. [DOI] [PubMed] [Google Scholar]
- 60.Himmelfarb J, McMonagle E, McMenamin E. Plasma protein thiol oxidation and carbonyl formation in chronic kidney failure. Kidney Int. 2000;58:2571–8. doi: 10.1046/j.1523-1755.2000.00443.x. [DOI] [PubMed] [Google Scholar]
- 61.Mohar DS, Barseghian A, Haider N, Domanski M, Narula J. Atherosclerosis in chronic kidney disease: lessons learned from glycation in diabetes. Med Clin North Am. 2012;96:57–65. doi: 10.1016/j.mcna.2011.11.005. [DOI] [PubMed] [Google Scholar]
- 62.Wang Z, Nicholls SJ, Rodriguez ER, et al. Protein carbamylation links inflammation, smoking, uremia and atherogenesis. Nat Med. 2007;13:1176–84. doi: 10.1038/nm1637. [DOI] [PubMed] [Google Scholar]
- 63.Shapiro RJ. Catabolism of low-density lipoprotein is altered in experimental chronic kidney failure. Metabolism. 1993;42:162–9. doi: 10.1016/0026-0495(93)90030-r. [DOI] [PubMed] [Google Scholar]
- 64.Horkko S, Savolainen MJ, Kervinen K, Kesaniemi YA. Carbamylation-induced alterations in low-density lipoprotein metabolism. Kidney Int. 1992;41:1175–81. doi: 10.1038/ki.1992.179. [DOI] [PubMed] [Google Scholar]
- 65.Horkko S, Huttunen K, Kervinen K, Kesaniemi YA. Decreased clearance of uraemic and mildly carbamylated low-density lipoprotein. Eur J Clin Invest. 1994;24:105–13. doi: 10.1111/j.1365-2362.1994.tb00974.x. [DOI] [PubMed] [Google Scholar]
- 66.Horkko S, Huttunen K, Kesaniemi YA. Decreased clearance of low-density lipoprotein in uremic patients under dialysis treatment. Kidney Int. 1995;47:1732–40. doi: 10.1038/ki.1995.239. [DOI] [PubMed] [Google Scholar]
- 67.Apostolov EO, Ray D, Savenka AV, Shah SV, Basnakian AG. Chronic uremia stimulates LDL carbamylation and atherosclerosis. J Am Soc Nephrol. 2010;21:1852–7. doi: 10.1681/ASN.2010040365. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Shah SV, Apostolov EO, Ok E, Basnakian AG. Novel mechanisms in accelerated atherosclerosis in kidney disease. J Ren Nutr. 2008;18:65–9. doi: 10.1053/j.jrn.2007.10.014. [DOI] [PubMed] [Google Scholar]
- 69.Apostolov EO, Shah SV, Ray D, Basnakian AG. Scavenger receptors of endothelial cells mediate the uptake and cellular proatherogenic effects of carbamylated LDL. Arterioscler Thromb Vasc Biol. 2009;29:1622–30. doi: 10.1161/ATVBAHA.109.189795. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Apostolov EO, Shah SV, Ok E, Basnakian AG. Carbamylated low-density lipoprotein induces monocyte adhesion to endothelial cells through intercellular adhesion molecule-1 and vascular cell adhesion molecule-1. Arterioscler Thromb Vasc Biol. 2007;27:826–32. doi: 10.1161/01.ATV.0000258795.75121.8a. [DOI] [PubMed] [Google Scholar]
- 71.Ok E, Basnakian AG, Apostolov EO, Barri YM, Shah SV. Carbamylated low-density lipoprotein induces death of endothelial cells: a link to atherosclerosis in patients with kidney disease. Kidney Int. 2005;68:173–8. doi: 10.1111/j.1523-1755.2005.00391.x. [DOI] [PubMed] [Google Scholar]
- 72.Apostolov EO, Basnakian AG, Yin X, Ok E, Shah SV. Modified LDLs induce proliferation-mediated death of human vascular endothelial cells through MAPK pathway. Am J Physiol Heart Circ Physiol. 2007;292:H1836–46. doi: 10.1152/ajpheart.01079.2006. [DOI] [PubMed] [Google Scholar]
- 73.Asci G, Basci A, Shah SV, et al. Carbamylated low-density lipoprotein induces proliferation and increases adhesion molecule expression of human coronary artery smooth muscle cells. Nephrology (Carlton) 2008;13:480–6. doi: 10.1111/j.1440-1797.2008.00948.x. [DOI] [PubMed] [Google Scholar]
- 74.Holzer M, Gauster M, Pfeifer T, et al. Protein carbamylation renders high-density lipoprotein dysfunctional. Antioxid Redox Signal. 2011;14:2337–46. doi: 10.1089/ars.2010.3640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Xiao S, Wagner L, Mahaney J, Baylis C. Uremic levels of urea inhibit L-arginine transport in cultured endothelial cells. Am J Physiol Kidney Physiol. 2001;280:F989–95. doi: 10.1152/ajprenal.2001.280.6.F989. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.El-Gamal D, Holzer M, Gauster M, et al. Cyanate is a novel inducer of endothelial icam-1 expression. Antioxid Redox Signal. 2011;16:129–37. doi: 10.1089/ars.2011.4090. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Koeth RA, Kalantar-Zadeh K, Wang Z, et al. Protein carbamylation predicts mortality in ESRD. J Am Soc Nephrol. 2013;24:853–61. doi: 10.1681/ASN.2012030254. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Nalbandian RM, Henry RL, Barnhart MI, Camp FR., Jr. Sickle cell disease: clinical advances by the Murayama molecular hypothesis. Mil Med. 1972;137:215–20. [PubMed] [Google Scholar]
- 79.Dean J, Schechter AN. Sickle-cell anemia: molecular and cellular bases of therapeutic approaches (first of three parts) N Engl J Med. 1978;299:752–63. doi: 10.1056/NEJM197810052991405. [DOI] [PubMed] [Google Scholar]
- 80.Nalbandian RM, Nichols BM, Stehouwer EJ, Camp FR., Jr. Urea, urease, cyanate, and the sickling of hemoglobin S. Clin Chem. 1972;18:961–4. [PubMed] [Google Scholar]
- 81.Gillette PN, Peterson CM, Lu YS, Cerami A. Sodium cyanate as a potential treatment for sickle-cell disease. N Engl J Med. 1974;290:654–60. doi: 10.1056/NEJM197403212901204. [DOI] [PubMed] [Google Scholar]
- 82.Harkness DR, Roth S. Clinical evaluation of cyanate in sickle cell anemia. Prog Hematol. 1975;9:157–84. [PubMed] [Google Scholar]
- 83.Nigen AM, Njikam N, Lee CK, Manning JM. Studies on the mechanism of action of cyanate in sickle cell disease. Oxygen affinity and gelling properties of hemoglobin S carbamylated on specific chains. J Biol Chem. 1974;249:6611–6. [PubMed] [Google Scholar]
- 84.Nicholson DH, Harkness DR, Benson WE, Peterson CM. Cyanate-induced cataracts in patients with sickle-cell hemoglobinopathies. Arch Ophthalmol. 1976;94:927–30. doi: 10.1001/archopht.1976.03910030465005. [DOI] [PubMed] [Google Scholar]
- 85.Lapko VN, Smith DL, Smith JB. In vivo carbamylation and acetylation of water-soluble human lens alphaB-crystallin lysine 92. Protein Sci. 2001;10:1130–6. doi: 10.1110/ps.40901. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Beswick HT, Harding JJ. Conformational changes induced in bovine lens alpha-crystallin by carbamylation. Relevance to cataract. Biochem J. 1984;223:221–7. doi: 10.1042/bj2230221. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Yan H, Zhang J, Harding JJ. Identification of the preferentially targeted proteins by carbamylation during whole lens incubation by using radio-labelled potassium cyanate and mass spectrometry. Int J Ophthalmol. 2010;3:104–11. doi: 10.3980/j.issn.2222-3959.2010.02.03. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Zhang J, Yan H, Harding JJ, et al. Identification of the primary targets of carbamylation in bovine lens proteins by mass spectrometry. Curr Eye Res. 2008;33:963–76. doi: 10.1080/02713680802455959. [DOI] [PubMed] [Google Scholar]
- 89.Derham BK, Harding JJ. Alpha-crystallin as a molecular chaperone. Prog Retin Eye Res. 1999;18:463–509. doi: 10.1016/s1350-9462(98)00030-5. [DOI] [PubMed] [Google Scholar]
- 90.Liu X, Li S. Carbamylation of human lens gamma-crystallins: relevance to cataract formation. Yan Ke Xue Bao. 1993;9:136–42. 57. [PubMed] [Google Scholar]
- 91.Harding JJ, Rixon KC. Carbamylation of lens proteins: a possible factor in cataractogenesis in some tropical countries. Exp Eye Res. 1980;31:567–71. doi: 10.1016/s0014-4835(80)80015-7. [DOI] [PubMed] [Google Scholar]
- 92.Kern HL, Bellhorn RW, Peterson CM. Sodium cyanate-induced ocular lesions in the beagle. J Pharmacol Exp Ther. 1977;200:10–6. [PubMed] [Google Scholar]
- 93.Steinbrecher UP, Fisher M, Witztum JL, Curtiss LK. Immunogenicity of homologous low density lipoprotein after methylation, ethylation, acetylation, or carbamylation: generation of antibodies specific for derivatized lysine. J Lipid Res. 1984;25:1109–16. [PubMed] [Google Scholar]
- 94.Mydel P, Wang Z, Brisslert M, et al. Carbamylation-dependent activation of T cells: a novel mechanism in the pathogenesis of autoimmune arthritis. J Immunol. 2010;184:6882–90. doi: 10.4049/jimmunol.1000075. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Shi J, Knevel R, Suwannalai P, et al. Autoantibodies recognizing carbamylated proteins are present in sera of patients with rheumatoid arthritis and predict joint damage. Proc Natl Acad Sci U S A. 2011;108:17372–7. doi: 10.1073/pnas.1114465108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Kummu O, Turunen SP, Wang C, et al. Carbamyl adducts on low-density lipoprotein induce IgG response in LDLR−/− mice and bind plasma autoantibodies in humans under enhanced carbamylation. Antioxid Redox Signal. 2013;19:1047–62. doi: 10.1089/ars.2012.4535. [DOI] [PubMed] [Google Scholar]
- 97.Shi J, van Veelen PA, Mahler M, et al. Carbamylation and antibodies against carbamylated proteins in autoimmunity and other pathologies. Autoimmun Rev. 2014;13:225–30. doi: 10.1016/j.autrev.2013.10.008. [DOI] [PubMed] [Google Scholar]
- 98.Muller PC, Anink J, Shi J, et al. Anticarbamylated protein (anti-CarP) antibodies are present in sera of juvenile idiopathic arthritis (JIA) patients. Ann Rheum Dis. 2013;72:2053–5. doi: 10.1136/annrheumdis-2013-203650. [DOI] [PubMed] [Google Scholar]
- 99.Shaykh M, Pegoraro AA, Mo W, et al. Carbamylated proteins activate glomerular mesangial cells and stimulate collagen deposition. J Lab Clin Med. 1999;133:302–8. doi: 10.1016/s0022-2143(99)90086-0. [DOI] [PubMed] [Google Scholar]
- 100.Farias G, Gonzalez-Billault C, Maccioni RB. Immunological characterization of epitopes on tau of Alzheimer's type and chemically modified tau. Mol Cell Biochem. 1997;168:59–66. doi: 10.1023/a:1006838626730. [DOI] [PubMed] [Google Scholar]
- 101.Roxborough HE, Millar CA, McEneny J, Young IS. Carbamylation inhibits the ferroxidase activity of caeruloplasmin. Biochem Biophys Res Commun. 1995;214:1073–8. doi: 10.1006/bbrc.1995.2395. [DOI] [PubMed] [Google Scholar]
- 102.Kimani S, Moterroso V, Lasarev M, et al. Carbamoylation correlates of cyanate neuropathy and cyanide poisoning: relevance to the biomarkers of cassava cyanogenesis and motor system toxicity. Springerplus. 2013;2:647. doi: 10.1186/2193-1801-2-647. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Kimani S, Sinei K, Bukachi F, Tshala-Katumbay D, Maitai C. Memory deficits associated with sublethal cyanide poisoning relative to cyanate toxicity in rodents. Metab Brain Dis. 2014 Mar;29(1):105–12. doi: 10.1007/s11011-013-9459-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Kassa RM, Kasensa NL, Monterroso VH, et al. On the biomarkers and mechanisms of konzo, a distinct upper motor neuron disease associated with food (cassava) cyanogenic exposure. Food Chem Toxicol. 2010;49:571–8. doi: 10.1016/j.fct.2010.05.080. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Tor-Agbidye J, Palmer VS, Lasarev MR, et al. Bioactivation of cyanide to cyanate in sulfur amino acid deficiency: relevance to neurological disease in humans subsisting on cassava. Toxicol Sci. 1999;50:228–35. doi: 10.1093/toxsci/50.2.228. [DOI] [PubMed] [Google Scholar]
- 106.Fluckiger R, Harmon W, Meier W, Loo S, Gabbay KH. Hemoglobin carbamylation in uremia. N Engl J Med. 1981;304:823–7. doi: 10.1056/NEJM198104023041406. [DOI] [PubMed] [Google Scholar]
- 107.Kwan JT, Carr EC, Barron JL, Bending MR. Carbamylated haemoglobin in normal, diabetic and uraemic patients. Ann Clin Biochem. 1992;29:206–9. doi: 10.1177/000456329202900215. Pt 2. [DOI] [PubMed] [Google Scholar]
- 108.Kwan JT, Carr EC, Barron JL, Bending MR. Carbamylated haemoglobin--a retrospective index of time-averaged urea concentration. Nephrol Dial Transplant. 1993;8:565–7. doi: 10.1093/ndt/8.6.565. [DOI] [PubMed] [Google Scholar]
- 109.Kwan JT, Carr EC, Neal AD, et al. Carbamylated haemoglobin, urea kinetic modelling and adequacy of dialysis in haemodialysis patients. Nephrol Dial Transplant. 1991;6:38–43. doi: 10.1093/ndt/6.1.38. [DOI] [PubMed] [Google Scholar]
- 110.Tarif N, Shaykh M, Stim J, Arruda JA, Dunea G. Carbamylated hemoglobin in hemodialysis patients. Am J Kidney Dis. 1997;30:361–5. doi: 10.1016/s0272-6386(97)90280-8. [DOI] [PubMed] [Google Scholar]
- 111.Davenport A, Jones S, Goel S, Astley JP, Feest TG. Carbamylated hemoglobin: a potential marker for the adequacy of hemodialysis therapy in end-stage kidney failure. Kidney Int. 1996;50:1344–51. doi: 10.1038/ki.1996.447. [DOI] [PubMed] [Google Scholar]
- 112.Hasuike Y, Nakanishi T, Maeda K, et al. Carbamylated hemoglobin as a therapeutic marker in hemodialysis. Nephron. 2002;91:228–34. doi: 10.1159/000058397. [DOI] [PubMed] [Google Scholar]
- 113.Jiao Y, Okumiya T, Saibara T, Park K, Sasaki M. Abnormally decreased HbA1c can be assessed with erythrocyte creatine in patients with a shortened erythrocyte age. Diabetes Care. 1998;21:1732–5. doi: 10.2337/diacare.21.10.1732. [DOI] [PubMed] [Google Scholar]
- 114.Nakao T, Matsumoto H, Okada T, et al. Influence of erythropoietin treatment on hemoglobin A1c levels in patients with chronic kidney failure on hemodialysis. Intern Med. 1998;37:826–30. doi: 10.2169/internalmedicine.37.826. [DOI] [PubMed] [Google Scholar]
- 115.Wang X, Peesapati SK, Renedo MF, Moktan S. Hemoglobin A1c levels in non-diabetic patients with end-stage kidney disease (ESRD) receiving hemodialysis. J Endocrinol Invest. 2004;27:733–5. doi: 10.1007/BF03347514. [DOI] [PubMed] [Google Scholar]
- 116.Freedman BI. A critical evaluation of glycated protein parameters in advanced nephropathy: a matter of life or death: time to dispense with the hemoglobin A1C in end-stage kidney disease. Diabetes Care. 2012;35:1621–4. doi: 10.2337/dc12-0027. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Stim J, Shaykh M, Anwar F, et al. Factors determining hemoglobin carbamylation in kidney failure. Kidney Int. 1995;48:1605–10. doi: 10.1038/ki.1995.454. [DOI] [PubMed] [Google Scholar]
- 118.Kwan JT, Carr EC, Bending MR, Barron JL. Determination of carbamylated hemoglobin by high-performance liquid chromatography. Clin Chem. 1990;36:607–10. [PubMed] [Google Scholar]
- 119.Wynckel A, Randoux C, Millart H, et al. Kinetics of carbamylated haemoglobin in acute kidney failure. Nephrol Dial Transplant. 2000;15:1183–8. doi: 10.1093/ndt/15.8.1183. [DOI] [PubMed] [Google Scholar]
- 120.Davenport A, Jones SR, Goel S, Astley JP, Hartog M. Differentiation of acute from chronic kidney impairment by detection of carbamylated haemoglobin. Lancet. 1993;341:1614–7. doi: 10.1016/0140-6736(93)90757-8. [DOI] [PubMed] [Google Scholar]
- 121.Han JS, Kim YS, Chin HJ, et al. Temporal changes and reversibility of carbamylated hemoglobin in kidney failure. Am J Kidney Dis. 1997;30:36–40. doi: 10.1016/s0272-6386(97)90562-x. [DOI] [PubMed] [Google Scholar]
- 122.Tang WH, Shrestha K, Wang Z, et al. Protein carbamylation in chronic systolic heart failure: relationship with kidney impairment and adverse long-term outcomes. J Card Fail. 2013;19:219–24. doi: 10.1016/j.cardfail.2013.02.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Raj DS, Zager P, Shah VO, et al. Protein turnover and amino acid transport kinetics in end-stage kidney disease. Am J Physiol Endocrinol Metab. 2004;286:E136–43. doi: 10.1152/ajpendo.00352.2003. [DOI] [PubMed] [Google Scholar]
- 124.Tattersall J. Do we need another Kt/V? Nephrol Dial Transplant. 2013;28:1963–6. doi: 10.1093/ndt/gft215. [DOI] [PubMed] [Google Scholar]
- 125.Owen WF, Jr., Meyer KB, Schmidt G, Alfred H. Methodological limitations of the ESRD Core Indicators Project: an ESRD network's experience with implementing an ESRD quality survey. Medical Review Board of the ESRD Network of New England. Am J Kidney Dis. 1997;30:349–55. doi: 10.1016/s0272-6386(97)90278-x. [DOI] [PubMed] [Google Scholar]
- 126.Holzer M, Zangger K, El-Gamal D, et al. Myeloperoxidase-derived chlorinating species induce protein carbamylation through decomposition of thiocyanate and urea: novel pathways generating dysfunctional high-density lipoprotein. Antioxid Redox Signal. 2012;17:1043–52. doi: 10.1089/ars.2011.4403. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Zil a R, Rahman MA. Serum thiocyanate levels in smokers, passive smokers and never smokers. J Pak Med Assoc. 2006;56:323–6. [PubMed] [Google Scholar]
- 128.Carracedo J, Merino A, Briceno C, et al. Carbamylated low-density lipoprotein induces oxidative stress and accelerated senescence in human endothelial progenitor cells. FASEB J. 2011;25:1314–22. doi: 10.1096/fj.10-173377. [DOI] [PubMed] [Google Scholar]

